[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (797)

Search Parameters:
Keywords = myofibroblast

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
37 pages, 4373 KiB  
Review
The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair?
by Fereshteh Sadat Younesi and Boris Hinz
Int. J. Mol. Sci. 2024, 25(16), 8712; https://doi.org/10.3390/ijms25168712 - 9 Aug 2024
Viewed by 295
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is [...] Read more.
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring—called fibrosis—that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies. Full article
Show Figures

Figure 1

Figure 1
<p>Tissue sources of therapeutic MSCs and stiffness-dependent differentiation. (<b>A</b>) The most prominently used tissue sources to isolate therapeutic MSCs from human biopsies include adipose tissues, bone marrow, and umbilical cord Wharton’s jelly. (<b>B</b>) The mechanical properties, i.e., softness or stiffness of tissues and those of the culture substrates used to grow and expand adhesive MSCs can influence MSC differentiation capacity and fate. MSCs cultured in soft cell culture environments matched to the elastic modulus (indicated in kPa) of normal fat and muscle tissue exhibit a high propensity for adipogenic and myogenic differentiation. In contrast, growth on stiffer culture substrates promotes the lineage commitment of MSCs towards cartilage and bone. One MSC fate, either representing a transitional state to osteogenesis or an independent scar-forming phenotype, is the activation of MSCs into fibrogenic myofibroblasts. Notably, the scars forming in response to the injury of soft tissues are always stiffer than the normal tissue texture (here schematized for skin), which drives the mechanically induced myofibroblast activation from resident and delivered mesenchymal cells. Cell culture plastic dishes are even stiffer (~10,000-times) than the stiff scar, which results in MSC-to-myofibroblast activation in vitro. Scheme produced with Biorender.</p>
Full article ">Figure 2
<p>MSC mechanoperception and nuclear mechanics. (<b>A</b>) The spreading area of mesenchymal stromal cells (MSCs) attaching to an adhesive substrate can be controlled using micropatterning; for instance, by transferring fibronectin protein (blue staining) in square shapes of different areas onto glass or plastic substrates using polydimethylsiloxane (PDMS) stamps. (<b>B</b>) Restricting MSC spreading limits the number and size of focal adhesions (green vinculin staining) and F-actin stress fibers (phalloidin, red), thus overall reducing MSC stress. (<b>C</b>) Another way to reduce stress on MSCs in culture is manipulating the elastic modulus of their substrate. MSCs perceive mechanical cues from the extracellular matrix (ECM) via transmembrane integrins; binding to extracellular ligands and intracellular F-actin shifts integrins from a low affinity inactive to a high-affinity active configuration. This integrin conformational switch prompts the assembly of complex focal adhesion structures comprising the cytosolic proteins talin, vinculin, focal adhesion kinase, paxillin, and filamin. Focal adhesions serve as hubs for mechanotransduction pathways, orchestrating the polymerization of G- into F-actin and the organization of vimentin monomers into intermediate filaments. Mechanical stress also opens stretch-activated channels (SACs) to allow the influx of Ca<sup>2+</sup> into the cytosol to trigger distinct signaling cascades. (<b>D</b>) The nuclei of MSCs grown on stiff surfaces are characterized by higher lamin A:C ratios in the inner nuclear membrane, more decondensed chromatin and higher histone acetylation compared to soft environments. A direct connection between ECM adhesions and the nucleus is established through the nucleoskeleton and cytoskeleton complex (LINC), containing nuclear envelope spectrin repeat proteins (nesprins) and Sad1p and UNC-84 homology (SUN) proteins that span the nuclear envelope. Nesprin-3 attaches SUN proteins to F-actin, whereas nesprins-1 and -2 link to intermediate filaments. Within the inner nuclear membrane, SUN dimers interact with lamin A bound to chromatin, causing organized DNA to unfold under high mechanical stress. High stress enhances the nuclear translocation of mechanosensitive transcription factors, such as MRTF-A, Runt-related transcription factor 2 (RUNX2), Yes-associated protein (YAP), and transcriptional coactivator with PDZ-binding motif (TAZ) via opening of the nuclear pore complex (NPC). The promoter binding of these transcription factors drives the expression of pro-fibrotic and osteogenic genes. Scheme elements produced using Biorender, immunofluorescence images produced by Nicole Berezyuk (Hinzlab).</p>
Full article ">Figure 3
<p>In vitro systems and mechanisms to generate mechanical memory in MSCs. (<b>A</b>) Seminal studies generated ‘long-term’ mechanical memory of lung fibroblasts [<a href="#B310-ijms-25-08712" class="html-bibr">310</a>] and MSC(M) [<a href="#B125-ijms-25-08712" class="html-bibr">125</a>] by culturing and adapting (‘priming’) cells for up to 3 weeks on either soft or stiff silicone elastomer substrates. Mechanical memory was defined as the capacity of MSCs to retain regenerative (soft) or pro-fibrotic and/or pro-osteogenic (stiff) features after switching to the respective substrate for another 2 weeks. (<b>B</b>) In the same study, growth on stiff culture substrates was shown to induce nuclear translocation of the mechanosensitive co-transcription factor myocardin-related transcription factor A (MRTF-A), where it drives the transcription of the profibrotic microRNA miR-21 [<a href="#B125-ijms-25-08712" class="html-bibr">125</a>]. Cytoplasmic miR-21 levels remain elevated for up to 2 weeks even after switching to soft substrates, whereas MRTF-A relocates to the cytosol within minutes. (<b>C</b>) In a different experimental approach to generate ‘short-term’ mechanical memory, MSCs and fibroblasts were cultured on stiff phototunable hydrogels for 10 d to acquire high levels of histone acetylation and low condensed chromatin [<a href="#B301-ijms-25-08712" class="html-bibr">301</a>,<a href="#B303-ijms-25-08712" class="html-bibr">303</a>]. Following in situ softening of the hydrogels using a light reaction, MSCs maintained high histone acetylation levels while showing increased chromatin condensation. The preserved histone acetylation can regulate chromatin accessibility and transcription profiles.</p>
Full article ">Figure 4
<p>Therapeutic effects of mechanically primed MSCs on rat wound healing. Skin wound healing was the first preclinical example to show a differential effect of soft- versus stiff-primed MSCs on tissue repair after transplantation [<a href="#B125-ijms-25-08712" class="html-bibr">125</a>]. Rat bone-marrow-derived MSCs (MSC(M)), primed for 3 weeks on either soft (5 kPa) or stiff (100 kPa) silicone culture substrates, were applied in a fibrin matrix to rat skin wounds, kept open, and made hypertrophic by a plastic frame splint. Shown are immunofluorescence images of 9-day-old wound tissue cross-sections. In this experimental model, soft primed MSC(M) suppress scar features such as enhanced wound tension which is not shown in the figure but in the published work [<a href="#B125-ijms-25-08712" class="html-bibr">125</a>], myofibroblast accumulation (red only, α-SMA), high vascularization (yellow, from co-staining of vascular smooth muscle for desmin, green, and α-SMA, red), and alignment of dense collagen extracellular matrix (only shown in the schematic). All these features are enhanced after delivery of stiff-primed MSC and even further accentuated in wounds that did not receive any MSCs.</p>
Full article ">
19 pages, 2347 KiB  
Review
Noncoding RNA-Mediated Epigenetic Regulation in Hepatic Stellate Cells of Liver Fibrosis
by Ruoyu Gao and Jingwei Mao
Non-Coding RNA 2024, 10(4), 44; https://doi.org/10.3390/ncrna10040044 - 7 Aug 2024
Viewed by 374
Abstract
Liver fibrosis is a significant contributor to liver-related disease mortality on a global scale. Despite this, there remains a dearth of effective therapeutic interventions capable of reversing this condition. Consequently, it is imperative that we gain a comprehensive understanding of the underlying mechanisms [...] Read more.
Liver fibrosis is a significant contributor to liver-related disease mortality on a global scale. Despite this, there remains a dearth of effective therapeutic interventions capable of reversing this condition. Consequently, it is imperative that we gain a comprehensive understanding of the underlying mechanisms driving liver fibrosis. In this regard, the activation of hepatic stellate cells (HSCs) is recognized as a pivotal factor in the development and progression of liver fibrosis. The role of noncoding RNAs (ncRNAs) in epigenetic regulation of HSCs transdifferentiation into myofibroblasts has been established, providing new insights into gene expression changes during HSCs activation. NcRNAs play a crucial role in mediating the epigenetics of HSCs, serving as novel regulators in the pathogenesis of liver fibrosis. As research on epigenetics expands, the connection between ncRNAs involved in HSCs activation and epigenetic mechanisms becomes more evident. These changes in gene regulation have attracted considerable attention from researchers in the field. Furthermore, epigenetics has contributed valuable insights to drug discovery and the identification of therapeutic targets for individuals suffering from liver fibrosis and cirrhosis. As such, this review offers a thorough discussion on the role of ncRNAs in the HSCs activation of liver fibrosis. Full article
(This article belongs to the Collection Feature Papers in Non-Coding RNA)
Show Figures

