1
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation during Wound Closure. ACS Biomater Sci Eng 2025; 11:2249-2262. [PMID: 40029610 DOI: 10.1021/acsbiomaterials.4c02019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. Extra domain A (EDA) Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, EDA Fn organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional coactivator, yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate whether YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity, but on stiffer substrates, they decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest that there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
2
|
Mo B, Ding Y, Ji Q. NLRP3 inflammasome in cardiovascular diseases: an update. Front Immunol 2025; 16:1550226. [PMID: 40079000 PMCID: PMC11896874 DOI: 10.3389/fimmu.2025.1550226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome is involved in numerous types of CVD. As part of innate immunity, the NLRP3 inflammasome plays a vital role, requiring priming and activation signals to trigger inflammation. The NLRP3 inflammasome leads both to the release of IL-1 family cytokines and to a distinct form of programmed cell death called pyroptosis. Inflammation related to CVD has been extensively investigated in relation to the NLRP3 inflammasome. In this review, we describe the pathways triggering NLRP3 priming and activation and discuss its pathogenic effects on CVD. This study also provides an overview of potential therapeutic approaches targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Binhai Mo
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yudi Ding
- First People’s Hospital of Nanning, Nanning, Guangxi, China
| | - Qingwei Ji
- People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
3
|
Van Heest A, Wang Y, Zhang L, Phillips LA, Karsen SD, Nelson C, Knight HL, Perper SJ, O’Brien S, Clements M, Sun VZ, Goodearl A, Schwartz Sterman A, Mitra S. Quantitative Assessment of Pulmonary Fibrosis in a Murine Model via a Multimodal Imaging Workflow. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:85-94. [PMID: 40018646 PMCID: PMC11863149 DOI: 10.1021/cbmi.4c00065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 03/01/2025]
Abstract
Disease-recapitulating animal models are valuable tools in preclinical development for the study of compounds. In the case of fibrotic pulmonary diseases such as idiopathic pulmonary fibrosis (IPF), the bleomycin model of lung injury in the mouse is widely used. To evaluate bleomycin-induced changes in the lung, we employed a quantitative, multimodal approach. Using in vivo microcomputed tomography (μCT), we demonstrated radiographic changes associated with disease progression in aeration levels of the lung parenchyma. There exists an unmet need for a quantitative, high-resolution imaging probe to detect pulmonary fibrosis, particularly that can differentiate between inflammatory and fibrotic components of the disease. Matrix remodeling and overexpression of extracellular matrix (ECM) proteins such as collagen and fibronectin are hallmarks of organ fibrosis. A splice variant of fibronectin containing extra domain A (FnEDA) is of particular interest in fibrosis due to its high level of expression in diseased tissue, which is confirmed here using immunohistochemistry (IHC) in mouse and human lungs. An antibody against FnEDA was evaluated for use as an imaging tool, particularly by using in vivo single-photon emission computed tomography (SPECT) and ex vivo near-infrared (NIR) fluorescence imaging. These data were further corroborated with histological tissue staining and fibrosis quantitation based on a Modified Ashcroft (MA) score and a digital image analysis of whole slide lung tissue sections. The fusion of these different approaches represents a robust integrated workflow combining anatomical and molecular imaging technologies to enable the visualization and quantitation of disease activity and treatment response with an inhibitor of the TGFβ signaling pathway.
Collapse
Affiliation(s)
| | | | - Liang Zhang
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Lucy A. Phillips
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Samuel D. Karsen
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Christine Nelson
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Heather L. Knight
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Stuart J. Perper
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Stephen O’Brien
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Meghan Clements
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Victor Z. Sun
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | - Andrew Goodearl
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| | | | - Soumya Mitra
- AbbVie Bioresearch Center, Worcester, Massachusetts 01605, United States
| |
Collapse
|
4
|
Lim YW, Quinn R, Bharti K, Ferrer M, Zarkoob H, Song MJ. Development of immunocompetent full thickness skin tissue constructs to model skin fibrosis for high-throughput drug screening. Biofabrication 2024; 17:015033. [PMID: 39622178 PMCID: PMC11638742 DOI: 10.1088/1758-5090/ad998c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
The lack of the immune component in most of the engineered skin models remains a challenge to study the interplay between different immune and non-immune cell types of the skin. Immunocompetent humanin vitroskin models offer potential advantages in recapitulatingin vivolike behavior which can serve to accelerate translational research and therapeutics development for skin diseases. Here we describe a three-dimensional human full-thickness skin (FTS) equivalent incorporating polarized M1 and M2 macrophages from human peripheral CD14+monocytes. This macrophage-incorporated FTS model demonstrates discernible immune responses with physiologically relevant cytokine production and macrophage plasticity under homeostatic and lipopolysaccharide stimulation conditions. M2-incorporated FTS recapitulates skin fibrosis phenotypes with transforming growth factor-β1 treatment as reflected by significant collagen deposition and myofibroblast expression, demonstrating a M2 potentiation effect. In conclusion, we successfully biofabricated an immunocompetent FTS with functional macrophages in a high-throughput (HT) amenable format. This model is the first step towards a HT-assay platform to develop new therapeutics for skin diseases.
Collapse
Affiliation(s)
- Yi Wei Lim
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Russell Quinn
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Hoda Zarkoob
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Min Jae Song
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| |
Collapse
|
5
|
Basalova NA, Vigovskiy MA, Popov VS, Lagereva EA, Grigorieva OA, Efimenko AY. The Role of Activated Stromal Cells in Fibrotic Foci Formation and Reversion. Cells 2024; 13:2064. [PMID: 39768155 PMCID: PMC11674712 DOI: 10.3390/cells13242064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
Fibrotic focus is a pivotal morphofunctional unit in developing fibrosis in various tissues. For most fibrotic diseases, including progressive forms, the foci are considered unable to remodel and contribute to the worsening of prognosis. Unfortunately, the dynamics of the fibrotic focus formation and resolution remains understudied. A number of data suggest that the key cell type for focus formation are activated stromal cells marked by fibroblast activated protein alpha (FAPα) due to their high capacity for extracellular matrix (ECM) remodeling. We evaluated the dynamics of fibrotic focus formation and the contribution of the main cell types, including FAPα+ cells, in this process using a murine model of bleomycin-induced lung fibrosis. We revealed the very early appearance of FAPα+ cells in lungs after injury and assumed their important involvement to the myofibroblast pool formation. During the first month after bleomycin administration, FAPα+ cells colocalize with CD206+ M2 macrophages. Interestingly, during the reversion stage, we unexpectedly observed the specific structured foci formed by CD90+FAPα+ cells, which we suggested calling "remodeling foci". Our findings highlight the crucial role of activated stromal cells in fibrosis initiation, progression, and reversion and provide emerging issues regarding the novel targets for antifibrotic therapy.
Collapse
Affiliation(s)
- Nataliya Andreevna Basalova
- Centre for Regenerative Medicine, Medical Research and Educational Institute, Lomonosov Moscow State University, 119192 Moscow, Russia; (M.A.V.); (V.S.P.); (E.A.L.); (O.A.G.); (A.Y.E.)
| | | | | | | | | | | |
Collapse
|
6
|
Zhang H, Thai PN, Shivnaraine RV, Ren L, Wu X, Siepe DH, Liu Y, Tu C, Shin HS, Caudal A, Mukherjee S, Leitz J, Wen WTL, Liu W, Zhu W, Chiamvimonvat N, Wu JC. Multiscale drug screening for cardiac fibrosis identifies MD2 as a therapeutic target. Cell 2024; 187:7143-7163.e22. [PMID: 39413786 PMCID: PMC11645214 DOI: 10.1016/j.cell.2024.09.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 08/13/2024] [Accepted: 09/19/2024] [Indexed: 10/18/2024]
Abstract
Cardiac fibrosis impairs cardiac function, but no effective clinical therapies exist. To address this unmet need, we employed a high-throughput screening for antifibrotic compounds using human induced pluripotent stem cell (iPSC)-derived cardiac fibroblasts (CFs). Counter-screening of the initial candidates using iPSC-derived cardiomyocytes and iPSC-derived endothelial cells excluded hits with cardiotoxicity. This screening process identified artesunate as the lead compound. Following profibrotic stimuli, artesunate inhibited proliferation, migration, and contraction in human primary CFs, reduced collagen deposition, and improved contractile function in 3D-engineered heart tissues. Artesunate also attenuated cardiac fibrosis and improved cardiac function in heart failure mouse models. Mechanistically, artesunate targeted myeloid differentiation factor 2 (MD2) and inhibited MD2/Toll-like receptor 4 (TLR4) signaling pathway, alleviating fibrotic gene expression in CFs. Our study leverages multiscale drug screening that integrates a human iPSC platform, tissue engineering, animal models, in silico simulations, and multiomics to identify MD2 as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Hao Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Phung N Thai
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | - Lu Ren
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xuekun Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dirk H Siepe
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chengyi Tu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hye Sook Shin
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Jeremy Leitz
- Greenstone Biosciences, Palo Alto, CA 94305, USA
| | - Wilson Tan Lek Wen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenqiang Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wenjuan Zhu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of California, Davis, Davis, CA 95616, USA; Department of Basic Medical Sciences and Translational Cardiovascular Research Center, University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Sadeghi Shaker M, Rokni M, Kavosi H, Enayati S, Madreseh E, Mahmoudi M, Farhadi E, Vodjgani M. Salirasib Inhibits the Expression of Genes Involved in Fibrosis in Fibroblasts of Systemic Sclerosis Patients. Immun Inflamm Dis 2024; 12:e70063. [PMID: 39601641 PMCID: PMC11600624 DOI: 10.1002/iid3.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/01/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Fibrosis is a principal sign of systemic sclerosis (SSc) which can affect several organs including the lung, heart, and dermis. Dermal fibroblasts of SSc patients are characterized by persistent and activated Ras and ERK1/2 signaling which stimulates extreme collagen and extracellular matrix synthesis. Salirasib is a Ras inhibitor that competitively prevents the adherence of GTP-bound Ras to the plasma membrane, that inhibits Ras signaling. This study intended to clarify whether salirasib can influence fibrotic mediators in SSc fibroblasts. MATERIALS AND METHODS Dermal fibroblasts from 10 SSc patients were treated with salirasib in the presence of TGF-β1, and mRNA levels of H-Ras and genes related to fibrosis, such as COL1A1, COL1A2, CTGF, TGF-β1, fibronectin, ACTA2, and MMP1 was measured by real-time PCR. The α-SMA protein expression was analyzed by immunofluorescence staining. RESULTS In dermal fibroblasts of SSc patients, salirasib treatment, markedly downregulated the H-Ras gene expression. In addition, the protein expression of α-SMA and gene expression of ACTA2 were inhibited upon salirasib treatment. Salirasib also significantly reduced the expression of COL1A1, and COL1A2 genes and augmented the gene expression of MMP1. The mRNA levels of other genes related to fibrosis such as FN1, CTGF, and TGF-β1 were significantly decreased upon salirasib treatment. CONCLUSION Considering salirasib significantly reduced the expression of genes related to the fibrosis process and α-SMA gene and protein expression, and given significant upregulation of MMP1 by salirasib, it can be considered as a new curative strategy for fibrotic diseases like SSc.
