1
|
Campitiello R, Soldano S, Gotelli E, Hysa E, Montagna P, Casabella A, Paolino S, Pizzorni C, Sulli A, Smith V, Cutolo M. The intervention of macrophages in progressive fibrosis characterizing systemic sclerosis: A systematic review. Autoimmun Rev 2024; 23:103637. [PMID: 39255852 DOI: 10.1016/j.autrev.2024.103637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND AND AIM Systemic sclerosis (SSc) is an immune mediated connective tissue disease characterized by microvascular dysfunction, aberrant immune response, and progressive fibrosis. Although the immuno-pathophysiological mechanisms underlying SSc are not fully clarified, they are often associated with a dysfunctional macrophage activation toward an alternative (M2) phenotype induced by cytokines [i.e., IL-4, IL-10, IL-13, and transforming growth factor (TGF-β)] involved in the fibrotic and anti-inflammatory process. A spectrum of macrophage activation state has been identified ranging from M1 to M2 phenotype, gene expression of phenotype markers, and functional aspects. This systematic review aims to analyze the importance of M2 macrophage polatization during the immune mediated process and the identification of specific pathways, cytokines, and chemokines involved in SSc pathogenesis. Moreover, this review provides an overview on the in vitro and in vivo studies aiming to test therapeutic strategies targeting M2 macrophages. METHODS A systematic literature review was performed according to the preferred Reported Items for Systematic Reviews and Meta-Analyses (PRISMA). The search encompassed the online medical databases PubMed and Embase up to the 30th of June 2024. Original research manuscripts (in vitro study, in vivo study), animal model and human cohort, were considered for the review. Exclusion criteria encompassed reviews, case reports, correspondences, and conference abstracts/posters. The eligible manuscripts main findings were critically analyzed, discussed, and summarized in the correspondent tables. RESULTS Out of the 77 screened abstracts, 49 papers were deemed eligible. Following a critical analysis, they were categorized according to the primary (29 original articles) and secondary (20 original articles) research objectives of this systematic review. The data from the present systematic review suggest the pivotal role of M2 macrophages differentiation and activation together with the dysregulation of the immune system in the SSc pathogenesis. Strong correlations have been found between M2 macrophage presence and clinical manifestations in both murine and human tissue samples. Interestingly, the presence of M2 cell surface markers on peripheral blood monocytes has been highlighted, suggesting a potential biomarker role for this finding. Therapeutic effects reducing M2 macrophage activities have been observed and/or tested for existing and for new drugs, demonstrating potential efficacy in modulating the pro-fibrotic immune response for treatment of SSc. CONCLUSIONS The increased M2 macrophage activation in course of SSc seems to offer new insights on the self-amplifying inflammatory and fibrotic response by the immune system on such disease. Therefore, the revaluation of immunomodulatory and ongoing antifibrotic therapies, as well as novel therapeutical approaches in SSc that contribute to limit the M2 macrophage activation are matter of intense investigations.
Collapse
Affiliation(s)
- Rosanna Campitiello
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Stefano Soldano
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy.
| | - Emanuele Gotelli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy
| | - Elvis Hysa
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | - Paola Montagna
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy.
| | - Andrea Casabella
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Sabrina Paolino
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Carmen Pizzorni
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Alberto Sulli
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, University of Ghent, Ghent, Belgium; Department of Internal Medicine, Ghent University Hospital, University of Ghent, Ghent, Belgium; Unit for Molecular Immunology and Inflammation, Flemish Institute for Biotechnology, Inflammation Research Center, Ghent, Belgium.
| | - Maurizio Cutolo
- Laboratory of Experimental Rheumatology and Academic Division of Clinical Rheumatology, Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genova, Genova, Italy; IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| |
Collapse
|
2
|
Owaki T, Iida T, Miyai Y, Kato K, Hase T, Ishii M, Ando R, Hinohara K, Akashi T, Mizutani Y, Ishikawa T, Mii S, Shiraki Y, Esaki N, Yamamoto M, Tsukamoto T, Nomura S, Murakami T, Takahashi M, Yuguchi Y, Maeda M, Sano T, Sassa N, Matsukawa Y, Kawashima H, Akamatsu S, Enomoto A. Synthetic retinoid-mediated preconditioning of cancer-associated fibroblasts and macrophages improves cancer response to immune checkpoint blockade. Br J Cancer 2024; 131:372-386. [PMID: 38849479 PMCID: PMC11263587 DOI: 10.1038/s41416-024-02734-3] [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] [Academic Contribution Register] [Received: 11/29/2023] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND The proliferation of cancer-associated fibroblasts (CAFs) hampers drug delivery and anti-tumor immunity, inducing tumor resistance to immune checkpoint blockade (ICB) therapy. However, it has remained a challenge to develop therapeutics that specifically target or modulate CAFs. METHODS We investigated the involvement of Meflin+ cancer-restraining CAFs (rCAFs) in ICB efficacy in patients with clear cell renal cell carcinoma (ccRCC) and urothelial carcinoma (UC). We examined the effects of Am80 (a synthetic retinoid) administration on CAF phenotype, the tumor immune microenvironment, and ICB efficacy in cancer mouse models. RESULTS High infiltration of Meflin+ CAFs correlated with ICB efficacy in patients with ccRCC and UC. Meflin+ CAF induction by Am80 administration improved ICB efficacy in the mouse models of cancer. Am80 exerted this effect when administered prior to, but not concomitant with, ICB therapy in wild-type but not Meflin-deficient mice. Am80-mediated induction of Meflin+ CAFs was associated with increases in antibody delivery and M1-like tumor-associated macrophage (TAM) infiltration. Finally, we showed the role of Chemerin produced from CAFs after Am80 administration in the induction of M1-like TAMs. CONCLUSION Our data suggested that Am80 administration prior to ICB therapy increases the number of Meflin+ rCAFs and ICB efficacy by inducing changes in TAM phenotype.