Figure 1

Figure 1
<p>The biological pathways and interactions of three ncRNAs. MiRNAs are produced from pri-miRNAs transcribed by RNA polymerase II from independent genes or introns of protein genes. The pri-miRNAs are processed by a complex involving Drosha and DiGeorge syndrome critical region 8 (DGCR8) to create pre-miRNAs, which are then exported to the cytoplasm by Exportin 5. In the cytoplasm, Dicer cleaves the pre-miRNA to form a miRNA duplex, with one strand being degraded and the other becoming the mature miRNA. The mature miRNA, along with Argonaute2 (AGO2) and glycine-tryptophan repeat-containing protein of 182 KDa (GW182), binds to the 3′-untranslated region (3′-UTR) of targeted mRNA to inhibit its translation. LncRNAs are transcribed by RNA polymerase II and have a structure similar to mRNA. They can be transported from the nucleus to the cytoplasm by nuclear RNA export factor 1 (NXF1). Some lncRNAs act as competitive endogenous RNAs (ceRNAs), binding to miRNAs to affect gene expression. LncRNAs can also interact with RBPs to stabilize and promote mRNA translation. CircRNAs are produced from pre-mRNA through backsplicing and can regulate gene expression by acting as ceRNAs to inhibit miRNA and increase expression of targeted genes.</p>
Full article ">Figure 2
<p>TGF-β1/Smad pathway mediated by identified ncRNAs. The TGF-β signaling pathway involves three isoforms, namely TGF-β1, TGF-β2, and TGF-β3. This discussion will focus specifically on the mechanism of TGF-β1. The precursor form of TGF-β contains a latency-associated peptide (LAP), which can be cleaved to allow binding to the mature TGF-β homodimer. This complex, along with the latent TGF-β-binding protein (LTBP), forms the latent TGF-β/LAP/LTBP complex, maintaining TGF-β in an inactive state unable to interact with TGF-βR I and TGF-βR II. Upon release of the TGF-β homodimer from the complex, it becomes active and can interact with TGF receptor II (TGFR II), leading to TGFR I activation and subsequent phosphorylation of Smad2 and Smad3. The phosphorylated forms of Smad2 and Smad3 form a complex with Smad4, translocate to the nucleus, and bind to the promoter regions of fibrotic genes, thereby inducing their transcription. Additionally, Smad7, acting as an inhibitory Smad, competitively interacts with TGFR I to inhibit the binding between TGFR I and Smad2/3, thereby suppressing the activation of the TGF-β1/Smad pathway. Furthermore, Smad7 recruits E3 ubiquitin ligase and phosphatases to facilitate the degradation and dephosphorylation of Smad2/3. Left–right determination factor 2 (LEFTY2) is a member of the TGF-β protein superfamily, which can inhibit TGF-β1/Smad3 signaling. Follistatin-like 1 (FSTL1) is an inducer of TGF-β1/Smad3 signaling through promoting the Smad3 phosphorylation. Some identified ncRNAs can also regulate the TGF-β/Smad pathway by targeting various components of this signaling cascade.</p>
Full article ">Figure 3
<p>Hippo and Hedgehog pathways mediated by identified ncRNAs. The Hippo pathway is activated by TAO kinase, which subsequently phosphorylates mammalian Ste20-like kinase 1/2 (MST1/2). Phosphorylated MST1/2 then phosphorylates scaffold proteins SAV1 and MOB1A/B, facilitating the recruitment and phosphorylation of the large tumor suppressor 1/2 (LATS1/2). Phosphorylated LATS1/2 facilitates the phosphorylation of transcriptional co-activators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ), leading to their cytoplasmic retention mediated by 14-3-3 and subsequent degradation. In the absence of Hippo pathway activation, the kinase cascades are not initiated, allowing YAP/TAZ co-activators to translocate into the nucleus and interact with the TEAD transcription factor family to modulate gene expression. In the absence of Hedgehog ligands, the twelve-pass transmembrane receptor Ptched (Ptch) is able to inhibit the activity of the seven-pass transmembrane receptor Smoothene (Smo). This inhibition of Smo leads to the inactivation of glioma-associated oncogene transcription factors (Glis) through the formation of a complex with suppressor of Fused (SuFu) and Kif7. This complex then promotes the phosphorylation of Glis by protein kinase (PKA), casein kinaseⅠɑ (CK1ɑ), and glycogen synthase kinase-3β (GSK3β), resulting in the repression of subsequent transcription. When the Hh protein binds to the Ptch receptor, it leads to a reduction in the inhibition of Smo, allowing for the phosphorylation of Smo by CK1 family kinase and GRK2. This phosphorylation event induces the release of Gli from the complex, enabling its translocation into the nucleus to regulate the expression of targeted genes. Additionally, the regulation of these pathways is influenced by ncRNAs, as depicted in this figure.</p>
Full article ">Figure 4
<p>DNA methylation usually occurs in the region enrichment with cytosine–phosphate–guanine (CpG) dinucleotides that are also called CpG islands. DNMTs catalyze the methyl group transference from S-adenyl methionine (SAM) to the fifth carbon of cytosine residue to form 5-methylcytosine (5mc). DNMT3a and DNMT3b catalyze the de novo methylation, namely, add the 5mc to the DNA directly. DNMT1 plays a role in maintaining methylation in DNA replication. A family of TET enzymes can superinduce a hydroxyl group to the 5mc; this process converts the 5mc to 5-hydroxymethylcytosine (5hmc). MecP2 can bind with methylated CpG to suppress or enhance gene expression. The regulation of these processes is influenced by ncRNAs, as depicted in this figure.</p>
Full article ">Figure 5
<p>Wnt/β-catenin pathway and associated ncRNAs. When the Wnt signaling pathway is not effectively inhibited by Dickkopf (DKK) or Wnt inhibitory protein (WIF), which interact with lipoprotein receptor-related 5/6 (LRP5/6) to disrupt LRP5/6 and Frizzled (FZD) receptor dimers, Wnt proteins are able to bind to their FZD receptors, leading to dimerization of FZD and LRP5/6 receptors. The formation of FZD/LRP heterodimers induces a conformational change in the receptors, leading to the binding of the cytoplasmic portion of FZD to disheveled (DVL) and phosphorylation of the LRP5/6 tail by GSK3. This phosphorylation event facilitates the recruitment of the scaffold protein Axin. DVL serves as a platform for enhanced interaction between Axin and the LRP5/6 tail. The interaction between Axin, LRP5/6, and DVL disrupts the Destructive Complex (DC), releasing β-catenin. Subsequently, β-catenin translocates into the nucleus, where it displaces corepressor Groucho/transducin with TCF/LEF to form a complex that mediates gene expression. The destruction complex (DC), composed of Axin, adenomatous polyposis (APC), glycogen synthase kinase 3β (GSK3β), and casein kinase 1ɑ (CK1ɑ), functions to maintain β-catenin in an inactive state through phosphorylation by CK1ɑ, facilitating GSK3β-mediated phosphorylation of β-catenin. Subsequent phosphorylation of β-catenin leads to recruitment of β-transducin repeat containing protein (β-Trcp), an E3 ubiquitin ligase that promotes ubiquitination of β-catenin for degradation. The Wnt/β-catenin pathway is also regulated by various ncRNAs.</p>
Full article ">
17 pages, 9371 KiB  
Article
Commercialization of the Xalkori Pediatric Multiparticulate Product Using Quality-by-Design Principles
by Jeremy Bartlett, Natalie Culver, Xiang Zhang, Brett Waybrant, Hannah Sullivan and Logan Howell
Pharmaceutics 2024, 16(8), 1027; https://doi.org/10.3390/pharmaceutics16081027 - 1 Aug 2024
Viewed by 459
Abstract
A pediatric dosage form for crizotinib (Xalkori) was commercialized using quality-by-design principles in a material-sparing fashion. The dosage form consists of spherical multiparticulates (microspheres or pellets) that are coated and encapsulated in capsules for opening. The crizotinib (Xalkori)-coated pellet product is approved in [...] Read more.
A pediatric dosage form for crizotinib (Xalkori) was commercialized using quality-by-design principles in a material-sparing fashion. The dosage form consists of spherical multiparticulates (microspheres or pellets) that are coated and encapsulated in capsules for opening. The crizotinib (Xalkori)-coated pellet product is approved in the US for pediatric patients 1 year of age and older and young adults with relapsed or refractory, systemic anaplastic large cell lymphoma (ALCL) and unresectable, recurrent, or refractory inflammatory myofibroblastic tumor (IMT) that is ALK-positive. The product is also approved in the US for adult patients with non-small cell lung cancer (NSCLC) who are unable to swallow intact capsules. The lipid multiparticulate is composed of a lipid matrix, a dissolution enhancer, and an active pharmaceutical ingredient (API). The API, which remains crystalline, is embedded within the microsphere at a 60% drug loading in the uncoated lipid multiparticulate to enable dose flexibility. The melt spray congealing technique using a rotary atomizer is used to manufacture the lipid multiparticulate. Following melt spray congealing, a barrier coating is applied via fluid bed coating. Due to their particle size and content uniformity, this dosage form provides the dosing flexibility and swallowability needed for the pediatric population. The required pediatric dose is achieved by opening the capsules and combining doses of different encapsulated dose strengths, followed by administration of the multiparticulates directly to the mouth. The encapsulation process was optimized through equipment modifications and by using a design of experiments approach to understand the operating space. A limited number of development batches produced using commercial-scale equipment were leveraged to design, understand, and verify the manufacturing process space. The quality by design and material-sparing approach taken to design the melt spray congeal and encapsulation manufacturing processes resulted in a pediatric product with exceptional content uniformity (a 95% confidence and 99% probability of passing USP <905> content uniformity testing for future batches). Full article
(This article belongs to the Special Issue Advanced Pediatric Drug Formulation Strategies)
Show Figures

