1
|
Lee B, Roh JS, Jeong H, Kim Y, Lee J, Yun C, Park J, Kim DS, Lee J, So MW, Kim A, Sohn DH, Lee SG. Ginkgo biloba extract ameliorates skin fibrosis in a bleomycin-induced mouse model of systemic sclerosis. Anim Cells Syst (Seoul) 2024; 28:152-160. [PMID: 38645438 PMCID: PMC11028018 DOI: 10.1080/19768354.2024.2337761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 03/24/2024] [Indexed: 04/23/2024] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by skin and internal organ fibrosis and obliterative vasculopathy. Few effective treatments are currently available for fibrosis in SSc, therefore, demand persists for novel therapies. Although use of Ginkgo biloba extract (GBE) has been reported to improve blood circulation and alleviate liver and lung fibrosis, its effect on skin fibrosis in SSc remains unclear. In this study, the effects and underlying mechanisms of GBE on skin fibrosis in bleomycin (BLM)-induced mouse model of SSc was investigated. GBE significantly reduced dermal thickness and protein levels of profibrotic factors in the BLM-induced SSc mouse model. Moreover, GBE inhibited the gene expression of profibrotic factors, such as COL1A1, α-SMA, and connective tissue growth factor (CTGF), in fibroblasts by suppressing transforming growth factor (TGF)-β signaling. Furthermore, GBE inhibited the transdifferentiation of adipocytes into myofibroblasts. Thus, our findings suggest that GBE is a promising therapeutic candidate for the treatment of SSc.
Collapse
Affiliation(s)
- Beomgu Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jong Seong Roh
- Department of Herbal Prescription, College of Korean Medicine, Daegu Haany University, Gyeongsan, Republic of Korea
| | - Hoim Jeong
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Yerin Kim
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jihyeon Lee
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Changun Yun
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
| | - Jiyoung Park
- Department of Biological Sciences, College of Information and Biotechnology, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Da-sol Kim
- Department of Dermatology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jungsoo Lee
- Department of Dermatology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Min Wook So
- Division of Rheumatology, Department of Internal Medicine, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Aran Kim
- Division of Rheumatology, Department of Internal Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| | - Dong Hyun Sohn
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Republic of Korea
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Seung-Geun Lee
- Division of Rheumatology, Department of Internal Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Republic of Korea
- Biomedical Research Institute, Pusan National University Hospital, Busan, Republic of Korea
| |
Collapse
|
2
|
Jussila A, Zhang B, Kirti S, Atit R. Tissue fibrosis associated depletion of lipid-filled cells. Exp Dermatol 2024; 33:e15054. [PMID: 38519432 PMCID: PMC10977660 DOI: 10.1111/exd.15054] [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: 10/03/2023] [Revised: 02/06/2024] [Accepted: 02/29/2024] [Indexed: 03/24/2024]
Abstract
Fibrosis is primarily described as the deposition of excessive extracellular matrix, but in many tissues it also involves a loss of lipid or lipid-filled cells. Lipid-filled cells are critical to tissue function and integrity in many tissues including the skin and lungs. Thus, loss or depletion of lipid-filled cells during fibrogenesis, has implications for tissue function. In some contexts, lipid-filled cells can impact ECM composition and stability, highlighting their importance in fibrotic transformation. Recent papers in fibrosis address this newly recognized fibrotic lipodystrophy phenomenon. Even in disparate tissues, common mechanisms are emerging to explain fibrotic lipodystrophy. These findings have implications for fibrosis in tissues composed of fibroblast and lipid-filled cell populations such as skin, lung, and liver. In this review, we will discuss the roles of lipid-containing cells, their reduction/loss during fibrotic transformation, and the mechanisms of that loss in the skin and lungs.
Collapse
Affiliation(s)
- Anna Jussila
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Brian Zhang
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Sakin Kirti
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Radhika Atit
- Department of Biology, College of Arts and Sciences, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
3
|
Jimenez SA, Piera-Velazquez S. Cellular Transdifferentiation: A Crucial Mechanism of Fibrosis in Systemic Sclerosis. Curr Rheumatol Rev 2024; 20:388-404. [PMID: 37921216 DOI: 10.2174/0115733971261932231025045400] [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: 05/12/2023] [Revised: 07/13/2023] [Accepted: 07/27/2023] [Indexed: 11/04/2023]
Abstract
Systemic Sclerosis (SSc) is a systemic autoimmune disease of unknown etiology with a highly complex pathogenesis that despite extensive investigation is not completely understood. The clinical and pathologic manifestations of the disease result from three distinct processes: 1) Severe and frequently progressive tissue fibrosis causing exaggerated and deleterious accumulation of interstitial collagens and other extracellular matrix molecules in the skin and various internal organs; 2) extensive fibroproliferative vascular lesions affecting small arteries and arterioles causing tissue ischemic alterations; and 3) cellular and humoral immunity abnormalities with the production of numerous autoantibodies, some with very high specificity for SSc. The fibrotic process in SSc is one of the main causes of disability and high mortality of the disease. Owing to its essentially universal presence and the severity of its clinical effects, the mechanisms involved in the development and progression of tissue fibrosis have been extensively investigated, however, despite intensive investigation, the precise molecular mechanisms have not been fully elucidated. Several recent studies have suggested that cellular transdifferentiation resulting in the phenotypic conversion of various cell types into activated myofibroblasts may be one important mechanism. Here, we review the potential role that cellular transdifferentiation may play in the development of severe and often progressive tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Sergio A Jimenez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| | - Sonsoles Piera-Velazquez
- Department of Dermatology and Cutaneous Biology, Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA
| |
Collapse
|
4
|
Sisto M, Lisi S. Immune and Non-Immune Inflammatory Cells Involved in Autoimmune Fibrosis: New Discoveries. J Clin Med 2023; 12:jcm12113801. [PMID: 37297996 DOI: 10.3390/jcm12113801] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/27/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Fibrosis is an important health problem and its pathogenetic activation is still largely unknown. It can develop either spontaneously or, more frequently, as a consequence of various underlying diseases, such as chronic inflammatory autoimmune diseases. Fibrotic tissue is always characterized by mononuclear immune cells infiltration. The cytokine profile of these cells shows clear proinflammatory and profibrotic characteristics. Furthermore, the production of inflammatory mediators by non-immune cells, in response to several stimuli, can be involved in the fibrotic process. It is now established that defects in the abilities of non-immune cells to mediate immune regulation may be involved in the pathogenicity of a series of inflammatory diseases. The convergence of several, not yet well identified, factors results in the aberrant activation of non-immune cells, such as epithelial cells, endothelial cells, and fibroblasts, that, by producing pro-inflammatory molecules, exacerbate the inflammatory condition leading to the excessive and chaotic secretion of extracellular matrix proteins. However, the precise cellular mechanisms involved in this process have not yet been fully elucidated. In this review, we explore the latest discoveries on the mechanisms that initiate and perpetuate the vicious circle of abnormal communications between immune and non-immune cells, responsible for fibrotic evolution of inflammatory autoimmune diseases.
