[go: up one dir, main page]

CN113577107A - Preparation method and application of stem cells - Google Patents

Preparation method and application of stem cells Download PDF

Info

Publication number
CN113577107A
CN113577107A CN202110557156.XA CN202110557156A CN113577107A CN 113577107 A CN113577107 A CN 113577107A CN 202110557156 A CN202110557156 A CN 202110557156A CN 113577107 A CN113577107 A CN 113577107A
Authority
CN
China
Prior art keywords
stem cells
cells
hmsc
serum
dental pulp
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110557156.XA
Other languages
Chinese (zh)
Inventor
施松涛
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sun Yat Sen University
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to CN202110557156.XA priority Critical patent/CN113577107A/en
Publication of CN113577107A publication Critical patent/CN113577107A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0664Dental pulp stem cells, Dental follicle stem cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/70Undefined extracts
    • C12N2500/80Undefined extracts from animals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/90Serum-free medium, which may still contain naturally-sourced components

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biotechnology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Hematology (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Rheumatology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

本发明属于生物医药领域,涉及一种干细胞的制备方法及其应用。本发明提供了干细胞的制备方法,所述干细胞使用的培养基为无血清培养基,所述干细胞为牙髓间充质干细胞,所述方法获得的干细胞用于抑制激活的T细胞以及免疫系统疾病的治疗。本发明研究发现,在无血清培养基培养下获得的hMSC‑DP,诱导T细胞凋亡的能力强于在有血清培养下获得的细胞,同时,将无血清培养的hMSC‑DP用于治疗DSS诱导的急性结肠炎,发现P5和P10代无血清培养的hMSC‑DP具有较强的治疗能力,提示无血清培养的hMSC‑DP具有运用于免疫疾病治疗的强大潜力。

Figure 202110557156

The invention belongs to the field of biomedicine, and relates to a preparation method and application of stem cells. The present invention provides a method for preparing stem cells, the medium used for the stem cells is serum-free medium, the stem cells are dental pulp mesenchymal stem cells, and the stem cells obtained by the method are used to inhibit activated T cells and immune system diseases Treatment. The research of the present invention found that hMSC-DP obtained under serum-free medium culture has a stronger ability to induce T cell apoptosis than cells obtained under serum-cultured culture, and at the same time, serum-free cultured hMSC-DP is used for the treatment of DSS Induced acute colitis, it was found that the serum-free cultured hMSC-DP of the P5 and P10 generations had a strong therapeutic ability, suggesting that the serum-free cultured hMSC-DP has a strong potential for the treatment of immune diseases.