Figure 1

Figure 1
<p>Process flow diagram of the melt spray congeal process. The red designates the heated path the melt follows from the extruder to the atomization disk. The cartoon depicts the droplets atomizing from the disk and cooling to the point of congealing, indicated by the color changing from yellow to blue, prior to impacting the collection bag.</p>
Full article ">Figure 2
<p>High-speed video footage of the crizotinib melt being dispensed onto the spinning disk and atomized on the rotary atomizer.</p>
Full article ">Figure 3
<p>Representative particle morphology for crizotinib lipid multiparticulates produced via MSC.</p>
Full article ">Figure 4
<p>Particle size (D[v,0.5] (µm)) (left) as a function of the two statistically significant process parameters. Potency (right) as a function of the one statistically significant process parameter (extruder screw speed). The black boxes in both figures denote the narrowed operating space to ensure consistent microsphere particle size. Note: <a href="#app1-pharmaceutics-16-01027" class="html-app">Supplement Table S1</a> reports the study design.</p>
Full article ">Figure 5
<p>Potency across three registrational stability batches of uncoated microspheres as a function of process time.</p>
Full article ">Figure 6
<p>Particle size distribution by laser diffraction for registrational stability lots of uncoated microspheres produced via melt spray congealing prior to screening.</p>
Full article ">Figure 7
<p>Mechanical scraper compared to air knife methods of removing the material cap.</p>
Full article ">Figure 8
<p>Left: Filled capsule weight RSD from the in-process control sampling for the (a) ICH campaign (mechanical scraper used) compared to (b) PPQ campaign (air knife used). Data are summarized for three batches of each campaign. Right: Capsule fill contents from extended uniformity of dosage units for the (a) ICH campaign (mechanical scraper used) compared to (b) PPQ campaign (air knife used).</p>
Full article ">Figure 9
<p>Volumetric dosing with a vacuum dosator. Process parameters are labeled and described in the text.</p>
Full article ">Figure 10
<p>Contour plots for filled capsule weight RSD (%) (shown as values on the contour lines; color spectrum from blue (0%) to red (2.5%) for the RSD values is shown at the bottom of the figure) for the statistically significant two-way interaction effects of (1) AC: air knife pressure and fill vacuum (left) and (2) AB: air knife pressure and infeed level (right) based on DoE Study 2. The stars denote additional process exploration, as explained below in <a href="#sec4dot3-pharmaceutics-16-01027" class="html-sec">Section 4.3</a>. Note: DoE Study 2 design and the results fitting and analysis are reported in <a href="#app1-pharmaceutics-16-01027" class="html-app">Supplement Tables S3 and S4</a>.</p>
Full article ">Figure 11
<p>Extended uniformity of dosage units across the demonstration batch among three different processing conditions.</p>
Full article ">Figure 12
<p>Capsule fill content uniformity for the ICH and PPQ campaigns, demonstrating the tight correlation between the fill content mass (X) and capsule assay (Y). Note that less than 2% of the capsules were less than 85 %LC and would be removed from the batch in the weight sorting process. Blue solid line: Y = 1.234 + 0.9921 × X (R<sup>2</sup> = 0.943); Black dashed line: Y = X for reference.</p>
Full article ">
18 pages, 2892 KiB  
Article
Pericardial Fluid Accumulates microRNAs That Regulate Heart Fibrosis after Myocardial Infarction
by Elsa D. Silva, Daniel Pereira-Sousa, Francisco Ribeiro-Costa, Rui Cerqueira, Francisco J. Enguita, Rita N. Gomes, João Dias-Ferreira, Cassilda Pereira, Ana Castanheira, Perpétua Pinto-do-Ó, Adelino F. Leite-Moreira and Diana S. Nascimento
Int. J. Mol. Sci. 2024, 25(15), 8329; https://doi.org/10.3390/ijms25158329 - 30 Jul 2024
Viewed by 526
Abstract
Pericardial fluid (PF) has been suggested as a reservoir of molecular targets that can be modulated for efficient repair after myocardial infarction (MI). Here, we set out to address the content of this biofluid after MI, namely in terms of microRNAs (miRs) that [...] Read more.
Pericardial fluid (PF) has been suggested as a reservoir of molecular targets that can be modulated for efficient repair after myocardial infarction (MI). Here, we set out to address the content of this biofluid after MI, namely in terms of microRNAs (miRs) that are important modulators of the cardiac pathological response. PF was collected during coronary artery bypass grafting (CABG) from two MI cohorts, patients with non-ST-segment elevation MI (NSTEMI) and patients with ST-segment elevation MI (STEMI), and a control group composed of patients with stable angina and without previous history of MI. The PF miR content was analyzed by small RNA sequencing, and its biological effect was assessed on human cardiac fibroblasts. PF accumulates fibrotic and inflammatory molecules in STEMI patients, namely causing the soluble suppression of tumorigenicity 2 (ST-2), which inversely correlates with the left ventricle ejection fraction. Although the PF of the three patient groups induce similar levels of fibroblast-to-myofibroblast activation in vitro, RNA sequencing revealed that PF from STEMI patients is particularly enriched not only in pro-fibrotic miRs but also anti-fibrotic miRs. Among those, miR-22-3p was herein found to inhibit TGF-β-induced human cardiac fibroblast activation in vitro. PF constitutes an attractive source for screening diagnostic/prognostic miRs and for unveiling novel therapeutic targets in cardiac fibrosis. Full article
(This article belongs to the Special Issue Cardiovascular Diseases: Molecular Mechanisms and Potential Therapy)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the study design. Pericardial fluid (PF) was collected from coronary patients undergoing CABG and subjected to an in vitro function assay to evaluate cardiac fibroblast activation and conduct a quantification of fibrosis-associated biomarkers and small RNA sequencing. The selected targets were validated and tested in vitro with respect to hCF activation.</p>
Full article ">Figure 2
<p>Pericardial fluid is a reservoir of fibrosis-related molecules and induces cardiac fibroblast activation. (<b>A</b>,<b>B</b>) Quantification of procollagen I C-terminal propeptide (PICP) and serum stimulation-2 (ST-2) in the pericardial fluid and plasma from the patient cohort (<span class="html-italic">n</span> ≥ 5/group). (<b>C</b>) Spearman correlation between left ventricle ejection fraction (LVEF) and ST-2 levels in the PF of STEMI patients (<span class="html-italic">n</span> = 11/group). (<b>D</b>) Schematic representation of the experimental design. (<b>E</b>) Expression of ACTA2, COL1A1, and CCN2 in hCFs cultured in 2% pericardial fluid (<span class="html-italic">n</span> ≥ 7/group). (<b>F</b>,<b>G</b>) Representative images and respective quantification of the alpha-smooth muscle actin (α-SMA) and collagen type I (COL I) of hCFs cultured in 2% pericardial fluid.</p>
Full article ">Figure 3
<p>Small RNA sequencing of PF collected from CTRL, NSTEMI, and STEMI patients undergoing CABG. miRNAs identified in all PF samples by small RNA sequencing (<span class="html-italic">n</span> = 8, CTRL; <span class="html-italic">n</span> = 9, NSTEMI; <span class="html-italic">n</span> = 5, STEMI). (<b>A</b>) Enrichment analysis (miRNet 2.0) of miRNA–function and miRNA–disease (based on TAM 2.0) of the 275 miRs present in all groups. (<b>B</b>) Venn diagram with the comparison between our miRs list, with miRs reported by Kuosmanen et al. [<a href="#B22-ijms-25-08329" class="html-bibr">22</a>]. (<b>C</b>) Venn diagram with the number and intersection of miRs with statistically different abundance in NSTEMI vs. CTRL, STEMI vs. CTRL, and STEMI vs. NSTEMI. (<b>D</b>) Enrichment analysis of the KEGG pathways of miR gene targets regulated by altered miRNAs in STEMI patients when compared with CTRL. In red, the KEGG pathways relevant for cardiac fibrosis. (<b>E</b>) Enrichment analysis (miRNet 2.0) of miR function associated with altered miRs in STEMI patients when compared with CTRL (based on TAM 2.0). (<b>F</b>) Heatmap of altered miRNAs in STEMI patients when compared with CTRL.</p>
Full article ">Figure 4
<p>miR-22-3p overexpression inhibits hCF activation in vitro. (<b>A</b>) qRT-PCR validation of the RNAseq results for mir-22-3p and miR-203-3p. Expression is presented through the 2-ΔCt method, with normalization of the expression of miR-99-p and miR-39-5p (<span class="html-italic">n</span> ≥ 5/group). (<b>B</b>) Schematic representation of the experimental design. (<b>C</b>) Expression of miR-22-3p in hCFs after transfection with 100 or 200nM of miR-22-3p or scramble miR precursor. (<b>D</b>) Expression of miR-22-3p in hCFs at 2, 6, and 8 days after transfection with miR-22-3p or scramble miR precursor. (<b>E</b>) Expression of ACTA2 and COL1A1 in hCFs at different time points after transfection (<span class="html-italic">n</span> = 2/group). (<b>F</b>) Representative figures of α-SMA and COL I in transfected hCFs following a 7-day activation assay. (<b>G</b>) Percentage of α-SMA + cells (<span class="html-italic">n</span> = 4/group) and area of COL I per cell (<span class="html-italic">n</span> = 2/group).</p>
Full article ">Figure 5
<p>A model for the role of miR-22-3p in cardiac fibrosis after MI. Several miRs previously shown to regulate pro-fibrotic or anti-fibrotic mechanisms were released, and they accumulate in the PF after MI. From those, miR-22-3p was shown to mitigate TGF-β-induced cardiac fibroblast activation in vitro, which may counterbalance the pro-fibrotic environment formed after MI in vivo.</p>
Full article ">
10 pages, 1919 KiB  
Article
Role of ATG4 Autophagy-Related Protein Family in the Lower Airways of Patients with Stable COPD
by Francesco Nucera, Antonino Di Stefano, Fabio Luigi Massimo Ricciardolo, Isabella Gnemmi, Cristina Pizzimenti, Francesco Monaco, Giovanni Tuccari, Gaetano Caramori and Antonio Ieni
Int. J. Mol. Sci. 2024, 25(15), 8182; https://doi.org/10.3390/ijms25158182 - 26 Jul 2024
Viewed by 404
Abstract
Autophagy is a complex physiological pathway mediating homeostasis and survival of cells degrading damaged organelles and regulating their recycling. Physiologic autophagy can maintain normal lung function, decrease lung cellular senescence, and inhibit myofibroblast differentiation. It is well known that autophagy is activated in [...] Read more.
Autophagy is a complex physiological pathway mediating homeostasis and survival of cells degrading damaged organelles and regulating their recycling. Physiologic autophagy can maintain normal lung function, decrease lung cellular senescence, and inhibit myofibroblast differentiation. It is well known that autophagy is activated in several chronic inflammatory diseases; however, its role in the pathogenesis of chronic obstructive pulmonary disease (COPD) and the expression of autophagy-related genes (ATGs) in lower airways of COPD patients is still controversial. The expression and localization of all ATG proteins that represented key components of the autophagic machinery modulating elongation, closure, and maturation of autophagosome membranes were retrospectively measured in peripheral lungs of patients with stable COPD (n = 10), control smokers with normal lung function (n = 10), and control nonsmoking subjects (n = 8) using immunohistochemical analysis. These results show an increased expression of ATG4 protein in alveolar septa and bronchiolar epithelium of stable COPD patients compared to smokers with normal lung function and non-smoker subjects. In particular, the genes in the ATG4 protein family (including ATG4A, ATG4B, ATG4C, and ATG4D) that have a key role in the modulation of the physiological autophagic machinery are the most important ATGs increased in the compartment of lower airways of stable COPD patients, suggesting that the alteration shown in COPD patients can be also correlated to impaired modulation of autophagic machinery modulating elongation, closure, and maturation of autophagosomes membranes. Statistical analysis was performed by the Kruskal–Wallis test and the Mann–Whitney U test for comparison between groups. A statistically significant increased expression of ATG4A (p = 0.0047), ATG4D (p = 0.018), and ATG5 (p = 0.019) was documented in the bronchiolar epithelium as well in alveolar lining for ATG4A (p = 0.0036), ATG4B (p = 0.0054), ATG4C (p = 0.0064), ATG4D (p = 0.0084), ATG5 (p = 0.0088), and ATG7 (p = 0.018) in patients with stable COPD compared to control groups. The ATG4 isoforms may be considered as additional potential targets for the development of new drugs in COPD. Full article
(This article belongs to the Special Issue The Role of Autophagy in Disease and Cancer)
Show Figures

