TW202505022A - Methods of producing tissue-derived epithelial organoids and uses thereof - Google Patents
Methods of producing tissue-derived epithelial organoids and uses thereof Download PDFInfo
- Publication number
- TW202505022A TW202505022A TW113122212A TW113122212A TW202505022A TW 202505022 A TW202505022 A TW 202505022A TW 113122212 A TW113122212 A TW 113122212A TW 113122212 A TW113122212 A TW 113122212A TW 202505022 A TW202505022 A TW 202505022A
- Authority
- TW
- Taiwan
- Prior art keywords
- tissue
- organoids
- derived epithelial
- hydrogel
- derived
- Prior art date
Links
Abstract
Description
本文所揭示之主題涉及組織來源之上皮類器官以及產生及使用此等類器官之方法。The subject matter disclosed herein relates to tissue-derived epithelial organoids and methods of generating and using such organoids.
組織來源之上皮類器官為三維 (3D) 多細胞橢球體,其重現 活體內組織之複雜性及功能,並已成為組織之生理相關 活體外模型。例如,腸類器官,也稱為“類腸 (enteroid)”或“類大腸 (colonoid)”,源自從原生腸組織 (primary intestinal tissue) 分離之成體幹細胞,並且可以輕鬆繁殖及冷凍保存以用於長期儲存。腸類器官可分化成各種腸細胞類型,進行上皮功能,諸如障壁維持、吸收、分泌及消化,並概括從其來源之患者之生物學特徵及臨床反應 (Clevers (2016) Cell 165, 1586–1597;Zachos 等人(2016) J Biol Chem 291, 3759–3766)。因此,許多腸類器官已被廣泛採用,取代傳統之轉化及永生化腸細胞株,從而產生基礎科學發現並促進在包括癌症生物學、傳染病及囊性纖維化的眾多領域中之轉化應用 (Clevers (2016);Schutgens 及 Clevers (2019) Annu Rev Pathology Mech Dis 15, 1–24)。 Tissue-derived epithelial organoids are three-dimensional (3D) multicellular spheroids that recapitulate the complexity and function of in vivo tissues and have become physiologically relevant in vitro models of tissues. For example, intestinal organoids, also called "enteroids" or "colonoids," are derived from adult stem cells isolated from primary intestinal tissue and can be easily propagated and cryopreserved for long-term storage. Intestinal organoids can differentiate into a variety of intestinal cell types, perform epithelial functions such as barrier maintenance, absorption, secretion, and digestion, and recapitulate the biological characteristics and clinical responses of the patients from which they are derived (Clevers (2016) Cell 165, 1586–1597; Zachos et al. (2016) J Biol Chem 291, 3759–3766). As a result, many intestinal organoids have been widely adopted to replace traditional transformed and immortalized intestinal cell lines, thereby generating basic science discoveries and promoting translational applications in many fields including cancer biology, infectious diseases, and cystic fibrosis (Clevers (2016); Schutgens and Clevers (2019) Annu Rev Pathology Mech Dis 15, 1–24).
組織來源之上皮類器官在諸如藥物開發等領域中的實施之挑戰係現有類器官培養技術難以規模化,需要繁瑣的手動方法或開發先進自動化基礎設施 (Louey 等人(2021) Slas Discov 26, 1138–1147)。在現有技術中,組織來源之上皮幹細胞 (從原生組織中分離或從已建立之類器官培養物中傳代) 係重懸於冷細胞外基質 (ECM) 溶液中,該溶液最常見為 CULTREX® 基底膜萃取物 (BME) 或 MATRIGEL® 水凝膠。將 ECM 沉積到盤表面上,然後溫熱以使 ECM 細胞溶液固化成表面接附之水凝膠圓頂,並以培養基覆蓋。每孔中之培養基每隔幾天更換一次,且類器官在 1-2 週時間內形成 (Mahe 等人Curr Protoc Mouse Biology 3, 217–240;Pleguezuelos‐Manzano 等人(2020) Curr Protoc Immunol 130, e106;Sato 等人(2009) Nature 459, 262–265;Sato 等人(2011) Gastroenterology 141, 1762–1772)。該技術難以規模化,因為它受到水凝膠圓頂形成可用表面積之限制,耗時且費力,並且容易出現使用者錯誤,並且水凝膠之擴散限制導致類器官生長及形態異質性 (Park 等人(2022) Nat Methods 19, 1449–1460;Ringel 等人(2020) Cell Stem Cell 26, 431-440.e8;Shin 等人(2020) iScience 23, 101372)。因此,本技術領域需要產生組織來源之上皮類器官之更有效及高生產量之方法。A challenge in the implementation of tissue-derived epithelial organoids in fields such as drug development is that existing organoid culture techniques are difficult to scale up, requiring either cumbersome manual methods or the development of advanced automation infrastructure (Louey et al. (2021) Slas Discov 26, 1138–1147). In existing techniques, tissue-derived epithelial stem cells (isolated from primary tissue or passaged from established organoid cultures) are resuspended in a cold extracellular matrix (ECM) solution, most commonly CULTREX® basement membrane extract (BME) or MATRIGEL® hydrogel. ECM is deposited onto the dish surface and then warmed to solidify the ECM cell solution into a surface-attached hydrogel dome and covered with medium. The medium in each well is changed every few days and organoids form within 1-2 weeks (Mahe et al. Curr Protoc Mouse Biology 3, 217–240; Pleguezuelos-Manzano et al. (2020) Curr Protoc Immunol 130, e106; Sato et al. (2009) Nature 459, 262–265; Sato et al. (2011) Gastroenterology 141, 1762–1772). The technique is difficult to scale up because it is limited by the surface area available for hydrogel dome formation, is time-consuming and labor-intensive, and is prone to user error, and the diffusion limitations of the hydrogel lead to heterogeneity in organoid growth and morphology (Park et al. (2022) Nat Methods 19, 1449–1460; Ringel et al. (2020) Cell Stem Cell 26, 431-440.e8; Shin et al. (2020) iScience 23, 101372). Therefore, there is a need for more efficient and high-throughput methods to generate tissue-derived epithelial organoids.
本文所揭示之主題涉及組織來源之上皮類器官及產生此等類器官之方法。本揭露進一步提供了使用組織來源之上皮類器官之方法及用於進行本文所揭示之方法之系統。The subject matter disclosed herein relates to tissue-derived epithelial organoids and methods of generating such organoids. The present disclosure further provides methods of using tissue-derived epithelial organoids and systems for performing the methods disclosed herein.
在某些實施例中,產生組織來源之上皮類器官之方法包括 (a) 使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物,(b) 將該水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物,以及 (c) 將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。在某些實施例中,使複數個組織來源之上皮幹細胞與該水凝膠接觸以產生該水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該方法進一步包括將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。In certain embodiments, a method of generating tissue-derived epithelial organoids comprises (a) contacting tissue-derived epithelial stem cells with hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture, (b) suspending the hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to generate a suspended hydrogel-tissue-derived epithelial stem cell mixture, and (c) culturing the suspended hydrogel-tissue-derived epithelial stem cell mixture in the culture medium to generate tissue-derived epithelial organoids. In some embodiments, a plurality of tissue-derived epithelial stem cells are contacted with the hydrogel to produce the hydrogel-tissue-derived epithelial stem cell mixture. In some embodiments, the method further comprises fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to produce a fragmented structure comprising the tissue-derived epithelial organoid.
在某些實施例中,該水凝膠係在與培養基接觸時 (upon contact) 固化。在某些實施例中,將該水凝膠-組織來源之上皮幹細胞混合物懸浮在該培養基中包括將含有該水凝膠-組織來源之上皮幹細胞混合物之分配裝置浸沒在該培養基中並將該水凝膠-組織來源之上皮幹細胞混合物分配到該培養基中。在某些實施例中,該培養基之溫度為約 25℃ 至約 50℃。在某些實施例中,該培養基之溫度為約 30℃ 至約 50℃。在某些實施例中,該培養基之溫度為約 30℃ 至約 40℃。在某些實施例中,該水凝膠-組織來源之上皮幹細胞混合物之溫度低於約 20℃。在某些實施例中,該水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 25℃。在某些實施例中,該水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 20℃。在某些實施例中,該水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 10℃。In some embodiments, the hydrogel solidifies upon contact with the culture medium. In some embodiments, suspending the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium includes immersing a dispensing device containing the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium and dispensing the hydrogel-tissue-derived epithelial stem cell mixture into the culture medium. In some embodiments, the temperature of the culture medium is about 25° C. to about 50° C. In some embodiments, the temperature of the culture medium is about 30° C. to about 50° C. In some embodiments, the temperature of the culture medium is about 30° C. to about 40° C. In some embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is less than about 20° C. In some embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is about 2° C. to about 25° C. In some embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is about 2° C. to about 20° C. In some embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is about 2° C. to about 10° C.
本揭露進一步提供了一種產生組織來源之上皮類器官之懸浮培養物之方法。在某些實施例中,該方法包括 (a) 將包含水凝膠及組織來源之上皮幹細胞之混合物引入到培養基中以產生經懸浮之混合物;以及 (b) 將該經懸浮之混合物在該培養基中培養以產生呈懸浮狀態之該等組織來源之上皮類器官。在某些實施例中,引入到該培養基中之該混合物包含該水凝膠及複數個組織來源之上皮幹細胞。在某些實施例中,該方法包括 (a) 將包含水凝膠及複數個組織來源之上皮幹細胞之混合物引入到培養基中以產生經懸浮之混合物,以及 (b) 將該混合物在該培養基中培養以產生呈懸浮狀態之該等組織來源之上皮類器官。在某些實施例中,該水凝膠係在與培養基接觸時固化。在某些實施例中,將該混合物引入該培養基中包含將含有該混合物之分配裝置浸沒在該培養基中並將該混合物分配到該培養基中。在某些實施例中,該培養基之溫度為約 25℃ 至約 50℃。在某些實施例中,該培養基之溫度為約 30℃ 至約 50℃。在某些實施例中,該培養基之溫度為約 25℃ 至約 40℃。在某些實施例中,該培養基之溫度為約 30℃ 至約 40℃。在某些實施例中,該混合物之溫度低於約 20℃。在某些實施例中,該混合物之溫度為約 2℃ 至約 25℃。在某些實施例中,該混合物之溫度為約 2℃ 至約 20℃。在某些實施例中,該混合物之溫度為約 2℃ 至約 10℃。在某些實施例中,該方法進一步包括將該經懸浮之混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。The present disclosure further provides a method for producing a suspended culture of tissue-derived epithelial organoids. In certain embodiments, the method comprises (a) introducing a mixture comprising a hydrogel and tissue-derived epithelial stem cells into a culture medium to produce a suspended mixture; and (b) culturing the suspended mixture in the culture medium to produce the tissue-derived epithelial organoids in a suspended state. In certain embodiments, the mixture introduced into the culture medium comprises the hydrogel and a plurality of tissue-derived epithelial stem cells. In some embodiments, the method comprises (a) introducing a mixture comprising a hydrogel and a plurality of tissue-derived epithelial stem cells into a culture medium to produce a suspended mixture, and (b) culturing the mixture in the culture medium to produce the tissue-derived epithelial organoids in a suspended state. In some embodiments, the hydrogel solidifies upon contact with the culture medium. In some embodiments, introducing the mixture into the culture medium comprises immersing a dispensing device containing the mixture in the culture medium and dispensing the mixture into the culture medium. In some embodiments, the temperature of the culture medium is about 25° C. to about 50° C. In some embodiments, the temperature of the culture medium is about 30° C. to about 50° C. In some embodiments, the temperature of the culture medium is about 25°C to about 40°C. In some embodiments, the temperature of the culture medium is about 30°C to about 40°C. In some embodiments, the temperature of the mixture is below about 20°C. In some embodiments, the temperature of the mixture is about 2°C to about 25°C. In some embodiments, the temperature of the mixture is about 2°C to about 20°C. In some embodiments, the temperature of the mixture is about 2°C to about 10°C. In some embodiments, the method further comprises fragmenting the suspended mixture to produce a fragmented structure comprising the tissue-derived epithelial organoid.
在某些實施例中,用於產生組織來源之上皮類器官之懸浮培養物之方法包括 (a) 使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物;(b) 將該水凝膠-組織來源之上皮幹細胞混合物沉積到基材上;(c) 將該水凝膠-組織來源之上皮幹細胞混合物固化以產生經固化之水凝膠-組織來源之上皮幹細胞混合物;(d) 將該經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物;以及 (e) 將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。在某些實施例中,使複數個組織來源之上皮幹細胞與該水凝膠接觸以產生該水凝膠-組織來源之上皮幹細胞混合物。例如但不限於,該方法包括 (a) 使複數個組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物,(b) 將該水凝膠-組織來源之上皮幹細胞混合物沉積到基材上,(c) 將該水凝膠-組織來源之上皮幹細胞混合物固化以產生經固化之水凝膠-組織來源之上皮幹細胞混合物,(d) 將該經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物,以及 (e) 將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。在某些實施例中,該方法進一步包括在將該經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在該培養基中之前,將該經固化之水凝膠-組織來源之上皮幹細胞混合物從該基材中取出。In certain embodiments, a method for generating a suspension culture of tissue-derived epithelial organoids comprises (a) contacting tissue-derived epithelial stem cells with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture; (b) depositing the hydrogel-tissue-derived epithelial stem cell mixture onto a substrate; (c) solidifying the hydrogel-tissue-derived epithelial stem cell mixture to generate a solidified hydrogel-tissue-derived epithelial stem cell mixture; (d) (e) culturing the suspended hydrogel-tissue-derived epithelial stem cell mixture in the culture medium to produce tissue-derived epithelial organoids. In certain embodiments, a plurality of tissue-derived epithelial stem cells are contacted with the hydrogel to produce the hydrogel-tissue-derived epithelial stem cell mixture. For example, but not limited to, the method includes (a) contacting a plurality of tissue-derived epithelial stem cells with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture, (b) depositing the hydrogel-tissue-derived epithelial stem cell mixture on a substrate, (c) solidifying the hydrogel-tissue-derived epithelial stem cell mixture to produce a solidified hydrogel-tissue-derived epithelial stem cell mixture, (d) suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue-derived epithelial stem cell mixture, and (e) The suspended hydrogel-tissue-derived epithelial stem cell mixture is cultured in the culture medium to produce tissue-derived epithelial organoids. In certain embodiments, the method further comprises removing the solidified hydrogel-tissue-derived epithelial stem cell mixture from the substrate before suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in the culture medium.
在某些實施例中,將該水凝膠-組織來源之上皮幹細胞混合物作為小滴沉積到該基材上。在某些實施例中,將該水凝膠-組織來源之上皮幹細胞混合物沉積到該基材上以具有絲狀結構。在某些實施例中,該絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,該方法進一步包括將該水凝膠-組織來源之上皮幹細胞混合物在該培養基中碎片化以產生包含該組織來源之上皮類器官之碎片化結構。In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is deposited onto the substrate as droplets. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is deposited onto the substrate to have a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine, or spiral shape. In some embodiments, the method further comprises fragmenting the hydrogel-tissue derived epithelial stem cell mixture in the culture medium to produce a fragmented structure comprising the tissue derived epithelial organoid.
在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物、經懸浮之混合物或經固化之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含大於約 0.1 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物、經懸浮之混合物或經固化之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含約 0.1 mm 至約 1,000 mm ( 例如,約 0.1 mm 至約 20 mm) 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物、經懸浮之混合物或經固化之水凝膠-組織來源之上皮幹細胞混合物呈小滴形式。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物、經懸浮之混合物或經固化之水凝膠-組織來源之上皮幹細胞混合物具有絲狀結構。在某些實施例中,該絲狀結構具有線形、蛇形或螺旋形形狀。 In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture, the suspended mixture, or the solidified hydrogel-tissue-derived epithelial stem cell mixture comprises a length, width, and/or diameter greater than about 0.1 mm. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture, the suspended mixture, or the solidified hydrogel-tissue-derived epithelial stem cell mixture comprises a length, width, and/or diameter of about 0.1 mm to about 1,000 mm ( e.g. , about 0.1 mm to about 20 mm). In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture, the suspended mixture, or the solidified hydrogel-tissue derived epithelial stem cell mixture is in the form of droplets. In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture, the suspended mixture, or the solidified hydrogel-tissue derived epithelial stem cell mixture has a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine, or spiral shape.
在某些實施例中,該組織來源之上皮幹細胞或該等複數個組織來源之上皮幹細胞含在組織碎片、類器官碎片或其組合內。在某些實施例中,該組織來源之上皮幹細胞或該等複數個組織來源之上皮幹細胞分離自原生上皮組織。在某些實施例中,該組織來源之上皮幹細胞係獲自組織之碎片,該組織選自由以下所組成之群組:淚腺、扁桃腺、唾液腺、胃腸組織、甲狀腺、肺、乳腺、肝、膽管、胃、腎、胰臟、子宮內膜、輸卵管、子宮頸、前列腺、膀胱、卵巢、味蕾、胎盤及其組合。可替代地或另外,該組織來源之上皮幹細胞係獲自類器官之碎片,該類器官係選自由以下所組成之群組:淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官、滋胚內層類器官及其組合。In some embodiments, the tissue-derived epithelial stem cell or the plurality of tissue-derived epithelial stem cells are contained in tissue fragments, organoid fragments, or a combination thereof. In some embodiments, the tissue-derived epithelial stem cell or the plurality of tissue-derived epithelial stem cells are isolated from native epithelial tissue. In certain embodiments, the tissue-derived epithelial stem cells are obtained from a fragment of a tissue selected from the group consisting of tear glands, tonsils, salivary glands, gastrointestinal tissue, thyroid, lung, breast, liver, bile duct, stomach, kidney, pancreas, endometrium, fallopian tube, cervix, prostate, bladder, ovary, taste buds, placenta, and combinations thereof. Alternatively or additionally, the tissue-derived epithelial stem cells are obtained from fragments of organoids selected from the group consisting of tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, trophoblastic organoids, and combinations thereof.
在某些實施例中,該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。 In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1×10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1×10 7 tissue-derived epithelial stem cells/ml hydrogel.
在某些實施例中,該水凝膠係選自由以下所組成之群組:合成水凝膠、天然水凝膠及其組合。在某些實施例中,該天然水凝膠包含基底膜萃取物 (BME) 或細胞外基質 (ECM) 成分。在某些實施例中,該水凝膠具有大於約 1 mg/ml 之蛋白質濃度。在某些實施例中,該水凝膠包含以 w/v % 計大於約 1 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。In some embodiments, the hydrogel is selected from the group consisting of: synthetic hydrogels, natural hydrogels, and combinations thereof. In some embodiments, the natural hydrogel comprises basement membrane extract (BME) or extracellular matrix (ECM) components. In some embodiments, the hydrogel has a protein concentration greater than about 1 mg/ml. In some embodiments, the hydrogel comprises greater than about 1 w/v % of a BME component, an ECM component, or a polymer in w/v %. In some embodiments, the hydrogel has a storage modulus G' equal to or greater than a loss modulus G''.
在某些實施例中,該培養基存在於容器中。在某些實施例中,該容器為培養皿、多孔盤、錐形管、貯器、培養袋、生物反應器或燒瓶。In some embodiments, the culture medium is present in a container. In some embodiments, the container is a culture dish, a multi-well dish, a conical tube, a container, a culture bag, a bioreactor or a flask.
本揭露進一步提供了一種藉由本文所揭示之方法產生之組織來源之上皮類器官。The present disclosure further provides a tissue-derived epithelial organoid produced by the methods disclosed herein.
在某些實施例中,與參考組織來源之上皮類器官相比,藉由本揭露之方法產生之組織來源之上皮類器官具有均勻形態。在某些實施例中,該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。在某些實施例中,組織來源之上皮類器官具有均勻尺寸。在某些實施例中,與該等參考組織來源之上皮類器官相比,該組織來源之上皮類器官之平均直徑更均勻。In certain embodiments, the tissue-derived epithelial organoids produced by the methods of the present disclosure have a uniform morphology compared to reference tissue-derived epithelial organoids. In certain embodiments, the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. In certain embodiments, the tissue-derived epithelial organoids have a uniform size. In certain embodiments, the average diameter of the tissue-derived epithelial organoids is more uniform than the reference tissue-derived epithelial organoids.
在某些實施例中,與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在藉由本揭露之方法產生之組織來源之上皮類器官之群體中以較高含量表現。在某些實施例中,與參考組織來源之上皮類器官之群體相比,分化標記物在藉由本揭露之方法產生之組織來源之上皮類器官之群體中以較低含量表現。在某些實施例中,該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。在某些實施例中,該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。在某些實施例中,該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。In certain embodiments, stem cell and/or proliferation markers are expressed at higher levels in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure compared to a population of reference tissue-derived epithelial organoids. In certain embodiments, differentiation markers are expressed at lower levels in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure compared to a population of reference tissue-derived epithelial organoids. In certain embodiments, the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. In certain embodiments, the stem cell and/or proliferation marker is selected from the group consisting of MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. In some embodiments, the differentiation marker is selected from the group consisting of keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA), and combinations thereof.
本揭露進一步提供了一種組成物,其包含組織來源之上皮類器官及培養基,其中該組織來源之上皮類器官嵌入在懸浮於該培養基中之水凝膠內。在某些實施例中,該水凝膠之幾何形狀包含大於約 0.1 mm 之長度、寬度及/或直徑。在某些實施例中,該水凝膠之幾何形狀包含約 0.1 mm 至約 1,000 mm ( 例如,約 0.1 mm 至約 20 mm) 之長度、寬度及/或直徑。在某些實施例中,該水凝膠為小滴。在某些實施例中,該水凝膠具有絲狀結構。在某些實施例中,該絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在組織來源之上皮類器官之群體中以較高含量表現。在某些實施例中,該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。在某些實施例中,與參考組織來源之上皮類器官之群體相比,分化標記物在上皮類器官群體中以較低含量表現。在某些實施例中,該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。在某些實施例中,該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 The present disclosure further provides a composition comprising a tissue-derived epithelial organoid and a culture medium, wherein the tissue-derived epithelial organoid is embedded in a hydrogel suspended in the culture medium. In some embodiments, the geometry of the hydrogel comprises a length, width, and/or diameter greater than about 0.1 mm. In some embodiments, the geometry of the hydrogel comprises a length, width, and/or diameter of about 0.1 mm to about 1,000 mm ( e.g. , about 0.1 mm to about 20 mm). In some embodiments, the hydrogel is a droplet. In some embodiments, the hydrogel has a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine, or spiral shape. In certain embodiments, stem cell and/or proliferation markers are expressed at higher levels in a population of tissue-derived epithelial organoids compared to a population of reference tissue-derived epithelial organoids. In certain embodiments, the stem cell and/or proliferation marker is selected from the group consisting of: MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. In certain embodiments, differentiation markers are expressed at lower levels in a population of epithelial organoids compared to a population of reference tissue-derived epithelial organoids. In some embodiments, the differentiation marker is selected from the group consisting of: keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA), and combinations thereof. In some embodiments, the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate.
本揭露進一步提供了一種用於篩選藥劑 ( 例如,治療劑) 之方法。在某些實施例中,該方法包括 (a) 使組織來源之上皮類器官或組織來源之上皮類器官之群體或組織來源之上皮類器官之組成物與藥劑 ( 例如,治療劑) 接觸,以及 (b) 分析該組織來源之上皮類器官或該等組織來源之上皮類器官之群體中指示該藥劑 ( 例如,治療劑) 之有效性、流佈及/或毒性之變化。在某些實施例中,使該藥劑 ( 例如,治療劑) 與該組織來源之上皮類器官或該等組織來源之上皮類器官之群體接觸約 1 分鐘至約 3 年, 例如,約 15 分鐘至約 3 年。在某些實施例中,該藥劑為治療劑。在某些實施例中,該治療劑為基於多肽之治療劑、小分子治療劑、基於細胞之治療劑、基因編輯系統、基於核酸之治療劑或其組合。在某些實施例中,該變化為性質的變化,該性質選自由以下所組成之群組:細胞生存力、細胞代謝、氧化還原電位、細胞增生、細胞形態、類器官形態、類器官尺寸、蛋白質表現含量、核酸表現含量、核酸修飾、轉譯後修飾、細胞傳訊路徑之活化、細胞傳訊路徑之壓制、酵素活性、障壁完整性及其組合。 The present disclosure further provides a method for screening an agent ( e.g. , a therapeutic agent). In certain embodiments, the method comprises (a) contacting a tissue-derived epithelial organoid, a population of tissue-derived epithelial organoids, or a composition of tissue-derived epithelial organoids with an agent ( e.g. , a therapeutic agent), and (b) analyzing the tissue-derived epithelial organoid or a population of such tissue-derived epithelial organoids for changes indicative of the effectiveness, distribution, and/or toxicity of the agent ( e.g. , a therapeutic agent). In some embodiments, the agent ( e.g. , a therapeutic agent) is contacted with the tissue-derived epithelial organoid or a population of such tissue-derived epithelial organoids for about 1 minute to about 3 years, e.g. , about 15 minutes to about 3 years. In some embodiments, the agent is a therapeutic agent. In some embodiments, the therapeutic agent is a polypeptide-based therapeutic agent, a small molecule therapeutic agent, a cell-based therapeutic agent, a gene editing system, a nucleic acid-based therapeutic agent, or a combination thereof. In certain embodiments, the change is a change in a property selected from the group consisting of cell viability, cell metabolism, redox potential, cell proliferation, cell morphology, organoid morphology, organoid size, protein expression level, nucleic acid expression level, nucleic acid modification, post-translational modification, activation of cell signaling pathways, repression of cell signaling pathways, enzyme activity, barrier integrity, and combinations thereof.
本揭露進一步提供了一種進行基因體篩選之方法。在某些實施例中,該方法包括 (a) 提供組織來源之上皮類器官或組織來源之上皮類器官之群體或組織來源之上皮類器官之組成物,(b) 在該組織來源之上皮類器官之一個或多個細胞之基因體中產生突變,以及 (c) 分析該組織來源之上皮類器官或該等組織來源之上皮類器官之群體中與該突變相關之變化。在某些實施例中,該突變係使用基因調節系統產生的。在某些實施例中,該基因調節系統為基因編輯系統。在某些實施例中,該基因編輯系統為 CRISPR 系統。在某些實施例中,該變化為性質的變化,該性質選自由以下所組成之群組:細胞生存力、細胞代謝、氧化還原電位、細胞增生、細胞形態、類器官形態、類器官尺寸、蛋白質表現含量、核酸表現含量、核酸修飾、轉譯後修飾、細胞傳訊路徑之活化、細胞傳訊路徑之壓制、酵素活性、障壁完整性及其組合。The present disclosure further provides a method for performing genomic screening. In some embodiments, the method includes (a) providing a tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids or a composition of tissue-derived epithelial organoids, (b) generating a mutation in the genome of one or more cells of the tissue-derived epithelial organoid, and (c) analyzing the changes associated with the mutation in the tissue-derived epithelial organoid or the group of tissue-derived epithelial organoids. In some embodiments, the mutation is generated using a gene regulation system. In some embodiments, the gene regulation system is a gene editing system. In some embodiments, the gene editing system is a CRISPR system. In certain embodiments, the change is a change in a property selected from the group consisting of cell viability, cell metabolism, redox potential, cell proliferation, cell morphology, organoid morphology, organoid size, protein expression level, nucleic acid expression level, nucleic acid modification, post-translational modification, activation of cell signaling pathways, repression of cell signaling pathways, enzyme activity, barrier integrity, and combinations thereof.
本揭露進一步提供了一種用於產生上皮細胞模型之方法。在某些實施例中,該方法包括 (a) 提供組織來源之上皮類器官或組織來源之上皮類器官之群體或組織來源之上皮類器官之組成物,(b) 將該組織來源之上皮類器官或該等組織來源之上皮類器官之群體消化成單細胞,以及 (c) 將該單細胞在培養基中培養以產生細胞單層。在某些實施例中,將該單細胞在可滲透細胞培養內件 (cell culture insert) 上培養。在某些實施例中,該培養基為分化培養基。在某些實施例中,該培養基為細胞生長培養基。在某些實施例中,該培養基為幹細胞促進培養基。The present disclosure further provides a method for generating an epithelial cell model. In some embodiments, the method includes (a) providing a tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids or a composition of tissue-derived epithelial organoids, (b) digesting the tissue-derived epithelial organoid or the group of such tissue-derived epithelial organoids into single cells, and (c) culturing the single cells in a culture medium to produce a cell monolayer. In some embodiments, the single cells are cultured on a permeable cell culture insert. In some embodiments, the culture medium is a differentiation medium. In some embodiments, the culture medium is a cell growth medium. In certain embodiments, the culture medium is a stem cell promoting medium.
本揭露進一步提供了一種使用藉由本文所述之方法產生之細胞單層之篩選方法。在某些實施例中,該篩選方法為用於篩選藥劑 ( 例如,治療劑) 之方法。例如但不限於,該方法可以包括 (a) 使藉由本文所述之方法產生之細胞單層與藥劑 ( 例如,治療劑) 接觸;以及 (b) 分析該細胞單層中指示該藥劑 ( 例如,治療劑) 之有效性、流佈及/或毒性之變化。在某些實施例中,使該藥劑 ( 例如,治療劑) 與該細胞單層接觸約 1 分鐘至約 3 年, 例如,約 15 分鐘至約 3 年。在某些實施例中,該藥劑為治療劑。在某些實施例中,該治療劑為基於多肽之治療劑、小分子治療劑、基於細胞之治療劑、基因編輯系統、基於核酸之治療劑或其組合。在某些實施例中,該篩選方法為基因體篩選。在某些實施例中,該進行基因體篩選之方法可包括 (a) 提供藉由如本文所述之方法產生之細胞單層;(b) 在該細胞單層之一個或多個細胞之基因體中產生突變;以及 (c) 分析該細胞單層中與該突變相關之變化。在某些實施例中,該突變係使用基因調節系統產生的。在某些實施例中,該基因調節系統為基因編輯系統。在某些實施例中,該基因編輯系統為 CRISPR 系統。在某些實施例中,該變化為性質的變化,該性質選自由以下所組成之群組:細胞生存力、細胞代謝、氧化還原電位、細胞增生、細胞形態、類器官形態、類器官尺寸、蛋白質表現含量、核酸表現含量、核酸修飾、轉譯後修飾、細胞傳訊路徑之活化、細胞傳訊路徑之壓制、酵素活性、障壁完整性及其組合。 The present disclosure further provides a screening method using a cell monolayer produced by the methods described herein. In some embodiments, the screening method is a method for screening an agent ( e.g. , a therapeutic agent). For example, but not limited to, the method can include (a) contacting a cell monolayer produced by the methods described herein with an agent ( e.g. , a therapeutic agent); and (b) analyzing changes in the cell monolayer that indicate the effectiveness, distribution, and/or toxicity of the agent ( e.g. , a therapeutic agent). In some embodiments, the agent ( e.g. , a therapeutic agent) is contacted with the cell monolayer for about 1 minute to about 3 years, for example , about 15 minutes to about 3 years. In some embodiments, the agent is a therapeutic agent. In some embodiments, the therapeutic agent is a polypeptide-based therapeutic agent, a small molecule therapeutic agent, a cell-based therapeutic agent, a gene editing system, a nucleic acid-based therapeutic agent, or a combination thereof. In some embodiments, the screening method is a genome screening. In some embodiments, the method for performing a genome screening may include (a) providing a cell monolayer produced by a method as described herein; (b) generating a mutation in a genome of one or more cells in the cell monolayer; and (c) analyzing changes in the cell monolayer associated with the mutation. In some embodiments, the mutation is generated using a gene regulation system. In some embodiments, the gene regulation system is a gene editing system. In some embodiments, the gene editing system is a CRISPR system. In some embodiments, the change is a change in a property selected from the group consisting of cell viability, cell metabolism, redox potential, cell proliferation, cell morphology, organoid morphology, organoid size, protein expression level, nucleic acid expression level, nucleic acid modification, post-translational modification, activation of cell signaling pathways, repression of cell signaling pathways, enzyme activity, barrier integrity, and combinations thereof.
在某些實施例中,本揭露之方法之一個或多個步驟可使用一個或多個機器人及/或自動化組件來進行。例如但不限於,如本文所述之產生組織來源之上皮類器官之方法、產生組織來源之上皮類器官之懸浮培養物之方法、篩選藥劑之方法、進行基因體篩選之方法、產生上皮細胞模型之方法及/或使用細胞單層之篩選方法之一個或多個步驟可使用一個或多個機器人及/或自動化組件來進行。在某些實施例中,一個或多個機器人及/或自動化組件選自由以下所組成之群組:液體處理機器人、3D 列印機、注射泵及其組合。在某些實施例中,該一個或多個機器人及/或自動化組件包含液體處理機器人。In some embodiments, one or more steps of the methods of the present disclosure may be performed using one or more robots and/or automated components. For example, but not limited to, one or more steps of the methods of generating tissue-derived epithelial organoids, methods of generating suspension cultures of tissue-derived epithelial organoids, methods of screening agents, methods of performing genomic screening, methods of generating epithelial cell models, and/or screening methods using cell monolayers as described herein may be performed using one or more robots and/or automated components. In some embodiments, the one or more robots and/or automated components are selected from the group consisting of: a liquid handling robot, a 3D printer, a syringe pump, and combinations thereof. In some embodiments, the one or more robots and/or automated components include a liquid handling robot.
本揭露進一步提供了用於培養組織來源之上皮類器官之系統。在某些實施例中,該系統包括組織來源之上皮類器官及培養基,其中該組織來源之上皮類器官嵌入在懸浮於該培養基中之水凝膠內。在某些實施例中,該水凝膠之幾何形狀包含大於約 0.1 mm 之長度、寬度及/或直徑。在某些實施例中,該水凝膠之幾何形狀包含約 0.1 mm 至約 1,000 mm ( 例如,約 0.1 mm 至約 20 mm) 之長度、寬度及/或直徑。在某些實施例中,該水凝膠為小滴。在某些實施例中,該水凝膠具有絲狀結構。在某些實施例中,該絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在該等組織來源之上皮類器官之群體中以較高含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。在某些實施例中,該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。在某些實施例中,與參考組織來源之上皮類器官之群體相比,分化標記物在該等組織來源之上皮類器官之群體中以較低含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。在某些實施例中,該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。在某些實施例中,該組織來源之上皮類器官係選自由以下所組成之群組:淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官、滋胚內層類器官及其組合。在某些實施例中,該系統包括用於產生及/或培養該組織來源之上皮類器官之一個或多個機器人及/或自動化組件。在某些實施例中,一個或多個機器人及/或自動化組件選自由以下所組成之群組:液體處理機器人、3D 列印機、注射泵及其組合。在某些實施例中,該一個或多個機器人及/或自動化組件包含液體處理機器人。 The present disclosure further provides a system for culturing tissue-derived epithelial organoids. In some embodiments, the system includes tissue-derived epithelial organoids and a culture medium, wherein the tissue-derived epithelial organoids are embedded in a hydrogel suspended in the culture medium. In some embodiments, the geometry of the hydrogel comprises a length, width, and/or diameter greater than about 0.1 mm. In some embodiments, the geometry of the hydrogel comprises a length, width, and/or diameter of about 0.1 mm to about 1,000 mm ( e.g. , about 0.1 mm to about 20 mm). In some embodiments, the hydrogel is a droplet. In some embodiments, the hydrogel has a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine, or helical shape. In some embodiments, stem cell and/or proliferation markers are expressed at higher levels in a population of tissue-derived epithelial organoids compared to a population of reference tissue-derived epithelial organoids, wherein the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. In some embodiments, the stem cell and/or proliferation marker is selected from the group consisting of MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. In certain embodiments, a differentiation marker is expressed at a lower level in a population of tissue-derived epithelial organoids compared to a population of reference tissue-derived epithelial organoids, wherein the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. In some embodiments, the differentiation marker is selected from the group consisting of: keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA) and combinations thereof. In some embodiments, the tissue-derived epithelial organoid is selected from the group consisting of tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, trophoblastic lining organoids, and combinations thereof. In some embodiments, the system includes one or more robots and/or automated components for generating and/or culturing the tissue-derived epithelial organoids. In some embodiments, the one or more robots and/or automated components are selected from the group consisting of: a liquid handling robot, a 3D printer, a syringe pump, and combinations thereof. In some embodiments, the one or more robots and/or automated components include a liquid handling robot.
本揭露進一步提供了用於進行本文揭示之方法之系統。例如但不限於,本揭露提供了用於進行以下之系統:產生組織來源之上皮類器官之方法、產生組織來源之上皮類器官之懸浮培養物之方法、篩選藥劑之方法、進行基因體篩選之方法、產生上皮細胞模型之方法及/或使用細胞單層之篩選方法。在某些實施例中,該系統包括用於產生及/或培養該組織來源之上皮類器官之一個或多個機器人及/或自動化組件。在某些實施例中,一個或多個機器人及/或自動化組件選自由以下所組成之群組:液體處理機器人、3D 列印機、注射泵及其組合。在某些實施例中,該一個或多個機器人及/或自動化組件包含液體處理機器人。The present disclosure further provides systems for performing the methods disclosed herein. For example, but not limited to, the present disclosure provides systems for performing the following: methods for generating tissue-derived epithelial organoids, methods for generating suspension cultures of tissue-derived epithelial organoids, methods for screening agents, methods for performing genomic screening, methods for generating epithelial cell models, and/or screening methods using cell monolayers. In some embodiments, the system includes one or more robots and/or automated components for generating and/or culturing the tissue-derived epithelial organoids. In some embodiments, the one or more robots and/or automated components are selected from the group consisting of: liquid handling robots, 3D printers, syringe pumps, and combinations thereof. In some embodiments, the one or more robots and/or automated components include a liquid handling robot.
相關申請之交叉引用Cross-references to related applications
本申請案主張 2023 年 6 月 14 日提交之美國臨時申請案第 63/508,132 號之優先權,該申請案之內容以引用方式全文併入本文中。This application claims priority to U.S. Provisional Application No. 63/508,132, filed on June 14, 2023, the contents of which are incorporated herein by reference in their entirety.
本揭露提供了於懸浮培養物中之組織來源之上皮類器官以及產生此等組織來源之上皮類器官之方法。本文所揭示之方法使得能夠產生大規模類器官培養物,而不需要繁瑣手動操作、專用設備或自動化。藉由在經懸浮之水凝膠而非習用表面接附之水凝膠圓頂中生長類器官細胞,可顯著增加水凝膠體積,且因此可顯著增加可在培養容器中生長之類器官細胞之數量。此外,由於目前所揭示之方法不需要將水凝膠沉積在二維 (2D) 表面上,因此該方法與各種培養容器 ( 例如培養瓶及培養袋) 相容,這允許進一步培養規模化,從而實現高生產量。 The present disclosure provides tissue-derived epithelial organoids in suspended cultures and methods of generating such tissue-derived epithelial organoids. The methods disclosed herein enable the generation of large-scale organoid cultures without the need for cumbersome manual manipulations, specialized equipment, or automation. By growing organoid cells in suspended hydrogels rather than using surface-attached hydrogel domes, the hydrogel volume can be significantly increased, and therefore the number of organoid cells that can be grown in a culture vessel can be significantly increased. Furthermore, since the presently disclosed method does not require the hydrogel to be deposited on a two-dimensional (2D) surface, the method is compatible with various culture containers ( eg, culture flasks and culture bags), which allows for further culture scalability, thereby achieving high throughput.
目前所揭示之方法允許使用各種幾何形狀之經懸浮水凝膠,這可以加快實際培養物製備時間。此外,由於類器官處於懸浮狀態,因此可以在培養過程中在不同時間對它們進行取樣、分割或收集,這對於固定在盤中之習用類器官培養物來說係困難的。如實例 1 所示,類器官在經懸浮之 BME 水凝膠中的生長比在習用表面接附之水凝膠圓頂中更均勻,在習用表面接附之水凝膠圓頂中,有限分子擴散導致營養素梯度 (Park 等人(2022);Shin 等人(2020))。本文所揭示之經懸浮之水凝膠培養方法產生更適合高生產量研究之組織來源之上皮類器官模型,這將有利於基礎科學及轉化領域二者。The methods disclosed herein allow the use of suspended hydrogels of various geometries, which can speed up actual culture preparation time. In addition, because organoids are in suspension, they can be sampled, split, or collected at different times during the culture process, which is difficult for conventional organoid cultures that are fixed in a dish. As shown in Example 1, organoids grew more uniformly in suspended BME hydrogels than in conventional surface-attached hydrogel domes, where limited molecular diffusion leads to nutrient gradients (Park et al. (2022); Shin et al. (2020)). The suspended hydrogel culture method disclosed herein generates epithelial organoid models of tissue origin that are more suitable for high-throughput studies, which will benefit both basic science and translational fields.
為求清楚,但不作為限制,將本文所揭示之主題之詳細描述分為以下小節: I. 定義; II. 類器官及其組成物; III. 生產類器官之方法; IV. 使用方法; V. 系統;以及 VI. 示例性實施例 I. 定義 For clarity, but not limitation, the detailed description of the subject matter disclosed herein is divided into the following subsections: I. Definitions; II. Organoids and Compositions Thereof; III. Methods of Producing Organoids; IV. Methods of Use; V. Systems; and VI. Exemplary Embodiments I. Definitions
除非另有定義,否則本文所用之全部技術及科學術語具有與一般熟習本揭露之主題所屬技術者通常所瞭解之含義相同之含義。下列參考文獻提供技術人員本揭露中所使用的許多術語之一般定義:Singleton 等人,Dictionary of Microbiology and Molecular Biology (第 2 版,1994);The Cambridge Dictionary of Science and Technology (Walker 編,1988);The Glossary of Genetics,第 5 版,R. Rieger 等人(編),Springer Verlag (1991);以及 Hale & Marham, The Harper Collins Dictionary of Biology (1991)。如本文所用,除非另有說明,否則以下術語具有以下賦予其等之含義。Unless otherwise defined, all technical and scientific terms used herein have the same meanings as those generally understood by those skilled in the art to which the subject matter of this disclosure belongs. The following references provide general definitions of many of the terms used in this disclosure by those skilled in the art: Singleton et al., Dictionary of Microbiology and Molecular Biology (2nd edition, 1994); The Cambridge Dictionary of Science and Technology (Walker ed., 1988); The Glossary of Genetics, 5th edition, R. Rieger et al. (eds.), Springer Verlag (1991); and Hale & Marham, The Harper Collins Dictionary of Biology (1991). As used herein, unless otherwise specified, the following terms have the meanings assigned to them below.
如本文所用,當「一」或「一種」一詞之使用與請求項及/或說明書中之術語「包含」結合使用時,其可意指「一個」,但亦與「一個或多個」、「至少一個」及「一個或大於一個」之含義一致。As used herein, when the terms "a" or "an" are used in conjunction with the term "comprising" in the claims and/or the specification, they may mean "one", but are also consistent with the meanings of "one or more", "at least one", and "one or more than one".
術語「約」或「大約」意指特定值處於本技術領域中具有通常知識者所判定之可接受之誤差範圍內,其部分地取決於如何測量或判定該值, 即,取決於測量系統之局限性。例如,按照本技術領域中之實務,「約」可意指 3 倍內或 3 倍以上之標準偏差。可替代地,「約」可意指給定值之至多 20%、較佳至多 10%、更佳至多 5% 並且更佳至多 1% 之範圍。可替代地,特別為關於生物系統或過程,該術語意指數值之一個數量級內, 例如在數值之 5 倍以內,或在數值之 2 倍以內。 The term "about" or "approximately" means that a particular value is within an acceptable range of error as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined, i.e. , on the limitations of the measurement system. For example, according to practice in the art, "about" can mean within 3 times or more of a standard deviation. Alternatively, "about" can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term means within an order of magnitude of a value, such as within 5 times of a value, or within 2 times of a value.
本文中之術語「抗體」以最廣義使用且涵蓋各種抗體結構,包括但不限於單株抗體、多株抗體、多特異性抗體 ( 例如,雙特異性抗體) 以及抗體片段,只要其等展示出預期抗原結合活性即可。 The term "antibody" herein is used in the broadest sense and covers various antibody structures, including but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies ( eg , bispecific antibodies) and antibody fragments, as long as they exhibit the intended antigen-binding activity.
「抗體片段」係指除完整抗體以外之分子,其包含結合完整抗體所結合抗原之完整抗體之一部分。抗體片段之實例包括但不限於 Fv、Fab、Fab'、Fab'-SH、F(ab') 2、雙功能抗體、線性抗體、單鏈抗體分子 ( 例如,scFv) 及由抗體片段形成之多特異性抗體。 "Antibody fragment" refers to a molecule other than an intact antibody, which comprises a portion of an intact antibody that binds to the antigen to which the intact antibody binds. Examples of antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab') 2 , bifunctional antibodies, linear antibodies, single-chain antibody molecules ( e.g. , scFv), and multispecific antibodies formed from antibody fragments.
如本文所用,術語「培養基 (culture medium)」或「培養基 (medium)」係指液體,其覆蓋培養容器 (諸如培養瓶及多孔盤) 中之細胞並含有滋養細胞之營養素。在某些實施例中,培養基亦可包括生長因子或分化因子以在細胞中產生所需變化。As used herein, the term "culture medium" or "medium" refers to a liquid that covers cells in a culture container (such as a culture bottle and a multiwell dish) and contains nutrients that nourish the cells. In certain embodiments, the culture medium may also include growth factors or differentiation factors to produce desired changes in the cells.
如本文所用,術語「包含」、「包括」、「具有 (having、has)」、「可」、「含有」及其變異體意欲為不排除其他行為或結構之開放式連接詞 (open-ended transitional phrase)、術語、或字。本揭露亦涵蓋「包含」本文所呈現之實施例或元件、「由其組成」及「基本上由其組成」之本文所呈現之實施例或元件,無論是否明確陳述。As used herein, the terms "comprise", "include", "having", "has", "may", "contain" and variations thereof are intended to be open-ended transitional phrases, terms, or words that do not exclude other actions or structures. The present disclosure also encompasses embodiments or elements presented herein that "comprise", "consist of", and "consist essentially of" the embodiments or elements presented herein, whether or not explicitly stated.
如本文所用,術語使細胞與化合物 例如治療劑「接觸」係指將細胞曝露於化合物,例如將化合物置於允許其接觸細胞之位置。接觸可使用任何合適方法來完成。例如但不限於,「接觸」可藉由將化合物添加到含有細胞之容器中來完成。接觸也可藉由將化合物添加到包含細胞之培養基中來完成。在某些實施例中,「接觸」係指將細胞 ( 例如組織來源之上皮類器官內或細胞單層中之細胞) 曝露於藥劑或化合物中。在某些實施例中,「接觸」係指將組織來源之上皮類器官或細胞單層曝露於潛在治療劑或所關注的治療劑中。 As used herein, the term "contacting" a cell with a compound , such as a therapeutic agent, refers to exposing the cell to the compound, such as placing the compound in a position that allows it to contact the cell. Contacting can be accomplished using any suitable method. For example, but not limited to, "contacting" can be accomplished by adding the compound to a container containing the cells. Contacting can also be accomplished by adding the compound to a culture medium containing the cells. In certain embodiments, "contacting" refers to exposing a cell ( e.g., a cell in an epithelial organoid or a cell monolayer of tissue origin) to an agent or compound. In certain embodiments, "contacting" refers to exposing the tissue-derived epithelial organoid or cell monolayer to a potential therapeutic agent or therapeutic agent of interest.
如本文所用,術語「來源自」或「建立自」或「分化自」當提及本文所揭示之任何細胞時,係指使用任何操作獲自細胞株、組織 (諸如解離組織) 或流體中之 ( 例如,經分離或純化的) 親代細胞之細胞。此等操作之非限制性實例包括單細胞分離、 活體外培養、使用 例如蛋白質、化學物質、輻射、病毒感染及核酸轉染之處理及/或誘變。在某些實施例中,可以藉由對生長因子、細胞介素、細胞介素治療之選定進程、黏附性、缺乏黏附性、分選程序或其組合之反應從混合群體中選擇來源細胞。 As used herein, the term "derived from" or "established from" or "differentiated from" when referring to any cell disclosed herein refers to a cell obtained from a cell line, tissue (such as a dissociated tissue), or a parent cell in a fluid ( e.g. , separated or purified) using any manipulation. Non-limiting examples of such manipulations include single cell isolation, in vitro culture, treatment and/or induction using, for example, proteins, chemicals, radiation, viral infection, and nucleic acid transfection. In certain embodiments, source cells can be selected from a mixed population by response to growth factors, cytokines, selected processes of cytokine treatment, adhesion, lack of adhesion, sorting procedures, or a combination thereof.
術語「檢測 (detection)」或「檢測 (detecting)」包括任何檢測手段,包括直接及間接檢測。The terms "detection" or "detecting" include any means of detection, including direct and indirect detection.
如本文所用,術語「小滴」當提及幾何形狀使用時係指球形或類球形形狀。As used herein, the term "droplet" when used in reference to a geometric shape refers to a spherical or spheroidal shape.
如本文所用,術語「嵌入」或「嵌入的」係指至少部分覆蓋或包圍細胞。在某些實施例中,如本文使用,術語「嵌入」或「嵌入的」係指 例如以水凝膠完全覆蓋或包圍組織來源之上皮類器官。 As used herein, the term "embedded" or "embedded" refers to at least partially covering or surrounding cells. In certain embodiments, as used herein, the term "embedded" or "embedded" refers to completely covering or surrounding tissue-derived epithelial organoids, such as with a hydrogel.
如本文所用,術語「表現 (expression)」或「表現 (express)」係指核苷酸序列之轉錄及/或轉譯。As used herein, the term "expression" or "express" refers to the transcription and/or translation of a nucleotide sequence.
術語「表現載體」用於表示線性或環狀之核酸分子,其中可以整合適當尺寸之另一核酸序列片段。此等核酸片段可包括提供由核酸序列片段編碼之基因轉錄之額外區段。額外區段可包括但不限於:啟動子、轉錄終止子、增強子、內部核醣體進入位點、非轉譯區、聚腺苷酸化訊號、可選擇標記物、複製起點等,如本技術領域已知的。表現載體通常來源自質粒、黏粒及病毒載體;載體通常為含有來自多種來源之核酸序列之重組分子。The term "expression vector" is used to denote a linear or circular nucleic acid molecule into which another nucleic acid sequence fragment of appropriate size can be incorporated. Such nucleic acid fragments may include additional segments that provide for transcription of the gene encoded by the nucleic acid sequence fragment. Additional segments may include, but are not limited to: promoters, transcription terminators, enhancers, internal ribosomal entry sites, non-translational regions, polyadenylation signals, selectable markers, origins of replication, etc., as known in the art. Expression vectors are usually derived from plasmids, cosmids and viral vectors; vectors are usually recombinant molecules containing nucleic acid sequences from a variety of sources.
如本文所用,術語「胃腸」係指口腔黏膜、咽 (喉)、食道、胃、小腸、大腸及直腸。As used herein, the term "gastrointestinal" refers to the oral mucosa, pharynx (throat), esophagus, stomach, small intestine, large intestine, and rectum.
如本文所用,術語「胃腸幹細胞」係指胃腸系統之幹細胞。As used herein, the term "gastrointestinal stem cells" refers to stem cells of the gastrointestinal system.
如本文所用,術語「受試者」或「個體」係指脊椎動物或無脊椎動物,諸如人類或非人類動物,例如哺乳動物。哺乳動物包括但不限於人類、非人類靈長類動物、農場動物、競賽動物、囓齒動物及寵物。非人類動物個體之非限制性實例包括囓齒動物,諸如小鼠、大鼠、倉鼠、天竺鼠、兔、狗、貓、綿羊、豬、山羊、牛、馬、猿及猴。在某些實施例中,受試者或個體為人類。As used herein, the term "subject" or "individual" refers to a vertebrate or invertebrate, such as a human or a non-human animal, such as a mammal. Mammals include, but are not limited to, humans, non-human primates, farm animals, game animals, rodents, and pets. Non-limiting examples of non-human animal individuals include rodents, such as mice, rats, hamsters, guinea pigs, rabbits, dogs, cats, sheep, pigs, goats, cows, horses, apes, and monkeys. In certain embodiments, the subject or individual is a human.
如本文所用,術語「腸的」或「腸」係指直腸、小腸及大腸。As used herein, the term "enteral" or "intestine" refers to the rectum, small intestine, and large intestine.
如本文所用,術語「腸幹細胞」係指腸之幹細胞。As used herein, the term "intestinal stem cells" refers to the stem cells of the intestine.
如本文所用,術語「 活體外」係指人工環境及在人工環境內發生之過程或反應。 活體外環境例如但不限於細胞培養物。 As used herein, the term " in vitro " refers to an artificial environment and processes or reactions that occur within an artificial environment. An in vitro environment includes, but is not limited to, a cell culture.
如本文所用,術語「 活體內」係指自然環境 ( 例如,動物或細胞) 及在自然環境中發生之過程或反應。 As used herein, the term " in vivo " refers to the natural environment ( eg , an animal or a cell) and processes or reactions that occur in the natural environment.
如本文所用,術語「線性」當提及幾何形狀使用時係指類似於線之形狀。在某些實施例中,該線可以為直線、曲線或任何形狀之線。As used herein, the term "linear" when used in reference to a geometric shape refers to a shape similar to a line. In some embodiments, the line can be a straight line, a curved line, or a line of any shape.
如本文所用,提及細胞 例如胃腸幹細胞之術語「經分離」係指已與其自然環境組分分離之細胞。 As used herein, the term "isolated" in reference to cells , such as gastrointestinal stem cells, refers to cells that have been separated from components of their natural environment.
如本文所用,「標記物」係指允許直接或間接檢測之藥劑。標記物包括但不限於螢光組成物、顯色標記、電子緻密標記、化學發光標記及放射性標記。標記物之非限制性實例包括綠色螢光蛋白 (「GFP」)、mCherry、dtTomato 或本技術領域中已知之其他螢光蛋白 (例如,Shaner 等人, A Guide to Choosing Fluorescent Proteins, Nature Methods 2(12):905-909 (2005),藉由引用併入本文), 32P、 14C、 125I、 3H 及 131I、螢光團 (諸如稀土螯合物或螢光黃及其衍生物)、玫瑰紅 (rhodamine) 及其衍生物,丹磺醯 (dansyl),繖形酮 (umbelliferone)、螢光素酶 (諸如螢火蟲螢光素酶及細菌螢光素純酶) (美國專利第 4,737,456 號)、螢光素、2,3-二氫呔𠯤二酮,以及產生可檢測訊號之酶, 例如山葵過氧化酶 (horseradish peroxidase, HRP)、鹼性磷酸酶、β 半乳糖苷酶、葡萄糖澱粉酶、溶菌酶、碳水化合物氧化酶 (諸如葡萄糖氧化酶、半乳糖氧化酶及葡萄糖-6-磷酸去氫酶 (G6PD)) 及雜環氧化酶 (諸如尿酸酶及黃嘌呤氧化酶)。 As used herein, "label" refers to an agent that allows direct or indirect detection. Labels include, but are not limited to, fluorescent compositions, chromogenic labels, electronically precise labels, chemiluminescent labels, and radioactive labels. Non-limiting examples of markers include green fluorescent protein ("GFP"), mCherry, dtTomato or other fluorescent proteins known in the art (e.g., Shaner et al., A Guide to Choosing Fluorescent Proteins, Nature Methods 2(12):905-909 (2005), incorporated herein by reference), 32 P, 14 C, 125 I, 3 H and 131 I, fluorophores (such as rare earth chelates or fluorescent yellow and its derivatives), rhodamine and its derivatives, dansyl, umbelliferone, luciferase (such as firefly luciferase and bacterial luciferin pure enzyme) (U.S. Patent No. 4,737,456 ), fluorescein, 2,3-dihydropyrrophenone, and enzymes that generate detectable signals, such as horseradish peroxidase (HRP), alkaline phosphatases, β-galactosidase, glucoamylase, lysozyme, carbohydrate oxidases (such as glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase (G6PD)), and heterocyclic oxidases (such as uricase and xanthine oxidase).
術語「核酸」或「多核苷酸」包括任何包含核苷酸聚合物之化合物及/或物質。各核苷酸由鹼基具體而言嘌呤或嘧啶鹼基 (即,胞嘧啶 (C)、鳥嘌呤 (G)、腺嘌呤 (A)、胸腺嘧啶 (T) 或尿嘧啶 (U))、糖 (即,去氧核糖或核糖) 及磷酸基團構成。通常,核酸分子藉由鹼基序列進行描述,其中該等鹼基代表核酸分子之一級結構 (線性結構)。鹼基序列通常由 5' 至 3' 表示。術語核酸包括:去氧核糖核酸 (DNA),包括例如,互補 DNA (cDNA) 及基因體 DNA;核糖核酸 (RNA),例如訊息 RNA (mRNA);DNA 或 RNA 之合成形式;以及包含這些分子中之兩者或更多者之混合聚合物。核酸分子可為線性或環狀的。此外,術語核酸包括有義股及反義股兩者,以及單股及雙股形式。此外,本文所述之核酸可含有天然存在或非天然存在之核苷酸。非天然存在之核苷酸之實例包括帶有衍生糖、磷酸鹽主鏈鍵結或化學修飾殘基之經修飾之核苷酸鹼基。 The term "nucleic acid" or "polynucleotide" includes any compound and/or substance comprising a polymer of nucleotides. Each nucleotide is composed of a base, specifically a purine or pyrimidine base (i.e., cytosine (C), guanine (G), adenine (A), thymine (T) or uracil (U)), a sugar (i.e., deoxyribose or ribose) and a phosphate group. Typically, nucleic acid molecules are described by a base sequence, wherein the bases represent the primary structure (linear structure) of the nucleic acid molecule. The base sequence is usually represented from 5' to 3'. The term nucleic acid includes: deoxyribonucleic acid (DNA), including, for example, complementary DNA (cDNA) and genomic DNA; ribonucleic acid (RNA), such as messenger RNA (mRNA); synthetic forms of DNA or RNA; and mixed polymers comprising two or more of these molecules. Nucleic acid molecules can be linear or circular. Furthermore, the term nucleic acid includes both sense and antisense strands, as well as single-stranded and double-stranded forms. Furthermore, the nucleic acids described herein can contain naturally occurring or non-naturally occurring nucleotides. Examples of non-naturally occurring nucleotides include modified nucleotide bases with derivatized sugars, phosphate backbone linkages, or chemically modified residues.
