Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (64)

Search Parameters:
Keywords = myofibroblast apoptosis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
31 pages, 1365 KB  
Review
Magnesium, Zinc and Copper in Lung Fibrosis: A Narrative Review
by Mihai Nechifor, Carmen Lacramioara Zamfir and Cristina Gales
Medicina 2026, 62(1), 10; https://doi.org/10.3390/medicina62010010 - 19 Dec 2025
Viewed by 495
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease with progressive evolution and high mortality. Magnesium, copper and zinc are essential biometals involved in numerous biological processes in all organs of the human body. A lower level of zinc and magnesium and a [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease with progressive evolution and high mortality. Magnesium, copper and zinc are essential biometals involved in numerous biological processes in all organs of the human body. A lower level of zinc and magnesium and a higher cooper/zinc ratio are frequently encountered in patients with idiopathic pulmonary fibrosis but also in other forms of pulmonary fibrosis. These imbalances are involved in the main pathogenic mechanisms of idiopathic pulmonary fibrosis: alveolar epithelial cell lesions, oxidative stress, inflammation, fibroblast and myofibroblast proliferation, mitochondrial activity, excessive extracellular matrix accumulation, high collagen production, alveolar macrophage dysfunctions, and apoptosis. A multitude of experimental and clinical studies have shown the importance of these bivalent cations for the synthesis or activity of some important endogenous active substances (fatty acids, eicosanoids, sirtuin1, p53 protein, interleukins, growth factors, some enzymes, and others) involved in one form or another in the pathogenesis of IPF. There are no randomized clinical trials yet, but some clinical and experimental results suggest that the association of zinc and magnesium with pirfenidone and nintedanib could be beneficial and should be assessed as soon as possible after the onset of this disease. The correction of hypomagnesemia and hypozincemia, whenever they exist, must be performed as soon as possible after the diagnosis of fibrosis. Full article
(This article belongs to the Section Pulmonology)
Show Figures

Figure 1

19 pages, 14660 KB  
Article
Reversal of Myofibroblast Apoptosis Resistance and Collagen Deposition by Phaseoloidin-Induced Autophagy Attenuates Pulmonary Fibrosis
by Siyuan Li, Jiazhen Qian, Lang Deng, Wei Liu, Siyuan Tang and Weixi Xie
Biomedicines 2025, 13(11), 2679; https://doi.org/10.3390/biomedicines13112679 - 31 Oct 2025
Viewed by 739
Abstract
Background and Objectives: Myofibroblast apoptosis resistance and excessive extracellular matrix (ECM) deposition are central drivers of the irreversibility of pulmonary fibrosis, and both are mechanistically linked to autophagy impairment. Phaseoloidin is a bioactive compound derived from Entada phaseoloides. This study aimed [...] Read more.
Background and Objectives: Myofibroblast apoptosis resistance and excessive extracellular matrix (ECM) deposition are central drivers of the irreversibility of pulmonary fibrosis, and both are mechanistically linked to autophagy impairment. Phaseoloidin is a bioactive compound derived from Entada phaseoloides. This study aimed to investigate the therapeutic potential of Phaseoloidin in bleomycin-induced pulmonary fibrosis and its underlying mechanisms. Methods:In vivo, the antifibrotic effects of Phaseoloidin were evaluated using a bleomycin-induced pulmonary fibrosis mouse model in male C57/BL mice. To further elucidate the mechanisms by which Phaseoloidin counteracts fibrosis, in vitro experiments were conducted using primary lung fibroblasts. Results: In vitro experiments showed that Phaseoloidin could activate the AMPK/mTOR pathway in autophagy-deficient myofibroblasts, effectively reversing autophagic defects and promoting collagen degradation. This autophagy activation selectively degraded PTPN13, a negative regulator of apoptosis, thereby enhancing the sensitivity of myofibroblasts to FasL-induced apoptosis and further facilitating fibrosis resolution. After AMPK gene knockout, the pro-autophagic effect of Phaseoloidin completely disappeared, and both collagen clearance and apoptosis recovery were blocked. In vivo experiments confirmed that Phaseoloidin exerted antifibrotic effects by activating AMPK-mediated autophagy in myofibroblasts, which significantly ameliorated pulmonary fibrosis. Conclusions: Phaseoloidin exerts a dual mechanism by activating AMPK-mediated autophagy in myofibroblasts: first, degrading PTPN13 to reverse myofibroblast apoptosis resistance; second, enhancing ECM turnover. These findings indicate that Phaseoloidin is a promising novel therapeutic candidate for pulmonary fibrosis. Full article
Show Figures

