1
|
Mikhailova EV, Romanova IV, Bagrov AY, Agalakova NI. Fli1 and Tissue Fibrosis in Various Diseases. Int J Mol Sci 2023; 24:ijms24031881. [PMID: 36768203 PMCID: PMC9915382 DOI: 10.3390/ijms24031881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 01/21/2023] Open
Abstract
Being initially described as a factor of virally-induced leukemias, Fli1 (Friend leukemia integration 1) has attracted considerable interest lately due to its role in both healthy physiology and a variety of pathological conditions. Over the past few years, Fli1 has been found to be one of the crucial regulators of normal hematopoiesis, vasculogenesis, and immune response. However, abnormal expression of Fli1 due to genetic predisposition, epigenetic reprogramming (modifications), or environmental factors is associated with a few diseases of different etiology. Fli1 hyperexpression leads to malignant transformation of cells and progression of cancers such as Ewing's sarcoma. Deficiency in Fli1 is implicated in the development of systemic sclerosis and hypertensive disorders, which are often accompanied by pronounced fibrosis in different organs. This review summarizes the initial findings and the most recent advances in defining the role of Fli1 in diseases of different origin with emphasis on its pro-fibrotic potential.
Collapse
Affiliation(s)
- Elena V. Mikhailova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Avenue, 194223 Saint-Petersburg, Russia
| | - Irina V. Romanova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Avenue, 194223 Saint-Petersburg, Russia
| | | | - Natalia I. Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Avenue, 194223 Saint-Petersburg, Russia
| |
Collapse
|
2
|
Shi R, Zhang Z, Zhu A, Xiong X, Zhang J, Xu J, Sy MS, Li C. Targeting Type I Collagen for Cancer Treatment. Int J Cancer 2022; 151:665-683. [PMID: 35225360 DOI: 10.1002/ijc.33985] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/11/2022] [Accepted: 02/14/2022] [Indexed: 11/07/2022]
Abstract
Collagen is the most abundant protein in animals. Interactions between tumor cells and collagen influence every step of tumor development. Type I collagen is the main fibrillar collagen in the extracellular matrix and is frequently up-regulated during tumorigenesis. The binding of type I collagen to its receptors on tumor cells promotes tumor cell proliferation, epithelial-mesenchymal transition, and metastasis. Type I collagen also regulates the efficacy of tumor therapies, such as chemotherapy, radiotherapy, and immunotherapy. Furthermore, type I collagen fragments are diagnostic markers of metastatic tumors and have prognostic value. Inhibition of type I collagen synthesis has been reported to have anti-tumor effects in animal models. However, collagen has also been shown to possess anti-tumor activity. Therefore, the roles that type I collagen plays in tumor biology are complex and tumor type-dependent. In this review, we discuss the expression and regulation of synthesis of type I collagen, as well as the role up-regulated type I collagen plays in various stages of cancer progression. We also discuss the role of collagen in tumor therapy. Finally, we highlight several recent approaches targeting type I collagen for cancer treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Run Shi
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Zhe Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Ankai Zhu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Xingxing Xiong
- Department of Operating Room, Jiangxi Cancer Hospital of Nanchang University, Nanchang, China
| | - Jie Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| | - Jiang Xu
- Department of Stomatology, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, China
| | - Man-Sun Sy
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Chaoyang Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong High Education Institute, Guangzhou, China
| |
Collapse
|
3
|
Cheng Z, Zhang J, Deng W, Lin S, Li D, Zhu K, Qi Q. Bushen Yijing Decoction (BSYJ) exerts an anti-systemic sclerosis effect via regulating MicroRNA-26a /FLI1 axis. Bioengineered 2021; 12:1212-1225. [PMID: 33843426 PMCID: PMC8806208 DOI: 10.1080/21655979.2021.1907128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/18/2021] [Accepted: 03/18/2021] [Indexed: 11/23/2022] Open
Abstract