術語“可操作地連接”,當應用於核酸序列時,例如表現載體中之核酸序列時,表示序列被排列使得它們協同發揮作用以實現其預期目的, 即,啟動子序列允許起始轉錄,該轉錄藉由鏈接編碼序列開展直至終止訊號。 The term "operably linked", when applied to nucleic acid sequences, e.g., those in an expression vector, means that the sequences are arranged so that they function together to achieve their intended purpose, i.e. , a promoter sequence allows initiation of transcription, which proceeds through the linked coding sequence until a termination signal.
如本文所用,術語「類器官」係指藉由幹細胞 ( 例如成體幹細胞) 之擴增而獲得之三維細胞結構,該幹細胞自組織並可分化成功能細胞類型。 參見Corro 等人(2020) Am. J. Physiol.Cell Physiol.319:C151-C165。 As used herein, the term "organoid" refers to a three-dimensional cell structure obtained by the expansion of stem cells ( e.g., adult stem cells) that self-organize and can differentiate into functional cell types. See Corro et al. (2020) Am. J. Physiol. Cell Physiol. 319:C151-C165.
如本文所用,術語「複數」係指數量大於一個。在某些實施例中,術語「複數個組織來源之上皮幹細胞」係指數量大於一個之組織來源之上皮幹細胞。例如但不限於,複數個組織來源之上皮幹細胞包括至少兩個組織來源之上皮幹細胞。在某些非限制性實施例中,複數個組織來源之上皮幹細胞可包括至少約 10 個、至少約 100 個、至少約 200 個、至少約 300 個、至少約 400 個、至少約 500 個、至少約 600 個、至少約 700 個、至少約 800 個、至少約 900 個、至少約 1000 個、至少約 5,000 個、至少約 10,000 個、至少約 100,000 個、至少約 1,000,000 個、至少約 10,000,000 個、至少約 100,000,000 個或至少約 1,000,000,000 個組織來源之上皮幹細胞。As used herein, the term "plurality" refers to a quantity greater than one. In certain embodiments, the term "plurality of tissue-derived epithelial stem cells" refers to a quantity greater than one tissue-derived epithelial stem cell. For example, but not limited to, the plurality of tissue-derived epithelial stem cells include at least two tissue-derived epithelial stem cells. In certain non-limiting embodiments, the plurality of tissue-derived epithelial stem cells may include at least about 10, at least about 100, at least about 200, at least about 300, at least about 400, at least about 500, at least about 600, at least about 700, at least about 800, at least about 900, at least about 1000, at least about 5,000, at least about 10,000, at least about 100,000, at least about 100,000, at least about 1,000,000, at least about 10,000,000, at least about 100,000,000, or at least about 1,000,000,000 tissue-derived epithelial stem cells.
如本文所用,術語「組織來源之上皮類器官之群體」係指至少兩種組織來源之上皮類器官之群組。在某些實施例中,「組織來源之上皮類器官之群體」係指藉由相同方法產生之組織來源之上皮類器官之群組。在某些非限制性實施例中,組織來源之上皮類器官之群體可包括至少約 10 個、至少約 100 個、至少約 200 個、至少約 300 個、至少約 400 個、至少約 500 個、至少約 600 個、至少約 700 個、至少約 800 個、至少約 900 個、至少約 1000 個、至少約 5,000 個、至少約 10,000 個、至少約 100,000 個、至少約 1,000,000 個、至少約 10,000,000 個、至少約 100,000,000 個或至少約 1,000,000,000 個組織來源之上皮類器官。As used herein, the term "a population of tissue-derived epithelial organoids" refers to a population of epithelial organoids from at least two tissues. In certain embodiments, a "population of tissue-derived epithelial organoids" refers to a population of tissue-derived epithelial organoids generated by the same method. In certain non-limiting embodiments, a population of tissue-derived epithelial organoids can include at least about 10, at least about 100, at least about 200, at least about 300, at least about 400, at least about 500, at least about 600, at least about 700, at least about 800, at least about 900, at least about 1000, at least about 5,000, at least about 10,000, at least about 100,000, at least about 1,000,000, at least about 10,000,000, at least about 100,000,000, or at least about 1,000,000,000 tissue-derived epithelial organoids.
如本文所用,術語「增生」係指細胞數量之增加。As used herein, the term "proliferation" refers to an increase in the number of cells.
如本文所使用,術語「啟動子」表示基因內之區域,轉錄因子及/或 RNA 聚合酶可與其結合以控制相關編碼序列之表現。啟動子通常 (但並非經常) 位於轉譯起始密碼子上游之基因 5' 非編碼區。基因之啟動子區可包括充當核酸結合蛋白之序列特異性核酸結合結構域之可辨識結合位點之一個或多個共同序列。然而,此等結合位點也可位於啟動子外部之區域中,例如位於內含子中或編碼序列下游之增強子區域中。As used herein, the term "promoter" refers to a region within a gene to which transcription factors and/or RNA polymerases can bind to control the expression of the associated coding sequence. The promoter is usually (but not always) located in the 5' non-coding region of the gene upstream of the translation start codon. The promoter region of a gene may include one or more common sequences that serve as recognizable binding sites for sequence-specific nucleic acid binding domains of nucleic acid binding proteins. However, these binding sites may also be located in regions outside the promoter, such as in introns or in enhancer regions downstream of the coding sequence.
如本文所用,術語「固化」或「凝固的」係指物質之硬化、增稠、聚合及/或剛性增加。As used herein, the term "cure" or "solidified" refers to the hardening, thickening, polymerization and/or increase in rigidity of a substance.
如本文所用,術語“蛇形”當提及幾何形狀使用時係指蛇形形狀。As used herein, the term "snake" when used in reference to a geometric shape refers to a serpentine shape.
如本文所用,術語“螺旋”當提及幾何形狀使用時係指圍繞中心點或圍繞軸線螺旋之連續曲線。As used herein, the term "helix" when used in reference to a geometric shape refers to a continuous curve that spirals around a center point or around an axis.
如本文所用,術語「子集」係指大量材料中之一小部分。As used herein, the term "subset" refers to a small portion of a larger body of material.
如本文所用,「組織來源之上皮幹細胞」係指獲自組織之上皮幹細胞。在某些實施例中,組織來源之上皮幹細胞不包括富潛能幹細胞 ( 例如,誘導富潛能幹細胞 (iPSC) 及胚胎幹細胞 (ESC))。 As used herein, "tissue-derived epithelial stem cells" refers to epithelial stem cells obtained from tissues. In certain embodiments, tissue-derived epithelial stem cells do not include high-potential stem cells ( eg , induced high-potential stem cells (iPSCs) and embryonic stem cells (ESCs)).
如本文所用,「治療」為用於獲得有益或期望結果包括臨床結果之方法。為本主題之目的,有益或期望臨床結果包括但不限於減輕或改善一種或多種體徵或症狀、減小疾病程度、穩定 ( 即,不惡化) 疾病狀態、預防疾病、延遲或減緩疾病進展、緩解疾病 ( 例如癌症) 及/或改善或減輕疾病狀態。該降低可為併發症、體徵或症狀之嚴重程度或進展至另一等級之可能性的至少 10%、20%、30%、40%、50%、60%、70%、80%、90%、95%、98% 或 99% 降低。在某些實施例中,「治療」亦可指抑制癌症之增生或進展至更高等級至少 10%、20%、30%、40%、50%、60%、70%、80%、90%、95%、98% 或 99%。 II. 類器官及其組成物 As used herein, "treatment" is a method for obtaining beneficial or desired results, including clinical results. For the purposes of the present subject matter, beneficial or desired clinical results include, but are not limited to, reduction or amelioration of one or more signs or symptoms, reduction in severity of disease, stabilization ( i.e. , not worsening) of the disease state, prevention of disease, delay or slow progression of disease, alleviation of disease ( e.g., cancer), and/or improvement or reduction of the disease state. The reduction may be at least a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99% reduction in the severity of complications, signs, or symptoms, or the likelihood of progression to another level. In certain embodiments, "treating" may also refer to inhibiting the proliferation or progression of cancer to a higher level by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 98%, or 99%. II. Organoids and their compositions
本揭露提供了組織來源之上皮類器官。在某些實施例中,組織來源之上皮類器官嵌入在不接附於基材 ( 例如培養容器之基材) 之水凝膠中。本揭露進一步提供了包括此等類器官之組成物。在某些實施例中,組織來源之上皮類器官藉由本文所揭示之方法產生, 例如第 III 節中所揭示之方法。 The present disclosure provides tissue-derived epithelial organoids. In certain embodiments, the tissue-derived epithelial organoids are embedded in a hydrogel that is not attached to a substrate ( e.g., a substrate of a culture vessel). The present disclosure further provides compositions comprising such organoids. In certain embodiments, the tissue-derived epithelial organoids are produced by a method disclosed herein, such as the method disclosed in Section III.
在某些實施例中,本揭露提供了包括組織來源之上皮類器官及培養基之組成物,其中組織來源之上皮類器官嵌入在水凝膠內。在某些實施例中,水凝膠不接附於基材表面, 例如細胞培養皿及/或多孔盤之表面。在某些實施例中,嵌入在水凝膠內之組織來源之上皮類器官懸浮在培養基中。 In certain embodiments, the present disclosure provides a composition comprising a tissue-derived epithelial organoid and a culture medium, wherein the tissue-derived epithelial organoid is embedded in a hydrogel. In certain embodiments, the hydrogel is not attached to a substrate surface, such as a cell culture dish and/or a multiwell dish. In certain embodiments, the tissue-derived epithelial organoid embedded in the hydrogel is suspended in the culture medium.
在某些實施例中,組織來源之上皮類器官為淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官或滋胚內層類器官。在某些實施例中,組織來源之上皮類器官為淚腺類器官。在某些實施例中,組織來源之上皮類器官為扁桃腺類器官。在某些實施例中,組織來源之上皮類器官為唾液腺類器官。在某些實施例中,組織來源之上皮類器官為胃腸類器官。在某些實施例中,組織來源之上皮類器官為甲狀腺類器官。在某些實施例中,組織來源之上皮類器官為肺類器官。在某些實施例中,組織來源之上皮類器官為乳腺類器官。在某些實施例中,組織來源之上皮類器官為肝類器官。在某些實施例中,組織來源之上皮類器官為膽管類器官。在某些實施例中,組織來源之上皮類器官為胃類器官。在某些實施例中,組織來源之上皮類器官為腎類器官。在某些實施例中,組織來源之上皮類器官為胰臟類器官。在某些實施例中,組織來源之上皮類器官為子宮內膜類器官。在某些實施例中,組織來源之上皮類器官為輸卵管類器官。在某些實施例中,組織來源之上皮類器官為子宮頸類器官。在某些實施例中,組織來源之上皮類器官為前列腺類器官。在某些實施例中,組織來源之上皮類器官為膀胱類器官。在某些實施例中,組織來源之上皮類器官為卵巢類器官。在某些實施例中,組織來源之上皮類器官為味蕾類器官。在某些實施例中,組織來源之上皮類器官為滋胚內層類器官。在某些實施例中,組織來源之上皮類器官選自由以下所組成之群組:肺類器官、胃腸類器官、肝類器官、胰臟類器官、乳腺類器官及其組合。In some embodiments, the tissue-derived epithelial organoid is a tear gland organoid, a tonsil organoid, a salivary gland organoid, a gastrointestinal organoid, a thyroid organoid, a lung organoid, a breast organoid, a liver organoid, a bile duct organoid, a stomach organoid, a kidney organoid, a pancreatic organoid, an endometrial organoid, a fallopian tube organoid, a cervical organoid, a prostate organoid, a bladder organoid, an ovarian organoid, a taste bud organoid, or a trophoblastic lining organoid. In some embodiments, the tissue-derived epithelial organoid is a tear gland organoid. In some embodiments, the tissue-derived epithelial organoid is a tonsil organoid. In some embodiments, the tissue-derived epithelial organoid is a salivary gland organoid. In some embodiments, the tissue-derived epithelial organoid is a gastrointestinal organoid. In some embodiments, the tissue-derived epithelial organoid is a thyroid organoid. In some embodiments, the tissue-derived epithelial organoid is a lung organoid. In some embodiments, the tissue-derived epithelial organoid is a mammary organoid. In some embodiments, the tissue-derived epithelial organoid is a liver organoid. In some embodiments, the tissue-derived epithelial organoid is a bile duct organoid. In some embodiments, the tissue-derived epithelial organoid is a gastric organoid. In some embodiments, the tissue-derived epithelial organoid is a kidney organoid. In some embodiments, the tissue-derived epithelial organoid is a pancreatic organoid. In some embodiments, the tissue-derived epithelial organoid is an endometrial organoid. In some embodiments, the tissue-derived epithelial organoid is a fallopian tube organoid. In some embodiments, the tissue-derived epithelial organoid is a cervical organoid. In some embodiments, the tissue-derived epithelial organoid is a prostate organoid. In some embodiments, the tissue-derived epithelial organoid is a bladder organoid. In some embodiments, the tissue-derived epithelial organoid is an ovarian organoid. In some embodiments, the tissue-derived epithelial organoid is a taste bud organoid. In some embodiments, the tissue-derived epithelial organoid is a trophoblastic lining organoid. In some embodiments, the tissue-derived epithelial organoid is selected from the group consisting of lung organoids, gastrointestinal organoids, liver organoids, pancreatic organoids, breast organoids, and combinations thereof.
在某些實施例中,組織來源之上皮類器官為肺類器官。在某些實施例中,組織來源之上皮類器官為肺泡 II 型 (ATII) 類器官。In certain embodiments, the tissue-derived epithelial organoid is a lung organoid. In certain embodiments, the tissue-derived epithelial organoid is an alveolar type II (ATII) organoid.
在某些實施例中,組織來源之上皮類器官為胃腸類器官。在某些實施例中,胃腸類器官為腸類器官。例如但不限於,組織來源之上皮類器官為大腸或迴腸類器官。在某些實施例中,組織來源之上皮類器官為大腸類器官。在某些實施例中,組織來源之上皮類器官為迴腸類器官。在某些實施例中,組織來源之上皮類器官為直腸類器官。在某些實施例中,組織來源之上皮類器官為食道類器官。在某些實施例中,組織來源之上皮類器官為口腔顎面類器官。在某些實施例中,胃腸類器官包含杯狀細胞及腸上皮細胞。In some embodiments, the tissue-derived epithelial organoids are gastrointestinal organoids. In some embodiments, the gastrointestinal organoids are intestinal organoids. For example, but not limited to, the tissue-derived epithelial organoids are colon or ileal organoids. In some embodiments, the tissue-derived epithelial organoids are colon organoids. In some embodiments, the tissue-derived epithelial organoids are ileal organoids. In some embodiments, the tissue-derived epithelial organoids are rectal organoids. In some embodiments, the tissue-derived epithelial organoids are esophageal organoids. In some embodiments, the tissue-derived epithelial organoids are oral maxillofacial organoids. In some embodiments, the gastrointestinal organoids comprise goblet cells and intestinal epithelial cells.
在某些實施例中,組織來源之上皮類器官為乳腺類器官。In certain embodiments, the tissue-derived epithelial organoid is a mammary organoid.
在某些實施例中,組織來源之上皮類器官為胰臟類器官。在某些實施例中,胰臟類器官包含導管細胞, 例如,如實例 6 所揭示的。 In some embodiments, the tissue-derived epithelial organoid is a pancreatic organoid. In some embodiments, the pancreatic organoid comprises vascular cells, for example , as disclosed in Example 6.
在某些實施例中,組織來源之上皮類器官為肝類器官。在某些實施例中,肝類器官包含肝細胞, 例如,如實例 7 所揭示的。 In some embodiments, the tissue-derived epithelial organoid is a liver organoid. In some embodiments, the liver organoid comprises liver cells, for example , as disclosed in Example 7.
在某些實施例中,本揭露之組成物可包括一種或多種不同類型之組織來源之上皮類器官。例如但不限於,本揭露之組成物可包括大腸及迴腸類器官。在某些實施例中,本揭露之組成物可包括肝及膽管類器官。In some embodiments, the compositions of the present disclosure may include epithelial organoids derived from one or more different types of tissues. For example, but not limited to, the compositions of the present disclosure may include colon and ileum organoids. In some embodiments, the compositions of the present disclosure may include liver and bile duct organoids.
在某些實施例中,本揭露之組成物包括約 1 個或多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官, 例如約 2 個或更多個、約 5 個或更多個、約 10 個或更多個、約 50 個或更多個、約 100 個或更多個、約 500 個或更多個、約 1,000 個或更多個、約 5,000 個或更多個、約 10,000 個或更多個、約 50,000 個或更多個、約 100,000 個或更多個、約 500,000 個或更多個、約 1,000,000 個或更多個、約 10,000,000 個或更多個、約 100,000,000 個或更多個或約 1,000,000,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 50 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 100 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 1,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 5,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 10,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 100,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 500,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 1,000,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 10,000,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。在某些實施例中,本揭露之組成物包括約 100,000,000 個或更多個嵌入懸浮於培養基中之水凝膠內之組織來源之上皮類器官。 In certain embodiments, the compositions of the present disclosure include about 1 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium, such as about 2 or more, about 5 or more, about 10 or more, about 50 or more, about 100 or more, about 500 or more, about 1,000 or more, about 5,000 or more, about 10,000 or more, about 50,000 or more, about 100,000 or more, about 500,000 or more, about 1,000,000 or more, about 10,000,000 or more, about 100,000,000 or more, or about 1,000,000,000 or more. In some embodiments, the compositions of the present disclosure include about 50 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In some embodiments, the compositions of the present disclosure include about 100 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In some embodiments, the compositions of the present disclosure include about 1,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 5,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 10,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 100,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 500,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 1,000,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 10,000,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. In certain embodiments, the compositions of the present disclosure include about 100,000,000 or more tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium.
在某些實施例中,水凝膠為三維 (3D) 支架。在某些實施例中,水凝膠由在大於約 10℃ 之溫度固化之材料構成。例如但不限於,水凝膠由在大於約 15℃、大於約 20℃、大於約 25℃、大於約 30℃、大於約 35℃、大於約 40℃、大於約 45℃ 或大於約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 25℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 30℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 35℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 40℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 45℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在大於約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在約 25℃ 至約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在約 25℃ 至約 40℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在約 30℃ 至約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠由在約 30℃ 至約 40℃, 例如約 37℃ 之溫度固化之材料構成。 In some embodiments, the hydrogel is a three-dimensional (3D) scaffold. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 10°C. For example, but not limited to, the hydrogel is composed of a material that cures at a temperature greater than about 15°C, greater than about 20°C, greater than about 25°C, greater than about 30°C, greater than about 35°C, greater than about 40°C, greater than about 45°C, or greater than about 50°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 25°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 30°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 35°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 40°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 45°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature greater than about 50°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature of about 25°C to about 50°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature of about 25°C to about 40°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature of about 30°C to about 50°C. In some embodiments, the hydrogel is composed of a material that cures at a temperature of about 30°C to about 40°C, such as about 37°C.
在某些實施例中,水凝膠為合成水凝膠、天然水凝膠或其組合。在某些實施例中,水凝膠為合成水凝膠。在某些實施例中,水凝膠為天然水凝膠。在某些實施例中,水凝膠可為合成水凝膠及天然水凝膠之混合物。在某些實施例中,水凝膠不包括化學交聯之蛋白質及/或聚合物。In some embodiments, the hydrogel is a synthetic hydrogel, a natural hydrogel, or a combination thereof. In some embodiments, the hydrogel is a synthetic hydrogel. In some embodiments, the hydrogel is a natural hydrogel. In some embodiments, the hydrogel may be a mixture of a synthetic hydrogel and a natural hydrogel. In some embodiments, the hydrogel does not include chemically cross-linked proteins and/or polymers.
在某些實施例中,水凝膠為天然水凝膠。在某些實施例中,天然水凝膠包括一種或多種天然存在組分。例如但不限於,天然水凝膠可包括一種或多種蛋白質, 例如醣蛋白及/或多醣。醣蛋白之非限制性實例包括膠原蛋白 ( 例如,I 型膠原蛋白、II 型膠原蛋白、III 型膠原蛋白、IV 型膠原蛋白、V 型膠原蛋白、VI 型膠原蛋白、VII 型膠原蛋白、VIII 型膠原蛋白、IX 型膠原蛋白、X 型膠原蛋白、 XI 型膠原蛋白及/或 XII 型膠原蛋白)、纖維連接蛋白、巢蛋白、腱生蛋白、玻連蛋白、纖網蛋白、透明質酸及層連結蛋白。在某些實施例中,天然水凝膠可進一步包括一種或多種組分,諸如但不限於多醣、水及/或彈性蛋白。在某些實施例中,本揭露之天然水凝膠包括層連結蛋白、巢蛋白及 IV 型膠原蛋白。在某些實施例中,本揭露之天然水凝膠包括層連結蛋白、巢蛋白、IV 型膠原蛋白及硫酸肝素蛋白聚醣。在某些實施例中,天然水凝膠包括由上皮細胞、內皮細胞、壁內胚層樣細胞及/或結締組織細胞分泌及/或來源之細胞外基質 (ECM)。 In some embodiments, the hydrogel is a natural hydrogel. In some embodiments, the natural hydrogel includes one or more naturally occurring components. For example, but not limited to, the natural hydrogel may include one or more proteins, such as glycoproteins and/or polysaccharides. Non-limiting examples of glycoproteins include collagen ( e.g. , type I collagen, type II collagen, type III collagen, type IV collagen, type V collagen, type VI collagen, type VII collagen, type VIII collagen, type IX collagen, type X collagen, type XI collagen and/or type XII collagen), fibronectin, entactin, tenascin, vitronectin, reticular protein, hyaluronic acid and laminin. In certain embodiments, the natural hydrogel may further include one or more components, such as but not limited to polysaccharides, water and/or elastin. In certain embodiments, the natural hydrogel disclosed herein includes laminin, entactin and type IV collagen. In certain embodiments, the natural hydrogel disclosed herein includes laminin, entactin, type IV collagen and heparin sulfate proteoglycans. In certain embodiments, the natural hydrogel includes extracellular matrix (ECM) secreted and/or derived from epithelial cells, endothelial cells, wall endodermal-like cells and/or connective tissue cells.
在某些實施例中,水凝膠為合成水凝膠。合成水凝膠之非限制性實例包括合成聚合物,諸如普羅結合蛋白 (ProNectin) (Sigma Z378666)、聚乙二醇 (PEG)、聚(甲基丙烯酸羥乙酯)、聚(乙烯亞胺)及聚乙烯醇 (PVA)。中揭示了合成水凝膠及此等合成水凝膠之聚合物之額外非限制性實例揭示於 Unal 及 West (2020) Bioconjugate Chem.31(10):2253-2271;以及 Madduma-Bandarage 及 Madihally (2020) J. of Applied Polymer Science 138(19):e50376 中,各篇之內容以引用方式全文併入本文中。In certain embodiments, the hydrogel is a synthetic hydrogel. Non-limiting examples of synthetic hydrogels include synthetic polymers such as ProNectin (Sigma Z378666), polyethylene glycol (PEG), poly(hydroxyethyl methacrylate), poly(ethyleneimine), and polyvinyl alcohol (PVA). Additional non-limiting examples of synthetic hydrogels and polymers of such synthetic hydrogels are disclosed in Unal and West (2020) Bioconjugate Chem. 31(10):2253-2271; and Madduma-Bandarage and Madihally (2020) J. of Applied Polymer Science 138(19):e50376, each of which is incorporated herein by reference in its entirety.
在某些實施例中,本揭露之水凝膠不包括藻酸鹽。In certain embodiments, the hydrogels disclosed herein do not include alginate.
在某些實施例中,水凝膠可為商業可獲得之 ECM。商業可獲得之 ECM 之非限制性實例包括 ECM 蛋白及來自 Engelbreth-Holm-Swarm (EHS) 小鼠肉瘤細胞之基底膜製劑。在某些實施例中,ECM 為 MATRIGEL™ (BD Biosciences),其包括層連結蛋白、巢蛋白及膠原蛋白 IV。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。BME 之非限制性實例為 CULTREX® 基底膜萃取物 2 型 (R&D Systems),包括層連結蛋白、巢蛋白、膠原蛋白 IV 及硫酸肝素蛋白聚醣。In some embodiments, the hydrogel can be a commercially available ECM. Non-limiting examples of commercially available ECM include ECM proteins and basement membrane preparations from Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells. In some embodiments, the ECM is MATRIGEL™ (BD Biosciences), which includes laminin, entactin, and collagen IV. In some embodiments, the ECM is basement membrane extract (BME), which is a soluble form of basement membrane. A non-limiting example of BME is CULTREX® Basement Membrane Extract Type 2 (R&D Systems), which includes laminin, entactin, collagen IV, and heparin sulfate proteoglycans.
在某些實施例中,水凝膠具有大於約 0.7 mg/ml 之蛋白質濃度, 例如醣蛋白濃度。在某些實施例中,水凝膠具有大於約 1 mg/ml 之蛋白質濃度, 例如醣蛋白濃度。在某些實施例中,水凝膠具有大於約 2 mg/ml 之蛋白質濃度, 例如醣蛋白濃度。在某些實施例中,該水凝膠具有大於約 3 mg/ml 之蛋白質濃度。在某些實施例中,該水凝膠具有大於約 4 mg/ml 之蛋白質濃度。在某些實施例中,該水凝膠具有大於約 5 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有大於約 6 mg/ml、大於約 7 mg/ml、大於約 8 mg/ml、大於約 9 mg/ml或大於約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有約 0.7 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有約 1 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有約 5 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有約 6 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有不小於 0.7 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有不小於 1 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有不小於 5 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠具有不小於 6 mg/ml 之蛋白質濃度。 In some embodiments, the hydrogel has a protein concentration, such as a glycoprotein concentration, greater than about 0.7 mg/ml. In some embodiments, the hydrogel has a protein concentration, such as a glycoprotein concentration, greater than about 1 mg/ml. In some embodiments, the hydrogel has a protein concentration, such as a glycoprotein concentration, greater than about 2 mg/ml. In some embodiments, the hydrogel has a protein concentration, such as a glycoprotein concentration, greater than about 3 mg/ml. In some embodiments, the hydrogel has a protein concentration greater than about 4 mg/ml. In some embodiments, the hydrogel has a protein concentration greater than about 5 mg/ml. In some embodiments, the hydrogel has a protein concentration of greater than about 6 mg/ml, greater than about 7 mg/ml, greater than about 8 mg/ml, greater than about 9 mg/ml, or greater than about 10 mg/ml. In some embodiments, the hydrogel has a protein concentration of about 0.7 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel has a protein concentration of about 1 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel has a protein concentration of about 5 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel has a protein concentration of about 6 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel has a protein concentration of not less than 0.7 mg/ml. In some embodiments, the hydrogel has a protein concentration of not less than 1 mg/ml. In some embodiments, the hydrogel has a protein concentration of not less than 5 mg/ml. In some embodiments, the hydrogel has a protein concentration of not less than 6 mg/ml.
在某些實施例中,水凝膠包含以 w/v % 計大於約 1 w/v % 之 BME 組分、ECM 組分或聚合物。 例如但不限於,水凝膠包含以 w/v % 計大於約 1 w/v %、大於約 1.5 w/v %、大於約 2 w/v %、大於約 2.5 w/v %、大於約 3 w/v %、大於約 3.5 w/v %、大於約 4 w/v %、大於約 4.5 w/v %、大於約 5 w/v %、大於約 5.5 w/v %、大於約 6 w/v %、大於約 6.5 w/v %、大於約 7 w/v %、大於約 7.5 w/v %、大於約 8 w/v %、大於約 8.5 w/v %、大於約 9 w/v %、大於約 9.5 w/v %、大於約 10 w/v %、大於約 10.5 w/v %、大於約 11 w/v %、大於約 11.5 w/v %、大於約 12 w/v %、大於約 12.5 w/v %、大於約 13 w/v %、大於約 13.5 w/v %、大於約 14 w/v %、大於約 14.5 w/v %、大於約 15 w/v %、大於約 15.5 w/v %、大於約 16 w/v %、大於約 16.5 w/v %、大於約 17 w/v %、大於約 17.5 w/v %、大於約 18 w/v %、大於約 18.5 w/v %、大於約 19 w/v %、大於約 19.5 w/v % 或大於約 20 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 1 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 2 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 5 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。 In certain embodiments, the hydrogel comprises greater than about 1 w/v % of a BME component, an ECM component, or a polymer. For example, but not limited to, the hydrogel comprises, in w/v%, greater than about 1 w/v%, greater than about 1.5 w/v%, greater than about 2 w/v%, greater than about 2.5 w/v%, greater than about 3 w/v%, greater than about 3.5 w/v%, greater than about 4 w/v%, greater than about 4.5 w/v%, greater than about 5 w/v%, greater than about 5.5 w/v%, greater than about 6 w/v%, greater than about 6.5 w/v%, greater than about 7 w/v%, greater than about 7.5 w/v%, greater than about 8 w/v%, greater than about 8.5 w/v%, greater than about 9 w/v%, greater than about 9.5 w/v%, greater than about 10 w/v%, greater than about 10.5 w/v%, greater than about %, greater than about 11 w/v %, greater than about 11.5 w/v %, greater than about 12 w/v %, greater than about 12.5 w/v %, greater than about 13 w/v %, greater than about 13.5 w/v %, greater than about 14 w/v %, greater than about 14.5 w/v %, greater than about 15 w/v %, greater than about 15.5 w/v %, greater than about 16 w/v %, greater than about 16.5 w/v %, greater than about 17 w/v %, greater than about 17.5 w/v %, greater than about 18 w/v %, greater than about 18.5 w/v %, greater than about 19 w/v %, greater than about 19.5 w/v %, or greater than about 20 w/v % of a BME component, an ECM component, or a polymer. In some embodiments, the hydrogel comprises about 1 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer. In some embodiments, the hydrogel comprises about 2 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer. In some embodiments, the hydrogel comprises about 5 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer.
在某些實施例中,該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。In certain embodiments, the hydrogel has a storage modulus G' that is equal to or greater than the loss modulus G''.
在某些實施例中,包含組織來源之上皮類器官並懸浮於培養基中之水凝膠具有幾何形狀。在某些實施例中,水凝膠之幾何形狀具有大於約 0.1 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有大於約 0.1 mm之長度。在某些實施例中,水凝膠之幾何形狀具有大於約 0.1 mm之寬度。在某些實施例中,水凝膠之幾何形狀具有大於約 0.1 mm 之直徑。例如但不限於,水凝膠之幾何形狀具有大於約 0.5 mm、大於約 1 mm、大於約 1.5 mm、大於約 2 mm、大於約 2.5 mm、大於約 3 mm、大於約 3.5 mm、大於約 4 mm、大於約 4.5 mm、大於約 5 mm、大於約 5.5 mm、大於約 6 mm、大於約 6.5 mm、大於約 7.5 mm、大於約 8 mm、大於約 8.5 mm、大於約 9 mm、大於約 9.5 mm、大於約 10 mm、大於約 10.5 mm、大於約 11 mm、大於約 11.5 mm、大於約 12 mm、大於約 12.5 mm、大於約 13 mm、大於約 13.5 mm、大於約 14 mm、大於約 14.5 mm、大於約 15 mm、大於約 15.5 mm、大於約 16 mm、大於約 16.5 mm、大於約 17.5 mm、大於約 18 mm、大於約 18.5 mm、大於約 19 mm、大於約 19.5 mm、大於約 20 mm、大於約 50 mm、大於約 100 mm、大於約 150 mm、大於約 200 mm、大於約 250 mm、大於約 300 mm、大於約 350 mm、大於約 400 mm、大於約 450 mm、大於約 500 mm、大於約 550 mm、大於約 600 mm、大於約 650 mm、大於約 700 mm、大於約 750 mm、大於約 800 mm、大於約 850 mm、大於約 900 mm、大於約 950 mm 或大於約 1,000 mm 之長度、寬度及/或直徑。In some embodiments, the hydrogel comprising the tissue-derived epithelial organoids and suspended in the culture medium has a geometry. In some embodiments, the geometry of the hydrogel has a length, width, and/or diameter greater than about 0.1 mm. In some embodiments, the geometry of the hydrogel has a length greater than about 0.1 mm. In some embodiments, the geometry of the hydrogel has a width greater than about 0.1 mm. In some embodiments, the geometry of the hydrogel has a diameter greater than about 0.1 mm. For example, but not limited to, the geometric shape of the hydrogel has a diameter greater than about 0.5 mm, greater than about 1 mm, greater than about 1.5 mm, greater than about 2 mm, greater than about 2.5 mm, greater than about 3 mm, greater than about 3.5 mm, greater than about 4 mm, greater than about 4.5 mm, greater than about 5 mm, greater than about 5.5 mm, greater than about 6 mm, greater than about 6.5 mm, greater than about 7.5 mm, greater than about 8 mm, greater than about 8.5 mm, greater than about 9 mm, greater than about 9.5 mm, greater than about 10 mm, greater than about 10.5 mm, greater than about 11 mm, greater than about 11.5 mm, greater than about 12 mm, greater than about 12.5 mm, greater than about 13 mm, greater than about 13.5 mm, greater than about 14 mm, greater than about 15 mm, greater than about 16 mm, greater than about 17 mm, greater than about 18 mm, greater than about 19 mm, greater than about 20 mm, greater than about 21 mm, greater than about 22 mm, greater than about 23 mm, greater than about 24 mm, greater than about 25 mm, greater than about 26 mm, greater than about 27 mm, greater than about 28 mm, greater than about 29 mm, greater than about 30 mm, greater than about 31 mm, greater than about 32 mm, greater than about 33 mm, greater than about 34 mm, greater than about 35 mm, greater than about 36 mm, greater than about 37 mm, greater than about 38 mm, greater than about 39 mm, 14.5 mm, greater than about 15 mm, greater than about 15.5 mm, greater than about 16 mm, greater than about 16.5 mm, greater than about 17.5 mm, greater than about 18 mm, greater than about 18.5 mm, greater than about 19 mm, greater than about 19.5 mm, greater than about 20 mm, greater than about 50 mm, greater than about 100 mm, greater than about 150 mm, greater than about 200 mm, greater than about 250 mm, greater than about 300 mm, greater than about 350 mm, greater than about 400 mm, greater than about 450 mm, greater than about 500 mm, greater than about 550 mm, greater than about 600 mm, greater than about 650 mm, greater than about 700 mm, greater than about 750 mm, greater than about 800 mm mm, greater than about 850 mm, greater than about 900 mm, greater than about 950 mm, or greater than about 1,000 mm in length, width, and/or diameter.
在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 1000 mm, 例如,約 0.1 mm 至約 500 mm、約 0.1 mm 至約 100 mm、約 0.1 mm 至約 50 mm、約 0.1 mm 至約 20 mm、約 0.1 mm 至約 10 mm、約 1 mm 至約 1000 mm、約 20 mm 至約 1000 mm、約 50 mm 至約 1000 mm、約 100 mm 至約 1000 mm、約 500 mm 至約 1000 mm、約 1 mm 至約 100 mm 或約 1 mm 至約 50 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 20.0 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 20.0 mm 之長度。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 20.0 mm 之寬度。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 20.0 mm 之直徑。例如但不限於,水凝膠之幾何形狀具有約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 1 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 4 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 0.1 mm 至約 5 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 1 mm 至約 20 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 1 mm 至約 10 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 1 mm 至約 5 mm 之長度、寬度及/或直徑。在某些實施例中,水凝膠之幾何形狀具有約 1 mm 至約 4 mm 之長度、寬度及/或直徑。 In certain embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 0.1 mm to about 1000 mm, e.g. , about 0.1 mm to about 500 mm, about 0.1 mm to about 100 mm, about 0.1 mm to about 50 mm, about 0.1 mm to about 20 mm, about 0.1 mm to about 10 mm, about 1 mm to about 1000 mm, about 20 mm to about 1000 mm, about 50 mm to about 1000 mm, about 100 mm to about 1000 mm, about 500 mm to about 1000 mm, about 1 mm to about 100 mm, or about 1 mm to about 50 mm. In some embodiments, the geometric shapes of the hydrogel have a length, width, and/or diameter of about 0.1 mm to about 20.0 mm. In some embodiments, the geometric shapes of the hydrogel have a length of about 0.1 mm to about 20.0 mm. In some embodiments, the geometric shapes of the hydrogel have a width of about 0.1 mm to about 20.0 mm. In some embodiments, the geometric shapes of the hydrogel have a diameter of about 0.1 mm to about 20.0 mm. For example, but not limited to, the geometric shape of the hydrogel has a diameter of about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 mm, about 0.5 mm to about 20 mm, about 1 mm to about 20 mm, about 2 mm to about 20 mm, about 3 mm to about 20 mm, about 4 mm to about 20 mm, about 5 mm to about 20 mm, about 6 mm to about 20 mm, about 7 mm to about 20 mm, about 8 mm to about 20 mm, about 9 mm to about 20 mm, about 10 mm to about 20 mm, about 11 mm to about 20 mm, about 12 mm to about 20 mm, about 13 mm to about 20 mm, about 14 mm to about 20 mm, about 15 mm to about 20 mm, about 16 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 10 mm, about 1 mm to about 5 mm, or about 1 mm to about 4 mm In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 0.1 mm to about 1 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 0.1 mm to about 4 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 0.1 mm to about 5 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 1 mm to about 20 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 1 mm to about 10 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 1 mm to about 5 mm. In some embodiments, the geometric shape of the hydrogel has a length, width, and/or diameter of about 1 mm to about 4 mm.
在某些實施例中,懸浮於培養基中之水凝膠之幾何形狀為小滴, 例如,如圖6 所示。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 1 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 4 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 20 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 10 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 4 mm 之直徑。在某些實施例中,各水凝膠小滴包括約 1 個或多個組織來源之上皮類器官, 例如,約 2 個或更多個、約 5 個或更多個、約 10 個或更多個、約 50 個或更多個、約 100 個或更多個、約 500 個或更多個,約 1,000 個或更多個、約 5,000 個或更多個、或約 10,000 個或更多個組織來源之上皮類器官。 In some embodiments, the geometry of the hydrogel suspended in the culture medium is a droplet, for example , as shown in FIG. 6 . In certain embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 20 mm, e.g. , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 1 mm. In some embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 4 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 20 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 10 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 4 mm. In some embodiments, each hydrogel droplet comprises about 1 or more tissue-derived epithelial organoids, e.g. , about 2 or more, about 5 or more, about 10 or more, about 50 or more, about 100 or more, about 500 or more, about 1,000 or more, about 5,000 or more, or about 10,000 or more tissue-derived epithelial organoids.
在某些實施例中,水凝膠具有絲狀結構, 例如,如圖6 所示。在某些實施例中,該絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,絲狀結構具有線形形狀。在某些實施例中,絲狀結構具有蛇形形狀。在某些實施例中,絲狀結構具有螺旋形形狀。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1000 mm, 例如,約 0.1 mm 至約 500 mm、約 0.1 mm 至約 100 mm、約 0.1 mm 至約 50 mm、約 0.1 mm 至約 20 mm、約 0.1 mm 至約 10 mm、約 1 mm 至約 1000 mm、約 20 mm 至約 1000 mm、約 50 mm 至約 1000 mm、約 100 mm 至約 1000 mm、約 500 mm 至約 1000 mm、約 1 mm 至約 100 mm 或約 1 mm 至約 50 mm 之長度及/或寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之長度及/或寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1 mm 之長度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 4 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 4 mm 之長度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1 mm 之寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 4 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 4 mm 之寬度。 In some embodiments, the hydrogel has a filamentary structure, for example , as shown in FIG6 . In some embodiments, the filamentary structure has a linear, serpentine, or spiral shape. In some embodiments, the filamentary structure has a linear shape. In some embodiments, the filamentary structure has a serpentine shape. In some embodiments, the filamentary structure has a spiral shape. In certain embodiments, the filamentous structure has a length and/or width of about 0.1 mm to about 1000 mm, e.g. , about 0.1 mm to about 500 mm, about 0.1 mm to about 100 mm, about 0.1 mm to about 50 mm, about 0.1 mm to about 20 mm, about 0.1 mm to about 10 mm, about 1 mm to about 1000 mm, about 20 mm to about 1000 mm, about 50 mm to about 1000 mm, about 100 mm to about 1000 mm, about 500 mm to about 1000 mm, about 1 mm to about 100 mm, or about 1 mm to about 50 mm. In certain embodiments, the filamentous structure has a diameter of about 0.1 mm to about 20 mm, for example , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the filamentous structure has a length and/or width of about 0.1 mm to about 4 mm. In some embodiments, the filamentous structure has a length of about 0.1 mm to about 1 mm. In some embodiments, the filamentous structure has a length of about 0.1 mm to about 4 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 20 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 10 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 4 mm. In some embodiments, the filamentous structure has a width of about 0.1 mm to about 1 mm. In some embodiments, the filamentous structure has a width of about 0.1 mm to about 4 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 20 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 10 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 4 mm.
在某些實施例中,絲狀結構具有約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 20 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 10 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 5 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 5 mm 之直徑。 In certain embodiments, the filamentous structure has a diameter of about 0.1 mm to about 20 mm, for example , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 20 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 10 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 5 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 20 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 10 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 5 mm.
在某些實施例中,各絲狀結構包括約 1 個或多個組織來源之上皮類器官, 例如,約 2 個或更多個、約 5 個或更多個、約 10 個或更多個、約 50 個或更多個、約 100 個或更多個、約 500 個或更多個、約 1,000 個或更多個、約 5,000 個或更多個、或約 10,000 個或更多個組織來源之上皮類器官。 In certain embodiments, each filamentous structure comprises about 1 or more tissue-derived epithelial organoids, e.g. , about 2 or more, about 5 or more, about 10 or more, about 50 or more, about 100 or more, about 500 or more, about 1,000 or more, about 5,000 or more, or about 10,000 or more tissue-derived epithelial organoids.
在某些實施例中,組成物包括任何合適的細胞培養基。在某些實施例中,細胞培養基含有對於支持培養細胞之維持而言重要的組分。 在某些實施例中,使用於本揭露之細胞培養基可為營養液,其包括標準細胞培養成分,諸如但不限於胺基酸、維生素、無機鹽、碳能源 ( 例如,葡萄糖) 及緩衝液。在某些實施例中,該培養基為分化培養基。在某些實施例中,培養基為生長培養基。在某些實施例中,該培養基為幹細胞促進培養基。實例中提供了細胞培養基之非限制性實例, 例如類器官生長培養基。 In some embodiments, the composition includes any suitable cell culture medium. In some embodiments, the cell culture medium contains components that are important for supporting the maintenance of cultured cells. In some embodiments, the cell culture medium used in the present disclosure may be a nutrient solution, which includes standard cell culture ingredients, such as but not limited to amino acids, vitamins, inorganic salts, carbon energy ( e.g. , glucose) and buffer. In some embodiments, the culture medium is a differentiation medium. In some embodiments, the culture medium is a growth medium. In some embodiments, the culture medium is a stem cell promotion medium. Non-limiting examples of cell culture mediums are provided in the examples, such as organoid growth medium.
在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 或更大。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 至約 1:100。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:5 至約 1:100、約 1:10 至約 1:100、約 1:15 至約 1:100、約 1:20 至約 1:100、約 1:25 至約 1:100、約 1:30 至約 1:100、約 1:35 至約 1:100、約 1:40 至約 1:100、約 1:45 至約 1:100、約 1:45 至約 1:100、約 1:45 至約 1:100、約 1:45 至約 1:100、約 1:50 至約 1:100、約 1:55 至約 1:100、約 1:60 至約 1:100、約 1:65 至約 1:100、約 1:70 至約 1:100、約 1:75 至約 1:100、約 1:80 至約 1:100、約 1:85 至約 1:100、約 1:90 至約 1:100、約 1:95 至約 1:100, 1:5 至約 1:50、約 1:10 至約 1:50、約 1:15 至約 1:50、約 1:20 至約 1:50、約 1:25 至約 1:50、約 1:30 至約 1:50、約 1:35 至約 1:50、約 1:40 至約 1:50、約 1:45 至約 1:50、約 1:1 至約 1:95、約 1:1 至約 1:90、約 1:1 至約 1:85、約 1:1 至約 1:80、約 1:1 至約 1:75、約 1:1 至約 1:70、約 1:1 至約 1:65、約 1:1 至約 1:60、約 1:1 至約 1:55、約 1:1 至約 1:50、約 1:1 至約 1:45、約 1:1 至約 1:40、約 1:1 至約 1:35、約 1:1 至約 1:30、約 1:1 至約 1:35、約 1:1 至約 1:30、約 1:1 至約 1:25、約 1:1 至約 1:20、約 1:1 至約 1:15、約 1:1 至約 1:10、約 1:1 至約 1:5、約 1:5 至約 1:75、約 1:5 至約 1:60、約 1:5 至約 1:50、約 1:1 至約 1:40、約 1:5 至約 1:30 或約 1:5 至約 1:20。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 至約 1:50。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:2 至約 1:50。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 至約 1:20。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 至約 1:15。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:1。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:2。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:5。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:10, 例如,如實例 8 所示。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:20。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:30。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:40。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:50。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:60。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:70。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:80。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:90。在某些實施例中,組成物中水凝膠體積與培養基體積之比率 (體積比) 為約 1:100。 In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1 or greater. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1 to about 1:100. In certain embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:5 to about 1:100, about 1:10 to about 1:100, about 1:15 to about 1:100, about 1:20 to about 1:100, about 1:25 to about 1:100, about 1:30 to about 1:100, about 1:35 to about 1:100, about 1:40 to about 1:100, about 1:45 to about 1:100, about 1:45 to about 1:100, about 1:45 to about 1:100, about 1:45 to about 1:100, about 1:50 to about 1:100, about 1:55 to about 1:100, about 1:60 to about 1:100, about 1:65 to about 1:100, about 1:70 to about 1:100, about 1:75 to about 1:100, about 1:80 to about 1:100, about 1:85 to about 1:100, about 1:90 to about 1:100, about 1:95 to about 1:100, 1:5 to about 1:50, about 1:10 to about 1:50, about 1:15 to about 1:50, about 1:20 to about 1:50, about 1:25 to about 1:50, about 1:30 to about 1:50, about 1:35 to about 1:50, about 1:40 to about 1:50, about 1:45 to about 1:50, about 1:1 to about 1:95, about 1:1 to about 1:90, about 1:1 to about 1:85, about 1:1 to about 1:80, about 1:1 to about 1:75, about 1:1 to about 1:70, about 1:1 to about 1:65, about 1:1 to about 1:60, about 1:1 to about 1:55, about 1:1 to about 1:50, about 1:1 to about 1:45, about 1:1 to about 1:40, about 1:1 to about 1:35, about 1:1 to about 1:30, about 1:1 to about 1:35, about 1:1 to about 1:30, about 1:1 to about 1:25, about 1:1 to about 1:20, about 1:1 to about 1:15, about 1:1 to about 1:10, about 1:1 to about 1:5, about 1:5 In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1 to about 1:50. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:2 to about 1:50. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1 to about 1:20. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1 to about 1:15. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:1. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:2. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:5. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:10, for example , as shown in Example 8. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:20. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:30. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:40. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:50. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:60. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:70. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:80. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:90. In some embodiments, the ratio of the volume of hydrogel to the volume of the culture medium in the composition (volume ratio) is about 1:100.
在某些實施例中,與參考組織來源之上皮類器官 ( 例如,嵌入接附於基材之水凝膠內之組織來源之上皮類器官) 相比,組織來源之上皮類器官在尺寸上更均勻。在某些實施例中,與參考組織來源之上皮類器官相比藉由本揭露之方法產生之組織來源之上皮類器官由於如實例 1 中所述之營養素利用率差異而在尺寸上更均勻。例如但不限於,與參考組織來源之上皮類器官 ( 例如,嵌入接附於基材之水凝膠內之組織來源之上皮類器官) 相比,藉由本揭露之方法產生之組織來源之上皮類器官在尺寸上在懸浮培養物小滴 ( 例如,經懸浮之水凝膠小滴) 之寬度上更均勻。在某些實施例中,參考組織來源之上皮類器官在如實例 1 所揭示之水凝膠圓頂中產生。 In certain embodiments, the tissue-derived epithelial organoids are more uniform in size compared to a reference tissue-derived epithelial organoid ( e.g. , a tissue-derived epithelial organoid embedded in a hydrogel attached to a substrate). In certain embodiments, the tissue-derived epithelial organoids produced by the methods of the present disclosure are more uniform in size compared to reference tissue-derived epithelial organoids due to differences in nutrient utilization as described in Example 1. For example, but not limited to, tissue-derived epithelial organoids produced by the methods of the present disclosure are more uniform in size across the width of a suspended culture droplet ( e.g. , a suspended hydrogel droplet) than a reference tissue-derived epithelial organoid ( e.g. , a tissue-derived epithelial organoid embedded in a hydrogel attached to a substrate). In certain embodiments, the reference tissue-derived epithelial organoid is produced in a hydrogel dome as disclosed in Example 1.
在某些實施例中,本揭露之組織來源之上皮類器 ( 例如,組織來源之上皮類器官之群體) 以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如,較高或較低) 的含量表現標記物。例如但不限於,與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比,本揭露之組織來源之上皮類器官 ( 例如,組織來源之上皮類器官之群體) 以較高含量表現標記物。可替代地或另外,在某些實施例中,與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比,本揭露之組織來源之上皮類器官 ( 例如,組織來源之上皮類器官之群體) 以較低之含量表現標記物。在某些實施例中,標記物為幹細胞及/或增生標記物, 例如,與幹細胞及/或增生相關之基因,如圖5A 所示。 例如但不限於,幹細胞及/或增生標記物為 MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1 及/或 CD44。在某些實施例中,標記物為分化標記物, 例如與分化相關之基因,如圖5B 所示。例如但不限於,分化標記物為角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、及/或平滑肌肌動蛋白 (SMA)。在某些實施例中,該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。例如但不限於,參考組織來源之上皮類器官為在如實例 1 中所揭示之水凝膠圓頂中產生之組織來源之上皮類器官。 In certain embodiments, a tissue-derived epithelial organoid ( e.g. , a population of tissue-derived epithelial organoids) of the present disclosure expresses a marker at a different ( e.g. , higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). For example, but not limited to, a tissue-derived epithelial organoid ( e.g. , a population of tissue-derived epithelial organoids) of the present disclosure expresses a marker at a higher level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). Alternatively or additionally, in some embodiments, the tissue-derived epithelial organoids ( e.g. , a population of tissue-derived epithelial organoids) of the present disclosure express a marker at a lower level compared to a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In some embodiments, the marker is a stem cell and/or proliferation marker, for example , a gene associated with stem cells and/or proliferation, as shown in FIG5A . For example, but not limited to, the stem cell and/or proliferation marker is MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1 and/or CD44. In some embodiments, the marker is a differentiation marker, such as a gene associated with differentiation, as shown in FIG5B . For example, but not limited to, the differentiation marker is keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), and/or smooth muscle actin (SMA). In some embodiments, the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. For example, but not limited to, the reference tissue-derived epithelial organoid is a tissue-derived epithelial organoid produced in a hydrogel dome as disclosed in Example 1.
在某些實施例中,組織來源之上皮類器官為胃腸類器官,且本揭露之胃腸類器官中之差異表現標記物為 MKI67、LGR5、SOX9、CD44、MUC2、MUC5B、TFF3、KRT20、FABP1、ALPI 及/或 CEACAM7。 In certain embodiments, the tissue-derived epithelial organoid is a gastrointestinal organoid, and the differentially expressed markers in the gastrointestinal organoids disclosed herein are MKI67, LGR5, SOX9, CD44, MUC2, MUC5B, TFF3, KRT20, FABP1, ALPI and/or CEACAM7.
在某些實施例中,組織來源之上皮類器官為乳腺類器官,且本揭露之乳腺類器官中之差異表現標記物為 EpCAM、CD49f、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14) 及/或平滑肌肌動蛋白 (SMA)。In certain embodiments, the tissue-derived epithelial organoid is a mammary organoid, and the differentially expressed markers in the mammary organoids of the present disclosure are EpCAM, CD49f, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), and/or smooth muscle actin (SMA).
在某些實施例中,組織來源之上皮類器官為胰臟類器官,且本揭露之胰臟類器官中之差異表現標記物為 CD133、LGR5、PDX1、SOX9、ALDH1A1、NEUROG3、NKX6.1、角蛋白 19 (KRT19)、MUC1、INS、GCG 及/或 AMY。In certain embodiments, the tissue-derived epithelial organoid is a pancreatic organoid, and the differentially expressed markers in the pancreatic organoids of the disclosure are CD133, LGR5, PDX1, SOX9, ALDH1A1, NEUROG3, NKX6.1, keratin 19 (KRT19), MUC1, INS, GCG and/or AMY.
在某些實施例中,組織來源之上皮類器官為肝類器官,且本揭露之肝類器官中之差異表現標記物為 LGR5、ALB、CYP3A4、HNF4A、KRT19、KRT7 及/或 SOX9。In certain embodiments, the tissue-derived epithelial organoid is a liver organoid, and the differentially expressed markers in the liver organoids of the disclosure are LGR5, ALB, CYP3A4, HNF4A, KRT19, KRT7 and/or SOX9.
在某些實施例中,與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物由本揭露之組織來源之上皮類器官之群體 ( 例如,本揭露之組成物中之組織來源之上皮類器官之群體) 以較高含量表現。幹細胞及/或增生標記物之非限制性實例包括 MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。在某些實施例中,幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、ASCL2、LGR5、SOX9、SMOC2、CD44 及其組合。在某些實施例中,幹細胞及/或增生標記物為 MKI67。在某些實施例中,幹細胞及/或增生標記物為 ASCL2。在某些實施例中,幹細胞及/或增生標記物為 LGR5。在某些實施例中,幹細胞及/或增生標記物為 SOX9。在某些實施例中,幹細胞及/或增生標記物為 SMOC2。在某些實施例中,幹細胞及/或增生標記物為 CD44。在某些實施例中,幹細胞及/或增生標記物為 EpCAM。在某些實施例中,幹細胞及/或增生標記物為 CD49f。在某些實施例中,幹細胞及/或增生標記物為 CD133。在某些實施例中,幹細胞及/或增生標記物為 ALDH1A1。在某些實施例中,幹細胞及/或增生標記物為 NEUROG3。在某些實施例中,幹細胞及/或增生標記物為 NKX6.1。在某些實施例中,幹細胞及/或增生標記物為 PDX1。在某些實施例中,幹細胞及/或增生標記物為 BMI1。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 10%、高至少 20%、高至少 30%、高至少 40%、高至少 50%、高至少 60%、高至少 70%、高至少 80%、高至少 90%、高至少 100%、高至少 110%、高至少 120%、高至少 130%、高至少 140%、高至少 150%、高至少 160%、高至少 170%、高至少 180%、高至少 190%、高至少 200%、高至少 210%、高至少 220%、高至少 230%、高至少 240%、高至少 250%、高至少 260%、高至少 270%、高至少 280%、高至少 290% 或高至少 300%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 50%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 100%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 200%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 300%。 In certain embodiments, stem cell and/or proliferation markers are expressed at higher levels by a population of tissue-derived epithelial organoids of the present disclosure ( e.g. , a population of tissue-derived epithelial organoids in a composition of the present disclosure) compared to a population of reference tissue-derived epithelial organoids. Non-limiting examples of stem cell and/or proliferation markers include MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. In certain embodiments, the stem cell and/or proliferation marker is selected from the group consisting of MKI67, ASCL2, LGR5, SOX9, SMOC2, CD44, and combinations thereof. In some embodiments, the stem cell and/or proliferation marker is MKI67. In some embodiments, the stem cell and/or proliferation marker is ASCL2. In some embodiments, the stem cell and/or proliferation marker is LGR5. In some embodiments, the stem cell and/or proliferation marker is SOX9. In some embodiments, the stem cell and/or proliferation marker is SMOC2. In some embodiments, the stem cell and/or proliferation marker is CD44. In some embodiments, the stem cell and/or proliferation marker is EpCAM. In some embodiments, the stem cell and/or proliferation marker is CD49f. In some embodiments, the stem cell and/or proliferation marker is CD133. In some embodiments, the stem cell and/or proliferation marker is ALDH1A1. In some embodiments, the stem cell and/or proliferation marker is NEUROG3. In some embodiments, the stem cell and/or proliferation marker is NKX6.1. In some embodiments, the stem cell and/or proliferation marker is PDX1. In some embodiments, the stem cell and/or proliferation marker is BMI1. In certain embodiments, the expression level of a stem cell and/or proliferation marker in a population of tissue-derived epithelial organoids of the present disclosure is at least 10% higher, at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 60% higher, at least 70% higher, at least 80% higher, at least 90% higher, at least 100% higher, at least 110% higher, at least 120% higher, at least 130% higher, at least 140% higher, at least 150% higher, at least 160% higher, at least 170% higher, at least 180% higher, at least 190% higher, at least 200% higher, at least 210% higher, at least 220% higher, at least 230% higher, at least 240% higher, at least 250% higher, at least 260% higher, at least 270% higher, at least 280% higher, at least 290% higher In some embodiments, the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue is at least 50% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue. In some embodiments, the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue is at least 100% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a tissue of the present disclosure is at least 200% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a tissue of the present disclosure is at least 300% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue.