Figure 1

Figure 1
<p>Immunoreactivity of ATGs in bronchial epithelium: a diffuse strong nuclear and cytoplasmic (score 6) staining was encountered with ATG4A (<b>A</b>, 400×), ATG4D (<b>B</b>, 400×), ATG5 (<b>C</b>, 400×) in patients with stable COPD; note the absence of immunoreaction in CNS/CS patients (<b>D</b>, 200×). Nuclear hemalum counterstain was made in all cases.</p>
Full article ">Figure 2
<p>Immunoreactivity of ATGs in alveolar lining: a diffuse strong nuclear and cytoplasmic (score 5–6) staining was encountered with ATG4A (<b>A</b>, 400×), ATG4B (<b>B</b>, 400×), ATG4C (<b>C</b>, 400×) ATG4D (<b>D</b>, 400×), ATG5 (<b>E</b>, 400×) and ATG7 (<b>F</b>, 400×) in patients with stable COPD, Note also the immunopositivity in alveolar macrophages. Nuclear hemalum counterstain was made in all cases.</p>
Full article ">
33 pages, 2382 KiB  
Review
Liver Fibrosis: From Basic Science towards Clinical Progress, Focusing on the Central Role of Hepatic Stellate Cells
by Hikmet Akkız, Robert K. Gieseler and Ali Canbay
Int. J. Mol. Sci. 2024, 25(14), 7873; https://doi.org/10.3390/ijms25147873 - 18 Jul 2024
Viewed by 738
Abstract
The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by [...] Read more.
The burden of chronic liver disease is globally increasing at an alarming rate. Chronic liver injury leads to liver inflammation and fibrosis (LF) as critical determinants of long-term outcomes such as cirrhosis, liver cancer, and mortality. LF is a wound-healing process characterized by excessive deposition of extracellular matrix (ECM) proteins due to the activation of hepatic stellate cells (HSCs). In the healthy liver, quiescent HSCs metabolize and store retinoids. Upon fibrogenic activation, quiescent HSCs transdifferentiate into myofibroblasts; lose their vitamin A; upregulate α-smooth muscle actin; and produce proinflammatory soluble mediators, collagens, and inhibitors of ECM degradation. Activated HSCs are the main effector cells during hepatic fibrogenesis. In addition, the accumulation and activation of profibrogenic macrophages in response to hepatocyte death play a critical role in the initiation of HSC activation and survival. The main source of myofibroblasts is resident HSCs. Activated HSCs migrate to the site of active fibrogenesis to initiate the formation of a fibrous scar. Single-cell technologies revealed that quiescent HSCs are highly homogenous, while activated HSCs/myofibroblasts are much more heterogeneous. The complex process of inflammation results from the response of various hepatic cells to hepatocellular death and inflammatory signals related to intrahepatic injury pathways or extrahepatic mediators. Inflammatory processes modulate fibrogenesis by activating HSCs and, in turn, drive immune mechanisms via cytokines and chemokines. Increasing evidence also suggests that cellular stress responses contribute to fibrogenesis. Recent data demonstrated that LF can revert even at advanced stages of cirrhosis if the underlying cause is eliminated, which inhibits the inflammatory and profibrogenic cells. However, despite numerous clinical studies on plausible drug candidates, an approved antifibrotic therapy still remains elusive. This state-of-the-art review presents cellular and molecular mechanisms involved in hepatic fibrogenesis and its resolution, as well as comprehensively discusses the drivers linking liver injury to chronic liver inflammation and LF. Full article
(This article belongs to the Special Issue Molecular Advances in Liver Fibrosis)
Show Figures

Figure 1

Figure 1
<p>Overview of the cellular and molecular mechanisms of liver fibrogenesis. During chronic liver injury, hepatocytes activate signaling via Janus kinase (JNK), Notch, osteopontin, and hedgehog and produce exosomes harboring microRNAs (miRNAs) to initiate HSC activation. Inflammation triggers KCs and recruits monocyte-derived macrophages through C-C motif chemokine receptor (CCR)9 and C-C motif chemokine ligand (CCL)2, CCl<sub>4</sub>, and CCL25. The crosstalk between C-X3-C motif chemokine ligand 1 (CX3CL1) and C-X3-C motif chemokine receptor 1 (CX3CR1) orchestrates macrophage survival, differentiation, and polarization. KCs trigger the HSC activation by TGF-β, PDGF, and IL-1-β. Activated HSCs produce ECM proteins and secrete inflammatory chemokines CCL2, CCL3, and CX3CL1, whereby accumulating proinflammatory monocytes. HSC-originated matrix metalloproteinase (MMP) and tissue inhibitor of metalloproteinase (TIMP) contribute to ECM perpetuation, remodeling, and fibrosis. Activated HSCs lead to portal hypertension by enhancing the hepatic sinusoids’ contractility. Some molecules and pathways, including endothelin 1, TGF-β, Jak2, and the Wnt/β/catenin pathway, affect sinusoidal contractility.</p>
Full article ">Figure 2
<p>Molecular pathways and cellular interactions involved in HSC activation and deactivation. Activated HSCs are the main effector cells during hepatic fibrosis. In the healthy liver, they metabolize and store retinoids. Upon activation by fibrogenic stimuli, quiescent HSCs transdifferentiate into myofibroblasts, lose their vitamin A, upregulate α-smooth muscle actin (αSMA), and produce collagen I. Various factors, including immune cell-derived fibrogenic molecules, growth factors, and lipopolysaccharide, as well as profibrotic lipid mediators such as lysophosphatidylinositol and lysophosphatidic acid, induce HSC activation in the course of chronic liver disease. TGF-β is the most HSC potent activator, which is produced by infiltrating lymphocytes and monocytes, Kupffer cells (KCs), and damaged hepatocytes. IL-17, produced by neutrophils and Th17 cells, sensitizes HSCs to TGF-β by upregulating TGF-β receptor II (TGF-βRII). In addition, platelet-derived growth factor (PDGF), which is produced by endothelial cells and macrophages, further promotes HSC activation. During fibrosis resolution, HSCs either die or revert to an inactive state by upregulating transcription factors such as peroxisome proliferator-activated receptor-γ (PPARγ), GATA-binding factor 4 (GATA4), GATA6, and transcription factor 21 (TCF21). NK and CD8<sup>+</sup> T cells can eliminate activated HSCs by inducing apoptosis (Further abbreviations: GM-CSF, granulocyte/macrophage colony-stimulating factor; HH, hedgehog ligands; IHH, Indian Hedgehog; LPA, lysophosphatidic acid; LPI, lysophosphatidylinositol; LPS, lipopolysaccharide; miRNA, microRNA; MSR1, macrophage scavenger receptor 1; NF-κB, nuclear factor κ-light chain-enhancer of activated B cells; OPN, osteopontin; oxLDL, oxidized low-density lipoprotein; ROS, reactive oxygen species; S1P, sphingosine-1-phosphate; SHH, sonic hedgehog; TLR4, Toll-like receptor 4). Modified from reference [<a href="#B5-ijms-25-07873" class="html-bibr">5</a>].</p>
Full article ">Figure 3
<p>The path towards liver fibrosis: Kupffer cell activation and macrophage recruitment in the chronic inflammatory microenvironment of the diseased liver. (<b>A</b>) Ingestion of fat-laden apoptotic hepatocytes and free cholesterol activates KCs by promoting the production of proinflammatory mediators. (<b>B</b>) The liver’s chronic inflammatory microenvironment recruits monocytes from the circulation, which, due to local proinflammatory signaling, differentiate into monocyte-derived KC-like inflammatory, as well as lipid-associated, macrophages. (<b>C</b>) Macrophage populations are the major contributors in shaping both profibrotic and antifibrotic drivers within the fibrotic niche. Relevant phenotypic markers of the macrophage populations detected in mouse models are indicated in the figure (Abbreviations: CEACAM1, carcinoembryonic antigen-related cell adhesion molecule 1; CLEC4F, C-type lectin domain family 4 member F; LAM, lipid-associated macrophage; Mac1, macrophage-1 antigen; Mar1, macrophage scavenger receptor1; MMP, matrix metalloproteinase; SAM, scar-associated macrophages; SatM, segregated nucleus-containing atypical monocytes; TGF-β, transforming growth factor-β; TNF-α, tumor necrosis factor-α; VSIG4, V-set and immunoglobulin domain containing 4).</p>
Full article ">
7 pages, 945 KiB  
Brief Report
The Novel Cytokine Interleukin-41/Meteorin-like Is Reduced in Diffuse Systemic Sclerosis
by Paul Freedman, Bettina Schock and Steven O’Reilly
Cells 2024, 13(14), 1205; https://doi.org/10.3390/cells13141205 - 17 Jul 2024
Viewed by 477
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disease with a triad of features that include vascular abnormalities, inflammation and skin and lung fibrosis. At the core of the disease is the activation of myofibroblasts from quiescent fibroblasts and this can be modified [...] Read more.
Systemic sclerosis (SSc) is an autoimmune connective tissue disease with a triad of features that include vascular abnormalities, inflammation and skin and lung fibrosis. At the core of the disease is the activation of myofibroblasts from quiescent fibroblasts and this can be modified by various cytokines. IL-41 is a recently described cytokine that was initially characterised as an adipokine as it was highly expressed in adipocytes and adipose tissue. However, it has recently been identified as being widely expressed and has immunomodulatory functions. This study examined the circulating levels of IL-41 and its expression in skin biopsies. We demonstrated significantly reduced levels of IL-41 in diffuse SSc that was also mirrored in the skin of SSc patients. AMPK has been proposed as a downstream target of IL-41, so we also measure mammalian target of rapamycin in skin and found that this is elevated in SSc patients. We speculate that IL-41 maybe an antifibrotic cytokine and its reduction may facilitate the activation of fibroblasts. Full article
(This article belongs to the Special Issue Fibrosis in Chronic Inflammatory Diseases)
Show Figures