Collapse
Affiliation(s)
- Mina Sadeghi Shaker
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Mohsen Rokni
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Department of ImmunologyUniversity of social Welfare and Rehabilitation SciencesTehranIran
| | - Hoda Kavosi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Samaneh Enayati
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
| | - Elham Madreseh
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Department of Epidemiology and Biostatistics, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Mahdi Mahmoudi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Elham Farhadi
- Rheumatology Research CenterTehran University of Medical SciencesTehranIran
- Research Center for Chronic Inflammatory DiseasesTehran University of Medical SciencesTehranIran
| | - Mohammad Vodjgani
- Department of Immunology, School of MedicineTehran University of Medical SciencesTehranIran
| |
Collapse
|
8
|
Patten J, Halligan P, Bashiri G, Kegel M, Bonadio JD, Wang K. EDA Fibronectin Microarchitecture and YAP Translocation During Wound Closure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614581. [PMID: 39386582 PMCID: PMC11463502 DOI: 10.1101/2024.09.23.614581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Fibronectin (Fn) is an extracellular matrix glycoprotein with mechanosensitive structure-function. EDA Fn, a Fn isoform, is not present in adult tissue but is required for tissue repair. Curiously, EDA Fn is linked to both regenerative and fibrotic tissue repair. Given that Fn mechanoregulates cell behavior, Fn EDA organization during wound closure might play a role in mediating these differing responses. One mechanism by which cells sense and respond to their microenvironment is by activating a transcriptional co-activator, Yes-associated protein (YAP). Interestingly, YAP activity is not only required for wound closure, but similarly linked to both regenerative and fibrotic repair. Therefore, this study aims to evaluate how, during normal and fibrotic wound closure, EDA Fn organization might modulate YAP translocation by culturing human dermal fibroblasts on polydimethylsiloxane (PDMS) substrates mimicking normal (soft: 18 kPa) and fibrotic (stiff: 146 kPa) wounded skin. On stiffer substrates mimicking fibrotic wounds, fibroblasts assembled an aligned EDA Fn matrix comprising thinner fibers, suggesting increased microenvironmental tension. To evaluate if cell binding to the EDA domain of Fn was essential to overall matrix organization, fibroblasts were treated with Irigenin, which inhibits binding to the EDA domain within Fn. Blocking adhesion to EDA led to randomly organized EDA Fn matrices with thicker fibers, suggesting reduced microenvironmental tension even during fibrotic wound closure. To evaluate if YAP signaling plays a role in EDA Fn organization, fibroblasts were treated with CA3, which suppresses YAP activity in a dose-dependent manner. Treatment with CA3 also led to randomly organized EDA Fn matrices with thicker fibers, suggesting a potential connected mechanism of reducing tension during fibrotic wound closure. Next, YAP activity was assessed to evaluate the impact of EDA Fn organization. Interestingly, fibroblasts migrating on softer substrates mimicking normal wounds increased YAP activity but on stiffer substrates, decreased YAP activity. When fibroblasts on stiffer substrates were treated with Irigenin or CA3, fibroblasts increased YAP activity. These results suggest there may be disrupted signaling between EDA Fn organization and YAP translocation during fibrotic wound closure that could be restored when reestablishing normal EDA Fn matrix organization to instead drive regenerative wound repair.
Collapse
Affiliation(s)
- Jennifer Patten
- Department of Bioengineering, Temple University, Pennsylvania
| | | | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Pennsylvania
| | - Michael Kegel
- Department of Bioengineering, Temple University, Pennsylvania
| | - Jacob D Bonadio
- Department of Bioengineering, Temple University, Pennsylvania
| | - Karin Wang
- Department of Bioengineering, Temple University, Pennsylvania
| |
Collapse
|
9
|
Man SM, Kanneganti TD. Innate immune sensing of cell death in disease and therapeutics. Nat Cell Biol 2024; 26:1420-1433. [PMID: 39223376 DOI: 10.1038/s41556-024-01491-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024]
Abstract
Innate immunity, cell death and inflammation underpin many aspects of health and disease. Upon sensing pathogens, pathogen-associated molecular patterns or damage-associated molecular patterns, the innate immune system activates lytic, inflammatory cell death, such as pyroptosis and PANoptosis. These genetically defined, regulated cell death pathways not only contribute to the host defence against infectious disease, but also promote pathological manifestations leading to cancer and inflammatory diseases. Our understanding of the underlying mechanisms has grown rapidly in recent years. However, how dying cells, cell corpses and their liberated cytokines, chemokines and inflammatory signalling molecules are further sensed by innate immune cells, and their contribution to further amplify inflammation, trigger antigen presentation and activate adaptive immunity, is less clear. Here, we discuss how pattern-recognition and PANoptosome sensors in innate immune cells recognize and respond to cell-death signatures. We also highlight molecular targets of the innate immune response for potential therapeutic development.
Collapse
Affiliation(s)
- Si Ming Man
- Division of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia.
| | | |
Collapse
|
10
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
11
|
Damerau A, Rosenow E, Alkhoury D, Buttgereit F, Gaber T. Fibrotic pathways and fibroblast-like synoviocyte phenotypes in osteoarthritis. Front Immunol 2024; 15:1385006. [PMID: 38895122 PMCID: PMC11183113 DOI: 10.3389/fimmu.2024.1385006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is the most common form of arthritis, characterized by osteophyte formation, cartilage degradation, and structural and cellular alterations of the synovial membrane. Activated fibroblast-like synoviocytes (FLS) of the synovial membrane have been identified as key drivers, secreting humoral mediators that maintain inflammatory processes, proteases that cause cartilage and bone destruction, and factors that drive fibrotic processes. In normal tissue repair, fibrotic processes are terminated after the damage has been repaired. In fibrosis, tissue remodeling and wound healing are exaggerated and prolonged. Various stressors, including aging, joint instability, and inflammation, lead to structural damage of the joint and micro lesions within the synovial tissue. One result is the reduced production of synovial fluid (lubricants), which reduces the lubricity of the cartilage areas, leading to cartilage damage. In the synovial tissue, a wound-healing cascade is initiated by activating macrophages, Th2 cells, and FLS. The latter can be divided into two major populations. The destructive thymocyte differentiation antigen (THY)1─ phenotype is restricted to the synovial lining layer. In contrast, the THY1+ phenotype of the sublining layer is classified as an invasive one with immune effector function driving synovitis. The exact mechanisms involved in the transition of fibroblasts into a myofibroblast-like phenotype that drives fibrosis remain unclear. The review provides an overview of the phenotypes and spatial distribution of FLS in the synovial membrane of OA, describes the mechanisms of fibroblast into myofibroblast activation, and the metabolic alterations of myofibroblast-like cells.
Collapse
Affiliation(s)
- Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Emely Rosenow
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- German Rheumatism Research Center Berlin, a Leibniz Institute, Glucocorticoids - Bioenergetics - 3R Research Lab, Berlin, Germany
| |
Collapse
|
12
|
Bonadio JD, Bashiri G, Halligan P, Kegel M, Ahmed F, Wang K. Delivery technologies for therapeutic targeting of fibronectin in autoimmunity and fibrosis applications. Adv Drug Deliv Rev 2024; 209:115303. [PMID: 38588958 PMCID: PMC11111362 DOI: 10.1016/j.addr.2024.115303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/29/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Fibronectin (FN) is a critical component of the extracellular matrix (ECM) contributing to various physiological processes, including tissue repair and immune response regulation. FN regulates various cellular functions such as adhesion, proliferation, migration, differentiation, and cytokine release. Alterations in FN expression, deposition, and molecular structure can profoundly impact its interaction with other ECM proteins, growth factors, cells, and associated signaling pathways, thus influencing the progress of diseases such as fibrosis and autoimmune disorders. Therefore, developing therapeutics that directly target FN or its interaction with cells and other ECM components can be an intriguing approach to address autoimmune and fibrosis pathogenesis.
Collapse
Affiliation(s)
- Jacob D Bonadio
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Ghazal Bashiri
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Patrick Halligan
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Michael Kegel
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Fatima Ahmed
- Department of Bioengineering, Temple University, Philadelphia, PA, United States
| | - Karin Wang
- Department of Bioengineering, Temple University, Philadelphia, PA, United States.
| |
Collapse
|
13
|
O'Reilly S. Emerging therapeutic targets in systemic sclerosis. J Mol Med (Berl) 2024; 102:465-478. [PMID: 38386070 DOI: 10.1007/s00109-024-02424-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 02/23/2024]
Abstract
Systemic sclerosis is an autoimmune connective tissue disease which is characterised by vascular perturbations, inflammation, and fibrosis. Although huge progress recently into the underlying molecular pathways that are perturbed in the disease, currently no therapy exists that targets the fibrosis element of the disease and consequently there is a huge unmet medical need. Emerging studies reveal new dimensions of complexity, and multiple aberrant pathways have been uncovered that have shed light on disturbed signalling in the disease, primarily in inflammatory pathways that can be targeted with repurposed drugs. Pre-clinical animal models using these inhibitors have yielded proof of concept for targeting these signalling systems and progressing to clinical trials. This review will examine the recent evidence of new perturbed pathways in SSc and how these can be targeted with new or repurposed drugs to target a currently intractable disease.
Collapse
Affiliation(s)
- Steven O'Reilly
- Department of Biosciences, Durham University, South Road, Durham, UK.
| |
Collapse
|
14
|
Laiva AL, O'Brien FJ, Keogh MB. Dual delivery gene-activated scaffold directs fibroblast activity and keratinocyte epithelization. APL Bioeng 2024; 8:016104. [PMID: 38283135 PMCID: PMC10821797 DOI: 10.1063/5.0174122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 01/12/2024] [Indexed: 01/30/2024] Open
Abstract
Fibroblasts are the most abundant cell type in dermal skin and keratinocytes are the most abundant cell type in the epidermis; both play a crucial role in wound remodeling and maturation. We aim to assess the functionality of a novel dual gene activated scaffold (GAS) on human adult dermal fibroblasts (hDFs) and see how the secretome produced could affect human dermal microvascular endothelial cells (HDMVECs) and human epidermal keratinocyte (hEKs) growth and epithelization. Our GAS is a collagen chondroitin sulfate scaffold loaded with pro-angiogenic stromal derived factor (SDF-1α) and/or an anti-aging β-Klotho plasmids. hDFs were grown on GAS for two weeks and compared to gene-free scaffolds. GAS produced a significantly better healing outcome in the fibroblasts than in the gene-free scaffold group. Among the GAS groups, the dual GAS induced the most potent pro-regenerative maturation in fibroblasts with a downregulation in proliferation (twofold, p < 0.05), fibrotic remodeling regulators TGF-β1 (1.43-fold, p < 0.01) and CTGF (1.4-fold, p < 0.05), fibrotic cellular protein α-SMA (twofold, p < 0.05), and fibronectin matrix deposition (twofold, p < 0.05). The dual GAS secretome also showed enhancements of paracrine keratinocyte pro-epithelializing ability (1.3-fold, p < 0.05); basement membrane regeneration through laminin (6.4-fold, p < 0.005) and collagen IV (8.7-fold, p < 0.005) deposition. Our findings demonstrate enhanced responses in dual GAS containing hDFs by proangiogenic SDF-1α and β-Klotho anti-fibrotic rejuvenating activities. This was demonstrated by activating hDFs on dual GAS to become anti-fibrotic in nature while eliciting wound repair basement membrane proteins; enhancing a proangiogenic HDMVECs paracrine signaling and greater epithelisation of hEKs.