Collapse
Affiliation(s)
- Takayuki Owaki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tadashi Iida
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Yuki Miyai
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Ando
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kunihiko Hinohara
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
| | - Tomohiro Akashi
- Division of Systems Biology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuyuki Mizutani
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shinji Mii
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Shiraki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobutoshi Esaki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masami Yamamoto
- Laboratory of Physiological Pathology, Nippon Veterinary and Life Science University, Tokyo, Japan
| | - Tetsuya Tsukamoto
- Division of Analytical Pathology, Oncology Innovation Center, Fujita Health University, Toyoake, Japan
| | - Sachiyo Nomura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Clinical Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Hoshi University, Tokyo, Japan
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Saitama, Japan
| | - Masahide Takahashi
- Department of Pathology, Fujita Health University, Toyoake, Japan
- International Center for Cell and Gene Therapy, Fujita Health University, Toyoake, Japan
| | - Yuri Yuguchi
- Department of Urology, Chukyo Hospital, Nagoya, Japan
| | | | - Tomoyasu Sano
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoto Sassa
- Department of Urology, Aichi Medical University, Nagakute, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Center for One Medicine Innovative Translational Research, Gifu University Institute for Advanced Study, Gifu, Japan.
| |
Collapse
|
3
|
Xu C, Li J. Tamibarotene targets heparin-binding protein for attenuating lung injury in sepsis. Allergol Immunopathol (Madr) 2023; 51:124-130. [PMID: 37422788 DOI: 10.15586/aei.v51i4.900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/04/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Excessively active pulmonary inflammation is a hallmark of sepsis-induced lung damage. A synthetic retinoid drug called tamibarotene reduces inflammation in a variety of conditions, including acute promyelocytic leukemia (APL), renal fibrosis, and neuroinflammation. Its effect on sepsis-related lung injury, however, has not been explained. PURPOSE The purpose of the study was to investigate how tamibarotene affected lung damage induced by cecal ligation and puncture (CLP) procedure. METHODS A CLP sepsis mouse model was developed, and tamibarotene was pretreated to determine whether it improved lung injury and survival. The degree of lung injury was evaluated using the Hematoxylin and eosin staining and lung injury score. In order to determine pulmonary vascular permeability, measurements were taken for total protein and cell content of bronchoalveolar lavage fluid (BALF), wet/dry ratio of the lung, and Evans blue stain. The BALF inflammatory mediators, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), IL-1β, and IL-17A were discovered by enzyme-linked immunosorbent serologic assay (ELISA). Then, the levels of heparin-binding protein (HBP), and phospho-nuclear factor kappa-B (p-NF-κB) P65, and NF-κB P65 were determined using ELISA and Western blot analysis, respectively. RESULTS Tamibarotene considerably increases survival and lessens lung damage stimulated by sepsis. Specifically, tamibarotene significantly relieves pulmonary vascular permeability and inhibits inflammation response in sepsis. Moreover, we further confirmed that these ameliorating effects of tamibarotene on sepsis may be exerted by targeting HBP and regulating the activation of NF-κB signaling pathway. CONCLUSION These findings demonstrated that tamibarotene lessens sepsis-induced lung injury, and the effect could be exerted by targeting HBP and thereby deregulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Chuanjie Xu
- Department of Neurocritical Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou Province, China
| | - Jianping Li
- Department of Neurocritical Medicine, The Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou Province, China;
| |
Collapse
|
4
|
The Therapeutic Effects of Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells on Scleroderma. Tissue Eng Regen Med 2021; 19:141-150. [PMID: 34784013 PMCID: PMC8782977 DOI: 10.1007/s13770-021-00405-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/28/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 11/15/2022] Open
Abstract
Background: Scleroderma is a multisystem disease in which tissue fibrosis is caused by inflammation and vascular damage. The mortality of scleroderma has remained high due to a lack of effective treatments. However, exosomes derived from human umbilical cord mesenchymal stem cells (HUMSCs)-Ex have been regarded as potential treatments for various autoimmune diseases, and may also act as candidates for treating scleroderma. Methods: Mice with scleroderma received a single 50 μg HUMSCs-Ex. HUMSCs-Ex was characterized using transmission electron microscopy, nanoparticle tracking analysis and nanoflow cytometry. The therapeutic efficacy was assessed using histopathology, immunohistochemistry, immunofluorescence, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay and western blot. Results: HUMSCs-Ex ameliorated the deposition of extracellular matrix and suppressed the epithelial-mesenchymal transition process, and the effects lasted at least three weeks. In addition, HUMSCs-Ex promoted M1 macrophage polarization and inhibited M2 macrophage polarization, leading to the restoration of the balance of M1/M2 macrophages. Conclusion: We investigated the potential antifibrotic and anti-inflammatory effects of HUMSCs-Ex in a bleomycin-induced mouse model of scleroderma. So HUMSCs-Ex could be considered as a candidate therapy for scleroderma. Supplementary Information The online version contains supplementary material available at 10.1007/s13770-021-00405-5.