Figure 1

26 pages, 5057 KB  
Article
Rapamycin Mitigates Corneal Damage in a Mouse Model of Alkali Burn Injury
by Basanta Bhujel, Woojune Hur, Seorin Lee, Hun Lee, Ho Seok Chung and Jae Yong Kim
Bioengineering 2025, 12(9), 998; https://doi.org/10.3390/bioengineering12090998 - 19 Sep 2025
Viewed by 1314
Abstract
Alkali burns to the cornea cause severe damage characterized by an intense inflammatory response driven by inflammatory cytokines, which orchestrate pathological processes, including neovascularization, fibrosis, apoptosis, abnormal cell proliferation, and disorganization of the extracellular matrix (ECM), often resulting in permanent vision impairment or [...] Read more.
Alkali burns to the cornea cause severe damage characterized by an intense inflammatory response driven by inflammatory cytokines, which orchestrate pathological processes, including neovascularization, fibrosis, apoptosis, abnormal cell proliferation, and disorganization of the extracellular matrix (ECM), often resulting in permanent vision impairment or loss. Rapamycin (RAPA), a well-known mTOR inhibitor with potent immunosuppressive activity and pleiotropic therapeutic effects, was investigated as a novel restorative modality for promoting corneal wound healing in a mouse model of alkali burn injury. Topical RAPA treatment significantly reduced clinical signs of inflammation and decreased the infiltration of F4/80+ macrophages and CD45+ leukocytes, along with suppressed expression of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-17A). RAPA also markedly downregulated angiogenic mediators, such as VEGF, and endothelial markers, like CD31, resulting in significant inhibition of neovascularization. Furthermore, it prevented fibrotic tissue formation and myofibroblast activation, as evidenced by reduced α-SMA levels, and attenuated pathological matrix remodeling through decreased MMP-9 expression. Notably, RAPA preserved epithelial barrier function by maintaining the tight junction protein ZO-1 and reduced both apoptotic cell death (TUNEL) and dysregulated proliferation (Ki67+), thereby preserving the functional and structural integrity of the cornea. In conclusion, RAPA represents a promising therapeutic candidate for managing severe corneal alkali burn injuries, with the potential to enhance corneal wound healing, minimize long-term complications, and protect visual function. Full article
(This article belongs to the Special Issue Bioengineering Strategies for Ophthalmic Diseases)
Show Figures

Graphical abstract

19 pages, 1479 KB  
Article
Blue Light (λ = 453 nm) Significantly Reduces TGF-β-Induced Fibroblast Differentiation Through Reversible Disruption of Mitochondrial Respiration, Glycolysis, and ATP Production Rate
by Pia Steentjes, Julia Krassovka, Christoph V. Suschek, Uwe Maus and Lisa Oezel
Biomedicines 2025, 13(9), 2231; https://doi.org/10.3390/biomedicines13092231 - 10 Sep 2025
Viewed by 916
Abstract
Background/Objectives: Abnormal differentiation of human skin fibroblasts into myofibroblasts contributes to fibrotic skin disorders such as hypertrophic scars, keloids, and Dupuytren’s disease. This process is characterized by increased fibroblast proliferation, enhanced differentiation into myofibroblasts, and reduced programmed cell death (apoptosis). We previously [...] Read more.
Background/Objectives: Abnormal differentiation of human skin fibroblasts into myofibroblasts contributes to fibrotic skin disorders such as hypertrophic scars, keloids, and Dupuytren’s disease. This process is characterized by increased fibroblast proliferation, enhanced differentiation into myofibroblasts, and reduced programmed cell death (apoptosis). We previously demonstrated that blue light irradiation (λ = 453 nm) significantly and dose-dependently inhibits both spontaneous and TGF-β-induced fibroblast differentiation. Methods: Because fibroblast differentiation depends on cellular energy metabolism, we investigated whether the inhibitory effect of blue light is linked to changes in the cells’ energy balance. Results: We found that blue light reduced TGF-β-induced differentiation, as shown by decreased levels of α-SMA and EDA-fibronectin, key markers of myofibroblast formation. This effect was strongly associated with almost complete inhibition of mitochondrial respiration, reduced glycolysis, a lower NAD+/NADH ratio, and decreased ATP production. ATP-dependent processes, including endocytosis and lysosomal activity, both essential parameters of fibroblast differentiation, were also strongly suppressed. Importantly, all these changes were fully reversible within 24 h after the last irradiation. Conclusions: Mechanistically, we propose that blue light triggers photochemical reduction in flavins in proteins of the respiratory chain and possibly the Krebs cycle, which temporarily alters cellular energy metabolism. These findings suggest that non-toxic blue light therapy (80 J/cm2) can effectively prevent factor-induced fibroblast differentiation and may serve as a standalone or supportive treatment to reduce fibrotic events such as scarring and keloid formation. Furthermore, our results indicate that targeting cellular energy metabolism, whether physically or pharmacologically, could be a promising strategy to prevent sclerotic skin disorders. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