Collapse
Affiliation(s)
| | | | - Michael B. Keogh
- Author to whom correspondence should be addressed:. Tel.: +973 17351450
| |
Collapse
|
15
|
Pang N, Laiva AL, Sulaiman NZ, Das P, O’Brien FJ, Keogh MB. Dual Glyoxalase-1 and β-Klotho Gene-Activated Scaffold Reduces Methylglyoxal and Reprograms Diabetic Adipose-Derived Stem Cells: Prospects in Improved Wound Healing. Pharmaceutics 2024; 16:265. [PMID: 38399319 PMCID: PMC10892312 DOI: 10.3390/pharmaceutics16020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Tissue engineering approaches aim to provide biocompatible scaffold supports that allow healing to progress often in healthy tissue. In diabetic foot ulcers (DFUs), hyperglycemia impedes ulcer regeneration, due to complications involving accumulations of cellular methylglyoxal (MG), a key component of oxidated stress and premature cellular aging which further limits repair. In this study, we aim to reduce MG using a collagen-chondroitin sulfate gene-activated scaffold (GAS) containing the glyoxalase-1 gene (GLO-1) to scavenge MG and anti-fibrotic β-klotho to restore stem cell activity in diabetic adipose-derived stem cells (dADSCs). dADSCs were cultured on dual GAS constructs for 21 days in high-glucose media in vitro. Our results show that dADSCs cultured on dual GAS significantly reduced MG accumulation (-84%; p < 0.05) compared to the gene-free controls. Similar reductions in profibrotic proteins α-smooth muscle actin (-65%) and fibronectin (-76%; p < 0.05) were identified in dual GAS groups. Similar findings were observed in the expression of pro-scarring structural proteins collagen I (-62%), collagen IV (-70%) and collagen VII (-86%). A non-significant decrease in the expression of basement membrane protein E-cadherin (-59%) was noted; however, the dual GAS showed a significant increase in the expression of laminin (+300%). We conclude that dual GAS-containing Glo-1 and β-klotho had a synergistic MG detoxification and anti-fibrotic role in dADSC's. This may be beneficial to provide better wound healing in DFUs by controlling the diabetic environment and rejuvenating the diabetic stem cells towards improved wound healing.
Collapse
Affiliation(s)
- Nadia Pang
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Ashang L. Laiva
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Noof Z. Sulaiman
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Priya Das
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| | - Fergal J. O’Brien
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123 St. Stephen’s Green, D02 YN77 Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Royal College of Surgeons in Ireland and Trinity College Dublin, D02 PN40 Dublin, Ireland
| | - Michael B. Keogh
- Tissue Engineering Research Group—Bahrain, Royal College of Surgeons in Ireland, Adliya P.O. Box 15503, Bahrain; (N.P.); (N.Z.S.); (P.D.)
| |
Collapse
|
16
|
Naha A, Driscoll TP. Fibronectin sensitizes activation of contractility, YAP, and NF-κB in nucleus pulposus cells. J Orthop Res 2024; 42:434-442. [PMID: 37525423 DOI: 10.1002/jor.25670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/21/2023] [Accepted: 07/26/2023] [Indexed: 08/02/2023]
Abstract
Intervertebral disc degeneration involves the breakdown of the discs of the spine due to genetics, aging, or faulty mechanical loading. As part of the progression of the disease, nucleus pulposus cells lose their phenotypic characteristics, inducing inflammation and extracellular matrix (ECM) alterations that result in a loss of disc mechanical homeostasis. Fibronectin is one ECM molecule that has been shown to be upregulated in disc degeneration and plays an important role in the progression of a wide variety of fibrotic diseases. Fragments of fibronectin have also long been associated with both osteoarthritis and disc degeneration. The goal of this work is to test the effects of fibronectin on disc cell phenotype, mechanosensing, and inflammatory signaling. We identify that fibronectin increases the activation of cellular contractility, the mechanosensitive transcription factor Yes-associated protein, and the inflammatory transcription factor nuclear factor-κB. This results in decreased production and expression of proteoglycans, which are required to maintain healthy disc function. Thus, fibronectin is a potential regulator of phenotypic changes in disc degeneration, and a potential target for treating disc degeneration at the cellular level. Understanding the role of fibronectin, and its potential as a therapeutic target, could provide new approaches for preventing or reversing disc degeneration.
Collapse
Affiliation(s)
- Ananya Naha
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| | - Tristan P Driscoll
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, Florida, USA
| |
Collapse
|
17
|
Afshar K, Sanaei MJ, Ravari MS, Pourbagheri-Sigaroodi A, Bashash D. An overview of extracellular matrix and its remodeling in the development of cancer and metastasis with a glance at therapeutic approaches. Cell Biochem Funct 2023; 41:930-952. [PMID: 37665068 DOI: 10.1002/cbf.3846] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
The extracellular matrix (ECM) is an inevitable part of tissues able to provide structural support for cells depending on the purpose of tissues and organs. The dynamic characteristics of ECM let this system fluently interact with the extrinsic triggers and get stiffed, remodeled, and/or degraded ending in maintaining tissue homeostasis. ECM could serve as the platform for cancer progression. The dysregulation of biochemical and biomechanical ECM features might take participate in some pathological conditions such as aging, tissue destruction, fibrosis, and particularly cancer. Tumors can reprogram how ECM remodels by producing factors able to induce protein synthesis, matrix proteinase expression, degradation of the basement membrane, growth signals and proliferation, angiogenesis, and metastasis. Therefore, targeting the ECM components, their secretion, and their interactions with other cells or tumors could be a promising strategy in cancer therapies. The present study initially introduces the physiological functions of ECM and then discusses how tumor-dependent dysregulation of ECM could facilitate cancer progression and ends with reviewing the novel therapeutic strategies regarding ECM.
Collapse
Affiliation(s)
- Kimiya Afshar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Sadat Ravari
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Basalova N, Alexandrushkina N, Grigorieva O, Kulebyakina M, Efimenko A. Fibroblast Activation Protein Alpha (FAPα) in Fibrosis: Beyond a Perspective Marker for Activated Stromal Cells? Biomolecules 2023; 13:1718. [PMID: 38136590 PMCID: PMC10742035 DOI: 10.3390/biom13121718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
The development of tissue fibrosis is a complex process involving the interaction of multiple cell types, which makes the search for antifibrotic agents rather challenging. So far, myofibroblasts have been considered the key cell type that mediated the development of fibrosis and thus was the main target for therapy. However, current strategies aimed at inhibiting myofibroblast function or eliminating them fail to demonstrate sufficient effectiveness in clinical practice. Therefore, today, there is an unmet need to search for more reliable cellular targets to contribute to fibrosis resolution or the inhibition of its progression. Activated stromal cells, capable of active proliferation and invasive growth into healthy tissue, appear to be such a target population due to their more accessible localization in the tissue and their high susceptibility to various regulatory signals. This subpopulation is marked by fibroblast activation protein alpha (FAPα). For a long time, FAPα was considered exclusively a marker of cancer-associated fibroblasts. However, accumulating data are emerging on the diverse functions of FAPα, which suggests that this protein is not only a marker but also plays an important role in fibrosis development and progression. This review aims to summarize the current data on the expression, regulation, and function of FAPα regarding fibrosis development and identify promising advances in the area.
Collapse
Affiliation(s)
- Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Natalya Alexandrushkina
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
| | - Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Educational Centre, Lomonosov Moscow State University, 119192 Moscow, Russia (O.G.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia;
| |
Collapse
|
19
|
Franco-Valls H, Tusquets-Uxó E, Sala L, Val M, Peña R, Iaconcig A, Villarino Á, Jiménez-Arriola M, Massó P, Trincado JL, Eyras E, Muro AF, Otero J, García de Herreros A, Baulida J. Formation of an invasion-permissive matrix requires TGFβ/SNAIL1-regulated alternative splicing of fibronectin. Breast Cancer Res 2023; 25:143. [PMID: 37964360 PMCID: PMC10647173 DOI: 10.1186/s13058-023-01736-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 10/30/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND As in most solid cancers, the emergence of cells with oncogenic mutations in the mammary epithelium alters the tissue homeostasis. Some soluble factors, such as TGFβ, potently modify the behavior of healthy stromal cells. A subpopulation of cancer-associated fibroblasts expressing a TGFβ target, the SNAIL1 transcription factor, display myofibroblastic abilities that rearrange the stromal architecture. Breast tumors with the presence of SNAIL1 in the stromal compartment, and with aligned extracellular fiber, are associated with poor survival prognoses. METHODS We used deep RNA sequencing and biochemical techniques to study alternative splicing and human tumor databases to test for associations (correlation t-test) between SNAIL1 and fibronectin isoforms. Three-dimensional extracellular matrices generated from fibroblasts were used to study the mechanical properties and actions of the extracellular matrices on tumor cell and fibroblast behaviors. A metastatic mouse model of breast cancer was used to test the action of fibronectin isoforms on lung metastasis. RESULTS In silico studies showed that SNAIL1 correlates with the expression of the extra domain A (EDA)-containing (EDA+) fibronectin in advanced human breast cancer and other types of epithelial cancers. In TGFβ-activated fibroblasts, alternative splicing of fibronectin as well as of 500 other genes was modified by eliminating SNAIL1. Biochemical analyses demonstrated that SNAIL1 favors the inclusion of the EDA exon by modulating the activity of the SRSF1 splicing factor. Similar to Snai1 knockout fibroblasts, EDA- fibronectin fibroblasts produce an extracellular matrix that does not sustain TGFβ-induced fiber organization, rigidity, fibroblast activation, or tumor cell invasion. The presence of EDA+ fibronectin changes the action of metalloproteinases on fibronectin fibers. Critically, in an mouse orthotopic breast cancer model, the absence of the fibronectin EDA domain completely prevents lung metastasis. CONCLUSIONS Our results support the requirement of EDA+ fibronectin in the generation of a metastasis permissive stromal architecture in breast cancers and its molecular control by SNAIL1. From a pharmacological point of view, specifically blocking EDA+ fibronectin deposition could be included in studies to reduce the formation of a pro-metastatic environment.