Collapse
Affiliation(s)
- Margherita Sisto
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Sabrina Lisi
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Section of Human Anatomy and Histology, University of Bari "Aldo Moro", 70124 Bari, Italy
| |
Collapse
|
5
|
Lee KI, Manuntag LJ, Kifayat A, Manuntag SE, Sperber K, Ash JY, Frishman WH, Wasserman A. Cardiovascular Manifestations of Systemic Sclerosis: An Overview of Pathophysiology, Screening Modalities, and Treatment Options. Cardiol Rev 2023; 31:22-27. [PMID: 34619709 DOI: 10.1097/crd.0000000000000420] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Systemic sclerosis, previously known as scleroderma, is a heterogeneous, systemic disease that is defined by its 3 pathological hallmarks: the production of autoantibodies, small vessel vasculopathy, and fibroblast dysfunction, leading to an increased deposition of extracellular matrix. We conducted a review of the available literature that covers the cardiovascular manifestations of SSc: electrical conduction abnormalities, pulmonary hypertension, pericardial disease, and atherosclerosis. Within each major category, we will discuss the definition, diagnostics, and available treatment options. Increased mortality from cardiovascular complications necessitates early screening and management. Annual screening with noninvasive modalities is encouraged. The current management of each complication generally follows the management algorithms of patients regardless of SSc status and is dependent on the severity of the patient's clinical presentation.
Collapse
Affiliation(s)
- Kyu-In Lee
- From the Department of Medicine, Westchester Medical Center, Valhalla, NY
| | - Levy Jo Manuntag
- From the Department of Medicine, Westchester Medical Center, Valhalla, NY
| | - Alina Kifayat
- Department of Rheumatology, Westchester Medical Center/New York Medical College, Valhalla, NY
| | | | - Kirk Sperber
- Department of Rheumatology, Westchester Medical Center/New York Medical College, Valhalla, NY
| | - Julia Yegudin Ash
- Department of Rheumatology, Westchester Medical Center/New York Medical College, Valhalla, NY
| | - William H Frishman
- Department of Allergy/Immunology, Westchester Medical Center/New York Medical College, Valhalla, NY
| | - Amy Wasserman
- Department of Rheumatology, Westchester Medical Center/New York Medical College, Valhalla, NY
| |
Collapse
|
6
|
Corano Scheri K, Liang X, Dalal V, Le Poole IC, Varga J, Hayashida T. SARA suppresses myofibroblast precursor transdifferentiation in fibrogenesis in a mouse model of scleroderma. JCI Insight 2022; 7:160977. [PMID: 36136606 PMCID: PMC9675568 DOI: 10.1172/jci.insight.160977] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 09/12/2022] [Indexed: 12/15/2022] Open
Abstract
We previously reported that Smad anchor for receptor activation (SARA) plays a critical role in maintaining epithelial cell phenotype. Here, we show that SARA suppressed myofibroblast precursor transdifferentiation in a mouse model of scleroderma. Mice overexpressing SARA specifically in PDGFR-β+ pericytes and pan-leukocytes (SARATg) developed significantly less skin fibrosis in response to bleomycin injection compared with wild-type littermates (SARAWT). Single-cell RNA-Seq analysis of skin PDGFR-β+ cells implicated pericyte subsets assuming myofibroblast characteristics under fibrotic stimuli, and SARA overexpression blocked the transition. In addition, a cluster that expresses molecules associated with Th2 cells and macrophage activation was enriched in SARAWT mice, but not in SARATg mice, after bleomycin treatment. Th2-specific Il-31 expression was increased in skin of the bleomycin-treated SARAWT mice and patients with scleroderma (or systemic sclerosis, SSc). Receptor-ligand analyses indicated that lymphocytes mediated pericyte transdifferentiation in SARAWT mice, while with SARA overexpression the myofibroblast activity of pericytes was suppressed. Together, these data suggest a potentially novel crosstalk between myofibroblast precursors and immune cells in the pathogenesis of SSc, in which SARA plays a critical role.
Collapse
Affiliation(s)
- Katia Corano Scheri
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Xiaoyan Liang
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Vidhi Dalal
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - I. Caroline Le Poole
- Departments of Dermatology and Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - John Varga
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Tomoko Hayashida
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Pediatric Nephrology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Utsunomiya A, Chino T, Kasamatsu H, Hasegawa T, Utsunomiya N, Luong VH, Matsushita T, Sasaki Y, Ogura D, Niwa SI, Oyama N, Hasegawa M. The compound LG283 inhibits bleomycin-induced skin fibrosis via antagonizing TGF-β signaling. Arthritis Res Ther 2022; 24:94. [PMID: 35488265 PMCID: PMC9052694 DOI: 10.1186/s13075-022-02773-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 04/11/2022] [Indexed: 11/15/2022] Open
Abstract
Background Systemic sclerosis (SSc) is a collagen disease that exhibits intractable fibrosis and vascular injury of the skin and internal organs. Transforming growth factor-β (TGF-β)/Smad signaling plays a central role in extracellular matrix (ECM) production by α-SMA-positive myofibroblasts. Myofibroblasts may be partially derived from various precursor cells in addition to resident fibroblasts. Recently, our high-throughput in vitro screening discovered a small compound, LG283, that may disrupt the differentiation of epithelial cells into myofibroblasts. This compound was originally generated as a curcumin derivative. Methods In this study, we investigated the effect of LG283 on inhibiting fibrosis and its mechanism. The action of LG283 on TGF-β-dependent fibrogenic activity and epithelial-mesenchymal transition (EMT) was analyzed in vitro. The effects of LG283 were also examined in a bleomycin-induced skin fibrosis mouse model. Results LG283 suppressed TGF-β-induced expression of ECM, α-SMA, and transcription factors Snail 1 and 2, and Smad3 phosphorylation in cultured human dermal fibroblasts. LG283 was also found to block EMT induction in cultured human epithelial cells. During these processes, Smad3 phosphorylation and/or expression of Snail 1 and 2 were inhibited by LG283 treatment. In the bleomycin-induced skin fibrosis model, oral administration of LG283 efficiently protected against the development of fibrosis and decrease of capillary vessels without significantly affecting cell infiltration or cytokine concentrations in the skin. No apparent adverse effects of LG283 were found. LG283 treatment remarkably inhibited the enhanced expression of α-SMA and phosphorylated Smad3, as well as those of Snail 1 and 2, in the bleomycin-injected skin. Conclusions The LG283 compound exhibits antagonistic activity on fibrosis and vascular injury through inhibition of TGF-β/Smad/Snail mesenchymal transition pathways and thus, may be a candidate therapeutic for the treatment of SSc. Although the involvement of EMT in the pathogenesis of SSc remains unclear, the screening of EMT regulatory compounds may be an attractive approach for SSc therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02773-2.
Collapse
Affiliation(s)
- Akira Utsunomiya
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Takenao Chino
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Hiroshi Kasamatsu
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Takumi Hasegawa
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Natsuko Utsunomiya
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Vu Huy Luong
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.,Department of Dermatology, Hanoi Medical University, Hanoi, Vietnam
| | - Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | - Dai Ogura
- Link Genomics, Inc., Chuo, Tokyo, Japan
| | | | - Noritaka Oyama
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan
| | - Minoru Hasegawa
- Department of Dermatology, Division of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui, 910-1193, Japan.
| |
Collapse
|
8
|
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]
|
9
|
Dai B, Ding L, Zhao L, Zhu H, Luo H. Contributions of Immune Cells and Stromal Cells to the Pathogenesis of Systemic Sclerosis: Recent Insights. Front Pharmacol 2022; 13:826839. [PMID: 35185577 PMCID: PMC8852243 DOI: 10.3389/fphar.2022.826839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/04/2022] [Indexed: 12/21/2022] Open
Abstract
Systemic sclerosis (SSc) is a multisystem rheumatic disease characterized by vascular dysfunction, autoimmune abnormalities, and progressive organ fibrosis. A series of studies in SSc patients and fibrotic models suggest that immune cells, fibroblasts, and endothelial cells participate in inflammation and aberrant tissue repair. Furthermore, the growing number of studies on single-cell RNA sequencing (scRNA-seq) technology in SSc elaborate on the transcriptomics and heterogeneities of these cell subsets significantly. In this review, we summarize the current knowledge regarding immune cells and stromal cells in SSc patients and discuss their potential roles in SSc pathogenesis, focusing on recent advances in the new subtypes by scRNA-seq.