Systemic sclerosis (SSc) refers to a group of autoimmune rheumatic diseases. Bushen Yijing decoction (BSYJ) is used for treating SSc. However, its underlying mechanism remains unknown. The present study aims to investigate potential roles of Friend leukemia integration factor 1 (FLI1) and microRNA in the beneficial effects of BSYJ on SSc. Primary skin fibroblasts were isolated from healthy individuals and SSc patients through tissue-explant technique and validated by immunocytochemistry. mRNA and microRNA levels were determined by quantitative RT-PCR. Protein expression was measured by western blotting. MiR-26a mimics or inhibitor were transfected to induce miR-26a overexpression or knockdown in vitro and in vivo, respectively. Histological changes of skin tissues from SSc mouse were evaluated by H&E and Masson trichrome staining. Results showed that FLI1 expression significantly decreased in primary skin fibroblasts of SSc patients. MiR-26a was predicted to target FLI1 untranslated region. Transfection of miR-26 mimics in SSc skin fibroblasts (SFB) leads to decrease in FLI1 expression and increase in collagen I gene expression and fibronectin accumulation. On the other hand, miR-26a knockdown increased FLI1 expression and decreased collagen I and fibronectin expression in SFB. In addition, BSYJ-containing rat serum suppressed miR-26a expression, while it elevated FLI1 expression and inhibited fibronectin and collagen I accumulation in SFB. In the mouse SSc model, BSYJ-containing serum inhibited dermal fibrosis by suppressing miR-26a expression and restoring FLI1 protein levels. Overall, our study demonstrates that BSYJ decoction exerts anti-dermal fibrosis in SSc patients via suppressing miR-26a level and thus to increase FLI1 expression in fibroblasts.
Collapse
Affiliation(s)
- Zixuan Cheng
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jialin Zhang
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wanying Deng
- Department of Dermatology, The First Affiliated Hospital, School of Clinical Medicine of Guangdong, Pharmaceutical University, Guangzhou, China
| | - Shaojian Lin
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Donghai Li
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ke Zhu
- Department of Dermatology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Qi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Dermatology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Yamashita K, Kawasaki A, Matsushita T, Furukawa H, Kondo Y, Okiyama N, Nagaoka S, Shimada K, Sugii S, Katayama M, Hirohata S, Okamoto A, Chiba N, Suematsu E, Setoguchi K, Migita K, Sumida T, Tohma S, Hamaguchi Y, Hasegawa M, Sato S, Kawaguchi Y, Takehara K, Tsuchiya N. Association of functional (GA)n microsatellite polymorphism in the FLI1 gene with susceptibility to human systemic sclerosis. Rheumatology (Oxford) 2021; 59:3553-3562. [PMID: 32696043 DOI: 10.1093/rheumatology/keaa306] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 04/08/2020] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVE Susceptibility genes that can account for characteristic features of SSc such as fibrosis, vasculopathy and autoimmunity remain to be determined. In mice, deficiency of Friend leukaemia integration 1 transcription factor (Fli1) causes SSc-like disease with these features. The human FLI1 gene contains (GA)n microsatellite, which has been shown to be associated with expression level. Because microsatellite polymorphisms are difficult to capture by genome-wide association studies, we directly genotyped FLI1 (GA)n microsatellite and examined its association with SSc. METHODS Genomic DNA from 639 Japanese SSc patients and 851 healthy controls was genotyped for (GA)n microsatellite using the fragment assay. The cut-off repeat number for susceptibility to SSc was determined by receiver operating characteristics (ROC) analysis. Association with susceptibility and clinical characteristics was examined using logistic regression analysis. FLI1 mRNA levels were determined using quantitative RT-PCR. RESULTS Based on the ROC analysis, (GA)n alleles with ≥22 repeats were collectively defined as L alleles and alleles with ≤21 repeats as S alleles. (GA)n L alleles were significantly associated with susceptibility to SSc (P = 5.0e-04, odds ratio 1.34, additive model). Significant association was observed both in diffuse cutaneous and limited cutaneous SSc. Among the SSc, (GA)n L alleles were significantly enriched in the patients with a modified Rodnan total skin thickness score ≥10 compared with those with a score <10. FLI1 mRNA levels were significantly decreased in healthy controls carrying (GA)n L alleles as compared with non-carriers. CONCLUSION Extended repeat alleles of FLI1 (GA)n microsatellite may be associated with lower FLI1 mRNA levels and susceptibility to human SSc.