13 pages, 7555 KB  
Article
Healed Perforated Corneal Ulcers in Human
by Yasser Helmy Mohamed, Masafumi Uematsu, Mao Kusano, Keiji Suzuki and Akio Oishi
Life 2025, 15(6), 939; https://doi.org/10.3390/life15060939 - 11 Jun 2025
Viewed by 966
Abstract
This study investigates the pathophysiological process of healed perforated corneal ulcers (HPCUs) in humans. All subjects underwent keratoplasty due to opacities or leakage from HPCUs. Half of each specimen was fixed with 4% glutaraldehyde for transmission electron microscope (TEM) examination. The other half [...] Read more.
This study investigates the pathophysiological process of healed perforated corneal ulcers (HPCUs) in humans. All subjects underwent keratoplasty due to opacities or leakage from HPCUs. Half of each specimen was fixed with 4% glutaraldehyde for transmission electron microscope (TEM) examination. The other half was fixed in 10% formaldehyde for immunofluorescence (IF) examination. TEM identified layered structures with two cell types (polygonal and elongated) connected by gap or adherent junctions during early stage of healing. Both apoptotic and mitotic changes were found in both types of cells. There were no endothelial cells or Descemet’s membrane (DM) present in early stage of healing. During the intermediate stage, the healed area comprised three layers: epithelium, Bowman’s layer, and stroma, with an increase in stromal collagen. Later, adjacent endothelial cells crept in, forming DM and completing the cornea’s 5-layer structure. IF examinations revealed that vimentin+ and α-smooth muscle actin (αSMA)+ myofibroblasts gathered around the damaged site. Proliferating cell nuclear antigen+ cells, which indicated cell proliferation, were found in both cells. Anti-phospho-histone H2AX antibodies were found in some epithelial cells. CK14-positive cells were only found in superficial polygonal cells. Corneal wound healing is a complex process that includes apoptosis, cell migration, mitosis, differentiation, and extracellular matrix remodeling. Full article
(This article belongs to the Special Issue Feature Paper in Physiology and Pathology: 2nd Edition)
Show Figures

Figure 1

23 pages, 5473 KB  
Article
Quercetin, Kaempferol and Capsaicin Counteract the TGF-β1-Induced Upregulation of αSMA and Collagen in Myoblasts
by Iris Cuijpers, Mireille M. J. P. E. Sthijns, Veerle A. R. van den Bogart, Joey Katsburg, Cliff F. M. Leenders and Freddy J. Troost
Int. J. Mol. Sci. 2025, 26(11), 5151; https://doi.org/10.3390/ijms26115151 - 27 May 2025
Cited by 2 | Viewed by 2075
Abstract
In fibrotic skeletal muscles, excessive extracellular matrix (ECM) deposition is a result of increased activation and decreased apoptosis of myofibroblasts. The aim of this study is to investigate whether treatment with quercetin, kaempferol or capsaicin can reduce the transforming growth factor-beta 1 (TGF-β1)-induced [...] Read more.
In fibrotic skeletal muscles, excessive extracellular matrix (ECM) deposition is a result of increased activation and decreased apoptosis of myofibroblasts. The aim of this study is to investigate whether treatment with quercetin, kaempferol or capsaicin can reduce the transforming growth factor-beta 1 (TGF-β1)-induced myofibroblast differentiation and fibrotic ECM expression in differentiated C2C12 cells. Two-day-differentiated C2C12 cells were treated with TGF-β1 for 48 h to induce myofibroblast differentiation. Twenty-four hours before (pre-treatment) and for forty-eight hours with (co-treatment) TGF-β1 treatment, cells were exposed to quercetin (25, 50 µM), kaempferol (10, 25, 50 µM) or capsaicin (25, 50 µM). The immunofluorescence intensity of alpha smooth muscle actin (αSMA) and collagen type I/III gene expression were assessed as myofibroblast markers. MyoD immunofluorescence intensity was measured as a myogenic marker. Co-treatment of TGF-β1 with the phytochemicals was most effective, resulting in a decreased number of αSMA-positive cells (all three compounds), decreased collagen type I (kaempferol, capsaicin) and type III (kaempferol) gene expression, and increased MyoD (kaempferol, capsaicin) protein expression compared to TGF-β1 treatment. This study demonstrates that treatment with quercetin, kaempferol or capsaicin can reduce myofibroblast markers. This suggests a possible anti-fibrotic effect of the phytochemicals in skeletal muscle. Full article
(This article belongs to the Special Issue The Effect of Phenolic Compounds in Human Diseases)
Show Figures