Figure 202110557156

Description

Preparation method and application of stem cells
Technical Field
The invention belongs to the field of biological medicines, and relates to a preparation method and application of stem cells.
Background
Mesenchymal Stem Cells (MSCs) have been widely used clinically for the treatment of various diseases[1]. MSCs can be isolated from various tissues, such as bone marrow, umbilical cord tissue, adipose tissue and dental pulp[2]. International Society for Cell Therapy (ISCT)) The minimum standard for MSC application was established in 2006[3]. However, no criteria have been established for quality assessment of MSCs from a particular organization.
Since the discovery of hMSC-DP[4,5]Since then, their physiological and functional properties have been extensively studied[6]. The excellent proliferative capacity, dentin/osteogenesis capacity and immunoregulatory capacity of hMSC-DP have been widely recognized[6-8]. In recent years, hMSC-DP has been used in preclinical and clinical studies for tissue regeneration and immunotherapy[7,9-13]. The quality and potency of MSCs is crucial to ensure their therapeutic effect[14]. However, the MSC minimum criteria established by ISCT may not provide sufficient guidance for clinical use of hMSC-DP[3]
Previous studies have shown that MSCs cultured using Fetal Bovine Serum (FBS) may cause some ethical and immune response problems[15-17]. Significant quality differences between FBS batches also limit the large-scale production of MSCs. Therefore, Serum (SF) free cultured MSCs are necessary for future clinical practice. FBS substitutes, including human platelet lysate and chemically-defined SF media, appear to provide sufficient nutritional support for culture expansion of hMSC-DP and maintain its tissue regeneration pluripotency[18-20]. However, the detailed culture effect of SF medium on hMSC-DP and its cellular function are not fully understood.
Disclosure of Invention
In some embodiments, the present invention provides the use of dental pulp mesenchymal stem cells obtained by culturing the dental pulp mesenchymal stem cells in a serum-free medium for the preparation of a modulator of T cell activity.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 15.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 10.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 5.
In some embodiments, the dental pulp mesenchymal stem cells comprise DPSC and SHED.
In some embodiments, the modulator of T cell activity is a modulator of induction of T cell apoptosis.
In some embodiments, the dental pulp stem cells are used to treat an immune disease.
In some embodiments, the dental pulp stem cells are used to treat enteritis.
In some embodiments, the invention provides the use of dental pulp mesenchymal stem cells obtained by culturing the dental pulp mesenchymal stem cells in a serum-free medium for the preparation of a medicament for treating an immune disease.
In some embodiments, the present invention provides a method of preparing a stem cell using a serum-free medium, the stem cell being a dental pulp mesenchymal stem cell, the stem cell obtained by the method for modulating T cell activity.
In some embodiments, the stem cells obtained by the methods are used to induce apoptosis of T cells.
In some embodiments, the stem cells obtained by the methods are used to treat immune diseases.
In some embodiments, the stem cells obtained by the methods are used to treat enteritis.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 15.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 10.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 5.
In some embodiments, the dental pulp mesenchymal stem cells comprise DPSC and SHED.
In some embodiments, the present invention provides a modulator of T cell activity comprising dental pulp mesenchymal stem cells; the stem cells are obtained by using serum-free culture medium for culture.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 15.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 10.
In some embodiments, the number of passages of the dental pulp mesenchymal stem cells is less than or equal to passage 5.
In some embodiments, the modulator of T cell activity is for inducing apoptosis of a T cell.
In some embodiments, the modulator of T cell activity is used to treat an immune disease.
In some embodiments, the modulator of T cell activity is used to treat enteritis.
In some embodiments, the colitis is acute colitis.
In some embodiments, the T cell is an activated T cell.
The prior art shows that the stronger the ability to induce apoptosis of T cells, the greater the potential to treat immune diseases. The research of the invention finds that the MSC-DP obtained in the state of the serum-free culture medium integrally shows better capability of inducing T cell apoptosis (compared with the state of the serum culture), and particularly shows a result which is obviously superior to that in the state of the serum culture for stem cells with low generation number (better quality and potential).
In some embodiments, the serum-free medium comprises human platelet lysate.
In some embodiments, the serum-free medium further comprises DMEM.
In some embodiments, the serum-free medium further comprises L-glutamine or an L-glutamine analog.
In some embodiments, the L-glutamine analog is a GlutaMAX additive.
In some embodiments, the present invention provides a method for preparing stem cells using serum-free medium, wherein the stem cells are DPSCs, and wherein the stem cells obtained by the method are used for osteogenic differentiation.
At the P20 generation, the DPSC cells cultured in a serum-free state still have better osteogenic differentiation capacity, while the DPSC cells cultured in the serum lose the osteogenic differentiation capacity at the 20 th generation. Embodies that the DPSC under the serum-free culture condition can obtain and maintain stronger osteogenic differentiation capacity.
In some embodiments, the present studies have found that osteogenic differentiation can be achieved after a high passage number of DPSCs has been achieved, without serum.
In some embodiments, the invention provides a method for preparing stem cells, the culture medium used by the stem cells is serum-containing medium, the stem cells are SHED, and the stem cells obtained by the method are used for adipogenic differentiation.
In some embodiments, the number of SHEDs is greater than 15.
The research of the invention finds that for SHED, a serum culture medium is better selected during adipogenic differentiation, and the adipogenic differentiation capacity is obviously enhanced when the generation number reaches 20.
Drawings
FIG. 1 morphological features of hMSC-DP. a. Schematic representation of the isolation and culture of hMSC-DP under SE or SF culture conditions. The forms of hMSC-DP in P5, P10 and P20 generations under SE or SF culture conditions. Scale bar 50 μm. c. Morphological analysis of cells with high content imaging. hMSC-DP was co-stained with pharloid (orange), hoechst (blue) and WGA (green) at a scale bar of 100 μm, with n of 3 per group. d. Cell senescence assay, the percentage of SA- β -gal positive cells was calculated and compared between the groups P5, P10 and P20. Black arrows indicate positive staining of SA-. beta. -gal. Scale bar 100 μm. Each group n is 3-5. SE, serum culture; SF, serum free culture, bars are shown as the mean of each group. P <0.05, p <0.01, p < 0.001. ns, no significant difference.
FIG. 2 morphological features of hMSC-DP under SF culture conditions. a-b cell morphology analysis by high content imaging. Area comparison of nuclear (a) and cellular regions of DPSCs and SHEDs under SE and SF culture conditions (b). Each group n is 3. c. Cell senescence assay. The percentage of SA- β -gal positive cells was calculated and compared between SE and SF culture conditions. Each group n is 3-5. SE, serum; SF, serum free. The graph bar shows the average values of a-c. ns, no significant difference.
FIG. 3 proliferation potency and chromosomal stability of hMSC-DP at different generations. a. The colony forming ability of DPSCs and SHED under SE and SF culture conditions was evaluated by CFU-F analysis, and the number of clones of P5, P10 and P20 was calculated and compared, with n-3 per group. b the proliferative capacity of three different groups of donor-derived DPSCs and three different groups of donor-derived SHEDs was analyzed by population doubling assay. c proliferation rate of hMSC-DP was assessed by EdU staining. The percentage of EdU-positive cells was calculated for P5, P10 and P20 passages. d. Chromosome stability was assessed by karyotyping. The DPSCs and SHED karyotypes of P5, P10, and P20 generations were examined. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. P <0.05, p <0.01, p < 0.001. ns, no significant difference.
FIG. 4 proliferation potency of hMSC-DP under SF culture conditions. CFU-F analysis. The number of hMSC-DP clones in SE or SF culture conditions was calculated and compared. Each group n is 3. b. Population doubling scores under SE and SF culture conditions in the DPSC and SHED groups were calculated and compared, respectively. Each group n is 3. Edu analysis. The percentage of EdU-positive cells between SE and SF culture conditions in the DPSC and SHED groups was calculated independently and compared. Each group n is 3. Scale bar 200 μm. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. P < 0.05. ns, no significant difference.