在某些實施例中,與參考組織來源之上皮類器官之群體相比,分化標記物由本揭露之組織來源之上皮類器官之群體 ( 例如,本揭露之組成物中之組織來源之上皮類器官之群體) 以較低含量表現。分化標記物之非限制性實例包括角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。在某些實施例中,分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、TFF3、ALPI、SI、CEACAM7 及其組合。在某些實施例中,分化標記物為角蛋白 20 (KRT20)。在某些實施例中,分化標記物為 FABP1。在某些實施例中,分化標記物為 MUC2。在某些實施例中,分化標記物為 MUC5B。在某些實施例中,分化標記物為 TFF3。在某些實施例中,分化標記物為 ALPI。在某些實施例中,分化標記物為 SI。在某些實施例中,分化標記物為 CEACAM7。在某些實施例中,分化標記物為角蛋白 19 (KRT19)。在某些實施例中,分化標記物為角蛋白 7 (KRT7)。在某些實施例中,分化標記物為 SOX9。在某些實施例中,分化標記物為 MUC1。在某些實施例中,分化標記物為 INS。在某些實施例中,分化標記物為 GCG。在某些實施例中,分化標記物為 AMY。在某些實施例中,分化標記物為 ALB。在某些實施例中,分化標記物為 CYP3A4。在某些實施例中,分化標記物為 HNF4A。在某些實施例中,分化標記物為細胞角蛋白 8 (K8)。在某些實施例中,分化標記物為細胞角蛋白 18 (K18)。在某些實施例中,分化標記物為細胞角蛋白 5 (K5)。在某些實施例中,分化標記物為細胞角蛋白 14 (K14)。在某些實施例中,分化標記物為平滑肌肌動蛋白 (SMA)。在某些實施例中,分化標記物為 MUC5AC。在某些實施例中,分化標記物為 MUC6。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 10%、低至少 20%、低至少 30%、低至少 40%、低至少 50%、低至少 60%、低至少 70%、低至少 80%、低至少 90%、低至少 100%、低至少 110%、低至少 120%、低至少 130%、低至少 140%、低至少 150%、低至少 160%、低至少 170%、低至少 180%、低至少 190%、低至少 200%、低至少 210%、低至少 220%、低至少 230%、低至少 240%、低至少 200%、低至少 250%、低至少 260%、低至少 270%、低至少 280%、低至少 290% 或低至少 300%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 50%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 100%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 200%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,本揭露之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 300%。 III. 產生類器官之方法 In certain embodiments, a differentiation marker is expressed at a lower level by a population of tissue-derived epithelial organoids of the disclosure ( eg , a population of tissue-derived epithelial organoids in a composition of the disclosure) compared to a population of reference tissue-derived epithelial organoids. Non-limiting examples of differentiation markers include keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA), and combinations thereof. In certain embodiments, the differentiation marker is selected from the group consisting of keratin 20 (KRT20), FABP1, MUC2, MUC5B, TFF3, ALPI, SI, CEACAM7, and combinations thereof. In some embodiments, the differentiation marker is keratin 20 (KRT20). In some embodiments, the differentiation marker is FABP1. In some embodiments, the differentiation marker is MUC2. In some embodiments, the differentiation marker is MUC5B. In some embodiments, the differentiation marker is TFF3. In some embodiments, the differentiation marker is ALPI. In some embodiments, the differentiation marker is SI. In some embodiments, the differentiation marker is CEACAM7. In some embodiments, the differentiation marker is keratin 19 (KRT19). In some embodiments, the differentiation marker is keratin 7 (KRT7). In some embodiments, the differentiation marker is SOX9. In some embodiments, the differentiation marker is MUC1. In some embodiments, the differentiation marker is INS. In some embodiments, the differentiation marker is GCG. In some embodiments, the differentiation marker is AMY. In some embodiments, the differentiation marker is ALB. In some embodiments, the differentiation marker is CYP3A4. In some embodiments, the differentiation marker is HNF4A. In some embodiments, the differentiation marker is cytokeratin 8 (K8). In some embodiments, the differentiation marker is cytokeratin 18 (K18). In some embodiments, the differentiation marker is cytokeratin 5 (K5). In some embodiments, the differentiation marker is cytokeratin 14 (K14). In some embodiments, the differentiation marker is smooth muscle actin (SMA). In some embodiments, the differentiation marker is MUC5AC. In some embodiments, the differentiation marker is MUC6. In certain embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids of the present disclosure is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 100% lower, at least 110% lower, at least 120% lower, at least 130% lower, at least 140% lower, at least 150% lower, at least 160% lower, at least 170% lower, at least 180% lower, at least 190% lower, at least 200% lower, at least 210% lower, at least 220% lower, at least 230% lower, at least 240% lower, at least 200% lower, at least 250% lower, at least 260% lower, at least 270% lower, at least 280% lower, at least 290% lower, or at least 300% lower than the expression level of a differentiation marker in a population of reference tissue-derived epithelial organoids. In certain embodiments, the expression level of differentiation markers in a population of epithelial organoids derived from a tissue of the present disclosure is at least 50% lower than the expression level of differentiation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of differentiation markers in a population of epithelial organoids derived from a tissue of the present disclosure is at least 100% lower than the expression level of differentiation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of differentiation markers in a population of epithelial organoids derived from a tissue of the present disclosure is at least 200% lower than the expression level of differentiation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of a differentiation marker in a population of epithelial organoids derived from a tissue of the present disclosure is at least 300% lower than the expression level of a differentiation marker in a population of epithelial organoids derived from a reference tissue. III. Methods of Generating Organoids
本揭露提供了產生組織來源之上皮類器官之方法。在某些實施例中,本揭露提供了用於產生嵌入在懸浮於培養基中之水凝膠中之組織來源之上皮類器官之方法。本揭露進一步提供了用於產生組織來源之上皮類器官之懸浮培養物之方法。在某些實施例中,可使用實例 1 及 4 至 8 以及圖10 及 11 中描述的方法來產生類器官。The present disclosure provides methods for generating tissue-derived epithelial organoids. In certain embodiments, the present disclosure provides methods for generating tissue-derived epithelial organoids embedded in a hydrogel suspended in a culture medium. The present disclosure further provides methods for generating suspension cultures of tissue-derived epithelial organoids. In certain embodiments, the methods described in Examples 1 and 4 to 8 and Figures 10 and 11 can be used to generate organoids.
在某些實施例中,本揭露提供了用於產生淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官或滋胚內層類器官之方法。在某些實施例中,本揭露提供了用於產生淚腺類器官之方法。在某些實施例中,本揭露提供了用於產生扁桃腺類器官之方法。在某些實施例中,本揭露提供了用於產生唾液腺類器官之方法。在某些實施例中,本揭露提供了用於產生胃腸類器官之方法。在某些實施例中,本揭露提供了用於產生甲狀腺類器官之方法。在某些實施例中,本揭露提供了用於產生肺類器官之方法。在某些實施例中,本揭露提供了用於產生乳腺類器官之方法。在某些實施例中,本揭露提供了用於產生肝類器官之方法。在某些實施例中,本揭露提供了用於產生膽管類器官之方法。在某些實施例中,本揭露提供了用於產生胃類器官之方法。在某些實施例中,本揭露提供了用於產生腎類器官之方法。在某些實施例中,本揭露提供了用於產生胰臟類器官之方法。在某些實施例中,本揭露提供了用於產生子宮內膜類器官之方法。在某些實施例中,本揭露提供了用於產生輸卵管類器官之方法。在某些實施例中,本揭露提供了用於產生子宮頸類器官之方法。在某些實施例中,本揭露提供了用於產生前列腺類器官之方法。在某些實施例中,本揭露提供了用於產生膀胱類器官之方法。在某些實施例中,本揭露提供了用於產生卵巢類器官之方法。在某些實施例中,本揭露提供了用於產生滋胚內層類器官之方法。在某些實施例中,本揭露提供了用於產生滋胚內層類器官之方法。In certain embodiments, the disclosure provides methods for producing tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, or trophoblastic lining organoids. In certain embodiments, the disclosure provides methods for producing tear gland organoids. In certain embodiments, the disclosure provides methods for producing tonsil organoids. In certain embodiments, the disclosure provides methods for producing salivary gland organoids. In certain embodiments, the disclosure provides methods for producing gastrointestinal organoids. In certain embodiments, the disclosure provides methods for producing thyroid organoids. In certain embodiments, the disclosure provides methods for producing lung organoids. In certain embodiments, the disclosure provides methods for producing mammary organoids. In certain embodiments, the disclosure provides methods for producing liver organoids. In certain embodiments, the disclosure provides methods for producing bile duct organoids. In certain embodiments, the disclosure provides methods for producing gastric organoids. In certain embodiments, the disclosure provides methods for producing kidney organoids. In certain embodiments, the disclosure provides methods for producing pancreatic organoids. In certain embodiments, the disclosure provides methods for producing endometrial organoids. In certain embodiments, the disclosure provides methods for producing fallopian tube organoids. In certain embodiments, the disclosure provides methods for producing cervical organoids. In certain embodiments, the disclosure provides methods for producing prostate organoids. In certain embodiments, the disclosure provides methods for producing bladder organoids. In certain embodiments, the disclosure provides methods for producing ovarian organoids. In certain embodiments, the disclosure provides methods for producing trophoblastic lining organoids. In certain embodiments, the disclosure provides methods for producing trophoblastic lining organoids.
在某些實施例中,本揭露提供了用於產生組織來源之上皮類器官之方法,該組織來源之上皮類器官選自由以下所組成之群組:肺類器官、胃腸類器官、肝類器官、胰臟類器官、乳腺類器官及其組合。In certain embodiments, the present disclosure provides methods for generating tissue-derived epithelial organoids selected from the group consisting of lung organoids, gastrointestinal organoids, liver organoids, pancreatic organoids, breast organoids, and combinations thereof.
在某些實施例中,本揭露提供了用於產生胃腸類器官之方法。在某些實施例中,本揭露提供了用於產生腸類器官之方法。例如但不限於,腸類器官為大腸或迴腸類器官。在某些實施例中,腸類器官為大腸類器官。在某些實施例中,腸類器官為迴腸類器官。在某些實施例中,本揭露提供了用於產生直腸類器官之方法。在某些實施例中,本揭露提供了用於產生食道類器官之方法。在某些實施例中,本揭露提供了用於產生口腔顎面類器官之方法。在某些實施例中,本揭露之方法可產生兩種或更多種不同類型之組織來源之上皮類器官, 例如大腸及迴腸類器官。 In certain embodiments, the present disclosure provides methods for producing gastrointestinal organoids. In certain embodiments, the present disclosure provides methods for producing intestinal organoids. For example, but not limited to, the intestinal organoids are colon or ileal organoids. In certain embodiments, the intestinal organoids are colon organoids. In certain embodiments, the intestinal organoids are ileal organoids. In certain embodiments, the present disclosure provides methods for producing rectal organoids. In certain embodiments, the present disclosure provides methods for producing esophageal organoids. In certain embodiments, the present disclosure provides methods for producing oral maxillofacial organoids. In certain embodiments, the methods of the present disclosure can produce epithelial organoids from two or more different types of tissue sources, such as colon and ileal organoids.
在某些實施例中,本揭露提供了用於產生肺類器官之方法。In certain embodiments, the present disclosure provides methods for generating lung organoids.
在某些實施例中,本揭露提供了用於產生肝類器官之方法。In certain embodiments, the present disclosure provides methods for generating liver organoids.
在某些實施例中,本揭露提供了用於產生胰臟類器官之方法。In certain embodiments, the present disclosure provides methods for generating pancreatic organoids.
在某些實施例中,本揭露提供了用於產生乳腺類器官之方法。In certain embodiments, the present disclosure provides methods for generating mammary organoids.
在某些實施例中,用於產生組織來源之上皮類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。In certain embodiments, a method for generating tissue-derived epithelial organoids comprises contacting tissue-derived epithelial stem cells with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells may be combined with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture.
在某些實施例中,複數個組織來源之上皮幹細胞包括至少兩個或更多個組織來源之上皮幹細胞。在某些實施例中,複數個組織來源之上皮幹細胞包括至少約 10 個或更多個組織來源之上皮幹細胞、至少約 100 個或更多個組織來源之上皮幹細胞、至少約 1,000 個或更多個組織來源之上皮幹細胞、至少約 10,000 個或更多個組織來源之上皮幹細胞、至少約 100,000 個或更多個組織來源之上皮幹細胞、至少約 100,000 個或更多個組織來源之上皮幹細胞、至少約 1,000,000 個或更多個組織來源之上皮幹細胞、至少約 10,000,000 個或更多個組織來源之上皮幹細胞、或至少約 100,000,000 個或更多個組織來源之上皮幹細胞、或至少約 1,000,000,000 個或更多個組織來源之上皮幹細胞。在某些實施例中,複數個組織來源之上皮幹細胞包括至少約 10 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 100 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 1,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 10,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 100,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 100,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 1,000,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 10,000,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、至少約 100,000,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠、或至少約 1,000,000,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,複數個組織來源之上皮幹細胞包括至少約 10,000 個或更多個組織來源之上皮幹細胞/ml 水凝膠。In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises at least two or more tissue-derived epithelial stem cells. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises at least about 10 or more tissue-derived epithelial stem cells, at least about 100 or more tissue-derived epithelial stem cells, at least about 1,000 or more tissue-derived epithelial stem cells, at least about 10,000 or more tissue-derived epithelial stem cells, at least about 100,000 or more tissue-derived epithelial stem cells, at least about 100,000 or more tissue-derived epithelial stem cells, at least about 1,000,000 or more tissue-derived epithelial stem cells, at least about 10 ... The invention relates to an epithelial cell transplantation method comprising at least about 100,000,000 or more tissue-derived epithelial stem cells, or at least about 1,000,000,000 or more tissue-derived epithelial stem cells. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises at least about 10 or more tissue-derived epithelial stem cells/ml hydrogel, at least about 100 or more tissue-derived epithelial stem cells/ml hydrogel, at least about 1,000 or more tissue-derived epithelial stem cells/ml hydrogel, at least about 10 ... In some embodiments, the plurality of tissue-derived epithelial stem cells comprises at least about 10,000,000 or more tissue-derived epithelial stem cells/ml hydrogel, at least about 100,000,000 or more tissue-derived epithelial stem cells/ml hydrogel, or at least about 1,000,000,000 or more tissue-derived epithelial stem cells/ml hydrogel. In some embodiments, the plurality of tissue-derived epithelial stem cells comprises at least about 10,000 or more tissue-derived epithelial stem cells/ml hydrogel.
在某些實施例中,複數個組織來源之上皮幹細胞包括約 1 × 10 4至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠。例如但不限於,複數個組織來源之上皮幹細胞包括約 1 × 10 4至約 1 × 10 9個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 104 至約 1 × 10 8個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 4至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 4至約 1 × 10 6個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 4至約 1 × 10 5個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 5至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 6至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 7至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 8至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 9至約 1 × 10 10個組織來源之上皮幹細胞/ml 水凝膠、約 1 × 10 5至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠、或約 1 × 10 5至約 1 × 10 8個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,複數個組織來源之上皮幹細胞包括約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,複數個組織來源之上皮幹細胞包括約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 6個組織來源之上皮幹細胞/ml 水凝膠。 In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1×10 4 to about 1×10 10 tissue-derived epithelial stem cells/ml hydrogel. For example, but not limited to, the plurality of tissue-derived epithelial stem cells include about 1 × 10 4 to about 1 × 10 9 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 4 to about 1 × 10 8 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 4 to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 4 to about 1 × 10 6 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 4 to about 1 × 10 5 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 5 to about 1 × 10 6 ... 10 10 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 6 to about 1 × 10 10 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 7 to about 1 × 10 10 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 8 to about 1 × 10 10 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 9 to about 1 × 10 10 tissue-derived epithelial stem cells/ml hydrogel, about 1 × 10 5 to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel, or about 1 × 10 5 to about 1 × 10 8 tissue-derived epithelial stem cells/ml hydrogel. In certain embodiments, the plurality of tissue-derived epithelial stem cells includes about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel. In certain embodiments, the plurality of tissue-derived epithelial stem cells includes about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 6 tissue-derived epithelial stem cells/ml hydrogel.
在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,將水凝膠-組織來源之上皮幹細胞混合物懸浮在該培養基中包括將含有水凝膠-組織來源之上皮幹細胞混合物之分配裝置浸沒在培養基中並將水凝膠-組織來源之上皮幹細胞混合物分配到培養基中。在某些實施例中,可以重複將水凝膠-組織來源之上皮幹細胞混合物分配到培養基中,以產生懸浮於培養基中之複數個且經分離之水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,分配裝置可為允許輸送混合物之任何裝置。在某些實施例中,輸送裝置以準確且受控方式分配混合物。分配裝置之非限制性實例包括移液管、滴管及注射器。在某些實施例中,分配裝置可手動操作或自動操作。例如但不限於,分配裝置可自動操作。在某些實施例中,分配裝置可為自動化液體處理器。在某些實施例中,分配裝置可為液體處理機器人。In some embodiments, the method may further include suspending the hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue-derived epithelial stem cell mixture. In some embodiments, suspending the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium includes immersing a dispensing device containing the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium and dispensing the hydrogel-tissue-derived epithelial stem cell mixture into the culture medium. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture can be repeatedly distributed into the culture medium to produce a plurality of separated hydrogel-tissue derived epithelial stem cell mixtures suspended in the culture medium. In some embodiments, the dispensing device can be any device that allows the mixture to be transported. In some embodiments, the transport device distributes the mixture in an accurate and controlled manner. Non-limiting examples of the dispensing device include a pipette, a dropper, and a syringe. In some embodiments, the dispensing device can be manually operated or automatically operated. For example, but not limited to, the dispensing device can be automatically operated. In some embodiments, the dispensing device can be an automated liquid handler. In some embodiments, the dispensing device can be a liquid handling robot.
在某些實施例中,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 1 µl。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可以為至少約 5 µl、至少約 10 µl、至少約 50 µl、至少約 100 µl、至少約 500 µl、至少約 1 ml、至少約 10 ml、至少約 50 ml、至少約 100 ml、至少約 500 ml、至少約 1 L、至少約 1.5 L、至少約 2 L、至少約 5 L 或至少約 10L。在某些實施例中,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為約 1 µl 至約 1 L。在某些實施例中,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為約 1 µl 至約 1 ml。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為約 1 µl 至約 900 µl、約 1 µl 至約 800 µl、約 1 µl 至約 700 µl、約 1 µl 至約 600 µl、約 1 µl 至約 500 µl、約 1 µl 至約 400 µl、約 1 µl 至約 300 µl、約 1 µl 至約 200 µl、約 1 µl 至約 100 µl、約 1 µl 至約 10 µl、約 1 µl 至約 900 µl、約 10 µl 至約 1 ml、約 100 µl 至約 1 ml、約 200 µl 至約 1 ml、約 300 µl 至約 1 ml、約 400 µl 至約 1 ml、約 500 µl 至約 1 ml、約 600 µl 至約 1 ml、約 700 µl 至約 1 ml、約 800 µl 至約 1 ml、約 900 µl 至約 1 ml、約 1 µl 至約 50 µl、約 5 µl 至約 20 µl、約 10 µl 至約 100 µl、約 10 µl 至約 500 µl、約 100 µl 至約 200 µl 或約 100 µl 至約 500 µl。在某些實施例中,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為約 1 µl 至約 100 µl。In certain embodiments, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium can be at least about 1 µl. For example, but not limited to, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium can be at least about 5 µl, at least about 10 µl, at least about 50 µl, at least about 100 µl, at least about 500 µl, at least about 1 ml, at least about 10 ml, at least about 50 ml, at least about 100 ml, at least about 500 ml, at least about 1 L, at least about 1.5 L, at least about 2 L, at least about 5 L, or at least about 10 L. In certain embodiments, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium may be about 1 µl to about 1 L. In certain embodiments, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium may be about 1 µl to about 1 ml. For example, but not limited to, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium may be about 1 µl to about 900 µl, about 1 µl to about 800 µl, about 1 µl to about 700 µl, about 1 µl to about 600 µl, about 1 µl to about 500 µl, about 1 µl to about 400 µl, about 1 µl to about 300 µl, about 1 µl to about 200 µl, about 1 µl to about 100 µl, about 1 µl to about 10 µl, about 1 µl to about 900 µl, about 10 µl to about 1 ml, about 100 µl to about 1 ml, about 200 µl to about 1 ml, or about 100 µl to about 1 ml. ml, about 300 µl to about 1 ml, about 400 µl to about 1 ml, about 500 µl to about 1 ml, about 600 µl to about 1 ml, about 700 µl to about 1 ml, about 800 µl to about 1 ml, about 900 µl to about 1 ml, about 1 µl to about 50 µl, about 5 µl to about 20 µl, about 10 µl to about 100 µl, about 10 µl to about 500 µl, about 100 µl to about 200 µl, or about 100 µl to about 500 µl. In certain embodiments, the volume of the hydrogel-tissue-derived epithelial stem cell mixture dispensed into the culture medium may be about 1 µl to about 100 µl.
在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 10℃ 之溫度固化之材料構成。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 15℃、大於約 20℃、大於約 25℃、大於約 30℃、大於約 35℃、大於約 40℃、大於約 45℃、或大於約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 25℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 30℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 35℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 40℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 45℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在約 25℃ 至約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在約 30℃ 至約 50℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在約 25℃ 至約 40℃, 例如約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在約 30℃ 至約 40℃, 例如約 37℃ 之溫度固化之材料構成。 In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 10°C. For example, but not limited to, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 15°C, greater than about 20°C, greater than about 25°C, greater than about 30°C, greater than about 35°C, greater than about 40°C, greater than about 45°C, or greater than about 50°C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 25°C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that cures at a temperature greater than about 30° C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that cures at a temperature greater than about 35° C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that cures at a temperature greater than about 40° C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that cures at a temperature greater than about 45° C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature of about 25°C to about 50°C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature of about 30°C to about 50°C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature of about 25°C to about 40°C, such as about 37°C. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature of about 30°C to about 40°C, such as about 37°C.
在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 25℃ 或更低之溫度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 20℃ 或更低之溫度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 15℃ 或更低之溫度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 10℃ 或更低之溫度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 5℃ 或更低之溫度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物在接觸培養基之前處於約 25℃、約 24℃、約 23℃、約 22℃、約 21℃、約 20℃、約 19℃、約 18℃、約 17℃、約 16℃、約 15℃、約 14℃、約 13℃、約 12℃、約 11℃、約 10℃、約 9℃、約 8℃、約 7℃、約 6℃、約 5℃、約 4℃、約 3℃ 或約 2℃ 之溫度。在某些實施例中,在接觸培養基之前,水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 25℃。在某些實施例中,在接觸培養基之前,水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 20℃。在某些實施例中,在接觸培養基之前,水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 15℃。在某些實施例中,在接觸培養基之前,水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 10℃。In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 25°C or less before contacting the culture medium. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 20°C or less before contacting the culture medium. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 15°C or less before contacting the culture medium. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 10°C or less before contacting the culture medium. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 5°C or less before contacting the culture medium. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is at a temperature of about 25°C, about 24°C, about 23°C, about 22°C, about 21°C, about 20°C, about 19°C, about 18°C, about 17°C, about 16°C, about 15°C, about 14°C, about 13°C, about 12°C, about 11°C, about 10°C, about 9°C, about 8°C, about 7°C, about 6°C, about 5°C, about 4°C, about 3°C, or about 2°C before contacting the culture medium. In certain embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture before contacting the culture medium is about 2° C. to about 25° C. In certain embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture before contacting the culture medium is about 2° C. to about 20° C. In certain embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture before contacting the culture medium is about 2° C. to about 15° C. In certain embodiments, the temperature of the hydrogel-tissue-derived epithelial stem cell mixture before contacting the culture medium is about 2° C. to about 10° C.
在某些實施例中,水凝膠可包括任何固化之材料。在某些實施例中,水凝膠為合成水凝膠、天然水凝膠或其組合。在某些實施例中,水凝膠為合成水凝膠。在某些實施例中,水凝膠為天然水凝膠。在某些實施例中,水凝膠可為合成水凝膠及天然水凝膠之混合物。在某些實施例中,水凝膠不包括化學交聯之蛋白質及/或聚合物。In some embodiments, the hydrogel may include any solidified material. In some embodiments, the hydrogel is a synthetic hydrogel, a natural hydrogel, or a combination thereof. In some embodiments, the hydrogel is a synthetic hydrogel. In some embodiments, the hydrogel is a natural hydrogel. In some embodiments, the hydrogel may be a mixture of a synthetic hydrogel and a natural hydrogel. In some embodiments, the hydrogel does not include chemically cross-linked proteins and/or polymers.
在某些實施例中,水凝膠為天然水凝膠。在某些實施例中,天然水凝膠包括一種或多種天然存在組分。例如但不限於,天然水凝膠可包括一種或多種蛋白質, 例如醣蛋白及/或多醣。醣蛋白之非限制性實例包括膠原蛋白 ( 例如,I 型膠原蛋白、II 型膠原蛋白、III 型膠原蛋白、IV 型膠原蛋白、V 型膠原蛋白、VI 型膠原蛋白、VII 型膠原蛋白、VIII 型膠原蛋白、IX 型膠原蛋白、X 型膠原蛋白、 XI 型膠原蛋白及/或 XII 型膠原蛋白)、纖維連接蛋白、巢蛋白、腱生蛋白、玻連蛋白、纖網蛋白、透明質酸及層連結蛋白。在某些實施例中,天然水凝膠可進一步包括一種或多種組分,諸如但不限於多醣、水及/或彈性蛋白。在某些實施例中,本揭露之天然水凝膠包括層連結蛋白、巢蛋白及 IV 型膠原蛋白。在某些實施例中,本揭露之天然水凝膠包括層連結蛋白、巢蛋白、IV 型膠原蛋白及硫酸肝素蛋白聚醣。在某些實施例中,天然水凝膠包括由上皮細胞、內皮細胞、壁內胚層樣細胞及/或結締組織細胞分泌及/或來源之細胞外基質 (ECM)。 In some embodiments, the hydrogel is a natural hydrogel. In some embodiments, the natural hydrogel includes one or more naturally occurring components. For example, but not limited to, the natural hydrogel may include one or more proteins, such as glycoproteins and/or polysaccharides. Non-limiting examples of glycoproteins include collagen ( e.g. , type I collagen, type II collagen, type III collagen, type IV collagen, type V collagen, type VI collagen, type VII collagen, type VIII collagen, type IX collagen, type X collagen, type XI collagen and/or type XII collagen), fibronectin, entactin, tenascin, vitronectin, reticular protein, hyaluronic acid and laminin. In certain embodiments, the natural hydrogel may further include one or more components, such as but not limited to polysaccharides, water and/or elastin. In certain embodiments, the natural hydrogel disclosed herein includes laminin, entactin and type IV collagen. In certain embodiments, the natural hydrogel disclosed herein includes laminin, entactin, type IV collagen and heparin sulfate proteoglycans. In certain embodiments, the natural hydrogel includes extracellular matrix (ECM) secreted and/or derived from epithelial cells, endothelial cells, wall endodermal-like cells and/or connective tissue cells.
在某些實施例中,水凝膠為合成水凝膠。合成水凝膠之非限制性實例包括合成聚合物,諸如普羅結合蛋白 (ProNectin) (Sigma Z378666)、聚乙二醇 (PEG)、聚(甲基丙烯酸羥乙酯)、聚(乙烯亞胺)及聚乙烯醇 (PVA)。中揭示了合成水凝膠及此等合成水凝膠之聚合物之額外非限制性實例揭示於 Unal 及 West (2020) Bioconjugate Chem.31(10):2253-2271;以及 Madduma-Bandarage 及 Madihally (2020) J. of Applied Polymer Science 138(19):e50376 中,各篇之內容以引用方式全文併入本文中。In certain embodiments, the hydrogel is a synthetic hydrogel. Non-limiting examples of synthetic hydrogels include synthetic polymers such as ProNectin (Sigma Z378666), polyethylene glycol (PEG), poly(hydroxyethyl methacrylate), poly(ethyleneimine), and polyvinyl alcohol (PVA). Additional non-limiting examples of synthetic hydrogels and polymers of such synthetic hydrogels are disclosed in Unal and West (2020) Bioconjugate Chem. 31(10):2253-2271; and Madduma-Bandarage and Madihally (2020) J. of Applied Polymer Science 138(19):e50376, each of which is incorporated herein by reference in its entirety.
在某些實施例中,本揭露之水凝膠不包括藻酸鹽。In certain embodiments, the hydrogels disclosed herein do not include alginate.
在某些實施例中,水凝膠可為商業可獲得之 ECM。商業可獲得之 ECM 之非限制性實例包括 ECM 蛋白及來自 Engelbreth-Holm-Swarm (EHS) 小鼠肉瘤細胞之基底膜製劑。在某些實施例中,ECM 為 MATRIGEL™ (BD Biosciences),其包括層連結蛋白、巢蛋白及膠原蛋白 IV。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。BME 之非限制性實例為 CULTREX® 基底膜萃取物 2 型 (R&D Systems),包括層連結蛋白、巢蛋白、膠原蛋白 IV 及硫酸肝素蛋白聚醣。In some embodiments, the hydrogel can be a commercially available ECM. Non-limiting examples of commercially available ECM include ECM proteins and basement membrane preparations from Engelbreth-Holm-Swarm (EHS) mouse sarcoma cells. In some embodiments, the ECM is MATRIGEL™ (BD Biosciences), which includes laminin, entactin, and collagen IV. In some embodiments, the ECM is basement membrane extract (BME), which is a soluble form of basement membrane. A non-limiting example of BME is CULTREX® Basement Membrane Extract Type 2 (R&D Systems), which includes laminin, entactin, collagen IV, and heparin sulfate proteoglycans.
在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 0.7 mg/ml 之蛋白質濃度, 例如,醣蛋白濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 1 mg/ml 之蛋白質濃度, 例如,醣蛋白濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 2 mg/ml 之蛋白質濃度, 例如,醣蛋白濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 3 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 4 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 5 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有大於約 6 mg/ml、大於約 7 mg/ml、大於約 8 mg/ml、大於約 9 mg/ml 或大於約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有約 0.7 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有約 1 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有約 5 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有約 6 mg/ml 至約 10 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有不小於 0.7 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有不小於 1 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有不小於 5 mg/ml 之蛋白質濃度。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物之水凝膠具有不小於 6 mg/ml 之蛋白質濃度。 In certain embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration, e.g. , glycoprotein concentration, greater than about 0.7 mg/ml. In certain embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration, e.g. , glycoprotein concentration, greater than about 1 mg/ml. In certain embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration, e.g., glycoprotein concentration, greater than about 2 mg/ml. In certain embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration, e.g. , glycoprotein concentration, greater than about 3 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration greater than about 4 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration greater than about 5 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration greater than about 6 mg/ml, greater than about 7 mg/ml, greater than about 8 mg/ml, greater than about 9 mg/ml, or greater than about 10 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration of about 0.7 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration of about 1 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration of about 5 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture has a protein concentration of about 6 mg/ml to about 10 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration of not less than 0.7 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration of not less than 1 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration of not less than 5 mg/ml. In some embodiments, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture has a protein concentration of not less than 6 mg/ml.
在某些實施例中,水凝膠包含以 w/v % 計大於約 1 w/v % 之 BME 組分、ECM 組分或聚合物。例如但不限於,水凝膠包含以 w/v % 計大於約 1 w/v %、大於約 1.5 w/v %、大於約 2 w/v %、大於約 2.5 w/v %、大於約 3 w/v %、大於約 3.5 w/v %、大於約 4 w/v %、大於約 4.5 w/v %、大於約 5 w/v %、大於約 5.5 w/v %、大於約 6 w/v %、大於約 6.5 w/v %、大於約 7 w/v %、大於約 7.5 w/v %、大於約 8 w/v %、大於約 8.5 w/v %、大於約 9 w/v %、大於約 9.5 w/v %、或大於約 10 w/v %、大於約 10.5 w/v %、大於約 11 w/v %、大於約 11.5 w/v %、大於約 12 w/v %、大於約 12.5 w/v %、大於約 13 w/v %、大於約 13.5 w/v %、大於約 14 w/v %、大於約 14.5 w/v %、大於約 15 w/v %、大於約 15.5 w/v %、大於約 16 w/v %、大於約 16.5 w/v %、大於約 17 w/v %、大於約 17.5 w/v %、大於約 18 w/v %、大於約 18.5 w/v %、大於約 19 w/v %、大於約 19.5 w/v % 或大於約 20 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 1 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 2 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。在某些實施例中,水凝膠包含以 w/v % 計約 5 w/v % 至約 10 w/v % 之 BME 組分、ECM 組分或聚合物。In certain embodiments, the hydrogel comprises greater than about 1 w/v % of a BME component, an ECM component, or a polymer. For example, but not limited to, the hydrogel comprises, in w/v%, greater than about 1 w/v%, greater than about 1.5 w/v%, greater than about 2 w/v%, greater than about 2.5 w/v%, greater than about 3 w/v%, greater than about 3.5 w/v%, greater than about 4 w/v%, greater than about 4.5 w/v%, greater than about 5 w/v%, greater than about 5.5 w/v%, greater than about 6 w/v%, greater than about 6.5 w/v%, greater than about 7 w/v%, greater than about 7.5 w/v%, greater than about 8 w/v%, greater than about 8.5 w/v%, greater than about 9 w/v%, greater than about 9.5 w/v%, or greater than about 10 w/v%, greater than about 10.5 w/v%, greater than about %, greater than about 11 w/v %, greater than about 11.5 w/v %, greater than about 12 w/v %, greater than about 12.5 w/v %, greater than about 13 w/v %, greater than about 13.5 w/v %, greater than about 14 w/v %, greater than about 14.5 w/v %, greater than about 15 w/v %, greater than about 15.5 w/v %, greater than about 16 w/v %, greater than about 16.5 w/v %, greater than about 17 w/v %, greater than about 17.5 w/v %, greater than about 18 w/v %, greater than about 18.5 w/v %, greater than about 19 w/v %, greater than about 19.5 w/v %, or greater than about 20 w/v % of a BME component, an ECM component, or a polymer. In certain embodiments, the hydrogel comprises about 1 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer. In certain embodiments, the hydrogel comprises about 2 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer. In certain embodiments, the hydrogel comprises about 5 w/v % to about 10 w/v % of a BME component, an ECM component, or a polymer.
在某些實施例中,該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。In certain embodiments, the hydrogel has a storage modulus G' that is equal to or greater than the loss modulus G''.
在某些實施例中,水凝膠體積與培養基體積之比率 (體積比) 為約 1:1 至約 1:50。在某些實施例中,水凝膠體積與培養基體積之比率 (體積比) 為約 1:2 至約 1:50。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:5 至約 1:50、約 1:10 至約 1:50、約 1:15 至約 1:50、約 1:20 至約 1:50、約 1:25 至約 1:50、約 1:30 至約 1:50、約 1:35 至約 1:50、約 1:40 至約 1:50、約 1:45 至約 1:50、約 1:2 至約 1:45、約 1:2 至約 1:40、約 1:2 至約 1:35、約 1:2 至約 1:30、約 1:2 至約 1:35、約 1:2 至約 1:30、約 1:2 至約 1:25、約 1:2 至約 1:20、約 1:2 至約 1:15、約 1:2 至約 1:10 或約 1:2 至約 1:5。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:2 至約 1:20。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:2。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:5。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:10, 例如,如實例 8 所示。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:20。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:30。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:40。在某些實施例中,水凝膠體積與培養基體積之比率為約 1:50。 In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium (volume ratio) is about 1:1 to about 1:50. In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium (volume ratio) is about 1:2 to about 1:50. In certain embodiments, the ratio of the volume of hydrogel to the volume of culture medium is about 1:5 to about 1:50, about 1:10 to about 1:50, about 1:15 to about 1:50, about 1:20 to about 1:50, about 1:25 to about 1:50, about 1:30 to about 1:50, about 1:35 to about 1:50, about 1:40 to about 1:50, about 1:45 to about 1:50, about 1:2 to about 1:45, about 1:2 to about 1:40, about 1:2 to about 1:35, about 1:2 to about 1:30, about 1:2 to about 1:35, about 1:2 to about 1:30, about 1:2 to about 1:25, about 1:2 to about 1:20, about 1:2 In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium is about 1:2 to about 1:20. In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium is about 1:2. In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium is about 1:5. In some embodiments, the ratio of the volume of the hydrogel to the volume of the culture medium is about 1:10, for example , as shown in Example 8. In some embodiments, the ratio of hydrogel volume to culture medium volume is about 1:20. In some embodiments, the ratio of hydrogel volume to culture medium volume is about 1:30. In some embodiments, the ratio of hydrogel volume to culture medium volume is about 1:40. In some embodiments, the ratio of hydrogel volume to culture medium volume is about 1:50.
在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。例如但不限於,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含大於約 0.1 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有大於約 0.1 mm 之長度。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有大於約 0.1 mm 之寬度。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有大於約 0.1 mm 之直徑。例如但不限於,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有大於約 0.5 mm、大於約 1 mm、大於約 1.5 mm、大於約 2 mm、大於約 2.5 mm、大於約 3 mm、大於約 3.5 mm、大於約 4 mm、大於約 4.5 mm、大於約 5 mm、大於約 5.5 mm、大於約 6 mm、大於約 6.5 mm、大於約 7.5 mm、大於約 8 mm、大於約 8.5 mm、大於約 9 mm、大於約 9.5 mm、大於約 10 mm、大於約 10.5 mm、大於約 11 mm、大於約 11.5 mm、大於約 12 mm、大於約 12.5 mm、大於約 13 mm、大於約 13.5 mm、大於約 14 mm、大於約 14.5 mm、大於約 15 mm、大於約 15.5 mm、大於約 16 mm、大於約 16.5 mm、大於約 17.5 mm、大於約 18 mm、大於約 18.5 mm、大於約 19 mm、大於約 19.5 mm、大於約 20 mm、大於約 50 mm、大於約 100 mm、大於約 150 mm、大於約 200 mm、大於約 250 mm、大於約 300 mm、大於約 350 mm、大於約 400 mm、大於約 450 mm、大於約 500 mm、大於約 550 mm、大於約 600 mm、大於約 650 mm、大於約 700 mm、大於約 750 mm、大於約 800 mm、大於約 850 mm、大於約 900 mm、大於約 950 mm 或大於約 1,000 mm 之長度、寬度及/或直徑。In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture is dispensed into a culture medium having a certain geometric shape. For example, but not limited to, the geometric shape of the suspended hydrogel-tissue derived epithelial stem cell mixture comprises a length, width, and/or diameter greater than about 0.1 mm. In some embodiments, the geometric shape of the suspended hydrogel-tissue derived epithelial stem cell mixture has a length greater than about 0.1 mm. In some embodiments, the geometric shape of the suspended hydrogel-tissue derived epithelial stem cell mixture has a width greater than about 0.1 mm. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture has a diameter greater than about 0.1 mm. For example, but not limited to, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is greater than about 0.5 mm, greater than about 1 mm, greater than about 1.5 mm, greater than about 2 mm, greater than about 2.5 mm, greater than about 3 mm, greater than about 3.5 mm, greater than about 4 mm, greater than about 4.5 mm, greater than about 5 mm, greater than about 5.5 mm, greater than about 6 mm, greater than about 6.5 mm, greater than about 7.5 mm, greater than about 8 mm, greater than about 8.5 mm, greater than about 9 mm, greater than about 9.5 mm, greater than about 10 mm, greater than about 10.5 mm, greater than about 11 mm, greater than about 11.5 mm, greater than about 12 mm, greater than about 12.5 mm, greater than about 13 mm, greater than about 14 mm, greater than about 15 mm, greater than about 16 mm, greater than about 17 mm, greater than about 18 mm, greater than about 19 mm, greater than about 20 mm, greater than about 21 mm, greater than about 22 mm, greater than about 23 mm, greater than about 24 mm, greater than about 25 mm, greater than about 26 mm, greater than about 27 mm, greater than about 28 mm, greater than about 29 mm, greater than about 30 mm, greater than about 31 mm, greater than about 32 mm, greater than about 33 mm, greater than about 34 mm, greater than about 35 mm, greater than about 36 mm, greater than about 37 mm, greater than about 38 mm, greater than about 39 mm, greater than about 40 mm, greater than about 41 mm, greater than about 13 mm, greater than about 13.5 mm, greater than about 14 mm, greater than about 14.5 mm, greater than about 15 mm, greater than about 15.5 mm, greater than about 16 mm, greater than about 16.5 mm, greater than about 17.5 mm, greater than about 18 mm, greater than about 18.5 mm, greater than about 19 mm, greater than about 19.5 mm, greater than about 20 mm, greater than about 50 mm, greater than about 100 mm, greater than about 150 mm, greater than about 200 mm, greater than about 250 mm, greater than about 300 mm, greater than about 350 mm, greater than about 400 mm, greater than about 450 mm, greater than about 500 mm, greater than about 550 mm, greater than about 600 mm, greater than about 650 mm, greater than about 700 mm mm, greater than about 750 mm, greater than about 800 mm, greater than about 850 mm, greater than about 900 mm, greater than about 950 mm, or greater than about 1,000 mm in length, width, and/or diameter.
在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含約 0.1 mm 至約 1000 mm, 例如,約 0.1 mm 至約 500 mm、約 0.1 mm 至約 100 mm、約 0.1 mm 至約 50 mm、約 0.1 mm 至約 20 mm、約 0.1 mm 至約 10 mm、約 1 mm 至約 1000 mm、約 20 mm 至約 1000 mm、約 50 mm 至約 1000 mm、約 100 mm 至約 1000 mm、約 500 mm 至約 1000 mm、約 1 mm 至約 100 mm 或約 1 mm 至約 50 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀包含約 0.1 mm 至約 4 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有約 0.1 mm 至約 1 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有約 0.1 mm 至約 4 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有約 1 mm 至約 20 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有約 1 mm 至約 10 mm 之長度、寬度及/或直徑。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀具有約 1 mm 至約 4 mm 之長度、寬度及/或直徑。 In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture comprises a length, width, and/or diameter of about 0.1 mm to about 1000 mm, e.g. , about 0.1 mm to about 500 mm, about 0.1 mm to about 100 mm, about 0.1 mm to about 50 mm, about 0.1 mm to about 20 mm, about 0.1 mm to about 10 mm, about 1 mm to about 1000 mm, about 20 mm to about 1000 mm, about 50 mm to about 1000 mm, about 100 mm to about 1000 mm, about 500 mm to about 1000 mm, about 1 mm to about 100 mm, or about 1 mm to about 50 mm. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture comprises about 0.1 mm to about 20 mm, for example , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 1 ... 20 mm, about 15 mm to about 20 mm, about 16 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 21 mm to about 20 mm, about 22 mm to about 20 mm, about 23 mm to about 20 mm, about 24 mm to about 20 mm, about 25 mm to about 20 mm, about 26 mm to about 20 mm, about 27 mm to about 20 mm, about 28 mm to about 20 mm, about 29 mm to about 20 mm, about 30 mm to about 20 mm, about 31 mm to about 20 mm, about 32 mm to about 20 mm, about 33 mm to about 20 mm, about 34 mm to about 20 mm, about 35 mm to about 20 mm, about 36 mm to about 20 mm, about 37 mm to about 20 mm, about 38 mm to about 20 mm, about 39 mm to about 20 mm mm, about 1 mm to about 15 mm, about 1 mm to about 10 mm, about 1 mm to about 5 mm, or about 1 mm to about 4 mm in length, width, and/or diameter. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture comprises a length, width, and/or diameter of about 0.1 mm to about 4 mm. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture has a length, width, and/or diameter of about 0.1 mm to about 1 mm. In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture has a length, width and/or diameter of about 0.1 mm to about 4 mm. In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture has a length, width and/or diameter of about 1 mm to about 20 mm. In some embodiments, the geometric shape of the suspended hydrogel-tissue derived epithelial stem cell mixture has a length, width and/or diameter of about 1 mm to about 10 mm. In certain embodiments, the geometry of the suspended hydrogel-tissue-derived epithelial stem cell mixture has a length, width, and/or diameter of about 1 mm to about 4 mm.
在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,絲狀結構具有線形形狀。在某些實施例中,絲狀結構具有蛇形形狀。在某些實施例中,絲狀結構具有螺旋形形狀。In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the filamentous structure has a linear shape. In some embodiments, the filamentous structure has a serpentine shape. In some embodiments, the filamentous structure has a spiral shape.
在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中 ( 參見例如圖10)。在某些實施例中,水凝膠小滴大於約 0.1 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 4 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 1 mm 之直徑。在某些實施例中,水凝膠小滴具有約 0.1 mm 至約 4 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 20 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 10 mm 之直徑。在某些實施例中,水凝膠小滴具有約 1 mm 至約 4 mm 之直徑。在某些實施例中,各水凝膠小滴包括約 1 個或多個組織來源之上皮幹細胞, 例如,約 5 個或更多個、約 10 個或更多個、約 50 個或更多個、約 100 個或更多個、約 500 個或更多個、約 1,000 個或更多個、約 5,000 個或更多個、約 10,000 個或更多個、約 100,000 個或更多個、約 200,000 個或更多個、約 300,000 個或更多個、約 400,000 個或更多個、約 500,000 個或更多個、約 600,000 個或更多個、約 700,000 個或更多個、約 800,000 個或更多個、約 900,000 個或更多個、約 1,000,000 個或更多個、約 2,000,000 個或更多個、約 3,000,000 個或更多個、約 4,000,000 個或更多個、約 5,000,000 個或更多個、約 6,000,000 個或更多個、約 7,000,000 個或更多個、約 8,000,000 個或更多個、約 9,000,000 個或更多個、約 10,000,000 個或更多個、約 100,000,000 個或更多個、或約 1,000,000,000 個或更多個組織來源之上皮幹細胞。 In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is suspended in a culture medium as a droplet ( see, e.g., FIG. 10 ). In some embodiments, the hydrogel droplet is larger than about 0.1 mm in diameter. In certain embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 20 mm, e.g. , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 4 mm. In some embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 1 mm. In some embodiments, the hydrogel droplets have a diameter of about 0.1 mm to about 4 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 20 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 10 mm. In some embodiments, the hydrogel droplets have a diameter of about 1 mm to about 4 mm. In certain embodiments, each hydrogel droplet comprises about 1 or more tissue-derived epithelial stem cells, e.g. , about 5 or more, about 10 or more, about 50 or more, about 100 or more, about 500 or more, about 1,000 or more, about 5,000 or more, about 10,000 or more, about 100,000 or more, about 200,000 or more, about 300,000 or more, about 400,000 or more, about 500,000 or more, about 600,000 or more, about 700,000 or more, about 800,000 or more, about 900,000 or more, about 1,000,000 or more, or about 2,000,000 or more. In some embodiments, the invention relates to epithelial stem cells of at least one tissue-derived epithelial cell. The epithelial stem cells may be about 1,000,000 or more, about 2,000,000 or more, about 3,000,000 or more, about 4,000,000 or more, about 5,000,000 or more, about 6,000,000 or more, about 7,000,000 or more, about 8,000,000 or more, about 9,000,000 or more, about 10,000,000 or more, about 100,000,000 or more, or about 1,000,000,000 or more tissue-derived epithelial stem cells.
在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有絲狀結構 ( 參見例如圖11)。在某些實施例中,絲狀結構具有大於約 0.1 mm 之長度及/或寬度。在某些實施例中,絲狀結構具有大於約 0.1 mm 之長度。在某些實施例中,絲狀結構具有大於約 0.1 mm 之寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1000 mm, 例如,約 0.1 mm 至約 500 mm、約 0.1 mm 至約 100 mm、約 0.1 mm 至約 50 mm、約 0.1 mm 至約 20 mm、約 0.1 mm 至約 10 mm、約 1 mm 至約 1000 mm、約 20 mm 至約 1000 mm、約 50 mm 至約 1000 mm、約 100 mm 至約 1000 mm、約 500 mm 至約 1000 mm、約 1 mm 至約 100 mm 或約 1 mm 至約 50 mm 之長度及/或寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之長度及/或寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1 mm 之長度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 4 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之長度。在某些實施例中,絲狀結構具有約 1 mm 至約 4 mm 之長度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 1 mm 之寬度。在某些實施例中,絲狀結構具有約 0.1 mm 至約 4 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之寬度。在某些實施例中,絲狀結構具有約 1 mm 至約 4 mm 之寬度。 In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture has a filamentous structure ( see, e.g., FIG. 11 ). In some embodiments, the filamentous structure has a length and/or width greater than about 0.1 mm. In some embodiments, the filamentous structure has a length greater than about 0.1 mm. In some embodiments, the filamentous structure has a width greater than about 0.1 mm. In certain embodiments, the filamentous structure has a length and/or width of about 0.1 mm to about 1000 mm, e.g. , about 0.1 mm to about 500 mm, about 0.1 mm to about 100 mm, about 0.1 mm to about 50 mm, about 0.1 mm to about 20 mm, about 0.1 mm to about 10 mm, about 1 mm to about 1000 mm, about 20 mm to about 1000 mm, about 50 mm to about 1000 mm, about 100 mm to about 1000 mm, about 500 mm to about 1000 mm, about 1 mm to about 100 mm, or about 1 mm to about 50 mm. In certain embodiments, the filamentous structure has a diameter of about 0.1 mm to about 20 mm, for example , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the filamentous structure has a length and/or width of about 0.1 mm to about 4 mm. In some embodiments, the filamentous structure has a length of about 0.1 mm to about 1 mm. In some embodiments, the filamentous structure has a length of about 0.1 mm to about 4 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 20 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 10 mm. In some embodiments, the filamentous structure has a length of about 1 mm to about 4 mm. In some embodiments, the filamentous structure has a width of about 0.1 mm to about 1 mm. In some embodiments, the filamentous structure has a width of about 0.1 mm to about 4 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 20 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 10 mm. In some embodiments, the filamentary structure has a width of about 1 mm to about 4 mm.
在某些實施例中,絲狀結構具有約 0.1 mm至約 20 mm, 例如,約 0.1 mm 至約 19 mm、約 0.1 mm 至約 18 mm、約 0.1 mm 至約 17 mm、約 0.1 mm 至約 16 mm、約 0.1 mm 至約 15 mm、約 0.1 mm 至約 14 mm、約 0.1 mm 至約 13 mm、約 0.1 mm 至約 12 mm、約 0.1 mm 至約 11 mm、約 0.1 mm 至約 10 mm、約 0.1 mm 至約 9 mm、約 0.1 mm 至約 8 mm、約 0.1 mm 至約 7 mm、約 0.1 mm 至約 6 mm、約 0.1 mm 至約 5 mm、約 0.1 mm 至約 4 mm、約 0.1 mm 至約 3 mm、約 0.1 mm 至約 2 mm、約 0.1 mm 至約 1 mm、約 0.5 mm 至約 20 mm、約 1 mm 至約 20 mm、約 2 mm 至約 20 mm、約 3 mm 至約 20 mm、約 4 mm 至約 20 mm、約 5 mm 至約 20 mm、約 6 mm 至約 20 mm、約 7 mm 至約 20 mm、約 8 mm 至約 20 mm、約 9 mm 至約 20 mm、約 10 mm 至約 20 mm、約 11 mm 至約 20 mm、約 12 mm 至約 20 mm、約 13 mm 至約 20 mm、約 14 mm 至約 20 mm、約 15 mm 至約 20 mm、約 16 mm 至約 20 mm、約 17 mm 至約 20 mm、約 18 mm 至約 20 mm、約 19 mm 至約 20 mm、約 1 mm 至約 15 mm、約 1 mm 至約 10 mm、約 1 mm 至約 5 mm 或約 1 mm 至約 4 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 20 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 10 mm 之直徑。在某些實施例中,絲狀結構具有約 0.1 mm 至約 5 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 20 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 10 mm 之直徑。在某些實施例中,絲狀結構具有約 1 mm 至約 5 mm 之直徑, 例如,如實例 8 所揭示的。 In certain embodiments, the filamentous structure has a diameter of about 0.1 mm to about 20 mm, for example , about 0.1 mm to about 19 mm, about 0.1 mm to about 18 mm, about 0.1 mm to about 17 mm, about 0.1 mm to about 16 mm, about 0.1 mm to about 15 mm, about 0.1 mm to about 14 mm, about 0.1 mm to about 13 mm, about 0.1 mm to about 12 mm, about 0.1 mm to about 11 mm, about 0.1 mm to about 10 mm, about 0.1 mm to about 9 mm, about 0.1 mm to about 8 mm, about 0.1 mm to about 7 mm, about 0.1 mm to about 6 mm, about 0.1 mm to about 5 mm, about 0.1 mm to about 4 mm, about 0.1 mm to about 3 mm, about 0.1 mm to about 2 mm, about 0.1 mm to about 1 20 mm, about 1 mm to about 20 mm, about 17 mm to about 20 mm, about 18 mm to about 20 mm, about 19 mm to about 20 mm, about 1 mm to about 15 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, about 1 mm to about 20 mm, or about 1 mm to about 15 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 4 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 20 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 10 mm. In some embodiments, the filamentary structure has a diameter of about 0.1 mm to about 5 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 20 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 10 mm. In some embodiments, the filamentary structure has a diameter of about 1 mm to about 5 mm, for example , as disclosed in Example 8.
在某些實施例中,各絲狀結構包括約 1 個或更多個組織來源之上皮幹細胞, 例如,約 5 個或更多個、約 10 個或更多個、約 50 個或更多個、約 100 個或更多個、約 500 個或更多個、約 1,000 個或更多個、約 5,000 個或更多個、約 10,000 個或更多個、或約 100,000 個或更多個、約 200,000 個或更多個、約 300,000 個或更多個、約 400,000 個或更多個、約 500,000 個或更多個、約 600,000 個或更多個、約 700,000 個或更多個、約 800,000 個或更多個、約 900,000 個或更多個、或約 1,000,000 個或更多個、約 2,000,000 個或更多個、約 3,000,000 個或更多個、約 4,000,000 個或更多個、約 5,000,000 個或更多個、約 6,000,000 個或更多個、約 7,000,000 個或更多個、約 8,000,000 個或更多個、約 9,000,000 個或更多個、約 10,000,000 個或更多個、約 100,000,000 個或更多個、約 1,000,000,000 個或更多個、或約 10,000,000,000 個或更多個組織來源之上皮幹細胞。 In certain embodiments, each filamentous structure comprises about 1 or more tissue-derived epithelial stem cells, for example , about 5 or more, about 10 or more, about 50 or more, about 100 or more, about 500 or more, about 1,000 or more, about 5,000 or more, about 10,000 or more, or about 100,000 or more, about 200,000 or more, about 300,000 or more, about 400,000 or more, about 500,000 or more, about 600,000 or more, about 700,000 or more, about 800,000 or more, about 900,000 or more, or about 1,000,000 or more. In some embodiments, the invention relates to epithelial stem cells of at least one tissue-derived epithelial cell. The epithelial stem cells may be about 1,000,000 or more, about 2,000,000 or more, about 3,000,000 or more, about 4,000,000 or more, about 5,000,000 or more, about 6,000,000 or more, about 7,000,000 or more, about 8,000,000 or more, about 9,000,000 or more, about 10,000,000 or more, about 100,000,000 or more, about 1,000,000,000 or more, or about 10,000,000,000 or more tissue-derived epithelial stem cells.
在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在培養基中培養以產生組織來源之上皮類器官。在某些實施例中,細胞培養基含有對於支持組織來源之上皮幹細胞及/或類器官之維持而言重要的組分。 在某些實施例中,使用於本揭露之細胞培養基可為營養液,其包括標準細胞培養成分,諸如但不限於胺基酸、維生素、無機鹽、碳能源 ( 例如,葡萄糖) 及緩衝液。在某些實施例中,該培養基為幹細胞促進培養基。在某些實施例中,該培養基為細胞生長培養基。在某些實施例中,該培養基為分化培養基。已知支持特定組織及細胞類型生長之培養基為本技術領域已知的並且可與本文所揭示之主題結合使用。例如但不限於,可在本揭露中使用之培養基之實例在 Calà 等人, Front.Bioeng.Biotechnol.11:1058970 (2023) ( 例如,Calà 等人之表 1) 中揭示,其內容特此以引用方式併入。在實例中, 例如實例 8 中,提供了使用於本揭露之培養基之額外非限制性實例。 In some embodiments, the method may further include culturing the tissue-derived epithelial stem cells of the suspended hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to produce tissue-derived epithelial organoids. In some embodiments, the cell culture medium contains components important for supporting the maintenance of tissue-derived epithelial stem cells and/or organoids. In some embodiments, the cell culture medium used in the present disclosure may be a nutrient solution, which includes standard cell culture ingredients, such as but not limited to amino acids, vitamins, inorganic salts, carbon energy ( e.g. , glucose) and buffer. In some embodiments, the culture medium is a stem cell promotion medium. In some embodiments, the medium is a cell growth medium. In some embodiments, the medium is a differentiation medium. Mediums known to support the growth of specific tissues and cell types are known in the art and can be used in conjunction with the subject matter disclosed herein. For example, but not limited to, examples of mediums that can be used in the present disclosure are disclosed in Calà et al., Front.Bioeng.Biotechnol.11:1058970 (2023) ( e.g. , Table 1 of Calà et al.), the contents of which are hereby incorporated by reference. In examples, such as Example 8, additional non-limiting examples of mediums used in the present disclosure are provided.