Figure 1

16 pages, 13174 KB  
Article
MicroRNA-150 Deletion from Adult Myofibroblasts Augments Maladaptive Cardiac Remodeling Following Chronic Myocardial Infarction
by Satoshi Kawaguchi, Marisa N. Sepúlveda, Jian-peng Teoh, Taiki Hayasaka, Bruno Moukette, Tatsuya Aonuma, Hyun Cheol Roh, Meena S. Madhur and Il-man Kim
Biomolecules 2024, 14(12), 1650; https://doi.org/10.3390/biom14121650 - 22 Dec 2024
Cited by 2 | Viewed by 1682
Abstract
MicroRNA (miR: small noncoding RNA)-150 is evolutionarily conserved and is downregulated in patients with diverse forms of heart failure (HF) and in multiple mouse models of HF. Moreover, miR-150 is markedly correlated with the outcome of patients with HF. We previously reported that [...] Read more.
MicroRNA (miR: small noncoding RNA)-150 is evolutionarily conserved and is downregulated in patients with diverse forms of heart failure (HF) and in multiple mouse models of HF. Moreover, miR-150 is markedly correlated with the outcome of patients with HF. We previously reported that systemic or cardiomyocyte-derived miR-150 in mice elicited myocardial protection through the inhibition of cardiomyocyte death, without affecting neovascularization and T cell infiltration. Our mechanistic studies also showed that the protective roles of miR-150 in ischemic mouse hearts and human cardiac fibroblasts were, in part, attributed to the inhibition of fibroblast activation via the repression of multiple profibrotic genes. However, the extent to which miR-150 expression in adult myofibroblasts (MFs) modulates the response to myocardial infarction (MI) remains unknown. Here, we develop a novel 4-hydroxytamoxifen-inducible MF-specific miR-150 conditional knockout mouse model and demonstrate that the mouse line exhibits worse cardiac dysfunction after MI. Our studies further reveal that miR-150 ablation selectively in adult MFs exacerbates cardiac damage and apoptosis after chronic MI. Lastly, MF-specific miR-150 deletion in adult mice promotes the expression of proinflammatory and profibrotic genes as well as cardiac fibrosis following chronic MI. Our findings indicate a key protective role for MF-derived miR-150 in modulating post-MI responses. Full article
(This article belongs to the Special Issue Heart Diseases: Molecular Mechanisms and New Therapies)
Show Figures