Collapse
Affiliation(s)
- Bingying Dai
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Liqing Ding
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Lijuan Zhao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Honglin Zhu, ; Hui Luo,
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Honglin Zhu, ; Hui Luo,
| |
Collapse
|
10
|
The role for miRNA146b-5p in the attenuation of dermal fibrosis and angiogenesis by targeting PDGFRα in skin wounds. J Invest Dermatol 2021; 142:1990-2002.e4. [DOI: 10.1016/j.jid.2021.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/03/2021] [Accepted: 11/26/2021] [Indexed: 11/22/2022]
|
11
|
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: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
12
|
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: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
13
|
Romano E, Rosa I, Fioretto BS, Cerinic MM, Manetti M. The Role of Pro-fibrotic Myofibroblasts in Systemic Sclerosis: from Origin to Therapeutic Targeting. Curr Mol Med 2021; 22:209-239. [PMID: 33823766 DOI: 10.2174/0929867328666210325102749] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/02/2021] [Accepted: 03/09/2021] [Indexed: 11/22/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex connective tissue disorder characterized by multisystem clinical manifestations resulting from immune dysregulation/autoimmunity, vasculopathy and, most notably, progressive fibrosis of the skin and internal organs. In recent years, it has emerged that the main drivers of SSc-related tissue fibrosis are myofibroblasts, a type of mesenchymal cells with both the extracellular matrix-synthesizing features of fibroblasts and the cytoskeletal characteristics of contractile smooth muscle cells. The accumulation and persistent activation of pro-fibrotic myofibroblasts during SSc development and progression result into elevated mechanical stress and reduced matrix plasticity within the affected tissues and may be ascribed to a reduced susceptibility of these cells to pro-apoptotic stimuli, as well as their increased formation from tissue-resident fibroblasts or transition from different cell types. Given the crucial role of myofibroblasts in SSc pathogenesis, finding the way to inhibit myofibroblast differentiation and accumulation by targeting their formation, function and survival may represent an effective approach to hamper the fibrotic process or even halt or reverse established fibrosis. In this review, we discuss the role of myofibroblasts in SSc-related fibrosis, with a special focus on their cellular origin and the signaling pathways implicated in their formation and persistent activation. Furthermore, we provide an overview of potential therapeutic strategies targeting myofibroblasts that may be able to counteract fibrosis in this pathological condition.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Marco Matucci Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence. Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence. Italy
| |
Collapse
|
14
|
Rosa I, Romano E, Fioretto BS, Matucci-Cerinic M, Manetti M. Adipose-derived stem cells: Pathophysiologic implications vs therapeutic potential in systemic sclerosis. World J Stem Cells 2021; 13:30-48. [PMID: 33584978 PMCID: PMC7859990 DOI: 10.4252/wjsc.v13.i1.30] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) residing in the stromal vascular fraction (SVF) of white adipose tissue are recently emerging as an alternative tool for stem cell-based therapy in systemic sclerosis (SSc), a complex connective tissue disorder affecting the skin and internal organs with fibrotic and vascular lesions. Several preclinical and clinical studies have reported promising therapeutic effects of fat grafting and autologous SVF/ADSC-based local treatment for facial and hand cutaneous manifestations of SSc patients. However, currently available data indicate that ADSCs may represent a double-edged sword in SSc, as they may exhibit a pro-fibrotic and anti-adipogenic phenotype, possibly behaving as an additional pathogenic source of pro-fibrotic myofibroblasts through the adipocyte-to-myofibroblast transition process. Thus, in the perspective of a larger employ of SSc-ADSCs for further therapeutic applications, it is important to definitely unravel whether these cells present a comparable phenotype and similar immunosuppressive, anti-inflammatory, anti-fibrotic and pro-angiogenic properties in respect to healthy ADSCs. In light of the dual role that ADSCs seem to play in SSc, this review will provide a summary of the most recent insights into the preclinical and clinical studies employing SVF and ADSCs for the treatment of the disease and, at the same time, will focus on the main findings highlighting the possible involvement of these stem cells in SSc-related fibrosis pathogenesis.
Collapse
Affiliation(s)
- Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence 50134, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Florence 50134, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence 50134, Italy.
| |
Collapse
|
15
|
Carvalheiro T, Malvar Fernández B, Ottria A, Giovannone B, Marut W, Reedquist KA, Garcia S, Radstake TR. Extracellular SPARC cooperates with TGF-β signalling to induce pro-fibrotic activation of systemic sclerosis patient dermal fibroblasts. Rheumatology (Oxford) 2021; 59:2258-2263. [PMID: 31840182 PMCID: PMC7449812 DOI: 10.1093/rheumatology/kez583] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 10/29/2019] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES SSc is an autoimmune disease characterized by inflammation, vascular injury and excessive fibrosis in multiple organs. Secreted protein acidic and rich in cysteine (SPARC) is a matricellular glycoprotein that regulates processes involved in SSc pathology, such as inflammation and fibrosis. In vivo and in vitro studies have implicated SPARC in SSc, but it is unclear if the pro-fibrotic effects of SPARC on fibroblasts are a result of intracellular signalling or fibroblast interactions with extracellular SPARC hampering further development of SPARC as a potential therapeutic target. This study aimed to analyse the potential role of exogenous SPARC as a regulator of fibrosis in SSc. METHODS Dermal fibroblasts from both healthy controls and SSc patients were stimulated with SPARC alone or in combination with TGF-β1, in the absence or presence of a TGF receptor 1 inhibitor. mRNA and protein expression of extracellular matrix components and other fibrosis-related mediators were measured by quantitative PCR and western blot. RESULTS Exogenous SPARC induced mRNA and protein expression of collagen I, collagen IV, fibronectin 1, TGF-β and SPARC by dermal fibroblasts from SSc patients, but not from healthy controls. Importantly, exogenous SPARC induced the activation of the tyrosine kinase SMAD2 and pro-fibrotic gene expression induced by SPARC in SSc fibroblasts was abrogated by inhibition of TGF-β signalling. CONCLUSION These results indicate that exogenous SPARC is an important pro-fibrotic mediator contributing to the pathology driving SSc but in a TGF-β dependent manner. Therefore, SPARC could be a promising therapeutic target for reducing fibrosis in SSc patients, even in late states of the disease.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| | | | - Andrea Ottria
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| | - Barbara Giovannone
- Department of Dermatology, University Medical Centre Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Wioleta Marut
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| | - Kris A Reedquist
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| | - Samuel Garcia
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| | - Timothy R Radstake
- Department of Rheumatology and Clinical Immunology.,Laboratory of Translational Immunology
| |
Collapse
|
16
|
Rosa I, Romano E, Fioretto BS, Manetti M. The contribution of mesenchymal transitions to the pathogenesis of systemic sclerosis. Eur J Rheumatol 2020; 7:S157-S164. [PMID: 31922472 PMCID: PMC7647682 DOI: 10.5152/eurjrheum.2019.19081] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022] Open
Abstract
Systemic sclerosis (SSc) is a multifaceted connective tissue disease characterized by widespread vasculopathy and autoimmune reactions that evolve into progressive interstitial, perivascular, and vessel wall fibrosis that affects the skin and multiple internal organs. Such an uncontrolled fibrotic process gradually disrupts the physiologic architecture of the affected tissues and frequently leads to significant organ dysfunction, thus representing a major cause of death in SSc patients. The main fibrosis orchestrators in SSc are represented by chronically activated myofibroblasts, a peculiar population of mesenchymal cells combining the extracellular matrix-synthesizing features of fibroblasts with cytoskeletal characteristics of contractile smooth muscle cells. Multiple lines of evidence support the notion that profibrotic myofibroblasts may derive not only from the activation of tissue resident fibroblasts but also from a variety of additional cell types, including pericytes, epithelial cells, vascular endothelial cells and preadipocytes/adipocytes. Here we overview an emerging picture that espouses that several cell transitional processes may be novel essential contributors to the pool of profibrotic myofibroblasts in SSc, potentially representing new suitable targets for therapeutic purposes. An in-depth dissection of the multiple origins of myofibroblasts and the underlying molecular mechanisms may be crucial in the process of deciphering the cellular bases of fibrosis persistence and refractoriness to the treatment and, therefore, may help in developing more effective and personalized therapeutic opportunities for SSc patients.