在某些實施例中,該培養基存在於容器中。容器之非限制性實例包括培養皿、多孔盤、錐形管、貯器、培養袋、生物反應器或燒瓶。在某些實施例中,錐形管為 50 ml 錐形管。在某些實施例中,多孔盤為 6 孔盤、12 孔盤、24 孔盤、48 孔盤、96 孔盤或 384 孔盤。在某些實施例中,貯器為自訂貯器。在某些實施例中,燒瓶為 25 ml 燒瓶、50 ml 燒瓶、250 ml 燒瓶或 600 ml 燒瓶。在某些實施例中,燒瓶為單一燒瓶或酒店燒瓶。在某些實施例中,容器由使組織來源之上皮幹細胞及/或水凝膠與容器表面之接附最小化之材料構成。可替代地或另外,容器可包括表面塗層,其使組織來源之上皮幹細胞及/或水凝膠與容器表面之接附最小化。In some embodiments, the medium is present in a container. Non-limiting examples of containers include culture dishes, multi-well plates, conical tubes, containers, culture bags, bioreactors, or flasks. In some embodiments, the conical tube is a 50 ml conical tube. In some embodiments, the multi-well plate is a 6-well plate, a 12-well plate, a 24-well plate, a 48-well plate, a 96-well plate, or a 384-well plate. In some embodiments, the container is a custom container. In some embodiments, the flask is a 25 ml flask, a 50 ml flask, a 250 ml flask, or a 600 ml flask. In some embodiments, the flask is a single flask or a hotel flask. In some embodiments, the container is composed of a material that minimizes the attachment of tissue-derived epithelial stem cells and/or hydrogel to the container surface. Alternatively or in addition, the container may include a surface coating that minimizes the attachment of tissue-derived epithelial stem cells and/or hydrogel to the container surface.
在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,與最初擠出之水凝膠-組織來源之上皮幹細胞混合物 ( 例如絲狀結構) 相比,將絲狀結構碎片化產生長度、寬度或二者短至少約 10%、至少約 15%、至少約 20%、至少約 25%、至少約 30%、至少約 35%、至少約 40%、至少約 45%、至少約 50%、至少約 55%、至少約 60%、至少約 65%、至少約 70%、至少約 75%、至少約 80%、至少約 85% 或至少約 90% 之結構。 In some embodiments, the method may further include fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures comprising the tissue-derived epithelial organoid. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce the fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting the filamentous structure produces a structure that is shorter in length, width, or both. In certain embodiments, the filamentous structure is fragmented to produce a structure that is at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, or at least about 90% shorter in length, width, or both than the hydrogel-tissue-derived epithelial stem cell mixture (e.g., filamentous structure) that was initially extruded.
目前所揭示之主題進一步提供了用於產生組織來源之上皮類器官之懸浮培養物之方法。在某些實施例中,該方法可包括將包含水凝膠及組織來源之上皮幹細胞之混合物引入到培養基中以產生經懸浮之混合物。在某些實施例中,引入到該培養基中之該混合物包含如本文所述之水凝膠及複數個組織來源之上皮幹細胞。在某些實施例中,該方法可進一步包括將混合物中之複數個組織來源之上皮幹細胞在培養基中培養以產生如本文所述之呈懸浮狀態之組織來源之上皮類器官。在某些實施例中,該方法可進一步包括將經懸浮之混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。The presently disclosed subject matter further provides methods for producing suspended cultures of tissue-derived epithelial organoids. In certain embodiments, the method may include introducing a mixture comprising a hydrogel and tissue-derived epithelial stem cells into a culture medium to produce a suspended mixture. In certain embodiments, the mixture introduced into the culture medium comprises a hydrogel as described herein and a plurality of tissue-derived epithelial stem cells. In certain embodiments, the method may further include culturing a plurality of tissue-derived epithelial stem cells in the mixture in a culture medium to produce a suspended tissue-derived epithelial organoid as described herein. In certain embodiments, the method may further comprise fragmenting the suspended mixture to produce a fragmented structure comprising the tissue-derived epithelial organoid.
在某些實施例中,用於產生組織來源之上皮類器官之懸浮培養物之方法可包括使胃腸幹細胞 ( 例如,複數個組織來源之上皮幹細胞) 與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物並將水凝膠-組織來源之上皮幹細胞混合物沉積到基材上。在某些實施例中,將該水凝膠-組織來源之上皮幹細胞混合物作為小滴沉積到該基材上。在某些實施例中,將該水凝膠-組織來源之上皮幹細胞混合物沉積到該基材上以具有絲狀結構。在某些實施例中,此方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物固化以產生經固化之水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該方法可包括將經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該方法包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。在某些實施例中,該方法可以進一步包括在將經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中之前,將經固化之水凝膠-組織來源之上皮幹細胞混合物從基材中取出。在某些實施例中,該方法可以進一步包括在將經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中之前或之後,將經固化之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含組織來源之上皮類器官之碎片化結構。 In certain embodiments, a method for generating a suspension culture of tissue-derived epithelial organoids may include contacting gastrointestinal stem cells ( e.g. , a plurality of tissue-derived epithelial stem cells) with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture and depositing the hydrogel-tissue-derived epithelial stem cell mixture onto a substrate. In certain embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is deposited onto the substrate as a droplet. In certain embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is deposited onto the substrate to have a filamentous structure. In some embodiments, the method may further include solidifying the hydrogel-tissue derived epithelial stem cell mixture to produce a solidified hydrogel-tissue derived epithelial stem cell mixture. In some embodiments, the method may include suspending the solidified hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. In some embodiments, the method includes culturing the suspended hydrogel-tissue derived epithelial stem cell mixture in the culture medium to produce tissue derived epithelial organoids. In some embodiments, the method may further include removing the solidified hydrogel-tissue-derived epithelial stem cell mixture from the substrate before suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in the culture medium. In some embodiments, the method may further include fragmenting the solidified hydrogel-tissue-derived epithelial stem cell mixture to generate a fragmented structure comprising tissue-derived epithelial organoids before or after suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in the culture medium.
在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在組織碎片、類器官碎片或其組合內。例如但不限於,包括組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞之組織碎片及/或類器官碎片可用於產生水凝膠-組織來源之上皮幹細胞混合物。可替代地或另外,組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞分離自組織 ( 例如,組織碎片)、類器官碎片 ( 例如,組織來源之上皮類器官碎片) 或其組合。在某些實施例中,組織來源之上皮幹細胞不包括富潛能幹細胞 ( 例如,誘導富潛能幹細胞 (iPSC) 及胚胎幹細胞 (ESC))。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞可獲自 活體外細胞培養物。在某些實施例中,組織碎片可獲自冷凍樣本或新鮮樣本, 例如冷凍或新鮮組織樣本及/或冷凍或新鮮組織類器官碎片。在某些實施例中,組織碎片可為原生組織碎片。在某些實施例中, 例如個體之組織 ( 例如組織碎片) 可係正常的 ( 例如非癌性的及/或非患病的)。在某些實施例中, 例如個體之組織 ( 例如組織碎片) 可係異常的 ( 例如癌性的及/或患病的)。在某些實施例中,組織來源之上皮幹細胞可分離自原生組織之碎片。 In certain embodiments, the tissue-derived epithelial stem cells or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained in tissue fragments, organoid fragments, or a combination thereof. For example, but not limited to, tissue fragments and/or organoid fragments including tissue-derived epithelial stem cells or a plurality of tissue-derived epithelial stem cells can be used to generate a hydrogel-tissue-derived epithelial stem cell mixture. Alternatively or additionally, the tissue-derived epithelial stem cells or a plurality of tissue-derived epithelial stem cells are isolated from a tissue ( e.g. , a tissue fragment), an organoid fragment ( e.g. , a tissue-derived epithelial organoid fragment), or a combination thereof. In some embodiments, the tissue-derived epithelial stem cells do not include enriched potential stem cells ( e.g. , induced enriched potential stem cells (iPSCs) and embryonic stem cells (ESCs)). In some embodiments, the tissue-derived epithelial stem cells used in the present disclosure may be obtained from an in vitro cell culture. In some embodiments, the tissue fragments may be obtained from a frozen sample or a fresh sample, such as a frozen or fresh tissue sample and/or a frozen or fresh tissue organoid fragment. In some embodiments, the tissue fragments may be primary tissue fragments. In some embodiments, for example , the tissue of an individual ( e.g., a tissue fragment) may be normal ( e.g., non-cancerous and/or non-disease). In certain embodiments, a tissue ( eg , a tissue fragment) from a subject may be abnormal ( eg, cancerous and/or diseased). In certain embodiments, tissue-derived epithelial stem cells may be isolated from a fragment of native tissue.
在某些實施例中,原生組織碎片可為淚腺、扁桃腺、唾液腺、胃腸組織、甲狀腺、肺、乳腺、肝、膽管、胃、腎、胰臟、子宮內膜、輸卵管、子宮頸、前列腺、膀胱、卵巢、味蕾或胎盤之碎片。在某些實施例中,組織來源之上皮幹細胞可分離自選自以下之組織 (或其碎片):淚腺、扁桃腺、唾液腺、胃腸組織、甲狀腺、肺、乳腺、肝、膽管、胃、腎、胰臟、子宮內膜、輸卵管、子宮頸、前列腺、膀胱、卵巢、味蕾、胎盤及其組合。在某些實施例中,原生組織碎片可為淚腺之碎片。在某些實施例中,原生組織碎片可為扁桃腺之碎片。在某些實施例中,原生組織碎片可為唾液腺之碎片。在某些實施例中,原生組織碎片可為胃腸組織之碎片。在某些實施例中,原發組織碎片可為甲狀腺之碎片。在某些實施例中,原生組織碎片可為肺之碎片。在某些實施例中,原生組織碎片可為乳腺之碎片。在某些實施例中,原生組織碎片可為肝之碎片。在某些實施例中,原生組織碎片可為膽管之碎片。在某些實施例中,主要組織碎片可為胃之碎片。在某些實施例中,原生組織碎片可為腎之碎片。在某些實施例中,原生組織碎片可為胰臟之碎片。在某些實施例中,原生組織碎片可為子宮內膜之碎片。在某些實施例中,原生組織碎片可為輸卵管之碎片。在某些實施例中,原生組織碎片可為子宮頸之碎片。在某些實施例中,原生組織碎片可為前列腺之碎片。在某些實施例中,原生組織碎片可為膀胱之碎片。 在某些實施例中,原生組織碎片可為卵巢之碎片。在某些實施例中,初級組織碎片可為味蕾。在某些實施例中,原生組織碎片可為胎盤之碎片。在某些實施例中,原生組織碎片選自由以下所組成之群組:肺組織碎片、肝組織碎片、胃腸組織碎片、乳腺組織碎片、胰臟組織碎片及其組合。In some embodiments, the primary tissue fragments can be fragments of tear glands, tonsils, salivary glands, gastrointestinal tissue, thyroid, lung, breast, liver, bile duct, stomach, kidney, pancreas, endometrium, fallopian tube, cervix, prostate, bladder, ovary, taste buds or placenta. In some embodiments, tissue-derived epithelial stem cells can be isolated from tissues (or fragments thereof) selected from the following: tear glands, tonsils, salivary glands, gastrointestinal tissue, thyroid, lung, breast, liver, bile duct, stomach, kidney, pancreas, endometrium, fallopian tube, cervix, prostate, bladder, ovary, taste buds, placenta and combinations thereof. In some embodiments, the primary tissue fragments may be fragments of lacrimal glands. In some embodiments, the primary tissue fragments may be fragments of tonsils. In some embodiments, the primary tissue fragments may be fragments of salivary glands. In some embodiments, the primary tissue fragments may be fragments of gastrointestinal tissue. In some embodiments, the primary tissue fragments may be fragments of thyroid glands. In some embodiments, the primary tissue fragments may be fragments of lungs. In some embodiments, the primary tissue fragments may be fragments of breasts. In some embodiments, the primary tissue fragments may be fragments of liver. In some embodiments, the primary tissue fragments may be fragments of bile ducts. In some embodiments, the primary tissue fragments may be fragments of stomach. In some embodiments, the primary tissue fragments may be fragments of kidneys. In some embodiments, the native tissue fragments may be fragments of the pancreas. In some embodiments, the native tissue fragments may be fragments of the endometrium. In some embodiments, the native tissue fragments may be fragments of the fallopian tube. In some embodiments, the native tissue fragments may be fragments of the cervix. In some embodiments, the native tissue fragments may be fragments of the prostate. In some embodiments, the native tissue fragments may be fragments of the bladder. In some embodiments, the native tissue fragments may be fragments of the ovary. In some embodiments, the primary tissue fragments may be taste buds. In some embodiments, the native tissue fragments may be fragments of the placenta. In certain embodiments, the primary tissue fragments are selected from the group consisting of lung tissue fragments, liver tissue fragments, gastrointestinal tissue fragments, breast tissue fragments, pancreatic tissue fragments, and combinations thereof.
在某些實施例中,原生組織碎片可為胃腸組織之碎片。在某些實施例中,組織碎片可為 例如個體之口腔黏膜、咽 (喉)、食道、胃、小腸、大腸及/或直腸之碎片。在某些實施例中,組織碎片可為 例如個體之口腔黏膜之碎片。在某些實施例中,組織碎片可為 例如個體之咽之碎片。在某些實施例中,組織碎片可為 例如個體之食道之碎片。在某些實施例中,組織碎片可為 例如個體之胃之碎片。在某些實施例中,組織碎片可為 例如個體之直腸之碎片。在某些實施例中,組織碎片可為 例如個體之小腸之碎片。在某些實施例中,組織碎片可為 例如個體之大腸之碎片。在某些實施例中,組織碎片可為 例如個體之大腸及/或迴腸組織之碎片。例如但不限於,組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞可分離自 例如個體之大腸及/或迴腸組織分離。在某些實施例中,組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞可分離自 ( 例如個體之) 大腸組織, 例如以產生大腸類器官。在某些實施例中,組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞可分離自 ( 例如個體之) 迴腸組織, 例如以產生迴腸類器官。在某些實施例中, 例如個體之組織可為正常的 ( 即,非癌性的)。例如但不限於, 例如個體之大腸及/或迴腸組織可為正常的 ( 即,非癌性的)。在某些實施例中, 例如個體之組織可為癌性的或患病的。例如但不限於, 例如個體之大腸及/或迴腸組織可為癌性的及/或患病的大腸及/或迴腸組織。在某些實施例中, 例如個體之食道組織可為癌性的及/或患病的食道組織。在某些實施例中, 例如個體之胃組織可為癌性的及/或患病的胃組織。在某些實施例中, 例如個體之直腸組織可為癌性的及/或患病的直腸組織。在某些實施例中,組織來源之上皮類器官可為個體來源之胃腸腫瘤類器官。 In certain embodiments, the primary tissue fragments may be fragments of gastrointestinal tissue. In certain embodiments, the tissue fragments may be fragments of, for example , the oral mucosa, pharynx (throat), esophagus, stomach, small intestine, large intestine and/or rectum of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the oral mucosa of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the pharynx of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the esophagus of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the stomach of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the rectum of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the small intestine of an individual. In certain embodiments, the tissue fragments may be fragments of, for example, the large intestine of an individual. In some embodiments, the tissue fragments can be fragments of colon and/or ileum tissue, for example , of an individual. For example, but not limited to, tissue-derived epithelial stem cells or a plurality of tissue-derived epithelial stem cells can be isolated from colon and/or ileum tissue, for example, of an individual. In some embodiments, tissue-derived epithelial stem cells or a plurality of tissue-derived epithelial stem cells can be isolated from colon tissue ( for example, of an individual), for example, to generate colon organoids. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells may be isolated from ileal tissue ( e.g., of an individual), e.g., to generate ileal organoids. In certain embodiments, the tissue of , e.g., an individual may be normal ( i.e. , non-cancerous). For example, but not limited to , the colon and/or ileal tissue of, e.g., an individual may be normal ( i.e. , non-cancerous). In certain embodiments, the tissue of, e.g. , an individual may be cancerous or diseased. For example, but not limited to, the colon and/or ileal tissue of , e.g., an individual may be cancerous and/or diseased colon and/or ileal tissue. In some embodiments, for example , the esophageal tissue of an individual may be cancerous and/or diseased esophageal tissue. In some embodiments, for example , the stomach tissue of an individual may be cancerous and/or diseased stomach tissue. In some embodiments, for example , the rectal tissue of an individual may be cancerous and/or diseased rectal tissue. In some embodiments, the tissue-derived epithelial organoid may be a gastrointestinal tumor organoid derived from an individual.
在某些實施例中,原生組織碎片為肺組織碎片。In certain embodiments, the native tissue fragments are lung tissue fragments.
在某些實施例中,原生組織碎片為肝組織碎片。In certain embodiments, the native tissue fragments are liver tissue fragments.
在某些實施例中,原生組織碎片為乳腺組織碎片。In certain embodiments, the native tissue fragment is a breast tissue fragment.
在某些實施例中,原生組織碎片為胰臟組織碎片。In certain embodiments, the native tissue fragments are pancreatic tissue fragments.
在某些實施例中,本揭露之方法產生與參考組織來源之上皮類器官 ( 例如,嵌入接附於基材之水凝膠內之組織來源之上皮類器官) 相比尺寸均勻之組織來源之上皮類器官。在某些實施例中,與參考組織來源之上皮類器官相比藉由本揭露之方法產生之組織來源之上皮類器官由於如實例 1 中所述之營養素利用率差異而在尺寸上更均勻。例如但不限於,與參考組織來源之上皮類器官 ( 例如,嵌入接附於基材之水凝膠內之組織來源之上皮類器官) 相比,藉由本揭露之方法產生之組織來源之上皮類器官在尺寸上在懸浮培養物小滴 ( 例如,經懸浮之水凝膠小滴) 之寬度上更均勻。在某些實施例中,參考組織來源之上皮類器官在如實例 1 所揭示之水凝膠圓頂中產生。 In certain embodiments, the methods of the present disclosure produce tissue-derived epithelial organoids that are uniform in size compared to a reference tissue-derived epithelial organoid ( e.g. , a tissue-derived epithelial organoid embedded in a hydrogel attached to a substrate). In certain embodiments, the tissue-derived epithelial organoids produced by the methods of the present disclosure are more uniform in size compared to the reference tissue-derived epithelial organoids due to differences in nutrient utilization as described in Example 1. For example, but not limited to, tissue-derived epithelial organoids produced by the methods of the present disclosure are more uniform in size across the width of a suspended culture droplet ( e.g. , a suspended hydrogel droplet) than a reference tissue-derived epithelial organoid ( e.g. , a tissue-derived epithelial organoid embedded in a hydrogel attached to a substrate). In certain embodiments, the reference tissue-derived epithelial organoid is produced in a hydrogel dome as disclosed in Example 1.
在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。例如但不限於,與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比,本揭露之組織來源之上皮類器官 ( 例如,組織來源之上皮類器官之群體) 以較高含量表現標記物。可替代地或另外,在某些實施例中,與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比,本揭露之組織來源之上皮類器官 ( 例如,組織來源之上皮類器官之群體) 以較低之含量表現標記物。在某些實施例中,標記物為幹細胞及/或增生標記物, 例如,與幹細胞及/或增生相關之基因。 例如但不限於,幹細胞及/或增生標記物為 MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1 及/或 CD44。在某些實施例中,標記物為分化標記物, 例如,與分化相關之基因。例如但不限於,分化標記物為角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、及/或平滑肌肌動蛋白 (SMA)。在某些實施例中,該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。例如但不限於,參考組織來源之上皮類器官為在如實例 1 中所揭示之水凝膠圓頂中產生之組織來源之上皮類器官。 In certain embodiments, the methods of the present disclosure generate a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). For example, but not limited to, a tissue-derived epithelial organoid ( e.g. , a population of tissue-derived epithelial organoids) of the present disclosure expresses a marker at a higher level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). Alternatively or additionally, in certain embodiments, the tissue-derived epithelial organoids ( e.g. , a population of tissue-derived epithelial organoids) of the present disclosure express a marker at a lower level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In certain embodiments, the marker is a stem cell and/or proliferation marker, e.g. , a gene associated with stem cells and/or proliferation. For example, but not limited to, the stem cell and/or proliferation marker is MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1 and/or CD44. In some embodiments, the marker is a differentiation marker, for example , a gene associated with differentiation. For example, but not limited to, the differentiation marker is keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), and/or smooth muscle actin (SMA). In some embodiments, the reference tissue-derived epithelial organoids are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. For example, but not limited to, the reference tissue-derived epithelial organoid is a tissue-derived epithelial organoid produced in a hydrogel dome as disclosed in Example 1.
在某些實施例中,本揭露之方法產生以與參考組織來源之上皮類器官之群體相比較高的含量表現幹細胞及/或增生標記物之組織來源之上皮類器官之群體。幹細胞及/或增生標記物之非限制性實例包括 MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、及/或 CD44 及其組合。在某些實施例中,幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、ASCL2、LGR5、SOX9、SMOC2、CD44 及其組合。在某些實施例中,幹細胞及/或增生標記物為 MKI67。在某些實施例中,幹細胞及/或增生標記物為 ASCL2。在某些實施例中,幹細胞及/或增生標記物為 LGR5。在某些實施例中,幹細胞及/或增生標記物為 SOX9。在某些實施例中,幹細胞及/或增生標記物為 SMOC2。在某些實施例中,幹細胞及/或增生標記物為 CD44。在某些實施例中,幹細胞及/或增生標記物為 EpCAM。在某些實施例中,幹細胞及/或增生標記物為 CD49f。在某些實施例中,幹細胞及/或增生標記物為 CD133。在某些實施例中,幹細胞及/或增生標記物為 ALDH1A1。在某些實施例中,幹細胞及/或增生標記物為 NEUROG3。在某些實施例中,幹細胞及/或增生標記物為 NKX6.1。在某些實施例中,幹細胞及/或增生標記物為 PDX1。在某些實施例中,幹細胞及/或增生標記物為 BMI1。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 10%、高至少 20%、高至少 30%、高至少 40%、高至少 50%、高至少 60%、高至少 70%、高至少 80%、高至少 90%、高至少 100%、高至少 110%、高至少 120%、高至少 130%、高至少 140%、高至少 150%、高至少 160%、高至少 170%、高至少 180%、高至少 190%、高至少 200%、高至少 210%、高至少 220%、高至少 230%、高至少 240%、高至少 250%、高至少 260%、高至少 270%、高至少 280%、高至少 290% 或高至少 300%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 50%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 100%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 200%。在某些實施例中,與參考組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中幹細胞及/或增生標記物之表現含量高至少 300%。In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express stem cell and/or proliferation markers at higher levels than a population of reference tissue-derived epithelial organoids. Non-limiting examples of stem cell and/or proliferation markers include MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, and/or CD44, and combinations thereof. In certain embodiments, the stem cell and/or proliferation marker is selected from the group consisting of MKI67, ASCL2, LGR5, SOX9, SMOC2, CD44, and combinations thereof. In some embodiments, the stem cell and/or proliferation marker is MKI67. In some embodiments, the stem cell and/or proliferation marker is ASCL2. In some embodiments, the stem cell and/or proliferation marker is LGR5. In some embodiments, the stem cell and/or proliferation marker is SOX9. In some embodiments, the stem cell and/or proliferation marker is SMOC2. In some embodiments, the stem cell and/or proliferation marker is CD44. In some embodiments, the stem cell and/or proliferation marker is EpCAM. In some embodiments, the stem cell and/or proliferation marker is CD49f. In some embodiments, the stem cell and/or proliferation marker is CD133. In some embodiments, the stem cell and/or proliferation marker is ALDH1A1. In some embodiments, the stem cell and/or proliferation marker is NEUROG3. In some embodiments, the stem cell and/or proliferation marker is NKX6.1. In some embodiments, the stem cell and/or proliferation marker is PDX1. In some embodiments, the stem cell and/or proliferation marker is BMI1. In certain embodiments, the expression level of a stem cell and/or proliferation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 10% higher, at least 20% higher, at least 30% higher, at least 40% higher, at least 50% higher, at least 60% higher, at least 70% higher, at least 80% higher, at least 90% higher, at least 100% higher, at least 110% higher, at least 120% higher, at least 130% higher, at least 140% higher, at least 150% higher, at least 160% higher, at least 170% higher, at least 180% higher, at least 190% higher, at least 200% higher, at least 210% higher, at least 220% higher, at least 230% higher, at least 240% higher, at least 250% higher, at least 260% higher, at least 270% higher, at least 280% higher, at least 290% higher, at least 300% higher, at least 310% higher, at least 320% higher, at least 330% higher, at least 340% higher, at least 350% higher, at least 360% higher, at least 370% higher, at least 380% higher, at least 390% higher, at least 400% higher, at least 410% higher, at least 420% higher, at least 430% higher, at least 440% higher, at least 450% higher, at least 460% higher, at least 470% higher, at least 480% higher, at least 490% higher In some embodiments, the expression level of stem cells and/or proliferation markers in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 50% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue. In some embodiments, the expression level of stem cells and/or proliferation markers in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 100% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids derived from a reference tissue. In certain embodiments, the expression level of stem cells and/or proliferation markers in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 200% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids from a reference tissue. In certain embodiments, the expression level of stem cells and/or proliferation markers in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 300% higher than the expression level of stem cells and/or proliferation markers in a population of epithelial organoids from a reference tissue.
在某些實施例中,本揭露之方法產生以與參考組織來源之上皮類器官之群體相比較低之含量表現分化標記物之組織來源之上皮類器官之群體。分化標記物之非限制性實例包括角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。在某些實施例中,分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、TFF3、ALPI、SI、CEACAM7 及其組合。在某些實施例中,分化標記物為角蛋白 20 (KRT20)。在某些實施例中,分化標記物為 FABP1。在某些實施例中,分化標記物為 MUC2。在某些實施例中,分化標記物為 MUC5B。在某些實施例中,分化標記物為 TFF3。在某些實施例中,分化標記物為 ALPI。在某些實施例中,分化標記物為 SI。在某些實施例中,分化標記物為 CEACAM7。在某些實施例中,分化標記物為角蛋白 19 (KRT19)。在某些實施例中,分化標記物為角蛋白 7 (KRT7)。在某些實施例中,分化標記物為 SOX9。在某些實施例中,分化標記物為 SOX9。在某些實施例中,分化標記物為 MUC1。在某些實施例中,分化標記物為 INS。在某些實施例中,分化標記物為 GCG。在某些實施例中,分化標記物為 AMY。在某些實施例中,分化標記物為 ALB。在某些實施例中,分化標記物為 CYP3A4。在某些實施例中,分化標記物為 HNF4A。在某些實施例中,分化標記物為細胞角蛋白 8 (K8)。在某些實施例中,分化標記物為細胞角蛋白 18 (K18)。在某些實施例中,分化標記物為細胞角蛋白 5 (K5)。在某些實施例中,分化標記物為細胞角蛋白 14 (K14)。在某些實施例中,分化標記物為平滑肌肌動蛋白 (SMA)。在某些實施例中,分化標記物為 MUC5AC。在某些實施例中,分化標記物為 MUC6。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 10%、低至少 20%、低至少 30%、低至少 40%、低至少 50%、低至少 60%、低至少 70%、低至少 80%、低至少 90%、低至少 100%、低至少 110%、低至少 120%、低至少 130%、低至少 140%、低至少 150%、低至少 160%、低至少 170%、低至少 180%、低至少 190%、低至少 200%、低至少 210%、低至少 220%、低至少 230%、低至少 240%、低至少 200%、低至少 250%、低至少 260%、低至少 270%、低至少 280%、低至少 290% 或低至少 300%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 50%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 100%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 200%。在某些實施例中,與參考組織來源之上皮類器官之群體中分化標記物之表現含量相比,藉由本揭露之方法產生之組織來源之上皮類器官之群體中分化標記物之表現含量低至少 300%。In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a differentiation marker at a lower level than a population of reference tissue-derived epithelial organoids. Non-limiting examples of differentiation markers include keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA), and combinations thereof. In some embodiments, the differentiation marker is selected from the group consisting of keratin 20 (KRT20), FABP1, MUC2, MUC5B, TFF3, ALPI, SI, CEACAM7, and combinations thereof. In some embodiments, the differentiation marker is keratin 20 (KRT20). In some embodiments, the differentiation marker is FABP1. In some embodiments, the differentiation marker is MUC2. In some embodiments, the differentiation marker is MUC5B. In some embodiments, the differentiation marker is TFF3. In some embodiments, the differentiation marker is ALPI. In some embodiments, the differentiation marker is SI. In some embodiments, the differentiation marker is CEACAM7. In some embodiments, the differentiation marker is keratin 19 (KRT19). In some embodiments, the differentiation marker is keratin 7 (KRT7). In some embodiments, the differentiation marker is SOX9. In some embodiments, the differentiation marker is SOX9. In some embodiments, the differentiation marker is MUC1. In some embodiments, the differentiation marker is INS. In some embodiments, the differentiation marker is GCG. In some embodiments, the differentiation marker is AMY. In some embodiments, the differentiation marker is ALB. In some embodiments, the differentiation marker is CYP3A4. In some embodiments, the differentiation marker is HNF4A. In some embodiments, the differentiation marker is cytokeratin 8 (K8). In some embodiments, the differentiation marker is cytokeratin 18 (K18). In some embodiments, the differentiation marker is cytokeratin 5 (K5). In some embodiments, the differentiation marker is cytokeratin 14 (K14). In some embodiments, the differentiation marker is smooth muscle actin (SMA). In some embodiments, the differentiation marker is MUC5AC. In some embodiments, the differentiation marker is MUC6. In certain embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 10% lower, at least 20% lower, at least 30% lower, at least 40% lower, at least 50% lower, at least 60% lower, at least 70% lower, at least 80% lower, at least 90% lower, at least 100% lower, at least 110% lower, at least 120% lower, at least 130% lower, at least 140% lower, at least 150% lower, at least 160% lower, at least 170% lower, at least 180% lower, at least 190% lower, at least 200% lower, at least 210% lower, at least 220% lower, at least 230% lower, at least 240% lower, at least 200% lower, at least 250% lower, at least 260% lower, at least 270% lower, at least 280% lower, at least 290% lower, at least 300% lower, at least 310% lower, at least 320% lower, at least 330% lower, at least 340% lower, at least 350% lower, at least 360% lower, at least 370% lower, at least 380% lower, at least 390% lower, at least 400% lower, at least 410% lower, at least 420% lower, at least 430% lower, at least 440% lower In some embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 50% lower than the expression level of a differentiation marker in a population of epithelial organoids from a reference tissue. In some embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 100% lower than the expression level of a differentiation marker in a population of epithelial organoids from a reference tissue. In certain embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 200% lower than the expression level of a differentiation marker in a population of epithelial organoids from a reference tissue. In certain embodiments, the expression level of a differentiation marker in a population of tissue-derived epithelial organoids produced by the methods of the present disclosure is at least 300% lower than the expression level of a differentiation marker in a population of epithelial organoids from a reference tissue.
在某些實施例中,用於產生胃腸類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 10 µL。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠可為商業可獲得之 ECM。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。BME 之非限制性實例為 CULTREX® 基底膜萃取物 2 型 (R&D Systems),包括層連結蛋白、巢蛋白、膠原蛋白 IV 及硫酸肝素蛋白聚醣。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在培養基 ( 例如,大腸傳代培養基 (Intesticult 類器官生長培養基 (OGM, StemCell Technologies 目錄號 06010) + 10 µM Y27632) 或迴腸培養基 (OGM + 10 µM Y27632 + 2.5 µM CHIR99021)) 中培養以分別產生大腸或迴腸類器官。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在類器官碎片內。在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。在某些實施例中,差異表現標記物為 MKI67、LGR5、SOX9、CD44、MUC2、MUC5B、TFF3、KRT20、FABP1、ALPI 及/或 CEACAM7。 In certain embodiments, the method for generating gastrointestinal organoids comprises contacting tissue-derived epithelial stem cells with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells can be combined with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1×10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1×10 7 tissue-derived epithelial stem cells/ml hydrogel. In some embodiments, the method may further include suspending the hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. For example, but not limited to, the volume of the hydrogel-tissue derived epithelial stem cell mixture dispensed into the culture medium may be at least about 10 μL. In some embodiments, the ratio of the volume of the hydrogel-tissue derived epithelial stem cell mixture to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 37°C. In some embodiments, the hydrogel can be a commercially available ECM. In some embodiments, the ECM is a basement membrane extract (BME), which is a soluble form of the basement membrane. A non-limiting example of BME is CULTREX® Basement Membrane Extract Type 2 (R&D Systems), including laminin, entactin, collagen IV, and heparin sulfate proteoglycans. In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture is distributed into a culture medium having a certain geometric shape. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is suspended in a culture medium in a droplet. In certain embodiments, the method may further comprise culturing the tissue-derived epithelial stem cells of the suspended hydrogel-tissue-derived epithelial stem cell mixture in a culture medium ( e.g. , colon passage medium (Intesticult Organoid Growth Medium (OGM, StemCell Technologies Catalog No. 06010) + 10 µM Y27632) or ileum culture medium (OGM + 10 µM Y27632 + 2.5 µM CHIR99021)) to generate colon or ileum organoids, respectively. In some embodiments, the method may further include fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures comprising the tissue-derived epithelial organoid. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce the fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting the filamentous structure produces a structure that is shorter in length, width, or both. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained in the organoid fragments. In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In certain embodiments, the differentially expressed marker is MKI67, LGR5, SOX9, CD44, MUC2, MUC5B, TFF3, KRT20, FABP1, ALPI, and/or CEACAM7.
在某些實施例中,用於產生肺類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 10 µL。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠可為商業可獲得之 ECM。ECM 之非限制性實例為 MATRIGEL®。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在培養基 ( 例如,含有 10 µM ROCK 抑制劑之 SFFF 培養基) 中培養。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在類器官碎片內。在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。 In certain embodiments, a method for generating lung organoids comprises contacting tissue-derived epithelial stem cells with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells may be combined with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1×10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1×10 7 tissue-derived epithelial stem cells/ml hydrogel. In some embodiments, the method may further include suspending the hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. For example, but not limited to, the volume of the hydrogel-tissue derived epithelial stem cell mixture dispensed into the culture medium may be at least about 10 μL. In some embodiments, the ratio of the volume of the hydrogel-tissue derived epithelial stem cell mixture to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue-derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 37°C. In some embodiments, the hydrogel can be a commercially available ECM. A non-limiting example of an ECM is MATRIGEL®. In some embodiments, the suspended hydrogel-tissue-derived epithelial stem cell mixture is distributed in a culture medium having a certain geometric shape. In some embodiments, the geometric shape of the suspended hydrogel-tissue-derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometric shape of the suspended hydrogel-tissue-derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the hydrogel-tissue-derived epithelial stem cell mixture is suspended in a culture medium in small drops. In some embodiments, the method may further include culturing the tissue-derived epithelial stem cells of the suspended hydrogel-tissue-derived epithelial stem cell mixture in a culture medium ( e.g. , SFFF culture medium containing 10 μM ROCK inhibitor). In some embodiments, the method may further include fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to produce a fragmented structure comprising the tissue-derived epithelial organoid. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting filamentous structures produces structures with shorter length, width, or both. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained within an organoid fragment. In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids).
在某些實施例中,用於產生乳腺類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 10 µL。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠可為商業可獲得之 ECM。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。BME 之非限制性實例為 CULTREX® 低生長因子 BME 2 型 (Trevigen, 3533-010-02)。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在培養基 ( 例如,含有 10 µM ROCK 抑制劑之 SFFF 培養基) 中培養。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在類器官碎片內。在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。在某些實施例中,差異表現之標記物為 EpCAM、CD49f、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14) 及/或平滑肌肌動蛋白 (SMA)。 In certain embodiments, a method for generating mammary organoids comprises contacting tissue-derived epithelial stem cells with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells may be combined with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel. In some embodiments, the method may further include suspending the hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. For example, but not limited to, the volume of the hydrogel-tissue derived epithelial stem cell mixture dispensed into the culture medium may be at least about 10 μL. In some embodiments, the ratio of the volume of the hydrogel-tissue derived epithelial stem cell mixture to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 37°C. In some embodiments, the hydrogel can be a commercially available ECM. In some embodiments, the ECM is a basement membrane extract (BME), which is a soluble form of the basement membrane. A non-limiting example of BME is CULTREX® Low Growth Factor BME Type 2 (Trevigen, 3533-010-02). In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture is distributed into a culture medium having a certain geometric shape. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is suspended in a culture medium in a droplet. In certain embodiments, the method may further include culturing the tissue-derived epithelial stem cells of the suspended hydrogel-tissue-derived epithelial stem cell mixture in a culture medium ( e.g. , SFFF medium containing 10 μM ROCK inhibitor). In certain embodiments, the method may further include fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to generate a fragmented structure comprising the tissue-derived epithelial organoid. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting filamentous structures produces structures with shorter length, width, or both. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained within an organoid fragment. In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In certain embodiments, the differentially expressed marker is EpCAM, CD49f, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), and/or smooth muscle actin (SMA).
在某些實施例中,用於產生胰臟類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 10 µL。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠可為商業可獲得之 ECM。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。BME 之非限制性實例為低生長因子 BME 2-RGF (基底膜萃取物 2 型 3533-010-02;AMSBIO,CULTREX®)。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中。在某些實施例中,該方法可進一步包括在前 7 天過程中將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在優化人類胰臟類器官擴增培養基 ( 例如,包括補充有 1X N2 及 1X B27 (皆來自 GIBCO)、1.25 mM N-乙醯半胱胺酸 (Sigma-Aldrich)、10% RSPO1 條件無血清培養基、10 nM 人類 [Leu 15]-胃泌激素 I (Sigma-Aldrich)、50 ng/mL EGF (Peprotech)、25 ng/mL Noggin (Peprotech)、100 ng/mL FGF10 (Peprotech)、10 mM 菸鹼醯胺 (Sigma-Aldrich)、5 μM A83.01 (Tocris)、10 μM FSK (Tocris) 及 3 μM PGE2 (Tocris) 且補充有 10 μM Rho 激酶抑制劑 (Y27632, Sigma-Aldrich) 之基礎培養基) 中培養。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在類器官碎片內。在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。在某些實施例中,組織來源之上皮類器官為胰臟類器官,且差異表現標記物為 CD133、LGR5、PDX1、SOX9、ALDH1A1、NEUROG3、NKX6.1、角蛋白 19 (KRT19)、MUC1、INS、GCG 及/或 AMY。 In certain embodiments, a method for generating pancreatic organoids comprises contacting tissue-derived epithelial stem cells with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells may be combined with a hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 1×10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1×10 7 tissue-derived epithelial stem cells/ml hydrogel. In some embodiments, the method may further include suspending the hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. For example, but not limited to, the volume of the hydrogel-tissue derived epithelial stem cell mixture dispensed into the culture medium may be at least about 10 μL. In some embodiments, the ratio of the volume of the hydrogel-tissue derived epithelial stem cell mixture to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 37°C. In some embodiments, the hydrogel can be a commercially available ECM. In some embodiments, the ECM is a basement membrane extract (BME), which is a soluble form of the basement membrane. A non-limiting example of BME is low growth factor BME 2-RGF (basement membrane extract type 2 3533-010-02; AMSBIO, CULTREX®). In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture is distributed into a culture medium having a certain geometric shape. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is suspended in a culture medium in a droplet. In certain embodiments, the method can further include culturing the suspended hydrogel-tissue-derived epithelial stem cells in an optimized human pancreatic organoid expansion medium ( e.g. , a serum-free medium supplemented with 1X N2 and 1X B27 (both from GIBCO), 1.25 mM N-acetylcysteine (Sigma-Aldrich), 10% RSPO1 conditioned medium, 10 nM human [Leu 15 ]-gastrin I (Sigma-Aldrich), 50 ng/mL EGF (Peprotech), 25 ng/mL Noggin (Peprotech), 100 ng/mL FGF10 (Peprotech), 10 mM niacinamide (Sigma-Aldrich), 5 μM A83.01 (Tocris), 10 μM FSK (Tocris) and 3 μM PGE2 (Tocris) supplemented with 10 μM Rho kinase inhibitor (Y27632, Sigma-Aldrich). In certain embodiments, the method may further comprise fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to generate a fragmented structure comprising the tissue-derived epithelial organoid. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting filamentous structures produces structures with shorter length, width, or both. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained within an organoid fragment. In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In certain embodiments, the tissue-derived epithelial organoids are pancreatic organoids, and the differentially expressed marker is CD133, LGR5, PDX1, SOX9, ALDH1A1, NEUROG3, NKX6.1, keratin 19 (KRT19), MUC1, INS, GCG, and/or AMY.
在某些實施例中,用於產生肝類器官之方法包括使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞可與水凝膠組合以產生水凝膠-組織來源之上皮幹細胞混合物。在某些實施例中,複數個組織來源之上皮幹細胞包括盤 ( 例如,48 孔盤) 中每孔約 3,000 至約 10,000 個組織來源之上皮幹細胞。在某些實施例中,該方法可進一步包括將水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物。 例如但不限於,分配到培養基中之水凝膠-組織來源之上皮幹細胞混合物之體積可為至少約 10 µL。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物體積與培養基體積之比率為約 1:2 至約 1:15。在某些實施例中,該水凝膠係在與培養基接觸時固化。例如但不限於,水凝膠-組織來源之上皮幹細胞混合物之水凝膠由在大於約 37℃ 之溫度固化之材料構成。在某些實施例中,水凝膠可為商業可獲得之 ECM。在某些實施例中,ECM 為基底膜萃取物 (BME),其為基底膜之可溶性形式。ECM 之非限制性實例為 MATRIGEL® (BD Biosciences) 或低生長因子 BME 2 (基底膜萃取物,2 型,Pathclear)。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物分配到具有某個幾何形狀之培養基中。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為球形或類球形。在某些實施例中,經懸浮之水凝膠-組織來源之上皮幹細胞混合物之幾何形狀為絲狀結構。在某些實施例中,絲狀結構具有線形、蛇形或螺旋形形狀。在某些實施例中,水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮於培養基中。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物之組織來源之上皮幹細胞在培養基中培養,該培養基 例如包含 AdDMEM/F12 (Invitrogen),其補充有 1% N2 (GIBCO) 及 1% B27 (GIBCO)、1.25 mM N-乙醯半胱胺酸 (Sigma)、10 nM 胃泌激素 (Sigma) 及生長因子:50 ng/ml EGF (Peprotech)、10% RSPO1 條件培養基 (自製)、100 ng/ml FGF10 (Peprotech)、25 ng/ml HGF (Peprotech)、10 mM 菸鹼醯胺 (Sigma)、5 µM A83.01 (Tocris) 及 10 µM FSK (Tocris),在分離後的前 3 天過程中,補充有 25 ng/ml Noggin (Peprotech)、30% Wnt 培養基 (如 Barker 等人Cell Stem Cell 6:25-36 (2010) 所述) 及 10 µM (Y27632, Sigma Aldrich) 或 hES 細胞選殖恢復溶液 (Stemgent) 以建立培養物。在某些實施例中,隨後將培養基更換為 例如不含 Noggin、Wnt、Y27632 及 hES 細胞選殖恢復溶液之培養基。在某些實施例中,將肝類器官在補充有 BMP7 (25 ng/ml) 之上述培養基中接種並培養 7 至 10 天。在某些實施例中,隨後將培養基更換為分化培養基, 例如,其包括補充有 1% N2 及 1% B27 並含有 EGF (50 ng/ml)、胃泌激素 (10 nM, Sigma)、HGF (25 ng/ml)、FGF19 (100 ng/ml)、A8301 (500 nM)、DAPT (10 µM)、BMP7 (25 ng/ml) 及地塞米松 (30 µM) 之 AdDMEM/F12 培養基,以產生肝細胞類器官。在某些實施例中,該方法可進一步包括將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化可包括剪切經懸浮之水凝膠-組織來源之上皮幹細胞混合物以產生碎片化結構, 例如藉由使含有經懸浮之水凝膠-組織來源之上皮幹細胞混合物之培養基上下移液。在某些實施例中,將經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化產生長度及/或寬度較短之結構。例如但不限於,將絲狀結構碎片化產生長度、寬度或二者較短之結構。在某些實施例中,使用於本揭露之組織來源之上皮幹細胞或複數個組織來源之上皮幹細胞含在類器官碎片內。在某些實施例中,本揭露之方法產生組織來源之上皮類器官之群體,其以與參考組織來源之上皮類器官 ( 例如,參考組織來源之上皮類器官之群體) 相比不同 ( 例如較高或較低) 的含量表現標記物。在某些實施例中,組織來源之上皮類器官為肝類器官,且差異表現標記物為 LGR5、ALB、CYP3A4、HNF4A、KRT19、KRT7 及/或 SOX9。 In certain embodiments, a method for generating a liver organoid comprises contacting a tissue-derived epithelial stem cell with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, a plurality of tissue-derived epithelial stem cells may be combined with a hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture. In certain embodiments, the plurality of tissue-derived epithelial stem cells comprises about 3,000 to about 10,000 tissue-derived epithelial stem cells per well in a dish ( e.g. , a 48-well dish). In some embodiments, the method may further include suspending the hydrogel-tissue derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue derived epithelial stem cell mixture. For example, but not limited to, the volume of the hydrogel-tissue derived epithelial stem cell mixture dispensed into the culture medium may be at least about 10 μL. In some embodiments, the ratio of the volume of the hydrogel-tissue derived epithelial stem cell mixture to the volume of the culture medium is about 1:2 to about 1:15. In some embodiments, the hydrogel solidifies when in contact with the culture medium. For example, but not limited to, the hydrogel of the hydrogel-tissue derived epithelial stem cell mixture is composed of a material that solidifies at a temperature greater than about 37°C. In some embodiments, the hydrogel can be a commercially available ECM. In some embodiments, the ECM is a basement membrane extract (BME), which is a soluble form of the basement membrane. Non-limiting examples of ECM are MATRIGEL® (BD Biosciences) or low growth factor BME 2 (basement membrane extract, type 2, Pathclear). In some embodiments, the suspended hydrogel-tissue derived epithelial stem cell mixture is distributed into a culture medium having a certain geometric shape. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is spherical or spherical. In some embodiments, the geometry of the suspended hydrogel-tissue derived epithelial stem cell mixture is a filamentous structure. In some embodiments, the filamentous structure has a linear, serpentine or spiral shape. In some embodiments, the hydrogel-tissue derived epithelial stem cell mixture is suspended in a culture medium in a droplet. In certain embodiments, the method may further comprise culturing the tissue-derived epithelial stem cells of the suspended hydrogel-tissue-derived epithelial stem cell mixture in a culture medium, such as AdDMEM/F12 (Invitrogen) supplemented with 1% N2 (GIBCO) and 1% B27 (GIBCO), 1.25 mM N-acetylcysteine (Sigma), 10 nM gastrin (Sigma), and growth factors: 50 ng/ml EGF (Peprotech), 10% RSPO1 conditioned medium (homemade), 100 ng/ml FGF10 (Peprotech), 25 ng/ml HGF (Peprotech), 10 mM niacinamide (Sigma), 5 µM A83.01 (Tocris) and 10 µM FSK (Tocris), supplemented with 25 ng/ml Noggin (Peprotech), 30% Wnt medium (as described in Barker et al. Cell Stem Cell 6:25-36 (2010)) and 10 µM (Y27632, Sigma Aldrich) or hES cell selective recovery solution (Stemgent) during the first 3 days after isolation to establish the culture. In certain embodiments, the medium is then changed to medium that does not contain Noggin, Wnt, Y27632, and hES cell selective recovery solution. In certain embodiments, hepatic organoids are plated and cultured in the above medium supplemented with BMP7 (25 ng/ml) for 7 to 10 days. In certain embodiments, the medium is then replaced with a differentiation medium, for example , comprising AdDMEM/F12 medium supplemented with 1% N2 and 1% B27 and containing EGF (50 ng/ml), gastrin (10 nM, Sigma), HGF (25 ng/ml), FGF19 (100 ng/ml), A8301 (500 nM), DAPT (10 µM), BMP7 (25 ng/ml) and dexamethasone (30 µM) to generate hepatocyte organoids. In certain embodiments, the method may further comprise fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to generate a fragmented structure comprising the tissue-derived epithelial organoids. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture may include shearing the suspended hydrogel-tissue derived epithelial stem cell mixture to produce fragmented structures, such as by pipetting a culture medium containing the suspended hydrogel-tissue derived epithelial stem cell mixture up and down. In some embodiments, fragmenting the suspended hydrogel-tissue derived epithelial stem cell mixture produces structures with shorter length and/or width. For example, but not limited to, fragmenting filamentous structures produces structures with shorter length, width, or both. In certain embodiments, a tissue-derived epithelial stem cell or a plurality of tissue-derived epithelial stem cells used in the present disclosure are contained within an organoid fragment. In certain embodiments, the methods of the present disclosure produce a population of tissue-derived epithelial organoids that express a marker at a different ( e.g., higher or lower) level than a reference tissue-derived epithelial organoid ( e.g. , a population of reference tissue-derived epithelial organoids). In certain embodiments, the tissue-derived epithelial organoid is a liver organoid, and the differentially expressed marker is LGR5, ALB, CYP3A4, HNF4A, KRT19, KRT7, and/or SOX9.
在某些實施例中,所揭示方法之一個或多個步驟可以使用機器人及/或自動化組件來進行。在某些實施例中,本揭露之方法可包括使用機器人及/或自動化組件來產生組織來源之上皮類器官。在某些實施例中,所揭示方法之一個或多個步驟可使用機器人及/或自動化組件來進行以產生組織來源之上皮類器官。可在所揭示之方法中使用之機器人及/或自動化組件之非限制性實例包括自動化液體處理器 ( 例如,液體處理機器人)、3D 列印機、注射泵、電子移液管 ( 例如,具有或不具有移液機器人) 或其組合。電子移液管 ( 例如,具有移液機器人) 之非限制性實例為來自 Integra Biosciences 之 Assist Plus。在某些實施例中,所揭示方法之一個或多個步驟可以藉由自動化液體處理器 ( 例如,液體處理機器人) 來進行。 In certain embodiments, one or more steps of the disclosed methods may be performed using a robot and/or automated components. In certain embodiments, the methods of the present disclosure may include the use of a robot and/or automated components to produce tissue-derived epithelial organoids. In certain embodiments, one or more steps of the disclosed methods may be performed using a robot and/or automated components to produce tissue-derived epithelial organoids. Non-limiting examples of robots and/or automated components that may be used in the disclosed methods include automated liquid handlers ( e.g. , liquid handling robots), 3D printers, syringe pumps, electronic pipettes ( e.g. , with or without a pipetting robot), or combinations thereof. A non-limiting example of an electronic pipette ( e.g. , with a pipetting robot) is the Assist Plus from Integra Biosciences. In certain embodiments, one or more steps of the disclosed methods may be performed by an automated liquid handler ( eg , a liquid handling robot).
在某些實施例中,藉由本揭露之方法產生之組織來源之上皮類器官培養物之傳代藉由機器人及/或自動化組件來進行。在某些實施例中,自動化機器人可以進行本揭露實例 1 之「類器官維持」中描述之方法, 例如,進行以下步驟中之任一者:添加 TrypLE Express、加熱類器官培養物、研磨類器官培養物以解離細胞、添加 PBS、沉澱細胞、將細胞重懸於 BME 中、冷卻培養物、平鋪細胞、以培養基覆蓋類器官或其組合。在某些實施例中,細胞解離可使用機器人及/或自動化組件來進行。在某些實施例中, 例如在類器官維持過程中或在組織來源之上皮類器官產生過程中之培養基更換可使用機器人及/或自動化組件來進行。 In certain embodiments, the passage of tissue-derived epithelial organoid cultures produced by the methods of the present disclosure is performed by a robot and/or automated components. In certain embodiments, an automated robot can perform the method described in "Organoid Maintenance" of Example 1 of the present disclosure, for example , performing any of the following steps: adding TrypLE Express, heating the organoid culture, grinding the organoid culture to dissociate cells, adding PBS, sedimenting cells, resuspending cells in BME, cooling the culture, flattening cells, covering the organoid with culture medium, or a combination thereof. In certain embodiments, cell dissociation can be performed using a robot and/or automated components. In certain embodiments, medium replacement, such as during organoid maintenance or during the generation of tissue-derived epithelial organoids, can be performed using robotic and/or automated components.
在某些實施例中,細胞培養基中 BOBA 及/或 SOBA 之產生藉由機器人及/或自動化組件來進行。在某些實施例中,自動化機器人可進行本揭露實例 1 之「經懸浮之水凝膠 BOBA 培養物」及/或「經懸浮之水凝膠 SOBA 碎片培養物」中描述之方法, 例如,進行以下步驟中之任一者:加熱培養基、將類器官細胞-BME 溶液作為小滴分配到溫熱培養基中以產生 BOBA,將類器官細胞-BME 溶液以 X-Y 平面中線形、蛇形或螺旋形運動方式分配到溫熱培養基中以產生 SOBA、研磨 SOBA 絲培養物以產生 SOBA 碎片、更換媒體或其組合。在某些實施例中,將類器官細胞-BME 溶液分配到溫熱培養基中以產生 BOBA 及/或 SOBA 可使用機器人及/或自動化組件來進行, 例如可藉由液體處理機器人來進行。在某些實施例中,更換培養基可使用機器人及/或自動化組件來進行, 例如可藉由液體處理機器人來進行。在某些實施例中,研磨 SOBA 絲培養物以產生 SOBA 碎片可使用機器人及/或自動化組件來進行, 例如可藉由液體處理機器人來進行。 In certain embodiments, the production of BOBA and/or SOBA in the cell culture medium is performed by a robot and/or automated assembly. In certain embodiments, the automated robot can perform the methods described in "Suspended hydrogel BOBA culture" and/or "Suspended hydrogel SOBA fragment culture" of Example 1 of the present disclosure, for example , performing any of the following steps: heating the culture medium, dispensing the organoid cell-BME solution as droplets into the warm culture medium to produce BOBA, dispensing the organoid cell-BME solution into the warm culture medium in a linear, serpentine or spiral motion in the XY plane to produce SOBA, grinding SOBA filament culture to produce SOBA fragments, changing the medium or a combination thereof. In some embodiments, dispensing the organoid cell-BME solution into the warm culture medium to produce BOBA and/or SOBA can be performed using a robot and/or automated components, such as a liquid handling robot. In some embodiments, changing the culture medium can be performed using a robot and/or automated components, such as a liquid handling robot. In some embodiments, grinding the SOBA silk culture to produce SOBA fragments can be performed using a robot and/or automated components, such as a liquid handling robot.
在某些實施例中, 例如本文所述之高生產量方法可使用機器人及/或自動化組件來進行。例如但不限於,本文所揭示之使用方法中任一者, 例如如第 IV 節所描述的,可部分地使用機器人及/或自動化組件來進行。在某些實施例中,用於篩選藥劑 例如治療劑之方法及用於使用所揭示之組織來源之上皮類器官進行基因體篩選之方法可部分地使用機器人及/或自動化組件來進行。 IV. 使用方法 In some embodiments, high throughput methods such as those described herein can be performed using robotics and/or automated components. For example, but not limited to, any of the methods of use disclosed herein, such as those described in Section IV, can be performed in part using robotics and/or automated components. In some embodiments, methods for screening agents such as therapeutic agents and methods for performing genomic screening using the disclosed tissue-derived epithelial organoids can be performed in part using robotics and/or automated components. IV. Methods of Use
本揭露提供了使用所揭示之類器官或包括此等類器官之組成物之方法。在某些實施例中,本揭露之組織來源之上皮類器官可用於篩選測定。例如但不限於,本揭露提供了用於篩選藥劑 ( 例如治療劑) 之方法,以及用於使用所揭示之組織來源之上皮類器官進行基因體篩選之方法。在某些實施例中,本揭露之組織來源之上皮類器官可用於產生基於類器官之模型。 The present disclosure provides methods of using the disclosed organoids or compositions comprising such organoids. In certain embodiments, the tissue-derived epithelial organoids of the present disclosure can be used in screening assays. For example, but not limited to, the present disclosure provides methods for screening agents ( e.g., therapeutic agents), and methods for performing genomic screening using the disclosed tissue-derived epithelial organoids. In certain embodiments, the tissue-derived epithelial organoids of the present disclosure can be used to generate organoid-based models.
在某些實施例中,本揭露之類器官或其組成物可用於鑑定具有治療效果之藥劑。在某些實施例中,本揭露之類器官或其組成物可用於鑑定可有效預防及/或治療疾病之治療劑。在某些實施例中,本揭露之類器官或其組成物可用於鑑定可有效改善疾病症狀之治療劑。In certain embodiments, the organoids or compositions thereof disclosed herein can be used to identify agents having therapeutic effects. In certain embodiments, the organoids or compositions thereof disclosed herein can be used to identify therapeutic agents that can effectively prevent and/or treat diseases. In certain embodiments, the organoids or compositions thereof disclosed herein can be used to identify therapeutic agents that can effectively improve disease symptoms.
在某些實施例中,本揭露之類器官或其組成物可用於研究疾病之生物學及/或發病機制。例如但不限於,本揭露之類器官或其組成物可與藥劑接觸以研究疾病之生物學及/或發病機制。In certain embodiments, the organoids or compositions thereof disclosed herein can be used to study the biology and/or pathogenesis of a disease. For example, but not limited to, the organoids or compositions thereof disclosed herein can be contacted with a drug to study the biology and/or pathogenesis of a disease.
在某些實施例中,本揭露之類器官或其組成物可用於鑑定可能有毒之藥劑, 例如治療劑。在某些實施例中,本揭露之類器官或其組成物可用於鑑定藥劑 ( 例如治療劑) 可能有毒的濃度。 In certain embodiments, the disclosed organoids or compositions thereof can be used to identify potentially toxic agents, such as therapeutic agents. In certain embodiments, the disclosed organoids or compositions thereof can be used to identify the concentrations at which agents ( such as therapeutic agents) may be toxic.
在某些實施例中,用於鑑定可有效預防及/或治療疾病之治療劑之方法、用於鑑定可有效改善疾病症狀之治療劑之方法及/或用於鑑定可能有毒之治療劑之方法可包括使組織來源之上皮類器官或組織來源之上皮類器官之群體與治療劑接觸。在某些實施例中,該方法可包括使包括組織來源之上皮類器官或組織來源之上皮類器官之群體之組成物與治療劑接觸。在某些實施例中,將組織來源之上皮類器官或組織來源之上皮類器官之群體嵌入在懸浮於之培養基中之水凝膠中,如本文所述。In certain embodiments, methods for identifying therapeutic agents that are effective in preventing and/or treating a disease, methods for identifying therapeutic agents that are effective in ameliorating symptoms of a disease, and/or methods for identifying therapeutic agents that are potentially toxic may include contacting a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids with a therapeutic agent. In certain embodiments, the method may include contacting a composition comprising a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids with a therapeutic agent. In certain embodiments, the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids is embedded in a hydrogel suspended in a culture medium, as described herein.