Figure 1

19 pages, 5454 KB  
Article
Evaluation of Antifibrotic Mechanisms of 3′5-Dimaleamylbenzoic Acid on Idiopathic Pulmonary Fibrosis: A Network Pharmacology and Molecular Docking Analysis
by Karina González-García, Jovito Cesar Santos-Álvarez, Juan Manuel Velázquez-Enríquez, Cecilia Zertuche-Martínez, Edilburga Reyes-Jiménez, Rafael Baltiérrez-Hoyos and Verónica Rocío Vásquez-Garzón
Drugs Drug Candidates 2024, 3(4), 860-878; https://doi.org/10.3390/ddc3040048 - 6 Dec 2024
Viewed by 2849
Abstract
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic, disabling disorder of unknown etiology, poor prognosis, and limited therapeutic options. Previously, 3′5-dimaleamylbenzoic acid (3′5-DMBA) was shown to exert resolving effects in IPF, offering a promising alternative for treating this disease; however, the molecular mechanisms [...] Read more.
Background: Idiopathic pulmonary fibrosis (IPF) is a chronic, disabling disorder of unknown etiology, poor prognosis, and limited therapeutic options. Previously, 3′5-dimaleamylbenzoic acid (3′5-DMBA) was shown to exert resolving effects in IPF, offering a promising alternative for treating this disease; however, the molecular mechanisms associated with this effect have not been explored. Objetive: We evaluated the potential antifibrotic mechanisms of 3′5-DMBA by network pharmacology (NP) and molecular docking (MD). Methods: 3′5-DMBA-associated targets were identified by screening in SwissTargetPrediction. IPF-associated targets were identified using lung tissue meta-analysis and public databases. Common targets were identified, and a protein–protein interaction (PPI) network was constructed; we ranked the proteins in the PPI network by topological analysis. MD validated the binding of 3′5-DMBA to the main therapeutic targets. Results: A total of 57 common targets were identified between 3′5-DMBA and IPF; caspase 8, 9, 3, and 7; myeloid leukemia-induced cell differentiation protein Mcl-1; and poly [ADP-ribose] polymerase 1 are primary targets regulating PPI networks. Functional analysis revealed that the common targets are involved in the pathological features of tissue fibrosis and primarily in the apoptotic process. MD revealed favorable interaction energies among the three main targets regulating PPI networks. Conclusions: NP results suggest that the antifibrotic effect of 3′5-DMBA is due to its regulation of the pathological features of IPF, mainly by modulating signaling pathways leading to apoptosis, suggesting its therapeutic potential to treat this disease. Full article
(This article belongs to the Section In Silico Approaches in Drug Discovery)
Show Figures

Figure 1

25 pages, 4278 KB  
Article
Exposure of Primary Human Skin Fibroblasts to Carbon Dioxide-Containing Solution Significantly Reduces TGF-β-Induced Myofibroblast Differentiation In Vitro
by Maxine Fleckner, Niklas K. Döhmen, Katharina Salz, Till Christophers, Joachim Windolf, Christoph V. Suschek and Lisa Oezel
Int. J. Mol. Sci. 2024, 25(23), 13013; https://doi.org/10.3390/ijms252313013 - 3 Dec 2024
Cited by 5 | Viewed by 2452
Abstract
Wound healing as a result of a skin injury involves a series of dynamic physiological processes, leading to wound closure, re-epithelialization, and the remodeling of the extracellular matrix (ECM). The primary scar formed by the new ECM never fully regains the original tissue’s [...] Read more.
Wound healing as a result of a skin injury involves a series of dynamic physiological processes, leading to wound closure, re-epithelialization, and the remodeling of the extracellular matrix (ECM). The primary scar formed by the new ECM never fully regains the original tissue’s strength or flexibility. Moreover, in some cases, due to dysregulated fibroblast activity, proliferation, and differentiation, the normal scarring can be replaced by pathological fibrotic tissue, leading to hypertrophic scars or keloids. These disorders can cause significant physical impairment and psychological stress and represent significant challenges in medical management in the wound-healing process. The present study aimed to investigate the therapeutic effects of exogenously applied carbon dioxide (CO2) on fibroblast behavior, focusing on viability, proliferation, migration, and differentiation to myofibroblasts. We found that CO2 exposure for up to 60 min did not significantly affect fibroblast viability, apoptosis rate, or proliferation and migration capacities. However, a notable finding was the significant reduction in α-smooth muscle actin (α-SMA) protein expression, indicative of myofibroblast differentiation inhibition, following CO2 exposure. This effect was specific to CO2 and concentration as well as time-dependent, with longer exposure durations leading to greater reductions in α-SMA expression. Furthermore, the inhibition of myofibroblast differentiation correlated with a statistically significantly reduced glycolytic and mitochondrial energy metabolism, and as a result, with a reduced ATP synthesis rate. This very noticeable decrease in cellular energy levels seemed to be specific to CO2 exposure and could not be observed in the control cultures using nitrogen (N2)-saturated solutions, indicating a unique and hypoxia-independent effect of CO2 on fibroblast metabolism. These findings suggest that exogenously applied CO2 may possess fibroblast differentiation-reducing properties by modulating fibroblast’s energy metabolism and could offer new therapeutic options in the prevention of scar and keloid development. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