Collapse
Affiliation(s)
- Héctor Franco-Valls
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Elsa Tusquets-Uxó
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
- Institute for Research in Biomedicine, Barcelona, Spain
| | - Laura Sala
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
- National Institutes of Health: Intramural Research Program, Bethesda, MD, USA
| | - Maria Val
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
- Vall Hebron Institute of Research, Barcelona, Spain
| | - Raúl Peña
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Alessandra Iaconcig
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Álvaro Villarino
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Martín Jiménez-Arriola
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Pere Massó
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
| | - Juan L Trincado
- Research Program of Biomedical Informatics, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Eduardo Eyras
- Research Program of Biomedical Informatics, Hospital del Mar Research Institute (IMIM), Barcelona, Spain
- John Curtin School of Medical Research, Australian National University, Canberra, Australia
| | - Andrés F Muro
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Jorge Otero
- Unitat Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Antonio García de Herreros
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain
- Departament de Medicina i Ciències de la Vida, Universitat Pompeu Fabra, Barcelona, Spain
| | - Josep Baulida
- Programa de Recerca en Càncer, Hospital del Mar Research Institute (IMIM), Dr. Aiguader, 88, 08003, Barcelona, Spain.
| |
Collapse
|
20
|
Bale S, Verma P, Varga J, Bhattacharyya S. Extracellular Matrix-Derived Damage-Associated Molecular Patterns (DAMP): Implications in Systemic Sclerosis and Fibrosis. J Invest Dermatol 2023; 143:1877-1885. [PMID: 37452808 PMCID: PMC11974346 DOI: 10.1016/j.jid.2023.04.030] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/24/2023] [Accepted: 04/07/2023] [Indexed: 07/18/2023]
Abstract
Damage-associated molecular patterns (DAMPs) are intracellular molecules released under cellular stress or recurring tissue injury, which serve as endogenous ligands for toll-like receptors (TLRs). Such DAMPs are either actively secreted by immune cells or passively released into the extracellular environment from damaged cells or generated as alternatively spliced mRNA variants of extracellular matrix (ECM) glycoproteins. When recognized by pattern recognition receptors (PRRs) such as TLRs, DAMPs trigger innate immune responses. Currently, the best-characterized PRRs include, in addition to TLRs, nucleotide-binding oligomerization domain-like receptors, RIG-I-like RNA helicases, C-type lectin receptors, and many more. Systemic sclerosis (SSc) is a chronic autoimmune condition characterized by inflammation and progressive fibrosis in multiple organs. Using an unbiased survey for SSc-associated DAMPs, we have identified the ECM glycoproteins fibronectin-containing extra domain A and tenascin C as the most highly upregulated in SSc skin and lung biopsies. These DAMPs activate TLR4 on resident stromal cells to elicit profibrotic responses and sustained myofibroblasts activation resulting in progressive fibrosis. This review summarizes the current understanding of the complex functional roles of DAMPs in the progression and failure of resolution of fibrosis in general, with a particular focus on SSc, and considers viable therapeutic approaches targeting DAMPs.
Collapse
Affiliation(s)
- Swarna Bale
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Swati Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
21
|
Zhu L, Liu L, Wang A, Liu J, Huang X, Zan T. Positive feedback loops between fibroblasts and the mechanical environment contribute to dermal fibrosis. Matrix Biol 2023; 121:1-21. [PMID: 37164179 DOI: 10.1016/j.matbio.2023.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/06/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Dermal fibrosis is characterized by excessive deposition of extracellular matrix in the dermis and affects millions of people worldwide and causes limited movement, disfigurement and psychological distress in patients. Fibroblast dysfunction of plays a central role in the pathogenesis of dermal fibrosis and is controlled by distinct factors. Recent studies support the hypothesis that fibroblasts can drive matrix deposition and stiffening, which in turn can exacerbate the functional dysregulation of fibroblasts. Ultimately, through a positive feedback loop, uncontrolled pathological fibrosis develops. This review aims to summarize the phenomenon and mechanism of the positive feedback loop in dermal fibrosis, and discuss potential therapeutic targets to help further elucidate the pathogenesis of dermal fibrosis and develop therapeutic strategies. In this review, fibroblast-derived compositional and structural changes in the ECM that lead to altered mechanical properties are briefly discussed. We focus on the mechanisms by which mechanical cues participate in dermal fibrosis progression. The mechanosensors discussed in the review include integrins, DDRs, proteoglycans, and mechanosensitive ion channels. The FAK, ERK, Akt, and Rho pathways, as well as transcription factors, including MRTF and YAP/TAZ, are also discussed. In addition, we describe stiffness-induced biological changes in the ECM on fibroblasts that contribute to the formation of a positive feedback loop. Finally, we discuss therapeutic strategies to treat the vicious cycle and present important suggestions for researchers conducting in-depth research.
Collapse
Affiliation(s)
- Liang Zhu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lechen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Aoli Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jinwen Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| | - Tao Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Bale S, Verma P, Yalavarthi B, Scarneo SA, Hughes P, Amin MA, Tsou PS, Khanna D, Haystead TA, Bhattacharyya S, Varga J. Pharmacological inhibition of TAK1 prevents and induces regression of experimental organ fibrosis. JCI Insight 2023; 8:e165358. [PMID: 37306632 PMCID: PMC10443806 DOI: 10.1172/jci.insight.165358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 05/31/2023] [Indexed: 06/13/2023] Open
Abstract
Multiorgan fibrosis in systemic sclerosis (SSc) accounts for substantial mortality and lacks effective therapies. Lying at the crossroad of TGF-β and TLR signaling, TGF-β-activated kinase 1 (TAK1) might have a pathogenic role in SSc. We therefore sought to evaluate the TAK1 signaling axis in patients with SSc and to investigate pharmacological TAK1 blockade using a potentially novel drug-like selective TAK1 inhibitor, HS-276. Inhibiting TAK1 abrogated TGF-β1 stimulation of collagen synthesis and myofibroblasts differentiation in healthy skin fibroblasts, and it ameliorated constitutive activation of SSc skin fibroblasts. Moreover, treatment with HS-276 prevented dermal and pulmonary fibrosis and reduced the expression of profibrotic mediators in bleomycin-treated mice. Importantly, initiating HS-276 treatment even after fibrosis was already established prevented its progression in affected organs. Together, these findings implicate TAK1 in the pathogenesis of SSc and identify targeted TAK1 inhibition using a small molecule as a potential strategy for the treatment of SSc and other fibrotic diseases.
Collapse
Affiliation(s)
- Swarna Bale
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Philip Hughes
- EydisBio Inc., Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - M. Asif Amin
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Pei-Suen Tsou
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dinesh Khanna
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Timothy A.J. Haystead
- EydisBio Inc., Durham, North Carolina, USA
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Swati Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
23
|
Baldwin SA, Haugh JM. Semi-autonomous wound invasion via matrix-deposited, haptotactic cues. J Theor Biol 2023; 568:111506. [PMID: 37094713 PMCID: PMC10393182 DOI: 10.1016/j.jtbi.2023.111506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023]
Abstract
Proper wound healing relies on invasion of fibroblasts via directed migration. While the related experimental and mathematical modeling literature has mainly focused on cell migration directed by soluble cues (chemotaxis), there is ample evidence that fibroblast migration is also directed by insoluble, matrix-bound cues (haptotaxis). Furthermore, numerous studies indicate that fibronectin (FN), a haptotactic ligand for fibroblasts, is present and dynamic in the provisional matrix throughout the proliferative phase of wound healing. In the present work, we show the plausibility of a hypothesis that fibroblasts themselves form and maintain haptotactic gradients in a semi-autonomous fashion. As a precursor to this, we examine the positive control scenario where FN is pre-deposited in the wound matrix, and fibroblasts maintain haptotaxis by removing FN at an appropriate rate. After developing conceptual and quantitative understanding of this scenario, we consider two cases in which fibroblasts activate the latent form of a matrix-loaded cytokine, TGFβ, which upregulates the fibroblasts' own secretion of FN. In the first of these, the latent cytokine is pre-patterned and released by the fibroblasts. In the second, fibroblasts in the wound produce the latent TGFβ, with the presence of the wound providing the only instruction. In all cases, wound invasion is more effective than a negative control model with haptotaxis disabled; however, there is a trade-off between the degree of fibroblast autonomy and the rate of invasion.
Collapse
Affiliation(s)
- Scott A Baldwin
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695, USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Campus Box 7905, Raleigh, NC 27695, USA.
| |
Collapse
|
24
|
Maddali P, Ambesi A, McKeown-Longo PJ. Induction of pro-inflammatory genes by fibronectin DAMPs in three fibroblast cell lines: Role of TAK1 and MAP kinases. PLoS One 2023; 18:e0286390. [PMID: 37228128 DOI: 10.1371/journal.pone.0286390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 05/13/2023] [Indexed: 05/27/2023] Open
Abstract
Changes in the organization and structure of the fibronectin matrix are believed to contribute to dysregulated wound healing and subsequent tissue inflammation and tissue fibrosis. These changes include an increase in the EDA isoform of fibronectin as well as the mechanical unfolding of fibronectin type III domains. In previous studies using embryonic foreskin fibroblasts, we have shown that fibronectin's EDA domain (FnEDA) and the partially unfolded first Type III domain (FnIII-1c) function as Damage Associated Molecular Pattern (DAMP) molecules to stimulate the induction of inflammatory cytokines by serving as agonists for Toll-Like Receptor-4 (TLR4). However, the role of signaling molecules downstream of TLR-4 such as TGF-β Activated Kinase 1 (TAK1) and Mitogen activated protein kinases (MAPK) in regulating the expression of fibronectin DAMP induced inflammatory genes in specific cell types is not known. In the current study, we evaluate the molecular steps regulating the fibronectin driven induction of inflammatory genes in three human fibroblast cell lines: embryonic foreskin, adult dermal, and adult kidney. The fibronectin derived DAMPs each induce the phosphorylation and activation of TAK1 which results in the activation of two downstream signaling arms, IKK/NF-κB and MAPK. Using the specific inhibitor 5Z-(7)-Oxozeanol as well as siRNA, we show TAK1 to be a crucial signaling mediator in the release of cytokines in response to fibronectin DAMPs in all three cell types. Finally, we show that FnEDA and FnIII-1c induce several pro-inflammatory cytokines whose expression is dependent on both TAK1 and JNK MAPK and highlight cell-type specific differences in the gene-expression profiles of the fibroblast cell-lines.
Collapse
Affiliation(s)
- Pranav Maddali
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| | - Anthony Ambesi
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| | - Paula J McKeown-Longo
- Department of Regenerative & Cancer Cell Biology, Albany Medical College, Albany, New York, United States of America
| |
Collapse
|
25
|
Fertala J, Wang ML, Rivlin M, Beredjiklian PK, Abboud J, Arnold WV, Fertala A. Extracellular Targets to Reduce Excessive Scarring in Response to Tissue Injury. Biomolecules 2023; 13:biom13050758. [PMID: 37238628 DOI: 10.3390/biom13050758] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Excessive scar formation is a hallmark of localized and systemic fibrotic disorders. Despite extensive studies to define valid anti-fibrotic targets and develop effective therapeutics, progressive fibrosis remains a significant medical problem. Regardless of the injury type or location of wounded tissue, excessive production and accumulation of collagen-rich extracellular matrix is the common denominator of all fibrotic disorders. A long-standing dogma was that anti-fibrotic approaches should focus on overall intracellular processes that drive fibrotic scarring. Because of the poor outcomes of these approaches, scientific efforts now focus on regulating the extracellular components of fibrotic tissues. Crucial extracellular players include cellular receptors of matrix components, macromolecules that form the matrix architecture, auxiliary proteins that facilitate the formation of stiff scar tissue, matricellular proteins, and extracellular vesicles that modulate matrix homeostasis. This review summarizes studies targeting the extracellular aspects of fibrotic tissue synthesis, presents the rationale for these studies, and discusses the progress and limitations of current extracellular approaches to limit fibrotic healing.