FIG. 5 surface marker phenotype and in vitro immunomodulatory capacity of hMSC-DP at different passage numbers. a. MSC surface marker analysis of hMSC-DP at P5, P10 and P20 under SE and SF culture conditions was assessed by flow cytometry, showing a positive rate of CD34, CD45, CD73, CD90 and CD105, with n-3 per group. b. The percentage of CD146 positive cells at P5, P10 and P20 was calculated in DPSC and SHED, respectively. Each group n is 3. c to assess in vitro immunomodulatory capacity, hMSC-DP at P5, P10 and P20 were co-cultured with T cells and the percentage of apoptotic T cells was detected by flow cytometry. Each group n is 3. The bars are shown as the average of the groups. Comparison with control group: # p <0.05, # p <0.01, # p < 0.001. NS has no obvious difference with the control group. Comparison of hMSC-DP at P5, P10 and P20: p <0.05, p <0.01, p < 0.001. ns, no significant difference.
FIG. 6 surface marker phenotype and in vitro immunomodulatory capacity of hMSC-DP. Flow cytometry showed the percentage of CD146 positive hMSC-DP at P5, P10 and P20 under SE or SF culture conditions (a). The percentage of CD146 positive cells under SE and SF culture conditions was compared (b). Each group n is 3. c. The percentage of apoptotic T cells was calculated under SE and SF culture conditions. Each group n is 3. SE, serum; SF, serum free. The bars are shown as the average of the groups. P < 0.05. ns, no significant difference.
FIG. 7 multilineage differentiation of hMSC-DP. Osteogenic capacity of a-b hMSC-DP. (a) Comparison of the percentage of alizarin red staining at P5, P10 and P20. (b) Western blot analysis showed the expression levels of the osteogenic markers Runx2 and ALP. Each group n is 3. (c-d) lipidogenic capacity of hMSC-DP. (c) The percentage of oil red o-positive cells at P5, P10 and P20 passages was calculated and analyzed. (d) Western blot analysis showed the expression levels of adipogenic markers (including LPL and PPAR γ), with n-3 per group. e neurogenic differentiation of hMSC-DP. Immunofluorescent staining and Western blot analysis showed expression of the neural markers β iii-tubulin and NeuN. Scale bar 100 μm. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. P <0.05, p <0.01, p < 0.001. ns, no significant difference.
FIG. 8 multilineage differentiation of hMSC-DP. a. Alizarin red staining method. The osteogenic capacity of P5, P10 and P20 were compared under SE and SF culture conditions. Each group n is 3. b. Oil red O staining assay. The lipid forming ability of hMSC-DP of P5, P10 and P20 under SE and SF culture conditions was compared. Each group n is 3. Se, serum-cultured at a scale of 50 μm; SF, serum free culture. The bars are shown as the average of the groups. P < 0.01. ns, no significant difference.
FIG. 9 immunotherapeutic Effect of hMSC-DP transplantation in Dextran Sodium Sulfate (DSS) -induced experimental colitis. a. Body weights were recorded daily for 8 days in both the DPSC and SHED transplant groups. The relative change in body weight was calculated. Each group n is 3. b. Disease Activity Index (DAI) was assessed on day 8 of DSS feeding. Each group n is 3. c. Colon length was measured on day 8. Each group n is 3. d. Histological scores based on H & E stained images were assessed on day 8. Histological scores were calculated for each group. Each group n is 3. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. Comparison with untreated DSS group: # p <0.05, # p <0.01, # p < 0.001. NS, no significant difference from DSS group. Comparison of DPSCs and SHED for P5, P10 and P20: p <0.05, p <0.01, p < 0.001. ns, no significant difference.
FIG. 10 therapeutic effect of hMSC-DP on experimental colitis. a. Disease Activity Index (DAI) was compared between treatment groups of DPSCs and SHED between SE and SF culture conditions at P5, P10 and P20 generations. Each group n is 3. b. Colon length after treatment of P5, P10 and P20 passages DPSCs and SHED under SE and SF culture conditions was compared. Each group n is 3. c. Histological structures were examined by H & E staining. Histological scores of the groups were compared under SE and SF culture conditions. Each group n is 3. Scale bar 100 μm. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. ns, no significant difference.
FIG. 11CD146 expression is associated with hMSC-DP proliferation, differentiation and immunomodulation. Western blot analysis showed expression of CD146 in P5, P10 and P20 in DPSC and SHED under SE or SF culture conditions. b. By Western blot analysis, scd 146 down-regulated the expression level of DPSCs and SHED for CD146 at P10 generation. c. Flow cytometry analysis showed a reduction in the percentage of CD146 positive cells after siCD146 treatment. Each group n is 3. EdU staining showed a decrease in the proliferation rate of hMSC-DP following siCD146 treatment. Each group n is 3. e. After coculture of siCD146 transfected hMSC-DP with T cells, the percentage of apoptotic T cells was reduced as assessed by flow cytometry. Each group n is 3. The expression of f-g knockdown CD146 reduced the osteogenic capacity of hMSC-DP, as assessed by the percentage of alizarin red staining (f) and Western blot for osteogenic markers (g). Each group n is 3. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. P <0.01, p < 0.001. ns, no significant difference.
FIG. 12 the expression level of CD146 is related to hMSC-DP characteristics. Correlation of cd146 with each experimental parameter. b. The linear regression plots show the correlation between the expression level of CD146 and various experimental parameters, including CFU-F clones, percent EdU positive cells, percent alizarin red positive area, percent SA- β -gal positive cells, DAI score, HAI score and colon length in DSS-induced colitis treated by each group of cells.
FIG. 13CD146 controls the proliferation and immunomodulatory capacity of hMSC-DP through the ERK pathway. Western blot analysis showed expression levels of the ERK pathway in P5, P10 and P20 passages of DPSC and SHED under SE or SF culture conditions. b Western blot analysis showed that ERK pathways were down-regulated by siCD146 treatment in hMSC-DP. c ERK pathway inhibitor PD98059 inhibits the expression level of p-ERK. EdU staining showed a decrease in the proliferation rate of hMSC-DP after PD98059 treatment. Each group n is 3. e. As shown by flow cytometry, the percentage of apoptotic T cells decreased after co-culturing PD98059 treated hMSC-DP with T cells. Each group n is 3. The osteogenesis of hMSC-DP was examined after pd98059 treatment and was assessed by the percentage of alizarin red staining (f) and the protein expression level of the osteogenic marker (g). Each group n is 3. SE, serum culture; SF, serum free culture. The bars are shown as the average of the groups. P <0.05, p <0.01, p < 0.001. ns, no significant difference.
Detailed Description
The technical solutions of the present invention are further illustrated by the following specific examples, which do not represent limitations to the scope of the present invention. Insubstantial modifications and adaptations of the present invention by others of the concepts fall within the scope of the invention.
List of abbreviations: hMSC-DP: human mesenchymal stem cells from dental pulp; and (4) DPSC: adult dental pulp stem cells; SHED: stem cells of deciduous teeth of a human being; and SE: serum; SF: serum-free; MSC: mesenchymal stem cells; ISCT: the international society for cell therapy; and (3) SOP: standard operating procedures; FBS: fetal bovine serum; EdU: 5-ethynyl-20-deoxyuridine; PD: multiplying the population; CFU: cloning a forming unit; WGA: wheat germ agglutinin; beta-gal: beta-galactosidase; SA- β -gal: senescence-associated beta-galactosidase enzyme; runx 2: runt-related transcription factor 2; ALP: alkaline phosphatase; PPAR γ: peroxisome proliferator activated receptor- γ 2; LPL: lipoprotein lipase; DSS: dextran sodium sulfate; DAI: an index of disease activity; HAI: histological activity index.
Materials and methods
1. Mouse
Male C57BL/6J mice were purchased from Zhongshan university, Guangzhou, China. All animal experiments were performed under agency approved protocols for animal studies (university of Zhongshan, SYSU-IACUC-2020-000394).
2. Isolated culture of hMSC-DP
Deciduous incisors and third molars, which were exfoliated by humans, were obtained from biological samples discarded in the oral hospital affiliated to Guanghua medical college, Zhongshan university.
All teeth were intact without any inflammation. The protocol (KQEC-2020-055-01) has been approved by the institutional medical Ethics Committee of the oral Hospital, university of Zhongshan. In the manner previously reported[4,5]DPSCs and SHED were isolated and cultured until passage 2 (P2). hMSC-DP from P2 generation was cultured in the presence of Serum (SE) or in the absence of Serum (SF) until P20 generation.
Serum culture of hMSC-DP culture was performed in the previously reported manner[4,5]. SF-cultured hMSC-DP was cultured in high glucose Dulbecco's modified Eagle Medium (DMEM, Invitrogen) supplemented with 5% volume fraction human platelet lysate (Compass biological), 100U/ml penicillin, 100mg/ml streptomycin (Gibco) and 1% volume fraction GlutaMAXTM supplement (Gibco). After the cells were confluent, they were digested and passaged by adding 0.25% trypsin (Gibco) containing 1mM EDTA and continuously cultured in SE or SF medium. At each passage, approximately 1X 10 was selected6hMSC-DP was seeded on 10cm dishes (Corning) and the excess cells were plated in serum-free cryopreservation medium (CELLBA)NKERTM2) Storing in a-80 deg.C refrigerator. hMSC-DP from generations P5, P10 and P20 was selected for subsequent experiments.