21 pages, 2580 KB  
Article
p53 and the E3 Ubiquitin Ligase MDM2 in Glaucomatous Lamina Cribrosa Cells
by Kealan McElhinney, Mustapha Irnaten, Jeffrey O’Callaghan and Colm O’Brien
Int. J. Mol. Sci. 2024, 25(22), 12173; https://doi.org/10.3390/ijms252212173 - 13 Nov 2024
Viewed by 1486
Abstract
Lamina cribrosa (LC) cells play an integral role in extracellular matrix remodeling and fibrosis in human glaucoma. LC cells bear similarities to myofibroblasts that adopt an apoptotic-resistant, proliferative phenotype, a process linked to dysregulation of tumor suppressor-gene p53 pathways, including ubiquitin-proteasomal degradation via [...] Read more.
Lamina cribrosa (LC) cells play an integral role in extracellular matrix remodeling and fibrosis in human glaucoma. LC cells bear similarities to myofibroblasts that adopt an apoptotic-resistant, proliferative phenotype, a process linked to dysregulation of tumor suppressor-gene p53 pathways, including ubiquitin-proteasomal degradation via murine-double-minute-2 (MDM2). Here, we investigate p53 and MDM2 in glaucomatous LC cells. Primary human LC cells were isolated from glaucomatous donor eyes (GLC) and age-matched normal controls (NLC) (n = 3 donors/group). LC cells were cultured under standard conditions ± 48-h treatment with p53-MDM2-interaction inhibitor RG-7112. Markers of p53-MDM2, fibrosis, and apoptosis were analyzed by real-time polymerase chain reaction (qRT-PCR), western blotting, and immunofluorescence. Cellular proliferation and viability were assessed using colorimetric methyl-thiazolyl-tetrazolium salt assays (MTS/MTT). In GLC versus NLC cells, protein expression of p53 was significantly decreased (p < 0.05), MDM2 was significantly increased, and immunofluorescence showed reduced p53 and increased MDM2 expression in GLC nuclei. RG-7112 treatment significantly increased p53 and significantly decreased MDM2 gene and protein expression. GLC cells had significantly increased protein expression of αSMA, significantly decreased caspase-3 protein expression, and significantly increased proliferation after 96 h. RG-7112 treatment significantly decreased COL1A1 and αSMA, significantly increased BAX and caspase-3 gene expression, and significantly decreased proliferation in GLC cells. MTT-assay showed equivocal cellular viability in NLC/GLC cells with/without RG-7112 treatment. Our data suggests that proliferation and the ubiquitin-proteasomal pathway are dysregulated in GLC cells, with MDM2-led p53 protein degradation negatively impacting its protective role. Full article
(This article belongs to the Special Issue Epigenetic Control of Apoptosis)
Show Figures

Figure 1

14 pages, 882 KB  
Review
Hedgehog Signalling Pathway and Its Role in Shaping the Architecture of Intestinal Epithelium
by Adrianna Konopka, Kamil Gawin and Marcin Barszcz
Int. J. Mol. Sci. 2024, 25(22), 12007; https://doi.org/10.3390/ijms252212007 - 8 Nov 2024
Cited by 5 | Viewed by 3562
Abstract
The hedgehog (Hh) signalling pathway plays a key role in both embryonic and postnatal development of the intestine and is responsible for gut homeostasis. It regulates stem cell renewal, formation of the villous–crypt axis, differentiation of goblet and Paneth cells, the cell cycle, [...] Read more.
The hedgehog (Hh) signalling pathway plays a key role in both embryonic and postnatal development of the intestine and is responsible for gut homeostasis. It regulates stem cell renewal, formation of the villous–crypt axis, differentiation of goblet and Paneth cells, the cell cycle, apoptosis, development of gut innervation, and lipid metabolism. Ligands of the Hh pathway, i.e., Indian hedgehog (Ihh) and Sonic hedgehog (Shh), are expressed by superficial enterocytes but act in the mesenchyme, where they are bound by a Patched receptor localised on myofibroblasts and smooth muscle cells. This activates a cascade leading to the transcription of target genes, including those encoding G1/S-specific cyclin-D2 and -E1, B-cell lymphoma 2, fibroblast growth factor 4, and bone morphogenetic protein 4. The Hh pathway is tightly connected to Wnt signalling. Ihh is the major ligand in the Hh pathway. Its activation inhibits proliferation, while its blocking induces hyperproliferation and triggers a wound-healing response. Thus, Ihh is a negative feedback regulator of cell proliferation. There are data indicating that diet composition may affect the expression of the Hh pathway genes and proteins, which in turn, induces changes in mucosal architecture. This was shown for fat, vitamin A, haem, berberine, and ovotransferrin. The Hh signalling is also affected by the intestinal microbiota, which affects the intestinal barrier integrity. This review highlights the critical importance of the Hh pathway in shaping the intestinal mucosa and summarises the results obtained so far in research on the effect of dietary constituents on the activity of this pathway. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