Collapse
Affiliation(s)
- Jolanta Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark L Wang
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Michael Rivlin
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Pedro K Beredjiklian
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Joseph Abboud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - William V Arnold
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
26
|
Benn MC, Pot SA, Moeller J, Yamashita T, Fonta CM, Orend G, Kollmannsberger P, Vogel V. How the mechanobiology orchestrates the iterative and reciprocal ECM-cell cross-talk that drives microtissue growth. SCIENCE ADVANCES 2023; 9:eadd9275. [PMID: 36989370 PMCID: PMC10058249 DOI: 10.1126/sciadv.add9275] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Controlled tissue growth is essential for multicellular life and requires tight spatiotemporal control over cell proliferation and differentiation until reaching homeostasis. As cells synthesize and remodel extracellular matrix, tissue growth processes can only be understood if the reciprocal feedback between cells and their environment is revealed. Using de novo-grown microtissues, we identified crucial actors of the mechanoregulated events, which iteratively orchestrate a sharp transition from tissue growth to maturation, requiring a myofibroblast-to-fibroblast transition. Cellular decision-making occurs when fibronectin fiber tension switches from highly stretched to relaxed, and it requires the transiently up-regulated appearance of tenascin-C and tissue transglutaminase, matrix metalloprotease activity, as well as a switch from α5β1 to α2β1 integrin engagement and epidermal growth factor receptor signaling. As myofibroblasts are associated with wound healing and inflammatory or fibrotic diseases, crucial knowledge needed to advance regenerative strategies or to counter fibrosis and cancer progression has been gained.
Collapse
Affiliation(s)
- Mario C. Benn
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Simon A. Pot
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Jens Moeller
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Tadahiro Yamashita
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Charlotte M. Fonta
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Gertraud Orend
- The Tumor Microenvironment Laboratory, INSERM U1109, Hôpital Civil, Institut d'Hématologie et d'Immunologie, 1 Place de l'Hôpital, Strasbourg 67091, France
- Université Strasbourg, Strasbourg 67000, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg 67000, France
| | - Philip Kollmannsberger
- Biomedical Physics, Heinrich-Heine-University Düsseldorf, Universitätsstrasse 1, Düsseldorf 40225, Germany
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| |
Collapse
|
27
|
Grigorieva O, Basalova N, Vigovskiy M, Arbatskiy M, Dyachkova U, Kulebyakina M, Kulebyakin K, Tyurin-Kuzmin P, Kalinina N, Efimenko A. Novel Potential Markers of Myofibroblast Differentiation Revealed by Single-Cell RNA Sequencing Analysis of Mesenchymal Stromal Cells in Profibrotic and Adipogenic Conditions. Biomedicines 2023; 11:biomedicines11030840. [PMID: 36979822 PMCID: PMC10045579 DOI: 10.3390/biomedicines11030840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) are the key regulators of tissue homeostasis and repair after damage. Accumulating evidence indicates the dual contribution of MSCs into the development of fibrosis induced by chronic injury: these cells can suppress the fibrotic process due to paracrine activity, but their promoting role in fibrosis by differentiating into myofibroblasts has also been demonstrated. Many model systems reproducing fibrosis have shown the ability of peroxisome proliferator-activated receptor (PPAR) agonists to reverse myofibroblast differentiation. Thus, the differentiation of multipotent cells into myofibroblasts and adipocytes can be considered as processes that require the activation of opposite patterns of gene expression. To test this hypothesis, we analyzed single cell RNA-Seq transcriptome of human adipose tissue MSCs after stimulation of the myofibroblast or adipogenic differentiation and revealed several genes that changed their expression in a reciprocal manner upon these conditions. We validated the expression of selected genes by RT-PCR, and evaluated the upregulation of several relevant proteins using immunocytochemistry, refining the results obtained by RNA-Seq analysis. We have shown, for the first time, the expression of neurotrimin (NTM), previously studied mainly in the nervous tissue, in human adipose tissue MSCs, and demonstrated its increased gene expression and clustering of membrane receptors upon the stimulation of myofibroblast differentiation. We also showed an increased level of CHD3 (Chromodomain-Helicase-DNA-binding protein 3) in MSCs under profibrotic conditions, while retinol dehydrogenase-10 (RDH10) was detected only in MSCs after adipogenic induction, which contradicted the data of transcriptomic analysis and again highlights the need to validate the data obtained by omics methods. Our findings suggest the further analysis of the potential contribution of neurotrimin and CHD3 in the regulation of myofibroblast differentiation and the development of fibrosis.
Collapse
Affiliation(s)
- Olga Grigorieva
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosovsky Ave., 27/10, 119192 Moscow, Russia; (N.B.); (M.V.); (K.K.); (A.E.)
- Correspondence:
| | - Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosovsky Ave., 27/10, 119192 Moscow, Russia; (N.B.); (M.V.); (K.K.); (A.E.)
| | - Maksim Vigovskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosovsky Ave., 27/10, 119192 Moscow, Russia; (N.B.); (M.V.); (K.K.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Mikhail Arbatskiy
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Uliana Dyachkova
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Konstantin Kulebyakin
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosovsky Ave., 27/10, 119192 Moscow, Russia; (N.B.); (M.V.); (K.K.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Natalia Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Lomonosovsky Ave., 27/10, 119192 Moscow, Russia; (N.B.); (M.V.); (K.K.); (A.E.)
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia; (M.A.); (U.D.); (M.K.); (P.T.-K.); (N.K.)
| |
Collapse
|
28
|
O'Reilly S. Toll-like receptor triggering in systemic sclerosis: time to target. Rheumatology (Oxford) 2023; 62:SI12-SI19. [PMID: 35863054 DOI: 10.1093/rheumatology/keac421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
SSc is an autoimmune disease that has features of vascular abnormalities, inflammation and skin and lung fibrosis. Toll-like receptors (TLRs) are sentinel receptors that serve to recognize pathogens or internal danger signals leading to downstream signalling pathways that ultimately lead to inflammation and modification of adaptive immunity. Inflammation and fibrosis appear intricately connected in this disease and TLR ligation on fibroblasts can directly activate these cells to produce copious amounts of collagen, a hallmark of disease. The presence of damage-associated molecular patterns in association with fibrosis has been highlighted. Given their prominent role in disease, this review discusses the evidence of their expression and role in disease pathogenesis and possible therapeutic intervention to mitigate fibrosis.
Collapse
|
29
|
Bhandari R, Yang H, Kosarek NN, Smith AE, Garlick JA, Hinchcliff M, Whitfield ML, Pioli PA. Human dermal fibroblast-derived exosomes induce macrophage activation in systemic sclerosis. Rheumatology (Oxford) 2023; 62:SI114-SI124. [PMID: 35946522 PMCID: PMC9910573 DOI: 10.1093/rheumatology/keac453] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 07/29/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES Prior work demonstrates that co-cultured macrophages and fibroblasts from patients with SSc engage in reciprocal activation. However, the mechanism by which these cell types communicate and contribute to fibrosis and inflammation in SSc is unknown. METHODS Fibroblasts were isolated from skin biopsies obtained from 7 SSc patients or 6 healthy age and gender-matched control subjects following written informed consent. Human donor-derived macrophages were cultured with exosomes isolated from control or SSc fibroblasts for an additional 48 h. Macrophages were immunophenotyped using flow cytometry, qRT-PCR and multiplex. For mutual activation studies, exosome-activated macrophages were co-cultured with SSc or healthy fibroblasts using Transwells. RESULTS Macrophages activated with dermal fibroblast-derived exosomes from SSc patients upregulated surface expression of CD163, CD206, MHC Class II and CD16 and secreted increased levels of IL-6, IL-10, IL-12p40 and TNF compared with macrophages incubated with healthy control fibroblasts (n = 7, P < 0.05). Exosome-stimulated macrophages and SSc fibroblasts engaged in reciprocal activation, as production of collagen and fibronectin was significantly increased in SSc fibroblasts receiving signals from SSc exosome-stimulated macrophages (n = 7, P < 0.05). CONCLUSION In this work, we demonstrate for the first time that human SSc dermal fibroblasts mediate macrophage activation through exosomes. Our findings suggest that macrophages and fibroblasts engage in cross-talk in SSc skin, resulting in mutual activation, inflammation, and extracellular matrix (ECM) deposition. Collectively, these studies implicate macrophages and fibroblasts as cooperative mediators of fibrosis in SSc and suggest therapeutic targeting of both cell types may provide maximal benefit in ameliorating disease in SSc patients.
Collapse
Affiliation(s)
| | - Heetaek Yang
- Department of Microbiology and Immunology
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Noelle N Kosarek
- Department of Microbiology and Immunology
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | - Avi E Smith
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA
| | - Jonathan A Garlick
- Department of Diagnostic Science, Tufts University School of Dental Medicine, Boston, MA
| | - Monique Hinchcliff
- Division of Rheumatology, Allergy, and Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Michael L Whitfield
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH
| | | |
Collapse
|
30
|
Chen L, Zhao J, Chao Y, Roy A, Guo W, Qian J, Xu W, Domsic RT, Lafyatis R, Lu B, Deng F, Wang QJ. Loss of Protein Kinase D2 Activity Protects Against Bleomycin-Induced Dermal Fibrosis in Mice. J Transl Med 2023; 103:100018. [PMID: 37039152 PMCID: PMC10507682 DOI: 10.1016/j.labinv.2022.100018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/22/2022] [Accepted: 09/20/2022] [Indexed: 01/11/2023] Open
Abstract
Protein kinase D (PKD) has been linked to inflammatory responses in various pathologic conditions; however, its role in inflammation-induced dermal fibrosis has not been evaluated. In this study, we aimed to investigate the roles and mechanisms of protein kinase D2 (PKD2) in inflammation-induced dermal fibrosis and evaluate the therapeutic potential of PKD inhibitors in this disease. Using homozygous kinase-dead PKD2 knock-in (KI) mice, we examined whether genetic ablation or pharmacologic inhibition of PKD2 activity affected dermal inflammation and fibrosis in a bleomycin (BLM)-induced skin fibrosis model. Our data showed that dermal thickness and collagen fibers were significantly reduced in BLM-treated PKD2 KI mice compared with that in wild-type mice, and so was the expression of α-smooth muscle actin and collagens and the mRNA levels of transforming growth factor-β1 and interleukin-6 in the KI mice. Corroboratively, pharmacologic inhibition of PKD by CRT0066101 also significantly blocked BLM-induced dermal fibrosis and reduced α-smooth muscle actin, collagen, and interleukin-6 expression. Further analyses indicated that loss of PKD2 activity significantly blocked BLM-induced infiltration of monocytes/macrophages and neutrophils in the dermis. Moreover, using bone marrow-derived macrophages, we demonstrated that PKD activity was required for cytokine production and migration of macrophages. We have further identified Akt as a major downstream target of PKD2 in the early inflammatory phase of the fibrotic process. Taken together, our findings indicate that PKD2 promotes dermal fibrosis via regulating immune cell infiltration, cytokine production, and downstream activation of Akt in lesional skin, and targeted inhibition of PKD2 may benefit the treatment of this condition.
