Molecular Mechanisms in Organ Fibrosis

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: closed (10 November 2021) | Viewed by 42149

Special Issue Editors


E-Mail
Guest Editor
Department of Biomedical Sciences, University of Padova, 35100 Padua, Italy
Interests: extra-cellular matrix (ECM) turn-over and remodeling; matrix–metalloproteinases (MMPs) and TIMPs; heparanase; tumor invasiveness and metastasis; epithelial to mesenchymal transition (EMT); SPATA2 (spermatogenesis-associated protein 2)
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
1. Department of Biomedical Sciences, University of Padova, 35100 Padua, Italy
2. Division of Nephrology and Dialysis, Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126 Verona, Italy
Interests: heparanase; organ fibrosis; inflammation; renal physiology; renal pathology; acute and chronic renal injury; epithelial to mesenchymal transition (EMT); molecular mechanisms in cancer progression
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear colleagues,

Fibrosis is a dynamic process which is mainly characterized by an excessive accumulation of extracellular matrix. Fibrosis generally occurs as an uncontrolled wound-healing response to multiple and chronic injuries. It induces the alteration of organ architecture and leads to loss of organ function. Fibrosis is a pathology that affects several organs such as the kidney, lungs, liver, skin, peritoneum, and heart. In addition, even if the main source of the extracellular matrix is activated myofibroblasts, a great many cell types modulate the phenomenon (epithelial and mesothelial cells, endothelial cells, pericytes, vascular smooth muscle cells, inflammatory cells, and mesenchymal stem cells). Commonly, the fibrotic process is initiated by parenchymal damage followed by an unsolved inflammation, and thus, some molecular mechanisms at the origin of fibrosis are shared by different organs. In this scenario, additional tissue-specific mechanisms are responsible for fibrosis. Keeping in mind that in certain situations, fibrosis could be a reversible process, a deeper knowledge of its foundation is highly necessary in order to develop new therapeutic strategies.

The aim of this Special Issue of Cells is to report new findings that advance knowledge of the molecular mechanisms of organ fibrosis. We welcome submissions of research papers and reviews that focus on aspects of known mechanisms/pathways such as epithelial–mesenchymal transition, TGF-beta signaling, myofibroblast activation, proliferation, and senescence, but also manuscripts exploring new aspects.

Prof. Maurizio Onisto
Dr. Valentina Masola
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Published Papers (11 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