Figure 1

Figure 1
<p>IL-41 is elevated in SSc serum. (<b>A</b>) Mean IL-41 levels in HC and diffuse SSc levels quantified by ELISA. Data are the mean and SEM from 18 donors; **** <span class="html-italic">p</span> = &lt;0.0001 for Mann–Whitney U test. (<b>B</b>) No correlation between IL-41 and mRSS in SSc patients; r = −0.007, <span class="html-italic">p</span> = 0.98; Pearson correlation is two-tailed.</p>
Full article ">Figure 2
<p>IL-41 is reduced in SSc skin. IL-41 was quantified in whole skin by qPCR and normalised to 18S gene and shown as fold change compared to HC. Data are the mean and SEM; * <span class="html-italic">p</span> = 0.04 for Students <span class="html-italic">t</span> test; <span class="html-italic">n</span> = 5.</p>
Full article ">Figure 3
<p>Elevated mTOR in SSc skin. (<b>A</b>) mRNA of mTOR in SSc or HC patient measured by qPCR and normalised to 18S gene and shown as fold change compared to HC. Data are the mean and SEM; * <span class="html-italic">p</span> = 0.022 for Student’s <span class="html-italic">t</span> test; <span class="html-italic">n</span> = 5. (<b>B</b>) Correlation analysis between IL-41 mRNA and mTOR mRNA expression; <span class="html-italic">p</span> = 0.48, r = 0.42; Pearsons correlation is two-tailed; <span class="html-italic">n</span> = 5.</p>
Full article ">
13 pages, 2899 KiB  
Article
The Influence of Sulfation Degree of Glycosaminoglycan-Functionalized 3D Collagen I Networks on Cytokine Profiles of In Vitro Macrophage–Fibroblast Cocultures
by Franziska Ullm, Alexander Renner, Uwe Freudenberg, Carsten Werner and Tilo Pompe
Gels 2024, 10(7), 450; https://doi.org/10.3390/gels10070450 - 9 Jul 2024
Viewed by 569
Abstract
Cell–cell interactions between fibroblasts and immune cells, like macrophages, are influenced by interaction with the surrounding extracellular matrix during wound healing. In vitro hydrogel models that mimic and modulate these interactions, especially of soluble mediators like cytokines, may allow for a more detailed [...] Read more.
Cell–cell interactions between fibroblasts and immune cells, like macrophages, are influenced by interaction with the surrounding extracellular matrix during wound healing. In vitro hydrogel models that mimic and modulate these interactions, especially of soluble mediators like cytokines, may allow for a more detailed investigation of immunomodulatory processes. In the present study, a biomimetic extracellular matrix model based on fibrillar 3D collagen I networks with a functionalization with heparin or 6-ON-desulfated heparin, as mimics of naturally occurring heparan sulfate, was developed to modulate cytokine binding effects with the hydrogel matrix. The constitution and microstructure of the collagen I network were found to be stable throughout the 7-day culture period. A coculture study of primary human fibroblasts/myofibroblasts and M-CSF-stimulated macrophages was used to show its applicability to simulate processes of progressed wound healing. The quantification of secreted cytokines (IL-8, IL-10, IL-6, FGF-2) in the cell culture supernatant demonstrated the differential impact of glycosaminoglycan functionalization of the collagen I network. Most prominently, IL-6 and FGF-2 were shown to be regulated by the cell culture condition and network constitution, indicating changes in paracrine and autocrine cell–cell communication of the fibroblast–macrophage coculture. From this perspective, we consider our newly established in vitro hydrogel model suitable for mechanistic coculture analyses of primary human cells to unravel the role of extracellular matrix factors in key events of tissue regeneration and beyond. Full article
(This article belongs to the Section Gel Applications)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Preparation and characterization of 3D GAG-modified Coll I matrices. (<b>A</b>) Schematic illustration of performed GAG modification of Coll I matrices. To maintain the 3D distribution of both cell types in the network, GAG solutions were applied after Coll I fibrillation was completed. Figure was generated using <a href="http://biorender.com" target="_blank">biorender.com</a> (accessed on 11 June 2024). (<b>B</b>) Turbidity measurement of Coll I fibrillogenesis performed over 120 min at 405 nm with and without cells present in the Coll I solution. (<b>C</b>,<b>D</b>) Representative cLSM images of Coll I matrices showing atto550-labelled variants of heparin and 6-ON-desulfated heparin (red) either 1 d (<b>C</b>) or 4 d (<b>D</b>) after reconstitution. Scale bar 20 µm. (<b>E</b>) Analysis of GAG amount in Coll I matrices 1 d and 4 d after reconstitution using papain digestion (<span class="html-italic">n</span> = 6; data is given as mean ± SE; Tukey’s multiple comparisons test followed by two-way ANOVA; * indicates <span class="html-italic">p</span> ≤ 0.05; ** indicates <span class="html-italic">p</span> ≤ 0.005 between the respective heparin variants; no significant differences between 1 d and 4 d comparison). (<b>F</b>) Analysis of cell viability after 4 d of cell culture exemplarily shown for heparin-modified Coll I matrices after 30 and 60 min of GAG incubation (<span class="html-italic">n</span> = 3, data is not significant after ordinary one-way ANOVA, Box-whisker plots show the 25 and 75 percentile range (box) with Tukey 95% confidence intervals (whiskers) and median values (transversal line); dotted line represents respective Coll I control without GAG modification).</p>
Full article ">Figure 2
<p>Cytokine secretion after 7 d in GAG-modified Coll I matrices. Secretion of (<b>A</b>) IL-8, (<b>B</b>) IL-10, (<b>C</b>) IL-6 and (<b>D</b>) FGF-2 by M-MΦ, MyoFb and in coculture. Concentration of cytokines (pg mL<sup>−1</sup>) in supernatants was determined by multiplex assay after 7 d using a standard curve. Data are shown as mean ± SD from 3 separate cell experiments. Levels of significance are shown against the respective coculture condition and were set to <span class="html-italic">p</span> ≤ 0.05 (*) using Tukey’s multiple comparisons test followed by two-way ANOVA, whereas <span class="html-italic">p</span> ≤ 0.01 (**) and <span class="html-italic">p</span> ≤ 0.001 (***) represents very and highly significant, respectively.</p>
Full article ">Figure 3
<p>Mediation of fibroblast–macrophage interactions by functionalization of 3D collagen I networks with sulfated glycosaminoglycans. For biomimetic 3D scaffold preparation, modification of Coll I fibrils with GAG variants was performed after fibrillogenesis. The figure describes the changes of the three main components of the culture system over the period of 7 d: Coll I matrix, myofibroblasts and macrophages. The figure was generated using <a href="http://biorender.com" target="_blank">biorender.com</a> (accessed on 11 June 2024).</p>
Full article ">
15 pages, 1345 KiB  
Review
Overcoming Chemoresistance in Cancer: The Promise of Crizotinib
by Sanaa Musa, Noor Amara, Adan Selawi, Junbiao Wang, Cristina Marchini, Abed Agbarya and Jamal Mahajna
Cancers 2024, 16(13), 2479; https://doi.org/10.3390/cancers16132479 - 7 Jul 2024
Viewed by 1243
Abstract
Chemoresistance is a major obstacle in cancer treatment, often leading to disease progression and poor outcomes. It arises through various mechanisms such as genetic mutations, drug efflux pumps, enhanced DNA repair, and changes in the tumor microenvironment. These processes allow cancer cells to [...] Read more.
Chemoresistance is a major obstacle in cancer treatment, often leading to disease progression and poor outcomes. It arises through various mechanisms such as genetic mutations, drug efflux pumps, enhanced DNA repair, and changes in the tumor microenvironment. These processes allow cancer cells to survive despite chemotherapy, underscoring the need for new strategies to overcome resistance and improve treatment efficacy. Crizotinib, a first-generation multi-target kinase inhibitor, is approved by the FDA for the treatment of ALK-positive or ROS1-positive non-small cell lung cancer (NSCLC), refractory inflammatory (ALK)-positive myofibroblastic tumors (IMTs) and relapsed/refractory ALK-positive anaplastic large cell lymphoma (ALCL). Crizotinib exists in two enantiomeric forms: (R)-crizotinib and its mirror image, (S)-crizotinib. It is assumed that the R-isomer is responsible for the carrying out various processes reviewed here The S-isomer, on the other hand, shows a strong inhibition of MTH1, an enzyme important for DNA repair mechanisms. Studies have shown that crizotinib is an effective multi-kinase inhibitor targeting various kinases such as c-Met, native/T315I Bcr/Abl, and JAK2. Its mechanism of action involves the competitive inhibition of ATP binding and allosteric inhibition, particularly at Bcr/Abl. Crizotinib showed synergistic effects when combined with the poly ADP ribose polymerase inhibitor (PARP), especially in ovarian cancer harboring BRCA gene mutations. In addition, crizotinib targets a critical vulnerability in many p53-mutated cancers. Unlike its wild-type counterpart, the p53 mutant promotes cancer cell survival. Crizotinib can cause the degradation of the p53 mutant, sensitizing these cancer cells to DNA-damaging substances and triggering apoptosis. Interestingly, other reports demonstrated that crizotinib exhibits anti-bacterial activity, targeting Gram-positive bacteria. Also, it is active against drug-resistant strains. In summary, crizotinib exerts anti-tumor effects through several mechanisms, including the inhibition of kinases and the restoration of drug sensitivity. The potential of crizotinib in combination therapies is emphasized, particularly in cancers with a high prevalence of the p53 mutant, such as triple-negative breast cancer (TNBC) and high-grade serous ovarian cancer (HGSOC). Full article
(This article belongs to the Collection Innovations in Cancer Drug Development Research)
Show Figures

Figure 1

Figure 1
<p>Chemical structure of (R)-crizotinib (<b>left</b>), and (S)-crizotinib (<b>right</b>). While (S)-crizotinib demonstrated initial promise, (R)-crizotinib emerged as the clinically preferred enantiomer due to its distinct properties. This review will therefore concentrate on the functions of (R)-crizotinib. Thus, crizotinib will refer to (R)-crizotinib in this context.</p>
Full article ">Figure 2
<p>Crizotinib activity. (R)-crizotinib structure is shown. FDA-approved indications are present in black writing. The abilities to modulate activity of other enzymes and processes are present in purple writing.</p>
Full article ">Figure 3
<p>Comparison of ATP competitive and allosteric inhibitors of kinases. This figure illustrates the distinct mechanisms by which ATP competitive and allosteric inhibitors regulate kinase activity. (<b>A</b>) The active site of the kinase is depicted, highlighting the ATP binding pocket crucial for phosphorylating target proteins. ATP competitive drugs directly compete with ATP for binding at this site, thereby inhibiting kinase activity. (<b>B</b>) The action of allosteric inhibitors. These inhibitors bind to a different regulatory site on the enzyme. Upon binding, they induce conformational changes that indirectly affect the ATP binding pocket, hindering ATP binding and ultimately inhibiting enzymatic activity.</p>
Full article ">
11 pages, 2117 KiB  
Article
Depletion of Activated Hepatic Stellate Cells and Capillarized Liver Sinusoidal Endothelial Cells Using a Rationally Designed Protein for Nonalcoholic Steatohepatitis and Alcoholic Hepatitis Treatment
by Falguni Mishra, Yi Yuan, Jenny J. Yang, Bin Li, Payton Chan and Zhiren Liu
Int. J. Mol. Sci. 2024, 25(13), 7447; https://doi.org/10.3390/ijms25137447 - 6 Jul 2024
Viewed by 927
Abstract
Nonalcoholic steatohepatitis (NASH) and alcoholic hepatitis (AH) affect a large part of the general population worldwide. Dysregulation of lipid metabolism and alcohol toxicity drive disease progression by the activation of hepatic stellate cells and the capillarization of liver sinusoidal endothelial cells. Collagen deposition, [...] Read more.
Nonalcoholic steatohepatitis (NASH) and alcoholic hepatitis (AH) affect a large part of the general population worldwide. Dysregulation of lipid metabolism and alcohol toxicity drive disease progression by the activation of hepatic stellate cells and the capillarization of liver sinusoidal endothelial cells. Collagen deposition, along with sinusoidal remodeling, alters sinusoid structure, resulting in hepatic inflammation, portal hypertension, liver failure, and other complications. Efforts were made to develop treatments for NASH and AH. However, the success of such treatments is limited and unpredictable. We report a strategy for NASH and AH treatment involving the induction of integrin αvβ3-mediated cell apoptosis using a rationally designed protein (ProAgio). Integrin αvβ3 is highly expressed in activated hepatic stellate cells (αHSCs), the angiogenic endothelium, and capillarized liver sinusoidal endothelial cells (caLSECs). ProAgio induces the apoptosis of these disease-driving cells, therefore decreasing collagen fibril, reversing sinusoid remodeling, and reducing immune cell infiltration. The reversal of sinusoid remodeling reduces the expression of leukocyte adhesion molecules on LSECs, thus decreasing leukocyte infiltration/activation in the diseased liver. Our studies present a novel and effective approach for NASH and AH treatment. Full article
(This article belongs to the Special Issue Chronic Liver Disease and Hepatocellular Carcinoma)
Show Figures