在某些實施例中,用於研究疾病之生物學及/或發病機制之方法可包括使組織來源之上皮類器官或組織來源之上皮類器官之群體與藥劑接觸。在某些實施例中,該方法可包括使包括組織來源之上皮類器官或組織來源之上皮類器官之群體之組成物與藥劑接觸。在某些實施例中,將組織來源之上皮類器官或組織來源之上皮類器官之群體嵌入在懸浮於之培養基中之水凝膠中,如本文所述。In certain embodiments, methods for studying the biology and/or pathogenesis of a disease may include contacting a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids with an agent. In certain embodiments, the method may include contacting a composition including a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids with an agent. In certain embodiments, the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids is embedded in a hydrogel suspended in a culture medium as described herein.
在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 3 年, 例如,約 15 分鐘至約 3 年、約 15 分鐘至約 2.5 年、約 15 分鐘至約 2 年、約 15 分鐘至約 1.5 年、約 15 分鐘至約 1 年、約 15 分鐘至約 183 天、約 15 分鐘至約 150 天、約 15 分鐘至約 100 天、約 15 分鐘至約 50 天、約 1 天至約 3 年、約 10 天至約 3 年、約 20 天至約 3 年、約 50 天至約 3 年、約 100 天至約 3 年、約 150 天至約 3 年、約 183 天至約 3 年、約 1 年至約 3 年、約 1.5 年至約 3 年、約 2 年至約 3 年、或約 2.5 年至約 3 年。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 100 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 15 分鐘至約 100 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 150 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 15 分鐘至約 150 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 1 年。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 15 分鐘至約 1 年。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 2 年。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 15 分鐘至約 2 年。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 分鐘至約 10 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 15 分鐘至約 10 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 1 小時至約 10 天、約 12 小時至約 10 天、約 1 天至約 10 天、約 2 天至約 10 天、約 3 天至約 10 天、約 4 天至約 10 天、約 5 天至約 10 天、約 6 天至約 10 天、約 7 天至約 10 天、約 8 天至約 10 天、約 9 天至約 10 天、約 15 分鐘至約 10 天、約 15 分鐘至約 9 天、約 15 分鐘至約 8 天、約 15 分鐘至約 7 天、約 15 分鐘至約 6 天、約 15 分鐘至約 5 天、約 15 分鐘至約 4 天、約 15 分鐘至約 3 天、約 15 分鐘至約 2 天、約 15 分鐘至約 1 天、約 1 天至約 5 天、約 1 天至約 2 天、約 2 天至約 5 天、或約 2 天至約 10 天。在某些實施例中,使藥劑 ( 例如治療劑) 與組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 接觸約 2 天至約 10 天。 In certain embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 3 years, e.g. , about 15 minutes to about 3 years, about 15 minutes to about 2.5 years, about 15 minutes to about 2 years, about 15 minutes to about 1.5 years, about 15 minutes to about 1 year, about 15 minutes to about 183 days, about 15 minutes to about 150 days, about 15 minutes to about 100 days, about 15 minutes to about 50 days, about 1 day to about 3 years, about 10 days to about 3 years, about 20 days to about 3 years, about 50 days to about 3 years, about 100 days to about 3 years, about 150 days to about 3 years, about 183 days to about 3 years, about 1 year to about 3 years years, about 1.5 to about 3 years, about 2 to about 3 years, or about 2.5 to about 3 years. In some embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 100 days. In some embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 15 minutes to about 100 days. In some embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 150 days. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 15 minutes to about 150 days. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 1 year. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 15 minutes to about 1 year. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 2 years. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 15 minutes to about 2 years. In some embodiments, an agent ( e.g., a therapeutic agent) is in contact with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 minute to about 10 days. In certain embodiments, an agent ( eg, a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 15 minutes to about 10 days. In certain embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 1 hour to about 10 days, about 12 hours to about 10 days, about 1 day to about 10 days, about 2 days to about 10 days, about 3 days to about 10 days, about 4 days to about 10 days, about 5 days to about 10 days, about 6 days to about 10 days, about 7 days to about 10 days, about 8 days to about 10 days, about 9 days to about 10 days, about 15 minutes to about 10 days, about 15 minutes to about 9 days, about 15 minutes to about 8 days, about 15 minutes to about 7 days, about 15 minutes to about 6 days, about 15 minutes to about 5 days, about 15 minutes to about 4 days, about 15 minutes to about 3 days, about 15 minutes to about 15 days, about 15 minutes to about 10 days, about 15 minutes to about 15 ... Minutes to about 2 days, about 15 minutes to about 1 day, about 1 day to about 5 days, about 1 day to about 2 days, about 2 days to about 5 days, or about 2 days to about 10 days. In certain embodiments, an agent ( e.g., a therapeutic agent) is contacted with a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or a composition thereof) for about 2 days to about 10 days.
在某些實施例中,該方法可包括使不同組織來源之上皮類器官之群體 (或其組成物) 與增加濃度之藥劑 ( 例如治療劑)接觸,以允許劑量反應研究。 In certain embodiments, the method may include contacting populations of epithelial organoids (or compositions thereof) of different tissue origin with increasing concentrations of an agent ( e.g., a therapeutic agent) to allow for dose-response studies.
在某些實施例中,藥劑可為任何所關注的藥劑。在某些實施例中,藥劑為已知影響疾病之生物學及/或發病機制之分子。可用於所揭示之方法中之藥劑之非限制性實例包括肽、多肽、小分子、細胞、基因編輯系統或核酸。在某些實施例中,此等藥劑可為影響細胞傳訊、核酸表現、蛋白質表現、細胞生長、細胞分化及/或細胞存活之藥劑。In some embodiments, the agent can be any agent of interest. In some embodiments, the agent is a molecule known to affect the biology and/or pathogenesis of the disease. Non-limiting examples of agents that can be used in the disclosed methods include peptides, polypeptides, small molecules, cells, gene editing systems, or nucleic acids. In some embodiments, such agents can be agents that affect cell signaling, nucleic acid expression, protein expression, cell growth, cell differentiation, and/or cell survival.
在某些實施例中,該藥劑為治療劑。在某些實施例中,治療劑可為任何所關注的治療劑。在某些實施例中,治療劑獲自潛在治療劑文庫。可使用所揭示之方法分析及/或鑑定之治療劑之非限制性實例包括基於肽之治療劑、基於多肽之治療劑、小分子治療劑、基於細胞之治療劑、基因編輯系統、基於核酸之治療劑及其組合。In some embodiments, the agent is a therapeutic agent. In some embodiments, the therapeutic agent can be any therapeutic agent of interest. In some embodiments, the therapeutic agent is obtained from a library of potential therapeutic agents. Non-limiting examples of therapeutic agents that can be analyzed and/or identified using the disclosed methods include peptide-based therapeutic agents, polypeptide-based therapeutic agents, small molecule therapeutic agents, cell-based therapeutic agents, gene editing systems, nucleic acid-based therapeutic agents, and combinations thereof.
在某些實施例中,治療劑為基於肽之治療劑。在某些實施例中,基於肽之治療劑包括分子量約 5,000 Da 或更小之肽。基於肽之治療劑之非限制性實例包括生長因子、抗感染劑、抗真菌劑、抗細菌劑、受體配體及酪胺酸激酶抑製劑。Wang 等人(2022) Signal Transduction and Targeted Therapy 7:48 ( 例如表 1 及表 2) 中揭示了肽治療劑之其他非限制性實例,其內容以引用方式全文併入本文中。 In some embodiments, the therapeutic agent is a peptide-based therapeutic agent. In some embodiments, the peptide-based therapeutic agent includes a peptide having a molecular weight of about 5,000 Da or less. Non-limiting examples of peptide-based therapeutic agents include growth factors, anti-infective agents, antifungal agents, antibacterial agents, receptor ligands, and tyrosine kinase inhibitors. Other non-limiting examples of peptide therapeutic agents are disclosed in Wang et al. (2022) Signal Transduction and Targeted Therapy 7:48 ( e.g., Tables 1 and 2), the contents of which are incorporated herein by reference in their entirety.
在某些實施例中,治療劑為基於多肽之治療劑。基於多肽之治療劑之非限制性實例包括基於抗體之治療劑,諸如抗體及抗體藥物結合物、激素及酵素。在某些實施例中,抗體可為促效劑抗體或拮抗劑抗體。在某些實施例中,抗體可為抗體片段。抗體片段之非限制性實例包括但不限於 Fv、Fab、Fab'、Fab'-SH、F(ab') 2、雙功能抗體、線性抗體、單鏈抗體分子 ( 例如,scFv) 及由抗體片段形成之多特異性抗體。 In certain embodiments, the therapeutic agent is a polypeptide-based therapeutic agent. Non-limiting examples of polypeptide-based therapeutic agents include antibody-based therapeutic agents, such as antibodies and antibody-drug conjugates, hormones, and enzymes. In certain embodiments, the antibody may be an agonist antibody or an antagonist antibody. In certain embodiments, the antibody may be an antibody fragment. Non-limiting examples of antibody fragments include, but are not limited to, Fv, Fab, Fab', Fab'-SH, F(ab') 2 , bifunctional antibodies, linear antibodies, single-chain antibody molecules ( e.g. , scFv), and multispecific antibodies formed from antibody fragments.
在某些實施例中,治療劑為小分子治療劑。例如但不限於,小分子治療劑為分子量小於約 1,000 Da 之化合物。在某些實施例中,小分子治療劑包括細胞週期調節劑、激酶調節劑 ( 例如,激酶抑制劑或活化劑)、酵素抑制劑、受體調節劑 ( 例如,受體抑制劑或活化劑)、抗感染劑、抗真菌劑、抗細菌劑、化學治療劑及抗炎劑。 In certain embodiments, the therapeutic agent is a small molecule therapeutic agent. For example, but not limited to, a small molecule therapeutic agent is a compound with a molecular weight of less than about 1,000 Da. In certain embodiments, small molecule therapeutic agents include cell cycle regulators, kinase regulators ( e.g. , kinase inhibitors or activators), enzyme inhibitors, receptor regulators ( e.g. , receptor inhibitors or activators), anti-infective agents, antifungal agents, antibacterial agents, chemotherapeutic agents, and anti-inflammatory agents.
在某些實施例中,治療劑為基於細胞之治療劑。 基於細胞之治療劑之非限制性實例包括細菌細胞及免疫細胞。免疫細胞之非限制性實例包括嗜中性球、嗜酸性球、嗜鹼性球、肥大細胞、單核細胞、巨噬細胞、樹突細胞、自然殺手細胞 (NK 細胞) 及淋巴球, 例如B 細胞及 T 細胞 ( 例如,細胞毒性 T 細胞、自然殺傷 T 細胞、調節性 T 細胞及輔助性 T 細胞)。在某些實施例中,免疫細胞可為經過基因工程改造以表現嵌合抗原受體 (CAR) ( 例如,CAR T 細胞及 CAR NK 細胞) 或 T 細胞受體 (TCR) ( 例如,異源性 TCR) 之經修飾之免疫細胞。 In certain embodiments, the therapeutic agent is a cell-based therapeutic agent. Non-limiting examples of cell-based therapeutic agents include bacterial cells and immune cells. Non-limiting examples of immune cells include neutrophils, eosinophils, basophils, mast cells, monocytes, macrophages, dendritic cells, natural killer cells (NK cells) and lymphocytes, such as B cells and T cells ( e.g. , cytotoxic T cells, natural killer T cells, regulatory T cells and helper T cells). In certain embodiments, the immune cell may be a modified immune cell that has been genetically engineered to express a chimeric antigen receptor (CAR) ( eg , CAR T cells and CAR NK cells) or a T cell receptor (TCR) ( eg , a heterologous TCR).
在某些實施例中,治療劑為基因調節系統及/或基因調節系統之組分。在某些實施例中,基因調節系統及/或基因調節系統之組分為基因編輯系統、CRISPRi、基因表現促進系統、基因抑制促進系統、基於核酸之治療劑、轉錄因子及/或轉錄後修飾之調節劑。In some embodiments, the therapeutic agent is a gene regulatory system and/or a component of a gene regulatory system. In some embodiments, the gene regulatory system and/or a component of a gene regulatory system is a gene editing system, CRISPRi, a gene expression promoting system, a gene suppression promoting system, a nucleic acid-based therapeutic agent, a transcription factor, and/or a regulator of post-transcriptional modification.
在某些實施例中,治療劑為基因編輯系統。基因編輯系統之非限制性實例包括歸巢核酸內切酶或大範圍核酸酶、鋅指核酸酶 (ZFN)、類轉錄活化因子效應核酸酶 (TALEN) 及 CRISPR 基因編輯系統。在某些實施例中,治療劑為 CRISPR 基因編輯系統, 例如CRISPR/Cas9 基因編輯系統。 In some embodiments, the therapeutic agent is a gene editing system. Non-limiting examples of gene editing systems include nested endonucleases or meganucleases, zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR gene editing systems. In some embodiments, the therapeutic agent is a CRISPR gene editing system, such as a CRISPR/Cas9 gene editing system.
在某些實施例中,治療劑為基於核酸之治療劑。基於核酸之治療劑之非限制性實例包括基於 RNA 之治療劑,包括 siRNA、微小 RNA、RNA 適體、核酶、RNA 誘餌及 RNAi。在某些實施例中,基於核酸之治療劑包括基於 DNA 之治療劑,包括反義寡核苷酸 (ASO) 及 DNA 適體。In certain embodiments, the therapeutic agent is a nucleic acid-based therapeutic agent. Non-limiting examples of nucleic acid-based therapeutic agents include RNA-based therapeutic agents, including siRNA, microRNA, RNA aptamers, ribozymes, RNA baits, and RNAi. In certain embodiments, nucleic acid-based therapeutic agents include DNA-based therapeutic agents, including antisense oligonucleotides (ASOs) and DNA aptamers.
在某些實施例中,該方法可進一步包括分析組織來源之上皮類器官或組織來源之上皮類器官 (或組織來源之上皮類器官之細胞) 之群體中之變化。在某些實施例中,該方法可進一步包括分析在藥劑存在下發生的組織來源之上皮類器官或組織來源之上皮類器官 (或組織來源之上皮類器官之細胞) 之群體中之變化。例如但不限於,該方法可進一步包括分析組織來源之上皮類器官或組織來源之上皮類器官 (或組織來源之上皮類器官之細胞) 之群體中之變化,該變化指示治療劑之有效性及/或毒性。在某些實施例中,該方法包括分析組織來源之上皮類器官或組織來源之上皮類器官之群體中之變化,該變化指示治療劑與未以治療劑治療之組織來源之上皮類器官或組織來源之上皮類器官之群體相比之有效性及/或毒性。In certain embodiments, the method may further comprise analyzing the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or cells of tissue-derived epithelial organoids) for changes. In certain embodiments, the method may further comprise analyzing the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or cells of tissue-derived epithelial organoids) for changes that occur in the presence of an agent. For example, but not limitation, the method may further comprise analyzing the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or cells of tissue-derived epithelial organoids) for changes that indicate effectiveness and/or toxicity of a therapeutic agent. In certain embodiments, the method comprises analyzing a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids for changes indicative of the effectiveness and/or toxicity of a therapeutic agent compared to a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids not treated with the therapeutic agent.
在某些實施例中,本揭露之類器官或其組成物可用於進行基因體篩選。在某些實施例中,基因體篩選可用於鑑定用於產生突變之基因編輯系統。突變之非限制性實例包括缺失、重複、插入及核苷酸取代。在某些實施例中,該方法可包括提供組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物) 並在組織來源之上皮類器官之一個或多個細胞之基因體中產生突變。在某些實施例中,使用基因調節系統及/或基因調節系統之組分產生突變。在某些實施例中,基因調節系統及/或基因調節系統之組分為基因編輯系統、CRISPRi、RNAi、基因表現促進系統、基因抑制促進系統、基於核酸之治療劑、轉錄因子及/或轉錄後修飾之調節劑。在某些實施例中,基因編輯系統為 CRISPR 系統, 例如,CRISPR/Cas9 基因編輯系統。在某些實施例中,該方法可進一步包括分析組織來源之上皮類器官或組織來源之上皮類器官之群體中與突變相關之變化。在某些實施例中,該方法包括分析與不具有突變之組織來源之上皮類器官或組織來源之上皮類器官之群體相比,組織來源之上皮類器官或組織來源之上皮類器官之群體中與突變相關之變化。 In certain embodiments, the organoids or compositions thereof disclosed herein can be used for genome screening. In certain embodiments, genome screening can be used to identify gene editing systems for generating mutations. Non-limiting examples of mutations include deletions, duplications, insertions, and nucleotide substitutions. In certain embodiments, the method can include providing a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids (or compositions thereof) and generating mutations in the genome of one or more cells of the tissue-derived epithelial organoids. In certain embodiments, mutations are generated using a gene regulation system and/or a component of a gene regulation system. In some embodiments, the gene regulation system and/or a component of a gene regulation system is a gene editing system, CRISPRi, RNAi, a gene expression promoting system, a gene suppression promoting system, a nucleic acid-based therapeutic agent, a transcription factor, and/or a regulator of post-transcriptional modification. In some embodiments, the gene editing system is a CRISPR system, for example , a CRISPR/Cas9 gene editing system. In some embodiments, the method may further include analyzing changes associated with mutations in a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids. In certain embodiments, the method comprises analyzing changes associated with the mutation in a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids compared to a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids that does not have the mutation.
在某些實施例中,本揭露進一步提供了用於使用本揭露之組織來源之上皮類器官產生上皮細胞模型之方法。在某些實施例中,該方法可包括提供組織來源之上皮類器官或組織來源之上皮類器官之群體 (或其組成物)、將組織來源之上皮類器官或組織來源之上皮類器官之群體消化成單細胞,以及將單細胞在培養基中培養以產生細胞單層。在某些實施例中,將該單細胞在可滲透細胞培養內件 (cell culture insert) 上培養。在某些實施例中,該培養基為分化培養基。在某些實施例中,該培養基為幹細胞促進培養基。在某些實施例中,該培養基為細胞生長培養基。In certain embodiments, the present disclosure further provides methods for generating epithelial cell models using tissue-derived epithelial organoids of the present disclosure. In certain embodiments, the method may include providing a tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids (or a composition thereof), digesting the tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids into single cells, and culturing the single cells in a culture medium to produce a cell monolayer. In certain embodiments, the single cells are cultured on a permeable cell culture insert. In certain embodiments, the culture medium is a differentiation medium. In certain embodiments, the culture medium is a stem cell promoting medium. In certain embodiments, the culture medium is a cell growth medium.
在某些實施例中,細胞單層可以用於本文所揭示之方法中任一者, 例如用於本文所揭示之篩選方法。例如但不限於,細胞單層可用於篩選治療劑及用於進行基因體篩選。在某些實施例中,本揭露之單層可用於研究疾病之生物學及/或發病機制。 In certain embodiments, the cell monolayer can be used in any of the methods disclosed herein, such as in the screening methods disclosed herein. For example, but not limited to, the cell monolayer can be used to screen therapeutic agents and to perform genomic screening. In certain embodiments, the monolayer of the present disclosure can be used to study the biology and/or pathogenesis of a disease.
在某些實施例中,用於使用本揭露之細胞單層篩選治療劑之方法可包括接觸細胞單層並分析細胞單層之變化,該變化指示治療劑之有效性、流佈及/或毒性。在某些實施例中,該方法包括與未以治療劑處理之細胞單層相比,分析細胞單層中之變化,該變化指示治療劑之有效性及/或毒性。In certain embodiments, methods for screening therapeutic agents using cell monolayers of the present disclosure may include contacting the cell monolayer and analyzing the cell monolayer for changes that indicate the effectiveness, distribution, and/or toxicity of the therapeutic agent. In certain embodiments, the method includes analyzing changes in the cell monolayer that indicate the effectiveness and/or toxicity of the therapeutic agent compared to a cell monolayer that has not been treated with the therapeutic agent.
在某些實施例中,用於使用本揭露之細胞單層進行基因體篩選之方法可包括提供藉由本文所述之方法產生之細胞單層,在細胞單層之一個或多個細胞之基因體中產生突變,以及分析細胞單層中與突變相關之變化。在某些實施例中,該方法包括分析與不具有突變之細胞單層相比,細胞單層中與突變相關之變化。In certain embodiments, methods for performing genomic screening using a cell monolayer of the present disclosure may include providing a cell monolayer produced by the methods described herein, generating a mutation in the genome of one or more cells in the cell monolayer, and analyzing the cell monolayer for changes associated with the mutation. In certain embodiments, the method includes analyzing the cell monolayer for changes associated with the mutation compared to a cell monolayer without the mutation.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為以下中之變化:細胞生存力、細胞增生、類器官尺寸、細胞形態、類器官形態、對水凝膠之侵襲性、運動性、分化狀態、突變狀態、核型、染色體畸變、核酸表現含量、蛋白質表現含量、核酸修飾 ( 例如,甲基化)、轉譯後修飾 ( 例如,磷酸化、泛蛋白化及/或醣基化)、細胞傳訊路徑之活化、細胞傳訊路徑之抑制、酵素活性 ( 例如,酵素裂解)、染色質可及性、組蛋白修飾及其他表觀遺傳變化、類器官之物理特性 (包括胃腸上皮障壁之滲透性、pH、氧張力以及管腔及基底膜中其他代謝物及水凝膠及/或培養基中之分泌因子之濃度)、細胞介素及激素之濃度、藥物靈敏度、藥物吸收及代謝藥物動力學及藥效學、力測量值、類器官、管腔及培養基/水凝膠內生物分子之間相互作用之測量值以及膜電位。 In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) can be changes in cell viability, cell proliferation, organoid size, cell morphology, organoid morphology, hydrogel invasiveness, motility, differentiation state, mutational state, karyotype, chromosomal aberration, nucleic acid expression level, protein expression level, nucleic acid modification ( e.g. , methylation), post-translational modification ( e.g. , phosphorylation, ubiquitination and/or glycosylation), activation of a cell signaling pathway, inhibition of a cell signaling pathway, enzyme activity ( e.g. , enzyme cleavage) , chromatin accessibility, histone modifications and other epigenetic changes, physical properties of organoids (including permeability of the gastrointestinal epithelial barrier, pH, oxygen tension, and concentrations of other metabolites in the lumen and basement membrane and secreted factors in the hydrogel and/or medium), concentrations of interleukins and hormones, drug sensitivity, pharmacokinetic and pharmacodynamics of drug absorption and metabolism, force measurements, measurements of biomolecular interactions within the organoid, lumen, and medium/hydrogel, and membrane potential.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為細胞生存力中之變化。In certain embodiments, the change in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be a change in cell viability.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為細胞增生中之變化。In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be changes in cell proliferation.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為類器官尺寸中之變化。In certain embodiments, the variation in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be a variation in organoid size.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為突變狀態中之變化。In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be changes in a mutant state.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為 RNA 表現含量及/或蛋白質表現含量中之變化。In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be changes in RNA expression levels and/or protein expression levels.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為幹細胞及/或增生標記物之 RNA 表現含量及/或蛋白質表現含量中之變化。在某些實施例中,組織來源之上皮類器官、組織來源之上皮類器官之群體或細胞單層中之變化可為幹細胞及/或增生標記物之蛋白質表現含量中之變化。在某些實施例中,組織來源之上皮類器官、組織來源之上皮類器官之群體或細胞單層中之變化可為幹細胞及/或增生標記物之 RNA 表現含量中之變化。例如但不限於,該變化可為 MKI67 表現中之變化、EpCAM 表現中之變化、CD49f 表現中之變化、ASCL2 表現中之變化、CD133 表現中之變化、LGR5 表現中之變化、SOX9 表現中之變化、ALDH1A1 表現中之變化、NEUROG3 表現中之變化、NKX6.1 表現中之變化、SMOC2 表現中之變化、PDX1 表現中之變化、BMI1 表現中之變化及/或 CD44 表現中之變化。在某些實施例中,變化可為 MKI67 表現中之變化。在某些實施例中,變化可為 ASCL2 表現中之變化。在某些實施例中,變化可為 LGR5 表現中之變化。在某些實施例中,變化可為 SOX9 表現中之變化。在某些實施例中,變化可為 SMOC2 表現中之變化。在某些實施例中,變化可以為 CD44 表現中之變化。在某些實施例中,變化可為 EpCAM 表現中之變化。在某些實施例中,變化可為 CD49f 表現中之變化。在某些實施例中,變化可為 CD133 表現中之變化。在某些實施例中,變化可為 ALDH1A1 表現中之變化。在某些實施例中,變化可為 NEUROG3 表現中之變化。在某些實施例中,變化可為 NKX6.1 表現中之變化。在某些實施例中,變化可為 PDX1 表現中之變化。在某些實施例中,變化可為 BMI1 表現中之變化。In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be changes in RNA expression levels and/or protein expression levels of stem cells and/or proliferation markers. In certain embodiments, the changes in a tissue-derived epithelial organoid, a population of tissue-derived epithelial organoids, or a cell monolayer may be changes in protein expression levels of stem cells and/or proliferation markers. In certain embodiments, the changes in a tissue-derived epithelial organoid, a population of tissue-derived epithelial organoids, or a cell monolayer may be changes in RNA expression levels of stem cells and/or proliferation markers. For example, but not limited to, the variation may be a variation in the expression of MKI67, a variation in the expression of EpCAM, a variation in the expression of CD49f, a variation in the expression of ASCL2, a variation in the expression of CD133, a variation in the expression of LGR5, a variation in the expression of SOX9, a variation in the expression of ALDH1A1, a variation in the expression of NEUROG3, a variation in the expression of NKX6.1, a variation in the expression of SMOC2, a variation in the expression of PDX1, a variation in the expression of BMI1, and/or a variation in the expression of CD44. In certain embodiments, the variation may be a variation in the expression of MKI67. In certain embodiments, the variation may be a variation in the expression of ASCL2. In certain embodiments, the variation may be a variation in the expression of LGR5. In certain embodiments, the variation may be a variation in the expression of SOX9. In some embodiments, the variation may be a variation in the expression of SMOC2. In some embodiments, the variation may be a variation in the expression of CD44. In some embodiments, the variation may be a variation in the expression of EpCAM. In some embodiments, the variation may be a variation in the expression of CD49f. In some embodiments, the variation may be a variation in the expression of CD133. In some embodiments, the variation may be a variation in the expression of ALDH1A1. In some embodiments, the variation may be a variation in the expression of NEUROG3. In some embodiments, the variation may be a variation in the expression of NKX6.1. In some embodiments, the variation may be a variation in the expression of PDX1. In some embodiments, the variation may be a variation in the expression of BMI1.
在某些實施例中,組織來源之上皮類器官 (或其細胞)、組織來源之上皮類器官 (或其細胞) 之群體或細胞單層 (或其細胞) 中之變化可為分化標記物之 RNA 表現含量及/或蛋白質表現含量中之變化。在某些實施例中,組織來源之上皮類器官、組織來源之上皮類器官之群體或細胞單層中之變化可為分化標記物之 RNA 表現含量中之變化。在某些實施例中,組織來源之上皮類器官、組織來源之上皮類器官之群體或細胞單層中之變化可為分化標記物之蛋白質表現含量中之變化。例如但不限於,該變化可為 角蛋白 20 (KRT20) 表現中之變化、FABP1 表現中之變化、MUC2 表現中之變化、MUC5B 表現中之變化、MUC5AC 表現中之變化、MUC6 表現中之變化、TFF3 表現中之變化、ALPI 表現中之變化、SI 表現中之變化、CEACAM7 表現中之變化、角蛋白 19 (KRT19) 表現中之變化、角蛋白 7 (KRT7) 表現中之變化、SOX9 表現中之變化、MUC1 表現中之變化、INS 表現中之變化、GCG 表現中之變化、AMY 表現中之變化、ALB 表現中之變化、CYP3A4 表現中之變化、HNF4A 表現中之變化、細胞角蛋白 8 (K8) 表現中之變化、細胞角蛋白 18 (K18) 表現中之變化、細胞角蛋白 5 (K5) 表現中之變化、細胞角蛋白 14 (K14) 表現中之變化及/或平滑肌肌動蛋白 (SMA) 表現中之變化。在某些實施例中,變化可為 KRT20 表現中之變化。在某些實施例中,變化可為 FABP1 表現中之變化。在某些實施例中,變化可為 MUC2 表現中之變化。在某些實施例中,變化可為 MUC5B 表現中之變化。在某些實施例中,變化可為 TFF3 表現中之變化。在某些實施例中,變化可為 ALPI 表現中之變化。在某些實施例中,變化可為 SI 表現中之變化。在某些實施例中,變化可為 CEACAM7 表現中之變化。在某些實施例中,變化可為角蛋白 19 (KRT19) 表現中之變化。在某些實施例中,變化可為角蛋白 7 (KRT7) 表現中之變化。在某些實施例中,變化可為 SOX9 表現中之變化。在某些實施例中,變化可為 MUC1 表現中之變化。在某些實施例中,變化可為 INS 表現中之變化。在某些實施例中,變化可為 GCG 表現中之變化。在某些實施例中,變化可為 AMY 表現中之變化。在某些實施例中,變化可為 ALB 表現中之變化。在某些實施例中,變化可為 CYP3A4 表現中之變化。在某些實施例中,變化可為 HNF4A 表現中之變化。在某些實施例中,變化可為細胞角蛋白 8 (K8) 表現中之變化。在某些實施例中,變化可為細胞角蛋白 18 (K18) 表現中之變化。在某些實施例中,變化可為細胞角蛋白 5 (K5) 表現中之變化。在某些實施例中,變化可為細胞角蛋白 14 (K14) 表現中之變化。在某些實施例中,變化可為平滑肌肌動蛋白 (SMA) 表現中之變化。在某些實施例中,變化可為 MUC5AC 表現中之變化。 在某些實施例中,變化可為 MUC6 表現中之變化。In certain embodiments, the changes in a tissue-derived epithelial organoid (or cells thereof), a population of tissue-derived epithelial organoids (or cells thereof), or a cell monolayer (or cells thereof) may be changes in RNA expression levels and/or protein expression levels of a differentiation marker. In certain embodiments, the changes in a tissue-derived epithelial organoid, a population of tissue-derived epithelial organoids, or a cell monolayer may be changes in RNA expression levels of a differentiation marker. In certain embodiments, the changes in a tissue-derived epithelial organoid, a population of tissue-derived epithelial organoids, or a cell monolayer may be changes in protein expression levels of a differentiation marker. For example, but not limited to, the changes may be changes in keratin 20 (KRT20) expression, changes in FABP1 expression, changes in MUC2 expression, changes in MUC5B expression, changes in MUC5AC expression, changes in MUC6 expression, changes in TFF3 expression, changes in ALPI expression, changes in SI expression, changes in CEACAM7 expression, changes in keratin 19 (KRT19) expression, changes in keratin 7 (KRT7) expression, changes in SOX9 expression, changes in MUC1 expression, changes in INS expression, changes in GCG expression, changes in AMY expression, changes in ALB expression, changes in CYP3A4 expression, changes in HNF4A expression, changes in cellular keratin 8 (K8) expression. In some embodiments, the changes may be changes in the expression of KRT20. In some embodiments, the changes may be changes in the expression of FABP1. In some embodiments, the changes may be changes in the expression of MUC2. In some embodiments, the changes may be changes in the expression of MUC5B. In some embodiments, the changes may be changes in the expression of TFF3. In some embodiments, the changes may be changes in the expression of ALPI. In some embodiments, the changes may be changes in the expression of SI. In certain embodiments, the variation may be a variation in the expression of CEACAM7. In certain embodiments, the variation may be a variation in the expression of keratin 19 (KRT19). In certain embodiments, the variation may be a variation in the expression of keratin 7 (KRT7). In certain embodiments, the variation may be a variation in the expression of SOX9. In certain embodiments, the variation may be a variation in the expression of MUC1. In certain embodiments, the variation may be a variation in the expression of INS. In certain embodiments, the variation may be a variation in the expression of GCG. In certain embodiments, the variation may be a variation in the expression of AMY. In certain embodiments, the variation may be a variation in the expression of ALB. In certain embodiments, the variation may be a variation in the expression of CYP3A4. In certain embodiments, the variation may be a variation in the expression of HNF4A. In certain embodiments, the changes may be changes in the expression of cytokeratin 8 (K8). In certain embodiments, the changes may be changes in the expression of cytokeratin 18 (K18). In certain embodiments, the changes may be changes in the expression of cytokeratin 5 (K5). In certain embodiments, the changes may be changes in the expression of cytokeratin 14 (K14). In certain embodiments, the changes may be changes in the expression of smooth muscle actin (SMA). In certain embodiments, the changes may be changes in the expression of MUC5AC. In certain embodiments, the changes may be changes in the expression of MUC6.
在某些實施例中,為了確定組織來源之上皮類器官、組織來源之上皮類器官之細胞及/或單層或單層細胞之變化,組織來源之上皮類器官培養物或單層培養物可藉由流式細胞分析技術、RNA 及蛋白質表現、類器官及培養基中之細胞介素及代謝物測量值、細胞生存力及增生測定、監測上皮細胞及免疫細胞運動之顯微術、障壁功能、遷移、增生、RNA、蛋白質及細胞器之亞細胞定位、細胞硬度及其他測定來進行分析。組織來源之上皮類器官、組織來源之上皮類器官之細胞、細胞單層及/或細胞單層之細胞可以染料標記或以編碼螢光蛋白及/或螢光素酶之核酸轉染,以使得能夠使用成像及生物發光測定進行細胞之視覺化及定量。In certain embodiments, tissue-derived epithelial organoid cultures or monolayer cultures may be analyzed by flow cytometry, RNA and protein expression, interleukin and metabolite measurements in organoids and culture media, cell viability and proliferation assays, microscopy to monitor epithelial and immune cell motility, barrier function, migration, proliferation, subcellular localization of RNA, proteins and organelles, cell stiffness, and other assays in order to determine changes in tissue-derived epithelial organoids, cells and/or monolayers or monolayers of tissue-derived epithelial organoids. Tissue-derived epithelial organoids, cells of tissue-derived epithelial organoids, cell monolayers, and/or cells of cell monolayers can be labeled with dyes or transfected with nucleic acids encoding fluorescent proteins and/or luciferases to enable visualization and quantification of cells using imaging and bioluminescence assays.
在某些實施例中,本揭露提供了用於鑑定可有效治療疾病之治療劑之方法。在某些實施例中,該方法可包括 (i) 使 (a) 組織來源之上皮類器官 (或其組成物),(b) 組織來源之上皮類器官之群體 (或其組成物),(c) 來源自組織來源之上皮類器官之細胞單層,或 (d) 組織來源之上皮類器官之群體與治療劑接觸,以及 (ii) 分析組織來源之上皮類器官或組織來源之上皮類器官之群體中之變化,該變化指示治療劑之有效性及/或毒性。在某些實施例中,變化指示治療劑之有效性。在某些實施例中,變化指示治療劑之毒性。例如但不限於,如果與未以治療劑治療之組織來源之上皮類器官或組織來源之上皮類器官之細胞之生存力相比,在治療劑存在下組織來源之上皮類器官或組織來源之上皮類器官之細胞之生存力降低,則指示治療劑係有毒的。In certain embodiments, the present disclosure provides methods for identifying therapeutic agents that are effective in treating a disease. In certain embodiments, the method may include (i) contacting (a) a tissue-derived epithelial organoid (or a composition thereof), (b) a population of tissue-derived epithelial organoids (or a composition thereof), (c) a cell monolayer from a tissue-derived epithelial organoid, or (d) a population of tissue-derived epithelial organoids with a therapeutic agent, and (ii) analyzing the tissue-derived epithelial organoid or the population of tissue-derived epithelial organoids for changes that indicate effectiveness and/or toxicity of the therapeutic agent. In certain embodiments, the changes indicate effectiveness of the therapeutic agent. In certain embodiments, the changes indicate toxicity of the therapeutic agent. For example, but not limited to, if the viability of the tissue-derived epithelial organoid or cells of the tissue-derived epithelial organoid is reduced in the presence of the therapeutic agent as compared to the viability of tissue-derived epithelial organoid or cells of the tissue-derived epithelial organoid not treated with the therapeutic agent, then that indicates that the therapeutic agent is toxic.
在某些實施例中,本揭露提供了進行基因體篩選之方法。在某些實施例中,該方法可包括 (i) 提供 (a) 組織來源之上皮類器官 (或其組成物),(b) 組織來源之上皮類器官之群體 (或其組成物),或 (c) 來源自組織來源之上皮類器官之細胞單層,(ii) 在組織來源之上皮類器官或細胞單層之一個或多個細胞之基因體中產生突變,以及 (iii) 分析組織來源之上皮類器官或組織來源之上皮類器官之群體中與突變相關之變化。例如但不限於,如果與參考細胞或組織來源之上皮類器官相比,在具有基因體突變之組織來源之上皮類器官或包括一個或多個細胞之細胞單層中觀察到變化,則這指示該變化為基因體突變之結果。 V. 系統 In certain embodiments, the present disclosure provides methods for performing genomic screening. In certain embodiments, the method may include (i) providing (a) a tissue-derived epithelial organoid (or a composition thereof), (b) a population of tissue-derived epithelial organoids (or a composition thereof), or (c) a cell monolayer from a tissue-derived epithelial organoid, (ii) generating a mutation in the genome of one or more cells of the tissue-derived epithelial organoid or the cell monolayer, and (iii) analyzing the changes associated with the mutation in the tissue-derived epithelial organoid or the population of tissue-derived epithelial organoids. For example, but not limited to, if a change is observed in an epithelial organoid or a cell monolayer comprising one or more cells of a tissue-derived organoid having a genome mutation compared to a reference cell- or tissue-derived epithelial organoid, this indicates that the change is a result of the genome mutation. V. Systems
本揭露進一步提供了使用於本揭露之系統。在某些實施例中,本揭露進一步提供了用於進行目前所揭示之方法之系統。在某些實施例中,本揭露提供了用於產生組織來源之上皮類器官之系統, 例如淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官或滋胚內層類器官。在某些實施例中,本揭露提供了用於鑑定可有效治療疾病之治療劑之系統。在某些實施例中,本揭露提供了用於進行基因體篩選之系統。在某些實施例中,本揭露提供了用於產生上皮細胞模型之系統。 The disclosure further provides systems for use in the disclosure. In certain embodiments, the disclosure further provides systems for performing the methods disclosed herein. In certain embodiments, the disclosure provides systems for generating tissue-derived epithelial organoids, such as tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, or trophoblastic lining organoids. In certain embodiments, the disclosure provides systems for identifying therapeutic agents that are effective in treating diseases. In certain embodiments, the disclosure provides systems for performing genomic screening. In certain embodiments, the present disclosure provides systems for generating epithelial cell models.
在某些實施例中,本揭露之系統可包括能夠進行目前所揭示之方法之機器人及/或自動化組件。在某些實施例中,本揭露之系統包括能夠產生組織來源之上皮類器官之機器人及/或自動化組件。在某些實施例中,本揭露之系統包括一個或多個機器人及/或自動化組件,其用於進行所揭示之用於產生組織來源之上皮類器官之方法之一個或多個步驟。機器人及/或自動化組件之非限制性實例包括自動化液體處理器 ( 例如,液體處理機器人)、3D 列印機、注射泵或其組合。在某些實施例中,本揭露之系統包括一個或多個自動化液體處理器。 In certain embodiments, the systems of the present disclosure may include robots and/or automated components capable of performing the presently disclosed methods. In certain embodiments, the systems of the present disclosure include robots and/or automated components capable of generating tissue-derived epithelial organoids. In certain embodiments, the systems of the present disclosure include one or more robots and/or automated components for performing one or more steps of the disclosed methods for generating tissue-derived epithelial organoids. Non-limiting examples of robots and/or automated components include automated liquid handlers ( e.g. , liquid handling robots), 3D printers, syringe pumps, or combinations thereof. In certain embodiments, the systems of the present disclosure include one or more automated liquid handlers.
在某些實施例中,機器人及/或自動化組件用於進行目前所揭示之方法及/或產生組織來源之上皮類器官以使用於高生產量研究。在某些實施例中,本揭露之系統可包括用於進行高生產量方法之一個或多個機器人及/或自動化組件, 例如,如本文所述。例如但不限於,本揭露之系統可包括一個或多個機器人及/或自動化組件,其可用於進行本文所揭示之使用方法中之任一者。在某些實施例中,本揭露之系統可包括一個或多個機器人及/或自動化組件,其用於篩選藥劑 ( 例如,治療劑),並用於使用所揭示之組織來源之上皮類器官進行基因體篩選。 In certain embodiments, robots and/or automated components are used to perform the presently disclosed methods and/or generate tissue-derived epithelial organoids for use in high-throughput studies. In certain embodiments, the systems of the present disclosure may include one or more robots and/or automated components for performing high-throughput methods, for example , as described herein. For example, but not limited to, the systems of the present disclosure may include one or more robots and/or automated components that can be used to perform any of the methods of use disclosed herein. In certain embodiments, the systems of the present disclosure may include one or more robots and/or automated components for screening agents ( e.g. , therapeutic agents) and for performing genomic screening using the disclosed tissue-derived epithelial organoids.
在某些實施例中,機器人及/或自動化組件能夠使組織來源之上皮類器官培養物傳代。在某些實施例中,自動化機器人可以進行本揭露實例 1 之「類器官維持」中描述之方法, 例如,進行以下步驟中之任一者:添加 TrypLE Express、加熱類器官培養物、研磨類器官培養物以解離細胞、添加 PBS、沉澱細胞、將細胞重懸於 BME 中、冷卻培養物、平鋪細胞、以培養基覆蓋類器官或其組合。在某些實施例中,本揭露之系統可包括一個或多個用於解離細胞之機器人及/或自動化組件。在某些實施例中,本揭露之系統可包括一個或多個機器人及/或自動化組件,其用於 例如在類器官維持過程中或在產生組織來源之上皮類器官過程中進行培養基更換。 In some embodiments, the robot and/or automated component is capable of passaging tissue-derived epithelial organoid cultures. In some embodiments, the automated robot can perform the method described in "Organoid Maintenance" of Example 1 of the present disclosure, for example , performing any of the following steps: adding TrypLE Express, heating the organoid culture, grinding the organoid culture to dissociate cells, adding PBS, sedimenting cells, resuspending cells in BME, cooling the culture, flattening cells, covering the organoid with culture medium, or a combination thereof. In some embodiments, the system of the present disclosure may include one or more robots and/or automated components for dissociating cells. In certain embodiments, the systems disclosed herein may include one or more robots and/or automated components for performing medium changes, for example, during organoid maintenance or during the generation of tissue-derived epithelial organoids.
在某些實施例中,機器人及/或自動化組件能夠產生 BOBA 及/或 SOBA。在某些實施例中,自動化機器人可進行本揭露實例 1 之「經懸浮之水凝膠 BOBA 培養物」及/或「經懸浮之水凝膠 SOBA 碎片培養物」中描述之方法, 例如,進行以下步驟中之任一者:加熱培養基、將類器官細胞-BME 溶液作為小滴分配到溫熱培養基中以產生 BOBA,將類器官細胞-BME 溶液以 X-Y 平面中線形、蛇形或螺旋形運動方式分配到溫熱培養基中以產生 SOBA、研磨 SOBA 絲培養物以產生 SOBA 碎片、更換媒體或其組合。在某些實施例中,本揭露之系統可包括一個或多個機器人及/或自動化組件,其用於將類器官細胞-BME 溶液分配到溫熱培養基中以產生 BOBA 及/或 SOBA。在某些實施例中,本揭露之系統可包括一個或多個液體處理機器人,其用於將類器官細胞-BME 溶液分配到溫熱培養基中以產生 BOBA 及/或 SOBA。在某些實施例中,本揭露之系統可包括用於進行媒介物更換之一個或多個機器人及/或自動化組件。 In some embodiments, the robot and/or automated component can produce BOBA and/or SOBA. In some embodiments, the automated robot can perform the method described in "suspended hydrogel BOBA culture" and/or "suspended hydrogel SOBA fragment culture" of Example 1 of the present disclosure, for example , performing any of the following steps: heating the culture medium, dispensing the organoid cell-BME solution as droplets into the warm culture medium to produce BOBA, dispensing the organoid cell-BME solution into the warm culture medium in a linear, serpentine or spiral motion in the XY plane to produce SOBA, grinding the SOBA filament culture to produce SOBA fragments, changing the medium or a combination thereof. In certain embodiments, the systems of the present disclosure may include one or more robots and/or automated components for dispensing the organoid cell-BME solution into the warm culture medium to produce BOBA and/or SOBA. In certain embodiments, the systems of the present disclosure may include one or more liquid handling robots for dispensing the organoid cell-BME solution into the warm culture medium to produce BOBA and/or SOBA. In certain embodiments, the systems of the present disclosure may include one or more robots and/or automated components for performing medium replacement.
在某些實施例中,本揭露之系統可包括組織來源之上皮類器官、組織來源之上皮類器官之群體及/或包括組織來源之上皮類器官之組成物, 例如淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官或滋胚內層類器官。在某些實施例中,組織來源之上皮類器官選自由以下所組成之群組:肺類器官、胃腸類器官、肝類器官、胰臟類器官、乳腺類器官及其組合。在某些實施例中,系統包括由一個或多個組織來源之上皮幹細胞產生之組織來源之上皮類器官。在某些實施例中,組織來源之上皮類器官以水凝膠形式提供。在某些實施例中,組織來源之上皮類器官可嵌入在水凝膠中並懸浮在培養基中。組織來源之上皮類器官 (或其組成物) 或產生此等組織來源之上皮類器官之方法之非限制性實例分別在第 II 節及第 III 節中揭示。在某些實施例中,組織來源之上皮類器官可提供在容器中, 例如培養容器。在某些實施例中,組織來源之上皮類器官可提供在培養容器中, 例如燒瓶及/或多孔盤。 In certain embodiments, the disclosed systems may include tissue-derived epithelial organoids, groups of tissue-derived epithelial organoids, and/or compositions comprising tissue-derived epithelial organoids, such as tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, or trophoblastic lining organoids. In certain embodiments, the tissue-derived epithelial organoids are selected from the group consisting of lung organoids, gastrointestinal organoids, liver organoids, pancreatic organoids, breast organoids, and combinations thereof. In some embodiments, the system includes tissue-derived epithelial organoids produced from one or more tissue-derived epithelial stem cells. In some embodiments, the tissue-derived epithelial organoids are provided in the form of a hydrogel. In some embodiments, the tissue-derived epithelial organoids may be embedded in a hydrogel and suspended in a culture medium. Non-limiting examples of tissue-derived epithelial organoids (or components thereof) or methods of producing such tissue-derived epithelial organoids are disclosed in Sections II and III, respectively. In some embodiments, the tissue-derived epithelial organoids may be provided in a container, such as a culture container. In some embodiments, the tissue-derived epithelial organoids may be provided in a culture container, such as a flask and/or a multiwell dish.
在某些實施例中,本揭露之系統可進一步包括用於使用組織來源之上皮類器官來確定待測試之治療劑是否有效治療疾病之說明。在某些實施例中,本揭露之系統可進一步包括用於使用組織來源之上皮類器官來確定基因體突變之影響之說明。在某些實施例中,本揭露之系統可進一步包括用於產生上皮細胞模型之說明。In some embodiments, the systems of the present disclosure may further include instructions for using tissue-derived epithelial organoids to determine whether a therapeutic agent to be tested is effective in treating a disease. In some embodiments, the systems of the present disclosure may further include instructions for using tissue-derived epithelial organoids to determine the effects of genomic mutations. In some embodiments, the systems of the present disclosure may further include instructions for generating epithelial cell models.