20 pages, 6332 KiB  
Article
Oncofetal Protein CRIPTO Is Involved in Wound Healing and Fibrogenesis in the Regenerating Liver and Is Associated with the Initial Stages of Cardiac Fibrosis
by Sofia Karkampouna, Danny van der Helm, Mario Scarpa, Bart van Hoek, Hein W. Verspaget, Marie-Jose Goumans, Minneke J. Coenraad, Boudewijn P.T. Kruithof and Marianna Kruithof-de Julio
Cells 2021, 10(12), 3325; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10123325 - 26 Nov 2021
Cited by 3 | Viewed by 2944
Abstract
Oncofetal protein, CRIPTO, is silenced during homeostatic postnatal life and often re-expressed in different neoplastic processes, such as hepatocellular carcinoma. Given the reactivation of CRIPTO in pathological conditions reported in various adult tissues, the aim of this study was to explore whether CRIPTO [...] Read more.
Oncofetal protein, CRIPTO, is silenced during homeostatic postnatal life and often re-expressed in different neoplastic processes, such as hepatocellular carcinoma. Given the reactivation of CRIPTO in pathological conditions reported in various adult tissues, the aim of this study was to explore whether CRIPTO is expressed during liver fibrogenesis and whether this is related to the disease severity and pathogenesis of fibrogenesis. Furthermore, we aimed to identify the impact of CRIPTO expression on fibrogenesis in organs with high versus low regenerative capacity, represented by murine liver fibrogenesis and adult murine heart fibrogenesis. Circulating CRIPTO levels were measured in plasma samples of patients with cirrhosis registered at the waitlist for liver transplantation (LT) and 1 year after LT. The expression of CRIPTO and fibrotic markers (αSMA, collagen type I) was determined in human liver tissues of patients with cirrhosis (on a basis of viral hepatitis or alcoholic disease), in cardiac tissue samples of patients with end-stage heart failure, and in mice with experimental liver and heart fibrosis using immuno-histochemical stainings and qPCR. Mouse models with experimental chronic liver fibrosis, induced with multiple shots of carbon tetrachloride (CCl4) and acute liver fibrosis (one shot of CCl4), were evaluated for CRIPTO expression and fibrotic markers. CRIPTO was overexpressed in vivo (Adenoviral delivery) or functionally sequestered by ALK4Fc ligand trap in the acute liver fibrosis mouse model. Murine heart tissues were evaluated for CRIPTO and fibrotic markers in three models of heart injury following myocardial infarction, pressure overload, and ex vivo induced fibrosis. Patients with end-stage liver cirrhosis showed elevated CRIPTO levels in plasma, which decreased 1 year after LT. Cripto expression was observed in fibrotic tissues of patients with end-stage liver cirrhosis and in patients with heart failure. The expression of CRIPTO in the liver was found specifically in the hepatocytes and was positively correlated with the Model for End-stage Liver Disease (MELD) score for end-stage liver disease. CRIPTO expression in the samples of cardiac fibrosis was limited and mostly observed in the interstitial cells. In the chronic and acute mouse models of liver fibrosis, CRIPTO-positive cells were observed in damaged liver areas around the central vein, which preceded the expression of αSMA-positive stellate cells, i.e., mediators of fibrosis. In the chronic mouse models, the fibrosis and CRIPTO expression were still present after 11 weeks, whereas in the acute model the liver regenerated and the fibrosis and CRIPTO expression resolved. In vivo overexpression of CRIPTO in this model led to an increase in fibrotic markers, while blockage of CRIPTO secreted function inhibited the extent of fibrotic areas and marker expression (αSMA, Collagen type I and III) and induced higher proliferation of residual healthy hepatocytes. CRIPTO expression was also upregulated in several mouse models of cardiac fibrosis. During myocardial infarction CRIPTO is upregulated initially in cardiac interstitial cells, followed by expression in αSMA-positive myofibroblasts throughout the infarct area. After the scar formation, CRIPTO expression decreased concomitantly with the αSMA expression. Temporal expression of CRIPTO in αSMA-positive myofibroblasts was also observed surrounding the coronary arteries in the pressure overload model of cardiac fibrosis. Furthermore, CRIPTO expression was upregulated in interstitial myofibroblasts in hearts cultured in an ex vivo model for cardiac fibrosis. Our results are indicative for a functional role of CRIPTO in the induction of fibrogenesis as well as a potential target in the antifibrotic treatments and stimulation of tissue regeneration. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