Collapse
Affiliation(s)
- Liping Chen
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinjun Zhao
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, India
| | - Wenjing Guo
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiabi Qian
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wanfu Xu
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Robyn T Domsic
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Lafyatis
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
31
|
Chu Y, Bao L, Teng Y, Yuan B, Ma L, Liu Y, Kang H. The Fibrotic Effects of LINC00663 in Human Hepatic Stellate LX-2 Cells and in Bile Duct-Ligated Cholestasis Mice Are Mediated through the Splicing Factor 2-Fibronectin. Cells 2023; 12:cells12020215. [PMID: 36672150 PMCID: PMC9857260 DOI: 10.3390/cells12020215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/25/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Hepatic fibrosis can develop into cirrhosis or even cancer without active therapy at an early stage. Long non-coding RNAs (lncRNAs) have been shown to be involved in the regulation of a wide variety of important biological processes. However, lncRNA mechanism(s) involved in cholestatic liver fibrosis remain unclear. RNA sequence data of hepatic stellate cells from bile duct ligation (BDL) mice or controls were analyzed by weighted gene co-expression network analysis (WGCNA). Based on WGCNA analysis, a competing endogenous RNA network was constructed. We identified LINC00663 and evaluated its function using a panel of assays, including a wound healing assay, a dual-luciferase reporter assay, RNA binding protein immunoprecipitation and chromatin immunoprecipitation. Functional research showed that LINC00663 promoted the activation, migration and epithelial-mesenchymal transition (EMT) of LX-2 cells and liver fibrosis in BDL mice. Mechanistically, LINC00663 regulated splicing factor 2 (SF2)-fibronectin (FN) alternative splicing through the sponging of hsa-miR-3916. Moreover, forkhead box A1 (FOXA1) specifically interacted with the promoter of LINC00663. In summary, we elaborated the fibrotic effects of LINC00663 in human hepatic stellate LX-2 cells and in bile duct-ligated cholestasis mice. We established a FOXA1/LINC00663/hsa-miR-3916/SF2-FN axis that provided a potential target for the diagnosis and targeted therapy of cholestatic liver fibrosis.
Collapse
Affiliation(s)
- Yang Chu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Linan Bao
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Yun Teng
- Department of Laboratory Medicine, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Bo Yuan
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Lijie Ma
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Ying Liu
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Hui Kang
- Department of Laboratory Medicine, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
- Correspondence:
| |
Collapse
|
32
|
Kong LX, Wang Z, Shou YK, Zhou XD, Zong YW, Tong T, Liao M, Han Q, Li Y, Cheng L, Ren B. The FnBPA from methicillin-resistant Staphylococcus aureus promoted development of oral squamous cell carcinoma. J Oral Microbiol 2022; 14:2098644. [PMID: 35859766 PMCID: PMC9291692 DOI: 10.1080/20002297.2022.2098644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is the most common tumor in the oral cavity. Methicillin-resistant Staphylococcus aureus (MRSA) were highly detected in OSCC patients; however, the interactions and mechanisms between drug-resistant bacteria (MRSA) and OSCC are not clear. Aim The aim of this study was to investigate the promotion of MRSA on the development of OSCC. Methods MRSA and MSSA (methicillin-susceptible) strains were employed to investigate the effect on the proliferation of OSCC in vitro and vivo. Results All of the MRSA strains significantly increased the proliferation of OSCC cells and MRSA arrested the cell cycles of OSCC cells in the S phase. MRSA activated the expression of TLR-4, NF-κB and c-fos in OSCC cells. MRSA also promoted the development of squamous cell carcinoma in vivo. The virulence factor fnbpA gene was significantly upregulated in all MRSA strains. By neutralizing FnBPA, the promotions of MRSA on OSCC cell proliferation and development of squamous cell carcinoma were significantly decreased. Meanwhile, the activation of c-fos and NF-κB by MRSA was also significantly decreased by FnBPA antibody. Conclusion MRSA promoted development of OSCC, and the FnBPA protein was the critical virulence factor. Targeting virulence factors is a new method to block the interaction between a drug-resistant pathogen and development of tumors.
Collapse
Affiliation(s)
- Li-Xin Kong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zheng Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yu-Ke Shou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xue-Dong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ya-Wen Zong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Ting Tong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Min Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Qi Han
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yan Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Biao Ren
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
33
|
Kim S, Park HJ, Lee SI. The Microbiome in Systemic Sclerosis: Pathophysiology and Therapeutic Potential. Int J Mol Sci 2022; 23:ijms232416154. [PMID: 36555792 PMCID: PMC9853331 DOI: 10.3390/ijms232416154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/06/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Systemic sclerosis (SSc), also known as scleroderma, is an autoimmune disease with unknown etiology characterized by multi-organ fibrosis. Despite substantial investigation on SSc-related cellular and molecular mechanisms, effective therapies are still lacking. The skin, lungs, and gut are the most affected organs in SSc, which act as physical barriers and constantly communicate with colonized microbiota. Recent reports have documented a unique microbiome signature, which may be the pathogenic trigger or driver of SSc. Since gut microbiota influences the efficacy and toxicity of oral drugs, evaluating drug-microbiota interactions has become an area of interest in disease treatment. The existing evidence highlights the potential of the microbial challenge as a novel therapeutic option in SSc. In this review, we have summarized the current knowledge about molecular mechanisms of SSc and highlighted the underlying role of the microbiome in SSc pathogenesis. We have also discussed the latest therapeutic interventions using microbiomes in SSc, including drug-microbiota interactions and animal disease models. This review aims to elucidate the pathophysiological connection and therapeutic potential of the microbiome in SSc. Insights into the microbiome will significantly improve our understanding of etiopathogenesis and developing therapeutics for SSc.
Collapse
|
34
|
Odell ID, Steach H, Gauld SB, Reinke-Breen L, Karman J, Carr TL, Wetter JB, Phillips L, Hinchcliff M, Flavell RA. Epiregulin is a dendritic cell-derived EGFR ligand that maintains skin and lung fibrosis. Sci Immunol 2022; 7:eabq6691. [PMID: 36490328 PMCID: PMC9840167 DOI: 10.1126/sciimmunol.abq6691] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immune cells are fundamental regulators of extracellular matrix (ECM) production by fibroblasts and have important roles in determining extent of fibrosis in response to inflammation. Although much is known about fibroblast signaling in fibrosis, the molecular signals between immune cells and fibroblasts that drive its persistence are poorly understood. We therefore analyzed skin and lung samples of patients with diffuse cutaneous systemic sclerosis, an autoimmune disease that causes debilitating fibrosis of the skin and internal organs. Here, we define a critical role of epiregulin-EGFR signaling between dendritic cells and fibroblasts to maintain elevated ECM production and accumulation in fibrotic tissue. We found that epiregulin expression marks an inducible state of DC3 dendritic cells triggered by type I interferon and that DC3-derived epiregulin activates EGFR on fibroblasts, driving a positive feedback loop through NOTCH signaling. In mouse models of skin and lung fibrosis, epiregulin was essential for persistence of fibrosis in both tissues, which could be abrogated by epiregulin genetic deficiency or a neutralizing antibody. Therapeutic administration of epiregulin antibody reversed fibrosis in patient skin and lung explants, identifying it as a previously unexplored biologic drug target. Our findings reveal epiregulin as a crucial immune signal that maintains skin and lung fibrosis in multiple diseases and represents a promising antifibrotic target.
Collapse
Affiliation(s)
- Ian D. Odell
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Holly Steach
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | | | - Monique Hinchcliff
- Department of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale School of Medicine, New Haven, CT, USA
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
35
|
Abstract
Renal fibrosis is a hallmark of end-stage chronic kidney disease. It is characterized by increased accumulation of extracellular matrix (ECM), which disrupts cellular organization and function within the kidney. Here, we review the bi-directional interactions between cells and the ECM that drive renal fibrosis. We will discuss the cells involved in renal fibrosis, changes that occur in the ECM, the interactions between renal cells and the surrounding fibrotic microenvironment, and signal transduction pathways that are misregulated as fibrosis proceeds. Understanding the underlying mechanisms of cell-ECM crosstalk will identify novel targets to better identify and treat renal fibrosis and associated renal disease.
Collapse
Affiliation(s)
- Kristin P. Kim
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Caitlin E. Williams
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Christopher A. Lemmon
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
36
|
Wang W, Bale S, Yalavarthi B, Verma P, Tsou PS, Calderone KM, Bhattacharyya D, Fisher GJ, Varga J, Bhattacharyya S. Deficiency of inhibitory TLR4 homolog RP105 exacerbates fibrosis. JCI Insight 2022; 7:e160684. [PMID: 36136452 PMCID: PMC9675479 DOI: 10.1172/jci.insight.160684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
Activation of TLR4 by its cognate damage-associated molecular patterns (DAMPs) elicits potent profibrotic effects and myofibroblast activation in systemic sclerosis (SSc), while genetic targeting of TLR4 or its DAMPs in mice accelerates fibrosis resolution. To prevent aberrant DAMP/TLR4 activity, a variety of negative regulators evolved to dampen the magnitude and duration of the signaling. These include radioprotective 105 kDa (RP105), a transmembrane TLR4 homolog that competitively inhibits DAMP recognition of TLR4, blocking TLR4 signaling in immune cells. The role of RP105 in TLR4-dependent fibrotic responses in SSc is unknown. Using unbiased transcriptome analysis of skin biopsies, we found that levels of both TLR4 and its adaptor protein MD2 were elevated in SSc skin and significantly correlated with each other. Expression of RP105 was negatively associated with myofibroblast differentiation in SSc. Importantly, RP105-TLR4 association was reduced, whereas TLR4-TLR4 showed strong association in fibroblasts from patients with SSc, as evidenced by PLA assays. Moreover, RP105 adaptor MD1 expression was significantly reduced in SSc skin biopsies and explanted SSc skin fibroblasts. Exogenous RP105-MD1 abrogated, while loss of RP105 exaggerated, fibrotic cellular responses. Importantly, ablation of RP105 in mice was associated with augmented TLR4 signaling and aggravated skin fibrosis in complementary disease models. Thus, we believe RP105-MD1 to be a novel cell-intrinsic negative regulator of TLR4-MD2-driven sustained fibroblast activation, representing a critical regulatory network governing the fibrotic process. Impaired RP105 function in SSc might contribute to persistence of progression of the disease.
Collapse
Affiliation(s)
- Wenxia Wang
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Swarna Bale
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Illinois, USA
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Priyanka Verma
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Pei-Suen Tsou
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Ken M. Calderone
- Derpartment of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Dibyendu Bhattacharyya
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Gary J. Fisher
- Derpartment of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Illinois, USA
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| | - Swati Bhattacharyya
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Illinois, USA
- Michigan Scleroderma Program, Division of Rheumatology, Department of Internal Medicine, and
| |
Collapse
|
37
|
Wang W, Bale S, Wei J, Yalavarthi B, Bhattacharyya D, Yan JJ, Abdala-Valencia H, Xu D, Sun H, Marangoni RG, Herzog E, Berdnikovs S, Miller SD, Sawalha AH, Tsou PS, Awaji K, Yamashita T, Sato S, Asano Y, Tiruppathi C, Yeldandi A, Schock BC, Bhattacharyya S, Varga J. Fibroblast A20 governs fibrosis susceptibility and its repression by DREAM promotes fibrosis in multiple organs. Nat Commun 2022; 13:6358. [PMID: 36289219 PMCID: PMC9606375 DOI: 10.1038/s41467-022-33767-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 09/29/2022] [Indexed: 02/04/2023] Open
Abstract
In addition to autoimmune and inflammatory diseases, variants of the TNFAIP3 gene encoding the ubiquitin-editing enzyme A20 are also associated with fibrosis in systemic sclerosis (SSc). However, it remains unclear how genetic factors contribute to SSc pathogenesis, and which cell types drive the disease due to SSc-specific genetic alterations. We therefore characterize the expression, function, and role of A20, and its negative transcriptional regulator DREAM, in patients with SSc and disease models. Levels of A20 are significantly reduced in SSc skin and lungs, while DREAM is elevated. In isolated fibroblasts, A20 mitigates ex vivo profibrotic responses. Mice haploinsufficient for A20, or harboring fibroblasts-specific A20 deletion, recapitulate major pathological features of SSc, whereas DREAM-null mice with elevated A20 expression are protected. In DREAM-null fibroblasts, TGF-β induces the expression of A20, compared to wild-type fibroblasts. An anti-fibrotic small molecule targeting cellular adiponectin receptors stimulates A20 expression in vitro in wild-type but not A20-deficient fibroblasts and in bleomycin-treated mice. Thus, A20 has a novel cell-intrinsic function in restraining fibroblast activation, and together with DREAM, constitutes a critical regulatory network governing the fibrotic process in SSc. A20 and DREAM represent novel druggable targets for fibrosis therapy.