3. Antibodies and reagents
anti-CD 34, CD45, CD73, CD90, CD105, CD146, and 7-AAD antibodies were purchased from BD Biosciences. anti-CD 3, CD8, and Annexin V antibodies were purchased from BioLegend. anti-Runx 2, ERK and p-ERK antibodies were purchased from Cell Signaling Technology. anti-CD 146, ALP, β III-tubulin and NeuN antibodies were purchased from Abcam. anti-LPL and β -actin antibodies were purchased from Invitrogen. anti-PPAR γ antibodies were purchased from Santa Cruz. WGA-488 was purchased from Biosharp. Hoechst 33342 was purchased from Sigma. The kFluor488-EDU cell proliferation assay kit was purchased from KeyGEN Biotech. LipofectamineTMRNAiMAX transfection reagent was purchased from Invitrogen. CD146 siRNA was purchased from rib Biotech. PD98059 was purchased from Beyotime. Cell senescence assay kits were purchased from Merck Millipore. Sodium dextran sulfate (DSS) was purchased from MP Biochemicals.
4. Flow cytometry analysis
For analysis of cell surface markers, 106hMSC-DP suspensions were prepared at a density of 100. mu.l for the P5, P10 and P20 generations. Thereafter, 1. mu.l of the antibody was added to 100. mu.l of the cell suspension and incubated at 4 ℃ for 30 minutes in the dark, followed by using a flow cytometer (ACEA novoCyte)TM) And (6) carrying out analysis. Apoptotic T cells were stained with CD3 and 7AAD antibodies for 30 min at 4 ℃, in the dark, washed twice with PBS, and then incubated with Annexin V antibody.
5. In vitro immunomodulatory capacity of hMSC-DP
Mixing P5, P10 and P20 (1X 10)5Per well) passage of hMSC-DP were seeded on 12-well culture plates (Corning) and incubated for 24 hours. T cells pre-stimulated by anti-CD 3 and CD8 antibodies were loaded directly onto hMSC-DP for 2 days of culture. Apoptotic T cell numbers were detected by flow cytometry.
6. Karyotyping analysis
Karyotyping of hMSC-DP, including chromosome number and G-band karyotype, was performed by Shanghai Haoge Biotechnology Inc. (Shanghai, China).
7. EdU dyeing method
Mixing hMSC-DP (2X 10 per well)4Individual cells) were seeded into 24-well plates (Corning) and cultured at 37 ℃ for 24 hours. Cells were treated with 200. mu.l of medium containing 50. mu.M of 5-ethynyl-20-deoxyuridine (EdU) for 2 hours. Cells were then detected using the EdU cell proliferation assay kit (KeyGEN, BioTECH) according to the manufacturer's instructions.
8. Population doubling assay
hMSC-DP generation P4 at 2X 105The individual cells were seeded in 60mm dishes (Corning) containing SE or SF medium. When the cells reach about 70% -80% confluence, the cells are harvested and inoculated with the same number of cells into the culture medium. The population doubling rate (PD) was calculated by the following formula: PD ═ log2 (number of harvested cells/number of seeded cells). The total number of accumulated PD was determined from P5 to the time when cell division ceased. Each group was tested in duplicate with 3 independent cells from different sources.
9. Clone Forming Unit (CFU) assay
A total of 1,000 hMSC-DP per group were plated in 6cm dishes (Corning). After 10 days, cells were fixed with 4% paraformaldehyde and stained with 1% crystal violet for 5 minutes. Clones containing 50 or more cells were selected for counting. This experiment was repeated three times.
10. Cellular morphological analysis by high content imaging
3,000 hMSC-DP of each group were seeded in 96-cell well plates (Corning) and cultured for 24 hours. After washing with PBS, fixation was performed for 10 minutes in 4% formaldehyde at room temperature. Cells were stained with Phalloidin, WGA and Hoechst 33342. The plates were scanned in an Operetta CLS and each group of cells was analyzed for cell size and shape using Harmony software (PerkinElmer).
11. Cellular senescence assay
Each group was given a total of 5X 104Each hMSC-DP was inoculated in 24-well plates (Corning) and cultured for 24 hours. The cellular senescence assay was then performed according to the protocol provided by the cellular senescence assay kit (Merck Millipore).
12. Inducing osteogenesis, adipogenesis and neurogenesis differentiation
About 3X 10 per group5Individual hMSC-DP were cultured in 6cm plates until complete fusion. In the previously reported manner [21,22 ]]Osteogenic and adipogenic differentiation induction and evaluation were performed. To induce neuro-differentiation, a total of 4X 10 were used4The individual cells were seeded on a 24-well culture plate (Corning) and cultured for 24 hours. The cells were then replaced with neural differentiation medium and cultured for 10 days. Neural differentiation assessment was performed as previously reported[5]
13. siRNA transfection and chemical treatment
For siRNA transfection, hMSC-DP (5X 10)5Per well) were inoculated in 6-well plates in low serum medium (Opti-MEM, Gibco) and treated with CD146 siRNA (RIBO, China) or lipofectamine reagent control vector siRNA (invitrogen) for 24 hours according to the manufacturer's instructions. For chemical treatment, hMSC-DP was treated with 10. mu.M PD 98059. The treated cells were collected or used for further experiments.
14. hMSC-DP transplantation in acute colitis mice
Acute colitis was induced in mice on drinking water containing 3% DSS (MP biochemicals) for 8 days. On day 3 after feeding with DSS water, the total number of each group was 1X 106One hMSC-DP was infused intravenously into colitis mice. All mice were euthanized and analyzed on day eight. Mice with induced colitis were evaluated as described previously[23]
15. Statistical analysis
GraphPad Prism 8 was used to perform statistical analysis. Comparisons between the two groups were analyzed using the independent student's two-tailed t-test, while comparisons between more than two groups were analyzed using one-way analysis of variance. P values <0.05 were considered statistically significant.
Example 1 morphological characterization of hMSC-DP
Experimental materials and methods are shown above in the materials and methods section.
A third molar from three donor sources was collected for DPSC isolation and a deciduous incisor from three donor sources was collected for SHED isolation. All donors were healthy volunteers, and teeth were free of caries and inflammation. According to the previous report[4,5]From toothhMSC-DP, including DPSC and SHED, was isolated from the marrow. The P2 generation hMSC-DP was isolated and propagated in conventional serum-containing FBS (SE) or serum-free (SF) culture conditions with human platelet lysate to obtain the DPSC-SE, DPSC-SF, SHED-SE and SHED-SF groups (FIG. 1 a).
Cell morphology is considered an indicator of MSC function[24]. hMSC-DP from all donors were adherent to petri dishes under SE and SF culture conditions, and P5 and P10 passages showed elongated spindle-shaped morphology. With serial passage to passage 20, hMSC-DP morphology became large, flat and irregular (fig. 1 b). To further analyze the morphological characteristics of the cells, we used high content imaging methods to analyze cell morphology, labeling the nucleus with hoechest, actin-labeled cytoskeleton and Wheat Germ Agglutinin (WGA) -labeled cell membrane[25]. More than 500 cells were analyzed per group. In DPSC-SE (P)<0.01),DPSC-SF(P<0.05),SHED-SE(P<0.05) and SHED-SF (P)<0.01) group, the nucleus size of hMSC-DP at P20 generation was significantly larger than P5. The cell size of the P20 generation hMSC-DP also showed a tendency to become larger compared to that of P10 (FIG. 1 c). There was no significant difference in nuclear size between SE and SF culture conditions (FIG. 2a), but the cell size of SHED tended to decrease in SF culture compared to SE culture (P10 for P)<0.05, P20 time P<0.01). (FIG. 2 b).
Since cellular senescence may be associated with changes in cell size and nuclear size[26]Therefore, we evaluated hMSC-DP for β -galactosidase (β -gal) positive senescing generations P5, P10, and P20. We found that the percentage of β -gal positive cells in each group increased from P5 to P20, and that this trend was statistically significant when comparing P5 to P20 (P20 in each group)<0.001) (fig. 1 d). However, there was no significant difference between SE and SF culture conditions (fig. 2 c). These data indicate that serial passage affects hMSC-DP characteristics, especially at P20.
Example 2 proliferative Capacity and chromosomal stability of hMSC-DP
Experimental materials and methods are shown above in the materials and methods section.
Proliferation potency is an important parameter for assessing the function of MSCs. We inoculated hMSC-DP at a density of 1,000 cells/plate and cultured it for 10 days. The number of clones decreased from P5 to P20 (FIG. 3 a). Interestingly, SHED-SE showed higher clone numbers compared to the P20 generation SHED-SF group (FIG. 4 a). To examine the proliferative potential of hMSC-DP, population doubling scores were calculated from three donor-derived DPSCs and three donor-derived SHEDs. These cells showed similar proliferation capacity (FIG. 3b), with no significant difference under SE or SF culture conditions (FIG. 4 b). In addition, staining with 5-ethynyl-2' -deoxyuridine (EdU) showed a decrease in proliferation rate of hMSC-DP with serial passage, significantly lower at P20 (fig. 3c) compared to P5 (P < 0.001). SE and SF cultures did not differ in the EdU positivity rate (FIG. 4 c).