14 pages, 2890 KiB  
Article
Pulmonary Alveolar Stem Cell Senescence, Apoptosis, and Differentiation by p53-Dependent and -Independent Mechanisms in Telomerase-Deficient Mice
by Kexiong Zhang, Lihui Wang, Xiaojing Hong, Hao Chen, Yao Shi, Yingying Liu, Jun Liu and Jun-Ping Liu
Cells 2021, 10(11), 2892; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10112892 - 26 Oct 2021
Cited by 5 | Viewed by 2613
Abstract
Pulmonary premature ageing and fibrogenesis as in idiopathic pulmonary fibrosis (IPF) occur with the DNA damage response in lungs deficient of telomerase. The molecular mechanism mediating pulmonary alveolar cell fates remains to be investigated. The present study shows that naturally occurring ageing is [...] Read more.
Pulmonary premature ageing and fibrogenesis as in idiopathic pulmonary fibrosis (IPF) occur with the DNA damage response in lungs deficient of telomerase. The molecular mechanism mediating pulmonary alveolar cell fates remains to be investigated. The present study shows that naturally occurring ageing is associated with the DNA damage response (DDR) and activation of the p53 signalling pathway. Telomerase deficiency induced by telomerase RNA component (TERC) knockout (KO) accelerates not only replicative senescence but also altered differentiation and apoptosis of the pulmonary alveolar stem cells (AEC2) in association with increased innate immune natural killer (NK) cells in TERC KO mice. TERC KO results in increased senescence-associated heterochromatin foci (SAHF) marker HP1γ, p21, p16, and apoptosis-associated cleaved caspase-3 in AEC2. However, additional deficiency of the tumour suppressor p53 in the Trp53−/− allele of the late generation of TERC KO mice attenuates the increased senescent and apoptotic markers significantly. Moreover, p53 deficiency has no significant effect on the increased gene expression of T1α (a marker of terminal differentiated AEC1) in AEC2 of the late generation of TERC KO mice. These findings demonstrate that, in natural ageing or premature ageing accelerated by telomere shortening, pulmonary senescence and IPF develop with alveolar stem cell p53-dependent premature replicative senescence, apoptosis, and p53-independent differentiation, resulting in pulmonary senescence-associated low-grade inflammation (SALI). Our studies indicate a natural ageing-associated molecular mechanism of telomerase deficiency-induced telomere DDR and SALI in pulmonary ageing and IPF. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

14 pages, 3051 KiB  
Article
IPF-Fibroblast Erk1/2 Activity Is Independent from microRNA Cluster 17-92 but Can Be Inhibited by Treprostinil through DUSP1
by Sabrina Blumer, Lei Fang, Wei-Chih Chen, Petra Khan, Katrin Hostettler, Michael Tamm, Michael Roth and Christopher Lambers
Cells 2021, 10(11), 2836; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10112836 - 21 Oct 2021
Cited by 9 | Viewed by 2215
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive terminal lung disease, and therapies aim to block fibrosis. Fibroblast proliferation is controlled by C/EBP-β, microRNA cluster 17-92 (miR17-92), and Erk1/2 mitogen-activated protein kinase. This study assessed the role of miR17-92 in IPF-fibroblast proliferation and its [...] Read more.
Idiopathic pulmonary fibrosis (IPF) is a progressive terminal lung disease, and therapies aim to block fibrosis. Fibroblast proliferation is controlled by C/EBP-β, microRNA cluster 17-92 (miR17-92), and Erk1/2 mitogen-activated protein kinase. This study assessed the role of miR17-92 in IPF-fibroblast proliferation and its modification by treprostinil. Fibroblasts were isolated from eight IPF patients, five interstitial lung fibrosis patients, and seven control lungs. Fibroblasts were stimulated with TGF-β1 over 24 h. The miR17-92 expression was analyzed by RT-qPCR, and protein expression by Western blotting. TGF-β1 upregulated C/EBP-β in all fibroblasts, which was reduced by treprostinil in control-fibroblasts, but not in IPF-fibroblasts. Compared to controls, the guide strands miR-19a-3p, miR-19b-3p, miR-20a-5p, and miR-92a-3p, as well as the passenger strands miR-17-3p, miR-18-3p, miR-19a-1-5p, and miR-92a-5p were significantly increased in IPF-fibroblasts. In controls, TGF-β1 and treprostinil significantly reduced specific miR17-92 members. IPF-fibroblast proliferation was inhibited by treprostinil through increased expression of the Erk1/2 inhibitor DUSP1. These data suggest that proliferation control via miR17-92 and C/EBP-β is disrupted in IPF-fibroblasts. Therefore, the inhibition of early stages of signaling cascades or specific mitogen receptors might be less effective. However, the increased proliferation is sensitive to Erk1/2 inhibition by treprostinil-induced DUSP1. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Graphical abstract