Collapse
Affiliation(s)
- Irene Rosa
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Mirko Manetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
17
|
Lerbs T, Cui L, King ME, Chai T, Muscat C, Chung L, Brown R, Rieger K, Shibata T, Wernig G. CD47 prevents the elimination of diseased fibroblasts in scleroderma. JCI Insight 2020; 5:140458. [PMID: 32814713 PMCID: PMC7455137 DOI: 10.1172/jci.insight.140458] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/08/2020] [Indexed: 12/26/2022] Open
Abstract
Scleroderma is a devastating fibrotic autoimmune disease. Current treatments are partly effective in preventing disease progression but do not remove fibrotic tissue. Here, we evaluated whether scleroderma fibroblasts take advantage of the "don't-eat-me-signal" CD47 and whether blocking CD47 enables the body's immune system to get rid of diseased fibroblasts. To test this approach, we used a Jun-inducible scleroderma model. We first demonstrated in patient samples that scleroderma upregulated transcription factor JUN and increased promoter accessibilities of both JUN and CD47. Next, we established our scleroderma model, demonstrating that Jun mediated skin fibrosis through the hedgehog-dependent expansion of CD26+Sca1- fibroblasts in mice. In a niche-independent adaptive transfer model, JUN steered graft survival and conferred increased self-renewal to fibroblasts. In vivo, JUN enhanced the expression of CD47, and inhibiting CD47 eliminated an ectopic fibroblast graft and increased in vitro phagocytosis. In the syngeneic mouse, depleting macrophages ameliorated skin fibrosis. Therapeutically, combined CD47 and IL-6 blockade reversed skin fibrosis in mice and led to the rapid elimination of ectopically transplanted scleroderma cells. Altogether, our study demonstrates the efficiency of combining different immunotherapies in treating scleroderma and provides a rationale for combining CD47 and IL-6 inhibition in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Tim Chai
- Institute for Stem Cell Biology and Regenerative Medicine, and
| | | | - Lorinda Chung
- Division of Immunology and Rheumatology, Department of Medicine, Stanford School of Medicine, Stanford, California, USA
| | | | | | | | - Gerlinde Wernig
- Department of Pathology.,Institute for Stem Cell Biology and Regenerative Medicine, and
| |
Collapse
|
18
|
Feng D, Gerarduzzi C. Emerging Roles of Matricellular Proteins in Systemic Sclerosis. Int J Mol Sci 2020; 21:E4776. [PMID: 32640520 PMCID: PMC7369781 DOI: 10.3390/ijms21134776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis is a rare chronic heterogenous disease that involves inflammation and vasculopathy, and converges in end-stage development of multisystem tissue fibrosis. The loss of tight spatial distribution and temporal expression of proteins in the extracellular matrix (ECM) leads to progressive organ stiffening, which is a hallmark of fibrotic disease. A group of nonstructural matrix proteins, known as matricellular proteins (MCPs) are implicated in dysregulated processes that drive fibrosis such as ECM remodeling and various cellular behaviors. Accordingly, MCPs have been described in the context of fibrosis in sclerosis (SSc) as predictive disease biomarkers and regulators of ECM synthesis, with promising therapeutic potential. In this present review, an informative summary of major MCPs is presented highlighting their clear correlations to SSc- fibrosis.
Collapse
Affiliation(s)
- Daniel Feng
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l’Université de Montréal, Montréal, QC H1T 2M4, Canada
| | - Casimiro Gerarduzzi
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l’Université de Montréal, Montréal, QC H1T 2M4, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
19
|
Liang M, Lv J, Jiang Z, He H, Chen C, Xiong Y, Zhu X, Xue Y, Yu Y, Yang S, Wang L, Li W, Guan M, Wan W, He R, Zou H. Promotion of Myofibroblast Differentiation and Tissue Fibrosis by the Leukotriene B 4 -Leukotriene B 4 Receptor Axis in Systemic Sclerosis. Arthritis Rheumatol 2020; 72:1013-1025. [PMID: 31872544 DOI: 10.1002/art.41192] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate the role of the inflammatory lipid mediator leukotriene B4 (LTB4 ) and its receptor, BLT1, in the development and progression of systemic sclerosis (SSc). METHODS Serum levels of LTB4 were compared in 64 patients with SSc and 80 healthy controls. Skin and lung tissue sections from patients with SSc and healthy donors were immunostained for leukotriene A4 hydrolase (LTA4 H), the critical enzyme for LTB4 synthesis, and BLT1, in combination with different cell markers. In mouse models of SSc using bleomycin or angiotensin II challenge or immunization with the DNA topoisomerase I, genetic or pharmacologic interruption of the LTB4 -BLT1 axis in mice was carried out to assess its effects on systemic disease features and myofibroblast markers. Immunoblotting was performed to examine the signaling pathway in fibroblasts and endothelial cells following stimulation with LTB4 or with serum from SSc patients. RESULTS Serum LTB4 levels were 44.93% higher in patients with SSc than in matched healthy controls (mean ± SD 220.3 ± 74.75 pg/ml versus 152.0 ± 68.05 pg/ml; P < 0.0001), and this was associated with the patient subsets of SSc-associated interstitial lung disease and diffuse cutaneous SSc. Levels of LTA4 H and BLT1 were increased in lesional areas of the skin and lungs of SSc patients, and both were abundant in myofibroblasts and endothelial cells. Interruption of the LTB4 -BLT1 axis in mouse models of SSc significantly mitigated dermal and pulmonary fibrosis, with 54.00% and 52.65% fewer α-smooth muscle actin-positive myofibroblasts accumulating in the skin and lungs of mice, respectively, after bleomycin challenge. Immunoblotting of cultures with recombinant LTB4 -stimulated fibroblasts and endothelial cells or with serum from SSc patients showed that fibroblast-myofibroblast and endothelial-mesenchymal transitions were promoted via BLT1, and that this was dependent on activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/mechanistic target of rapamycin (mTOR) pathway but independent of the release of transforming growth factor β (TGFβ) by fibroblasts or endothelial cells. CONCLUSION The LTB4 -BLT1 axis may contribute to fibrosis in SSc by directly promoting myofibroblast differentiation via the PI3K/Akt/mTOR pathway, and this appears to operate independently of autocrine secretion of TGFβ.