Collapse
|
5
|
Li L, Luo R, Yang Y, Cheng Y, Ge S, Xu G. Tamibarotene inhibit the accumulation of fibrocyte and alleviate renal fibrosis by IL-17A. Ren Fail 2021; 42:1173-1183. [PMID: 33213229 PMCID: PMC7737677 DOI: 10.1080/0886022x.2020.1847145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022] Open
Abstract
Renal fibrosis is a common pathological process in the progression of chronic kidney disease. Accumulating evidence suggests that interleukin-17A (IL‐17A) and fibrocytes play crucial roles in the pathogenesis of fibrosis. However, the role of IL-17A in the regulation of renal fibrocytes in renal fibrosis has rarely been reported. Here, we report that the plasma IL-17A level is increased in immunoglobulin A nephropathy (IgAN) patients and is correlated with clinical parameters. Using a mouse model of unilateral ureteral obstruction (UUO), we found that both IL-17A expression and fibrocyte infiltration were increased in the kidneys of UUO mice. Besides, IL-17A enhanced fibrosis and fibrocyte-associated chemokine and activator expression in vitro. Furthermore, inhibition of IL-17A using Am80 (Tamibarotene) decreased fibrocytes and fibrocyte-associated chemokine and activator expression and significantly attenuated renal fibrosis in the UUO mice. Our findings suggest that Am80, which inhibits the accumulation of fibrocytes and alleviates renal fibrosis mediated by IL-17A, maybe a novel therapeutic drug for renal fibrosis.
Collapse
Affiliation(s)
- Lixi Li
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Luo
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Yang
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yichun Cheng
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuwang Ge
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Yang N, Chen Z, Zhang X, Shi Y. Novel Targeted Biological Agents for the Treatment of Atopic Dermatitis. BioDrugs 2021; 35:401-415. [PMID: 34213742 DOI: 10.1007/s40259-021-00490-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 06/14/2021] [Indexed: 02/04/2023]
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatologic disease clinically characterized by intense itch, recurrent eczematous lesions, and a chronic or relapsing disease course. Mild-to-moderate AD can be controlled by using moisturizers and topical immunomodulators such as topical corticosteroids and calcineurin inhibitors. If topical therapies fail, phototherapy and systemic immunosuppressant therapies, such as ciclosporin, methotrexate, and azathioprine, can be considered. However, relapse and side effects could still occur. The pathogenesis of AD involves epidermal barrier dysfunction, skin microbiome abnormalities, and cutaneous inflammation. Inflammatory mediators, such as interleukin (IL)-4, IL-13, IL-31, IL-33, IL-17, IL-23, and thymic stromal lymphopoietin, are involved in AD development. Therefore, a series of biological agents targeting these cytokines are promising approaches for treating AD. Dupilumab is the first biological agent approved for the treatment of AD in patients aged 6 years and older in the United States. Tralokinumab, lebrikizumab, and nemolizumab have also been confirmed to have significant efficacy against AD in phase III or IIb clinical trials. Also, fezakinumab was effective in severe AD patients in a phase IIa trial. However, phase II trials of ustekinumab, tezepelumab, etokimab, secukinumab, and omalizumab have failed to meet their primary endpoints. Phase II trials of GBR 830 and KHK 4083 are ongoing. In general, further studies are needed to explore new therapeutic targets and improve the efficacy of biological agents.