14 pages, 4138 KiB  
Article
Indole-3-Carboxaldehyde Restores Gut Mucosal Integrity and Protects from Liver Fibrosis in Murine Sclerosing Cholangitis
by Fiorella D’Onofrio, Giorgia Renga, Matteo Puccetti, Marilena Pariano, Marina Maria Bellet, Ilaria Santarelli, Claudia Stincardini, Paolo Mosci, Maurizio Ricci, Stefano Giovagnoli, Claudio Costantini and Luigina Romani
Cells 2021, 10(7), 1622; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10071622 - 29 Jun 2021
Cited by 27 | Viewed by 3904
Abstract
Primary sclerosing cholangitis (PSC) is a long-term liver disease characterized by a progressive course of cholestasis with liver inflammation and fibrosis. Intestinal barrier dysfunction has been implicated in the pathogenesis of PSC. According to the “leaky gut” hypothesis, gut inflammation alters the permeability [...] Read more.
Primary sclerosing cholangitis (PSC) is a long-term liver disease characterized by a progressive course of cholestasis with liver inflammation and fibrosis. Intestinal barrier dysfunction has been implicated in the pathogenesis of PSC. According to the “leaky gut” hypothesis, gut inflammation alters the permeability of the intestinal mucosa, with the translocation of gut-derived products that enter the enterohepatic circulation and cause hepatic inflammation. Thus, the administration of molecules that preserve epithelial barrier integrity would represent a promising therapeutic strategy. Indole-3-carboxaldehyde (3-IAld) is a microbial-derived product working at the interface between the host and the microbiota and is able to promote mucosal immune homeostasis in a variety of preclinical settings. Herein, by resorting to a murine model of PSC, we found that 3-IAld formulated for localized delivery in the gut alleviates hepatic inflammation and fibrosis by modulating the intestinal microbiota and activating the aryl hydrocarbon receptor-IL-22 axis to restore mucosal integrity. This study points to the therapeutic potential of 3-IAld in liver pathology. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Graphical abstract

19 pages, 11654 KiB  
Article
Eritoran Attenuates Hepatic Inflammation and Fibrosis in Mice with Chronic Liver Injury
by Yun-Cheng Hsieh, Kuei-Chuan Lee, Pei-Shan Wu, Teh-Ia Huo, Yi-Hsiang Huang, Ming-Chih Hou and Han-Chieh Lin
Cells 2021, 10(6), 1562; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061562 - 21 Jun 2021
Cited by 11 | Viewed by 3054
Abstract
Toll-like receptor 4 (TLR4) signaling plays a key role in liver inflammation and fibrosis. The therapeutic effects of eritoran, a TLR4 antagonist, in mice with chronic liver injury remained unclear. C57BL/6 mice were fed a fast-food diet (FFD) or treated with carbon tetrachloride [...] Read more.
Toll-like receptor 4 (TLR4) signaling plays a key role in liver inflammation and fibrosis. The therapeutic effects of eritoran, a TLR4 antagonist, in mice with chronic liver injury remained unclear. C57BL/6 mice were fed a fast-food diet (FFD) or treated with carbon tetrachloride (CCl4) to induce chronic liver injury. Eritoran (10 mg/kg) or a vehicle was randomly intraperitoneally administered to the FFD-fed mice and the CCl4-injured mice. Primary mouse liver cells were cultured with lipopolysaccharide (LPS) or eritoran. In both FFD and CCl4 mouse models, eritoran significantly reduced serum ALT levels and decreased hepatic inflammatory cell infiltration without altering hepatic steatosis. Additionally, eritoran attenuated liver fibrosis by decreasing hepatic stellate cells (HSCs) activation and the abundance of α-smooth muscle actin and transforming growth factor-β1. Hepatic TLR4 downstream signaling including MyD88 expression, NF-κB p65 nuclear translocation, p38 and JNK phosphorylation were successfully inhibited by eritoran. In the in vitro study, LPS-induced nuclear translocation of NF-κB in primary HSCs and Kupffer cells was significantly suppressed by eritoran. In conclusion, eritoran attenuated hepatic inflammation and fibrosis by inhibition of the TLR4 signaling pathway in mice with chronic liver injury. Eritoran may serve as a potential drug for chronic liver disease. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Graphical abstract