After culture amplification, it is crucial to use clinical grade human MSCs with normal genotype and chromosomal stability[27]. We evaluated the karyotypes of P5, P10 and P20 passages DPSCs and SHED under SE and SF culture conditions. This is the same as the previous report that human MSCs maintain a relatively stable genome during culture amplification, and no chromosomal aberrations were detected in any of the test groups (fig. 3d)[28,29]
Example 3 surface phenotype and in vitro immunomodulatory Capacity of hMSC-DP
Experimental materials and methods are shown above in the materials and methods section.
Next, we analyzed a series of stem cell surface markers, including those described by ISCT[3]. hMSC-DPs from P5, P10 and P20 generations expressed the MSC surface markers CD73, CD90 and CD105, whereas they did not express CD34 and CD45 under SE and SF culture conditions (FIG. 5 a). Notably, the expression level of CD146 showed heterogeneity between groups (fig. 5a, fig. 6 a). We observed a gradual decrease in CD146 expression from P5 to P10 and from P10 to P20. CD146 expression at P20 was significantly reduced compared to P5 (DPSC-SE, DPSC-SF, SHED-SE and SHED-SF: P)<0.001) (fig. 5 b). Neither SE nor SF culture conditions showed an effect on CD146 expression levels (fig. 6 b). Previous studies have shown that CD146 is not only a melanoma cell adhesion molecule, but also a cell surface receptor for various ligands, involved in many physiological and pathological processes of cells[30]. Of importanceThat is, CD146 is also expressed in MSCs.
To evaluate the in vitro immunomodulatory capacity of hMSC-DP, we co-cultured hMSC-DP with activated T cells for 2 days. Flow cytometry analysis showed that the hMSC-DP of P5 and P10 passages were able to significantly induce the level of T cell apoptosis compared to the control group, but the hMSC-DP of P20 lost the ability to induce T cell apoptosis (FIG. 5 c). hMSC-DP of P5 and P10 showed higher immunomodulatory capacity under SF culture compared to SE cultured cells. DPSCs of P5 passage showed significantly reduced immunomodulatory capacity (P <0.05) (fig. 6 c). These results indicate that hMSC-DP cultured in SF medium may be more suitable for immunotherapy.
Example 4 multilineage differentiation of hMSC-DP
Experimental materials and methods are shown above in the materials and methods section.
The multi-lineage differentiation potential is a key parameter for assessing MSC function. To evaluate the osteogenic capacity of hMSC-DP, we cultured hMSC-DP for two weeks at generations P5, P10 and P20 in SE and SF osteogenic induction medium. Alizarin red staining showed that hMSC-DP showed a reduced tendency to mineralized nodule formation from P5 to P20 passages, indicating that continued passaging impaired the osteogenic differentiation capacity of hMSC-DP (fig. 7a, fig. 8 a). Western blot analysis confirmed that serial passage reduced the expression levels of run-associated transcription factor 2(Runx2) and alkaline phosphatase (ALP) (FIG. 7 b). Interestingly, P20 passage SHED still had a relatively strong osteogenic capacity under SF culture compared to SE cultured DPSCs, whereas P20 passage DPSCs had a significantly stronger capacity to form mineralized nodules under SF culture. All these results indicate that SF medium has an advantage in maintaining the osteogenic ability of hMSC-DP.
Our previous studies showed that hMSC-DP was able to differentiate into adipocytes, but with limited capacity[4,5]. Here, we show that hMSC-DP from P5 to P20 passages showed limited adipogenic differentiation capacity after 6 weeks of adipogenic induction in SE or SF medium (FIG. 7c, FIG. 8 b). Western blot analysis confirmed the expression of two adipocyte-specific marker proteins in each group, peroxisome proliferator-activated receptor- γ 2(PPAR γ) and lipoprotein lipase (LPL) (fig. 7 d).
Previous studies have shown that DPSCs and SHEDs are capable of differentiating into neural cells, presumably because they originate from the neural crest[31-33]. hMSC-DP maintained neuro-differentiation ability from P5 to P20 under SE or SF culture conditions, and expression of the neuronal markers β III tubulin and NeuN was also demonstrated by Western blot and immunostaining evaluation (fig. 7 e).
Example 5hMSC-DP demonstrates immunotherapeutic Effect in Dextran Sodium Sulfate (DSS) -induced experimental colitis
Experimental materials and methods are shown above in the materials and methods section.
DSS-induced experimental colitis has been widely used to assess the in vivo immunomodulatory capacity of mouse MSCs[23,34]. To further evaluate the immunomodulatory capacity of hMSC-DP, we injected SE and SF cultured P5, P10 and P20 passages of hMSC-DP into experimental colitis mice by tail vein infusion on day 3 after induction of 3% DSS. Experimental colitis mice were sacrificed 8 days after DSS induction. It was observed at day 8 that DPSC-SE of P5 generation and all DPSCs cultured under SF improved the body weight of the mice; wherein with P10 (P)<0.05) and P20 (P)<0.05) cells, better body weight recovery (P) was achieved with the P5 generation of DPSC-SF<0.01). The SHED-SF of P20 lost its ability to improve body weight to some extent (FIG. 9 a). Treatment with hMSC-DP at generation P5 significantly improved the Disease Activity Index (DAI) compared to the untreated DSS group, whereas hMSC-DP at generation P20 did not improve DAI (FIG. 9 b). Anatomical images of each group of colons showed that both DPSC-SE and SF at P5 had therapeutic effect on DSS-induced colitis; SHED-SE in P5 and SHED-SF in P5 and P10 improved colon length compared to untreated group (FIG. 9c, FIG. 10 b). Histological scoring of HE stained images showed that except DPSC-SE and SHED at P20, hMSC-DP of the other groups had therapeutic effect on DSS-induced colitis (fig. 9d, fig. 10 c). In general, the immunoregulatory capacity of stem cells gradually decreases with continued passage: in experimental colitis mice, the therapeutic effect of hMSC-DP at P10 was weaker than that of hMSC-DP at P5, while hMSC-DP at P20 showed little therapeutic effect. The DPSC cultured by SF shows stronger immunity on the whole than the DPSC cultured by SEModulation (fig. 10).
Example 6 functional surface molecule CD146 prediction of hMSC-DP function
Experimental materials and methods are shown above in the materials and methods section.
CD146 is one of the surface markers of hMSC-DP[35]. Previous studies have shown that MSCs with high CD146 expression have better osteogenic and immunomodulatory capacity than MSCs with low CD146 expression[36-38]. We found that the expression level of CD146 in hMSC-DP decreased with passage (FIG. 11 a). To further explore the role of CD146 in hMSC-DP, we blocked the expression of CD146 in hMSC-DP with siCD146 as shown by Western blotting and flow cytometry (FIG. 11b, c). Following siCD146 interference, EdU staining, T cell apoptosis rate, alizarin red staining, and Western blot analysis of ALP and Runx2 demonstrated significant decreases in proliferation, in vitro immunomodulation, and osteogenic capacity (FIG. 11 d-g). Furthermore, we analyzed the correlation between these potency indices and CD146 expression levels and found that the potency of hMSC-DP was positively correlated with CD146 expression levels, particularly in terms of proliferation, osteogenesis and in vivo immunoregulatory capacity (fig. 12). These results indicate that CD146 is a functional surface marker, reflecting the function of hMSC-DP.
It is well known that the ERK/p-ERK pathway plays a key role in the regulation of MSC biological behavior[22,39,40]. We found that the expression level of p-ERK, but not ERK, was consistent with the level of CD146 in hMSC-DP (FIG. 13 a). Western blot results showed that siCD146 treatment down-regulated the expression of p-ERK in hMSC-DP (FIG. 13b), indicating that CD146 may maintain hMSC-DP function through the ERK/p-ERK pathway. To validate this hypothesis, we used the ERK inhibitor PD98059 to reduce the expression level of pERK in hMSC-DP as assessed by Western blot experiments (FIG. 13 c). As shown by EdU staining and T cell apoptosis rate, we found that it significantly reduced proliferation rate and immunosuppressive capacity (fig. 13d, e). Interestingly, osteogenic differentiation was not affected in the DPSCs group but increased in the SHED group after treatment with p-ERK inhibitors (FIG. 13f, g)[22,41]. These results indicate that CD146 can be maintained fine by the ERK pathway in hMSC-DPCell proliferation and immunoregulatory ability, but not the osteogenic effect.
Reference to the literature
1Pittenger MF,Discher DE,Péault BM et al.Mesenchymal stem cell perspective:cell biology to clinical progress.NPJ Regen Med 2019;4:22.
2Marquez-Curtis LA,Janowska-Wieczorek A,McGann LE et al.Mesenchymal stromal cells derived from various tissues:Biological,clinical and cryopreservation aspects.Cryobiology 2015;71(2):181-197.
3Dominici M,Le Blanc K,Mueller I et al.Minimal criteria for defining multipotent mesenchymal stromal cells.The International Society for Cellular Therapy position statement.Cytotherapy 2006;8(4):315-317.
4Gronthos S,Mankani M,Brahim J et al.Postnatal human dental pulp stem cells(DPSCs)in vitro and in vivo.Proc Natl Acad Sci U S A 2000;97(25):13625-13630.
5Miura M,Gronthos S,Zhao M et al.SHED:stem cells from human exfoliated deciduous teeth.Proc Natl Acad Sci U S A 2003;100(10):5807-5812.
6Sui B,Wu D,Xiang L et al.Dental Pulp Stem Cells:From Discovery to Clinical Application.Journal of Endodontics 2020;46(9):S46-S55.
7Botelho J,Cavacas MA,Machado V et al.Dental stem cells:recent progresses in tissue engineering and regenerative medicine.Ann Med 2017;49(8):644-651.