Collapse
Affiliation(s)
- Minrui Liang
- Huashan Hospital and Fudan University, Shanghai, China
| | - Jiaoyan Lv
- Fudan University, Shanghai, China, and Tsinghua University School of Medicine, Beijing, China
| | - Zhixing Jiang
- Huashan Hospital and Fudan University, Shanghai, China
| | - Hang He
- Fudan University, Shanghai, China
| | - Chen Chen
- Huashan Hospital and Fudan University, Shanghai, China
| | | | - Xiaoxia Zhu
- Huashan Hospital and Fudan University, Shanghai, China
| | - Yu Xue
- Huashan Hospital and Fudan University, Shanghai, China
| | - Yiyun Yu
- Huashan Hospital and Fudan University, Shanghai, China
| | - Sen Yang
- Huashan Hospital and Fudan University, Shanghai, China
| | - Lingbiao Wang
- Huashan Hospital and Fudan University, Shanghai, China
| | | | - Ming Guan
- Huashan Hospital and Fudan University, Shanghai, China
| | - Weiguo Wan
- Huashan Hospital and Fudan University, Shanghai, China
| | - Rui He
- Fudan University, Shanghai, China
| | - Hejian Zou
- Huashan Hospital and Fudan University, Shanghai, China
| |
Collapse
|
20
|
Tsai CY, Hsieh SC, Wu TH, Li KJ, Shen CY, Liao HT, Wu CH, Kuo YM, Lu CS, Yu CL. Pathogenic Roles of Autoantibodies and Aberrant Epigenetic Regulation of Immune and Connective Tissue Cells in the Tissue Fibrosis of Patients with Systemic Sclerosis. Int J Mol Sci 2020; 21:ijms21093069. [PMID: 32349208 PMCID: PMC7246753 DOI: 10.3390/ijms21093069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a multi-system autoimmune disease with tissue fibrosis prominent in the skin and lung. In this review, we briefly describe the autoimmune features (mainly autoantibody production and cytokine profiles) and the potential pathogenic contributors including genetic/epigenetic predisposition, and environmental factors. We look in detail at the cellular and molecular bases underlying tissue-fibrosis which include trans-differentiation of fibroblasts (FBs) to myofibroblasts (MFBs). We also state comprehensively the pro-inflammatory and pro-fibrotic cytokines relevant to MFB trans-differentiation, vasculopathy-associated autoantibodies, and fibrosis-regulating microRNAs in SSc. It is conceivable that tissue fibrosis is mainly mediated by an excessive production of TGF-β, the master regulator, from the skewed Th2 cells, macrophages, fibroblasts, myofibroblasts, and keratinocytes. After binding with TGF-β receptors on MFB, the downstream Wnt/β-catenin triggers canonical Smad 2/3 and non-canonical Smad 4 signaling pathways to transcribe collagen genes. Subsequently, excessive collagen fiber synthesis and accumulation as well as tissue fibrosis ensue. In the later part of this review, we discuss limited data relevant to the role of long non-coding RNAs (lncRNAs) in tissue-fibrosis in SSc. It is expected that these lncRNAs may become the useful biomarkers and therapeutic targets for SSc in the future. The prospective investigations in the development of novel epigenetic modifiers are also suggested.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Cheng-Shiun Lu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| |
Collapse
|
21
|
Eckel-Mahan K, Ribas Latre A, Kolonin MG. Adipose Stromal Cell Expansion and Exhaustion: Mechanisms and Consequences. Cells 2020; 9:cells9040863. [PMID: 32252348 PMCID: PMC7226766 DOI: 10.3390/cells9040863] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue (AT) is comprised of a diverse number of cell types, including adipocytes, stromal cells, endothelial cells, and infiltrating leukocytes. Adipose stromal cells (ASCs) are a mixed population containing adipose progenitor cells (APCs) as well as fibro-inflammatory precursors and cells supporting the vasculature. There is growing evidence that the ability of ASCs to renew and undergo adipogenesis into new, healthy adipocytes is a hallmark of healthy fat, preventing disease-inducing adipocyte hypertrophy and the spillover of lipids into other organs, such as the liver and muscles. However, there is building evidence indicating that the ability for ASCs to self-renew is not infinite. With rates of ASC proliferation and adipogenesis tightly controlled by diet and the circadian clock, the capacity to maintain healthy AT via the generation of new, healthy adipocytes appears to be tightly regulated. Here, we review the contributions of ASCs to the maintenance of distinct adipocyte pools as well as pathogenic fibroblasts in cancer and fibrosis. We also discuss aging and diet-induced obesity as factors that might lead to ASC senescence, and the consequences for metabolic health.
Collapse
Affiliation(s)
- Kristin Eckel-Mahan
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
| | - Aleix Ribas Latre
- Helmholtz Institute for Metabolic, Obesity and Vascular Research Center, D-04103 Leipzig, Germany;
| | - Mikhail G. Kolonin
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX 77030, USA;
- Correspondence:
| |
Collapse
|
22
|
LeBleu VS, Neilson EG. Origin and functional heterogeneity of fibroblasts. FASEB J 2020; 34:3519-3536. [PMID: 32037627 DOI: 10.1096/fj.201903188r] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/12/2022]
Abstract
The inherent plasticity and resiliency of fibroblasts make this cell type a conventional tool for basic research. But where do they come from, are all fibroblasts the same, and how do they function in disease? The first fibroblast lineages in mammalian development emerge from the ooze of primary mesenchyme during gastrulation. They are cells that efficiently create and negotiate the extracellular matrix of the mesoderm in order to migrate and meet their developmental fate. Mature fibroblasts in epithelial tissues live in the interstitial spaces between basement membranes that spatially delimit complex organ structures. While the function of resident fibroblasts in healthy tissues is largely conjecture, the accumulation of fibroblasts in pathologic lesions offers insight into biologic mechanisms that control their function; fibroblasts are poised to coordinate fibrogenesis in tissue injury, neoplasia, and aging. Here, we examine the developmental origin and plasticity of fibroblasts, their molecular and functional definitions, the epigenetic control underlying their identity and activation, and the evolution of their immune regulatory functions. These topics are reviewed through the lens of fate mapping using genetically engineered mouse models and from the perspective of single-cell RNA sequencing. Recent observations suggest dynamic and heterogeneous functions for fibroblasts that underscore their complex molecular signatures and utility in injured tissues.
Collapse
Affiliation(s)
- Valerie S LeBleu
- Departments of Medicine and Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eric G Neilson
- Departments of Medicine and Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
23
|
Manetti M, Matucci-Cerinic M. In search for the ideal anatomical composition of vascularised human skin equivalents for systemic sclerosis translational research: should we recruit the telocytes? Ann Rheum Dis 2019; 80:e149. [PMID: 31582376 DOI: 10.1136/annrheumdis-2019-216371] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 01/11/2023]
Affiliation(s)
- Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Section of Rheumatology, University of Florence, Florence, Italy
| |
Collapse
|
24
|
Wollin L, Distler JHW, Denton CP, Gahlemann M. Rationale for the evaluation of nintedanib as a treatment for systemic sclerosis-associated interstitial lung disease. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2019; 4:212-218. [PMID: 35382502 PMCID: PMC8922567 DOI: 10.1177/2397198319841842] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/25/2019] [Indexed: 03/23/2024]
Abstract
Interstitial lung disease is a common manifestation of systemic sclerosis. Systemic sclerosis-associated interstitial lung disease is characterized by progressive pulmonary fibrosis and a reduction in pulmonary function. Effective treatments for systemic sclerosis-associated interstitial lung disease are lacking. In addition to clinical similarities, systemic sclerosis-associated interstitial lung disease shows similarities to idiopathic pulmonary fibrosis in the pathophysiology of the underlying fibrotic processes. Idiopathic pulmonary fibrosis and systemic sclerosis-associated interstitial lung disease culminate in a self-sustaining pathway of pulmonary fibrosis in which fibroblasts are activated, myofibroblasts accumulate, and the excessive extracellular matrix is deposited. Nintedanib is a tyrosine kinase inhibitor that has been approved for the treatment of idiopathic pulmonary fibrosis. In patients with idiopathic pulmonary fibrosis, nintedanib slows disease progression by decreasing the rate of lung function decline. In this review, we summarize the antifibrotic, anti-inflammatory, and attenuated vascular remodeling effects of nintedanib demonstrated in in vitro studies and in animal models of aspects of systemic sclerosis. Nintedanib interferes at multiple critical steps in the pathobiology of systemic sclerosis-associated interstitial lung disease, providing a convincing rationale for its investigation as a potential therapy. Finally, we summarize the design of the randomized placebo-controlled SENSCIS® trial that is evaluating the efficacy and safety of nintedanib in patients with systemic sclerosis-associated interstitial lung disease.