在某些非限制性實施例中,本揭露之系統可進一步包括一種或多種藥劑及其他組分 ( 例如,染料、抗體、引體、探針等) 以確定變化, 例如蛋白質或核酸表現、組織來源之上皮類器官、組織來源之上皮類器官之細胞、細胞單層或細胞單層之細胞中之變化。此等變化之非限制性實例在第 III 節中揭示。在某些實施例中,本揭露之系統可包括一個或多個機器人及/或自動化組件,其用於分析組織來源之上皮類器官、組織來源之上皮類器官之細胞、細胞單層或細胞單層之細胞中之變化。 VI. 示例性實施例A. 目前所揭示之主題提供了一種產生組織來源之上皮類器官之方法,該方法包含: a) 使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物; b) 將該水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物;以及 c) 將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。 A1.如 A 之方法,其中使複數個組織來源之上皮幹細胞與該水凝膠接觸以產生該水凝膠-組織來源之上皮幹細胞混合物。 A2.如 A1 之方法,其中該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。 A3.如 A1 或 A2 之方法,其中該等複數個組織來源之上皮幹細胞包含在組織碎片、類器官碎片或其組合內。 A4.如 A 至 A2 之方法,其中該組織來源之上皮幹細胞或該等複數個組織來源之上皮幹細胞分離自原生上皮組織。 A5.如 A 至 A4 中任一項之方法,其中該水凝膠係在與該培養基接觸時固化。 A6.如 A 至 A5 中任一項之方法,其中將該水凝膠-組織來源之上皮幹細胞混合物懸浮在該培養基中包含將含有該水凝膠-組織來源之上皮幹細胞混合物之分配裝置浸沒在該培養基中並將該水凝膠-組織來源之上皮幹細胞混合物分配到該培養基中。 A7.如 A 至 A6 中任一項之方法,其中該培養基之溫度為約 25℃ 至約 50℃。 A7-1.如 A 至 A7 中任一項之方法,其中該培養基之溫度為約 30℃ 至約 50℃。 A8.如 A 至 A7-1 中任一項之方法,其中該水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 25℃。 A8-1.如 A 至 A8 中任一項之方法,其中該水凝膠-組織來源之上皮幹細胞混合物之溫度為約 2℃ 至約 20℃。 A9.如 A 至 A8-1 中任一項之方法,其中該水凝膠係選自由以下所組成之群組:合成水凝膠、天然水凝膠及其組合。 A10.如 A9 之方法,其中該天然水凝膠包含基底膜萃取物 (BME) 或細胞外基質 (ECM) 成分。 A11.如 A 至 A10 中任一項之方法,其中該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。 A12.如 A 至 A11 中任一項之方法,其中該經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有包含大於約 0.1 mm 之長度、寬度及/或直徑之幾何形狀。 A13.如 A12 之方法,其中該經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有包含約 0.1 mm 至約 1,000 mm 之長度、寬度及/或直徑之幾何形狀。 A14.如 A 至 A13 中任一項之方法,其中該水凝膠-組織來源之上皮幹細胞混合物係以小滴懸浮在該培養基中。 A15.如 A 至 A13 中任一項之方法,其中該經懸浮之水凝膠-組織來源之上皮幹細胞混合物具有絲狀結構。 A16.如 A15 之方法,其中該絲狀結構具有線形、蛇形或螺旋形形狀。 A17.如 A 至 A16 中任一項之方法,其進一步包含將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。 B. 一種產生組織來源之上皮類器官之懸浮培養物之方法,該方法包含: a) 將包含水凝膠及組織來源之上皮幹細胞之混合物引入到培養基中以產生經懸浮之混合物;以及 b) 將該經懸浮之混合物在該培養基中培養以產生呈懸浮狀態之該等組織來源之上皮類器官。 B1.如 B 之方法,其中引入到該培養基中之該混合物包含該水凝膠及複數個組織來源之上皮幹細胞。 B2.如 B1 之方法,其中該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。 B3.如 B1 或 B2 之方法,其中該等複數個組織來源之上皮幹細胞包含在組織碎片、類器官碎片或其組合內。 B4.如 B 至 B2 中任一項之方法,其中該組織來源之上皮幹細胞或該等複數個組織來源之上皮幹細胞分離自原生上皮組織。 B5.如 B 至 B4 中任一項之方法,其中該水凝膠係在與該培養基接觸時固化。 B6.如 B 至 B5 中任一項之方法,其中將混合物引入該培養基中包含將含有該混合物之分配裝置浸沒在該培養基中並將該混合物分配到該培養基中。 B7.如 B 至 B6 中任一項之方法,其中該培養基之溫度為約 25℃ 至約 50℃。 B7-1.如 B 至 B7 中任一項之方法,其中該培養基之溫度為約 30℃ 至約 50℃。 B8.如 B 至 B7-1 中任一項之方法,其中該混合物之溫度為約 2℃ 至約 25℃。 B8-1.如 B 至 B8 中任一項之方法,其中該混合物之溫度為約 2℃ 至約 20℃。 B9.如 B 至 B8-1 中任一項之方法,其中該水凝膠係選自由以下所組成之群組:合成水凝膠、天然水凝膠及其組合。 B10.如 B9 之方法,其中該天然水凝膠包含基底膜萃取物 (BME) 或細胞外基質 (ECM) 成分。 B11.如 B 至 B10 中任一項之方法,其中該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。 B12.如 B 至 B11 中任一項之方法,其中該經懸浮之混合物具有包含大於約 0.1 mm 之長度、寬度及/或直徑之幾何形狀。 B13.如 B12 之方法,其中該經懸浮之混合物具有包含約 0.1 mm 至約 1,000 mm 之長度、寬度及/或直徑之幾何形狀。 B14.如 B 至 B13 中任一項之方法,其中該混合物係以小滴引入到該培養基中。 B15.如 B 至 B3 中任一項之方法,其中該混合物係以絲狀結構形式引入到該培養基中。 B16.如 B15 之方法,其中該絲狀結構具有線形、蛇形或螺旋形形狀。 B17.如 B 至 B16 中任一項之方法,其進一步包含將該經懸浮之混合物碎片化以產生包含該組織來源之上皮類器官之碎片化結構。 B18.如 A 至 B17 中任一項之方法,其中該培養基存在於容器中,該容器選自由以下所組成之群組:培養皿、多孔盤、錐形管、貯器、培養袋、生物反應器或燒瓶。 C. 一種產生組織來源之上皮類器官之懸浮培養物之方法,該方法包含: a) 使組織來源之上皮幹細胞與水凝膠接觸以產生水凝膠-組織來源之上皮幹細胞混合物; b) 將該水凝膠-組織來源之上皮幹細胞混合物沉積到基材上; c) 將該水凝膠-組織來源之上皮幹細胞混合物固化以產生經固化之水凝膠-組織來源之上皮幹細胞混合物; d) 將該經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在培養基中以產生經懸浮之水凝膠-組織來源之上皮幹細胞混合物;以及 e) 將該經懸浮之水凝膠-組織來源之上皮幹細胞混合物在該培養基中培養以產生組織來源之上皮類器官。 C1.如 C 之方法,其中使複數個組織來源之上皮幹細胞與該水凝膠接觸以產生該水凝膠-組織來源之上皮幹細胞混合物。 C2.如 C1 之方法,其中該等複數個組織來源之上皮幹細胞包含約 1 × 10 4個組織來源之上皮幹細胞/ml 水凝膠至約 1 × 10 7個組織來源之上皮幹細胞/ml 水凝膠。 C3.如 C1 或 C2 之方法,其中該等複數個組織來源之上皮幹細胞包含在組織碎片、類器官碎片或其組合內。 C4.如 C 至 C2 中任一項之方法,其中該組織來源之上皮幹細胞或該等複數個組織來源之上皮幹細胞分離自原生上皮組織。 C5.如 C 至 C3 中任一項之方法,其進一步包含在將該經固化之水凝膠-組織來源之上皮幹細胞混合物懸浮在該培養基中之前,將該經固化之水凝膠-組織來源之上皮幹細胞混合物從該基材中取出。 C6.如 C 至 C4 中任一項之方法,其中該水凝膠係選自由以下所組成之群組:合成水凝膠、天然水凝膠及其組合。 C7.如 C6 之方法,其中該天然水凝膠包含基底膜萃取物 (BME) 或細胞外基質 (ECM) 成分。 C8.如 C 至 C7 中任一項之方法,其中該水凝膠具有等於或大於損耗模數 G'' 之儲存模數 G'。 C9.如 C 至 C8 中任一項之方法,其中該經固化之水凝膠-組織來源之上皮幹細胞混合物具有包含大於約 0.1 mm 之長度、寬度及/或直徑之幾何形狀。 C10.如 C9 之方法,其中該經固化之水凝膠-組織來源之上皮幹細胞混合物具有包含約 0.1 mm 至約 1,000 mm 之長度、寬度及/或直徑之幾何形狀。 C11.如 C 至 C10 中任一項之方法,其中將該水凝膠-組織來源之上皮幹細胞混合物作為小滴沉積到該基材上。 C12.如 C 至 C10 中任一項之方法,其中將該水凝膠-組織來源之上皮幹細胞混合物沉積到該基材上以具有絲狀結構。 C13.如 C12 之方法,其中該絲狀結構具有線形、蛇形或螺旋形形狀。 C14.如 C 至 C13 中任一項之方法,其進一步包含將該水凝膠-組織來源之上皮幹細胞混合物在該培養基中碎片化以產生包含該組織來源之上皮類器官之碎片化結構。 C15.如 C 至 C14 中任一項之方法,其中與參考組織來源之上皮類器官相比,該組織來源之上皮類器官具有均勻形態,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 C16.如 C15 之方法,其中該組織來源之上皮類器官具有均勻尺寸。 C17.如 C16 之方法,其中與該等參考組織來源之上皮類器官相比,該組織來源之上皮類器官之平均直徑更均勻。 C18.如 A 至 C17 中任一項之方法,其中與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在該等組織來源之上皮類器官之群體中以較高含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 C19.如 C18 之方法,其中該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。 C20.如 A 至 C18 中任一項之方法,其中與參考組織來源之上皮類器官之群體相比,分化標記物在該等組織來源之上皮類器官之群體中以較低含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 C21.如 C20 之方法,其中該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。 D. 一種組織來源之上皮類器官,其藉由如 A 至 C21 中任一項之方法產生。 D1.如 D 之組織來源之上皮類器官,其中該組織來源之上皮類器官係選自由以下所組成之群組:淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官、滋胚內層類器官及其組合。 E. 一種組成物,其包含組織來源之上皮類器官及培養基,其中該組織來源之上皮類器官嵌入在懸浮於該培養基中之水凝膠內。 E1.如 E 之組成物,其中該水凝膠具有包含大於約 0.1 mm 之長度、寬度及/或直徑之幾何形狀。 E2.如 E1 之組成物,其中該水凝膠具有包含約 0.1 mm 至約 1,000 mm 之長度、寬度及/或直徑之幾何形狀。 E3.如 E 至 E2 中任一項之組成物,其中該水凝膠為小滴。 E4.如 E 至 E2 中任一項之組成物,其中該水凝膠具有絲狀結構。 E5.如 E4 之組成物,其中該絲狀結構具有線形、蛇形或螺旋形形狀。 E6.如 E 至 E5 中任一項之組成物,其中與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在該等組織來源之上皮類器官之群體中以較高含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 E7.如 E6 之組成物,其中該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。 E8.如 E 至 E5 中任一項之組成物,其中與參考組織來源之上皮類器官之群體相比,分化標記物在該等組織來源之上皮類器官之群體中以較低含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 E9.如 E8 之組成物,其中該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。 F. 一種篩選藥劑之方法,該方法包含: a) 使如 D 或 D1 之組織來源之上皮類器官或組織來源之上皮類器官之群體或如 E 至 E9 中任一項之組成物與藥劑接觸;以及 b) 分析該組織來源之上皮類器官或該等組織來源之上皮類器官之群體中指示該藥劑之有效性及/或毒性之變化。 F1.如 F 之方法,其中使該藥劑與該組織來源之上皮類器官或該等組織來源之上皮類器官之群體接觸約 1 分鐘至約 3 年。 F1-1.如 F1 之方法,其中使該藥劑與該組織來源之上皮類器官或該等組織來源之上皮類器官之群體接觸約 15 分鐘至約 3 年。 F2.如 F、F1 或 F1-1 之方法,其中該藥劑為治療劑。 F3.如 F2 之方法,其中該治療劑為基於多肽之治療劑、小分子治療劑、細胞治療劑、基因編輯系統、基於核酸之治療劑或其組合。 G. 一種進行基因體篩選之方法,該方法包含: a) 提供如 D 或 D1 之組織來源之上皮類器官或組織來源之上皮類器官之群體或如 E 至 E9 中任一項之組成物; b) 在該組織來源之上皮類器官之一個或多個細胞之基因體中產生突變;以及 c) 分析該組織來源之上皮類器官或該組織來源之上皮類器官之群體的與該突變相關之變化。 G1.如 G 之方法,其中該突變係使用基因調節系統產生的。 G2.如 G1 之方法,其中該基因調節系統為基因編輯系統。 G3.如 G2 之方法,其中該基因編輯系統為 CRISPR 系統。 G4.如 F 至 G2 中任一項之方法,其中該變化為性質的變化,該性質選自由以下所組成之群組:細胞生存力、細胞代謝、氧化還原電位、細胞增生、細胞形態、類器官形態、類器官尺寸、蛋白質表現含量、核酸表現含量、核酸修飾、轉譯後修飾、細胞傳訊路徑之活化、細胞傳訊路徑之壓制、酵素活性、障壁完整性及其組合。 H. 一種用於產生上皮細胞模型之方法,該方法包含: a) 提供如 D 之組織來源之上皮類器官或組織來源之上皮類器官之群體; b) 將該組織來源之上皮類器官或該組織來源之上皮類器官之群體消化成單細胞;以及 c) 將該等單細胞在培養基中培養以產生細胞單層。 H1.如 H 之方法,其中將該單細胞在可滲透細胞培養內件上培養。 H2.如 H 或 H1 之方法,其中該培養基為分化培養基。 H3.如 H 或 H1 之方法,其中該培養基為細胞生長或幹細胞促進培養基。 I. 一種篩選藥劑之方法,該方法包含: a) 使藉由如 H 至 H3 中任一項之方法產生之細胞單層與藥劑接觸;以及 b) 分析該細胞單層的指示該藥劑之有效性、流佈及/或毒性之變化。 I1.如 I 之方法,其中使該藥劑與該細胞單層接觸約 1 分鐘至約 3 年。 I1-1.如 I1 之方法,其中使該藥劑與該細胞單層接觸約 15 分鐘至約 3 年。 I2.如 I、I1 或 I1-1 之方法,其中該藥劑為治療劑。 I3.如 I2 之方法,其中該治療劑為基於多肽之治療劑、小分子治療劑、細胞治療劑、基因編輯系統、基於核酸之治療劑或其組合。 J. 一種進行基因體篩選之方法,該方法包含: a) 提供藉由如 H 至 H3 中任一項之方法產生之細胞單層; b) 在該細胞單層之一個或多個細胞之基因體中產生突變;以及 c) 分析該細胞單層的與該突變相關之變化。 J1.如 J 之方法,其中該突變係使用基因調節系統產生的。 J2.如 J1 之方法,其中該基因調節系統為基因編輯系統。 J3.如 J2 之方法,其中該基因編輯系統為 CRISPR 系統。 J4.如 J 至 J3 中任一項之方法,其中該變化為性質的變化,該性質選自由以下所組成之群組:細胞生存力、細胞代謝、氧化還原電位、細胞增生、細胞形態、類器官形態、類器官尺寸、蛋白質表現含量、核酸表現含量、核酸修飾、轉譯後修飾、細胞傳訊路徑之活化、細胞傳訊路徑之壓制、酵素活性、障壁完整性及其組合。 J1.如 A 至 C21 及 F 至 J4 中任一項之方法或如 E 至 E9 中任一項之組成物,其中該組織碎片為來自組織之碎片,該組織係選自由以下所組成之群組:淚腺、扁桃腺、唾液腺、胃腸組織、甲狀腺、肺、乳腺、肝、膽管、胃、腎、胰臟、子宮內膜、輸卵管、子宮頸、前列腺、膀胱、味蕾、卵巢、胎盤及其組合。 J5.如 A 至 C21 及 F 至 J4 中任一項之方法或如 E 至 E9 中任一項之組成物,其中該組織來源之上皮幹細胞係獲自類器官之碎片,該類器官係選自由以下所組成之群組:淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官、滋胚內層類器官及其組合。 K. 一種用於培養組織來源之上皮類器官之系統,該系統包含組織來源之上皮類器官及培養基,其中該組織來源之上皮類器官嵌入在懸浮於該培養基中之水凝膠內。 K1.如 K 之系統,其中該組織來源之上皮類器官為腸類器官。 K2.如 K 或 K1 之系統,其中該水凝膠具有包含大於約 0.1 mm 之長度、寬度及/或直徑之幾何形狀。 K3.如 K 至 K2 中任一項之系統,其中該水凝膠具有包含約 0.1 mm 至約 1,000 mm 之長度、寬度及/或直徑之幾何形狀。 K4.如 K 至 K3 中任一項之系統,其中該水凝膠為小滴。 K5.如 K 至 K3 中任一項之系統,其中該水凝膠具有絲狀結構。 K6.如 K5 之系統,其中該絲狀結構具有線形、蛇形或螺旋形形狀。 K7.如 K 至 K6 中任一項之系統,其中與參考組織來源之上皮類器官之群體相比,幹細胞及/或增生標記物在該等組織來源之上皮類器官之群體中以較高含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 K8.如 K7 之系統,其中該幹細胞及/或增生標記物選自由以下所組成之群組:MKI67、EpCAM、BMI1、CD49f、ASCL2、CD133、LGR5、SOX9、ALDH1A1、NEUROG3、NKX6.1、SMOC2、PDX1、CD44 及其組合。 K9.如 K 至 K6 中任一項之系統,其中與參考組織來源之上皮類器官之群體相比,分化標記物在該等組織來源之上皮類器官之群體中以較低含量表現,其中該等參考組織來源之上皮類器官為嵌入在接附至基材之水凝膠內之組織來源之上皮類器官。 K10.如 K9 之系統,其中該分化標記物選自由以下所組成之群組:角蛋白 20 (KRT20)、FABP1、MUC2、MUC5B、MUC5AC、MUC6、TFF3、ALPI、SI、CEACAM7、角蛋白 19 (KRT19)、角蛋白 7 (KRT7)、SOX9、MUC1、INS、GCG、AMY、ALB、CYP3A4、HNF4A、細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角蛋白 5 (K5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA) 及其組合。 K11.如 K 至 K10 中任一項之系統,其中該組織來源之上皮類器官係選自由以下所組成之群組:淚腺類器官、扁桃腺類器官、唾液腺類器官、胃腸類器官、甲狀腺類器官、肺類器官、乳腺類器官、肝類器官、膽管類器官、胃類器官、腎類器官、胰臟類器官、子宮內膜類器官、輸卵管類器官、子宮頸類器官、前列腺類器官、膀胱類器官、卵巢類器官、味蕾類器官、滋胚內層類器官及其組合。 K12.如 K 至 K11 中任一項之系統,其進一步包含機器人及/或自動化組件。 K13.如 K12 之系統,其中該機器人及/或自動化組件包含液體處理機器人、3D 列印機、注射泵或其組合。 L. 如 A 至 C21 及 F 至 J5 中任一項之方法,其中該方法之一個或多個步驟藉由機器人及/或自動化組件進行。 L1.如 L 之方法,其中該機器人及/或自動化組件包含液體處理機器人、3D 列印機、注射泵或其組合。 L2.如 L 或 L1 之方法,其中該機器人及/或自動化組件為液體處理機器人。 M. 一種產生如 D 至 D1 之組織來源之上皮類器官或如 E 至 E9 之組成物之方法,其中該方法之一個或多個步驟藉由機器人及/或自動化組件進行。 M1.如 M 之方法,其中該機器人及/或自動化組件包含液體處理機器人、3D 列印機、注射泵或其組合。 N. 如 B 至 B18 中任一項之方法,其中將包含水凝膠及組織來源之上皮幹細胞之混合物引入到培養基中以產生經懸浮之混合物藉由機器人及/或自動化組件進行。 N1.如 B 至 B18 中任一項之方法,其中將經懸浮之混合物在該培養基中培養以產生呈懸浮狀態之該等組織來源之上皮類器官藉由機器人及/或自動化組件進行。 N2.如 N 或 N1 之方法,其中該機器人及/或自動化組件包含液體處理機器人、3D 列印機、注射泵或其組合。 N3.如 N2 之方法,其中該機器人及/或自動化組件包含一個或多個液體處理機器人。 實例 In certain non-limiting embodiments, the systems of the present disclosure may further include one or more agents and other components ( e.g. , dyes, antibodies, primers, probes, etc.) to determine changes, such as protein or nucleic acid expression, changes in tissue-derived epithelial organoids, cells of tissue-derived epithelial organoids, cell monolayers, or cells of cell monolayers. Non-limiting examples of such changes are disclosed in Section III. In certain embodiments, the systems of the present disclosure may include one or more robots and/or automated components for analyzing changes in tissue-derived epithelial organoids, cells of tissue-derived epithelial organoids, cell monolayers, or cells of cell monolayers. VI. Exemplary Embodiments A. The presently disclosed subject matter provides a method for producing tissue-derived epithelial organoids, the method comprising: a) contacting tissue-derived epithelial stem cells with hydrogel to produce a hydrogel-tissue-derived epithelial stem cell mixture; b) suspending the hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue-derived epithelial stem cell mixture; and c) culturing the suspended hydrogel-tissue-derived epithelial stem cell mixture in the culture medium to produce tissue-derived epithelial organoids. A1. The method of A, wherein a plurality of tissue-derived epithelial stem cells are contacted with the hydrogel to produce the hydrogel-tissue-derived epithelial stem cell mixture. A2. The method of A1, wherein the plurality of tissue-derived epithelial stem cells comprise about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel. A3. The method of A1 or A2, wherein the plurality of tissue-derived epithelial stem cells are contained in tissue fragments, organoid fragments, or a combination thereof. A4. The method of A to A2, wherein the tissue-derived epithelial stem cell or the plurality of tissue-derived epithelial stem cells are isolated from native epithelial tissue. A5. The method of any one of A to A4, wherein the hydrogel solidifies when in contact with the culture medium. A6. The method of any one of A to A5, wherein suspending the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium comprises immersing a dispensing device containing the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium and dispensing the hydrogel-tissue-derived epithelial stem cell mixture into the culture medium. A7. The method of any one of A to A6, wherein the temperature of the culture medium is about 25°C to about 50°C. A7-1. The method of any one of A to A7, wherein the temperature of the culture medium is about 30°C to about 50°C. A8. The method of any one of A to A7-1, wherein the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is about 2°C to about 25°C. A8-1. The method of any one of A to A8, wherein the temperature of the hydrogel-tissue-derived epithelial stem cell mixture is about 2°C to about 20°C. A9. The method of any one of A to A8-1, wherein the hydrogel is selected from the group consisting of: synthetic hydrogels, natural hydrogels, and combinations thereof. A10. The method of A9, wherein the natural hydrogel comprises basement membrane extract (BME) or extracellular matrix (ECM) components. A11. The method of any one of A to A10, wherein the hydrogel has a storage modulus G' equal to or greater than the loss modulus G''. A12. The method of any one of A to A11, wherein the suspended hydrogel-tissue-derived epithelial stem cell mixture has a geometric shape comprising a length, width and/or diameter greater than about 0.1 mm. A13. The method of A12, wherein the suspended hydrogel-tissue derived epithelial stem cell mixture has a geometric shape comprising a length, width and/or diameter of about 0.1 mm to about 1,000 mm. A14. The method of any one of A to A13, wherein the hydrogel-tissue derived epithelial stem cell mixture is suspended in the culture medium in small drops. A15. The method of any one of A to A13, wherein the suspended hydrogel-tissue derived epithelial stem cell mixture has a filamentous structure. A16. The method of A15, wherein the filamentous structure has a linear, serpentine or spiral shape. A17. The method of any one of A to A16, further comprising fragmenting the suspended hydrogel-tissue-derived epithelial stem cell mixture to produce a fragmented structure comprising the tissue-derived epithelial organoid. B. A method for producing a suspended culture of tissue-derived epithelial organoids, the method comprising: a) introducing a mixture comprising hydrogel and tissue-derived epithelial stem cells into a culture medium to produce a suspended mixture; and b) culturing the suspended mixture in the culture medium to produce the tissue-derived epithelial organoids in a suspended state. B1. The method of B, wherein the mixture introduced into the culture medium comprises the hydrogel and a plurality of tissue-derived epithelial stem cells. B2. The method of B1, wherein the plurality of tissue-derived epithelial stem cells comprises about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel. B3. The method of B1 or B2, wherein the plurality of tissue-derived epithelial stem cells are contained in tissue fragments, organoid fragments, or a combination thereof. B4. The method of any one of B to B2, wherein the tissue-derived epithelial stem cell or the plurality of tissue-derived epithelial stem cells are isolated from native epithelial tissue. B5. The method of any one of B to B4, wherein the hydrogel solidifies upon contact with the culture medium. B6. The method of any one of B to B5, wherein introducing the mixture into the culture medium comprises immersing a dispensing device containing the mixture in the culture medium and dispensing the mixture into the culture medium. B7. The method of any one of B to B6, wherein the temperature of the culture medium is about 25°C to about 50°C. B7-1. The method of any one of B to B7, wherein the temperature of the culture medium is about 30°C to about 50°C. B8. The method of any one of B to B7-1, wherein the temperature of the mixture is about 2°C to about 25°C. B8-1. The method of any one of B to B8, wherein the temperature of the mixture is about 2°C to about 20°C. B9. The method of any one of B to B8-1, wherein the hydrogel is selected from the group consisting of: synthetic hydrogels, natural hydrogels, and combinations thereof. B10. The method of B9, wherein the natural hydrogel comprises basement membrane extract (BME) or extracellular matrix (ECM) components. B11. The method of any one of B to B10, wherein the hydrogel has a storage modulus G' that is equal to or greater than the loss modulus G''. B12. The method of any one of B to B11, wherein the suspended mixture has a geometric shape comprising a length, width and/or diameter greater than about 0.1 mm. B13. The method of B12, wherein the suspended mixture has a geometric shape comprising a length, width and/or diameter from about 0.1 mm to about 1,000 mm. B14. The method of any one of B to B13, wherein the mixture is introduced into the culture medium as a droplet. B15. The method of any one of B to B3, wherein the mixture is introduced into the culture medium in the form of a filamentary structure. B16. The method of B15, wherein the filamentary structure has a linear, serpentine or spiral shape. B17. The method of any one of B to B16, further comprising fragmenting the suspended mixture to produce fragmented structures comprising the tissue-derived epithelial organoid. B18. The method of any one of A to B17, wherein the culture medium is present in a container selected from the group consisting of a culture dish, a multi-well dish, a conical tube, a container, a culture bag, a bioreactor or a flask. C. A method for generating a suspension culture of tissue-derived epithelial organoids, the method comprising: a) contacting tissue-derived epithelial stem cells with hydrogel to generate a hydrogel-tissue-derived epithelial stem cell mixture; b) depositing the hydrogel-tissue-derived epithelial stem cell mixture onto a substrate; c) solidifying the hydrogel-tissue-derived epithelial stem cell mixture to generate a solidified hydrogel-tissue-derived epithelial stem cell mixture; d) suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in a culture medium to produce a suspended hydrogel-tissue-derived epithelial stem cell mixture; and e) culturing the suspended hydrogel-tissue-derived epithelial stem cell mixture in the culture medium to produce tissue-derived epithelial organoids. C1. The method of C, wherein a plurality of tissue-derived epithelial stem cells are contacted with the hydrogel to produce the hydrogel-tissue-derived epithelial stem cell mixture. C2. The method of C1, wherein the plurality of tissue-derived epithelial stem cells comprises about 1 × 10 4 tissue-derived epithelial stem cells/ml hydrogel to about 1 × 10 7 tissue-derived epithelial stem cells/ml hydrogel. C3. The method of C1 or C2, wherein the plurality of tissue-derived epithelial stem cells are contained in tissue fragments, organoid fragments, or a combination thereof. C4. The method of any one of C to C2, wherein the tissue-derived epithelial stem cells or the plurality of tissue-derived epithelial stem cells are isolated from native epithelial tissue. C5. The method of any one of C to C3, further comprising removing the solidified hydrogel-tissue-derived epithelial stem cell mixture from the substrate before suspending the solidified hydrogel-tissue-derived epithelial stem cell mixture in the culture medium. C6. The method of any one of C to C4, wherein the hydrogel is selected from the group consisting of: synthetic hydrogels, natural hydrogels and combinations thereof. C7. The method of C6, wherein the natural hydrogel comprises basement membrane extract (BME) or extracellular matrix (ECM) components. C8. The method of any one of C to C7, wherein the hydrogel has a storage modulus G' that is equal to or greater than the loss modulus G''. C9. The method of any one of C to C8, wherein the cured hydrogel-tissue derived epithelial stem cell mixture has a geometry comprising a length, width and/or diameter greater than about 0.1 mm. C10. The method of C9, wherein the cured hydrogel-tissue derived epithelial stem cell mixture has a geometry comprising a length, width and/or diameter of about 0.1 mm to about 1,000 mm. C11. The method of any one of C to C10, wherein the hydrogel-tissue derived epithelial stem cell mixture is deposited onto the substrate as a droplet. C12. The method of any one of C to C10, wherein the hydrogel-tissue-derived epithelial stem cell mixture is deposited on the substrate to have a filamentous structure. C13. The method of C12, wherein the filamentous structure has a linear, serpentine or spiral shape. C14. The method of any one of C to C13, further comprising fragmenting the hydrogel-tissue-derived epithelial stem cell mixture in the culture medium to produce a fragmented structure comprising the tissue-derived epithelial organoid. C15. The method of any one of C to C14, wherein the tissue-derived epithelial organoid has a uniform morphology compared to a reference tissue-derived epithelial organoid, wherein the reference tissue-derived epithelial organoid is a tissue-derived epithelial organoid embedded in a hydrogel attached to a substrate. C16. The method of C15, wherein the tissue-derived epithelial organoid has a uniform size. C17. The method of C16, wherein the average diameter of the tissue-derived epithelial organoid is more uniform than that of the reference tissue-derived epithelial organoid. C18. The method of any one of A to C17, wherein the stem cell and/or proliferation marker is expressed at a higher level in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. C19. The method of C18, wherein the stem cell and/or proliferation marker is selected from the group consisting of: MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. C20. The method of any one of A to C18, wherein the differentiation marker is expressed at a lower level in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. C21. The method of C20, wherein the differentiation marker is selected from the group consisting of: keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA) and combinations thereof. D. A tissue-derived epithelial organoid produced by the method of any one of A to C21. D1. The tissue-derived epithelial organoid of D, wherein the tissue-derived epithelial organoid is selected from the group consisting of tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, trophoblastic lining organoids, and combinations thereof. E. A composition comprising tissue-derived epithelial organoids and a culture medium, wherein the tissue-derived epithelial organoid is embedded in a hydrogel suspended in the culture medium. E1. The composition of E, wherein the hydrogel has a geometry comprising a length, width and/or diameter greater than about 0.1 mm. E2. The composition of E1, wherein the hydrogel has a geometry comprising a length, width and/or diameter from about 0.1 mm to about 1,000 mm. E3. The composition of any one of E to E2, wherein the hydrogel is a droplet. E4. The composition of any one of E to E2, wherein the hydrogel has a filamentous structure. E5. The composition of E4, wherein the filamentous structure has a linear, serpentine or helical shape. E6. The composition of any one of E to E5, wherein the stem cell and/or proliferation marker is expressed at a higher level in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. E7. The composition of E6, wherein the stem cell and/or proliferation marker is selected from the group consisting of: MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. E8. The composition of any of E to E5, wherein a differentiation marker is expressed at a lower level in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are embedded in a hydrogel attached to a substrate. E9. The composition of E8, wherein the differentiation marker is selected from the group consisting of: keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA) and a combination thereof. F. A method for screening an agent, the method comprising: a) contacting a tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids as D or D1, or a composition as any one of E to E9, with an agent; and b) analyzing the tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids for changes indicative of the effectiveness and/or toxicity of the agent. F1. The method of F, wherein the agent is contacted with the tissue-derived epithelial organoid or a group of tissue-derived epithelial organoids for about 1 minute to about 3 years. F1-1. The method of F1, wherein the agent is contacted with the tissue-derived epithelial organoid or a population of such tissue-derived epithelial organoids for about 15 minutes to about 3 years. F2. The method of F, F1 or F1-1, wherein the agent is a therapeutic agent. F3. The method of F2, wherein the therapeutic agent is a polypeptide-based therapeutic agent, a small molecule therapeutic agent, a cell-based therapeutic agent, a gene editing system, a nucleic acid-based therapeutic agent, or a combination thereof. G. A method for performing genomic screening, the method comprising: a) providing a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids as described in D or D1 or a composition as described in any one of E to E9; b) generating a mutation in the genome of one or more cells of the tissue-derived epithelial organoid; and c) analyzing the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids for changes associated with the mutation. G1. The method of G, wherein the mutation is generated using a gene regulation system. G2. The method of G1, wherein the gene regulation system is a gene editing system. G3. The method of G2, wherein the gene editing system is a CRISPR system. G4. The method of any one of F to G2, wherein the change is a change in a property selected from the group consisting of cell viability, cell metabolism, redox potential, cell proliferation, cell morphology, organoid morphology, organoid size, protein expression level, nucleic acid expression level, nucleic acid modification, post-translational modification, activation of cell signaling pathways, repression of cell signaling pathways, enzyme activity, barrier integrity, and combinations thereof. H. A method for generating an epithelial cell model, the method comprising: a) providing a tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids as in D; b) digesting the tissue-derived epithelial organoid or a population of tissue-derived epithelial organoids into single cells; and c) culturing the single cells in a culture medium to generate a cell monolayer. H1. The method as in H, wherein the single cells are cultured on a permeable cell culture inner. H2. The method as in H or H1, wherein the culture medium is a differentiation medium. H3. The method as in H or H1, wherein the culture medium is a cell growth or stem cell promotion medium. I. A method for screening an agent, the method comprising: a) contacting a cell monolayer produced by any of the methods of H to H3 with an agent; and b) analyzing the cell monolayer for changes indicative of the effectiveness, distribution and/or toxicity of the agent. I1. The method of I, wherein the agent is contacted with the cell monolayer for about 1 minute to about 3 years. I1-1. The method of I1, wherein the agent is contacted with the cell monolayer for about 15 minutes to about 3 years. I2. The method of I, I1 or I1-1, wherein the agent is a therapeutic agent. I3. The method of I2, wherein the therapeutic agent is a polypeptide-based therapeutic agent, a small molecule therapeutic agent, a cell therapeutic agent, a gene editing system, a nucleic acid-based therapeutic agent, or a combination thereof. J. A method for performing genome screening, the method comprising: a) providing a cell monolayer produced by a method as described in any one of H to H3; b) generating a mutation in the genome of one or more cells in the cell monolayer; and c) analyzing the cell monolayer for changes associated with the mutation. J1. The method of J, wherein the mutation is generated using a gene regulation system. J2. The method of J1, wherein the gene regulation system is a gene editing system. J3. The method of J2, wherein the gene editing system is a CRISPR system. J4. The method of any one of J to J3, wherein the change is a change in a property, the property being selected from the group consisting of: cell viability, cell metabolism, redox potential, cell proliferation, cell morphology, organoid morphology, organoid size, protein expression level, nucleic acid expression level, nucleic acid modification, post-translational modification, activation of cell signaling pathways, repression of cell signaling pathways, enzyme activity, barrier integrity, and combinations thereof. J1. The method of any one of A to C21 and F to J4 or the composition of any one of E to E9, wherein the tissue fragment is a fragment from a tissue selected from the group consisting of lacrimal glands, tonsils, salivary glands, gastrointestinal tissue, thyroid, lung, breast, liver, bile duct, stomach, kidney, pancreas, endometrium, fallopian tube, cervix, prostate, bladder, taste buds, ovaries, placenta and combinations thereof. J5. The method of any one of A to C21 and F to J4 or the composition of any one of E to E9, wherein the tissue-derived epithelial stem cells are obtained from fragments of organoids, the organoids being selected from the group consisting of tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, gastric organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, trophoblastic organoids, and combinations thereof. K. A system for culturing tissue-derived epithelial organoids, the system comprising tissue-derived epithelial organoids and a culture medium, wherein the tissue-derived epithelial organoids are embedded in a hydrogel suspended in the culture medium. K1. The system of K, wherein the tissue-derived epithelial organoids are intestinal organoids. K2. The system of K or K1, wherein the hydrogel has a geometry comprising a length, width, and/or diameter greater than about 0.1 mm. K3. The system of any one of K to K2, wherein the hydrogel has a geometry comprising a length, width, and/or diameter of about 0.1 mm to about 1,000 mm. K4. The system of any of K to K3, wherein the hydrogel is a droplet. K5. The system of any of K to K3, wherein the hydrogel has a filamentous structure. K6. The system of K5, wherein the filamentous structure has a linear, serpentine, or helical shape. K7. The system of any of K to K6, wherein stem cell and/or proliferation markers are expressed at higher levels in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. K8. The system of K7, wherein the stem cell and/or proliferation marker is selected from the group consisting of: MKI67, EpCAM, BMI1, CD49f, ASCL2, CD133, LGR5, SOX9, ALDH1A1, NEUROG3, NKX6.1, SMOC2, PDX1, CD44, and combinations thereof. K9. The system of any one of K to K6, wherein the differentiation marker is expressed at a lower level in a population of epithelial organoids derived from tissues compared to a population of epithelial organoids derived from reference tissues, wherein the epithelial organoids derived from reference tissues are tissue-derived epithelial organoids embedded in a hydrogel attached to a substrate. K10. A system as described in K9, wherein the differentiation marker is selected from the group consisting of: keratin 20 (KRT20), FABP1, MUC2, MUC5B, MUC5AC, MUC6, TFF3, ALPI, SI, CEACAM7, keratin 19 (KRT19), keratin 7 (KRT7), SOX9, MUC1, INS, GCG, AMY, ALB, CYP3A4, HNF4A, cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (K5), cytokeratin 14 (K14), smooth muscle actin (SMA) and combinations thereof. K11. The system of any one of K to K10, wherein the tissue-derived epithelial organoid is selected from the group consisting of tear gland organoids, tonsil organoids, salivary gland organoids, gastrointestinal organoids, thyroid organoids, lung organoids, breast organoids, liver organoids, bile duct organoids, stomach organoids, kidney organoids, pancreatic organoids, endometrial organoids, fallopian tube organoids, cervical organoids, prostate organoids, bladder organoids, ovarian organoids, taste bud organoids, trophoblastic lining organoids, and combinations thereof. K12. The system of any one of K to K11, further comprising a robot and/or automated components. K13. A system as in K12, wherein the robot and/or automated component comprises a liquid handling robot, a 3D printer, a syringe pump, or a combination thereof. L. A method as in any one of A to C21 and F to J5, wherein one or more steps of the method are performed by a robot and/or automated component. L1. A method as in L, wherein the robot and/or automated component comprises a liquid handling robot, a 3D printer, a syringe pump, or a combination thereof. L2. A method as in L or L1, wherein the robot and/or automated component is a liquid handling robot. M. A method for producing epithelial organoids derived from tissues as in D to D1 or compositions as in E to E9, wherein one or more steps of the method are performed by a robot and/or automated component. M1. The method of M, wherein the robot and/or automated component comprises a liquid handling robot, a 3D printer, a syringe pump, or a combination thereof. N. The method of any one of B to B18, wherein the mixture comprising the hydrogel and the tissue-derived epithelial stem cells is introduced into the culture medium to produce a suspended mixture by a robot and/or an automated component. N1. The method of any one of B to B18, wherein the suspended mixture is cultured in the culture medium to produce the tissue-derived epithelial organoids in a suspended state by a robot and/or an automated component. N2. The method of N or N1, wherein the robot and/or automated component comprises a liquid handling robot, a 3D printer, a syringe pump, or a combination thereof. N3. The method of N2, wherein the robot and/or automated component comprises one or more liquid handling robots.
藉由參考以下實例將更好地理解本文所揭示之主題,這些實例是作為本文所揭示之主題的例示而不是作為限制而提供的。The subject matter disclosed herein will be better understood by reference to the following examples, which are provided by way of illustration and not by way of limitation of the subject matter disclosed herein.
實例Examples 11 :腸類器官之形成:Formation of intestinal organoids
該實例提供了一種在各種幾何形狀之經懸浮之基底膜萃取物 (BME) 水凝膠中培養腸類器官之方法。該方法簡化了協議,增加了可擴展性,實現了動力學採樣並改善了培養物均勻性,而無需專用設備或額外專業知識。此方法與多種培養物形式相容,並且藉由此方法產生之類器官可用於下游應用,諸如實施中等生產量藥物篩選及產生用於障壁評估之 Transwell 單層,如實例 2 及 3 所示。經懸浮之 BME 水凝膠培養方法使腸類器官之使用範圍比以前可能的使用範圍更廣泛且生產量比以前可能的生產量更高。This example provides a method for culturing intestinal organoids in suspended basement membrane extract (BME) hydrogels in a variety of geometries. The method simplifies the protocol, increases scalability, enables kinetic sampling, and improves culture homogeneity without the need for specialized equipment or additional expertise. The method is compatible with a variety of culture formats, and organoids generated by this method can be used in downstream applications such as performing mid-throughput drug screening and generating Transwell monolayers for barrier assessment, as shown in Examples 2 and 3. The suspended BME hydrogel culture method enables the use of intestinal organoids to a wider range of uses and at higher throughputs than previously possible.
方法method
人類腸類器官來源。 類器官來源如前所述 (Pleguezuelos-Manzano 等人(2020)),其中進行了一些修改。來自已故供體之未鑑定之人類大腸及迴腸組織樣本藉由西部供體網路獲得。組織以 Advanced DMEM/F12 培養基 (ThermoFisher) 洗滌,切成 5cm x 5cm 區段,然後將上皮從黏膜下層刮到培養基中並用刀片切碎。將溶液以 450 x g 沉澱 5 分鐘,然後在不含 Mg 2+或 Ca 2+之 PBS 中於 2.5 µM EDTA 中於 37°C 培育 9 分鐘 (迴腸) 或 12 分鐘 (大腸),每 3 至 4 分鐘渦旋一次,直至釋放隱窩。將隱窩以 450 x g 沉澱 5 分鐘,在 PBS 中洗滌,藉由無菌紗布過濾,然後藉由 100 µm 細胞過濾器過濾以去除碎片,並以 450 x g 沉澱 5 分鐘。將隱窩重懸於冰上之 CULTREX® 低生長因子基底膜基質 II 型 (BME, R&D Systems 目錄號 3533-010-02) 中,置於 24 孔盤中之 50 µL 圓頂中,在 37℃ 固化 15 至 30 分鐘,然後以 500 µL 大腸傳代培養基 (Intesticult 類器官生長培養基 (OGM, StemCell Technologies 目錄號 06010) + 10 µM Y27632) 或迴腸培養基 (OGM + 10 µM Y27632 + 2.5 µM CHIR99021) 覆蓋。於培養物中前 2 至 3 天後,每 2 至 3 天一次更換培養基,或當培養基變黃時更換一次培養基,對於大腸培養物更換為普通 OGM,對於迴腸培養物更換為迴腸培養基。 Human intestinal organoids source. Organoids were sourced as previously described (Pleguezuelos-Manzano et al. (2020)) with some modifications. Unidentified human colon and ileum tissue samples from deceased donors were obtained through the Western Donor Network. Tissues were washed with Advanced DMEM/F12 medium (ThermoFisher), cut into 5 cm x 5 cm segments, and the epithelium was scraped from the submucosal layer into the medium and minced with a razor blade. The solution was pelleted at 450 x g for 5 min and then incubated in 2.5 µM EDTA in PBS without Mg 2+ or Ca 2+ at 37°C for 9 min (ileum) or 12 min (colon), vortexing every 3 to 4 min until the crypts were released. The crypts were pelleted at 450 x g for 5 min, washed in PBS, filtered through sterile gauze, then filtered through a 100 µm cell filter to remove debris, and pelleted at 450 x g for 5 min. Nests were resuspended in CULTREX® Reduced Growth Factor Basement Membrane Matrix Type II (BME, R&D Systems Catalog No. 3533-010-02) on ice, placed in 50 µL domes in 24-well plates, solidified at 37°C for 15 to 30 minutes, and then overlaid with 500 µL of either colonic medium (Intesticult Organoid Growth Medium (OGM, StemCell Technologies Catalog No. 06010) + 10 µM Y27632) or ileal medium (OGM + 10 µM Y27632 + 2.5 µM CHIR99021). After the first 2 to 3 days in culture, change the medium every 2 to 3 days or when the medium turns yellow, to plain OGM for colon cultures and to ileal medium for ileal cultures.
類器官維持。 每 1 至 2 週藉由以 TrypLE Express (ThermoFisher) 在 37°C 消化 10 分鐘來使類器官培養物傳代,然後以 P1000 移液管研磨。如有必要,重複培育最多 2 次以獲得單細胞懸浮液。TrypLE Express 藉由以 PBS 稀釋來滅話,並將細胞以 450 x g 沉澱 3 分鐘。將細胞以 6 x 10 5個細胞/mL 重懸於冰上之 BME 中,在 24 孔盤中在 50 µL 圓頂中平鋪,並在 37°C 固化 15 至 30 分鐘。前 2 至 3 天以大腸傳代培養基或迴腸培養基覆蓋圓頂,並且每 2 至 3 天一次以普通 OGM 更換大腸之培養基或以迴腸培養基更換迴腸之培養基。對於大腸類器官分化,以 Advanced DMEM/F12 培養基洗滌培養物,然後以 Intesticult 類器官分化培養基 (ODM, StemCell Technologies 目錄號 100-0214) + 5 µM DAPT 覆蓋 5 天,每 2 至 3 天一次更換培養基。 Organoid maintenance. Organoid cultures were passaged every 1 to 2 weeks by digestion with TrypLE Express (ThermoFisher) at 37°C for 10 min, followed by trituration with a P1000 pipette. If necessary, the culture was repeated up to 2 times to obtain a single-cell suspension. TrypLE Express was quenched by diluting with PBS and the cells were pelleted at 450 x g for 3 min. Cells were resuspended at 6 x 10 5 cells/mL in BME on ice, plated in 50 µL domes in a 24-well plate, and fixed at 37°C for 15 to 30 min. For the first 2 to 3 days, cover the domes with colon passage medium or ileum medium and replace the medium with plain OGM for colon or ileum medium every 2 to 3 days. For colon organoid differentiation, wash the cultures with Advanced DMEM/F12 medium and then cover with Intesticult Organoid Differentiation Medium (ODM, StemCell Technologies Catalog No. 100-0214) + 5 µM DAPT for 5 days, replacing the medium every 2 to 3 days.
經懸浮之水凝膠 BOBA 培養物。 如上所述,在冰上製備 BME 中之單類器官細胞。將預熱之大腸傳代培養基或迴腸培養基添加至 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15-30 mL/皿) 或 50 mL 錐形管 (30 mL/管) 中,並保持在 37°C 之溫熱珠浴上。超低接附 (ULA) 或標準組織培養盤產生類似結果。圖10 提供了顯示 BOBA 之產生之示例性示意圖。為了產生經懸浮之 BME 小滴或 BOBA,使用帶有寬孔或切割移液管尖端 (開口約 2 mm) 之電子連續移液管 (Integra VIAFLO 300) 以慢速至中速將類器官細胞-BME 溶液以 10 µL 體積直接分配到溫熱培養基中,以避免形成絲或細帶而不是小滴。在分配過程中,尖端立即浸入液體表面下方,然後在每次分配後提起,以確保小滴分離。對於較大形式之培養物,藉由血清移液管或傾析將 BOBA 轉移至燒瓶中。每 2 至 3 天一次以普通 OGM 更換大腸之培養基或以迴腸培養基更換迴腸之培養基。在 6 孔盤培養中,將無菌 70 µm 細胞過濾器放入孔中,傾斜盤並透過過濾器輕輕吸出 4 mL 培養基。在燒瓶培養物中,將燒瓶傾斜一定角度,將 BOBA 放置在角落,然後以血清移液管更換大約 2/3 體積之已用培養基。 Suspended hydrogel BOBA cultures. Single organoids in BME were prepared on ice as described above. Pre-warmed colon passage medium or ileal medium was added to 6-well plates (5 mL/well), 100 cm culture dishes (15-30 mL/dish), or 50 mL conical tubes (30 mL/tube) and maintained in a warm bead bath at 37°C. Ultra-low attachment (ULA) or standard tissue culture dishes produced similar results. Figure 10 provides an exemplary schematic showing the generation of BOBA. To generate suspended BME droplets or BOBAs, dispense the organoid cell-BME solution in 10 µL volumes directly into the warm medium using an electronic continuous pipette (Integra VIAFLO 300) with a wide-bore or cut pipette tip (opening approximately 2 mm) at a slow to moderate speed to avoid the formation of threads or ribbons instead of droplets. During the dispensing process, the tip is immediately dipped below the surface of the liquid and then lifted after each dispense to ensure the separation of the droplets. For larger cultures, transfer the BOBA to a flask by serological pipette or decanting. Replace the medium with normal OGM for colon or ileum medium every 2 to 3 days. In 6-well plate cultures, place a sterile 70 µm cell filter into the well, tilt the plate and gently aspirate 4 mL of medium through the filter. In flask cultures, tilt the flask at an angle, place BOBA in a corner, and replace approximately 2/3 of the volume of used medium with a serological pipette.
經懸浮之水凝膠 SOBA 及 SOBA 碎片培養物。 如上所述,在冰上製備 BME 中之單類器官細胞。將經預熱之大腸傳代培養基添加至 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15-30 mL/皿) 中,並保持在溫熱珠浴上。圖11 提供了顯示產生 SOBA 絲之示例性示意圖。為了產生經懸浮之 SOBA 絲,將細胞-BME 溶液輕輕吸入帶有 15 號鈍尖端針之注射器中,然後直接擠出到溫熱培養基中,同時以 X-Y 平面中線形、蛇形或螺旋形運動方式移動浸沒針。X-Y 平面中之擠出速度及運動可影響絲長度及/或寬度。為了產生 SOBA 碎片,使用 10 mL 血清移液管或寬孔 P1000 移液管尖端輕輕研磨 SOBA 絲培養物兩次。添加額外培養基以使最終 BME 與培養基之比率達到 1:10。如上對 BOBA 培養物所述進行培養基更換。 Suspended hydrogel SOBA and SOBA fragment cultures. Single organoid cells in BME were prepared on ice as described above. Prewarmed colon passage medium was added to 6-well plates (5 mL/well), 100 cm culture dishes (15-30 mL/dish) and kept on a warm bead bath. Figure 11 provides an exemplary schematic showing the generation of SOBA filaments. To generate suspended SOBA filaments, the cell-BME solution was gently aspirated into a syringe with a 15-gauge blunt-tip needle and then extruded directly into the warm medium while moving the submerged needle in a linear, serpentine, or spiral motion in the XY plane. Extrusion speed and motion in the XY plane can affect filament length and/or width. To generate SOBA fragments, gently triturate SOBA filament cultures twice using a 10 mL serological pipette or wide-bore P1000 pipette tip. Add additional medium to achieve a final BME to medium ratio of 1:10. Perform medium changes as described above for BOBA cultures.
明視野顯微術及影像分析。 使用具有 2.5X、4X 或 10X 物鏡之 THUNDER DMi8 倒置光學顯微鏡 (Leica) 及 DFC9000 GTC 相機 (Leica) 藉由明視野顯微術對培養物進行成像。使用 Imaris 影像分析軟體及 Imaris Batch 軟體套件 (Oxford Instruments) 分析影像。對於自動化類器官直徑測量值,Imaris Surfaces 檢測模組與倒置明視野影像一起使用。使用背景扣除步驟 (滾球,直徑 19.5 µm) 排除背景及失焦類器官。然後根據四個標準對檢測到之表面進行過濾:軟體指定之品質指標 (>3000 A.U.)、短軸長度 (>35 µm - 排除碎片)、長方形圓度 (>0.2 - 排除陰影) 及長軸長度 (35 µm 與 600 µm 之間,排除重疊類器官之假檢測)。在剩餘表面 (每個分析影像至少 40 個) 中,直徑被報告為最小物件定向包圍框之最長邊。然後在所有分析之影像上批量進行該過程。計算每個影像之平均值,並繪製 n = 3 次重複之三個總實驗之平均值。 Bright field microscopy and image analysis. Cultures were imaged by bright field microscopy using a THUNDER DMi8 inverted light microscope (Leica) with 2.5X, 4X, or 10X objectives and a DFC9000 GTC camera (Leica). Images were analyzed using Imaris image analysis software and Imaris Batch software suite (Oxford Instruments). For automated organoid diameter measurements, the Imaris Surfaces detection module was used with inverted bright field images. Background and out-of-focus organoids were excluded using a background subtraction step (rolling ball, diameter 19.5 µm). The detected surfaces were then filtered according to four criteria: software-specified quality metrics (>3000 AU), minor axis length (>35 µm - to exclude debris), rectangular circularity (>0.2 - to exclude shadows), and major axis length (between 35 µm and 600 µm, to exclude false detections of overlapping organoids). In the remaining surfaces (at least 40 per analyzed image), the diameter was reported as the longest side of the smallest object oriented bounding box. This process was then performed in batches on all analyzed images. The average was calculated for each image and plotted for three total experiments with n = 3 replicates.
使用 FIJI (ImageJ) 進行空間類器官均勻性類器官直徑分析。在各影像中心設置含量矩形 (1.5 mm x 9 mm) ROI,並在 X 軸中分成 1 mm 部分。對於各部分,手動測量跨越各類器官最寬點之直徑。Spatial organoid homogeneity Organoid diameter analysis was performed using FIJI (ImageJ). A rectangular (1.5 mm x 9 mm) ROI was set at the center of each image and divided into 1 mm sections in the X-axis. For each section, the diameter across the widest point of each organoid was manually measured.
免疫螢光樣本製備及共焦顯微術。 24 孔圓頂培養物以於 PBS 中之 2% 多聚甲醛 (PFA) 固定。使用抹刀將圓頂從盤上分離,並使用切割 P1000 移液管尖端轉移到微量離心管中。對於 BOBA 培養物,使用切割 P1000 移液管尖端將 500 µL 培養物轉移至微量離心管中,除去培養基並添加於 PBS 中之 2% PFA。將樣本在固定劑中室溫培育 15 至 30 分鐘,然後在 PBS 中洗滌 3 次。將樣本在微量離心管中以在封閉/透化緩衝液 (3% BSA、0.1% Triton X-100、0.02% 疊氮化鈉,於 PBS 中) 中稀釋之初級抗體在室溫染色至少 4 小時,然後在 PBS 中洗滌 3 次。所使用之初級抗體如下:ɑ-Ki67 (Invitrogen 目錄號 MA5-14520)、ɑ-MUC2 (Millipore 目錄號 MABF1989)、ɑ-FABP1 (Novus 目錄號 NBP-87695) 及 ɑ-CHGA (Novus 目錄號 NB120-15160)。然後將樣本與於室溫在封閉/透化緩衝液中稀釋之二級抗體 (驢 ɑ-兔 Alexa Fluor 488 (ThermoFisher 目錄號 A-21206) 或山羊 ɑ-小鼠 Alexa Fluor 594 (ThermoFisher 目錄號 A-11032))、DAPI 及 AlexaFluor 660 蠅虎蕈鹼一起在室溫培育至少 2 小時。使用 40X 物鏡在 Stellaris 8 共焦顯微鏡 (Leica) 上收集影像,並使用 Imaris 影像分析軟體 (Oxford Instruments) 進行 3D 重建。 Immunofluorescence Sample Preparation and Confocal Microscopy. 24-well dome cultures were fixed with 2% paraformaldehyde (PFA) in PBS. Domes were detached from the plate using a spatula and transferred to a microcentrifuge tube using a cut P1000 pipette tip. For BOBA cultures, 500 µL of culture was transferred to a microcentrifuge tube using a cut P1000 pipette tip, the medium was removed and 2% PFA in PBS was added. Samples were incubated in fixative at room temperature for 15 to 30 minutes and then washed three times in PBS. Samples were stained in microcentrifuge tubes with primary antibodies diluted in blocking/permeabilization buffer (3% BSA, 0.1% Triton X-100, 0.02% sodium azide in PBS) for at least 4 hours at room temperature and then washed three times in PBS. The primary antibodies used were as follows: ɑ-Ki67 (Invitrogen Catalog No. MA5-14520), ɑ-MUC2 (Millipore Catalog No. MABF1989), ɑ-FABP1 (Novus Catalog No. NBP-87695), and ɑ-CHGA (Novus Catalog No. NB120-15160). Samples were then incubated with secondary antibodies (donkey ɑ-rabbit Alexa Fluor 488 (ThermoFisher Catalog No. A-21206) or goat ɑ-mouse Alexa Fluor 594 (ThermoFisher Catalog No. A-11032)), DAPI, and AlexaFluor 660 agaricus diluted in blocking/permeabilization buffer at room temperature for at least 2 hours. Images were collected on a Stellaris 8 confocal microscope (Leica) using a 40X objective and 3D reconstructions were performed using Imaris image analysis software (Oxford Instruments).
轉錄組分析。 對於 RNA 分離,使用 RNeasy Micro Plus 套組 (Qiagen)。將 RLT+ 裂解緩衝液添加至 BME 圓頂或沉澱 BOBA 中並儲存在 -80℃。使用 QiaCube Connect (Qiagen) 進行 RNA 分離,並使用 Nanodrop 8000 (ThermoFisher) 定量 RNA。批量 mRNA-seq (NovaSeq PE150) 及分析藉由 Novogene 進行。使用 HISAT2 (Mortazavi 等人 2008) 比對讀段,使用 DESeq2 (Anders 等人2014) 進行差異基因表現分析,並使用負二項分佈模型及 Benjamini-Hochberg FDR 校正計算統計顯著性。 Transcriptome analysis. For RNA isolation, the RNeasy Micro Plus kit (Qiagen) was used. RLT+ lysis buffer was added to BME domes or pelleted BOBA and stored at -80°C. RNA isolation was performed using QiaCube Connect (Qiagen), and RNA was quantified using Nanodrop 8000 (ThermoFisher). Bulk mRNA-seq (NovaSeq PE150) and analysis were performed by Novogene. Reads were aligned using HISAT2 (Mortazavi et al. 2008), differential gene expression analysis was performed using DESeq2 (Anders et al. 2014), and statistical significance was calculated using a negative binomial distribution model with Benjamini-Hochberg FDR correction.
96 孔盤懸浮之 BME 類器官變異性。 培養 9 天後,收集 225 cm 2燒瓶中之大腸類器官 SOBA 碎片,並使用血清移液管輕輕研磨兩次,以均質化樣本,而不會破壞完整類器官。將類器官混合物轉移至試劑貯器中,且然後使用帶有寬孔移液管尖端之 P200 多通道移液管以 100 µL/孔平鋪於 96 孔盤中。如上所述製備圓頂培養物,並使用連續移液管將其平鋪於 96 孔盤各孔中心之 5 µL 圓頂中。在進行生存力測量之前,使培養物生長 7 天。所有生存力測量均使用 Cell Titer Glo 3D Assay 套組 (Promega) 進行,並在 Ensight 酶標儀 (Perkin Elmer) 上測量發光。 Variability of BME organoids suspended in 96 -well plates. After 9 days of culture, colon organoid SOBA fragments in 225 cm2 flasks were collected and gently triturated twice using a serological pipette to homogenize the sample without disrupting the intact organoids. The organoid mixture was transferred to a reagent container and then plated at 100 µL/well in a 96-well plate using a P200 multichannel pipette with a wide-bore pipette tip. Dome cultures were prepared as described above and plated in a 5 µL dome in the center of each well of a 96-well plate using a continuous pipette. Cultures were grown for 7 days before viability measurements. All viability measurements were performed using the Cell Titer Glo 3D Assay kit (Promega), and luminescence was measured on an Ensight microplate reader (Perkin Elmer).
統計學分析。 除非另有說明,否則所有統計學分析均使用 Prism 9 軟體 (Graphpad) 進行。統計學檢驗、n 及 p 值在圖例中標示。 Statistical analysis. Unless otherwise stated, all statistical analyses were performed using Prism 9 software (Graphpad). Statistical tests, n, and p values are indicated in the figure legends.
結果:result:
用於人類腸類器官培養之經懸浮之 BME 水凝膠方法。 習用腸類器官培養方法需要將冷 ECM 中之類器官細胞溶液沉積到塑膠表面,在培養箱中固化形成以水凝膠圓頂,然後以生長培養基覆蓋 (圖1A-1B) (Mahe 等人(2013);Pleguezuelos‐Manzano 等人(2020);Sato 等人(2009);Sato 等人(2011))。為了解決規模化該技術之局限性,開發了一種方法,其中冷細胞 ECM 溶液立即固化為懸浮於溫熱培養基中之漂浮水凝膠。 已經顯示了一種使用經懸浮之 BME 小滴之方法,BME 為 Engelbreth-Holm-Swarm (EHS) 細胞來源之 ECM,相當於 MATRIGEL®,並且該方法稱為 BOBA (BME 嵌入類器官珠組裝體) 培養。 Suspended BME hydrogel method for human intestinal organoid culture. The conventional intestinal organoid culture method requires that a solution of organoid cells in cold ECM be deposited onto a plastic surface, solidified in an incubator to form a hydrogel dome, and then covered with growth medium (Figure 1A-1B) (Mahe et al. (2013); Pleguezuelos-Manzano et al. (2020); Sato et al. (2009); Sato et al. (2011)). To address the limitations of scaling up this technique, a method was developed in which the cold cell-ECM solution immediately solidifies into a floating hydrogel suspended in warm medium. A method has been demonstrated using suspended droplets of BME, an Engelbreth-Holm-Swarm (EHS) cell-derived ECM equivalent to MATRIGEL®, and is termed BOBA (BME Embedded Organoid Bead Assembly) culture.
首先在經懸浮之 BOBA 培養方法中評估腸類器官生長,並與習用表面接附 BME 圓頂方法 (此處稱為圓頂培養) 進行比較。對於圓頂培養,將來自消化類器官之單細胞懸浮在冷 BME 中,並作為 50 µL 圓頂平鋪在 24 孔盤中,在 37°C 培養箱中固化 15 至 30 分鐘,然後以生長培養基 (Intesticult 類器官生長培養基) 覆蓋。每 2 至 3 天一次對各孔單獨進行培養基更換。對於 BOBA 培養,使用帶有寬孔尖端之電子連續移液管將單細胞 BME 溶液作為 10 µL 小滴直接沉積到預熱培養基中,其中水凝膠立即固化成經懸浮之 BOBA。BOBA 可以在培養皿、盤或錐形管中產生,並且經由血清移液管或傾析輕鬆轉移到更大容器,如細胞培養瓶中 (圖1A-1B)。藉由允許 BOBA 沉降,然後以新鮮培養基替換頂部 75% 體積之已用培養基來對整個燒瓶進行培養基更換。Intestinal organoid growth was first assessed in a suspended BOBA culture method and compared to the conventional surface-attached BME dome method (referred to here as dome culture). For dome culture, single cells from digested organoids were suspended in cold BME and plated as 50 µL domes in a 24-well plate, solidified in a 37°C incubator for 15 to 30 minutes, and then covered with growth medium (Intesticult Organoid Growth Medium). Medium changes were performed on individual wells every 2 to 3 days. For BOBA culture, single-cell BME solutions are deposited as 10 µL drops directly into pre-warmed medium using an electronic continuous pipette with a wide-bore tip, where the hydrogel immediately solidifies into suspended BOBA. BOBA can be produced in culture dishes, plates, or conical tubes and easily transferred to larger containers, such as cell culture flasks, via serological pipette or decanting (Figure 1A-1B). Perform a medium change for the entire flask by allowing the BOBA to settle and then replacing the top 75% of the volume of spent medium with fresh medium.
將人類腸類器官細胞-BME 溶液平鋪於平行圓頂或 BOBA 培養物中,並在生長培養基中生長 9 天。類器官生長及尺寸在兩種方法中相似,如藉由明視野顯微術所觀察到的 (圖1C) 並以類器官直徑測量進行定量 (圖1D)。類器官細胞增生也係可比較的,如藉由定量表現增生標記物 Ki-67 之細胞豐度所確定的 (圖1E-1F)。BOBA 方法似乎使得能夠在每 cm 2表面積上實現更多類器官細胞生長,儘管僅在小腸 (迴腸) 類器官中觀察到統計學顯著性 (圖1G-1H)。對於大腸類器官,圓頂培養產生平均 2.9 x 10 5個活細胞/孔或 24 孔盤中 1.5 x 10 5個細胞/cm 2,而 BOBA 培養產生平均 2.2 x 10 7個活細胞/孔或 75 cm 2燒瓶中 2.9 x 10 5個細胞/cm 2(圖 1G)。對於迴腸類器官,圓頂培養產生平均 4.8 x 10 5個活細胞/孔或 24 孔盤中 2.6 x 10 5個細胞/cm 2,而 BOBA 培養產生平均 2.9 x 10 7個活細胞/孔或 75 cm 2燒瓶中 3.9 x 10 5個細胞/cm 2(圖1H)。每 µL BME 水凝膠之活細胞數量相似,指示兩種方法在固定接種密度下之生長量係可比較的 (圖1G)。 Human intestinal organoid cell-BME solutions were plated in parallel domes or BOBA cultures and grown in growth medium for 9 days. Organoid growth and size were similar in both methods, as observed by bright field microscopy (Fig. 1C) and quantified by organoid diameter measurement (Fig. 1D). Organoid cell proliferation was also comparable, as determined by quantification of cell abundance expressing the proliferation marker Ki-67 (Fig. 1E-1F). The BOBA method appeared to enable the growth of more organoid cells per cm2 surface area, although statistical significance was only observed in small intestinal (ileal) organoids (Fig. 1G-1H). For colon organoids, dome culture yielded an average of 2.9 x 10 5 viable cells/well or 1.5 x 10 5 cells/cm 2 in a 24-well plate, while BOBA culture yielded an average of 2.2 x 10 7 viable cells/well or 2.9 x 10 5 cells/cm 2 in a 75 cm 2 flask (Figure 1G). For ileal organoids, dome culture yielded an average of 4.8 x 10 5 viable cells/well or 2.6 x 10 5 cells/cm 2 in a 24-well plate, while BOBA culture yielded an average of 2.9 x 10 7 viable cells/well or 3.9 x 10 5 cells/cm 2 in a 75 cm 2 flask (Figure 1H). The number of viable cells per µL of BME hydrogel was similar, indicating that the two methods were comparable in growth at a fixed seeding density (Figure 1G).
BOBA 培養中之類器官分化。 腸類器官模型之主要優點係能夠藉由改變培養基組成 (例如藉由取出幹細胞促進因子) 分化成各種腸上皮細胞類型 (Clevers (2016);Schutgens 及 Clevers (2019);Zachos 等人(2016))。比較了圓頂及 BOBA 培養中之類器官分化。使類器官在生長培養基中生長 7 天,然後洗滌並轉移到分化培養基 (含有 5 µM DAPT 之 Intesticut 類器官分化培養基) 中,持續 5 天。明視野顯微術顯示,在兩種培養形式中,生長培養基中之增生類器官表現出具有大管腔之隱窩形態 (圖2A) 且分化類器官表現出具有細長柱狀細胞及小管腔之緻密球狀形態 (圖2A)。 Organoid differentiation in BOBA culture. A major advantage of the intestinal organoid model is the ability to differentiate into various intestinal epithelial cell types by changing the medium composition (e.g., by removing stem cell-promoting factors) (Clevers (2016); Schutgens and Clevers (2019); Zachos et al. (2016)). Organoid differentiation in dome and BOBA culture was compared. Organoids were grown in growth medium for 7 days and then washed and transferred to differentiation medium (Intesticut Organoid Differentiation Medium containing 5 µM DAPT) for 5 days. Bright field microscopy showed that in both culture formats, proliferating organoids in growth medium exhibited a cryptic morphology with large lumens (Figure 2A) and differentiated organoids exhibited a compact spherical morphology with elongated columnar cells and small lumens (Figure 2A).