8Shi X,Mao J,Liu Y.Pulp stem cells derived from human permanent and deciduous teeth:Biological characteristics and therapeutic applications.Stem Cells Transl Med 2020;9(4):445-464.
9Bakopoulou A,About I.Stem Cells of Dental Origin:Current Research Trends and Key Milestones towards Clinical Application.Stem Cells Int 2016;2016:4209891.
10Yamada Y,Nakamura-Yamada S,Kusano K et al.Clinical Potential and Current Progress of Dental Pulp Stem Cells for Various Systemic Diseases in Regenerative Medicine:A Concise Review.Int J Mol Sci2019;20(5).
11Xuan K,Li B,Guo H et al.Deciduous autologous tooth stem cells regenerate dental pulp after implantation into injured teeth.Sci Transl Med 2018;10(455).
12Prasad MGS,Ramakrishna J,Babu DN.Allogeneic stem cells derived from human exfoliated deciduous teeth(SHED)for the management of periapical lesions in permanent teeth:Two case reports of a novel biologic alternative treatment.J Dent Res Dent Clin Dent Prospects 2017;11(2):117-122.
13Manimaran K,Sankaranarayanan S,Ravi VR et al.Treatment of osteoradionecrosis of mandible with bone marrow concentrate and with dental pulp stem cells.Ann Maxillofac Surg 2014;4(2):189-192.
14Ankrum J,Karp JM.Mesenchymal stem cell therapy:Two steps forward,one step back.Trends Mol Med2010;16(5):203-209.
15Jin X,Xu Q,Champion K et al.Endotoxin contamination of apolipoprotein A-I:effect on macrophage proliferation--a cautionary tale.Atherosclerosis 2015;240(1):121-124.
16Tekkatte C,Gunasingh GP,Cherian KM et al."Humanized"stem cell culture techniques:the animal serum controversy.Stem Cells Int 2011;2011:504723.
17Spees JL,Gregory CA,Singh H et al.Internalized antigens must be removed to prepare hypoimmunogenic mesenchymal stem cells for cell and gene therapy.Mol Ther 2004;9(5):747-756.
18Coates DE,Alansary M,Friedlander L et al.Dental pulp stem cells in serum-free medium for regenerative medicine.Journal of the Royal Society of New Zealand 2019;50(1):80-90.
19Qu C,Brohlin M,Kingham PJ et al.Evaluation of growth,stemness,and angiogenic properties of dental pulp stem cells cultured in cGMP xeno-/serum-free medium.Cell Tissue Res 2020;380(1):93-105.
20Iwanaka T,Yamaza T,Sonoda S et al.A model study for the manufacture and validation of clinical-grade deciduous dental pulp stem cells for chronic liver fibrosis treatment.Stem Cell Res Ther 2020;11(1):134.21Matsui M,Kobayashi T,Tsutsui TW.CD146 positive human dental pulp stem cells promote regeneration of dentin/pulp-like structures.Hum Cell 2018;31(2):127-138.
22Liu Y,Jing H,Kou X et al.PD-1 is required to maintain stem cell properties in human dental pulp stem cells.Cell Death Differ 2018;25(7):1350-1360.
23Akiyama K,Chen C,Wang D et al.Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis.Cell Stem Cell 2012;10(5):544-555.
24Haasters F,Prall WC,Anz D et al.Morphological and immunocytochemical characteristics indicate the yield of early progenitors and represent a quality control for human mesenchymal stem cell culturing.JAnat 2009;214(5):759-767.
25Bray M-A,Singh S,Han H et al.Cell Painting,a high-content image-based assay for morphological profiling using multiplexed fluorescent dyes.Nature Protocols 2016;11(9):1757-1774.
26Zhao H,Darzynkiewicz Z.Biomarkers of cell senescence assessed by imaging cytometry.Methods in molecular biology(Clifton,NJ)2013;965:83-92.
27Colter DC,Sekiya I,Prockop DJ.Identification of a subpopulation of rapidly self-renewing and multipotential adult stem cells in colonies of human marrow stromal cells.Proc Natl Acad Sci U S A2001;98(14):7841-7845.
28Bernardo ME,Zaffaroni N,Novara F et al.Human bone marrow derived mesenchymal stem cells do not undergo transformation after long-term in vitro culture and do not exhibit telomere maintenance mechanisms.Cancer Res 2007;67(19):9142-9149.
29Lange C,Cakiroglu F,Spiess AN et al.Accelerated and safe expansion of human mesenchymal stromal cells in animal serum-free medium for transplantation and regenerative medicine.J Cell Physiol2007;213(1):18-26.
30Wang Z,Xu Q,Zhang N et al.CD146,from a melanoma cell adhesion molecule to a signaling receptor.Signal Transduct Target Ther 2020;5(1):148.
31Mayo V,Sawatari Y,Huang CY et al.Neural crest-derived dental stem cells--where we are and where we are going.J Dent 2014;42(9):1043-1051.
32Li D,Zou X-Y,El-Ayachi I et al.Human Dental Pulp Stem Cells and Gingival Mesenchymal Stem Cells Display Action Potential Capacity In Vitro after Neuronogenic Differentiation.Stem Cell Reviews and Reports 2019;15(1):67-81.
33Anoop M,Datta I.Stem Cells Derived from Human Exfoliated Deciduous teeth[shed]in Neuronal Disorders:A Review.Curr Stem Cell Res Ther 2020.
34Shuai Y,Liao L,Su X et al.Melatonin Treatment Improves Mesenchymal Stem Cells Therapy by Preserving Stemness during Long-term In Vitro Expansion.Theranostics 2016;6(11):1899-1917.
35Shi S,Gronthos S.Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp.Journal of Bone and Mineral Research 2003;18(4):696-704.
36Wangler S,Menzel U,Li Z et al.CD146/MCAM distinguishes stem cell subpopulations with distinct migration and regenerative potential in degenerative intervertebral discs.Osteoarthritis Cartilage2019;27(7):1094-1105.
37Wu CC,Liu FL,Sytwu HK et al.CD146+mesenchymal stem cells display greater therapeutic potential than CD146-cells for treating collagen-induced arthritis in mice.Stem Cell Res Ther 2016;7:23.
38Samsonraj RM,Rai B,Sathiyanathan P et al.Establishing criteria for human mesenchymal stem cell potency.Stem Cells 2015;33(6):1878-1891.
39Higuchi C,Myoui A,Hashimoto N et al.Continuous inhibition of MAPK signaling promotes the early osteoblastic differentiation and mineralization of the extracellular matrix.J Bone Miner Res 2002;17(10):1785-1794.
40Schindeler A,Little DG.Ras-MAPK signaling in osteogenic differentiation:friend or foe?.J Bone Miner Res 2006;21(9):1331-1338.
41Li B,Qu C,Chen C et al.Basic fibroblast growth factor inhibits osteogenic differentiation of stem cells from human exfoliated deciduous teeth through ERK signaling.Oral Dis 2012;18(3):285-292.
42Xuan K,Li B,Guo H et al.Deciduous autologous tooth stem cells regenerate dental pulp after implantation into injured teeth.Sci Transl Med 2018;10(455).
43Astori G,Amati E,Bambi F et al.Platelet lysate as a substitute for animal serum for the ex-vivo expansion of mesenchymal stem/stromal cells:present and future.Stem Cell Res Ther 2016;7(1):93.
44Capelli C,Pedrini O,Valgardsdottir R et al.Clinical grade expansion of MSCs.Immunol Lett 2015;168(2):222-227.
45Paliwal S,Chaudhuri R,Agrawal A et al.Human tissue-specific MSCs demonstrate differential mitochondria transfer abilities that may determine their regenerative abilities.Stem Cell Res Ther 2018;9(1):298.
46Ma L,Makino Y,Yamaza H et al.Cryopreserved dental pulp tissues of exfoliated deciduous teeth is a feasible stem cell resource for regenerative medicine.PLoS One 2012;7(12):e51777.
47Andrukhov O,Behm C,Blufstein A et al.Immunomodulatory properties of dental tissue-derived mesenchymal stem cells:Implication in disease and tissue regeneration.World J Stem Cells 2019;11(9):604-617.
48Li N,Hua J.Interactions between mesenchymal stem cells and the immune system.Cell Mol Life Sci 2017;74(13):2345-2360.
49Shi Y,Wang Y,Li Q et al.Immunoregulatory mechanisms of mesenchymal stem and stromal cells in inflammatory diseases.Nat Rev Nephrol 2018;14(8):493-507.
50Krampera M,Galipeau J,Shi Y et al.Immunological characterization of multipotent mesenchymal stromal cells--The International Society for Cellular Therapy(ISCT)working proposal.Cytotherapy 2013;15(9):1054-1061.
51Wang Z,Xu Q,Zhang N et al.CD146,from a melanoma cell adhesion molecule to a signaling receptor.Signal Transduction and Targeted Therapy 2020;5(1).
52Bowles AC,Kouroupis D,Willman MA et al.Signature quality attributes of CD146(+)mesenchymal stem/stromal cells correlate with high therapeutic and secretory potency.Stem Cells 2020;38(8):1034-1049.
53Wang XT,Rao NQ,Fang TJ et al.[Comparison of the properties of CD146 positive and CD146 negative subpopulations of stem cells from human exfoliated deciduous teeth][in chi].Beijing Da Xue Xue Bao Yi Xue Ban 2018;50(2):284-292.
54Almalki SG,Agrawal DK.ERK signaling is required for VEGF-A/VEGFR2-induced differentiation of porcine adipose-derived mesenchymal stem cells into endothelial cells.Stem Cell Research&Therapy 2017;8(1):113.
55Zhang S,Chuah SJ,Lai RC et al.MSC exosomes mediate cartilage repair by enhancing proliferation,attenuating apoptosis and modulating immune reactivity.Biomaterials 2018;156:16-27.
56Ma Y,Zhang H,Xiong C et al.CD146 mediates an E-cadherin-to-N-cadherin switch during TGF-βsignaling-induced epithelial-mesenchymal transition.Cancer Lett 2018;430:201-214.
57Jiang T,Zhuang J,Duan H et al.CD146 is a coreceptor for VEGFR-2 in tumor angiogenesis.Blood 2012;120(11):2330-2339.
58Jaiswal RK,Jaiswal N,Bruder SP et al.Adult human mesenchymal stem cell differentiation to the osteogenic or adipogenic lineage is regulated by mitogen-activated protein kinase.J Biol Chem2000;275(13):9645-9652.
59Murakami J,Ishii M,Suehiro F et al.Vascular endothelial growth factor-C induces osteogenic differentiation of human mesenchymal stem cells through the ERK and RUNX2 pathway.Biochem Biophys Res Commun 2017;484(3):710-718.
60Baker N,Sohn J,Tuan RS.Promotion of human mesenchymal stem cell osteogenesis by PI3-kinase/Akt signaling,and the influence of caveolin-1/cholesterol homeostasis.Stem Cell Research&Therapy2015;6(1):238.
61Xu C,Liu H,He Y et al.Endothelial progenitor cells promote osteogenic differentiation in co-cultured with mesenchymal stem cells via the MAPK-dependent pathway.Stem Cell Research&Therapy2020;11(1):537.