Collapse
Affiliation(s)
- Nan Yang
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Zeyu Chen
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200072, China.,Department of Dermatology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Xilin Zhang
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.,Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China. .,Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
7
|
Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. New Insights into Profibrotic Myofibroblast Formation in Systemic Sclerosis: When the Vascular Wall Becomes the Enemy. Life (Basel) 2021; 11:610. [PMID: 34202703 PMCID: PMC8307837 DOI: 10.3390/life11070610] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2021] [Revised: 06/21/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
In systemic sclerosis (SSc), abnormalities in microvessel morphology occur early and evolve into a distinctive vasculopathy that relentlessly advances in parallel with the development of tissue fibrosis orchestrated by myofibroblasts in nearly all affected organs. Our knowledge of the cellular and molecular mechanisms underlying such a unique relationship between SSc-related vasculopathy and fibrosis has profoundly changed over the last few years. Indeed, increasing evidence has suggested that endothelial-to-mesenchymal transition (EndoMT), a process in which profibrotic myofibroblasts originate from endothelial cells, may take center stage in SSc pathogenesis. While in arterioles and small arteries EndoMT may lead to the accumulation of myofibroblasts within the vessel wall and development of fibroproliferative vascular lesions, in capillary vessels it may instead result in vascular destruction and formation of myofibroblasts that migrate into the perivascular space with consequent tissue fibrosis and microvessel rarefaction, which are hallmarks of SSc. Besides endothelial cells, other vascular wall-resident cells, such as pericytes and vascular smooth muscle cells, may acquire a myofibroblast-like synthetic phenotype contributing to both SSc-related vascular dysfunction and fibrosis. A deeper understanding of the mechanisms underlying the differentiation of myofibroblasts inside the vessel wall provides the rationale for novel targeted therapeutic strategies for the treatment of SSc.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy;
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy;
| |
Collapse
|
8
|
Di Benedetto P, Ruscitti P, Berardicurti O, Vomero M, Navarini L, Dolo V, Cipriani P, Giacomelli R. Endothelial-to-mesenchymal transition in systemic sclerosis. Clin Exp Immunol 2021; 205:12-27. [PMID: 33772754 DOI: 10.1111/cei.13599] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/27/2021] [Revised: 02/19/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by significant vascular alterations and multi-organ fibrosis. Microvascular alterations are the first event of SSc and injured endothelial cells (ECs) may transdifferentiate towards myofibroblasts, the cells responsible for fibrosis and collagen deposition. This process is identified as endothelial-to-mesenchymal transition (EndMT), and understanding of its development is pivotal to identify early pathogenetic events and new therapeutic targets for SSc. In this review, we have highlighted the molecular mechanisms of EndMT and summarize the evidence of the role played by EndMT during the development of progressive fibrosis in SSc, also exploring the possible therapeutic role of its inhibition.
Collapse
Affiliation(s)
- P Di Benedetto
- Clinical Pathology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Ruscitti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - O Berardicurti
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - M Vomero
- Unit of Rheumatology and Clinical Immunology, University of Rome 'Campus Biomedico', Rome, Italy
| | - L Navarini
- Unit of Rheumatology and Clinical Immunology, University of Rome 'Campus Biomedico', Rome, Italy
| | - V Dolo
- Clinical Pathology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - P Cipriani
- Division of Rheumatology, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - R Giacomelli
- Unit of Rheumatology and Clinical Immunology, University of Rome 'Campus Biomedico', Rome, Italy
| |
Collapse
|
9
|
Watanabe H, Bi J, Murata R, Fujimura R, Nishida K, Imafuku T, Nakamura Y, Maeda H, Mukunoki A, Takeo T, Nakagata N, Kurauchi Y, Katsuki H, Tanaka M, Matsushita K, Fukagawa M, Maruyama T. A synthetic retinoic acid receptor agonist Am80 ameliorates renal fibrosis via inducing the production of alpha-1-acid glycoprotein. Sci Rep 2020; 10:11424. [PMID: 32651445 PMCID: PMC7351735 DOI: 10.1038/s41598-020-68337-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/20/2019] [Accepted: 06/09/2020] [Indexed: 01/30/2023] Open
Abstract
Renal fibrosis is a major factor in the progression of chronic kidney disease and the final common pathway of kidney injury. Therefore, the effective therapies against renal fibrosis are urgently needed. The objective of this study was to investigate the effect of Am80, a synthetic retinoic acid receptor (RAR) agonist, in the treatment of renal interstitial fibrosis using unilateral ureteral obstruction (UUO) mice. The findings indicate that Am80 treatment suppressed renal fibrosis and inflammation to the same degree as the naturally-occuring retinoic acid, all-trans retinoic acid (atRA). But the adverse effect of body weight loss in Am80-treated mice was lower compared to the atRA treatment. The hepatic mRNA levels of alpha-1-acid glycoprotein (AGP), a downstream molecule of RAR agonist, was increased following administration of Am80 to healthy mice. In addition, increased AGP mRNA expression was also observed in HepG2 cells and THP-1-derived macrophages that had been treated with Am80. AGP-knockout mice exacerbated renal fibrosis, inflammation and macrophage infiltration in UUO mice, indicating endogenous AGP played an anti-fibrotic and anti-inflammatory role during the development of renal fibrosis. We also found that no anti-fibrotic effect of Am80 was observed in UUO-treated AGP-knockout mice whereas atRA treatment tended to show a partial anti-fibrotic effect. These collective findings suggest that Am80 protects against renal fibrosis via being involved in AGP function.