Collapse
Affiliation(s)
- Wenxia Wang
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Swarna Bale
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jun Wei
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Bharath Yalavarthi
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Dibyendu Bhattacharyya
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jing Jing Yan
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Dan Xu
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Hanshi Sun
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Roberta G Marangoni
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA
| | - Erica Herzog
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Sergejs Berdnikovs
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Amr H Sawalha
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Pei-Suen Tsou
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kentaro Awaji
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Chinnaswamy Tiruppathi
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, IL, USA
| | - Anjana Yeldandi
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bettina C Schock
- Wellcome-Wolfson Institute for Experimental Medicine, Queens University Belfast, Belfast, UK
| | - Swati Bhattacharyya
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA.
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - John Varga
- Northwestern Scleroderma Program, Department of Medicine, Feinberg School of Medicine, Chicago, IL, USA.
- Michigan Scleroderma Program, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
38
|
Tuncer F, Bulik M, Villandre J, Lear T, Chen Y, Tuncer B, Kass DJ, Valenzi E, Morse C, Sembrat J, Lafyatis R, Chen B, Evankovich J. Fibronectin-EDA accumulates via reduced ubiquitination downstream of Toll-like receptor 9 activation in SSc-ILD fibroblasts. Am J Physiol Lung Cell Mol Physiol 2022; 323:L484-L494. [PMID: 35997276 PMCID: PMC9550569 DOI: 10.1152/ajplung.00019.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022] Open
Abstract
Accumulation of excessive extracellular matrix (ECM) components from lung fibroblasts is a feature of systemic sclerosis-associated interstitial lung disease (SSc-ILD), and there is increasing evidence that innate immune signaling pathways contribute to these processes. Toll-like receptors (TLRs) are innate immune sensors activated by danger signals derived from pathogens or host molecular patterns. Several damage-associated molecular pattern (DAMP) molecules are elevated in SSc-ILD plasma, including ligands that activate TLR9, an innate immune sensor recently implicated in driving profibrotic responses in fibroblasts. Fibronectin and the isoform fibronectin-extra domain A (FN-EDA) are prominent in pathological extracellular matrix accumulation, but mechanisms promoting FN-EDA accumulation are only partially understood. Here, we show that TLR9 activation increases FN-EDA accumulation in MRC5 and SSc-ILD fibroblasts, but that this effect is independent of changes in FN-EDA gene transcription. Rather, we describe a novel mechanism where TLR9 activation inhibits FN-EDA turnover via reduced FN-EDA ubiquitination. TLR9 ligand ODN2006 reduces ubiquitinated FN-EDA destined for lysosomal degradation, an effect abrogated with TLR9 knockdown or inhibition. Taken together, these results provide rationale for disrupting the TLR9 signaling axis or FN-EDA degradation pathways to reduce FN-EDA accumulation in SSc-ILD fibroblasts. More broadly, enhancing intracellular degradation of ECM components through TLR9 inhibition or enhanced ECM turnover could be a novel strategy to attenuate pathogenic ECM accumulation in SSc-ILD.
Collapse
Affiliation(s)
- Ferhan Tuncer
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melissa Bulik
- Division of Rheumatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Villandre
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Travis Lear
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yanwen Chen
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Beyza Tuncer
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Daniel J Kass
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christina Morse
- Division of Rheumatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert Lafyatis
- Simmons Center for Interstitial Lung Disease, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Rheumatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bill Chen
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Evankovich
- Aging Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
39
|
Liu X, Lu F, Chen X. Examination of the role of necroptotic damage-associated molecular patterns in tissue fibrosis. Front Immunol 2022; 13:886374. [PMID: 36110858 PMCID: PMC9468929 DOI: 10.3389/fimmu.2022.886374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022] Open
Abstract
Fibrosis is defined as the abnormal and excessive deposition of extracellular matrix (ECM) components, which leads to tissue or organ dysfunction and failure. However, the pathological mechanisms underlying fibrosis remain unclear. The inflammatory response induced by tissue injury is closely associated with tissue fibrosis. Recently, an increasing number of studies have linked necroptosis to inflammation and fibrosis. Necroptosis is a type of preprogrammed death caused by death receptors, interferons, Toll-like receptors, intracellular RNA and DNA sensors, and other mediators. These activate receptor-interacting protein kinase (RIPK) 1, which recruits and phosphorylates RIPK3. RIPK3 then phosphorylates a mixed lineage kinase domain-like protein and causes its oligomerization, leading to rapid plasma membrane permeabilization, the release of cellular contents, and exposure of damage-associated molecular patterns (DAMPs). DAMPs, as inflammatory mediators, are involved in the loss of balance between extensive inflammation and tissue regeneration, leading to remodeling, the hallmark of fibrosis. In this review, we discuss the role of necroptotic DAMPs in tissue fibrosis and highlight the inflammatory responses induced by DAMPs in tissue ECM remodeling. By summarizing the existing literature on this topic, we underscore the gaps in the current research, providing a framework for future investigations into the relationship among necroptosis, DAMPs, and fibrosis, as well as a reference for later transformation into clinical treatment.
Collapse
Affiliation(s)
| | - Feng Lu
- *Correspondence: Feng Lu, ; Xihang Chen,
| | | |
Collapse
|
40
|
Dooling LJ, Saini K, Anlaş AA, Discher DE. Tissue mechanics coevolves with fibrillar matrisomes in healthy and fibrotic tissues. Matrix Biol 2022; 111:153-188. [PMID: 35764212 PMCID: PMC9990088 DOI: 10.1016/j.matbio.2022.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/16/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022]
Abstract
Fibrillar proteins are principal components of extracellular matrix (ECM) that confer mechanical properties to tissues. Fibrosis can result from wound repair in nearly every tissue in adults, and it associates with increased ECM density and crosslinking as well as increased tissue stiffness. Such fibrotic tissues are a major biomedical challenge, and an emerging view posits that the altered mechanical environment supports both synthetic and contractile myofibroblasts in a state of persistent activation. Here, we review the matrisome in several fibrotic diseases, as well as normal tissues, with a focus on physicochemical properties. Stiffness generally increases with the abundance of fibrillar collagens, the major constituent of ECM, with similar mathematical trends for fibrosis as well as adult tissues from soft brain to stiff bone and heart development. Changes in expression of other core matrisome and matrisome-associated proteins or proteoglycans contribute to tissue stiffening in fibrosis by organizing collagen, crosslinking ECM, and facilitating adhesion of myofibroblasts. Understanding how ECM composition and mechanics coevolve during fibrosis can lead to better models and help with antifibrotic therapies.
Collapse
Affiliation(s)
- Lawrence J Dooling
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Karanvir Saini
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Alişya A Anlaş
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA
| | - Dennis E Discher
- Molecular and Cellular Biophysics Lab, University of Pennsylvania,Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Tenascin-C in fibrosis in multiple organs: Translational implications. Semin Cell Dev Biol 2022; 128:130-136. [PMID: 35400564 PMCID: PMC10119770 DOI: 10.1016/j.semcdb.2022.03.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 12/28/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex disease with a pathogenic triad of autoimmunity, vasculopathy, and fibrosis involving the skin and multiple internal organs [1]. Because fibrosis accounts for as much as 45% of all deaths worldwide and appears to be increasing in prevalence [2], understanding its pathogenesis and progression is an urgent scientific challenge. Fibroblasts and myofibroblasts are the key effector cells executing physiologic tissue repair on one hand, and pathological fibrogenesis leading to chronic fibrosing conditions on the other. Recent studies identify innate immune signaling via toll-like receptors (TLRs) as a key driver of persistent fibrotic response in SSc. Repeated injury triggers the in-situ generation of "damage-associated molecular patterns" (DAMPs) or danger signals. Sensing of these danger signals by TLR4 on resident cells elicits potent stimulatory effects on fibrotic gene expression and myofibroblast differentiation triggering the self-limited tissue repair response to self-sustained pathological fibrosis characteristic of SSc. Our unbiased survey for DAMPs associated with SSc identified extracellular matrix glycoprotein tenascin-C as one of the most highly up-regulated ECM proteins in SSc skin and lung biopsies [3,4]. Furthermore, tenascin C is responsible for driving sustained fibroblasts activation, thereby progression of fibrosis [3]. This review summarizes recent studies examining the regulation and complex functional role of tenascin C, presenting tenascin-TLR4 axis in pathological fibrosis, and novel anti-fibrotic approaches targeting their signaling.
Collapse
|
42
|
Fibronectin Functions as a Selective Agonist for Distinct Toll-like Receptors in Triple-Negative Breast Cancer. Cells 2022; 11:cells11132074. [PMID: 35805158 PMCID: PMC9265717 DOI: 10.3390/cells11132074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/17/2022] [Accepted: 06/25/2022] [Indexed: 02/04/2023] Open
Abstract
The microenvironment of tumors is characterized by structural changes in the fibronectin matrix, which include increased deposition of the EDA isoform of fibronectin and the unfolding of the fibronectin Type III domains. The impact of these structural changes on tumor progression is not well understood. The fibronectin EDA (FnEDA) domain and the partially unfolded first Type III domain of fibronectin (FnIII-1c) have been identified as endogenous damage-associated molecular pattern molecules (DAMPs), which induce innate immune responses by serving as agonists for Toll-Like Receptors (TLRs). Using two triple-negative breast cancer (TNBC) cell lines MDA-MB-468 and MDA-MB-231, we show that FnEDA and FnIII-1c induce the pro-tumorigenic cytokine, IL-8, by serving as agonists for TLR5 and TLR2, the canonical receptors for bacterial flagellin and lipoprotein, respectively. We also find that FnIII-1c is not recognized by MDA-MB-468 cells but is recognized by MDA-MB-231 cells, suggesting a cell type rather than ligand specific utilization of TLRs. As IL-8 plays a major role in the progression of TNBC, these studies suggest that tumor-induced structural changes in the fibronectin matrix promote an inflammatory microenvironment conducive to metastatic progression.
Collapse
|
43
|
Shutova MS, Boehncke WH. Mechanotransduction in Skin Inflammation. Cells 2022; 11:2026. [PMID: 35805110 PMCID: PMC9265324 DOI: 10.3390/cells11132026] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/16/2022] Open
Abstract
In the process of mechanotransduction, the cells in the body perceive and interpret mechanical stimuli to maintain tissue homeostasis and respond to the environmental changes. Increasing evidence points towards dysregulated mechanotransduction as a pathologically relevant factor in human diseases, including inflammatory conditions. Skin is the organ that constantly undergoes considerable mechanical stresses, and the ability of mechanical factors to provoke inflammatory processes in the skin has long been known, with the Koebner phenomenon being an example. However, the molecular mechanisms and key factors linking mechanotransduction and cutaneous inflammation remain understudied. In this review, we outline the key players in the tissue's mechanical homeostasis, the available data, and the gaps in our current understanding of their aberrant regulation in chronic cutaneous inflammation. We mainly focus on psoriasis as one of the most studied skin inflammatory diseases; we also discuss mechanotransduction in the context of skin fibrosis as a result of chronic inflammation. Even though the role of mechanotransduction in inflammation of the simple epithelia of internal organs is being actively studied, we conclude that the mechanoregulation in the stratified epidermis of the skin requires more attention in future translational research.