Collapse
Affiliation(s)
- Keita Yamashita
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Laboratory Medicine, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Aya Kawasaki
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | - Hiroshi Furukawa
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
| | - Yuya Kondo
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Naoko Okiyama
- Department of Dermatology, University of Tsukuba, Tsukuba, Japan
| | - Shouhei Nagaoka
- Department of Rheumatology, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Kota Shimada
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Shoji Sugii
- Department of Rheumatology, Tokyo Metropolitan Tama Medical Center, Fuchu, Japan
| | - Masao Katayama
- Department of Internal Medicine, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Shunsei Hirohata
- Department of Rheumatology and Infectious Diseases, Kitasato University School of Medicine, Sagamihara, Japan
| | - Akira Okamoto
- Department of Rheumatology, National Hospital Organization Himeji Medical Center, Himeji, Japan
| | - Noriyuki Chiba
- Department of Rheumatology, National Hospital Organization Morioka Medical Center, Morioka, Japan
| | - Eiichi Suematsu
- Department of Internal Medicine and Rheumatology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Keigo Setoguchi
- Allergy and Immunological Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takayuki Sumida
- Department of Internal Medicine, University of Tsukuba, Tsukuba, Japan
| | - Shigeto Tohma
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization Sagamihara Hospital, Sagamihara, Japan.,Department of Rheumatology, National Hospital Organization Tokyo National Hospital, Kiyose, Japan
| | | | - Minoru Hasegawa
- Department of Dermatology, University of Fukui, Fukui, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo, Tokyo, Japan
| | - Yasushi Kawaguchi
- Department of Rheumatology, Tokyo Women's Medical University School of Medicine, Tokyo, Japan
| | | | - Naoyuki Tsuchiya
- Molecular and Genetic Epidemiology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan.,Doctoral Program in Biomedical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
5
|
Wang X, Lennard Richard M, Li P, Henry B, Schutt S, Yu XZ, Fan H, Zhang W, Gilkeson G, Zhang XK. Expression of GM-CSF Is Regulated by Fli-1 Transcription Factor, a Potential Drug Target. THE JOURNAL OF IMMUNOLOGY 2020; 206:59-66. [PMID: 33268481 DOI: 10.4049/jimmunol.2000664] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022]
Abstract
Friend leukemia virus integration 1 (Fli-1) is an ETS transcription factor and a critical regulator of inflammatory mediators, including MCP-1, CCL5, IL-6, G-CSF, CXCL2, and caspase-1. GM-CSF is a regulator of granulocyte and macrophage lineage differentiation and a key player in the pathogenesis of inflammatory/autoimmune diseases. In this study, we demonstrated that Fli-1 regulates the expression of GM-CSF in both T cells and endothelial cells. The expression of GM-CSF was significantly reduced in T cells and endothelial cells when Fli-1 was reduced. We found that Fli-1 binds directly to the GM-CSF promoter using chromatin immunoprecipitation assay. Transient transfection assays indicated that Fli-1 drives transcription from the GM-CSF promoter in a dose-dependent manner, and mutation of the Fli-1 DNA binding domain resulted in a significant loss of transcriptional activation. Mutation of a known phosphorylation site within the Fli-1 protein led to a significant increase in GM-CSF promoter activation. Thus, direct binding to the promoter and phosphorylation are two important mechanisms behind Fli-1-driven activation of the GM-CSF promoter. In addition, Fli-1 regulates GM-CSF expression in an additive manner with another transcription factor Sp1. Finally, we demonstrated that a low dose of a chemotherapeutic drug, camptothecin, inhibited expression of Fli-1 and reduced GM-CSF production in human T cells. These results demonstrate novel mechanisms for regulating the expression of GM-CSF and suggest that Fli-1 is a critical druggable regulator of inflammation and immunity.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.,Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Mara Lennard Richard
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425; and
| | - Brittany Henry
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Steven Schutt
- Department of Microbiology and Immunology, Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425; and
| | - Weiru Zhang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Gary Gilkeson
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Xian K Zhang
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425;
| |
Collapse
|
6
|