Collapse
Affiliation(s)
- Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| | - Jing Bi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Program for Leading Graduate Schools "HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program", Kumamoto University, Kumamoto, Japan
| | - Ryota Murata
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Rui Fujimura
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Program for Leading Graduate Schools "HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program", Kumamoto University, Kumamoto, Japan
| | - Kento Nishida
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Tadashi Imafuku
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Program for Leading Graduate Schools "HIGO (Health Life Science: Interdisciplinary and Global Oriented) Program", Kumamoto University, Kumamoto, Japan
| | - Yuka Nakamura
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Ayumi Mukunoki
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Motoko Tanaka
- Department of Nephrology, Akebono Clinic, Kumamoto, Japan
| | | | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, Kanagawa, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| |
Collapse
|
10
|
Park GT, Kwon YW, Lee TW, Kwon SG, Ko HC, Kim MB, Kim JH. Formyl Peptide Receptor 2 Activation Ameliorates Dermal Fibrosis and Inflammation in Bleomycin-Induced Scleroderma. Front Immunol 2019; 10:2095. [PMID: 31552041 PMCID: PMC6733889 DOI: 10.3389/fimmu.2019.02095] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/10/2019] [Accepted: 08/20/2019] [Indexed: 02/02/2023] Open
Abstract
Systemic sclerosis is a profibrotic autoimmune disease mediated by the dysregulation of extracellular matrix synthesis. Formyl peptide receptor 2 (Fpr2) is a G protein-coupled receptor that modulates inflammation and host defense by regulating the activation of inflammatory cells, such as macrophages. However, the role of Fpr2 in the development and therapy of scleroderma is still unclear. The present study was conducted to investigate the effects of Fpr2 activation in the treatment of scleroderma fibrosis. We found that intradermal administration of WKYMVm, an Fpr2-specific agonist, alleviated bleomycin-induced scleroderma fibrosis in mice and decreased dermal thickness in scleroderma skin. WKYMVm-treated scleroderma skin tissues displayed reduced numbers of myofibroblasts expressing α-smooth muscle actin, Vimentin, and phosphorylated SMAD3. WKYMVm treatment attenuated macrophage infiltration in scleroderma skin and reduced the number of M2 macrophages. The therapeutic effects of WKYMVm in scleroderma-associated fibrosis and inflammation were completely abrogated in Fpr2 knockout mice. Moreover, WKYMVm treatment reduced the serum levels of inflammatory cytokines, such as tumor necrosis factor-α, and interferon-γ, in the scleroderma model of wild-type mice but not in Fpr2 knockout mice. These results suggest that WKYMVm-induced activation of Fpr2 leads to alleviation of fibrosis by stimulating immune resolution in systemic sclerosis.
Collapse
Affiliation(s)
- Gyu Tae Park
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Yang Woo Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Tae Wook Lee
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Seong Gyu Kwon
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Hyun-Chang Ko
- Department of Dermatology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Moon Bum Kim
- Department of Dermatology, Pusan National University School of Medicine, Yangsan-si, South Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan-si, South Korea.,Research Institute of Convergence Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan-si, South Korea
| |
Collapse
|
11
|
Yamashita T, Asano Y, Saigusa R, Taniguchi T, Nakamura K, Miura S, Toyama T, Takahashi T, Ichimura Y, Hirabayashi M, Yoshizaki A, Miyagaki T, Sugaya M, Sato S. Increased expression of aquaporin-1 in dermal fibroblasts and dermal microvascular endothelial cells possibly contributes to skin fibrosis and edema in patients with systemic sclerosis. J Dermatol Sci 2018; 93:24-32. [PMID: 30270117 DOI: 10.1016/j.jdermsci.2018.09.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/26/2018] [Revised: 08/27/2018] [Accepted: 09/18/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND Aquaporin-1 (AQP1), a water channel protein controlling the water contents of cells and tissues, exerts pleiotropic effects on various biological activities, including inflammation, angiogenesis, and extracellular matrix remodeling, by regulating cell behaviors and tissue water balance. OBJECTIVE To investigate AQP1 roles in systemic sclerosis (SSc) which is characterized by autoimmune inflammation, vasculopathy, and tissue fibrosis. METHODS AQP1 expression was evaluated by immunohistochemistry and quantitative reverse transcription PCR in skin samples from human and animal models and by immunoblotting in cultured cells. Fli1 binding to the AQP1 promoter was evaluated by chromatin immunoprecipitation. Cell migration was assessed by scratch assay. RESULTS Dermal fibroblasts and endothelial cells highly expressed AQP1 in SSc lesional skin, and AQP1 expression in dermal fibroblasts and endothelial cells positively correlated with the degrees of tissue fibrosis and edema, respectively. Consistently, SSc dermal fibroblasts up-regulated AQP1 compared with normal dermal fibroblasts in vitro. Furthermore, TGF-β stimulation induced AQP1 expression in normal dermal fibroblasts, while TGF-β1 antisense oligonucleotide suppressed AQP1 expression in SSc dermal fibroblasts. In endothelial cells, Fli1 deficiency resulted in AQP1 up-regulation in vivo and in vitro and Fli1 bound to the AQP1 promoter. Importantly, SSc dermal fibroblasts and FLI1 siRNA-treated endothelial cells had a pro-migratory property, which was remarkably diminished by gene silencing of AQP1. CONCLUSION AQP1 is up-regulated in SSc dermal fibroblasts and SSc endothelial cells at least partially due to autocrine TGF-β stimulation and Fli1 deficiency, respectively, possibly contributing to inflammation, vasculopathy, and tissue fibrosis by regulating tissue edema and cell migration.
Collapse
Affiliation(s)
- Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan.