Figure 1

Figure 1
<p>ProAgio decreases hepatic fibrosis in NASH mice. (<b>A</b>) The scheme illustrates the ProAgio or vehicle treatment regimen of the NASH mice. (<b>B</b>,<b>C</b>) Representative images (<b>B</b>) and quantifications (<b>C</b>) of Sirius Red staining of liver sections from the treated mice. ET, early treatment; LT late treatment. (<b>D</b>,<b>E</b>) Representative images (<b>D</b>) and quantifications (<b>E</b>) of IHC staining of αSMA in liver sections from the treated mice. Quantitation of collagen by Sirius Red and αSMA levels by IHC of αSMA staining using Fiji software version 2.14.0/1.54f. Four randomly selected tissue sections per animal, three randomly selected view fields in each section, and six randomly selected animals (n = 6) were quantified. The quantity of collagen and αSMA levels are presented as % of staining positive area. (<b>F</b>) Blood glucose levels (mM/L) of five mice (n = 5, showing in different color) treated with the indicated agents were measured at indicated time points before (time 0) and after i.v. injection of 2 g/kg glucose. The mice were fasted overnight before the glucose injection and measurements. (<b>G</b>) Representative images of H&amp;E-stained liver sections from NASH mice treated with the indicated agents. Hepatic ballooning was scored by a hepatic pathologist based on H&amp;E staining (see the arrows in (<b>G</b>) for examples; 5 randomly selected sections per animal were scored n = 5): normal and ProAgio-treated animals score 0; vehicle-treated animals score 2 (number in each panel). The error bars in (<b>C</b>,<b>E</b>) are the standard deviations of five independent mice. Statistical analysis of data was performed by a one-way Student’s <span class="html-italic">t</span>-test. (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>ProAgio reduces inflammation in NASH mouse liver. (<b>A</b>,<b>B</b>) Representative images (<b>A</b>) and quantifications (<b>B</b>) of F4/80 staining of liver sections from mice treated with indicated agents. The quantity of total macrophage levels is presented as % of staining positive area. (<b>C</b>,<b>D</b>) Total population (%) of macrophages (<b>C</b>) and polymorphonuclear neutrophils (PMNs) (<b>D</b>) in liver tissues from NASH mice treated with indicated agents were analyzed by FACS. For macrophages in (<b>C</b>), CD45<sup>+</sup>/CD11b<sup>+</sup>/F4/80<sup>+</sup>/Ly-6C<sup>−</sup> cells were used. For PMN in (D), CD45<sup>+</sup>/CD11b<sup>+</sup>/Ly-6G<sup>+</sup> were used. (<b>E</b>,<b>F</b>) Representative images (<b>E</b>) and quantifications (<b>F</b>) of IHC staining of CD44 in liver sections from NASH mice treated with indicated agents. The quantity of CD44 levels is presented as % of staining positive area. Con in (<b>D</b>) and Controls in (<b>E</b>,<b>F</b>) means the mice were normal healthy mice without any disease induction and subsequent treatment. The error bars in (<b>B</b>–<b>D</b>,<b>F</b>) are standard deviations of measurements of 4 mice for (<b>B</b>,<b>C</b>) and 6 mice for (<b>D</b>,<b>F</b>). Statistical analysis of data was performed via a Student’s <span class="html-italic">t</span>-test for a two-group comparison by a one-way ANOVA with Tukey’s multiple comparison test. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>ProAgio decreases hepatic fibrosis in AH mice. (<b>A</b>) The scheme illustrates the ProAgio or vehicle treatment regimen for the AH mice. (<b>B</b>,<b>C</b>) Representative images (<b>B</b>) and quantifications (<b>C</b>) of Sirius Red staining of liver sections from mice treated with indicated agents. (<b>D</b>,<b>E</b>) Representative images (<b>D</b>) and quantifications (<b>E</b>) of IHC staining of αSMA in liver sections from the treated mice. (<b>F</b>,<b>G</b>) Representative images (<b>F</b>) and quantifications (<b>G</b>) of IHC staining of CD44 in liver sections from mice treated with indicated agents. The quantities in (<b>C</b>,<b>E</b>,<b>G</b>) are presented as % of staining positive area. Three randomly selected view fields per section, four randomly selected sections per animal, and six randomly selected animals (n = 6) were quantified. Veh in (<b>G</b>) is the vehicle-treated group. Control in (<b>G</b>) means the mice were normal healthy mice without disease induction and subsequent treatments. The error bars in (<b>C</b>,<b>E</b>,<b>G</b>) are standard deviations of 6 independent mice. Statistical analysis of data was performed by a Student’s <span class="html-italic">t</span>-test for a two-group comparison or a one-way ANOVA with Tukey’s multiple comparison test. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 4
<p>ProAgio reduces inflammation in AH mouse liver. (<b>A</b>) The total population of neutrophils (Neutrophil%) in the liver tissues from the AH mice that were treated with indicated agents was analyzed by FACS (CD11b<sup>+</sup> Ly-6G<sup>+</sup>). (<b>B</b>,<b>C</b>) Representative images (<b>B</b>) and quantifications (<b>C</b>) of F4/80 staining of liver sections from mice treated with indicated agents. The quantity of total macrophage levels is presented as % of staining positive area. (<b>D</b>,<b>E</b>) Representative images (<b>D</b>) and quantifications (<b>E</b>) of IHC staining of SE-1 in liver sections from the NASH and AH mice that were treated with indicated agents. The quantity of differentiated healthy LSEC levels in (<b>E</b>) is presented as % of SE-1-positive staining area. The error bars in (<b>A</b>,<b>C</b>,<b>E</b>) are standard deviations of measurements of 6 mice. Statistical analysis of data was performed by a one-way ANOVA with Tukey’s multiple comparison test or an unpaired Student’s <span class="html-italic">t</span>-test for two-group comparisons. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>ProAgio decreases immune cell attachment molecules in NASH and AH mice (<b>A</b>–<b>D</b>) Representative images (<b>A</b>,<b>C</b>) and quantifications (<b>B</b>,<b>D</b>) of IHC staining of LYVE-1 (<b>A</b>,<b>B</b>) or VAP-1 (<b>C</b>,<b>D</b>) of liver sections from NASH or AH mice treated with indicated agents. The total LYVE-1 or VAP-1 levels are presented as % of staining positive area. Con and control mean the sections from normal healthy mice without any disease induction or subsequent treatment. (<b>E</b>) Scheme illustrating the drug actions of ProAgio in steatohepatitis. The error bars in (<b>B</b>,<b>D</b>) are standard deviations of measurements of 6 mice. Statistical analysis of data was performed by a one-way ANOVA with Tukey’s multiple comparison test. (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">
1 pages, 622 KiB  
Correction
Correction: Petroll et al. Experimental Models for Investigating Intra-Stromal Migration of Corneal Keratocytes, Fibroblasts and Myofibroblasts. J. Funct. Biomater. 2012, 3, 183–198
by Walter Matthew Petroll, Neema Lakshman and Lisha Ma
J. Funct. Biomater. 2024, 15(7), 182; https://doi.org/10.3390/jfb15070182 - 2 Jul 2024
Viewed by 437
Abstract
In the original publication [...] Full article
Show Figures

Figure 6

Figure 6
<p>Maximum intensity projection images (~50 microns thick) of Live/Dead staining after 1 day of culture following freeze injury using sandwich construct. Live cells are labeled green and dead cells are labeled red. (<b>A</b>) 1 day after freeze injury, induced by pushing on the surface of the matrix using a cold 3 mm diameter probe; (<b>B</b>) 1 day control sample, in which a room temperature probe was used.</p>
Full article ">
18 pages, 2758 KiB  
Review
The Influence of Metabolic Risk Factors on the Inflammatory Response Triggered by Myocardial Infarction: Bridging Pathophysiology to Treatment
by Lisaidy Ramos-Regalado, Sebastià Alcover, Lina Badimon and Gemma Vilahur
Cells 2024, 13(13), 1125; https://doi.org/10.3390/cells13131125 - 29 Jun 2024
Viewed by 720
Abstract
Myocardial infarction (MI) sets off a complex inflammatory cascade that is crucial for effective cardiac healing and scar formation. Yet, if this response becomes excessive or uncontrolled, it can lead to cardiovascular complications. This review aims to provide a comprehensive overview of the [...] Read more.
Myocardial infarction (MI) sets off a complex inflammatory cascade that is crucial for effective cardiac healing and scar formation. Yet, if this response becomes excessive or uncontrolled, it can lead to cardiovascular complications. This review aims to provide a comprehensive overview of the tightly regulated local inflammatory response triggered in the early post-MI phase involving cardiomyocytes, (myo)fibroblasts, endothelial cells, and infiltrating immune cells. Next, we explore how the bone marrow and extramedullary hematopoiesis (such as in the spleen) contribute to sustaining immune cell supply at a cardiac level. Lastly, we discuss recent findings on how metabolic cardiovascular risk factors, including hypercholesterolemia, hypertriglyceridemia, diabetes, and hypertension, disrupt this immunological response and explore the potential modulatory effects of lifestyle habits and pharmacological interventions. Understanding how different metabolic risk factors influence the inflammatory response triggered by MI and unraveling the underlying molecular and cellular mechanisms may pave the way for developing personalized therapeutic approaches based on the patient’s metabolic profile. Similarly, delving deeper into the impact of lifestyle modifications on the inflammatory response post-MI is crucial. These insights may enable the adoption of more effective strategies to manage post-MI inflammation and improve cardiovascular health outcomes in a holistic manner. Full article
Show Figures