Penke LR, Peters-Golden M. Molecular determinants of mesenchymal cell activation in fibroproliferative diseases. Cell Mol Life Sci 2019; 76:4179-4201. [PMID: 31563998 PMCID: PMC6858579 DOI: 10.1007/s00018-019-03212-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/01/2019] [Accepted: 06/26/2019] [Indexed: 02/06/2023]
Abstract
Uncontrolled scarring, or fibrosis, can interfere with the normal function of virtually all tissues of the body, ultimately leading to organ failure and death. Fibrotic diseases represent a major cause of death in industrialized countries. Unfortunately, no curative treatments for these conditions are yet available, highlighting the critical need for a better fundamental understanding of molecular mechanisms that may be therapeutically tractable. The ultimate indispensable effector cells responsible for deposition of extracellular matrix proteins that comprise scars are mesenchymal cells, namely fibroblasts and myofibroblasts. In this review, we focus on the biology of these cells and the molecular mechanisms that regulate their pertinent functions. We discuss key pro-fibrotic mediators, signaling pathways, and transcription factors that dictate their activation and persistence. Because of their possible clinical and therapeutic relevance, we also consider potential brakes on mesenchymal cell activation and cellular processes that may facilitate myofibroblast clearance from fibrotic tissue-topics that have in general been understudied.
Collapse
Affiliation(s)
- Loka R Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
7
|
Control of lung myofibroblast transformation by monovalent ion transporters. CURRENT TOPICS IN MEMBRANES 2019. [PMID: 31196603 DOI: 10.1016/bs.ctm.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Myofibroblast differentiation is a critical process in the pathogenesis of tissue fibrosis. We focus our mini-review on recent data showing an implication of monovalent ion transporters in fibroblast to myofibroblast transformation of human lung fibroblasts (HLF). In cultured HLF, cardiotonic steroids (CTS) known as potent inhibitors of Na+,K+-ATPase suppress myofibroblast differentiation in parallel with up- and down-regulated expression of cyclooxygenase-2 (COX-2) and TGF-β receptor subunit TGFBR2, respectively. K+-free medium mimics antifibrotic action of CTS indicating a key role of elevated intracellular [Na+]i/[K+]i ratio. Augmented expression of COX-2 is abolished by inhibition of Na+/Ca2+ exchanger. Side-by-side with CTS acting via elevation of the [Na+]i/[K+]i ratio fibroblast to myofibroblast transformation is also suppressed by potent inhibitors of Ca2+-activated chloride channels tannic acid and K+,Cl- cotransporter DIOA. The relative impact of [Formula: see text] -mediated and -independent signaling triggered by elevated [Na+]i/[K+]i ratio and altered intracellular anion handling in transcriptomic changes involved in myofibroblast differentiation should be examined further.
Collapse
|
8
|
Abstract
Myofibroblast activation is a critical process in the pathogenesis of tissue fibrosis accounting for 45% of all deaths. No effective therapies are available for the treatment of fibrotic diseases. We focus our mini-review on recent data showing that cardiotonic steroids (CTS) that are known as potent inhibitors of Na+,K+-ATPase affect myofibroblast differentiation in a cell type-specific manner. In cultured human lung fibroblasts (HLF), epithelial cells, and cancer-associated fibroblasts, CTS blocked myofibroblast differentiation triggered by profibrotic cytokine TGF-β. In contrast, in the absence of TGF-β, CTS augmented myofibroblast differentiation of cultured cardiac fibroblasts. The cell type-specific action of CTS in myofibroblast differentiation is consistent with data obtained in in vivo studies. Thus, infusion of ouabain via osmotic mini-pumps attenuated the development of lung fibrosis in bleomycintreated mice, whereas marinobufagenin stimulated renal and cardiac fibrosis in rats with experimental renal injury. In TGF-β-treated HLF, suppression of myofibroblast differentiation by ouabain is mediated by elevation of the [Na+]i/[K+]i ratio and is accompanied by upregulation of cyclooxygenase COX-2 and downregulation of TGF-β receptor TGFBR2. Augmented expression of COX-2 is abolished by inhibition of Na+/Ca2+ exchanger, suggesting a key role of [Ca2+]i-mediated signaling. What is the relative impact in tissue fibrosis of [Na+]i,[K+]iindependent signaling documented in several types of CTS-treated cells? Do the different conformational transitions of Na+,K+-ATPase α1 subunit in the presence of ouabain and marinobufagenin contribute to their distinct involvement in myofibroblast differentiation? Additional experiments should be done to answer these questions and to develop novel pharmacological approaches for the treatment of fibrosis-related disorders.