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Shunsuke Miura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Megumi Hirabayashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Tomomitsu Miyagaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Makoto Sugaya
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo 113-8655, Japan
| |
Collapse
|
12
|
Thuan DTB, Zayed H, Eid AH, Abou-Saleh H, Nasrallah GK, Mangoni AA, Pintus G. A Potential Link Between Oxidative Stress and Endothelial-to-Mesenchymal Transition in Systemic Sclerosis. Front Immunol 2018; 9:1985. [PMID: 30283435 PMCID: PMC6156139 DOI: 10.3389/fimmu.2018.01985] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/13/2018] [Accepted: 08/13/2018] [Indexed: 12/18/2022] Open
Abstract
Systemic sclerosis (SSc), an autoimmune disease that is associated with a number of genetic and environmental risk factors, is characterized by progressive fibrosis and microvasculature damage in the skin, lungs, heart, digestive system, kidneys, muscles, joints, and nervous system. These abnormalities are associated with altered secretion of growth factor and profibrotic cytokines, such as transforming growth factor-beta (TGF-β), interleukin-4 (IL-4), platelet-derived growth factor (PDGF), and connective-tissue growth factor (CTGF). Among the cellular responses to this proinflammatory environment, the endothelial cells phenotypic conversion into activated myofibroblasts, a process known as endothelial to mesenchymal transition (EndMT), has been postulated. Reactive oxygen species (ROS) might play a key role in SSs-associated fibrosis and vascular damage by mediating and/or activating TGF-β-induced EndMT, a phenomenon that has been observed in other disease models. In this review, we identified and critically appraised published studies investigating associations ROS and EndMT and the presence of EndMT in SSc, highlighting a potential link between oxidative stress and EndMT in this condition.
Collapse
Affiliation(s)
- Duong Thi Bich Thuan
- Department of Biochemistry, Hue University of Medicine and Pharmacy, University of Hue, Hue, Vietnam
| | - Hatem Zayed
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar.,Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Haissam Abou-Saleh
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Gheyath K Nasrallah
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Arduino A Mangoni
- Department of Clinical Pharmacology, College of Medicine and Public Health, Flinders Medical Centre, Flinders University, Adelaide, SA, Australia
| | - Gianfranco Pintus
- Department of Biomedical Sciences, College of Health Sciences, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Yamashita T, Lakota K, Taniguchi T, Yoshizaki A, Sato S, Hong W, Zhou X, Sodin-Semrl S, Fang F, Asano Y, Varga J. An orally-active adiponectin receptor agonist mitigates cutaneous fibrosis, inflammation and microvascular pathology in a murine model of systemic sclerosis. Sci Rep 2018; 8:11843. [PMID: 30087356 PMCID: PMC6081386 DOI: 10.1038/s41598-018-29901-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/17/2017] [Accepted: 06/04/2018] [Indexed: 12/22/2022] Open
Abstract
The hallmarks of systemic sclerosis (SSc) are autoimmunity, microangiopathy and fibrosis. Skin fibrosis is accompanied by attrition of the dermal white adipose tissue layer, and alterations in the levels and function of adiponectin. Since these findings potentially implicate adiponectin in the pathogenesis of SSc, we employed a novel pharmacological approach to augment adiponectin signaling using AdipoRon, an orally active adiponectin receptor agonist. Chronic treatment with AdipoRon significantly ameliorated bleomycin-induced dermal fibrosis in mice. AdipoRon attenuated fibroblast activation, adipocyte-to-myofibroblast transdifferentiation, Th2/Th17-skewed polarization of the immune response, vascular injury and endothelial-to-mesenchymal transition within the lesional skin. In vitro, AdipoRon abrogated profibrotic responses elicited by TGF-β in normal fibroblasts, and reversed the inherently-activated profibrotic phenotype of SSc fibroblasts. In view of these broadly beneficial effects on all three cardinal pathomechanisms underlying the clinical manifestations of SSc, pharmacological augmentation of adiponectin signaling might represent a novel strategy for the treatment of SSc.
Collapse
Affiliation(s)
- Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia.,Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, USA
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Wen Hong
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, USA
| | - Xingchun Zhou
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, USA
| | - Snezn Sodin-Semrl
- Department of Rheumatology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Feng Fang
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, USA
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
14
|
Hong L, Du X, Li W, Mao Y, Sun L, Li X. EndMT: A promising and controversial field. Eur J Cell Biol 2018; 97:493-500. [PMID: 30082099 DOI: 10.1016/j.ejcb.2018.07.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/09/2018] [Revised: 07/03/2018] [Accepted: 07/27/2018] [Indexed: 12/17/2022] Open
Abstract
The endothelial to mesenchymal transition (EndMT) is the process by which endothelial cells lose a portion of their cellular features and obtain certain characteristics of mesenchymal cells, including loss of tight junctions, increased motility, and increased secretion of extracellular matrix proteins. EndMT is involved in cardiac development and a variety of diseases processes, such as vascular or tissue fibrosis and tumor. However, its role in specific diseases remains under debate. This review summarizes EndMT-related diseases, existing controversies, different types of EndMT, and molecules and signaling pathways associated with the process.