對圓頂及 BOBA 培養中之增生及分化類器官進行批量 RNA-seq 分析。在兩種培養方法中,相對於增生類器官,分化類器官下調幹細胞及增生標記物 ( MKI67、 LGR5、 SOX9及 CD44) 之表現,且上調杯狀細胞 ( MUC2、 MUC5B、 TFF3) 及腸上皮細胞 ( KRT20、 FABP1、 ALPI及 CEACAM7) 之分化標記物之表現 (圖2B)。也藉由免疫螢光 (IF) 共焦顯微術在兩種培養形式中觀察到分化細胞類型 (圖2C)。 Bulk RNA-seq analysis was performed on proliferative and differentiated organoids in dome and BOBA culture. In both culture methods, differentiated organoids downregulated the expression of stem cell and proliferation markers ( MKI67 , LGR5 , SOX9 , and CD44 ) and upregulated the expression of differentiation markers for goblet cells ( MUC2 , MUC5B , TFF3 ) and intestinal epithelial cells ( KRT20 , FABP1 , ALPI , and CEACAM7 ) relative to proliferative organoids (Figure 2B). Differentiated cell types were also observed in both culture formats by immunofluorescence (IF) confocal microscopy (Figure 2C).
BOBA 培養條件之表徵及優化。 接下來,評估了在 BOBA 方法中不同培養參數將如何影響類器官生長。將培養物接種在 6 孔盤 (圖3A-3B) 或 25 cm 2燒瓶 (圖3C) 中,該接種係以各種 BME 體積及 6 x 10 5個細胞/mL BME 之固定細胞接種密度進行。所有 BOBA 均以 10 µL 小滴形式在 5 mL 生長培養基中產生,並藉由定量培養 9 天後之類器官直徑及活細胞數量來評估類器官生長 (圖3B-3C)。在 6 孔盤中,隨著每孔總 BME 體積從 0.5 mL 增加到 2 mL,類器官直徑降低,且活細胞與 BME 體積之比率降低 (儘管不具有統計學顯著性)。儘管在較高 BME 體積條件下接種細胞數量較高,但在所有條件下,活細胞總數及每 cm 2表面積之活細胞比率係可比較的,指示每個接種細胞之增生較少。總之,這些資料表明,當 BME 體積超過 6 孔盤中固定量培養基之閾值時,類器官生長就會受到損害 (圖3B)。 Characterization and Optimization of BOBA Culture Conditions. Next, we evaluated how different culture parameters would affect organoid growth in the BOBA approach. Cultures were seeded in 6-well plates (Figures 3A-3B) or 25 cm2 flasks (Figure 3C) at various BME volumes and a fixed cell seeding density of 6 x 105 cells/mL BME. All BOBAs were produced as 10 µL droplets in 5 mL growth medium, and organoid growth was assessed by quantifying organoid diameter and number of viable cells after 9 days of culture (Figures 3B-3C). In 6-well plates, as the total BME volume per well increased from 0.5 mL to 2 mL, organoid diameter decreased and the ratio of viable cells to BME volume decreased (although not statistically significant). Although the number of cells seeded was higher under the higher BME volume conditions, the total number of viable cells and the ratio of viable cells per cm2 of surface area were comparable under all conditions, indicating less proliferation per seeded cell. Together, these data suggest that organoid growth is impaired when the BME volume exceeds a threshold of a fixed amount of medium in 6-well plates (Figure 3B).
有趣的是,對於 25 cm 2燒瓶中之 BOBA 培養物,類器官在所有測試條件下都生長得一樣好 (圖3C)。隨著每個燒瓶之總 BME 體積從 0.5 mL 增加到 2 mL,類器官直徑以及活細胞與 BME 體積之比率相似。在培養物中 BME 體積似乎增加時,活細胞總數及每 cm 2表面積之活細胞比率增加 (儘管由於實驗間差異,不具有統計學顯著性),這表明對於 6 孔盤及 25 cm 2燒瓶,BME 體積與培養基之閾值係不同的。BME 與培養基之比率及容器類型都應針對特定應用進行優化。 Interestingly, for BOBA cultures in 25 cm2 flasks, organoids grew equally well under all conditions tested (Figure 3C). As the total BME volume per flask increased from 0.5 mL to 2 mL, the organoid diameter and the ratio of viable cells to BME volume were similar. The total number of viable cells and the ratio of viable cells per cm2 surface area appeared to increase as the BME volume in the culture increased (although not statistically significant due to inter-experimental variability), suggesting that the thresholds for BME volume and medium are different for 6-well plates and 25 cm2 flasks. Both the BME to medium ratio and the vessel type should be optimized for specific applications.
BOBA 培養中之類器官均勻性。 已知習用圓頂方法會導致類器官異質性 (Pleguezuelos-Manzano 等人(2020);Ringel 等人(2020))。天然 ECM 水凝膠限制氣體及分子擴散,導致營養物梯度及異質的類器官生長 (Colom 等人(2014) J Biomed Mater Res A 102, 2776–2784;Park 等人(2022);Shin 等人(2020))。據觀察,類器官在 BOBA 培養中比在圓頂培養中生長得更均勻 (圖4)。藉由明視野顯微術在每種形式的最深點-圓頂之底部 Z 平面或 BOBA 之中心 Z 平面-對大腸類器官培養物進行成像,並定量各水凝膠中心跨越矩形 ROI 的平均類器官直徑 (圖4C)。與先前報告一致 (Park 等人(2022);Shin 等人(2020)),圓頂培養類器官在水凝膠邊緣的生長較大,且在核心的生長較小 (圖4B-4E)。然而,BOBA 中之類器官跨越水凝膠之生長尺寸係可比較的 (圖4B-E)。 Organoid Homogeneity in BOBA Culture. The conventional dome approach is known to result in organoid heterogeneity (Pleguezuelos-Manzano et al. (2020); Ringel et al. (2020)). The native ECM hydrogel restricts gas and molecular diffusion, leading to nutrient gradients and heterogeneous organoid growth (Colom et al. (2014) J Biomed Mater Res A 102 , 2776–2784; Park et al. (2022); Shin et al. (2020)). Organoids were observed to grow more uniformly in BOBA culture than in dome culture (Figure 4). Colon organoid cultures were imaged by bright field microscopy at the deepest point of each format—the bottom Z plane of the dome or the center Z plane of the BOBA—and the average organoid diameter across a rectangular ROI at the center of each hydrogel was quantified (Figure 4C). Consistent with previous reports (Park et al. (2022); Shin et al. (2020)), dome-cultured organoids had larger growth at the edges of the hydrogel and smaller growth in the core (Figures 4B-4E). However, the growth size of organoids in the BOBA across the hydrogel was comparable (Figures 4B-E).
也觀察到,圓頂培養物核心中之類器官通常具有緻密球狀管腔較少之形態,這通常指示分化 (圖4B)。批量 RNA-seq 分析指示,相對於其 BOBA 培養對應物,圓頂培養類器官之幹細胞及增生標記物表現較低,且腸上皮細胞標記物表現較高,這支持圓頂培養物中可能存在分化類器官亞群之假設 (圖5)。It was also observed that organoids in the core of dome cultures generally had a dense spherical morphology with less lumen, which is generally indicative of differentiation (Figure 4B). Bulk RNA-seq analysis indicated that dome-cultured organoids had lower expression of stem cell and proliferation markers and higher expression of intestinal epithelial cell markers relative to their BOBA-cultured counterparts, supporting the hypothesis that a subpopulation of differentiated organoids may exist in dome cultures (Figure 5).
經懸浮之 BME 水凝膠之替代幾何形狀。 雖然 BOBA 方法比習用圓頂方法具有顯著規模化優勢,但在沒有自動化液體處理器之情況下經懸浮之 BOBA 水凝膠小滴之大規模培養擴增仍然為一項勞動密集型工作。為了減少經懸浮之 BME 水凝膠培養所需之時間及勞動力,設計了替代的水凝膠幾何形狀,具體而言為水凝膠絲。擠出絲已在生物列印領域用於在空間上控制細胞生長或構建 3D 水凝膠結構之逐層組裝體,但通常依賴接附於表面 (Kolesky 等人(2014) Adv.Mater.26, 2966–2966;Kolesky 等人(2016) Proc National Acad Sci 113, 3179–3184)。為了產生含有類器官細胞之水凝膠絲,稱為 SOBA (注射器擠出之類器官 BME 組裝體),將冷細胞-BME 溶液加載到帶有15 號 (1.37 mm 內徑) 鈍尖端針之注射器中,然後直接注入溫熱培養基,同時跨越 X-Y 平面移動尖端 ( 例如,以線形、蛇形或螺旋形圖案)。藉由以寬孔 P1000 尖端或 10 mL 血清移液管輕輕研磨 SOBA 培養物,也產生了更密切接近 BOBA 幾何形狀之絲碎片,稱為 SOBA 碎片。 Alternative geometries for suspended BME hydrogels. While the BOBA approach has significant scalability advantages over the conventional dome approach, scale-up of large-scale cultures of suspended BOBA hydrogel droplets remains labor-intensive without automated liquid handlers. To reduce the time and labor required for suspended BME hydrogel cultures, alternative hydrogel geometries, specifically hydrogel filaments, have been designed. Extruded filaments have been used in the bioprinting field to spatially control cell growth or to construct layer-by-layer assemblies of 3D hydrogel structures, but they generally rely on attachment to a surface (Kolesky et al. (2014) Adv. Mater. 26, 2966–2966; Kolesky et al. (2016) Proc National Acad Sci 113, 3179–3184). To generate hydrogel filaments containing organoid cells, called SOBAs (syringe-extruded organoid BME assemblies), cold cell-BME solutions were loaded into a syringe with a 15-gauge (1.37 mm inner diameter) blunt-tip needle and then injected directly into warm medium while moving the tip across the XY plane ( e.g. , in a linear, serpentine, or spiral pattern). Silk fragments that more closely resemble the BOBA geometry and are termed SOBA fragments were also generated by gently triturating SOBA cultures with a wide-bore P1000 tip or a 10 mL serological pipette.
在 BOBA、SOBA 或 SOBA 碎片培養物中生長 9 天之類器官顯示出相似生長,如藉由明視野顯微術 (圖6A)、類器官直徑測量 (圖6B 至 6C) 及活細胞計數 (圖6D)。相對於 BOBA 小滴,SOBA 及 SOBA 碎片形式產生可比較的類器官生長,同時實現更快且更省力之培養物製備。與 SOBA 培養物相比,SOBA 碎片更均勻地分散在培養基中,從而使單一培養物更容易分割、采樣或等分。Organoids grown in BOBA, SOBA, or SOBA fragment cultures for 9 days showed similar growth as measured by bright field microscopy (Figure 6A), organoid diameter measurement (Figures 6B to 6C), and viable cell counts (Figure 6D). SOBA and SOBA fragment formats produced comparable organoid growth relative to BOBA droplets while enabling faster and less laborious culture preparation. SOBA fragments were more evenly dispersed in the medium compared to SOBA cultures, making it easier to divide, sample, or aliquot a single culture.
討論:Discuss:
該實例描述了用於人類腸上皮類器官之經懸浮之 BME 水凝膠培養方法之開發,該方法克服了若干與習用表面接附之圓頂水凝膠培養方法相關之挑戰。與圓頂方法相比,BOBA、SOBA 及 SOBA 絲方法簡化並加快了協議,使得能夠與可擴展之培養容器相容,允許動態學培養採樣並改善類器官培養均勻性。This example describes the development of a suspended BME hydrogel culture method for human intestinal epithelial organoids that overcomes several challenges associated with conventional surface-attached dome hydrogel culture methods. Compared to the dome method, the BOBA, SOBA, and SOBA-filament methods simplify and expedite the protocol, enable compatibility with scalable culture vessels, allow for kinetic culture sampling, and improve organoid culture uniformity.
已經針對腸類器官及類腫瘤規模化提出了若干懸浮培養協議,但在這些方法中,類器官係在含有不會形成完整水凝膠之低濃度溶解 MATRIGEL® (類似於 BME) 之液體培養基中培養的 (Hirokawa 等人(2021) Commun Biology 4, 1067;Price 等人(2022) Sci Rep-Uk 12, 5571)。這與目前所揭示之方法形成對比,該方法依賴於形成完全固化的不溶性水凝膠。此外,先前研究顯示,雖然類器官及類腫瘤可在 5% 可溶性 MATRIGEL® 溶液中生長,但濃度高於 5% 會導致生長減少。此外,在 5% MATRIGEL® 中生長之腸類器官表現出反向上皮極性 (Hirokawa 等人(2021)),這與先前關於低 ECM 條件下類器官極性反轉之報告一致 (Co 等人(2019) Cell Reports 26, 2509-2520.e4)。Several suspension culture protocols have been proposed for intestinal organoid and tumorigenesis scale-up, but in these approaches, organoids are cultured in liquid media containing low concentrations of dissolved MATRIGEL® (similar to BME) that do not form complete hydrogels (Hirokawa et al. (2021) Commun Biology 4, 1067; Price et al. (2022) Sci Rep-Uk 12, 5571). This is in contrast to the currently described method, which relies on the formation of a fully solidified insoluble hydrogel. In addition, previous studies have shown that while organoids and tumorigenesis can be grown in 5% soluble MATRIGEL® solutions, concentrations above 5% result in reduced growth. Furthermore, intestinal organoids grown in 5% MATRIGEL® exhibited reversed epithelial polarity (Hirokawa et al. (2021)), which is consistent with previous reports of organoid polarity reversal under low ECM conditions (Co et al. (2019) Cell Reports 26, 2509-2520.e4).
與習用圓頂方法相比,BOBA 方法提供了若干優點。首先,它簡化了協議。由於水凝膠直接在培養基中固化,因此無需單獨固化培育步驟,從而節省時間及勞力。其次,它減少了容器表面積量及平鋪圓頂所需之技術精度。藉由減輕對可用表面積之依賴,BOBA 方法利用了培養容器之所有 3 個維度,從而增加了水凝膠體積及每個培養物之類器官細胞。BOBA 方法有助於大幅規模化培養,因為它使得能夠與燒瓶相容,燒瓶可得尺寸較大,更易於操作,並且使得能夠快速更換培養基。例如,在 BOBA 培養中,可在單一 225 cm
2燒瓶中培養 10 mL BME,並且可簡單藉由以血清移液管更換上清液來更換培養基。然而,使用圓頂方法,等同培養物需要九個 24 孔盤 (50 µL 圓頂之 200 孔),並且需要為各孔單獨更換培養基。這也使塑膠消耗量減少超過 70% (225 cm
2燒瓶含有 152.7 g 塑膠,且代替九個 24 孔盤中之 562.5 g 塑膠,資料未顯示)。可使用可容納數升培養物體積之多層燒瓶或「細胞工廠」來實現進一步規模化。表 1 提供了針對若干容器類型,所揭示之經懸浮之 BME 水凝膠培養設置之一般指南。據報導,培養物容器類型會影響如氣體傳輸的參數 (Allen 等人(2001) Am J Physiol-Lung C 281, L1021–L1027),以及諸如培養基配方、水凝膠組成及類器官株特異性生長速率的因素都可影響類器官生長。
表 1
BOBA 方法也克服了圓頂培養物中產生的因水凝膠擴散限製而產生之培養物異質性 (Park 等人(2022);Shin 等人(2020))。一些協議建議平鋪較小之 10-15 µL 水凝膠圓頂 (Pleguezuelos-Manzano 等人(2020);Stewart 等人(2020) Methods Mol Biology 2121, 185–198),由此減少分子轉運之擴散路徑。然而,即使在每孔平鋪多個圓頂時,平鋪較小圓頂也會減少每孔之水凝膠總體積。克服水凝膠擴散限制之另一方法係在水凝膠固化步驟過程中翻轉盤,使重力引起細胞沉降在圓頂頂部表面附近,其中圓頂核心中幾乎沒有或沒有細胞 (Pleguezuelos-Manzano 等人(2020))。這導致昂貴水凝膠體積之次優使用,並且最終每 µL 水凝膠可接種之類器官細胞較少。已經開發出複雜生物工程方法來增加水凝膠表面積以改善分子轉運 (Park 等人(2022)),但這些難以規模化,並且仍然依賴將水凝膠錨定到 2D 表面。BOBA 方法產生更均質的培養物,而不會犧牲每孔之水凝膠量或每 µL 水凝膠之細胞數量。不限於特定理論,所觀察到之類器官培養均勻性改善可藉由 BOBA 水凝膠來解釋,該 BOBA 水凝膠具有 (1) 較小直徑,且因此較短擴散路徑,及 (2) 所有外表面都曝露於培養基中,使得分子甚至能夠均勻擴散到水凝膠中。The BOBA approach also overcomes the culture heterogeneity that arises from hydrogel diffusion limitations in dome cultures (Park et al. (2022); Shin et al. (2020)). Some protocols recommend plating smaller 10-15 µL hydrogel domes (Pleguezuelos-Manzano et al. (2020); Stewart et al. (2020) Methods Mol Biology 2121, 185–198) to reduce diffusion pathways for molecular transport. However, plating smaller domes reduces the total volume of hydrogel per well, even when plating multiple domes per well. Another approach to overcome the diffusion limitations of hydrogels is to flip the dish during the hydrogel solidification step, allowing gravity to cause cells to settle near the top surface of the dome, with few or no cells in the dome core (Pleguezuelos-Manzano et al. (2020)). This results in suboptimal use of expensive hydrogel volume and ultimately fewer organoid cells can be seeded per µL of hydrogel. Complex bioengineering methods have been developed to increase hydrogel surface area to improve molecular transport (Park et al. (2022)), but these are difficult to scale and still rely on anchoring the hydrogel to a 2D surface. The BOBA approach produces more homogeneous cultures without sacrificing the amount of hydrogel per well or the number of cells per µL of hydrogel. Without being bound by a particular theory, the observed improvement in organoid culture uniformity may be explained by BOBA hydrogels having (1) smaller diameters and therefore shorter diffusion pathways, and (2) all external surfaces exposed to the culture medium, allowing for even diffusion of molecules into the hydrogel.
水凝膠圓頂中類器官形態異質性之另一有記錄之挑戰係位於盤底部附近之類器官可接附於盤表面、展開及變平,並失去其 3D 結構 (Pleguezuelos-Manzano 等人(2020);Price 等人(2022))。由於經懸浮之水凝膠小滴不會與盤表面直接接觸,因此不會發生類器官展開及變平。Another documented challenge to organoid morphological heterogeneity in hydrogel domes is that organoids located near the bottom of the dish can attach to the dish surface, unfold and flatten, and lose their 3D structure (Pleguezuelos-Manzano et al. (2020); Price et al. (2022)). Since the suspended hydrogel droplets do not come into direct contact with the dish surface, organoid unfolding and flattening do not occur.
除了 BOBA 水凝膠小滴外,類器官還可以生長為 SOBA 絲及 SOBA 絲碎片。類器官在所有 3 種配置中的生長相似,證明了懸浮 BME 培養方法之穩健性。SOBA 方法有利於並加快培養物製備,因為可將大體積細胞-BME 溶液加載到單一注射器中以產生 SOBA 水凝膠絲。一分鐘內可產生約 10 mL SOBA,而等同圓頂培養則需要超過 15 分鐘進行平鋪,並額外需要 15 至 30 分鐘進行固化。儘管 SOBA 之長度比 BOBA 長得多,但沒有觀察到類器官生長之異質性 (如在圓頂培養物中觀察到的),這可能係因為 SOBA 直徑較小 (通常小於 2 mm) 並且使得能夠從培養基有效轉運營養物。SOBA 碎片在尺寸及幾何形狀上更密切接近 BOBA,但產生速度要快得多。與 BOBA 一樣,SOBA 碎片均勻地分散在整個培養物中,這對於動態採樣或劃分培養物以進行多次應用或讀取可能係有用的。In addition to BOBA hydrogel droplets, organoids were grown as SOBA filaments and SOBA filament fragments. Organoid growth was similar in all 3 configurations, demonstrating the robustness of the suspended BME culture method. The SOBA method facilitates and speeds up culture preparation because a large volume of cell-BME solution can be loaded into a single syringe to produce SOBA hydrogel filaments. Approximately 10 mL of SOBA can be produced in one minute, while the equivalent dome culture requires more than 15 minutes for leveling and an additional 15 to 30 minutes for solidification. Despite the much longer length of SOBAs than BOBAs, no heterogeneity in organoid growth (as observed in dome cultures) was observed, likely due to the smaller diameter of SOBAs (typically <2 mm) and the ability to efficiently transport nutrients from the culture medium. SOBA fragments more closely resemble BOBAs in size and geometry, but are generated much more rapidly. Like BOBAs, SOBA fragments are evenly dispersed throughout the culture, which may be useful for dynamic sampling or for partitioning cultures for multiple applications or readouts.
整體而言,經懸浮之水凝膠培養方法使得能夠進行大規模類器官規模化,改善類器官培養均勻性,並節省勞力、時間及資源。這些培養改善使腸類器官更適合於實施如化合物或基因體篩選的高生產量應用。該方法可能擴展到培養患病腸類器官及類腫瘤、來自其他物種之腸類器官以及來自不同組織類型之類器官。因此,BOBA、SOBA 及 SOBA 絲有潛力推動及擴大類器官技術之採用。Overall, the suspended hydrogel culture method enables large-scale organoid scalability, improves organoid culture uniformity, and saves labor, time, and resources. These culture improvements make intestinal organoids more suitable for high-throughput applications such as compound or genomic screening. The method may be extended to the culture of diseased intestinal organoids and tumor-like structures, intestinal organoids from other species, and organoids from different tissue types. Therefore, BOBA, SOBA, and SOBA filaments have the potential to promote and expand the adoption of organoid technology.
實例Examples 22 :類器官在篩選方法中之用途:The use of organoids in screening methods
該實例揭示了藉由實例 1 之方法產生之腸類器官在細胞毒性測定中之用途。This example discloses the use of intestinal organoids produced by the method of Example 1 in cytotoxicity assays.
方法:method:
96 孔盤懸浮之 BME 類器官細胞毒性測定。 將來自均質化大腸類器官 SOBA 碎片之兩個 96 孔盤以每孔 90 µl 接種到225 cm 2燒瓶中。將化合物原液在 Intesticult OGM 中稀釋至 10 倍最終濃度,並向各孔中添加 10 µL。對從 100 µM 開始 5 倍稀釋之 8 劑量稀釋系列在技術上一式四份評估。3 天處理後,使用 Cell Titer Glo 3D 測定套組 (Promega) 進行生存力測量,並在 Ensight 酶標儀 (Perkin Elmer) 上測量發光。使用 Prism (GraphPad) 產生 4PL 擬合曲線。 Cytotoxicity assay of suspended BME organoids in 96 -well plates. Two 96-well plates were seeded with 90 µl per well from homogenized colon organoid SOBA fragments into 225 cm2 flasks. Compound stocks were diluted to 10-fold final concentration in Intesticult OGM and 10 µL was added to each well. An 8-dose dilution series starting at 100 µM with 5-fold dilutions was evaluated in technical quadruplicate. After 3 days of treatment, viability was measured using the Cell Titer Glo 3D assay kit (Promega) and luminescence was measured on an Ensight ELISA reader (Perkin Elmer). 4PL fit curves were generated using Prism (GraphPad).
結果:result:
將懸浮之 BME 水凝膠類器官在藥物毒性篩選測定中之應用。 以 BOBA、SOBA 或 SOBA 碎片形式產生之腸類器官可直接用於下游測定,無需額外類器官消化或傳代步驟。作為概念驗證,在藥物毒性篩檢中證明了這些類器官之實施。SOBA 碎片培養之類器官在 225 cm 2燒瓶中生長 (圖7A-7B),藉由以血清移液管研磨均質化,然後轉移至 96 孔盤 (圖7A-7C)。藉由基於 Cell Titer Glo 3D ATP 之生存力測定測量之孔間變異性與平鋪在 96 孔盤中之 SOBA 絲生長類器官及圓頂培養物係可比較的 (圖7D)。 Application of suspended BME hydrogel organoids in drug toxicity screening assays. Intestinal organoids generated as BOBA, SOBA, or SOBA fragments can be used directly in downstream assays without the need for additional organoid digestion or passaging steps. As a proof of concept, the implementation of these organoids was demonstrated in drug toxicity screening. Organoids cultured in SOBA fragments were grown in 225 cm2 flasks (Figures 7A-7B), homogenized by trituration with a serological pipette, and then transferred to 96-well plates (Figures 7A-7C). Inter-well variability measured by the Cell Titer Glo 3D ATP-based viability assay was comparable to SOBA filament-grown organoids and dome cultures plated in 96-well plates ( FIG. 7D ).
對於藥物毒性篩選,將 96 孔盤中之 SOBA 絲生長類器官以已知引起腸毒性之化合物 (雙醋瑞因、索拉非尼、SN-38 或多西紫杉醇) 或 DMSO 媒介物對照處理 3 天。生存力被確定為 ATP 之量度並產生劑量反應曲線 (圖7E)。這係經懸浮之 BME 水凝膠培養物如何直接用於下游應用之一個實例。For drug toxicity screening, SOBA filament-grown organoids in 96-well plates were treated for 3 days with compounds known to cause enterotoxicity (diacerein, sorafenib, SN-38, or docetaxel) or DMSO vehicle control. Viability was determined as a measure of ATP and a dose-response curve was generated (Figure 7E). This is an example of how suspended BME hydrogel cultures can be used directly in downstream applications.
經懸浮之 BME 水凝膠生長培養物之實用性在此處在兩個應用中得到了證明。首先,SOBA 絲培養物可以經均質化以產生低孔間類器官變異性之 96 孔盤培養物。作為概念驗證,進行了藥物毒性篩檢;此方法可用於其他中高生產量篩選。The utility of suspended BME hydrogel-grown cultures was demonstrated here in two applications. First, SOBA filament cultures could be homogenized to generate 96-well plate cultures with low well-to-well organoid variability. As a proof of concept, drug toxicity screening was performed; this approach can be applied to other medium- to high-throughput screens.
實例Examples 33 :類器官來源模型之產生:Generation of organoid-derived models
該實例揭示了藉由實例 1 之方法產生之腸類器官在提供替代類器官來源模型中之用途。This example discloses the use of intestinal organoids produced by the method of Example 1 in providing an alternative organoid source model.
方法:method:
Transwell 單層。 將 225 cm 2燒瓶中之大腸類器官 SOBA 碎片收集到 50 mL 錐形管中,並以 800 x g 沉澱 3 分鐘。移除上清液,並藉由在 TrypLE Express 中於 37°C 水浴中培育 10 分鐘,將類器官消化成單細胞,然後用 P1000 移液管研磨。根據需要重複培育及研磨最多 2 次。將細胞沉澱並以 PBS 洗滌,然後重懸於單層生長培養基 (Intesticult OGM + 10 µM Y-27632) 或單層分化培養基 (Intesticult ODM + 10 µM Y-27632) 中。對於 96 孔 PET Transwell 盤 (孔徑 0.4 µm,Corning 目錄號 3450) 之各孔,將 100 µL 培養基中之 2.0 x 10 5個細胞接種到頂部室中,並將 200 µL 培養基添加到基部室中。兩個室中之培養基每 2 至 3 天一次更換。使用 10X 物鏡以及 THUNDER 顯微鏡 (Leica) 與 DFC9000 GTC 相機 (Leica) 進行明視野成像。藉由以伏特/歐姆計 (EVOM3, WPI) 測量電阻,然後將電阻乘以 Transwell 表面積 (0.143 cm 2) 來確定跨上皮電阻 (TEER)。 Transwell monolayer. Collect colon organoid SOBA fragments from 225 cm2 flasks into 50 mL conical tubes and pellet at 800 xg for 3 min. Remove supernatant and digest organoids into single cells by incubating in TrypLE Express in a 37°C water bath for 10 min, then triturate with a P1000 pipette. Repeat incubation and trituration up to 2 times as needed. Pellet cells and wash with PBS, then resuspend in monolayer growth medium (Intesticult OGM + 10 µM Y-27632) or monolayer differentiation medium (Intesticult ODM + 10 µM Y-27632). For each well of a 96-well PET Transwell plate (pore size 0.4 µm, Corning catalog #3450), 2.0 x 10 5 cells in 100 µL of medium were seeded into the apical chamber, and 200 µL of medium was added to the basal chamber. The medium in both chambers was changed every 2 to 3 days. Bright field imaging was performed using a 10X objective and a THUNDER microscope (Leica) and a DFC9000 GTC camera (Leica). Transepithelial electrical resistance (TEER) was determined by measuring the electrical resistance with a volt/ohmmeter (EVOM3, WPI) and then multiplying the resistance by the Transwell surface area (0.143 cm 2 ).
結果:result:
經懸浮 BME 之水凝膠培養物有利於替代類器官來源模型。 除了直接以其經懸浮之 BME 水凝膠形式使用類器官外,這些類器官還可以有利於產生其他類器官來源模型,尤其係該等需要大細胞輸入之模型,如單層及微生理系統 (MPS) 裝置。經懸浮之 BME 水凝膠類器官用於產生大腸上皮 Transwell 單層,並在兩種條件下評估上皮障壁功能。將於 225 cm 2燒瓶中培養之 SOBA 絲類器官消化成單細胞,在匯合時接種在 96 孔 Transwell 小室上,並在單層生長或單層分化培養基中培養 7 天 (圖8A)。這兩種條件產生具有不同形態之 Transwell 上皮培養物:在生長培養基中,所得上皮形成從單層表面突出之 3D 結構,而在分化培養基中,這些結構不存在,並且可見具有成熟緊密連接之分化上皮細胞之典型「鵝卵石」細胞形態 (圖8B)。與此形態一致,到第 3 天時,與生長培養基相比,分化培養基中之 Transwell 單層之藉由跨上皮電阻 (TEER) 測量定量之上皮障壁功能較高 (圖8C)。該實驗為使用 BOBA、SOBA 或 SOBA 絲方法生長之類器官如何能夠產生替代腸類器官來源之 活體外模型之實例。 Suspended BME hydrogel cultures facilitate alternative organoid-derived models. In addition to using organoids directly in their suspended BME hydrogel format, these organoids can facilitate the generation of other organoid-derived models, particularly those that require large cell input, such as monolayers and microphysiological system (MPS) devices. Suspended BME hydrogel organoids were used to generate colon epithelial Transwell monolayers and assess epithelial barrier function under two conditions. SOBA filament organoids cultured in 225 cm2 flasks were digested into single cells, plated on 96-well Transwell inserts at confluence, and cultured for 7 days in either monolayer growth or monolayer differentiation media (Figure 8A). The two conditions produced Transwell epithelial cultures with different morphologies: in growth medium, the resulting epithelium formed 3D structures protruding from the monolayer surface, whereas in differentiation medium these structures were absent and the typical "cobblestone" cell morphology of differentiated epithelial cells with mature tight junctions was seen (Figure 8B). Consistent with this morphology, by day 3, epithelial barrier function, as quantified by transepithelial electrical resistance (TEER) measurements, was higher in Transwell monolayers in differentiation medium compared to growth medium (Figure 8C). This experiment is an example of how organoids grown using the BOBA, SOBA, or SOBA filament approach can generate an in vitro model of an alternative source of intestinal organoids.
SOBA 絲類器官可用於產生類器官來源之 Transwell 單層,其提供同時進入頂端及基底外側之優勢,但難以規模化,因為它們需要大量細胞來接種。藉由實現大量類器官擴增,經懸浮之 BME 水凝膠培養方法可有利於具有更高組織傳真度及實驗優勢之複雜腸類器官來源模型之開發及實施。SOBA filament organoids can be used to generate organoid-derived Transwell monolayers, which offer the advantage of access to both the apical and basolateral sides, but are difficult to scale up because they require large numbers of cells for seeding. By enabling large-scale organoid expansion, the suspended BME hydrogel culture method may facilitate the development and implementation of complex intestinal organoid-derived models with greater tissue fidelity and experimental advantages.
實例Examples 44 :肺類器官之產生:Generation of lung organoids
該實例揭示了使用本文所述之 BME 懸浮方法產生肺類器官。This example discloses the generation of lung organoids using the BME suspension method described herein.
方法:method:
人類肺 ATII 細胞分離及類器官來源。 來自已故供體之未鑑定之人類肺組織藉由西部供體網路獲得。先前描述了肺解離為單細胞及類器官建立 (Konishi 等人,2022)。簡而言之,以 HBSS 緩衝液洗滌組織並以消化緩衝液 (I 型膠原酶:450 單位/mL;分散酶:5 單位/mL;DNase I:10 單位/mL) 膨脹。去除胸膜及小氣道後,用單刃刀片切碎剩餘組織,轉移到含有溫熱消化緩衝液之標準管中,且在每 15 分鐘一次攪拌及劇烈混合下於 37℃ 培育總共 1 小時。使溶液通過 100 µm 細胞過濾器,並在 4℃ 以 450 g 沉澱 10 分鐘。將細胞沉澱重懸於 5 mL ACK 緩衝液中以裂解紅血球,並在室溫培育 3 至 5 分鐘。藉由添加 DMEM/F12+10%FBS 來終止反應。隨後將細胞懸浮液透過 40 µm 細胞過濾器過濾,並在 4℃ 以 450g 沉澱 5 分鐘。然後以 CD45 磁性微珠 (Miltenyi Biotech,目錄號 130-045-801) 標記細胞,並將該細胞加載到 MACS® 柱上,將該柱放置在 MACS 分離器之磁場中 (根據製造協議)。磁性標記之 CD45+ 細胞保留在柱內並丟棄,並收集通過柱之未標記細胞。將這些細胞在 Gentle MACS 緩衝液中洗滌,在 4℃ 以 450g 沉澱 5 分鐘,並與含有 50 nM Lysotracker 綠 (Invitrogen,目錄號 L7526) 之 DMEM+10%FBS 一起在 37℃ 培育 30 分鐘。將以下抗體添加到冰上之細胞中,持續 30 分鐘:預結合之 HTII280+Alexa 647 (Terrance Biotech;Invitrogen 目錄號 A20186); EpcamPE-Cy7 (BioLegend,目錄號 324222); CD45-Alexa700 (BioLegend,目錄號 304024),CD31-Alexa594 (BioLegend,目錄號 303126)。亦添加了活/死染色探針 Sytox 藍 (Invitrogen)。然後將細胞沉澱、洗滌、重懸於 PBS+2%FBS 中,並藉由針對 Sytox 藍-、CD45-、CD31-、EpCAM+、HTII-280+、lysotracker 綠+ 進行分選來分離 ATII 細胞。將 ATII 細胞重懸於 MATRIGEL® (低生長因子,不含酚紅;Corning,目錄號 356231) 中,平鋪於 6 孔盤之 50 µl 圓頂中 (60 x 10 4個細胞/mL,或 3000 個細胞/50 µl MATRIGEL®),在 37°C 固化 30 分鐘,並且然後以含有 10 µM ROCK 抑制劑 Y27632 (Selleckchem,目錄號 S1049) 之 3 ml 無血清無飼養層 (SFFF) 培養基 (Konishi 等人,2022) 覆蓋 3 天。培養前 2 至 3 天後,每 2 至 3 天一次更換不含 ROCK 抑制劑之 SFFF 培養基。 Human lung ATII cell isolation and organoid source. Unidentified human lung tissue from deceased donors was obtained through the Western Donor Network. Lung dissociation for single cells and organoid establishment has been described previously (Konishi et al., 2022). Briefly, tissue was washed with HBSS buffer and expanded with digestion buffer (collagenase type I: 450 units/mL; dispase: 5 units/mL; DNase I: 10 units/mL). After removal of the pleura and small airways, the remaining tissue was minced with a single-edged blade, transferred to a standard tube containing warm digestion buffer, and incubated at 37°C for a total of 1 hour with stirring and vigorous mixing every 15 minutes. Pass the solution through a 100 µm cell filter and pellet at 450 g for 10 minutes at 4°C. Resuspend the cell pellet in 5 mL ACK buffer to lyse red blood cells and incubate at room temperature for 3 to 5 minutes. Terminate the reaction by adding DMEM/F12+10%FBS. Then filter the cell suspension through a 40 µm cell filter and pellet at 450 g for 5 minutes at 4°C. The cells were then labeled with CD45 magnetic microbeads (Miltenyi Biotech, catalog number 130-045-801) and loaded onto a MACS® column, which was placed in the magnetic field of a MACS separator (according to the manufacturer's protocol). The magnetically labeled CD45+ cells were retained in the column and discarded, and the unlabeled cells that passed through the column were collected. These cells were washed in Gentle MACS buffer, pelleted at 450g for 5 minutes at 4°C, and incubated with DMEM + 10% FBS containing 50 nM Lysotracker Green (Invitrogen, catalog number L7526) at 37°C for 30 minutes. The following antibodies were added to the cells on ice for 30 minutes: pre-conjugated HTII280+Alexa 647 (Terrance Biotech; Invitrogen Catalog No. A20186); EpcamPE-Cy7 (BioLegend, Catalog No. 324222); CD45-Alexa700 (BioLegend, Catalog No. 304024), CD31-Alexa594 (BioLegend, Catalog No. 303126). The live/dead staining probe Sytox Blue (Invitrogen) was also added. Cells were then pelleted, washed, resuspended in PBS + 2% FBS, and ATII cells were isolated by sorting against Sytox Blue-, CD45-, CD31-, EpCAM+, HTII-280+, lysotracker Green+. ATII cells were resuspended in MATRIGEL® (reduced growth factor, without phenol red; Corning, catalog number 356231), plated in 50 µl domes in 6-well plates (60 x 10 4 cells/mL, or 3000 cells/50 µl MATRIGEL®), fixed at 37°C for 30 min, and then overlaid with 3 ml serum-free feeder-free (SFFF) medium (Konishi et al., 2022) containing 10 µM ROCK inhibitor Y27632 (Selleckchem, catalog number S1049) for 3 days. After the first 2 to 3 days of culture, SFFF medium without ROCK inhibitor was replaced every 2 to 3 days.
類器官維持。 類器官培養物每 10 至 14 天一次傳代。為了傳代,首先將培養物與 Accutase (Cell Stem Cell Technologies,目錄號 07922) 一起培育 15 分鐘以軟化 MATRIGEL®。然後將圓頂及類器官收集在 15 ml 管中,並在 37°C 攪拌下培育 15 分鐘。在 4°C 以 450g 離心 5 分鐘後,以 TrypLE Express (Gibco,目錄號 12604-021) 在 37°C 處理細胞 10 分鐘。TrypLE Express 藉由以 PBS 稀釋來滅活,並在 4°C 以 450 x g 沉澱細胞 5 分鐘。將細胞以 60 x 10 4個細胞/mL 重懸於冰上之 MATRIGEL® 中,平鋪於 6 孔盤之 50 µL 圓頂中,並在 37°C 固化 30 分鐘。前 3 天以含有 10 µM ROCK 抑制劑之 SFFF 培養基覆蓋圓頂,並且然後添加不含 ROCK 抑制劑之 SFFF 培養基,且每 2 至 3 天一次更換。 Organoid maintenance. Organoid cultures were passaged every 10 to 14 days. For passaging, cultures were first incubated with Accutase (Cell Stem Cell Technologies, catalog #07922) for 15 minutes to soften the MATRIGEL®. Domes and organoids were then collected in 15 ml tubes and incubated at 37°C for 15 minutes with agitation. After centrifugation at 450 g for 5 minutes at 4°C, cells were treated with TrypLE Express (Gibco, catalog #12604-021) for 10 minutes at 37°C. TrypLE Express was inactivated by diluting with PBS and the cells were pelleted at 450 x g for 5 minutes at 4°C. Cells were resuspended in MATRIGEL® on ice at 60 x 104 cells/mL, plated in 50 µL domes in 6-well plates, and fixed at 37°C for 30 minutes. Domes were covered with SFFF medium containing 10 µM ROCK inhibitor for the first 3 days and then SFFF medium without ROCK inhibitor was added and changed every 2 to 3 days.
經懸浮之水凝膠 BOBA 培養物。 如上所述,在冰上製備 MATRIGEL® 中之單類器官細胞。將含有 10 µM ROCK 抑制劑之預熱 SFFF 培養基添加至 6 孔盤 (4 mL/孔) 中,並保持在 37°C 溫浴上。為了產生經懸浮之 BME 小滴或 BOBA,使用帶有寬孔或切割移液管尖端 (開口約 2 mm) 之電子連續移液管 (參考) 將類器官細胞-MATRIGEL® 溶液以10 µL 體積直接分配到溫熱培養基中。在分配過程中,尖端立即浸入液體表面下方,然後在每次分配後提起,以確保小滴分離。3 天後,添加 SFFF 培養基並每 2 至 3 天一次更換。對於培養基交換,將無菌 70 µm 細胞過濾器放入孔中,傾斜盤並透過過濾器輕輕吸出 3 mL 培養基。 Suspended hydrogel BOBA cultures. Single organoids in MATRIGEL® were prepared on ice as described above. Pre-warmed SFFF medium containing 10 µM ROCK inhibitor was added to a 6-well plate (4 mL/well) and kept in a 37°C incubator. To generate suspended BME droplets or BOBA, the organoid-MATRIGEL® solution was dispensed in 10 µL volumes directly into the warm medium using an electronic continuous pipette (reference) with a wide-bore or cut pipette tip (opening approximately 2 mm). During the dispensing process, the tip was immediately dipped below the liquid surface and then lifted after each dispense to ensure droplet separation. After 3 days, add SFFF medium and change it every 2 to 3 days. For medium exchange, place a sterile 70 µm cell filter into the well, tilt the plate and gently aspirate 3 mL of medium through the filter.
結果:result:
如圖9 所示,使用分離自嵌入懸浮於培養基中之水凝膠中的肺組織之肺泡 II 型 (ATII) 幹細胞成功產生肺類器官。As shown in Figure 9, lung organoids were successfully generated using alveolar type II (ATII) stem cells isolated from lung tissue embedded in hydrogel suspended in culture medium.
實例Examples 55 :乳腺類器官之產生:Generation of Mammary Organoids
該實例提供了使用本文所述之 BME 懸浮方法產生乳腺類器官之方法。This example provides a method for generating mammary organoids using the BME suspension method described herein.
方法:method:
人類乳腺類器官來源。 乳腺類器官如 Sachs 等人Cell 172(1-2):373-389.e10 (2018) 之前所述來源,其內容全文併入本文中。將乳腺組織切成 1-3 mm 3塊,切碎並以 10 mL AdDF+++ (含有 1x Glutamax、10 mM HEPES 及抗生素之 Advanced DMEM/F12) 洗滌。隨後將組織在迴旋振盪器上在含有 1 至 2 mg/ml 膠原蛋白酶 (Sigma,C9407) 之 10 mL 類器官培養基中於 37°C 消化 1 至 2 小時。使用塑膠及火焰玻璃 Pasteur 移液管依序剪切消化組織懸浮液,並且在各剪切步驟後,將懸浮液在過濾器 ( 例如,100 μm 過濾器) 上過濾。以約 10ml AdDF+++ 對過濾後保留之組織塊進行後續剪切步驟。將 2% FCS 添加到過濾懸浮液中,且然後以 400 rcf 離心 。將沉澱重懸於 10 ml AdDF+++ 中,並再次以 400 rcf 離心 。如果可見紅色沉澱,則將紅血球在 2 mL 紅血球裂解緩衝液 (Roche,11814389001) 中在室溫裂解 5 分鐘,然後添加 10 ml AdDF+++ 並以 400 rcf 離心 。將沉澱重懸於 10 mg/ml 冷 BME ( 例如,CULTREX® 低生長因子 BME 2 型 (Trevigen,3533-010-02)) 中,並使 40 µL BME-細胞懸浮液滴在預熱 24 孔懸浮液培養盤 (Greiner,M9312) 上於 37°C 固化 20 分鐘。凝膠化完成時,向各孔添加 400 µL 類器官培養基,並將盤轉移至 2% 或環境 O 2之濕潤 37°C/5% CO 2培養箱中。培養基每 4 天一次更換。類器官培養基含有 R-Spondin 1 條件培養基 (10%) 或 R-Spondin 3 (250 ng/ml) 與神經調節蛋白 1 (5 nM)、FGF7 (5 ng/ml)、FGF10 (20 ng/ ml)、EGF (5 ng/ml)、Noggin (100 ng/ml)、A83-01 (500 nM)、Y-27632 (5 µM)、SB202190 (500 nM)、B27 補充劑 (1x)、N-乙醯半胱胺酸 (1.25 mM)、菸鹼醯胺 (5 mM)、GlutaMax 100x (1x)、HEPES (10 nM)、青黴素/鏈黴素 (100 U/ml/100 µg)、Primocin (50 µg/ml) 及 Advanced DMEM/F12 (1x)。 Human mammary organoids were sourced. Mammary organoids were sourced as previously described by Sachs et al. Cell 172(1-2):373-389.e10 (2018), which is incorporated herein in its entirety. Mammary tissue was cut into 1-3 mm3 pieces, minced, and washed with 10 mL AdDF+++ (Advanced DMEM/F12 containing 1x Glutamax, 10 mM HEPES, and antibiotics). Tissues were then digested in 10 mL organoid medium containing 1 to 2 mg/ml collagenase (Sigma, C9407) at 37°C on an orbital shaker for 1 to 2 hours. Sequentially shear the digested tissue suspension using a plastic and flamed glass Pasteur pipette and after each shearing step, filter the suspension over a filter ( e.g. , 100 μm filter). Perform the subsequent shearing step on the tissue pieces retained after filtration with approximately 10 ml of AdDF+++. Add 2% FCS to the filtered suspension and then centrifuge at 400 rcf . Resuspend the pellet in 10 ml of AdDF+++ and centrifuge again at 400 rcf . If a red precipitate is visible, lyse the erythrocytes in 2 mL erythrocyte lysis buffer (Roche, 11814389001) at room temperature for 5 minutes, then add 10 ml AdDF+++ and centrifuge at 400 rcf . Resuspend the precipitate in 10 mg/ml cold BME ( e.g. , CULTREX® Reduced Growth Factor BME Type 2 (Trevigen, 3533-010-02)) and allow 40 µL of the BME-cell suspension to solidify on a pre-warmed 24-well suspension plate (Greiner, M9312) at 37°C for 20 minutes. When gelation is complete, add 400 µL of organoid medium to each well and transfer the plate to a humidified 37°C/5% CO 2 incubator with 2% or ambient O 2. Change the medium every 4 days. Organoid culture medium contained R-Spondin 1 conditioned medium (10%) or R-Spondin 3 (250 ng/ml) with neuromodulin 1 (5 nM), FGF7 (5 ng/ml), FGF10 (20 ng/ ml), EGF (5 ng/ml), Noggin (100 ng/ml), A83-01 (500 nM), Y-27632 (5 µM), SB202190 (500 nM), B27 supplement (1x), N-acetylcysteine (1.25 mM), niacinamide (5 mM), GlutaMax 100x (1x), HEPES (10 nM), penicillin/streptomycin (100 U/ml/100 µg), primocin (50 µg/ml) and Advanced DMEM/F12 (1x).
類器官維持。 類器官每 1 至 4 週一次傳代。將囊性類器官重懸於 2 mL 冷 AdDF+++ 中,並透過火焰玻璃 Pasteur 移液管進行機械剪切。緻密類器官藉由重懸於 2 mL TrypLE Express (Invitrogen,12605036) 中來解離,在室溫培育 1 至 5 分鐘,並藉由火焰玻璃 Pasteur 移液管進行機械剪切。添加 10 mL AdDF+++ 並以 300 rcf 或 400 rcf 離心後,將類器官碎片重懸於冷 BME 中,並以上述比率 ( 例如,1:1 至 1:6) 重新接種,從而形成新類器官。 Organoid maintenance. Organoids were passaged every 1 to 4 weeks. Cystic organoids were resuspended in 2 mL of cold AdDF+++ and mechanically sheared by a flamed glass Pasteur pipette. Delicate organoids were dissociated by resuspending in 2 mL of TrypLE Express (Invitrogen, 12605036), incubated at room temperature for 1 to 5 minutes, and mechanically sheared by a flamed glass Pasteur pipette. After adding 10 mL of AdDF+++ and centrifuging at 300 rcf or 400 rcf, organoid fragments were resuspended in cold BME and replated at the above ratios ( e.g. , 1:1 to 1:6) to form new organoids.
經懸浮之水凝膠 BOBA 培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱類器官培養基添加到 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15 至 30 mL/皿) 或 50 mL 錐形管 (30 mL/管) 中,並保持在 37°C 溫熱珠浴上。為了產生經懸浮之 BME 小滴或 BOBA,使用帶有寬孔或切割移液管尖端 (開口約 2 mm) 之電子連續移液管 (Integra VIAFLO 300) 將類器官細胞-BME 溶液以 10 µL 體積直接分配到溫熱培養基中。在分配過程中,尖端立即浸入液體表面下方,然後在各分配後提起,以確保小滴分離。對於較大形式培養物,藉由血清移液管或傾析將 BOBA 轉移至燒瓶中。培養基每 2 至 4 天一次更換。在 6 孔盤培養物中,將無菌 70 µm 細胞過濾器放入孔中,傾斜盤並透過過濾器輕輕吸出 4 mL 培養基。在燒瓶培養物中,將燒瓶傾斜一定角度,將 BOBA 放置在角落,然後以血清移液管更換大約 2/3 體積之已用培養基。 Suspended hydrogel BOBA cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed organoid medium to 6-well plates (5 mL/well), 100 cm dishes (15 to 30 mL/dish), or 50 mL conical tubes (30 mL/tube) and keep in a 37°C warm bead bath. To generate suspended BME droplets or BOBA, dispense the organoid cell-BME solution in 10 µL volumes directly into the warm medium using an electronic continuous pipette (Integra VIAFLO 300) with a wide-bore or cut pipette tip (opening approximately 2 mm). During dispensing, the tip is dipped immediately below the surface of the liquid and then lifted after each dispense to ensure droplet separation. For larger format cultures, transfer BOBA to flasks by serological pipette or decanting. Change medium every 2 to 4 days. In 6-well dish cultures, place a sterile 70 µm cell filter in the well, tilt the dish and gently aspirate 4 mL of medium through the filter. In flask cultures, tilt the flask at an angle, place BOBA in a corner, and replace approximately 2/3 of the volume of used medium with a serological pipette.
經懸浮之水凝膠 SOBA 及 SOBA 碎片培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱類器官培養基添加至 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15-30 mL/皿) 中,並保持在溫熱珠浴上。為了產生經懸浮之 SOBA 絲,將細胞-BME 溶液輕輕吸入帶有 15 號鈍尖端針之注射器中,然後直接擠出到溫熱培養基中,同時以 X-Y 平面中線形、蛇形或螺旋形運動方式移動浸沒針。為了產生 SOBA 碎片,使用 10 mL 血清移液管或寬孔 P1000 移液管尖端輕輕研磨 SOBA 絲培養物兩次。添加額外培養基以使最終 BME 與培養基之比率達到 1:10。如上對 BOBA 培養物所述進行培養基更換。 Suspended hydrogel SOBA and SOBA fragment cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed organoid medium to 6-well plates (5 mL/well), 100 cm dishes (15-30 mL/dish) and keep on a warm bead bath. To generate suspended SOBA filaments, gently draw the cell-BME solution into a syringe with a 15-gauge blunt-tip needle and extrude directly into the warm medium while moving the submerged needle in a linear, serpentine, or spiral motion in the XY plane. To generate SOBA fragments, gently triturate the SOBA filament culture twice using a 10 mL serological pipette or a wide-bore P1000 pipette tip. Additional medium was added to give a final BME to medium ratio of 1:10. Medium changes were performed as described above for BOBA cultures.
免疫螢光樣本製備及共焦顯微術。 24 孔圓頂培養物以於 PBS 中之 2% 多聚甲醛 (PFA) 固定。使用抹刀將圓頂從盤上分離,並使用切割 P1000 移液管尖端轉移到微量離心管中。對於 BOBA 培養物,使用切割 P1000 移液管尖端將 500 µL 培養物轉移至微量離心管中,除去培養基並添加於 PBS 中之 2% PFA。將樣本在固定劑中室溫培育 15 至 30 分鐘,然後在 PBS 中洗滌 3 次。將樣本在微量離心管中以在封閉/透化緩衝液 (3% BSA、0.1% Triton X-100、0.02% 疊氮化鈉,於 PBS 中) 中稀釋之初級抗體在室溫染色至少 4 小時,然後在 PBS 中洗滌 3 次。然後將樣本與於室溫在封閉/透化緩衝液中稀釋之二級抗體、DAPI 以及 AlexaFluor 660 蠅虎蕈鹼在室溫培育至少 2 小時。使用 40X 物鏡在 Stellaris 8 共焦顯微鏡 (Leica) 上收集影像,並使用 Imaris 影像分析軟體 (Oxford Instruments) 進行 3D 重建。 Immunofluorescence Sample Preparation and Confocal Microscopy. 24-well dome cultures were fixed with 2% paraformaldehyde (PFA) in PBS. Domes were detached from the plate using a spatula and transferred to a microcentrifuge tube using a cut P1000 pipette tip. For BOBA cultures, 500 µL of culture was transferred to a microcentrifuge tube using a cut P1000 pipette tip, the medium was removed and 2% PFA in PBS was added. Samples were incubated in fixative at room temperature for 15 to 30 minutes and then washed three times in PBS. Samples were stained in microcentrifuge tubes with primary antibodies diluted in blocking/permeabilization buffer (3% BSA, 0.1% Triton X-100, 0.02% sodium azide in PBS) for at least 4 hours at room temperature and then washed three times in PBS. Samples were then incubated with secondary antibodies, DAPI, and AlexaFluor 660 agaricus diluted in blocking/permeabilization buffer for at least 2 hours at room temperature. Images were collected on a Stellaris 8 confocal microscope (Leica) using a 40X objective and 3D reconstruction was performed using Imaris image analysis software (Oxford Instruments).
轉錄組分析。 對於 RNA 分離,使用 RNeasy Micro Plus 套組 (Qiagen)。將 RLT+ 裂解緩衝液添加至 BME 圓頂或沉澱 BOBA 中並儲存在 -80℃。使用 QiaCube Connect (Qiagen) 進行 RNA 分離,並使用 Nanodrop 8000 (ThermoFisher) 定量 RNA。批量 mRNA-seq (NovaSeq PE150) 及分析藉由 Novogene 進行。使用 HISAT2 (Mortazavi 等人 2008) 比對讀段,使用 DESeq2 (Anders 等人2014) 進行差異基因表現分析,並使用負二項分佈模型及 Benjamini-Hochberg FDR 校正計算統計顯著性。分析幹細胞及增生標記物 (EpCAM 及 CD49f +) 以及分化標記物 (細胞角蛋白 8 (K8)、細胞角蛋白 18 (K18)、細胞角質白 5 (k5)、細胞角蛋白 14 (K14)、平滑肌肌動蛋白 (SMA))。 Transcriptome analysis. For RNA isolation, the RNeasy Micro Plus kit (Qiagen) was used. RLT+ lysis buffer was added to BME domes or pelleted BOBA and stored at -80°C. RNA isolation was performed using QiaCube Connect (Qiagen), and RNA was quantified using Nanodrop 8000 (ThermoFisher). Bulk mRNA-seq (NovaSeq PE150) and analysis were performed by Novogene. Reads were aligned using HISAT2 (Mortazavi et al. 2008), differential gene expression analysis was performed using DESeq2 (Anders et al. 2014), and statistical significance was calculated using a negative binomial distribution model with Benjamini-Hochberg FDR correction. Stem cell and proliferation markers (EpCAM and CD49f + ) and differentiation markers (cytokeratin 8 (K8), cytokeratin 18 (K18), cytokeratin 5 (k5), cytokeratin 14 (K14), smooth muscle actin (SMA)) were analyzed.
實例Examples 66 :胰臟類器官之產生:Generation of pancreatic organoids
該實例提供了使用本文所述之 BME 懸浮方法產生胰臟類器官之方法。This example provides a method for generating pancreatic organoids using the BME suspension method described herein.