Collapse
Affiliation(s)
- Lutz Wollin
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Jörg HW Distler
- Department of Internal Medicine 3 Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | | | | |
Collapse
|
25
|
Carvalheiro T, Affandi AJ, Malvar-Fernández B, Dullemond I, Cossu M, Ottria A, Mertens JS, Giovannone B, Bonte-Mineur F, Kok MR, Marut W, Reedquist KA, Radstake TR, García S. Induction of Inflammation and Fibrosis by Semaphorin 4A in Systemic Sclerosis. Arthritis Rheumatol 2019; 71:1711-1722. [PMID: 31012544 PMCID: PMC6790618 DOI: 10.1002/art.40915] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/18/2019] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To analyze the potential role of semaphorin 4A (Sema4A) in inflammatory and fibrotic processes involved in the pathology of systemic sclerosis (SSc). METHODS Sema4A levels in the plasma of healthy controls (n = 11) and SSc patients (n = 20) were determined by enzyme-linked immunosorbent assay (ELISA). The expression of Sema4A and its receptors in monocytes and CD4+ T cells from healthy controls and SSc patients (n = 6-7 per group) was determined by ELISA and flow cytometry. Th17 cytokine production by CD4+ T cells (n = 5-7) was analyzed by ELISA and flow cytometry. The production of inflammatory mediators and extracellular matrix (ECM) components by dermal fibroblast cells (n = 6) was analyzed by quantitative polymerase chain reaction, ELISA, Western blotting, confocal microscopy, and ECM deposition assay. RESULTS Plasma levels of Sema4A, and Sema4A expression by circulating monocytes and CD4+ T cells, were significantly higher in SSc patients than in healthy controls (P < 0.05). Inflammatory mediators significantly up-regulated the secretion of Sema4A by monocytes and CD4+ T cells from SSc patients (P < 0.05 versus unstimulated SSc cells). Functional assays showed that Sema4A significantly enhanced the expression of Th17 cytokines induced by CD3/CD28 in total CD4+ T cells as well in different CD4+ T cell subsets (P < 0.05 versus unstimulated SSc cells). Finally, Sema4A induced a profibrotic phenotype in dermal fibroblasts from both healthy controls and SSc patients, which was abrogated by blocking or silencing the expression of Sema4A receptors. CONCLUSION Our findings indicate that Sema4A plays direct and dual roles in promoting inflammation and fibrosis, 2 main features of SSc, suggesting that Sema4A might be a novel therapeutic target in SSc.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Alsya J Affandi
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | | | - Ilse Dullemond
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Marta Cossu
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Andrea Ottria
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jorre S Mertens
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Barbara Giovannone
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | | | - Marc R Kok
- Maasstad Hospital Rotterdam, Rotterdam, The Netherlands
| | - Wioleta Marut
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Kris A Reedquist
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Timothy R Radstake
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Samuel García
- University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| |
Collapse
|
26
|
Manetti M, Romano E, Rosa I, Fioretto BS, Praino E, Guiducci S, Iannone F, Ibba-Manneschi L, Matucci-Cerinic M. Systemic Sclerosis Serum Steers the Differentiation of Adipose-Derived Stem Cells Toward Profibrotic Myofibroblasts: Pathophysiologic Implications. J Clin Med 2019; 8:E1256. [PMID: 31430950 PMCID: PMC6723717 DOI: 10.3390/jcm8081256] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/06/2019] [Accepted: 08/16/2019] [Indexed: 12/16/2022] Open
Abstract
Systemic sclerosis (SSc; scleroderma) is characterized by life-threatening progressive multiorgan fibrosis orchestrated by profibrotic myofibroblasts originating from different sources. Because recent data demonstrated that the majority of myofibroblasts in a murine scleroderma model arise from adipocytic progenitors through the adipocyte-myofibroblast transition process, we sought to determine whether the SSc microenvironment may affect the differentiation potential of adipose-derived stem cells (ADSC). Normal human ADSC from three donors were treated with serum from SSc patients (n = 6), serum from healthy individuals (n = 6), or recombinant human transforming growth factor-β1 (TGFβ1) as positive control of myofibroblastic phenotype induction. ADSC were subjected to in vitro adipogenic differentiation for up to 21 days in the presence of different stimuli followed by lipid content quantification. In selected experiments, adipocytic and mesenchymal/myofibroblast marker gene and protein expression levels were assessed by Real-Time PCR, immunoblotting and immunofluorescence after administration of different stimuli for 72 and 96 h, respectively. Cell contractile phenotype was assayed by collagen gel contraction assay. Likewise stimulation with TGFβ1, SSc serum was able to significantly inhibit the adipocyte differentiation of ADSC as testified by a strong decrease in red-colored lipid droplets after 21 days of adipogenic induction. Treatment of ADSC either with SSc serum or TGFβ1 resulted in the acquisition of a myofibroblast-like phenotype characterized by a reduced expression of the adipocytic markers perilipin and adiponectin, a significant upregulation of the mesenchymal/myofibroblast markers α-SMA+ stress fibers, S100A4 and type I collagen, and an ability to effectively contract collagen gels. In SSc, the pathologic environment may favor the differentiation of ADSC into profibrotic and contractile myofibroblast-like cells. These findings strengthen the notion that the generation of myofibroblasts from ADSC may be relevant in SSc pathophysiology potentially representing a new target for the prevention/treatment of multiorgan fibrosis.
Collapse
Affiliation(s)
- Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy.
| | - Eloisa Romano
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Bianca Saveria Fioretto
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Emanuela Praino
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, 70121 Bari, Italy
| | - Serena Guiducci
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Florenzo Iannone
- Rheumatology Unit, Department of Emergency and Organ Transplantation, University of Bari, 70121 Bari, Italy
| | - Lidia Ibba-Manneschi
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Matucci-Cerinic
- Division of Rheumatology and Scleroderma Unit, AOUC, Department of Geriatric Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
27
|
Korman B. Evolving insights into the cellular and molecular pathogenesis of fibrosis in systemic sclerosis. Transl Res 2019; 209:77-89. [PMID: 30876809 PMCID: PMC6545260 DOI: 10.1016/j.trsl.2019.02.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/27/2019] [Accepted: 02/20/2019] [Indexed: 01/11/2023]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex multisystem disease characterized by autoimmunity, vasculopathy, and most notably, fibrosis. Multiple lines of evidence demonstrate a variety of emerging cellular and molecular pathways which are relevant to fibrosis in SSc. The myofibroblast remains the key effector cell in SSc. Understanding the development, differentiation, and function of the myofibroblast is therefore crucial to understanding the fibrotic phenotype of SSc. Studies now show that (1) multiple cell types give rise to myofibroblasts, (2) fibroblasts and myofibroblasts are heterogeneous, and (3) that a large number of (primarily immune) cells have important influences on the transition of fibroblasts to an activated myofibroblasts. In SSc, this differentiation process involves multiple pathways, including well known signaling cascades such as TGF-β and Wnt/β-Catenin signaling, as well as epigenetic reprogramming and a number of more recently defined cellular pathways. After reviewing the major and emerging cellular and molecular mechanisms underlying SSc, this article looks to identify clinical applications where this new molecular knowledge may allow for targeted treatment and personalized medicine approaches.