19 pages, 1700 KB  
Review
Ca2+ Signaling in Cardiovascular Fibroblasts
by Andreas Rinne and Florentina Pluteanu
Biomolecules 2024, 14(11), 1365; https://doi.org/10.3390/biom14111365 - 27 Oct 2024
Cited by 3 | Viewed by 3116
Abstract
Fibrogenesis is a physiological process required for wound healing and tissue repair. It is induced by activation of quiescent fibroblasts, which first proliferate and then change their phenotype into migratory, contractile myofibroblasts. Myofibroblasts secrete extracellular matrix proteins, such as collagen, to form a [...] Read more.
Fibrogenesis is a physiological process required for wound healing and tissue repair. It is induced by activation of quiescent fibroblasts, which first proliferate and then change their phenotype into migratory, contractile myofibroblasts. Myofibroblasts secrete extracellular matrix proteins, such as collagen, to form a scar. Once the healing process is terminated, most myofibroblasts undergo apoptosis. However, in some tissues, such as the heart, myofibroblasts remain active and sensitive to neurohumoral factors and inflammatory mediators, which lead eventually to excessive organ fibrosis. Many cellular processes involved in fibroblast activation, including cell proliferation, protein secretion and cell contraction, are highly regulated by intracellular Ca2+ signals. This review summarizes current research on Ca2+ signaling pathways underlying fibroblast activation. We present receptor- and ion channel-mediated Ca2+ signaling pathways, discuss how localized Ca2+ signals of the cell nucleus may be involved in fibroblast activation and present Ca2+-sensitive transcription pathways relevant for fibroblast biology. When investigated, we highlight how the function of Ca2+-handling proteins changes during cardiac and pulmonary fibrosis. Many aspects of Ca2+ signaling remain unexplored in different types of cardiovascular fibroblasts in relation to pathologies, and a better understanding of Ca2+ signaling in fibroblasts will help to design targeted therapies against fibrosis. Full article
(This article belongs to the Special Issue Advances in Cellular Biophysics: Transport and Mechanics)
Show Figures

Figure 1

15 pages, 1345 KB  
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
Cited by 16 | Viewed by 4997
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

20 pages, 951 KB  
Review
Recent Insights into Cellular and Molecular Mechanisms of Defective Angiogenesis in Systemic Sclerosis
by Eloisa Romano, Irene Rosa, Bianca Saveria Fioretto and Mirko Manetti
Biomedicines 2024, 12(6), 1331; https://doi.org/10.3390/biomedicines12061331 - 14 Jun 2024
Cited by 10 | Viewed by 2834
Abstract
In systemic sclerosis (SSc, or scleroderma), defective angiogenesis, clinically manifesting with abnormal capillary architecture and severe capillary reduction, represents a hallmark of early-stage disease, usually preceding the onset of tissue fibrosis, and is caused by several cellular and molecular mechanisms affecting microvascular endothelial [...] Read more.
In systemic sclerosis (SSc, or scleroderma), defective angiogenesis, clinically manifesting with abnormal capillary architecture and severe capillary reduction, represents a hallmark of early-stage disease, usually preceding the onset of tissue fibrosis, and is caused by several cellular and molecular mechanisms affecting microvascular endothelial cells with different outcomes. Indeed, once damaged, endothelial cells can be dysfunctionally activated, thus becoming unable to undergo angiogenesis and promoting perivascular inflammation. They can also undergo apoptosis, transdifferentiate into profibrotic myofibroblasts, or acquire a senescence-associated secretory phenotype characterized by the release of exosomes and several profibrotic and proinflammatory mediators. In this narrative review, we aimed to give a comprehensive overview of recent studies dealing with the cellular and molecular mechanisms underlying SSc defective angiogenesis and the related endothelial cell dysfunctions, mainly the endothelial-to-mesenchymal transition process. We also discussed potential novel vascular treatment strategies able to restore the angiogenic process and reduce the endothelial-to-mesenchymal transition in this complex disease. Full article
(This article belongs to the Special Issue Angiogenesis)
Show Figures