Claims (10)

1.牙髓干细胞在制备T细胞活性调节剂或治疗免疫性疾病中的应用,其特征在于,所述牙髓间充质干细胞为使用无血清培养基培养获得的。1. The application of dental pulp stem cells in the preparation of T cell activity regulators or the treatment of immune diseases, wherein the dental pulp mesenchymal stem cells are obtained by culturing in a serum-free medium. 2.如权利要求1所述的应用,其特征在于,所述牙髓间充质干细胞的代数小于或等于第15代;2. The application according to claim 1, wherein the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 15th generation; 优选地,所述牙髓间充质干细胞的代数小于或等于第10代;Preferably, the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 10th generation; 优选地,所述牙髓间充质干细胞的代数小于或等于第5代。Preferably, the passage number of the dental pulp mesenchymal stem cells is less than or equal to the fifth passage. 3.如权利要求1所述的应用,其特征在于,所述牙髓间充质干细胞为DPSC或SHED;3. application as claimed in claim 1, is characterized in that, described dental pulp mesenchymal stem cell is DPSC or SHED; 优选地,所述的T细胞活性调节剂为T细胞活性抑制剂;Preferably, the T cell activity regulator is a T cell activity inhibitor; 优选地,所述T细胞为活化的T细胞;Preferably, the T cells are activated T cells; 优选地,所述免疫性疾病为肠炎。Preferably, the immune disease is enteritis. 4.如权利要求3所述的应用,其特征在于,所述肠炎为急性结肠炎。4. The use according to claim 3, wherein the enteritis is acute colitis. 5.一种制备干细胞的方法,其特征在于,所述干细胞使用的培养基为无血清培养基,所述干细胞为牙髓间充质干细胞,所述方法获得的干细胞用于调节T细胞活性;5. A method for preparing stem cells, wherein the culture medium used by the stem cells is a serum-free medium, the stem cells are dental pulp mesenchymal stem cells, and the stem cells obtained by the method are used to regulate T cell activity; 优选地,所述方法获得的干细胞用于诱导T细胞的凋亡;Preferably, the stem cells obtained by the method are used to induce apoptosis of T cells; 优选地,所述T细胞为活化的T细胞。Preferably, the T cells are activated T cells. 6.如权利要求5所述的方法,其特征在于,所述方法获得的干细胞用于治疗免疫性疾病;6. The method of claim 5, wherein the stem cells obtained by the method are used for the treatment of immune diseases; 优选地,所述方法获得的干细胞用于治疗肠炎;Preferably, the stem cells obtained by the method are used for the treatment of enteritis; 优选地,所述肠炎为急性结肠炎。Preferably, the enteritis is acute colitis. 7.如权利要求5所述的方法,其特征在于,所述牙髓间充质干细胞的代数小于或等于第15代;7. The method of claim 5, wherein the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 15th generation; 优选地,所述牙髓间充质干细胞的代数小于或等于第10代;Preferably, the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 10th generation; 优选地,所述牙髓间充质干细胞的代数为小于或等于第5代;Preferably, the generation of the dental pulp mesenchymal stem cells is less than or equal to the fifth generation; 优选地,所述牙髓间充质干细胞为DPSCs或SHED。Preferably, the dental pulp mesenchymal stem cells are DPSCs or SHEDs. 8.一种T细胞活性调节剂,其特征在于,所述调节剂中含有牙髓间充质干细胞;所述干细胞使用无血清培养基培养获得的;8. A T cell activity regulator, characterized in that the regulator contains dental pulp mesenchymal stem cells; the stem cells are obtained by culturing in a serum-free medium; 优选地,所述牙髓间充质干细胞的代数小于或等于第15代;Preferably, the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 15th generation; 优选地,所述牙髓间充质干细胞的代数小于或等于第10代;Preferably, the generation number of the dental pulp mesenchymal stem cells is less than or equal to the 10th generation; 优选地,所述牙髓间充质干细胞的代数为小于或等于第5代。Preferably, the passage number of the dental pulp mesenchymal stem cells is less than or equal to the fifth passage. 9.如权利要求8所述的T细胞活性调节剂,其特征在于,所述T细胞活性调节剂用于诱导T细胞的凋亡;9. The T cell activity regulator of claim 8, wherein the T cell activity regulator is used to induce apoptosis of T cells; 优选地,所述T细胞为活化的T细胞;Preferably, the T cells are activated T cells; 优选地,所述T细胞活性调节剂用于治疗免疫性疾病;Preferably, the T cell activity modulator is used for the treatment of immune diseases; 优选地,所述T细胞活性调节剂用于治疗肠炎;Preferably, the T cell activity modulator is used to treat enteritis; 优选地,所述肠炎为急性结肠炎。Preferably, the enteritis is acute colitis. 10.权利要求1-4任一所述的应用,或权利要求5-7任一所述的方法,或权利要求8-9任一所述的T细胞活性调节剂,其特征在于,所述的无血清培养基含有人血小板裂解物;10. The use according to any one of claims 1-4, or the method according to any one of claims 5-7, or the T cell activity regulator according to any one of claims 8-9, wherein the The serum-free medium contains human platelet lysate; 优选地,所述无血清培养基还含有DMEM;Preferably, the serum-free medium further contains DMEM; 优选地,所述无血清培养基还含有L-谷氨酰胺或L-谷氨酰胺类似物;Preferably, the serum-free medium further contains L-glutamine or L-glutamine analogs; 优选地,所述L-谷氨酰胺类似物为GlutaMAX添加剂。Preferably, the L-glutamine analog is a GlutaMAX additive.
CN202110557156.XA 2021-05-21 2021-05-21 Preparation method and application of stem cells Pending CN113577107A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110557156.XA CN113577107A (en) 2021-05-21 2021-05-21 Preparation method and application of stem cells