Collapse
Affiliation(s)
- Lei Hong
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| | - Xiaolong Du
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| | - Wendong Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Youjun Mao
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Lili Sun
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Soochow University, Sanxiang Road, Suzhou 215000, JiangSu, China.
| |
Collapse
|
15
|
Shrishrimal S, Kosmacek EA, Chatterjee A, Tyson MJ, Oberley-Deegan RE. The SOD Mimic, MnTE-2-PyP, Protects from Chronic Fibrosis and Inflammation in Irradiated Normal Pelvic Tissues. Antioxidants (Basel) 2017; 6:antiox6040087. [PMID: 29113120 PMCID: PMC5745497 DOI: 10.3390/antiox6040087] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/29/2017] [Revised: 10/25/2017] [Accepted: 10/31/2017] [Indexed: 02/07/2023] Open
Abstract
Pelvic radiation for cancer therapy can damage a variety of normal tissues. In this study, we demonstrate that radiation causes acute changes to pelvic fibroblasts such as the transformation to myofibroblasts and the induction of senescence, which persist months after radiation. The addition of the manganese porphyrin, MnTE-2-PyP, resulted in protection of these acute changes in fibroblasts and this protection persisted months following radiation exposure. Specifically, at two months post-radiation, MnTE-2-PyP inhibited the number of α-smooth muscle actin positive fibroblasts induced by radiation and at six months post-radiation, MnTE-2-PyP significantly reduced collagen deposition (fibrosis) in the skin and bladder tissues of irradiated mice. Radiation also resulted in changes to T cells. At two months post-radiation, there was a reduction of Th1-producing splenocytes, which resulted in reduced Th1:Th2 ratios. MnTE-2-PyP maintained Th1:Th2 ratios similar to unirradiated mice. At six months post-radiation, increased T cells were observed in the adipose tissues. MnTE-2-PyP treatment inhibited this increase. Thus, MnTE-2-PyP treatment maintains normal fibroblast function and T cell immunity months after radiation exposure. We believe that one of the reasons MnTE-2-PyP is a potent radioprotector is due to its protection of multiple cell types from radiation damage.
Collapse
Affiliation(s)
- Shashank Shrishrimal
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Elizabeth A Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Arpita Chatterjee
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - McDonald J Tyson
- Department of Physics & Cancer Research Center, Hampton University, Hampton, VA 23668, USA.
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
16
|
Schmidt KG, Herrero San Juan M, Trautmann S, Berninger L, Schwiebs A, Ottenlinger FM, Thomas D, Zaucke F, Pfeilschifter JM, Radeke HH. Sphingosine-1-Phosphate Receptor 5 Modulates Early-Stage Processes during Fibrogenesis in a Mouse Model of Systemic Sclerosis: A Pilot Study. Front Immunol 2017; 8:1242. [PMID: 29033951 PMCID: PMC5626866 DOI: 10.3389/fimmu.2017.01242] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2017] [Accepted: 09/19/2017] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a rare multi-organ autoimmune disease characterized by progressive skin fibrosis. Inflammation, type 2 immunity, and fibrogenic processes are involved in disease development and may be affected by sphingolipids. However, details about early-stage pathophysiological mechanisms and implicated mediators remain elusive. The sphingolipid sphingosine-1-phosphate (S1P) is elevated in the sera of SSc patients, and its receptor S1P5 is expressed in skin tissue. Nevertheless, almost nothing is known about the dermatological contribution of S1P5 to inflammatory and pro-fibrotic processes leading to the pathological changes seen in SSc. In this study, we observed a novel effect of S1P5 on the inflammatory processes during low-dose bleomycin (BLM)-induced fibrogenesis in murine skin. By comparing 2-week-treated skin areas of wild-type (WT) and S1P5-deficient mice, we found that S1P5 is important for the transcriptional upregulation of the Th2 characteristic transcription factor GATA-3 under treatment-induced inflammatory conditions, while T-bet (Th1) and FoxP3 (Treg) mRNA expression was regulated independently of S1P5. Additionally, treatment caused a regulation of S1P receptor 1 and S1P receptor 3 mRNA as well as a regulation of long-chain ceramide profiles, which both differ significantly between the genotypes. Despite S1P5-dependent differences regarding inflammatory processes, similar macroscopic evidence of fibrosis was detected in the skin histology of WT and S1P5-deficient mice after 4 weeks of subcutaneous BLM treatment. However, at the earlier 2-week point in time, the mRNA data of pro-collagen type 1 and SMAD7 indicate a pro-fibrotic S1P5 contribution in the applied SSc mouse model. In conclusion, we propose that S1P5 plays a role as a novel modulator during the early phase of BLM-caused fibrogenesis in murine skin. An immediate relationship between dermal S1P5 expression and fibrotic processes leading to skin alterations, such as formative for SSc pathogenesis, is indicated but should be studied more profound in further investigations. Therefore, this study is an initial step in understanding the role of S1P5-mediated effects during early stages of fibrogenesis, which may encourage the ongoing search for new therapeutic options for SSc patients.