13 pages, 273 KiB  
Article
Lack of Relationship between Fibrosis-Related Biomarkers and Cardiac Magnetic Resonance-Assessed Replacement and Interstitial Fibrosis in Dilated Cardiomyopathy
by Paweł Rubiś, Ewa Dziewięcka, Magdalena Szymańska, Robert Banyś, Małgorzata Urbańczyk-Zawadzka, Maciej Krupiński, Małgorzata Mielnik, Sylwia Wiśniowska-Śmiałek, Aleksandra Karabinowska, Piotr Podolec, Mateusz Winiarczyk, Matylda Gliniak, Monika Kaciczak, Jan Robak, Arman Karapetyan and Ewa Wypasek
Cells 2021, 10(6), 1295; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10061295 - 23 May 2021
Cited by 4 | Viewed by 2470
Abstract
The relationship between circulating fibrosis-related molecules and magnetic resonance-assessed cardiac fibrosis in dilated cardiomyopathy (DCM) is poorly understood. To compare circulating biomarkers between DCM patients with high and low fibrosis burdens, we performed a prospective, single-center, observational study. The study population was composed [...] Read more.
The relationship between circulating fibrosis-related molecules and magnetic resonance-assessed cardiac fibrosis in dilated cardiomyopathy (DCM) is poorly understood. To compare circulating biomarkers between DCM patients with high and low fibrosis burdens, we performed a prospective, single-center, observational study. The study population was composed of 100 DCM patients (87 male, mean age 45.2 ± 11.8 years, mean ejection fraction 29.7% ± 10.1%). Replacement fibrosis was quantified by means of late gadolinium enhancement (LGE), whereas interstitial fibrosis was assessed via extracellular volume (ECV). Plasma concentrations of cardiotrophin-1, growth differentiation factor-15, platelet-derived growth factor, procollagen I C-terminal propeptide, procollagen III N-terminal propeptide, and C-terminal telopeptide of type I collagen were measured. There were 44% patients with LGE and the median ECV was 27.7%. None of analyzed fibrosis serum biomarkers were associated with the LGE or ECV, whereas NT-proBNP was independently associated with both LGE and ECV, and troponin T was associated with ECV. None of the circulating fibrosis markers differentiated between DCM patients with and without replacement fibrosis, or patients stratified according to median ECV. However, cardiac-specific markers, such as NT-proBNP and hs-TnT, were associated with fibrosis. Levels of circulating markers of fibrosis seem to have no utility in the diagnosis and monitoring of cardiac fibrosis in DCM. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Graphical abstract