Collapse
Affiliation(s)
- Benjamin Korman
- Division of Allergy/Immunology & Rheumatology, University of Rochester Medical Center, Rochester, New York.
| |
Collapse
|
28
|
Shook BA, Wasko RR, Rivera-Gonzalez GC, Salazar-Gatzimas E, López-Giráldez F, Dash BC, Muñoz-Rojas AR, Aultman KD, Zwick RK, Lei V, Arbiser JL, Miller-Jensen K, Clark DA, Hsia HC, Horsley V. Myofibroblast proliferation and heterogeneity are supported by macrophages during skin repair. Science 2018; 362:eaar2971. [PMID: 30467144 PMCID: PMC6684198 DOI: 10.1126/science.aar2971] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 07/20/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022]
Abstract
During tissue repair, myofibroblasts produce extracellular matrix (ECM) molecules for tissue resilience and strength. Altered ECM deposition can lead to tissue dysfunction and disease. Identification of distinct myofibroblast subsets is necessary to develop treatments for these disorders. We analyzed profibrotic cells during mouse skin wound healing, fibrosis, and aging and identified distinct subpopulations of myofibroblasts, including adipocyte precursors (APs). Multiple mouse models and transplantation assays demonstrate that proliferation of APs but not other myofibroblasts is activated by CD301b-expressing macrophages through insulin-like growth factor 1 and platelet-derived growth factor C. With age, wound bed APs and differential gene expression between myofibroblast subsets are reduced. Our findings identify multiple fibrotic cell populations and suggest that the environment dictates functional myofibroblast heterogeneity, which is driven by fibroblast-immune interactions after wounding.
Collapse
Affiliation(s)
- Brett A Shook
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA.
| | - Renee R Wasko
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | | | | | | | - Biraja C Dash
- Department of Surgery (Plastic), Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Krystal D Aultman
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Rachel K Zwick
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Vivian Lei
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
| | - Jack L Arbiser
- Department of Dermatology, Atlanta Veterans Administration Health Center, Emory University, Atlanta, GA 30322, USA
| | - Kathryn Miller-Jensen
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Damon A Clark
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT 06511, USA
| | - Henry C Hsia
- Department of Surgery (Plastic), Yale School of Medicine, New Haven, CT 06510, USA
| | - Valerie Horsley
- Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA.
- Department of Dermatology, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
29
|
Liakouli V, Cipriani P, Di Benedetto P, Ruscitti P, Carubbi F, Berardicurti O, Panzera N, Giacomelli R. The role of extracellular matrix components in angiogenesis and fibrosis: Possible implication for Systemic Sclerosis. Mod Rheumatol 2018; 28:922-932. [DOI: 10.1080/14397595.2018.1431004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Vasiliki Liakouli
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Paola Cipriani
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Paola Di Benedetto
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Piero Ruscitti
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Francesco Carubbi
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Onorina Berardicurti
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Noemi Panzera
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| | - Roberto Giacomelli
- Department of Biotechnological and Applied Clinical Science, Rheumatology Unit, School of Medicine, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
30
|
Schulz JN, Plomann M, Sengle G, Gullberg D, Krieg T, Eckes B. New developments on skin fibrosis - Essential signals emanating from the extracellular matrix for the control of myofibroblasts. Matrix Biol 2018; 68-69:522-532. [PMID: 29408278 DOI: 10.1016/j.matbio.2018.01.025] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/29/2018] [Accepted: 01/29/2018] [Indexed: 02/06/2023]
Abstract
Many different diseases are associated with fibrosis of the skin. The clinical symptoms can vary considerably with a broad range from isolated small areas to the involvement of the entire integument. Fibrosis is triggered by a multitude of different stimuli leading to activation of the immune and vascular system that then initiate fibroblast activation and formation of matrix depositing and remodeling myofibroblasts. Ultimately, myofibroblasts deposit excessive amounts of extracellular matrix with a pathological architecture and alterations in growth factor binding and biomechanical properties, which culminates in skin hardening and loss of mobility. Treatment depends certainly on the specific type and cause of the disease, for the autoimmune driven localized and systemic scleroderma therapeutic options are still limited, but recent research has pointed out diverse molecular targets and mechanisms that can be exploited for the development of novel antifibrotic therapy.
Collapse
Affiliation(s)
| | - Markus Plomann
- Center for Biochemistry, University of Cologne, Medical Faculty, Cologne, Germany
| | - Gerhard Sengle
- Center for Biochemistry, University of Cologne, Medical Faculty, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Donald Gullberg
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), University of Bergen, Bergen, Norway
| | - Thomas Krieg
- Department of Dermatology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany; Cluster of Excellence in Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Beate Eckes
- Department of Dermatology, University of Cologne, Cologne, Germany; Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.
| |
Collapse
|
31
|
Abstract
Purpose of Review This review provides a summary of recent insights into the role of the local white adipose tissue (WAT) in systemic sclerosis. Recent Findings Adipocytes located in an interfacial WAT area adjacent to fibrotic lesions have an intermediate phenotype and special properties implicated in fibrotic pathology in systemic sclerosis (SSc). The important role of these cells is recognized in different pathologies, such as wound healing, psoriasis, breast cancer, and prostate cancer. Additionally, both immature and mature adipocytes are involved in the appearance of fibroblast-like cells but exhibit different phenotypes and synthetic properties. Summary Adipocytes from interfacial WAT adjacent to the fibrotic area in SSc are phenotypically different from bulk adipocytes and are involved in pathogenesis of SSc. Immature and mature adipocytes from this WAT layer differentiate into various types of fibroblast-like cells, making the local ratio of immature to mature adipocytes in interfacial WAT of particular importance in SSc pathogenesis.
Collapse
|
32
|
Ibba-Manneschi L, Rosa I, Manetti M. Telocytes in Chronic Inflammatory and Fibrotic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 913:51-76. [PMID: 27796880 DOI: 10.1007/978-981-10-1061-3_4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Telocytes are a peculiar stromal (interstitial) cell type implicated in tissue homeostasis and development, as well as in the pathophysiology of several disorders. Severe damage and reduction of telocytes have been reported during fibrotic remodeling of multiple organs in various diseases, including scleroderma, Crohn's disease, ulcerative colitis, and liver fibrosis, as well as in chronic inflammatory lesions like those of primary Sjögren's syndrome and psoriasis. Owing to their close relationship with stem cells, telocytes are also supposed to contribute to tissue repair/regeneration. Indeed, telocytes are universally considered as "connecting cells" mostly oriented to intercellular signaling. On the basis of recent promising experimental findings, in the near future, telocyte transplantation might represent a novel therapeutic opportunity to control the evolution of chronic inflammatory and fibrotic diseases. Notably, there is evidence to support that telocytes could help in preventing abnormal activation of immune cells and fibroblasts, as well as in attenuating the altered matrix organization during the fibrotic process. By targeting telocytes alone or in tandem with stem cells, we might be able to promote regeneration and prevent the evolution to irreversible tissue injury. Besides exogenous transplantation, exploring pharmacological or non-pharmacological methods to enhance the growth and/or survival of telocytes could be an additional therapeutic strategy for many disorders.
Collapse
Affiliation(s)
- Lidia Ibba-Manneschi
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, Florence, 50134, Italy.
| |
Collapse
|
33
|
Abstract
Excess ECM and fibrosis of white adipose tissue (WAT) is associated with tissue dysfunction and type 2 diabetes. In this issue of Cell Metabolism, Marcelin et al. (2017) elucidate a key mechanism behind WAT fibrosis in which the activation of PDGFRα on adipocyte precursors drives this population toward a fibrotic phenotype.