Figure 1

23 pages, 11147 KB  
Article
Comparison between the Regenerative and Therapeutic Impacts of Bone Marrow Mesenchymal Stem Cells and Adipose Mesenchymal Stem Cells Pre-Treated with Melatonin on Liver Fibrosis
by Ahmed Elzainy, Abir El Sadik and Waleed Mohammad Altowayan
Biomolecules 2024, 14(3), 297; https://doi.org/10.3390/biom14030297 - 1 Mar 2024
Cited by 12 | Viewed by 4176
Abstract
Background: The distinctive feature of liver fibrosis is the progressive replacement of healthy hepatic cells by the extracellular matrix protein, which is abundant in collagen I and III, with impaired matrix remodeling. The activation of myofibroblastic cells enhances the fibrogenic response of complex [...] Read more.
Background: The distinctive feature of liver fibrosis is the progressive replacement of healthy hepatic cells by the extracellular matrix protein, which is abundant in collagen I and III, with impaired matrix remodeling. The activation of myofibroblastic cells enhances the fibrogenic response of complex interactions of hepatic stellate cells, fibroblasts, and inflammatory cells to produce the excessive deposition of the extracellular protein matrix. This process is activated by multiple fibrogenic mediators and cytokines, such as TNF-α and IL-1β, accompanied with a decrease in the anti-fibrogenic factor NF-κβ. Mesenchymal stem cells (MSCs) represent a promising therapy for liver fibrosis, allowing for a more advanced regenerative influence when cultured with extrinsic or intrinsic proliferative factors, cytokines, antioxidants, growth factors, and hormones such as melatonin (MT). However, previous studies showed conflicting findings concerning the therapeutic effects of adipose (AD) and bone marrow (BM) MSCs; therefore, the present work aimed to conduct a comparative and comprehensive study investigating the impact of MT pre-treatment on the immunomodulatory, anti-inflammatory, and anti-apoptotic effects of AD- and BM-MSCs and to critically analyze whether MT-pre-treated AD-MSCs and BM-MSCs reveal equal or different therapeutic and regenerative potentials in a CCl4-injured liver experimental rat model. Materials and methods: Six groups of experimental rats were used, with ten rats in each group: group I (control group), group II (CCl4-treated group), group III (CCl4- and BM-MSC-treated group), group IV (CCl4 and MT-pre-treated BM-MSC group), group V (CCl4- and AD-MSC-treated group), and group VI (CCl4 and MT-pre-treated AD-MSC group). Liver function tests and the gene expression of inflammatory, fibrogenic, apoptotic, and proliferative factors were analyzed. Histological and immunohistochemical changes were assessed. Results: The present study compared the ability of AD- and BM-MSCs, with and without MT pre-treatment, to reduce hepatic fibrosis. Both types of MSCs improved hepatocyte function by reducing the serum levels of ALT, aspartate aminotransferase (AST), alkaline phosphatase (AKP), and total bilirubin (TBIL). In addition, the changes in the hepatocellular architecture, including the hepatocytes, liver sinusoids, central veins, portal veins, biliary ducts, and hepatic arteries, showed a decrease in hepatocyte injury and cholestasis with a reduction in inflammation, apoptosis, and necrosis of the hepatic cells, together with an inhibition of liver tissue fibrosis. These results were augmented by an analysis of the expression of the pro-inflammatory cytokines TNFα and IL-1β, the anti-fibrogenic factor NF-κβ, the apoptotic factor caspase-3, and the proliferative indicators antigen Ki-67 and proliferating cell nuclear antigen (PCNA). These findings were found to be statistically significant, with the restoration of normal parameters in the rats that received AD-MSCs pre-treated with MT, denoting optimal regenerative and therapeutic effects. Conclusions: AD-MSCs pre-treated with MT are the preferred choice in improving hepatic fibrosis and promoting the therapeutic and regenerative ability of liver tissue. They represent a very significant tool for future stem cell use in the tissue regeneration strategy for the treatment of liver diseases. Full article
(This article belongs to the Special Issue Recent Developments in Mesenchymal Stem Cells)
Show Figures

Figure 1

Back to TopTop