23 pages, 4585 KiB  
Article
Modulation of microRNome by Human Cytomegalovirus and Human Herpesvirus 6 Infection in Human Dermal Fibroblasts: Possible Significance in the Induction of Fibrosis in Systemic Sclerosis
by Irene Soffritti, Maria D’Accolti, Gloria Ravegnini, Maria-Cristina Arcangeletti, Clara Maccari, Flora De Conto, Adriana Calderaro and Elisabetta Caselli
Cells 2021, 10(5), 1060; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10051060 - 29 Apr 2021
Cited by 10 | Viewed by 2481
Abstract
Human cytomegalovirus (HCMV) and Human herpesvirus 6 (HHV-6) have been reportedly suggested as triggers of the onset and/or progression of systemic sclerosis (SSc), a severe autoimmune disorder characterized by multi-organ fibrosis. The etiology and pathogenesis of SSc are still largely unknown but virological [...] Read more.
Human cytomegalovirus (HCMV) and Human herpesvirus 6 (HHV-6) have been reportedly suggested as triggers of the onset and/or progression of systemic sclerosis (SSc), a severe autoimmune disorder characterized by multi-organ fibrosis. The etiology and pathogenesis of SSc are still largely unknown but virological and immunological observations support a role for these beta-herpesviruses, and we recently observed a direct impact of HCMV and HHV-6 infection on the expression of cell factors associated with fibrosis at the cell level. Since miRNA expression has been found profoundly deregulated at the tissue level, here we aimed to investigate the impact on cell microRNome (miRNome) of HCMV and HHV-6 infection in in vitro infected primary human dermal fibroblasts, which represent one of the main SSc target cells. The analysis, performed by Taqman arrays detecting and quantifying 754 microRNAs (miRNAs), showed that both herpesviruses significantly modulated miRNA expression in infected cells, with evident early and late effects and deep modulation (>10 fold) of >40 miRNAs at each time post infection, including those previously recognized for their key function in fibrosis. The correlation between these in vitro results with in vivo observations is strongly suggestive of a role of HCMV and/or HHV-6 in the multistep pathogenesis of fibrosis in SSc and in the induction of fibrosis-signaling pathways finally leading to tissue fibrosis. The identification of specific miRNAs may open the way to their use as biomarkers for SSc diagnosis, assessment of disease progression and possible antifibrotic therapies. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

Review

Jump to: Research

19 pages, 1846 KiB  
Review
Therapeutic Targeting of Intestinal Fibrosis in Crohn’s Disease
by Giovanni Santacroce, Marco Vincenzo Lenti and Antonio Di Sabatino
Cells 2022, 11(3), 429; https://0-doi-org.brum.beds.ac.uk/10.3390/cells11030429 - 26 Jan 2022
Cited by 23 | Viewed by 6182
Abstract
Intestinal fibrosis is one of the most threatening complications of Crohn’s disease. It occurs in more than a third of patients with this condition, is associated with increased morbidity and mortality, and surgery often represents the only available therapeutic option. The mechanisms underlying [...] Read more.
Intestinal fibrosis is one of the most threatening complications of Crohn’s disease. It occurs in more than a third of patients with this condition, is associated with increased morbidity and mortality, and surgery often represents the only available therapeutic option. The mechanisms underlying intestinal fibrosis are partly known. Studies conducted so far have shown a relevant pathogenetic role played by mesenchymal cells (especially myofibroblasts), cytokines (e.g., transforming growth factor-β), growth factors, microRNAs, intestinal microbiome, matrix stiffness, and mesenteric adipocytes. Further studies are still necessary to elucidate all the mechanisms involved in intestinal fibrosis, so that targeted therapies can be developed. Although several pre-clinical studies have been conducted so far, no anti-fibrotic therapy is yet available to prevent or reverse intestinal fibrosis. The aim of this review is to provide an overview of the main therapeutic targets currently identified and the most promising anti-fibrotic therapies, which may be available in the near future. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