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110557156.XA CN113577107A (en) 2021-05-21 2021-05-21 Preparation method and application of stem cells

Publications (1)

Publication Number Publication Date
CN113577107A true CN113577107A (en) 2021-11-02

Family

ID=78243179

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110557156.XA Pending CN113577107A (en) 2021-05-21 2021-05-21 Preparation method and application of stem cells

Country Status (1)

Country Link
CN (1) CN113577107A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120128636A1 (en) * 2009-01-20 2012-05-24 University Of Southern California Gingiva Derived Stem Cell And Its Application In Immunodulation And Reconstruction
WO2013146992A1 (en) * 2012-03-29 2013-10-03 日本ケミカルリサーチ株式会社 Method for producing pluripotent stem cells derived from dental pulp
CN104694464A (en) * 2015-02-09 2015-06-10 安徽新生命干细胞科技有限公司 Clinical dental pulp stem cell and preparation method thereof
CN107058219A (en) * 2017-04-13 2017-08-18 上海莱馥生命科学技术有限公司 A kind of method that application stem cell self-characteristic prepares dental pulp stem cell
CN112159793A (en) * 2020-09-11 2021-01-01 希诺谷(江苏)生物科技有限公司 Application of human deciduous tooth pulp stem cells

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120128636A1 (en) * 2009-01-20 2012-05-24 University Of Southern California Gingiva Derived Stem Cell And Its Application In Immunodulation And Reconstruction
WO2013146992A1 (en) * 2012-03-29 2013-10-03 日本ケミカルリサーチ株式会社 Method for producing pluripotent stem cells derived from dental pulp
CN104694464A (en) * 2015-02-09 2015-06-10 安徽新生命干细胞科技有限公司 Clinical dental pulp stem cell and preparation method thereof
CN107058219A (en) * 2017-04-13 2017-08-18 上海莱馥生命科学技术有限公司 A kind of method that application stem cell self-characteristic prepares dental pulp stem cell
CN112159793A (en) * 2020-09-11 2021-01-01 希诺谷(江苏)生物科技有限公司 Application of human deciduous tooth pulp stem cells

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
Y. ZHAO等: "Fas ligand regulates the Immunomodulatory Properties of Dental Pulp stem cells"", 《J DENT RES》, vol. 91, no. 10, pages 948 - 954 *
张苗等: ""牙髓干细胞免疫特性及其应用研究进展"", 《中华实用诊断与治疗杂志》, vol. 30, no. 10, pages 937 - 939 *
霍冬梅等: ""人血小板裂解液对牙髓间充质细胞成骨分化的影响"", 《口腔医学研究》, vol. 36, no. 6, pages 539 - 543 *
霍冬梅等: ""人血小板裂解液对牙髓间充质细胞成骨分化的影响"", 《口轻医学研究》, vol. 36, no. 6, pages 539 - 543 *
黄智庆等: ""人牙髓间充质干细胞的规范化鉴定"", 《2020年中华口腔医学会口腔生物医学专业委员会第十次全国口腔生物医学学术年会暨第六次全国口腔杰青优青论坛论文汇编》, 2 November 2020 (2020-11-02), pages 385 - 386 *

Similar Documents

Publication Publication Date Title
Ma et al. CD146 controls the quality of clinical grade mesenchymal stem cells from human dental pulp
Ji et al. Comparison of immunomodulatory properties of exosomes derived from bone marrow mesenchymal stem cells and dental pulp stem cells
Mennan et al. A comprehensive characterisation of large-scale expanded human bone marrow and umbilical cord mesenchymal stem cells
JP7571109B2 (en) Improved Stem Cell Compositions
Mareddy et al. Clonal isolation and characterization of bone marrow stromal cells from patients with osteoarthritis
Alge et al. Donor‐matched comparison of dental pulp stem cells and bone marrow‐derived mesenchymal stem cells in a rat model
Lindroos et al. Serum-free, xeno-free culture media maintain the proliferation rate and multipotentiality of adipose stem cells in vitro
Koerner et al. Equine peripheral blood-derived progenitors in comparison to bone marrow-derived mesenchymal stem cells
Meligy et al. The efficiency of in vitro isolation and myogenic differentiation of MSCs derived from adipose connective tissue, bone marrow, and skeletal muscle tissue
Hsu et al. Isolation of the multipotent MSC subpopulation from human gingival fibroblasts by culturing on chitosan membranes
JP6348848B2 (en) Proliferation of mesenchymal stem cells
Dissanayaka et al. Characterization of dental pulp stem cells isolated from canine premolars
Konala et al. Secretome studies of mesenchymal stromal cells (MSCs) isolated from three tissue sources reveal subtle differences in potency
US10736922B2 (en) High telomerase activity bone marrow mesenchymal stem cells, methods of producing the same and pharmaceuticals and treatment methods based thereon
JP5155855B2 (en) Isolation of multipotent stem cells
Huo et al. Differentiation of dermal multipotent cells into odontogenic lineage induced by embryonic and neonatal tooth germ cell–conditioned medium
Xiao et al. The establishment of a chemically defined serum-free culture system for human dental pulp stem cells
Zheng et al. Effects of sEV derived from SHED and DPSC on the proliferation, migration and osteogenesis of PDLSC
CN113462765B (en) Method and kit for identifying stem cells
KR20110095218A (en) CD4, which promotes proliferation, pluripotency, and reprogramming of adult stem cells through the PI3 / APT / KS3 pathway
WO2016029267A1 (en) A method for culturing mesenchymal stem cells
KR100808546B1 (en) Method of obtaining mesenchymal stem cells by sonication
Ma et al. Identification and characterization of pulmonary mesenchymal stem cells derived from rat fetal lung tissue
Feter et al. Dental mesenchymal stem cell: Its role in tooth development, types, surface antigens and differentiation potential
CN113577107A (en) Preparation method and application of stem cells

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right
TA01 Transfer of patent application right

Effective date of registration: 20240425

Address after: 510275 No. 135 West Xingang Road, Guangzhou, Guangdong, Haizhuqu District

Applicant after: SUN YAT-SEN University

Country or region after: China

Address before: 501, Floor 5, Building 1, No. 6, Nanjiang Second Road, the Pearl River Street, Nansha District, Guangzhou, Guangdong 510000

Applicant before: Shi Songtao

Country or region before: U.S.A.