Collapse
Affiliation(s)
- Brett Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
| | - Matthew S Rodeheffer
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA; Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
34
|
Manetti M, Romano E, Rosa I, Guiducci S, Bellando-Randone S, De Paulis A, Ibba-Manneschi L, Matucci-Cerinic M. Endothelial-to-mesenchymal transition contributes to endothelial dysfunction and dermal fibrosis in systemic sclerosis. Ann Rheum Dis 2017; 76:924-934. [DOI: 10.1136/annrheumdis-2016-210229] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/15/2016] [Accepted: 12/17/2016] [Indexed: 12/15/2022]
Abstract
ObjectiveSystemic sclerosis (SSc) features multiorgan fibrosis orchestrated predominantly by activated myofibroblasts. Endothelial-to-mesenchymal transition (EndoMT) is a transdifferentiation by which endothelial cells (ECs) lose their specific morphology/markers and acquire myofibroblast-like features. Here, we determined the possible contribution of EndoMT to the pathogenesis of dermal fibrosis in SSc and two mouse models.MethodsSkin sections were immunostained for endothelial CD31 or vascular endothelial (VE)-cadherin in combination with α-smooth muscle actin (α-SMA) myofibroblast marker. Dermal microvascular ECs (dMVECs) were prepared from SSc and healthy skin (SSc-dMVECs and H-dMVECs). H-dMVECs were treated with transforming growth factor-β1 (TGFβ1) or SSc and healthy sera. Endothelial/mesenchymal markers were assessed by real-time PCR, immunoblotting and immunofluorescence. Cell contractile phenotype was assayed by collagen gel contraction.ResultsCells in intermediate stages of EndoMT were identified in dermal vessels of either patients with SSc or bleomycin-induced and urokinase-type plasminogen activator receptor (uPAR)-deficient mouse models. At variance with H-dMVECs, SSc-dMVECs exhibited a spindle-shaped appearance, co-expression of lower levels of CD31 and VE-cadherin with myofibroblast markers (α-SMA+ stress fibres, S100A4 and type I collagen), constitutive nuclear localisation of the EndoMT driver Snail1 and an ability to effectively contract collagen gels. Treatment of H-dMVECs either with SSc sera or TGFβ1 resulted in the acquisition of a myofibroblast-like morphology and contractile phenotype and downregulation of endothelial markers in parallel with the induction of mesenchymal markers. Matrix metalloproteinase-12-dependent uPAR cleavage was implicated in the induction of EndoMT by SSc sera.ConclusionsIn SSc, EndoMT may be a crucial event linking endothelial dysfunction and development of dermal fibrosis.
Collapse
|
35
|
Skin fibrosis: Models and mechanisms. Curr Res Transl Med 2016; 64:185-193. [PMID: 27939457 DOI: 10.1016/j.retram.2016.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 02/06/2023]
Abstract
Matrix synthesis, deposition and remodeling are complex biological processes that are critical in development, maintenance of tissue homeostasis and repair of injured tissues. Disturbances in the regulation of these processes can result in severe pathological conditions which are associated with tissue fibrosis as e.g. in Scleroderma, cutaneous Graft-versus-Host-Disease, excessive scarring after trauma or carcinogenesis. Therefore, finding efficient treatments to limit skin fibrosis is of major clinical importance. However the pathogenesis underlying the development of tissue fibrosis is still not entirely resolved. In recent years progress has been made unraveling the complex cellular and molecular mechanisms that determine fibrosis. Here we provide an overview of established and more recently developed mouse models that can be used to investigate the mechanisms of skin fibrosis and to test potential therapeutic approaches.
Collapse
|
36
|
Shook B, Rivera Gonzalez G, Ebmeier S, Grisotti G, Zwick R, Horsley V. The Role of Adipocytes in Tissue Regeneration and Stem Cell Niches. Annu Rev Cell Dev Biol 2016; 32:609-631. [PMID: 27146311 PMCID: PMC5157158 DOI: 10.1146/annurev-cellbio-111315-125426] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Classically, white adipose tissue (WAT) was considered an inert component of connective tissue but is now appreciated as a major regulator of metabolic physiology and endocrine homeostasis. Recent work defining how WAT develops and expands in vivo emphasizes the importance of specific locations of WAT or depots in metabolic regulation. Interestingly, mature white adipocytes are integrated into several tissues. A new perspective regarding the in vivo regulation and function of WAT in these tissues has highlighted an essential role of adipocytes in tissue homeostasis and regeneration. Finally, there has been significant progress in understanding how mature adipocytes regulate the pathology of several diseases. In this review, we discuss these novel roles of WAT in the homeostasis and regeneration of epithelial, muscle, and immune tissues and how they contribute to the pathology of several disorders.
Collapse
Affiliation(s)
- Brett Shook
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Guillermo Rivera Gonzalez
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Sarah Ebmeier
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | | | - Rachel Zwick
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
| | - Valerie Horsley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut 06520;
- Department of Dermatology, Yale University, New Haven, Connecticut 06520
| |
Collapse
|
37
|
Furtado MB, Nim HT, Boyd SE, Rosenthal NA. View from the heart: cardiac fibroblasts in development, scarring and regeneration. Development 2016; 143:387-97. [PMID: 26839342 DOI: 10.1242/dev.120576] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the adult, tissue repair after injury is generally compromised by fibrosis, which maintains tissue integrity with scar formation but does not restore normal architecture and function. The process of regeneration is necessary to replace the scar and rebuild normal functioning tissue. Here, we address this problem in the context of heart disease, and discuss the origins and characteristics of cardiac fibroblasts, as well as the crucial role that they play in cardiac development and disease. We discuss the dual nature of cardiac fibroblasts, which can lead to scarring, pathological remodelling and functional deficit, but can also promote heart function in some contexts. Finally, we review current and proposed approaches whereby regeneration could be fostered by interventions that limit scar formation.
Collapse
Affiliation(s)
- Milena B Furtado
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Hieu T Nim
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Sarah E Boyd
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia
| | - Nadia A Rosenthal
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia Systems Biology Institute (SBI) Australia, Monash University, Clayton, Victoria 3800, Australia National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK The Jackson Laboratory, Bar Harbor, ME 04609, USA
| |
Collapse
|
38
|
Ibba-Manneschi L, Rosa I, Manetti M. Telocyte implications in human pathology: An overview. Semin Cell Dev Biol 2016; 55:62-9. [PMID: 26805444 DOI: 10.1016/j.semcdb.2016.01.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 01/18/2016] [Indexed: 12/23/2022]
Abstract
Telocytes are a recently described interstitial cell population widely distributed in the stromal compartment of many organs in vertebrates, including humans. Owing to their close spatial relationship with multiple cell types, telocytes are universally considered as 'connecting cells' mostly committed to intercellular signaling by converting the interstitium into an integrated system that drives organ development and contributes to the maintenance of local tissue homeostasis. Increasing evidence indicates that telocytes may cooperate with tissue-resident stem cells to foster organ repair and regeneration, and that telocyte damage and dysfunction may occur in several disorders. The goal of this review is to provide an overview of the most recent findings concerning the implication of telocytes in a variety of pathologic conditions in humans, including heart disease, chronic inflammation and multiorgan fibrosis. Based on recent promising experimental data, there is realistic hope that by targeting telocytes alone or in tandem with stem cells, we might be able to promote organ regeneration and/or prevent irreversible end-stage organ damage in different pathologies.
Collapse
Affiliation(s)
- Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, 50134 Florence, Italy.
| |
Collapse
|