20 pages, 400 KiB  
Review
Metformin and Glaucoma—Review of Anti-Fibrotic Processes and Bioenergetics
by Daire J. Hurley, Mustapha Irnaten and Colm O’Brien
Cells 2021, 10(8), 2131; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10082131 - 19 Aug 2021
Cited by 7 | Viewed by 3489
Abstract
Glaucoma is the leading cause of irreversible blindness globally. With an aging population, disease incidence will rise with an enormous societal and economic burden. The treatment strategy revolves around targeting intraocular pressure, the principle modifiable risk factor, to slow progression of disease. However, [...] Read more.
Glaucoma is the leading cause of irreversible blindness globally. With an aging population, disease incidence will rise with an enormous societal and economic burden. The treatment strategy revolves around targeting intraocular pressure, the principle modifiable risk factor, to slow progression of disease. However, there is a clear unmet clinical need to find a novel therapeutic approach that targets and halts the retinal ganglion cell (RGC) degeneration that occurs with fibrosis. RGCs are highly sensitive to metabolic fluctuations as a result of multiple stressors and thus their viability depends on healthy mitochondrial functioning. Metformin, known for its use in type 2 diabetes, has come to the forefront of medical research in multiple organ systems. Its use was recently associated with a 25% reduced risk of glaucoma in a large population study. Here, we discuss its application to glaucoma therapy, highlighting its effect on fibrotic signalling pathways, mitochondrial bioenergetics and NAD oxidation. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
27 pages, 1258 KiB  
Review
Hypoxia, Hypoxia-Inducible Factors and Liver Fibrosis
by Beatrice Foglia, Erica Novo, Francesca Protopapa, Marina Maggiora, Claudia Bocca, Stefania Cannito and Maurizio Parola
Cells 2021, 10(7), 1764; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10071764 - 13 Jul 2021
Cited by 35 | Viewed by 5296
Abstract
Liver fibrosis is a potentially reversible pathophysiological event, leading to excess deposition of extracellular matrix (ECM) components and taking place as the net result of liver fibrogenesis, a dynamic and highly integrated process occurring during chronic liver injury of any etiology. Liver fibrogenesis [...] Read more.
Liver fibrosis is a potentially reversible pathophysiological event, leading to excess deposition of extracellular matrix (ECM) components and taking place as the net result of liver fibrogenesis, a dynamic and highly integrated process occurring during chronic liver injury of any etiology. Liver fibrogenesis and fibrosis, together with chronic inflammatory response, are primarily involved in the progression of chronic liver diseases (CLD). As is well known, a major role in fibrogenesis and fibrosis is played by activated myofibroblasts (MFs), as well as by macrophages and other hepatic cell populations involved in CLD progression. In the present review, we will focus the attention on the emerging pathogenic role of hypoxia, hypoxia-inducible factors (HIFs) and related mediators in the fibrogenic progression of CLD. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

57 pages, 3903 KiB  
Review
Extracellular Vesicles in Organ Fibrosis: Mechanisms, Therapies, and Diagnostics
by David R. Brigstock
Cells 2021, 10(7), 1596; https://0-doi-org.brum.beds.ac.uk/10.3390/cells10071596 - 25 Jun 2021
Cited by 31 | Viewed by 6230
Abstract
Fibrosis is the unrelenting deposition of excessively large amounts of insoluble interstitial collagen due to profound matrigenic activities of wound-associated myofibroblasts during chronic injury in diverse tissues and organs. It is a highly debilitating pathology that affects millions of people globally and leads [...] Read more.
Fibrosis is the unrelenting deposition of excessively large amounts of insoluble interstitial collagen due to profound matrigenic activities of wound-associated myofibroblasts during chronic injury in diverse tissues and organs. It is a highly debilitating pathology that affects millions of people globally and leads to decreased function of vital organs and increased risk of cancer and end-stage organ disease. Extracellular vesicles (EVs) produced within the chronic wound environment have emerged as important vehicles for conveying pro-fibrotic signals between many of the cell types involved in driving the fibrotic response. On the other hand, EVs from sources such as stem cells, uninjured parenchymal cells, and circulation have in vitro and in vivo anti-fibrotic activities that have provided novel and much-needed therapeutic options. Finally, EVs in body fluids of fibrotic individuals contain cargo components that may have utility as fibrosis biomarkers, which could circumvent current obstacles to fibrosis measurement in the clinic, allowing fibrosis stage, progression, or regression to be determined in a manner that is accurate, safe, minimally-invasive, and conducive to repetitive testing. This review highlights the rapid and recent progress in our understanding of EV-mediated fibrotic pathogenesis, anti-fibrotic therapy, and fibrosis staging in the lung, kidney, heart, liver, pancreas, and skin. Full article
(This article belongs to the Special Issue Molecular Mechanisms in Organ Fibrosis)
Show Figures

Figure 1

Back to TopTop