Collapse
Affiliation(s)
- Maria S. Shutova
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland;
- Department of Dermatology, Geneva University Hospitals, 1211 Geneva, Switzerland
| |
Collapse
|
44
|
Fibronectin containing alternatively spliced extra domain A interacts at the central and c-terminal domain of Toll-like receptor-4. Sci Rep 2022; 12:9662. [PMID: 35690624 PMCID: PMC9188610 DOI: 10.1038/s41598-022-13622-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/18/2022] [Indexed: 11/08/2022] Open
Abstract
Extra domain A of cellular fibronectin (FN-EDA) is known to cause insulin resistance, atherosclerosis, tissue fibrosis, ischemic stroke and exaggerated myocardial reperfusion injury through Toll-like receptor 4 (TLR4). However, the FN-EDA-TLR4 interacting site is not well established. Therefore, in-silico approaches have been used to study FN-EDA and TLR4 interactions at the interface. In the present study, molecular docking studies of FN-EDA with TLR4-myeloid differentiation factor 2 (MD2) heterodimer have been performed to unravel the FN-EDA-TLR4 interacting sequence. Furthermore, the modulatory role of FN-EDA adjacent domains FNIII(11) and FNIII(12) on its interaction with TLR4-MD2 was investigated. The results show that FN-EDA interacting sequence “SPEDGIRELF” selectively interacts with TLR4 directly near its central and C-terminal domain region. The regulatory domains, FN type III 11 facilitate and 12 impede the FN-EDA-TLR4 interaction. Furthermore, the molecular dynamic simulation studies confirmed that FN-EDA forms a stable complex with TLR4-MD2 heterodimer. In conclusion, FN-EDA interacts and forms a stable complex through its “SPEDGIRELF” sequence at the central and C-terminal domain region of TLR4. The revelation of FN-EDA and TLR4 interacting sites may help design novel therapeutics for drug discovery research.
Collapse
|
45
|
Menko AS, Romisher A, Walker JL. The Pro-fibrotic Response of Mesenchymal Leader Cells to Lens Wounding Involves Hyaluronic Acid, Its Receptor RHAMM, and Vimentin. Front Cell Dev Biol 2022; 10:862423. [PMID: 35386200 PMCID: PMC8977891 DOI: 10.3389/fcell.2022.862423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/08/2022] [Indexed: 12/31/2022] Open
Abstract
Hyaluronic Acid/Hyaluronan (HA) is a major component of the provisional matrix deposited by cells post-wounding with roles both in regulating cell migration to repair a wound and in promoting a fibrotic outcome to wounding. Both are mediated through its receptors CD44 and RHAMM. We now showed that HA is present in the provisional matrix assembled on the substrate surface in a lens post-cataract surgery explant wound model in which mesenchymal leader cells populate the wound edges to direct migration of the lens epithelium across the adjacent culture substrate onto which this matrix is assembled. Inhibiting HA expression with 4-MU blocked assembly of FN-EDA and collagen I by the wound-responsive mesenchymal leader cells and their migration. These cells express both the HA receptors CD44 and RHAMM. CD44 co-localized with HA at their cell-cell interfaces. RHAMM was predominant in the lamellipodial protrusions extended by the mesenchymal cells at the leading edge, and along HA fibrils organized on the substrate surface. Within a few days post-lens wounding the leader cells are induced to transition to αSMA+ myofibroblasts. Since HA/RHAMM is implicated in both cell migration and inducing fibrosis we examined the impact of blocking HA synthesis on myofibroblast emergence and discovered that it was dependent on HA. While RHAMM has not been previously linked to the intermediate filament protein vimentin, our studies with these explant cultures have shown that vimentin in the cells’ lamellipodial protrusions regulate their transition to myofibroblast. PLA studies now revealed that RHAMM was complexed with both HA and vimentin in the lamellipodial protrusions of leader cells, implicating this HA/RHAMM/vimentin complex in the regulation of leader cell function post-wounding, both in promoting cell migration and in the transition of these cells to myofibroblasts. These results increase our understanding of how the post-wounding matrix environment interacts with receptor/cytoskeletal complexes to determine whether injury outcomes are regenerative or fibrotic.
Collapse
Affiliation(s)
- A Sue Menko
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alison Romisher
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Janice L Walker
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
46
|
Bolourani S, Sari E, Brenner M, Wang P. The role of eCIRP in bleomycin-induced pulmonary fibrosis in mice. PLoS One 2022; 17:e0266163. [PMID: 35377906 PMCID: PMC8979429 DOI: 10.1371/journal.pone.0266163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 03/15/2022] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE AND DESIGN We examined the role of eCIRP in the pathogenesis of bleomycin-induced pulmonary fibrosis (PF). MATERIAL AND METHODS Publicly available gene expression omnibus datasets were analyzed for the expression of CIRP in lung samples from patients with PF. Wild type (WT) or CIRP-/- mice received daily injections of 10 μg/g bleomycin for 10 days. A subset of bleomycin-injected WT mice was treated with the eCIRP antagonist C23 (8 μg/g/day) from day 10 to day 19. At three weeks, transthoracic echocardiography was performed to measure the degree of pulmonary hypertension, and lung tissues were collected and analyzed for markers of fibrosis. RESULTS Analysis of the mRNA data of human lung samples showed a significant positive correlation between CIRP and α-smooth muscle actin (α-SMA), an important marker of fibrosis. Moreover, the expression of CIRP was higher in patients with acute exacerbation of PF than in patients with stable PF. CIRP-/- mice showed attenuated induction of α-SMA and collagens (Col1a1, Col3a1), reduced hydroxyproline content, decreased histological fibrosis scores, and improved pulmonary hypertension as compared to WT mice. WT mice treated with C23 also had significant attenuation of the above endpoint measure. CONCLUSIONS Our study demonstrates that eCIRP plays a key role in promoting the development of PF, and blocking eCIRP with C23 can significantly attenuate this process.
Collapse
Affiliation(s)
- Siavash Bolourani
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States of America
| | - Ezgi Sari
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States of America
| | - Max Brenner
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States of America
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States of America
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States of America
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States of America
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States of America
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States of America
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States of America
| |
Collapse
|
47
|
Ocon A, Lokineni S, Korman B. Understanding and Therapeutically Targeting the Scleroderma Myofibroblast. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2022. [DOI: 10.1007/s40674-021-00189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
48
|
Rosendahl AH, Schönborn K, Krieg T. Pathophysiology of systemic sclerosis (scleroderma). Kaohsiung J Med Sci 2022; 38:187-195. [PMID: 35234358 DOI: 10.1002/kjm2.12505] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 01/18/2022] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (scleroderma) is an autoimmune-triggered chronic fibrosing disease that affects the skin and many other organs. Its pathophysiology is complex and involves an early endothelial damage, an inflammatory infiltrate and a resulting fibrotic reaction. Based on a predisposing genetic background, an altered balance of the acquired and the innate immune system leads to the release of many cytokines and chemokines as well as autoantibodies, which induce the activation of fibroblasts with the formation of myofibroblasts and the deposition of a stiff and rigid connective tissue. A curative treatment is still not available but remarkable progress has been made in the management of organ complications. In addition, several breakthroughs in the pathophysiology have led to new therapeutic concepts. Based on these, many new compounds have been developed during the last years, which target these different pathways and offer specific therapeutic approaches.
Collapse
Affiliation(s)
- Ann-Helen Rosendahl
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Katrin Schönborn
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Department of Dermatology, University Hospital of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Xu D, Bhattacharyya S, Wang W, Ifergan I, Chiang Wong MYA, Procissi D, Yeldandi A, Bale S, Marangoni RG, Horbinski C, Miller SD, Varga J. PLG nanoparticles target fibroblasts and MARCO+ monocytes to reverse multi-organ fibrosis. JCI Insight 2022; 7:151037. [PMID: 35104243 PMCID: PMC8983146 DOI: 10.1172/jci.insight.151037] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 01/26/2022] [Indexed: 11/17/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic, multisystem orphan disease with a highly variable clinical course, high mortality rate, and a poorly understood complex pathogenesis. We have identified an important role for a subpopulation of monocytes and macrophages characterized by surface expression of the scavenger receptor macrophage receptor with collagenous structure (MARCO) in chronic inflammation and fibrosis in SSc and in preclinical disease models. We show that MARCO+ monocytes and macrophages accumulate in lesional skin and lung in topographic proximity to activated myofibroblasts in patients with SSc and in the bleomycin-induced mouse model of SSc. Short-term treatment of mice with a potentially novel nanoparticle, poly(lactic-co-glycolic) acid (PLG), which is composed of a carboxylated, FDA-approved, biodegradable polymer and modulates activation and trafficking of MARCO+ inflammatory monocytes, markedly attenuated bleomycin-induced skin and lung inflammation and fibrosis. Mechanistically, in isolated cells in culture, PLG nanoparticles inhibited TGF-dependent fibrotic responses in vitro. Thus, MARCO+ monocytes are potent effector cells of skin and lung fibrosis and can be therapeutically targeted in SSc using PLG nanoparticles.
Collapse
Affiliation(s)
- Dan Xu
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Swati Bhattacharyya
- Department of Internal Medicine, University of Michigan, Ann Arbor, United States of America
| | - Wenxia Wang
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Igal Ifergan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Ming-Yi Alice Chiang Wong
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Daniele Procissi
- Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Anjana Yeldandi
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Swarna Bale
- Department of Internal Medicine, University of Michigan, Ann Arbor, United States of America
| | - Roberta G Marangoni
- Northwestern Scleroderma Program, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Stephen D Miller
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - John Varga
- Department of Internal Medicine, University of Michigan, Ann Arbor, United States of America
| |
Collapse
|
50
|
Keller KE, Peters DM. Pathogenesis of glaucoma: Extracellular matrix dysfunction in the trabecular meshwork-A review. Clin Exp Ophthalmol 2022; 50:163-182. [PMID: 35037377 DOI: 10.1111/ceo.14027] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/19/2021] [Accepted: 11/26/2021] [Indexed: 12/11/2022]
Abstract
The trabecular meshwork regulates aqueous humour outflow from the anterior chamber of the eye. It does this by establishing a tunable outflow resistance, defined by the interplay between cells and their extracellular matrix (ECM) milieu, and the molecular interactions between ECM proteins. During normal tissue homeostasis, the ECM is remodelled and trabecular cell behaviour is modified, permitting increased aqueous fluid outflow to maintain intraocular pressure (IOP) within a relatively narrow physiological pressure. Dysfunction in the normal homeostatic process leads to increased outflow resistance and elevated IOP, which is a primary risk factor for glaucoma. This review delineates some of the changes in the ECM that lead to gross as well as some more subtle changes in the structure and function of the ECM, and their impact on trabecular cell behaviour. These changes are discussed in the context of outflow resistance and glaucoma.
Collapse
Affiliation(s)
- Kate E Keller
- Casey Eye Institute, Oregon Health &Science University, Portland, Oregon, USA
| | - Donna M Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin, USA
| |
Collapse
|