Collapse
Affiliation(s)
- Sergei N. Orlov
- Faculty of Biology, Lomonosov Moscow State University, Russian Federation
| | - Jennifer La
- Department of Medicine, The University of Chicago, IL, United States
| | | | - Nickolai O. Dulin
- Department of Medicine, The University of Chicago, IL, United States
| |
Collapse
|
9
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|
10
|
Furue M, Mitoma C, Mitoma H, Tsuji G, Chiba T, Nakahara T, Uchi H, Kadono T. Pathogenesis of systemic sclerosis-current concept and emerging treatments. Immunol Res 2018; 65:790-797. [PMID: 28488090 DOI: 10.1007/s12026-017-8926-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is an intractable multifaceted disease with high mortality. Although its pathogenesis is not fully understood, recent studies have advanced our knowledge on SSc. The cardinal pathological features of SSc are autoimmunity, vasculopathy, and fibrosis. The B cells in SSc are constitutively activated and lead to the production of a plethora of autoantibodies, such as anti-topoisomerase I and anti-centromere antibodies. In addition to these autoantibodies, which are valuable for diagnostic criteria or biomarkers, many other autoantibodies targeting endothelial cells, including endothelin type A receptor and angiotensin II type I receptor, are known to be functional and induce activation or apoptosis of endothelial cells. The autoantibody-mediated endothelial cell perturbation facilitates inflammatory cell infiltration, cytokine production, and myofibroblastic transformation of fibroblasts and endothelial cells. Profibrotic cytokines, such as transforming growth factor β, connective tissue growth factor, interleukin 4/interleukin 13, and interleukin 6, play a pivotal role in collagen production from myofibroblasts. Specific treatments targeting these causative molecules may improve the clinical outcomes of patients with SSc. In this review, we summarize recent topics on the pathogenesis (autoantibodies, vasculopathy, and fibrosis), animal models, and emerging treatments for SSc.
Collapse
Affiliation(s)
- Masutaka Furue
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan. .,Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka, Japan. .,Division of Skin Surface Sensing, Department of Dermatology, Kyushu University, Fukuoka, Japan.
| | - Chikage Mitoma
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan.,Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka, Japan
| | - Hiroki Mitoma
- Department of Clinical Immunology and Rheumatology/Infectious Disease, Kyushu University, Fukuoka, Japan
| | - Gaku Tsuji
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan.,Research and Clinical Center for Yusho and Dioxin, Kyushu University, Fukuoka, Japan
| | - Takahito Chiba
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan.,Division of Skin Surface Sensing, Department of Dermatology, Kyushu University, Fukuoka, Japan
| | - Hiroshi Uchi
- Department of Dermatology, Kyushu University, Maidashi 3-1-1, Higashiku, Fukuoka, 812-8582, Japan
| | - Takafumi Kadono
- Department of Dermatology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
11
|
Fli1-haploinsufficient dermal fibroblasts promote skin-localized transdifferentiation of Th2-like regulatory T cells. Arthritis Res Ther 2018; 20:23. [PMID: 29415756 PMCID: PMC5803841 DOI: 10.1186/s13075-018-1521-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 01/19/2018] [Indexed: 12/15/2022] Open
Abstract
Background Friend leukemia virus integration 1 (Fli1) deficiency, a predisposing factor of systemic sclerosis (SSc), induces SSc-like phenotypes in various cell types. A recent study demonstrated the transdifferentiation of T helper type 2 cell (Th2)-like regulatory T cells (Tregs) in SSc lesional skin through interleukin (IL)-33 produced by fibroblasts. Therefore, we investigated the role of Fli1 deficiency in dermal fibroblast-mediated transdifferentiation of Tregs. Methods Cytokine expression was assessed in Tregs by flow cytometry and in skin samples and cultivated cells by immunostaining, immunoblotting, and/or qRT-PCR. Fli1 binding to the target gene promoters was examined by chromatin