Figure 1

Figure 1
<p>Necrotic cardiomyocytes generate danger-associated molecular patterns (DAMPs) acting as danger signals on different immune cells through pattern recognition receptors (PRRs), enhancing cytokine release and local inflammatory response. High-mobility group box 1 protein (HMGB1); toll-like receptors (TLRs); receptors for advanced glycation end products (RAGEs); heat-shock proteins (HSPs); tumor necrosis factor-alpha (TNF-α); interleukin (IL); mitochondrial DNA (mtDNA); nuclear factor-kappa light chain enhancer of activated B cells (NFκB); receptor-associated kinase 1 (IRAK1); mitogen-activated protein kinase (MAPK); Nucleotide-binding Oligomerization Domain (NOD)-like receptors pyrin domain containing 3 (NLRP3). Illustration created with BioRender.</p>
Full article ">Figure 2
<p>Bone marrow and splenic hematopoiesis fuel the cardiac inflammatory response during myocardial infarction (MI). In the bone marrow, granulocyte colony-stimulating factor (G-CSF) and granulocyte–macrophage colony-stimulating factor (GM-CSF), along with various inflammatory mediators, stimulate the proliferation and release of granulocytes and monocytes into the bloodstream. This process is further supported by sympathetic nervous system fibers since they decrease chemokine motif ligand 12 (CXCL12) activity, promoting Hematopoietic stem cells (HSC) release. In the spleen, angiotensin II (Ang II) enhances the motility and migration of resident monocyte populations, while interleukin-1β (IL-1β) triggers extramedullary hematopoiesis. This process involves the interaction of bone marrow-released HSCs with CD169+ macrophages via vascular cell adhesion molecule 1 (VCAM-1). Consequently, inflammatory cells migrate into the infarcted heart. Neutrophils infiltrate the cardiac tissue through β2-integrin and P-selectin, producing reactive oxygen species (ROS) and chemokines that facilitate the transmigration of dendritic cells and monocytes into the damaged tissue, creating a positive feedback loop. Additionally, neutrophils also release extracellular traps (NETs) and extracellular vesicles (EVs), further sustaining the inflammatory response. Epigenetic modifications, including microRNAs and DNA methylation, also contribute to the modulation of the overall inflammatory response post-MI. Danger-associated molecular patterns (DAMPs); interleukin (IL); angiotensin receptor (AT-1; AT-2); chemokine (C-C motif) ligand 7 (CCL7). Illustration created with BioRender.</p>
Full article ">Figure 3
<p>The role of macrophages, dendritic cells, and lymphocytes in the healing process post-myocardial infarction (MI). Monocytes are recruited to the infarcted heart, where they differentiate into activated macrophages, increasing immune cell recruitment and promoting the inflammatory response. Over time, macrophages remove necrotic cells and shift toward an anti-inflammatory phenotype, producing interleukin 10 (IL-10). Dendritic cells present antigenic proteins that shift the fibroblast phenotype toward a proinflammatory state, producing extracellular matrix proteins to maintain myocardial integrity. T and B lymphocyte activation triggers myocardial destruction and interferon gamma (IFN-γ) and IL-17 release. Activated B cells increase C-C motif ligand 7 and release IL-10, which enhance cardiac repair and wound healing. Reactive oxygen species (ROS); monocyte chemoattractant protein-1 (MCP-1); C-C motif chemokine receptor 2 (CCR2); vascular cell adhesion molecule 1 (VCAM-1); tumor necrosis factor-alpha (TNF-α); toll-like receptors (TLRs); receptors for advanced glycation end products (RAGEs). Illustration created with BioRender.</p>
Full article ">
14 pages, 7521 KiB  
Article
Evaluation of Immunological Response to TLR2 and α-SMA in Crohn’s Disease and Ulcerative Colitis
by Anthea Miller, Giorgia Pia Lombardo, Giuseppina Rizzo, Magdalena Kotanska, Giuseppinella Melita, Socrate Pallio, Alba Migliorato, Giuseppina Cutroneo and Simona Pergolizzi
Gastroenterol. Insights 2024, 15(3), 541-554; https://doi.org/10.3390/gastroent15030040 - 28 Jun 2024
Viewed by 536
Abstract
Inflammatory bowel diseases (IBDs) represent multifactorial chronic inflammatory conditions of the gastrointestinal tract. The main IBDs are Crohn’s disease (CD) and ulcerative colitis (UC). CD may cause perforation, stricture or transmural inflammation, which can occur discontinuously in the entire gastrointestinal tract (GIT). UC [...] Read more.
Inflammatory bowel diseases (IBDs) represent multifactorial chronic inflammatory conditions of the gastrointestinal tract. The main IBDs are Crohn’s disease (CD) and ulcerative colitis (UC). CD may cause perforation, stricture or transmural inflammation, which can occur discontinuously in the entire gastrointestinal tract (GIT). UC leads to mucosal inflammation as well as mucosal atrophy in the rectum and the colon. Innate immunity is considered the first line of defense against microbial invasion; among Toll-like receptors, TLR2 is the most important for defense against mycobacterial infection. TLR2 has been reported to have a lot of functions in infectious diseases and in other pathologies, such as chronic and acute inflammatory diseases. Alfa-Smooth Muscle Actin (α-SMA) is an important biomarker in IBDs. All myofibroblasts express α-SMA, which has been found to be upregulated in CD and UC. Paraformaldehyde-fixed intestinal tissues, from patients with CD and patients with UC, were analyzed by immunostaining for TLR2 and α-SMA. Our results showed that, in the samples obtained from UC patients with inflamed mucosa, TLR2-positive epithelial cells concentrated on the mucosal surface and scattered immune cells in the connective tissue; furthermore, numerous α-SMA-positive cells (subepithelial myofibroblasts) were detected in the lamina propria and around glands, while some myofibroblasts co-localizing with α-SMA and TLR2 could be inflammatory macrophages. In CD patients, TLR2-positive enterocytes and α-SMA-positive myofibroblasts in the lamina propria of the villus have been observed. In control samples, a low positivity to α-SMA and TLR2 was observed in subepithelial myofibroblasts and scattered immune cells of the lamina propria. These data showed the recall of α-SMA-positive myofibroblasts during the inflammatory state; in addition, TLR2 expression has been observed to change in the intestinal epithelium in IBDs, demonstrating that alterations in the innate system response may contribute to the pathogenesis of these diseases. Full article
(This article belongs to the Section Gastrointestinal Disease)
Show Figures

Figure 1

Figure 1
<p>The Mayo Endoscopic Score (ESM) is the most widely used endoscopic index to evaluate the stage of ulcerative colitis. Normal mucosa (<b>1</b>).</p>
Full article ">Figure 2
<p>Mild activity (erythema, thinned/distorted vascular reticulum) (<b>2</b>).</p>
Full article ">Figure 3
<p>Severe activity (spontaneous bleeding, extensive ulceration) (<b>3</b>).</p>
Full article ">Figure 4
<p>Normal mucosa (<b>4</b>).</p>
Full article ">Figure 5
<p>Mild activity with ulcerated surfaces (<b>5</b>).</p>
Full article ">Figure 6
<p>Severe activity with inflamed mucosa due to Crohn’s disease (<b>6</b>).</p>
Full article ">Figure 7
<p>Graph of the power analysis data.</p>
Full article ">Figure 8
<p>UC human colon biopsy samples, 40×, scale bar 20 µm. TLR2-positive epithelial cells (arrowheads), between goblet cells (gb) and scattered immune cells (red arrows) in the connective tissue are present. Numerous positive cells (subepithelial myofibroblasts α-SMA positive (green arrows) in the lamina propria and in the connective tissue can be noted. Some myofibroblasts in the connective tissue (yellow arrows) co-localize with TLR2 and α-SMA. TL = transmitted light, to highlight tissue morphology.</p>
Full article ">Figure 9
<p>UC human colon biopsy samples, 40×, scale bar 20 µm. α-SMA positive myofibroblasts (green arrows) forming a pericryptal fibroblast sheath around glands (green arrows) can be seen. TL = transmitted light, to highlight tissue morphology.</p>
Full article ">Figure 10
<p>Control group colon, 40×, scale bar 20 µm. Low reactivity to TLR2 and α-SMA is shown in healthy colonic mucosa. TL = transmitted light, to highlight tissue morphology.</p>
Full article ">Figure 11
<p>Samples of inflamed ileum in CD patients, 40×, scale bar 20 µm. TLR2-positive enterocytes (arrowheads) are present. α-SMA-positive myofibroblasts are present in the lamina propria of the villus (green arrows), which form a continuous layer in the muscularis mucosae. TL = transmitted light, to highlight tissue morphology.</p>
Full article ">Figure 12
<p>Samples of inflamed ileum in CD patients, 40×, scale bar 20 µm. α-SMA-positive myofibroblasts are present in the lamina propria of the villus (green arrows). TL = transmitted light, to highlight tissue morphology.</p>
Full article ">Figure 13
<p>Control group ileum, 40×, scale bar 20 µm. Low reactivity to TLR2 and α-SMA is shown in samples of the healthy gut. TL = transmitted light, to highlight tissue morphology.</p>
Full article ">
22 pages, 9362 KiB  
Article
Reversed Corneal Fibroblasts Therapy Restores Transparency of Mouse Cornea after Injury
by Maria A. Surovtseva, Kristina Yu. Krasner, Irina I. Kim, Nikolay V. Surovtsev, Elena V. Chepeleva, Natalia A. Bondarenko, Alexander P. Lykov, Nataliya P. Bgatova, Alina A. Alshevskaya, Alexander N. Trunov, Valery V. Chernykh and Olga V. Poveshchenko
Int. J. Mol. Sci. 2024, 25(13), 7053; https://doi.org/10.3390/ijms25137053 - 27 Jun 2024
Viewed by 2248
Abstract
Cell-based therapies using corneal stromal stem cells (CSSC), corneal keratocytes, or a combination of both suppress corneal scarring. The number of quiescent keratocytes in the cornea is small; it is difficult to expand them in vitro in quantities suitable for transplantation. This study [...] Read more.
Cell-based therapies using corneal stromal stem cells (CSSC), corneal keratocytes, or a combination of both suppress corneal scarring. The number of quiescent keratocytes in the cornea is small; it is difficult to expand them in vitro in quantities suitable for transplantation. This study examined the therapeutic effect of corneal fibroblasts reversed into keratocytes (rCF) in a mouse model of mechanical corneal injury. The therapeutic effect of rCF was studied in vivo (slit lamp, optical coherence tomography) and ex vivo (transmission electron microscopy and immunofluorescence staining). Injection of rCF into the injured cornea was accompanied by recovery of corneal thickness, improvement of corneal transparency, reduction of type III collagen in the stroma, absence of myofibroblasts, and the improvement in the structural organization of collagen fibers. TEM results showed that 2 months after intrastromal injection of cells, there was a decrease in the fibril density and an increase in the fibril diameter and the average distance between collagen fibrils. The fibrils were well ordered and maintained the short-range order and the number of nearest-neighbor fibrils, although the averaged distance between them increased. Our results demonstrated that the cell therapy of rCF from ReLEx SMILe lenticules promotes the recovery of transparent corneal stroma after injury. Full article
Show Figures