Collapse
Affiliation(s)
- Katrin G Schmidt
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| | - Martina Herrero San Juan
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| | - Sandra Trautmann
- pharmazentrum frankfurt/ZAFES, Institute for Clinical Pharmacology, Hospital of the Goethe University, Frankfurt, Germany
| | - Lucija Berninger
- Dr Rolf M Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital, Friedrichsheim gGmbH, Frankfurt, Germany
| | - Anja Schwiebs
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| | - Florian M Ottenlinger
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| | - Dominique Thomas
- pharmazentrum frankfurt/ZAFES, Institute for Clinical Pharmacology, Hospital of the Goethe University, Frankfurt, Germany
| | - Frank Zaucke
- Dr Rolf M Schwiete Research Unit for Osteoarthritis, Orthopedic University Hospital, Friedrichsheim gGmbH, Frankfurt, Germany
| | - Josef M Pfeilschifter
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| | - Heinfried H Radeke
- pharmazentrum frankfurt/ZAFES, Institute of Pharmacology and Toxicology, Hospital of the Goethe University, Frankfurt, Germany
| |
Collapse
|
17
|
Early inflammatory players in cutaneous fibrosis. J Dermatol Sci 2017; 87:228-235. [DOI: 10.1016/j.jdermsci.2017.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/06/2017] [Revised: 06/09/2017] [Accepted: 06/13/2017] [Indexed: 01/12/2023]
|
18
|
Toyama T, Asano Y, Miyagawa T, Nakamura K, Hirabayashi M, Yamashita T, Saigusa R, Miura S, Ichimura Y, Takahashi T, Taniguchi T, Yoshizaki A, Sato S. The impact of transcription factor Fli1 deficiency on the regulation of angiogenesis. Exp Dermatol 2017; 26:912-918. [PMID: 28370536 DOI: 10.1111/exd.13341] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 03/22/2017] [Indexed: 12/18/2022]
Abstract
The insufficiency of Friend leukaemia virus integration 1 (Fli1), a member of the Ets family transcription factors, is implicated in the pathogenesis of vasculopathy associated with systemic sclerosis (SSc). Fli1 deficiency accelerates early steps of angiogenesis, including detachment of pre-existing pericytes and extracellular matrix degradation by endothelial proteinases, but the impact of Fli1 deficiency on the other steps of angiogenesis has not been investigated. Therefore, we evaluated the effect of Fli1 deficiency on migration, proliferation, cell survival and tube formation of human dermal microvascular endothelial cells (HDMECs). HDMECs transfected with FLI1 siRNA exhibited a greater migratory property in scratch assay and transwell migration assay and a higher proliferation rate in BrdU assay than HDMECs transfected with non-silencing scrambled RNA. In flow cytometry-based apoptosis assay, FLI1 siRNA-transduced HDMECs revealed the decreased number of annexin and propidium iodide-double-positive apoptotic cells compared with control cells, reflecting the promotion of cell survival. On the other hand, tubulogenic activity on Matrigel was remarkably suppressed in Fli1-deficient HDMECs relative to control cells. These results indicate that Fli1 deficiency promotes migration, proliferation and cell survival, while abating tube formation of endothelial cells, suggesting that Fli1 deficiency is potentially attributable to the development of both proliferative obliterative vasculopathy (occlusion of arterioles and small arteries) and destructive vasculopathy (loss of small vessels) characteristic of SSc vasculopathy.
Collapse
Affiliation(s)
- Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takuya Miyagawa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Megumi Hirabayashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shunsuke Miura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Glycyrrhizin Ameliorates Fibrosis, Vasculopathy, and Inflammation in Animal Models of Systemic Sclerosis. J Invest Dermatol 2016; 137:631-640. [PMID: 27777101 DOI: 10.1016/j.jid.2016.08.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/27/2016] [Revised: 08/06/2016] [Accepted: 08/17/2016] [Indexed: 02/08/2023]
Abstract
Systemic sclerosis (SSc) is a multisystem inflammatory and vascular disease resulting in extensive tissue fibrosis. Glycyrrhizin, clinically used for chronic hepatic diseases and itching dermatitis, modulates the pathological processes of inflammation, vasculopathy, and fibrosis in human diseases and their animal models. Therefore, we investigated a potential impact of glycyrrhizin on the key pathological manifestations of SSc, including inflammation, vasculopathy, and tissue fibrosis, with bleomycin-treated mice mimicking the fibrotic and inflammatory components of SSc and endothelial cell-specific Fli1-knockout mice recapitulating SSc vasculopathy. Glycyrrhizin significantly ameliorated dermal fibrosis in bleomycin-treated mice, which was partly attributable to blockade of transforming growth factor-β signaling in dermal fibroblasts through the down-regulation of thrombospondin 1, a latent transforming growth factor-β receptor, and transcription factors Smad3 and Ets1. Furthermore, bleomycin-dependent induction of T helper type 2-skewed immune polarization, M2 macrophage infiltration, and endothelial-to-mesenchymal transition were greatly suppressed in mice administered glycyrrhizin. Glycyrrhizin also improved vascular permeability of endothelial cell-specific Fli1-knockout mice by increasing the expression of molecules regulating vascular integrity. These results indicate that glycyrrhizin ameliorates bleomycin-induced dermal fibrosis through the inhibition of fibroblast activation, T helper type 2-skewed immune polarization, M2 macrophage infiltration, and endothelial-to-mesenchymal transition and improves endothelial Fli1 deficiency-dependent vascular disintegrity, implying its potential as a disease-modifying drug for SSc.
Collapse
|