方法:method:
人類 胰臟類器官來源及維持。 胰臟類器官如 Georgakopoulos 等人BMC Developmental Biology 20:4 (2020) 之前所述產生,其內容全文併入本文中。將約 5 mg 胰臟組織手動切碎並以 gentleMACS 解離器 (Miltenyi Biotec) 解離總共 2 分鐘。將切碎組織在洗滌培養基中洗滌兩次,並在 40 mL 消化溶液中消化,並在 37℃ 放置 1 至 2 小時。洗滌溶液包括杜爾貝科改良伊格爾培養基 (DMEM)、高葡萄糖、GlutaMAX、丙酮酸鹽 (Life Technologies),其補充有 1% 胎牛血清 (FBS) (Life Technologies) 及 1% 青黴素/鏈黴素 (10,000 U/mL) (Life Technologies) Technologies),且消化溶液包括膠原蛋白酶 I 型 (Sigma-Aldrich) 及分散酶 II (Life Technologies),在含有 0.1 mg/mL DNase I (Sigma-Aldrich) 之 DMEM 中之濃度為 0.125 mg/mL。分離導管以移液管手工挑選,或以 100 μm 孔尼龍細胞過濾器 (Falcon) 過濾整個消化混合物。導管碎片在基礎培養基 (Advanced DMEM/F12 (Life Technologies),其補充有 1% 青黴素/鏈黴素、1% Glutamax 100x (Life Technologies) 及 HEPES (Life Technologies) 10 mM) 中洗滌,並以 200 g 旋轉 5 分鐘。將細胞沉澱與低生長因子 BME 2-RGF (基底膜萃取物 2 型 3533-010-02;AMSBIO,CULTREX®) 混合,接種於 24 孔盤中,並以優化人類胰臟類器官擴增培養基覆蓋。優化人類胰臟類器官擴增培養基包括基礎培養基,其補充有 1X N2 及 1X B27 (皆來自 GIBCO)、1.25 mM N-乙醯半胱胺酸 (Sigma-Aldrich)、10% RSPO1 條件無血清培養基、10 nM 人類 [Leu 15]-胃泌激素 I (Sigma-Aldrich)、50 ng/mL EGF (Peprotech)、25 ng/mL Noggin (Peprotech)、100 ng/mL FGF10 (Peprotech)、10 mM 菸鹼醯胺 (Sigma-Aldrich)、5 μM A83.01 (Tocris)、10 μM FSK (Tocris) 及 3 μM PGE2 (Tocris)。在前 7 天過程中,給優化人類胰臟類器官擴增培養基補充 10 μM Rho 激酶抑制劑 (Y27632,Sigma-Aldrich)。14 天後,如 Broutier 等人Nat Protoc.11:1724–43 (2016) 之前所述進行傳代,其內容全文併入本文中。 Human pancreatic organoids source and maintenance. Pancreatic organoids were generated as previously described by Georgakopoulos et al. BMC Developmental Biology 20:4 (2020), which is incorporated herein in its entirety. Approximately 5 mg of pancreatic tissue was minced manually and dissociated with a gentleMACS dissociator (Miltenyi Biotec) for a total of 2 minutes. The minced tissue was washed twice in wash medium and digested in 40 mL of digestion solution and placed at 37°C for 1 to 2 hours. The wash solution consisted of Dulbecco's modified Eagle's medium (DMEM), high glucose, GlutaMAX, pyruvate (Life Technologies) supplemented with 1% fetal bovine serum (FBS) (Life Technologies) and 1% penicillin/streptomycin (10,000 U/mL) (Life Technologies), and the digestion solution consisted of collagenase type I (Sigma-Aldrich) and dispase II (Life Technologies) at a concentration of 0.125 mg/mL in DMEM containing 0.1 mg/mL DNase I (Sigma-Aldrich). The detached tubes were either manually picked with a pipette or the entire digestion mixture was filtered with a 100 μm pore nylon cell filter (Falcon). Duct fragments were washed in basal medium (Advanced DMEM/F12 (Life Technologies) supplemented with 1% penicillin/streptomycin, 1% Glutamax 100x (Life Technologies), and HEPES (Life Technologies) 10 mM) and spun at 200 g for 5 min. Cell pellets were mixed with reduced growth factor BME 2-RGF (Basement Membrane Extract Type 2 3533-010-02; AMSBIO, CULTREX®), seeded in 24-well plates, and overlaid with optimized human pancreatic organoid expansion medium. Optimized human pancreatic organoid expansion medium included basal medium supplemented with 1X N2 and 1X B27 (both from GIBCO), 1.25 mM N-acetylcysteine (Sigma-Aldrich), 10% RSPO1 conditioned serum-free medium, 10 nM human [Leu 15 ]-gastrin I (Sigma-Aldrich), 50 ng/mL EGF (Peprotech), 25 ng/mL Noggin (Peprotech), 100 ng/mL FGF10 (Peprotech), 10 mM niacinamide (Sigma-Aldrich), 5 μM A83.01 (Tocris), 10 μM FSK (Tocris), and 3 μM PGE2 (Tocris). During the first 7 days, optimized human pancreatic organoid expansion medium was supplemented with 10 μM Rho kinase inhibitor (Y27632, Sigma-Aldrich). After 14 days, cells were passaged as previously described by Broutier et al. Nat Protoc. 11:1724–43 (2016), which is incorporated herein in its entirety.
經懸浮之水凝膠 BOBA 培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱培養基添加到 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15 至 30 mL/皿) 或 50 mL 錐形管 (30 mL/管) 中,並保持在 37°C 溫熱珠浴上。為了產生經懸浮之 BME 小滴或 BOBA,使用帶有寬孔或切割移液管尖端 (開口約 2 mm) 之電子連續移液管 (Integra VIAFLO 300) 將類器官細胞-BME 溶液以 10 µL 體積直接分配到溫熱培養基中。在分配過程中,尖端立即浸入液體表面下方,然後在各分配後提起,以確保小滴分離。對於較大形式培養物,藉由血清移液管或傾析將 BOBA 轉移至燒瓶中。培養基每 2 至 4 天一次更換。在 6 孔盤培養物中,將無菌 70 µm 細胞過濾器放入孔中,傾斜盤並透過過濾器輕輕吸出 4 mL 培養基。在燒瓶培養物中,將燒瓶傾斜一定角度,將 BOBA 放置在角落,然後以血清移液管更換大約 2/3 體積之已用培養基。 Suspended hydrogel BOBA cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed medium to 6-well plates (5 mL/well), 100 cm dishes (15 to 30 mL/dish), or 50 mL conical tubes (30 mL/tube) and keep in a 37°C warm bead bath. To generate suspended BME droplets or BOBA, dispense the organoid cell-BME solution in 10 µL volumes directly into the warm medium using an electronic continuous pipette (Integra VIAFLO 300) with a wide-bore or cut pipette tip (opening approximately 2 mm). During dispensing, the tip is dipped immediately below the surface of the liquid and then lifted after each dispense to ensure droplet separation. For larger format cultures, transfer BOBA to flasks by serological pipette or decanting. Change medium every 2 to 4 days. In 6-well dish cultures, place a sterile 70 µm cell filter in the well, tilt the dish and gently aspirate 4 mL of medium through the filter. In flask cultures, tilt the flask at an angle, place BOBA in a corner, and replace approximately 2/3 of the volume of used medium with a serological pipette.
經懸浮之水凝膠 SOBA 及 SOBA 碎片培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱培養基添加至 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15-30 mL/皿) 中,並保持在溫熱珠浴上。為了產生經懸浮之 SOBA 絲,將細胞-BME 溶液輕輕吸入帶有 15 號鈍尖端針之注射器中,然後直接擠出到溫熱培養基中,同時以 X-Y 平面中線形、蛇形或螺旋形運動方式移動浸沒針。為了產生 SOBA 碎片,使用 10 mL 血清移液管或寬孔 P1000 移液管尖端輕輕研磨 SOBA 絲培養物兩次。添加額外培養基以使最終 BME 與培養基之比率達到 1:10。如上對 BOBA 培養物所述進行培養基更換。 Suspended hydrogel SOBA and SOBA fragment cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed medium to 6-well plates (5 mL/well), 100 cm dishes (15-30 mL/dish) and keep on a warm bead bath. To generate suspended SOBA filaments, gently draw the cell-BME solution into a syringe with a 15-gauge blunt-tip needle and extrude directly into the warm medium while moving the submerged needle in a linear, serpentine, or spiral motion in the XY plane. To generate SOBA fragments, gently triturate the SOBA filament culture twice using a 10 mL serological pipette or a wide-bore P1000 pipette tip. Additional medium was added to give a final BME to medium ratio of 1:10. Medium changes were performed as described above for BOBA cultures.
免疫螢光樣本製備及共焦顯微術。 24 孔圓頂培養物以於 PBS 中之 2% 多聚甲醛 (PFA) 固定。使用抹刀將圓頂從盤上分離,並使用切割 P1000 移液管尖端轉移到微量離心管中。對於 BOBA 培養物,使用切割 P1000 移液管尖端將 500 µL 培養物轉移至微量離心管中,除去培養基並添加於 PBS 中之 2% PFA。將樣本在固定劑中室溫培育 15 至 30 分鐘,然後在 PBS 中洗滌 3 次。將樣本在微量離心管中以在封閉/透化緩衝液 (3% BSA、0.1% Triton X-100、0.02% 疊氮化鈉,於 PBS 中) 中稀釋之初級抗體在室溫染色至少 4 小時,然後在 PBS 中洗滌 3 次。然後將樣本與於室溫在封閉/透化緩衝液中稀釋之二級抗體、DAPI 以及 AlexaFluor 660 蠅虎蕈鹼在室溫培育至少 2 小時。使用 40X 物鏡在 Stellaris 8 共焦顯微鏡 (Leica) 上收集影像,並使用 Imaris 影像分析軟體 (Oxford Instruments) 進行 3D 重建。 Immunofluorescence Sample Preparation and Confocal Microscopy. 24-well dome cultures were fixed with 2% paraformaldehyde (PFA) in PBS. Domes were detached from the plate using a spatula and transferred to a microcentrifuge tube using a cut P1000 pipette tip. For BOBA cultures, 500 µL of culture was transferred to a microcentrifuge tube using a cut P1000 pipette tip, the medium was removed and 2% PFA in PBS was added. Samples were incubated in fixative at room temperature for 15 to 30 minutes and then washed three times in PBS. Samples were stained in microcentrifuge tubes with primary antibodies diluted in blocking/permeabilization buffer (3% BSA, 0.1% Triton X-100, 0.02% sodium azide in PBS) for at least 4 hours at room temperature and then washed three times in PBS. Samples were then incubated with secondary antibodies, DAPI, and AlexaFluor 660 agaricus diluted in blocking/permeabilization buffer for at least 2 hours at room temperature. Images were collected on a Stellaris 8 confocal microscope (Leica) using a 40X objective and 3D reconstruction was performed using Imaris image analysis software (Oxford Instruments).
轉錄組分析。 對於 RNA 分離,使用 RNeasy Micro Plus 套組 (Qiagen)。將 RLT+ 裂解緩衝液添加至 BME 圓頂或沉澱 BOBA 中並儲存在 -80℃。使用 QiaCube Connect (Qiagen) 進行 RNA 分離,並使用 Nanodrop 8000 (ThermoFisher) 定量 RNA。批量 mRNA-seq (NovaSeq PE150) 及分析藉由 Novogene 進行。使用 HISAT2 (Mortazavi 等人 2008) 比對讀段,使用 DESeq2 (Anders 等人2014) 進行差異基因表現分析,並使用負二項分佈模型及 Benjamini-Hochberg FDR 校正計算統計顯著性。分析幹細胞及增生標記物 ( CD133、 LGR5、 PDX1、 SOX9、 ALDH1A1、 NEUROG3及 NKX6.1) 以及分化標記物 ( 角蛋白 19 (KRT19)、 MUC1、 INS、 GCG及 AMY)。 Transcriptome analysis. For RNA isolation, the RNeasy Micro Plus kit (Qiagen) was used. RLT+ lysis buffer was added to BME domes or pelleted BOBA and stored at -80°C. RNA isolation was performed using QiaCube Connect (Qiagen), and RNA was quantified using Nanodrop 8000 (ThermoFisher). Bulk mRNA-seq (NovaSeq PE150) and analysis were performed by Novogene. Reads were aligned using HISAT2 (Mortazavi et al. 2008), differential gene expression analysis was performed using DESeq2 (Anders et al. 2014), and statistical significance was calculated using a negative binomial distribution model with Benjamini-Hochberg FDR correction. Stem cell and proliferation markers ( CD133 , LGR5 , PDX1 , SOX9 , ALDH1A1 , NEUROG3 , and NKX6.1 ) and differentiation markers ( keratin 19 (KRT19) , MUC1 , INS , GCG , and AMY ) were analyzed.
實例Examples 77 :肝類器官之產生:Generation of liver organoids
該實例提供了使用本文所述之 BME 懸浮方法產生肝類器官之方法。This example provides a method for generating liver organoids using the BME suspension method described herein.
方法:method:
人類肝類器官來源及維持。 肝類器官如 Huch 等人Cell 160(1-2):299-312 (2015) 之前所述產生,其內容全文併入本文中。 尺寸範圍為 0.5 至 1 cm 3之肝活體組織切片獲自肝移植過程中之供體及外植體肝。藉由切碎肝活體組織切片,以含 1% FCS 之 DMEM (GIBCO) 沖洗 2 次,並與消化溶液 (於 EBSS (Hyclone, Thermoscientific) 中之 2.5 mg/ml 膠原蛋白酶 D (Roche) 及 0.1 mg/ml DNase I (Sigma)) 一起在 37°C 培育 20 至 40 分鐘來從肝活體組織切片中分離肝細胞。藉由添加含有 1% FCS 之冷 DMEM 來停止消化。將消化懸浮液透過 70 µm 尼龍細胞過濾器過濾,並以 300 至 400 × g 旋轉 5 分鐘。將所得沉澱重懸於含有 1% FCS 之 DMEM 中並保持冷卻。如果材料保留在過濾器上,則在 37°C 在 Accutase (GIBCO) 中進一步消化此等材料 10 分鐘,然後停止並如上所述收集細胞。隨後將不同級分混合並以冷 Advanced DMEM/F12 洗滌,並以 300 至 400 × g 旋轉 5 分鐘。將細胞沉澱與水凝膠 ( 例如,MATRIGEL® (BD Biosciences) 或低生長因子 BME 2 (基底膜萃取物,2 型,Pathclear)) 混合,並將約 3,000 至 10,000 個細胞接種到盤 ( 例如,48 孔盤) 中之每個孔中。然後在 MATRIGEL® 或 BME 固化後添加培養基。培養基基於 AdDMEM/F12 (Invitrogen),其補充有 1% N2 (GIBCO) 及 1% B27 (GIBCO)、1.25 mM N-乙醯半胱氨酸 (Sigma)、10 nM 胃泌激素 (Sigma) 及生長因子:50 ng/ml EGF (Peprotech)、10% RSPO1 條件培養基 (自製)、100 ng/ml FGF10 (Peprotech)、25 ng/ml HGF (Peprotech)、10 mM 菸鹼醯胺 (Sigma)、5 µM A83.01 (Tocris) 及 10 µM FSK (Tocris)。在分離後前 3 天過程中,給培養基補充 25 ng/ml Noggin (Peprotech)、30% Wnt 培養基 (如 Barker 等人Cell Stem Cell 6:25-36 (2010) 所述) 及 10 µM (Y27632, Sigma Aldrich) 或 hES 細胞選殖恢復溶液 (Stemgent) 以建立培養物。隨後將培養基更換為不含 Noggin、Wnt、Y27632 及 hES 細胞選殖恢復溶液之培養基。10 至 14 天後,將類器官從 MATRIGEL® 或 BME 中取出,機械解離成小碎片並轉移到新鮮基質中。以分流比 ( 例如,1:4 至 1:8) 每 7 至 10 天一次進行傳代,持續至少 6 個月。為了製備冷凍原液,將類器官培養物解離並與恢復細胞培養冷凍培養基 (GIBCO) 混合,並依照標準程序冷凍。使用標準解凍程序解凍培養物並如上所述進行培養。在解凍後前 3 天內,給培養基補充 Y-27632 (10 μM)。 Human liver organoids source and maintenance. Liver organoids were generated as previously described by Huch et al. Cell 160(1-2):299-312 (2015), which is incorporated herein in its entirety. Liver biopsies ranging in size from 0.5 to 1 cm3 were obtained from donor and explant livers during liver transplantation. Hepatocytes were isolated from liver biopsies by mincing the liver biopsies, rinsing twice in DMEM (GIBCO) containing 1% FCS, and incubating with digestion solution (2.5 mg/ml collagenase D (Roche) and 0.1 mg/ml DNase I (Sigma) in EBSS (Hyclone, Thermoscientific)) at 37°C for 20 to 40 minutes. The digestion was stopped by adding cold DMEM containing 1% FCS. The digestion suspension was filtered through a 70 µm nylon cell filter and spun at 300 to 400 × g for 5 min. The resulting pellet was resuspended in DMEM containing 1% FCS and kept cold. If material remained on the filter, this was further digested in Accutase (GIBCO) at 37°C for 10 min before stopping and collecting the cells as described above. The different fractions were then pooled and washed with cold Advanced DMEM/F12 and spun at 300 to 400 × g for 5 min. The cell pellet is mixed with a hydrogel ( e.g. , MATRIGEL® (BD Biosciences) or reduced growth factor BME 2 (basement membrane extract, type 2, Pathclear)) and approximately 3,000 to 10,000 cells are plated per well in a plate ( e.g. , a 48-well plate). The medium is then added after the MATRIGEL® or BME solidifies. The culture medium was based on AdDMEM/F12 (Invitrogen) supplemented with 1% N2 (GIBCO) and 1% B27 (GIBCO), 1.25 mM N-acetylcysteine (Sigma), 10 nM gastrin (Sigma), and growth factors: 50 ng/ml EGF (Peprotech), 10% RSPO1 conditioned medium (homemade), 100 ng/ml FGF10 (Peprotech), 25 ng/ml HGF (Peprotech), 10 mM niacinamide (Sigma), 5 µM A83.01 (Tocris), and 10 µM FSK (Tocris). During the first 3 days after isolation, cultures were established by supplementing the medium with 25 ng/ml Noggin (Peprotech), 30% Wnt medium (as described by Barker et al. Cell Stem Cell 6:25-36 (2010)), and 10 µM (Y27632, Sigma Aldrich) or hES cell selective recovery solution (Stemgent). The medium was then changed to medium without Noggin, Wnt, Y27632, and hES cell selective recovery solution. After 10 to 14 days, organoids were removed from MATRIGEL® or BME, mechanically dissociated into small fragments, and transferred to fresh medium. Passage every 7 to 10 days for at least 6 months at a split ratio ( e.g. , 1:4 to 1:8). To prepare frozen stocks, lyse organoid cultures and mix with Recovery Cell Culture Freezing Medium (GIBCO) and freeze according to standard procedures. Thaw cultures using standard thawing procedures and culture as described above. Supplement media with Y-27632 (10 μM) within the first 3 days after thawing.
對於肝細胞分化,,將肝類器官在補充有 BMP7 (25 ng/ml) 之上述肝培養基中接種並培養 7 至 10 天。將培養物分開並接種在此補充有 BMP7 之培養基中至少約 2 至 4 天。隨後,將培養基更換為分化培養基,其包括補充有 1% N2 及 1% B27 並含有 EGF (50 ng/ml)、胃泌激素 (10 nM, Sigma)、HGF (25 ng/ml)、FGF19 (100 ng/ml)、A8301 (500 nM)、DAPT (10 µM)、BMP7 (25 ng/ml) 及地塞米松 (30 µM) 之 AdDMEM/F12 培養基。在 11 至 13 天之時段內,每 2 至 3 天一次更換分化培養基。For hepatocyte differentiation, hepatic organoids were plated and cultured in the above hepatic medium supplemented with BMP7 (25 ng/ml) for 7 to 10 days. The cultures were split and plated in this BMP7-supplemented medium for at least about 2 to 4 days. Subsequently, the medium was changed to differentiation medium, which included AdDMEM/F12 medium supplemented with 1% N2 and 1% B27 and containing EGF (50 ng/ml), gastrin (10 nM, Sigma), HGF (25 ng/ml), FGF19 (100 ng/ml), A8301 (500 nM), DAPT (10 µM), BMP7 (25 ng/ml), and dexamethasone (30 µM). Change the differentiation medium every 2 to 3 days over a period of 11 to 13 days.
經懸浮之水凝膠 BOBA 培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱培養基添加到 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15 至 30 mL/皿) 或 50 mL 錐形管 (30 mL/管) 中,並保持在 37°C 溫熱珠浴上。為了產生經懸浮之 BME 小滴或 BOBA,使用帶有寬孔或切割移液管尖端 (開口約 2 mm) 之電子連續移液管 (Integra VIAFLO 300) 將類器官細胞-BME 溶液以 10 µL 體積直接分配到溫熱培養基中。在分配過程中,尖端立即浸入液體表面下方,然後在各分配後提起,以確保小滴分離。對於較大形式培養物,藉由血清移液管或傾析將 BOBA 轉移至燒瓶中。每 2 至 3 天一次以培養基更換培養基。在 6 孔盤培養物中,將無菌 70 µm 細胞過濾器放入孔中,傾斜盤並透過過濾器輕輕吸出 4 mL 培養基。在燒瓶培養物中,將燒瓶傾斜一定角度,將 BOBA 放置在角落,然後以血清移液管更換大約 2/3 體積之已用培養基。 Suspended hydrogel BOBA cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed medium to 6-well plates (5 mL/well), 100 cm dishes (15 to 30 mL/dish), or 50 mL conical tubes (30 mL/tube) and keep in a 37°C warm bead bath. To generate suspended BME droplets or BOBA, dispense the organoid cell-BME solution in 10 µL volumes directly into the warm medium using an electronic continuous pipette (Integra VIAFLO 300) with a wide-bore or cut pipette tip (opening approximately 2 mm). During dispensing, the tip is dipped immediately below the surface of the liquid and then lifted after each dispense to ensure droplet separation. For larger format cultures, transfer BOBA to flasks by serological pipette or decanting. Replace medium with medium every 2 to 3 days. In 6-well dish cultures, place a sterile 70 µm cell filter in the well, tilt the dish and gently aspirate 4 mL of medium through the filter. In flask cultures, tilt the flask at an angle, place BOBA in a corner, and replace approximately 2/3 of the volume of used medium with a serological pipette.
經懸浮之水凝膠 SOBA 及 SOBA 碎片培養物。 如上所述,在冰上製備 BME 中之單類器官細胞或類器官碎片。將預熱培養基添加至 6 孔盤 (5 mL/孔)、100 cm 培養皿 (15-30 mL/皿) 中,並保持在溫熱珠浴上。為了產生經懸浮之 SOBA 絲,將細胞-BME 溶液輕輕吸入帶有 15 號鈍尖端針之注射器中,然後直接擠出到溫熱培養基中,同時以 X-Y 平面中線形、蛇形或螺旋形運動方式移動浸沒針。為了產生 SOBA 碎片,使用 10 mL 血清移液管或寬孔 P1000 移液管尖端輕輕研磨 SOBA 絲培養物兩次。添加額外培養基以使最終 BME 與培養基之比率達到 1:10。如上對 BOBA 培養物所述進行培養基更換。 Suspended hydrogel SOBA and SOBA fragment cultures. Prepare single organoid cells or organoid fragments in BME on ice as described above. Add pre-warmed medium to 6-well plates (5 mL/well), 100 cm dishes (15-30 mL/dish) and keep on a warm bead bath. To generate suspended SOBA filaments, gently draw the cell-BME solution into a syringe with a 15-gauge blunt-tip needle and extrude directly into the warm medium while moving the submerged needle in a linear, serpentine, or spiral motion in the XY plane. To generate SOBA fragments, gently triturate the SOBA filament culture twice using a 10 mL serological pipette or a wide-bore P1000 pipette tip. Additional medium was added to give a final BME to medium ratio of 1:10. Medium changes were performed as described above for BOBA cultures.
免疫螢光樣本製備及共焦顯微術。 24 孔圓頂培養物以於 PBS 中之 2% 多聚甲醛 (PFA) 固定。使用抹刀將圓頂從盤上分離,並使用切割 P1000 移液管尖端轉移到微量離心管中。對於 BOBA 培養物,使用切割 P1000 移液管尖端將 500 µL 培養物轉移至微量離心管中,除去培養基並添加於 PBS 中之 2% PFA。將樣本在固定劑中室溫培育 15 至 30 分鐘,然後在 PBS 中洗滌 3 次。將樣本在微量離心管中以在封閉/透化緩衝液 (3% BSA、0.1% Triton X-100、0.02% 疊氮化鈉,於 PBS 中) 中稀釋之初級抗體在室溫染色至少 4 小時,然後在 PBS 中洗滌 3 次。然後將樣本與於室溫在封閉/透化緩衝液中稀釋之二級抗體、DAPI 以及 AlexaFluor 660 蠅虎蕈鹼在室溫培育至少 2 小時。使用 40X 物鏡在 Stellaris 8 共焦顯微鏡 (Leica) 上收集影像,並使用 Imaris 影像分析軟體 (Oxford Instruments) 進行 3D 重建。 Immunofluorescence Sample Preparation and Confocal Microscopy. 24-well dome cultures were fixed with 2% paraformaldehyde (PFA) in PBS. Domes were detached from the plate using a spatula and transferred to a microcentrifuge tube using a cut P1000 pipette tip. For BOBA cultures, 500 µL of culture was transferred to a microcentrifuge tube using a cut P1000 pipette tip, the medium was removed and 2% PFA in PBS was added. Samples were incubated in fixative at room temperature for 15 to 30 minutes and then washed three times in PBS. Samples were stained in microcentrifuge tubes with primary antibodies diluted in blocking/permeabilization buffer (3% BSA, 0.1% Triton X-100, 0.02% sodium azide in PBS) for at least 4 hours at room temperature and then washed three times in PBS. Samples were then incubated with secondary antibodies, DAPI, and AlexaFluor 660 agaricus diluted in blocking/permeabilization buffer for at least 2 hours at room temperature. Images were collected on a Stellaris 8 confocal microscope (Leica) using a 40X objective and 3D reconstruction was performed using Imaris image analysis software (Oxford Instruments).
轉錄組分析。 對於 RNA 分離,使用 RNeasy Micro Plus 套組 (Qiagen)。將 RLT+ 裂解緩衝液添加至 BME 圓頂或沉澱 BOBA 中並儲存在 -80℃。使用 QiaCube Connect (Qiagen) 進行 RNA 分離,並使用 Nanodrop 8000 (ThermoFisher) 定量 RNA。批量 mRNA-seq (NovaSeq PE150) 及分析藉由 Novogene 進行。使用 HISAT2 (Mortazavi 等人 2008) 比對讀段,使用 DESeq2 (Anders 等人2014) 進行差異基因表現分析,並使用負二項分佈模型及 Benjamini-Hochberg FDR 校正計算統計顯著性。分析幹細胞及增生標記物 ( LGR5) 以及分化標記物 (針對肝細胞之 ALB、 CYP3A4及 HNF4A,以及針對導管細胞之 KRT19、 KRT7及 SOX9)。 Transcriptome analysis. For RNA isolation, the RNeasy Micro Plus kit (Qiagen) was used. RLT+ lysis buffer was added to BME domes or pelleted BOBA and stored at -80°C. RNA isolation was performed using QiaCube Connect (Qiagen), and RNA was quantified using Nanodrop 8000 (ThermoFisher). Bulk mRNA-seq (NovaSeq PE150) and analysis were performed by Novogene. Reads were aligned using HISAT2 (Mortazavi et al. 2008), differential gene expression analysis was performed using DESeq2 (Anders et al. 2014), and statistical significance was calculated using a negative binomial distribution model with Benjamini-Hochberg FDR correction. Stem cell and proliferation markers ( LGR5 ) and differentiation markers ( ALB , CYP3A4 , and HNF4A for hepatocytes, and KRT19 , KRT7 , and SOX9 for ductal cells) were analyzed.
實例Examples 88 :注射器擠出之類器官:Organoids squeezed out of a syringe BMEBME 組裝體Assembly (SOBA)(SOBA) 培養方法Cultivation method
該實例提供了使用 SOBA 絲及 SOBA 絲碎片產生腸類器官並維持此等腸類器官之方法。SOBA 方法有利於並加快培養物製備,因為可將大體積細胞-BME 溶液加載到單一注射器中以產生 SOBA 水凝膠絲。整體而言,SOBA 方法使得能夠進行大規模類器官規模化,改善類器官培養均勻性,並節省勞力、時間及資源。This example provides methods for generating intestinal organoids using SOBA filaments and SOBA filament fragments and maintaining such intestinal organoids. The SOBA method facilitates and speeds up culture preparation because a large volume of cell-BME solution can be loaded into a single syringe to generate SOBA hydrogel filaments. Overall, the SOBA method enables large-scale organoid scalability, improves organoid culture uniformity, and saves labor, time, and resources.
材料:Material:
該方法使用了以下材料:10 cm 或 15 cm 培養皿、5 mL 注射器、15 號鈍尖端針 (藉由浸入 70% EtOH 中滅菌)、燒瓶 (T25-T225 或 6 孔盤/培養皿皆相容)、50 mL 錐形管、離心機、含 5% CO 2之培養箱、TC 倒置光學顯微鏡及 37℃ 水浴。 The method uses the following materials: 10 cm or 15 cm culture dishes, 5 mL syringes, 15-gauge blunt-tip needles (sterilized by immersion in 70% EtOH), flasks (T25-T225 or 6-well plates/culture dishes are compatible), 50 mL conical tubes, centrifuge, incubator with 5% CO 2 , TC inverted light microscope, and 37°C water bath.
藥劑:Drugs:
CULTREX® 低生長因子基底膜萃取物 2 型 (BME,R&D Systems 3533-010-2;必須保存在冰上) 或 MATRIGEL®;TrypLE Express;PBS (不含 Ca 2+/Mg 2+);類器官生長培養基:人類腸類器官生長培養基 (OGM,Stem Cell Tech 06010) 及 Primocin®(1:1000, InvivoGen ant-pm-1);以及類器官傳代培養基:類器官生長培養基及 10 µM Y-27632 (10 µM;1:1000 的於 PBS 中之 10 mM 原液)。 CULTREX® Reduced Growth Factor Basement Membrane Extract Type 2 (BME, R&D Systems 3533-010-2; must be kept on ice) or MATRIGEL®; TrypLE Express; PBS (without Ca 2+ /Mg 2+ ); Organoid Growth Medium: Human Intestinal Organoid Growth Medium (OGM, Stem Cell Tech 06010) and Primocin® (1:1000, InvivoGen ant-pm-1); and Organoid Passaging Medium: Organoid Growth Medium and 10 µM Y-27632 (10 µM; 1:1000 of a 10 mM stock solution in PBS).
低滯留移液管尖端可用於所有步驟,以防止類器官損失。如果沒有低滯留尖端且不可接受類器官損失,則可在使用前給管及/或尖端包被 1% BSA/PBS。Low retention pipette tips can be used in all steps to prevent organoid loss. If low retention tips are not available and organoid loss is unacceptable, tubes and/or tips can be coated with 1% BSA/PBS prior to use.
方法:method:
細胞製備 。如本文所述產生胃腸類器官,並在類器官生長培養基中培養 7 至 14 天,該培養基包括人 Intesticult 類器官生長培養基 (OGM,Stem Cell Tech 06010) 及 Primocin® (1:1000,InvivoGen ant-pm-1)。對活細胞進行計數 (通常觀察到約 50% 至 70% 生存力),並且視情況地,藉由重懸於 10mL DMEM 或 PBS 中並以 500 x g 離心 3 分鐘來洗滌細胞。這改善了生存力百分比,但有一些總體細胞損失。為了產生細胞-BME 溶液,將類器官細胞沉澱以 6 x 10 5個細胞/mL BME 之密度重懸於冰上之 BME 中 (這相當於 24 孔盤每孔 3 x 10 4個細胞,接種密度可能需要針對各行進行調整)。以 P1000 移液管將細胞沉澱輕輕重懸於 1 mL BME 中,而不引入氣泡。然後以 1 mL 增量添加額外 BME,每次均輕輕研磨。 Cell preparation . Gastrointestinal organoids were generated as described herein and cultured for 7 to 14 days in organoid growth medium, which included human Intesticult Organoid Growth Medium (OGM, Stem Cell Tech 06010) and Primocin® (1:1000, InvivoGen ant-pm-1). Viable cells were counted (about 50% to 70% viability was usually observed) and cells were washed by resuspending in 10 mL DMEM or PBS and centrifuging at 500 x g for 3 minutes, as appropriate. This improved the viability percentage, but there was some overall cell loss. To create the cell-BME solution, resuspend the organoid cell pellet in BME on ice at a density of 6 x 10 5 cells/mL BME (this is equivalent to 3 x 10 4 cells per well of a 24-well plate; seeding density may need to be adjusted for individual rows). Gently resuspend the cell pellet in 1 mL of BME using a P1000 pipette without introducing air bubbles. Then add additional BME in 1 mL increments, triturating gently each time.
經懸浮之水凝膠 SOBA 及 SOBA 碎片培養物。 將預熱類器官傳代培養基 (在 37℃ 水或珠浴中預熱) 添加到培養皿 (10 cm 培養皿中 30 mL 培養基或 15 cm 培養皿中 60 mL 培養基) 中並保持在溫熱珠浴上以維持溫度,該培養基包括類器官生長培養基及 10 µM Y-27632 (10 µM;1:1000 的於 PBS 中之 10 mM 原液)。如果迅速進行,則 SOBA 產生可在室溫在實驗台上進行,而不需要溫熱珠浴。 Suspended hydrogel SOBA and SOBA fragment cultures. Pre-warmed organoid subculture medium (pre-warmed in 37°C water or bead bath) consisting of organoid growth medium and 10 µM Y-27632 (10 µM; 1:1000 of a 10 mM stock in PBS) is added to the culture dishes (30 mL medium in a 10 cm dish or 60 mL medium in a 15 cm dish) and kept on a warm bead bath to maintain temperature. SOBA production can be performed at room temperature on the benchtop without the need for a warm bead bath if performed rapidly.
為了產生經懸浮之 SOBA 絲,將冷細胞-BME 溶液加載到帶有 15 號 (或更寬) 鈍尖端針或大體積移液管尖端之注射器中。將細胞-BME 溶液直接擠出到溫熱培養基中,同時在 X-Y 平面中以線形、蛇形或螺旋形運動移動浸沒針或尖端以產生 SOBA 絲。類器官在直徑 1-5 mm 絲中成功生長,其中藉由較慢分配速度及較慢 X-Y 運動產生更寬絲。更寬且更長絲可改善培養基更換過程中之 SOBA 重力沉降。如果培養基不夠溫熱或 BME 溶液太冷,則 SOBA 絲可能無法很好地形成。如果培養基不夠溫熱,則可能無法將 BME 溫熱到足以在分配過程中足夠快地聚合。如果 BME-細胞溶液太冷,則溫熱培養基可能無法立即將 BME 溫熱到足夠溫度以進行正確固化。在產生 SOBA 之前,可將 BME-細胞溶液在手中短暫溫熱,或可從冰中取出至室溫,持續 30 秒到一分鐘 (取決於 BME 體積)。To generate suspended SOBA filaments, load cold cell-BME solution into a syringe with a 15-gauge (or wider) blunt-tip needle or large-volume pipette tip. Extrude the cell-BME solution directly into warm medium while moving the submerged needle or tip in a linear, serpentine, or spiral motion in the X-Y plane to generate SOBA filaments. Organoids have been successfully grown in filaments 1-5 mm in diameter, with wider filaments generated by slower dispensing speeds and slower X-Y motions. Wider and longer filaments improve gravity settling of SOBA during medium changes. SOBA filaments may not form well if the medium is not warm enough or the BME solution is too cold. If the medium is not warm enough, it may not be able to warm the BME enough to polymerize quickly enough during the dispensing process. If the BME-cell solution is too cold, the warm medium may not immediately warm the BME to a sufficient temperature for proper solidification. The BME-cell solution may be briefly warmed in the hands or may be removed from ice to room temperature for 30 seconds to a minute (depending on the volume of BME) prior to generating the SOBA.
為了將 SOBA 絲及培養基轉移到燒瓶或其他培養容器中,使用寬開口血清移液管緩慢吸出並分配 SOBA 絲,以防止 SOBA 絲碎片化。然後將類器官傳代培養基添加到培養容器中,以使最終 BME 與培養基之比率達到 1:10。To transfer the SOBA filament and medium to a flask or other culture vessel, use a wide-opening serological pipette to slowly aspirate and dispense the SOBA filament to prevent fragmentation of the SOBA filament. Then add the Organoid Subculture Medium to the culture vessel to achieve a final BME to medium ratio of 1:10.
培養基變化 。為了使 SOBA 絲藉由重力沉降,(a) 將培養容器 ( 例如,燒瓶) 垂直支撐並對角傾斜 5 分鐘 (例如,燒瓶可支撐在管架或冰桶中) ,或 (b) 將培養物輕輕傾析入 50 mL 錐形管中,其中藉由血清移液管轉移 SOBA 絲之次數應受到限制,以盡量減少碎片化。如果 SOBA 碎片非常小,則它們可保持漂浮在培養基中,在這種情況下,將培養物轉移到錐形管中以有利於 SOBA 沉降。將錐形管以 500 x g 離心 3 分鐘,關閉制動以促進沉降,並以 PBS 稀釋培養物並再次離心。在某些情況下,可以 PBS 稀釋培養物並再次離心。使用血清移液管,每 2 至 3 天用新鮮類器官傳代培養基更換約 50% 至 75% 培養基。如果將培養物轉移到錐形管中,則將培養物輕輕傾析回原始培養容器中。 Medium changes . To allow SOBA filaments to settle by gravity, (a) support the culture vessel ( e.g. , flask) vertically and tilt diagonally for 5 minutes (e.g., flask can be supported in a tube rack or ice bucket), or (b) gently decant the culture into a 50 mL conical tube, where transfers of SOBA filaments by serological pipette should be limited to minimize fragmentation. If SOBA fragments are very small, they may remain floating in the medium, in which case, transfer the culture to a conical tube to facilitate settling of the SOBA. Centrifuge the tube at 500 x g for 3 minutes, turn off the brake to promote sedimentation, and dilute the culture with PBS and centrifuge again. In some cases, the culture may be diluted with PBS and centrifuged again. Using a serological pipette, replace approximately 50% to 75% of the medium with fresh organoid subculture medium every 2 to 3 days. If the culture was transferred to a conical tube, gently decant the culture back into the original culture vessel.
使 SOBA 培養物傳代。 類器官通常在接種單類器官細胞後 7 至 14 天即可傳代。作物初始步驟,藉由重力沉降 SOBA 碎片。如上所討論的,為了使 SOBA 絲藉由重力沉降,(a) 將培養容器 ( 例如,燒瓶) 垂直支撐並對角傾斜 5 分鐘,或 (b) 將培養物輕輕傾析入 50 mL 錐形管中。藉由血清移液管除去盡可能多的培養基。如果 SOBA 碎片仍漂浮在培養基中,則將培養物稀釋,轉移至錐形管並以 PBS 稀釋。將錐形管以 500 x g 離心 3 分鐘,關閉制動以促進沉降。 Passaging SOBA cultures. Organoids are typically ready for passage 7 to 14 days after inoculation of a single organoid cell. Initially, allow SOBA fragments to settle by gravity. As discussed above, to allow SOBA filaments to settle by gravity, (a) support the culture vessel ( e.g. , flask) vertically and tilt it diagonally for 5 minutes, or (b) gently decant the culture into a 50 mL conical tube. Remove as much medium as possible using a serological pipette. If SOBA fragments are still floating in the medium, dilute the culture, transfer to a conical tube, and dilute with PBS. Centrifuge the tube at 500 x g for 3 minutes with the brake off to promote sedimentation.
將預熱 TrypLE Express 以至少 10 mL TrypLE Express 與 SOBA 培養物中 1 mL BME 之比率添加到錐形管中,並以血清移液管研磨溶液以打碎 SOBA 絲。然後將溶液在 37°C 水浴中培育 30 分鐘至 1 小時,視情況地在培育期間藉由血清移液管劇烈研磨。培育後,以血清移液管劇烈研磨溶液,並目視觀察類器官解離 ( 例如,使用倒置顯微鏡)。然後藉由向錐形管中添加 PBS 來滅活 TrypLE Express。 Pre-warmed TrypLE Express is added to a conical tube at a ratio of at least 10 mL TrypLE Express to 1 mL BME in SOBA culture medium, and the solution is triturated with a serological pipette to break up SOBA threads. The solution is then incubated in a 37°C water bath for 30 minutes to 1 hour, with vigorous trituration with a serological pipette during the incubation period as appropriate. After incubation, the solution is triturated vigorously with a serological pipette and visually inspected for organoid dissociation ( e.g. , using an inverted microscope). TrypLE Express is then inactivated by adding PBS to the conical tube.
採用這種技術,活細胞可根據需要進行計數、稀釋或等分。在某些實施例中,細胞可根據需要傳代、冷凍或用於實驗。 * * * * * * * * Using this technique, live cells can be counted, diluted, or aliquoted as desired. In certain embodiments, cells can be passaged, frozen, or used in experiments as desired. * * * * * * * * *
儘管已詳細描述目前揭露之主題及其優點,但應理解,在不脫離本揭露的精神和範圍的情況下,可在本文中進行各種改變、替換和變更。此外,本案的範圍不意圖限於說明書中所述的製程、機器、製造和物質組成、手段、方法和步驟的特定實施例。本技術領域中具有通常知識者將容易地從本文所揭示之主題的發明中輕易理解,可根據本文所揭示的主題,利用目前存在或今後將開發的進行與在本文描述的對應實施例基本上相同的功能或實現基本上相同的結果的製程、機器、製造、物質組成、手段、方法或步驟。因此,所附的申請專利範圍旨在將該等製程、機器、製造、物質組成、手段、方法或步驟包括在該申請專利範圍的範圍內。Although the subject matter and advantages of the present disclosure have been described in detail, it should be understood that various changes, substitutions and modifications may be made herein without departing from the spirit and scope of the present disclosure. In addition, the scope of the present case is not intended to be limited to the specific embodiments of the processes, machines, manufactures and material compositions, means, methods and steps described in the specification. A person with ordinary knowledge in the art will easily understand from the invention of the subject matter disclosed herein that a process, machine, manufacture, material composition, means, method or step that currently exists or will be developed in the future can be used according to the subject matter disclosed herein to perform substantially the same function or achieve substantially the same result as the corresponding embodiment described herein. Therefore, the appended claims are intended to include within the scope of such claims such processes, machines, manufacture, compositions of matter, means, methods or steps.
本案通篇引用了各種專利、專利申請案、出版物、產品說明、方案和序列登錄號,出於所有目的,其內容藉由引用整體而併入本文。Various patents, patent applications, publications, product descriptions, protocols, and serial accession numbers are cited throughout this application, the contents of which are incorporated herein by reference in their entirety for all purposes.
圖 1A-1H 顯示經懸浮之 BME 水凝膠中腸類器官之規模化。習用圓頂培養物 (上) 及經懸浮之 BOBA (嵌入 BME 之類器官珠組裝體) 培養物 (下) 中人類大腸類器官之 (A) 示意圖、(B) 照片及 (C) 明視野顯微術。顯微照片之比例尺為 500 µm。(D) 明視野影像中圓頂及 BOBA 培養物之類器官直徑。(E) 共焦影像中 Ki67 陽性細胞之百分比。所呈現之資料為平均值 ± SD,學生 t 檢驗,n = 3,各 10 個視野。(F) 圓頂或 BOBA 培養物中大腸類器官之 3D 重建共焦影像。Ki67 為綠色,細胞核為藍色,肌動蛋白為白色,且比例尺為 20 µm。(G, H) (G) 大腸及 (H) 迴腸類器官之總活細胞、每 cm 2表面積之細胞或每 µL BME 之細胞。所有所呈現之資料為平均值 ± SD,學生 t 檢驗,*p ≤ 0.05,****p ≤ 0.0001,n = 3 次實驗。 圖 2A-2C 顯示 BOBA 培養物中之類器官分化。(A) 圓頂 (上) 及 BOBA (下) 培養物中大腸類器官之明視野影像顯示增生及分化類器官之相當形態。比例尺為 100 µm。(B) 批量 RNA-seq 顯示,從生長培養基過渡到分化培養基後,圓頂及 BOBA 培養之大腸類器官類似地下調幹細胞及前驅細胞標記物並上調分化標記物。(C) 針對分化上皮細胞類型之標記物 (針對杯狀細胞之 MUC2、針對腸上皮細胞之 FABP1 及針對腸內分泌細胞之 CHGA) 在以圓頂 (上) 及 BOBA (下) 二者形式培養之類器官中表現。細胞核為藍色,肌動蛋白為白色,且比例尺為 10 µm。 圖 3A-3C 顯示 BME 體積及培養容器可影響經懸浮之 BME 水凝膠培養物中之類器官生長。(A) 24 孔盤中表面接附之圓頂 (0.5 mL 培養基中之 50 µL BME) 中大腸類器官之明視野影像,或 6 孔盤孔中之 BOBA 培養物 (5 mL 培養基中之 0.5、1 或 2 mL BME) 中之大腸類器官之明視野影像。比例尺為 200 µm。(B,C) (B) 6 孔盤或 (C) 25 cm 2燒瓶中類器官直徑、總活細胞、每 cm 2表面積之活細胞及每 µL BME 之活細胞之定量。所呈現之資料為平均值 ± SD,單因素 ANOVA 多重比較檢驗,n = 3 次實驗;*p ≤ 0.05,**p ≤ 0.01,***p ≤ 0.001,****p ≤ 0.0001。 圖 4A-4E 顯示 BOBA 培養物中類器官尺寸均勻性。(A) 描述用於均勻性分析之成像策略之示意圖。(B,C) 50 µl BME 圓頂或 10 µl 經懸浮之 BOBA 水凝膠中最深平面處之大腸類器官之明視野影像。比例尺為 (B) 200 µm 及 (C) 1 mm。(D) 依跨 X 軸之位置,圓頂或 BOBA 水凝膠之水平 ROI 中之類器官直徑之定量。所呈現之資料為平均值 ± SD,並且在 3 次實驗之代表性實驗中 n=3 次重複。(E) 圓頂及 BOBA 培養物邊緣或核心處平均類器官直徑之定量。所有所呈現之資料為平均值 ± SD,雙因素 ANOVA,並且在 3 次實驗之代表性實驗中 n=3 次重複。 圖 5A-5B 顯示圓頂及 BOBA 培養物中類器官之基因表現。(A,B) 批量 RNA-seq 分析顯示,在生長培養基中培養 7 天後,在圓頂培養物或經懸浮之 BOBA 培養物中之大腸類器官之間 (A) 幹細胞及增生標記物與 (B) 腸上皮細胞標記物之基因表現差異。所呈現之資料為藉由 DESeq2 確定之標準化計數之平均值 ± SD,n = 3 次重複,且藉由負二項分佈模型進行統計分析,其中 BH 調整 p 值,*padj ≤ 0.05,** padj ≤ 0.01。 圖 6A-6D 顯示替代的經懸浮之 BME 水凝膠培養物形式具有相當的類器官生長。BOBA、SOBA (注射器擠出之類器官 BME 組裝體) 或 SOBA 碎片形式之 6 孔盤培養物中大腸類器官之 (A) 照片及 (B) 明視野影像。替代形式使得能夠加快培養物製備以用於規模化。比例尺為 1 mm。(C) 類器官直徑之定量及 (D) 每孔之活細胞。所呈現之資料為平均值 ± SD,單因素 ANOVA 多重比較檢驗,n = 3 次實驗。 圖 7A-7E 顯示經懸浮之 BME 水凝膠類器官培養物在中等生產量篩選中之應用。(A) 實驗示意圖。將 225 cm 2燒瓶中之 SOBA 碎片類器官培養物研磨以達到均勻類器官懸浮液,然後接種到 96 孔盤中。(B) 在 225 cm 2燒瓶中培養之 SOBA 碎片培養物之明視野影像。比例尺為 1 mm。(C) 跨 96 孔盤隨機選擇之孔之明視野影像顯示相當的孔間類器官密度。比例尺為 1 mm。(D) Cell Titer Glo 3D (CTG) 生存力讀數顯示 96 孔盤中圓頂與經懸浮之 BME 培養物之間相似的孔間變異性。所呈現之資料為平均值 ± SD。學生 t 檢驗。(E) 以雙醋瑞因、索拉非尼、SN-38 或多西紫杉醇處理 3 天之經懸浮之 BME 類器官之代表性劑量反應生存力曲線 (CTG 測定)。所呈現之資料為平均值 ± SD,n = 4。 圖 8A-8C 顯示使用經懸浮之 BME 水凝膠類器官來產生 Transwell 單層。(A) 實驗示意圖。將 225 cm 2燒瓶中之 SOBA 碎片類器官培養物消化成單細胞懸浮液,然後在匯合時接種到 96 孔 BME 包被之 Transwell 小室中。(B) 接種後 3 天之 SOBA 碎片類器官來源之 Transwell 單層之明視野影像。Transwell 係以單層生長培養基或單層分化培養基建立。比例尺為 100 µm。(C) 在單層生長培養基 (實心圓圈) 或單層分化培養基 (空心圓圈) 中培養之 Transwell 單層之跨上皮電阻 (TEER)。所呈現之資料為平均值 ± SD,n = 6 孔。 圖 9提供嵌入懸浮於 BOBA 培養物之培養基中之水凝膠中之肺 AT2 類器官之影像。 圖 10提供描繪用於產生水凝膠小滴之示例性條件之示意圖,該水凝膠小滴在本文中被稱為基底膜類器官珠組裝體 (BOBA)。懸浮於冷液體水凝膠中之類器官或經解離之類器官細胞作為小滴分配到室溫或更溫暖之培養基中,這使得水凝膠在分配到培養基中時能夠立即固化。 圖 11提供描繪用於產生水凝膠絲之示例性條件之示意圖,水凝膠絲在本文中被稱為注射器擠出之類器官基底膜組裝體 (SOBA)。懸浮於冷液體水凝膠中之類器官或經解離之類器官細胞作為絲擠出 (藉由注射器、移液管或任何其他分配裝置) 到室溫或更溫暖之培養基中,這使得水凝膠在分配到培養基中時能夠立即固化。 Figures 1A-1H show the scaling of intestinal organoids in suspension in BME hydrogel. (A) Schematic, (B) photographs, and (C) bright field microscopy of human intestinal organoids in conventional dome cultures (top) and suspended BOBA (organoid bead assemblies embedded in BME) cultures (bottom). Scale bar for micrographs is 500 µm. (D) Diameter of organoids in dome and BOBA cultures in bright field images. (E) Percentage of Ki67-positive cells in confocal images. Data presented are mean ± SD, Student's t test, n = 3, 10 fields each. (F) 3D reconstructed confocal images of colon organoids in dome or BOBA culture. Ki67 is green, nuclei are blue, actin is white, and the scale bar is 20 µm. (G, H) Total viable cells, cells per cm2 surface area, or cells per µL BME for (G) colon and (H) ileum organoids. All data presented are mean ± SD, Student’s t test, *p ≤ 0.05, ****p ≤ 0.0001, n = 3 experiments. Figures 2A-2C show organoid differentiation in BOBA culture. (A) Bright field images of colon organoids in dome (top) and BOBA (bottom) cultures show comparable morphology of proliferating and differentiating organoids. Scale bar, 100 µm. (B) Bulk RNA-seq shows that colon organoids cultured in dome and BOBA similarly downregulate stem cell and progenitor cell markers and upregulate differentiation markers following transition from growth medium to differentiation medium. (C) Markers specific for differentiated epithelial cell types (MUC2 for goblet cells, FABP1 for intestinal epithelial cells, and CHGA for intestinal endocrine cells) are expressed in organoids cultured in both dome (top) and BOBA (bottom) formats. Nuclei are blue, actin is white, and the scale bar is 10 µm. Figures 3A-3C show that BME volume and culture vessel can affect organoid growth in suspended BME hydrogel culture. (A) Bright field images of intestinal organoids in surface-attached domes (50 µL BME in 0.5 mL medium) in a 24-well plate or in BOBA culture (0.5, 1, or 2 mL BME in 5 mL medium) in a 6-well plate. Scale bar is 200 µm. (B, C) Quantification of organoid diameter, total viable cells, viable cells per cm2 surface area, and viable cells per µL BME in (B) 6-well plates or (C) 25 cm2 flasks. Data presented are mean ± SD, one-way ANOVA multiple comparison test, n = 3 experiments; *p ≤ 0.05, **p ≤ 0.01, ***p ≤ 0.001, ****p ≤ 0.0001. Figures 4A-4E show organoid size homogeneity in BOBA culture. (A) Schematic depicting the imaging strategy used for homogeneity analysis. (B, C) Bright field images of colon organoids in the deepest plane of a 50 µl BME dome or 10 µl suspended BOBA hydrogel. Scale bars are (B) 200 µm and (C) 1 mm. (D) Quantification of organoid diameter in the horizontal ROI of the dome or BOBA hydrogel according to position across the X-axis. Data presented are mean ± SD, and n = 3 replicates in a representative experiment of 3 experiments. (E) Quantification of mean organoid diameter at the edge or core of the dome and BOBA culture. All data presented are mean ± SD, two-way ANOVA, and n = 3 replicates in a representative experiment of 3 experiments. Figures 5A-5B show gene expression in organoids in dome and BOBA culture. (A, B) Bulk RNA-seq analysis shows differences in gene expression of (A) stem cell and proliferation markers and (B) intestinal epithelial cell markers between colon organoids in dome culture or suspended BOBA culture after 7 days of culture in growth medium. Data presented are means ± SD of normalized counts determined by DESeq2, n = 3 replicates, and statistical analysis was performed by negative binomial distribution model with BH adjusted p value, *padj ≤ 0.05, **padj ≤ 0.01. Figures 6A-6D show that alternative suspended BME hydrogel culture formats have comparable organoid growth. (A) Photographs and (B) bright field images of colon organoids in 6-well plate cultures in BOBA, SOBA (syringe-extruded BME organoid assemblies), or SOBA fragment formats. The alternative formats enable faster culture preparation for scale-up. Scale bar is 1 mm. (C) Quantification of organoid diameter and (D) viable cells per well. Data presented are mean ± SD, one-way ANOVA multiple comparison test, n = 3 experiments. Figures 7A-7E show the use of suspended BME hydrogel organoid cultures for mid-production screening. (A) Schematic diagram of the experiment. SOBA chip organoid cultures in 225 cm2 flasks were ground to achieve a homogenous organoid suspension and then seeded into 96-well plates. (B) Bright field image of SOBA chip cultures grown in 225 cm2 flasks. Scale bar is 1 mm. (C) Bright field image of randomly selected wells across a 96-well plate showing comparable organoid density between wells. Scale bar is 1 mm. (D) Cell Titer Glo 3D (CTG) viability readouts show similar well-to-well variability between dome and suspended BME cultures in 96-well plates. Data presented are mean ± SD. Student’s t test. (E) Representative dose-response viability curves (CTG assay) of suspended BME organoids treated with diacerein, sorafenib, SN-38, or docetaxel for 3 days. Data presented are mean ± SD, n = 4. Figures 8A-8C show the generation of Transwell monolayers using suspended BME hydrogel organoids . (A) Schematic diagram of the experiment. SOBA chip organoid cultures in 225 cm2 flasks were digested into single-cell suspensions and then seeded into 96-well BME-coated Transwell chambers at confluence. (B) Bright field images of SOBA chip organoid-derived Transwell monolayers 3 days after seeding. Transwells were set up in either monolayer growth medium or monolayer differentiation medium. Scale bar is 100 µm. (C) Transepithelial electrical resistance (TEER) of Transwell monolayers cultured in either monolayer growth medium (filled circles ) or monolayer differentiation medium (open circles). Data presented are mean ± SD, n = 6 wells. FIG9 provides images of lung AT2 organoids embedded in hydrogel suspended in BOBA medium. FIG10 provides a schematic depicting exemplary conditions for generating hydrogel droplets, referred to herein as basement membrane organoid bead assemblies (BOBAs). Organoids or dissociated organoid cells suspended in cold liquid hydrogel are dispensed as droplets into a room temperature or warmer culture medium, which allows the hydrogel to solidify immediately upon dispensing into the culture medium. FIG. 11 provides a schematic diagram depicting exemplary conditions for producing hydrogel filaments, which are referred to herein as syringe-extruded organoid basement membrane assemblies (SOBAs). Organoids or dissociated organoid cells suspended in cold liquid hydrogel are extruded (by a syringe, pipette, or any other dispensing device) as filaments into a room temperature or warmer culture medium, which allows the hydrogel to solidify immediately upon dispensing into the culture medium.
Claims (117)
Applications Claiming Priority (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US63/508,132 | 2023-06-14 |
Publications (1)
Publication Number | Publication Date |
---|---|
TW202505022A true TW202505022A (en) | 2025-02-01 |
Family
ID=
Similar Documents
Publication | Publication Date | Title |
---|---|---|
US11920158B2 (en) | Cardiomyocyte maturation | |
Szot et al. | 3D in vitro bioengineered tumors based on collagen I hydrogels | |
Lee et al. | Engineering liver tissue spheroids with inverted colloidal crystal scaffolds | |
Li et al. | Generation of offspring-producing 3D ovarian organoids derived from female germline stem cells and their application in toxicological detection | |
Hookway et al. | Aggregate formation and suspension culture of human pluripotent stem cells and differentiated progeny | |
JP5894276B2 (en) | Somatic stem cells | |
Sakib et al. | Generation of porcine testicular organoids with testis specific architecture using microwell culture | |
JP2021500929A (en) | Compositions and methods | |
US10526575B2 (en) | Cell suspension medium and cell suspension medium additive for the three dimensional growth of cells | |
WO2014148562A1 (en) | Method for obtaining cell mass containing cancer stem cell | |
Hu et al. | Porcine uterine luminal fluid-derived extracellular vesicles improve conceptus-endometrial interaction during implantation | |
Pryzhkova et al. | Adaptation of human testicular niche cells for pluripotent stem cell and testis development research | |
van Loo et al. | Mass production of lumenogenic human embryoid bodies and functional cardiospheres using in-air-generated microcapsules | |
Frenster et al. | Establishing primary human glioblastoma tumorsphere cultures from operative specimens | |
JP7251595B2 (en) | METHOD FOR GENERATING DRUG-RESISTANT CELLS AND METHOD FOR SCREENING ANTICANCER AGENT | |
US20150240209A1 (en) | Methods for the preparation of fibroblasts | |
TW202505022A (en) | Methods of producing tissue-derived epithelial organoids and uses thereof | |
Zhao et al. | The trophoblast compartment helps maintain embryonic pluripotency and delays differentiation towards cardiomyocytes | |
WO2024259104A1 (en) | Methods of producing tissue-derived epithelial organoids and uses thereof | |
Gonçalves et al. | Human menstrual blood-derived mesenchymal cells improve mouse embryonic development | |
IL268268A (en) | Methods and compositions for tumor assessment | |
Di Nisio et al. | Silk-Ovarioids: Establishment and characterization of human ovarian primary cells 3D-model system | |
US20250019665A1 (en) | Scalable biomanufacturing of tumor organoids | |
Candelaria et al. | Three-dimensional culture in a bioengineered matrix and somatic cell complementation to improve growth and survival of bovine preantral follicles | |
US20210380934A1 (en) | Deterministic Culturing of Single Cells |