Figure 1

Figure 1
<p>Immunofluorescence microscopy of fibroblasts reversed into keratocytes (rCF) and fibroblasts (CFs). The images show the comparative expression of various markers in cells: red—stained with antibodies to keratocan (arrow); green—staining with antibodies to lumican (arrow) or collagen type I (arrow); blue—DAPI staining of nuclei. The scale bar is 50 µm for keratocan and lumican (the image was captured at 20×) and 20 µm for collagen type (the image was captured at 40×).</p>
Full article ">Figure 2
<p>Corneal opacity mouse model. (<b>A</b>) Slit-lamp biomicroscopy with fluorescein. (<b>B</b>) Slit-lamp biomicroscopy without fluorescein.</p>
Full article ">Figure 3
<p>Corneal thickness 2 months after injury and hCSC injection. (<b>A</b>) Representative OCT images at months 2 post injury and cell treatment. (<b>B</b>) Corneal thickness (µm) at 2 months post injury and cell treatment. Data presented as [Me (Q1; Q3)], (<span class="html-italic">n</span> = 8 in each group). * <span class="html-italic">p</span> &lt; 0.05 compared to the control (healthy cornea), ** <span class="html-italic">p</span> = 0.01 compared to the control (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the injury + saline.</p>
Full article ">Figure 4
<p>Corneal transparency 2 months after injury and hCSC injection. Mean gray value analysis of OCT data was performed using ImageJ 1.48 v software. Data are presented as [Me (Q1; Q3)], (<span class="html-italic">n</span> = 8 in each group). * <span class="html-italic">p</span> &lt; 0.001 compared to the control group (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the injury group, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.001 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> &lt; 0.05 compared to the injury + saline group, <sup>!!</sup> <span class="html-italic">p</span> &lt; 0.001 compared to the injury + saline group.</p>
Full article ">Figure 5
<p>Immunofluorescence analysis of mouse corneal stroma after 2 months of injury and of hCSC injection. (<b>A</b>) Immunofluorescence staining of collagen type III in mouse corneal stroma. (<b>B</b>) Immunofluorescence staining of α-SMA in mouse corneal stroma. The images show the comparative expression of various markers in cells: green—staining with antibodies to collagen type III; red—stained with antibodies to α-SMA; blue—DAPI staining of nuclei. The scale bar is 50 µm and 100 µm.</p>
Full article ">Figure 6
<p>Ultrastructure of keratocytes and extracellular collagen matrix in the mouse cornea 2 months after injury and hCSC injection. (<b>a</b>–<b>c</b>) “control (healthy cornea)”: (<b>a</b>)—ordered arrangement of collagen fibrils around the keratocyte; (<b>b</b>)—regular distance between collagen fibrils; (<b>c</b>)—keratocyte between collagen fibers (arrow). (<b>d</b>–<b>f</b>) “injury”: (<b>d</b>)—low density and random arrangement of collagen fibrils around the keratocyte (arrow); (<b>e</b>)—random arrangement of collagen fibrils (arrow); (<b>f</b>)—keratocyte with an increased content of mitochondria in the cytoplasm. (<b>g</b>–<b>i</b>) “injury + saline”: (<b>g</b>)—elongated keratocyte with a disordered arrangement of collagen fibrils and irregular distance between fibrils (arrow); (<b>h</b>)—neutrophils in the corneal stroma; (<b>i</b>)–keratocyte with swollen mitochondria (arrows), disordered arrangement of collagen fibrils and irregular distance between fibrils. (<b>j</b>–<b>l</b>) “injury + CF”: (<b>j</b>)—low density and random arrangement of collagen fibrils around the keratocyte; (<b>k</b>)—structure of the collagen matrix–heterogeneity in the thickness of the collagen fiber bundles; (<b>l</b>)—keratocyte with a high content of mitochondria. (<b>m</b>–<b>o</b>) “injury + rCF”: (<b>m</b>)—ordered arrangement of collagen fibrils around the keratocyte; (<b>n</b>)—perpendicular arrangement of collagen fiber bundles in the stroma; (<b>o</b>)—convoluted form of keratocyte, corresponding to the course of collagen fibers.</p>
Full article ">Figure 7
<p>Fibril diameters 2 months after injury and hCSC injection determined with the ImageJ 1.48 v software from TEM images. Five corneas were analyzed with five images per cornea. For each image, at least 100 fibrils were evaluated. (<b>A</b>) Data are presented as means ± SDs (<span class="html-italic">n</span> &gt; 500 in each group). * <span class="html-italic">p</span> = 0.00 compared to the control group (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> = 0.00 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> = 0.00 compared to the injury + saline, ^ <span class="html-italic">p</span> = 0.00 compared to the injury + CF. (<b>B</b>) Frequency of occurrence of collagen fibril diameter in mouse cornea.</p>
Full article ">Figure 8
<p>Fibril density 2 months after injury and hCSC injection determined with the ImageJ 1.48 v software from TEM images. Five corneas were analyzed with five images per cornea. For each image, at least 100 fibrils were evaluated. (<b>A</b>) Data are presented as means ± SDs (<span class="html-italic">n</span> &gt; 500 in each group). * <span class="html-italic">p</span> = 0.00 compared to the control (healthy cornea), ** <span class="html-italic">p</span> &lt; 0.05 compared to the control (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> = 0.00 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> = 0.00 compared to the injury + saline group, ^ <span class="html-italic">p</span> = 0.00 compared to the injury + CF group. (<b>B</b>) Frequency of occurrence of collagen fibril density in the mouse cornea.</p>
Full article ">Figure 8 Cont.
<p>Fibril density 2 months after injury and hCSC injection determined with the ImageJ 1.48 v software from TEM images. Five corneas were analyzed with five images per cornea. For each image, at least 100 fibrils were evaluated. (<b>A</b>) Data are presented as means ± SDs (<span class="html-italic">n</span> &gt; 500 in each group). * <span class="html-italic">p</span> = 0.00 compared to the control (healthy cornea), ** <span class="html-italic">p</span> &lt; 0.05 compared to the control (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> = 0.00 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> = 0.00 compared to the injury + saline group, ^ <span class="html-italic">p</span> = 0.00 compared to the injury + CF group. (<b>B</b>) Frequency of occurrence of collagen fibril density in the mouse cornea.</p>
Full article ">Figure 9
<p>Interfibrillar distance analysis 2 months after injury and hCSC injection determined with the ImageJ 1.48 v software from TEM images. Five corneas in each study group with five or more images per cornea were analyzed. At least 300 fibrils were evaluated for each image. (<b>A</b>) Mean distance to the nearest neighbor between fibrils. Data are presented as a means ± SDs (<span class="html-italic">n</span> &gt; 500 in each group). * <span class="html-italic">p</span> = 0.00 compared to the control group (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> = 0.00 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> = 0.00 compared to the injury + saline group, ^ <span class="html-italic">p</span> = 0.00 compared to the injury + CF group. (<b>B</b>) Distributions of the nearest neighbor interfibrillar distances. Different curves in each frame correspond to calculations from different photographs. Arrows indicate apparent maxima of the distributions. The thick black lines are the averages of the different curves. (<b>C</b>) Fibril pair correlation function <span class="html-italic">g</span>(<span class="html-italic">r</span>). Each symbolized curve corresponds to data from one photograph. From 300 to 500 fibrils were analyzed in each photograph. (<b>D</b>) Averaged pair correlation functions. The arrows show the radii of the circles within which the four closest neighboring fibrils are located.</p>
Full article ">Figure 9 Cont.
<p>Interfibrillar distance analysis 2 months after injury and hCSC injection determined with the ImageJ 1.48 v software from TEM images. Five corneas in each study group with five or more images per cornea were analyzed. At least 300 fibrils were evaluated for each image. (<b>A</b>) Mean distance to the nearest neighbor between fibrils. Data are presented as a means ± SDs (<span class="html-italic">n</span> &gt; 500 in each group). * <span class="html-italic">p</span> = 0.00 compared to the control group (healthy cornea), <sup>#</sup> <span class="html-italic">p</span> = 0.00 compared to the injury group, <sup>!</sup> <span class="html-italic">p</span> = 0.00 compared to the injury + saline group, ^ <span class="html-italic">p</span> = 0.00 compared to the injury + CF group. (<b>B</b>) Distributions of the nearest neighbor interfibrillar distances. Different curves in each frame correspond to calculations from different photographs. Arrows indicate apparent maxima of the distributions. The thick black lines are the averages of the different curves. (<b>C</b>) Fibril pair correlation function <span class="html-italic">g</span>(<span class="html-italic">r</span>). Each symbolized curve corresponds to data from one photograph. From 300 to 500 fibrils were analyzed in each photograph. (<b>D</b>) Averaged pair correlation functions. The arrows show the radii of the circles within which the four closest neighboring fibrils are located.</p>
Full article ">
21 pages, 625 KiB  
Review
TGF-β-Based Therapies for Treating Ocular Surface Disorders
by Fernando T. Ogata, Sudhir Verma, Vivien J. Coulson-Thomas and Tarsis F. Gesteira
Cells 2024, 13(13), 1105; https://doi.org/10.3390/cells13131105 - 26 Jun 2024
Viewed by 1005
Abstract
The cornea is continuously exposed to injuries, ranging from minor scratches to deep traumas. An effective healing mechanism is crucial for the cornea to restore its structure and function following major and minor insults. Transforming Growth Factor-Beta (TGF-β), a versatile signaling molecule that [...] Read more.
The cornea is continuously exposed to injuries, ranging from minor scratches to deep traumas. An effective healing mechanism is crucial for the cornea to restore its structure and function following major and minor insults. Transforming Growth Factor-Beta (TGF-β), a versatile signaling molecule that coordinates various cell responses, has a central role in corneal wound healing. Upon corneal injury, TGF-β is rapidly released into the extracellular environment, triggering cell migration and proliferation, the differentiation of keratocytes into myofibroblasts, and the initiation of the repair process. TGF-β-mediated processes are essential for wound closure; however, excessive levels of TGF-β can lead to fibrosis and scarring, causing impaired vision. Three primary isoforms of TGF-β exist—TGF-β1, TGF-β2, and TGF-β3. Although TGF-β isoforms share many structural and functional similarities, they present distinct roles in corneal regeneration, which adds an additional layer of complexity to understand the role of TGF-β in corneal wound healing. Further, aberrant TGF-β activity has been linked to various corneal pathologies, such as scarring and Peter’s Anomaly. Thus, understanding the molecular and cellular mechanisms by which TGF-β1-3 regulate corneal wound healing will enable the development of potential therapeutic interventions targeting the key molecule in this process. Herein, we summarize the multifaceted roles of TGF-β in corneal wound healing, dissecting its mechanisms of action and interactions with other molecules, and outline its role in corneal pathogenesis. Full article
(This article belongs to the Special Issue Mechanism of Cell Signaling during Eye Development and Diseases)
Show Figures

Figure 1

Figure 1
<p>Extracellular dimeric TGF-β binds to its transmembrane receptor (TGFβR), triggering the transphosphorylation of the receptor. The activated receptor recruits and phosphorylates R-Smads, downstream proteins in the TGF-β signaling pathway. Activated R-Smads migrate to the nucleus, where they interact with other co-activators, forming the transcription complex. Three approaches have been used to disrupt this pathway as follows: (1) Monoclonal antibodies with high affinity and neutralizing capability against TGF-β. (2) Small molecules that inhibit downstream signaling from the receptor to Smads. (3) Antisense oligonucleotides developed against either TGF-β or its receptor to impair this signaling pathway.</p>
Full article ">
Back to TopTop