immunoprecipitation. Murine dermal fibroblasts and Tregs were cocultured with or without blocking antibodies against target cytokines. Results Th2- and Th17-like cell proportions in skin-homing Tregs were increased in bleomycin-treated Fli1+/− mice compared with bleomycin-treated wild-type mice, whereas Th1-, Th2-, and Th17-like cell proportions in splenic Tregs were comparable. Fli1+/− fibroblasts overproduced IL-33 and IL-6, in particular IL-33, and Fli1 occupied the IL33 and IL6 promoters in dermal fibroblasts. Importantly, the IL-4-producing cell proportion was significantly higher in wild-type Tregs cocultured with Fli1+/− fibroblasts than in those cocultured with wild-type fibroblasts, which were canceled by neutralizing anti-IL-33 antibody. Under the same coculture condition, an increased tendency of IL-17A-producing cell proportion, which was possibly mediated by IL-6, was evident. Conclusions Fli1 haploinsufficiency increases the proportions of Th2- and Th17-like Tregs in bleomycin-induced profibrotic skin conditions, in which IL-33-producing dermal fibroblasts contribute to Th2-like Treg transdifferentiation, suggesting a critical role of Fli1 deficiency in the interaction of dermal fibroblasts with immune cells in pathological skin fibrosis. Electronic supplementary material The online version of this article (10.1186/s13075-018-1521-3) contains supplementary material, which is available to authorized users.
Collapse
|
12
|
Asano Y. What can we learn from Fli1-deficient mice, new animal models of systemic sclerosis? JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2018; 3:6-13. [PMID: 35382130 DOI: 10.1177/2397198318758221] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2018] [Indexed: 01/06/2023]
Abstract
Systemic sclerosis is a complex multifactorial disease characterized by autoimmunity, vasculopathy, and selective organ fibrosis. A series of genetic and epidemiological studies have demonstrated that environmental influences play a central role in the onset of systemic sclerosis, while genetic factors determine the susceptibility to and the severity of this disease. Therefore, the identification of predisposing factors related to environmental influences would provide us with an informative clue to better understand the pathological process of this disease. Based on this concept, the deficiency of transcription factor Friend leukemia virus integration 1, which is epigenetically suppressed in systemic sclerosis, seems to be a potential candidate acting as the predisposing factor of this disease. Indeed, Fli1-mutated mice serve as a set of useful disease models to disclose the complex pathology of systemic sclerosis. This article overviews the recent advancement in systemic sclerosis animal models associated with Friend leukemia virus integration 1 deficiency.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, Graduate School of Medicine, The University of Tokyo, Tokyo - Japan
| |
Collapse
|
13
|
Aslani S, Sobhani S, Gharibdoost F, Jamshidi A, Mahmoudi M. Epigenetics and pathogenesis of systemic sclerosis; the ins and outs. Hum Immunol 2018; 79:178-187. [PMID: 29330110 DOI: 10.1016/j.humimm.2018.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 12/25/2017] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
Abstract
The pathogenesis of many diseases is influenced by environmental factors which can affect human genome and be inherited from generation to generation. Adverse environmental stimuli are recognized through the epigenetic regulatory complex, leading to gene expression alteration, which in turn culminates in disease outcomes. Three epigenetic regulatory mechanisms modulate the manifestation of a gene, namely DNA methylation, histone changes, and microRNAs. Both epigenetics and genetics have been implicated in the pathogenesis of systemic sclerosis (SSc) disease. Genetic inheritance rate of SSc is low and the concordance rate in both monozygotic (MZ) and dizygotic (DZ) twins is little, implying other possible pathways in SSc pathogenesis scenario. Here, we provide an extensive overview of the studies regarding different epigenetic events which may offer insights into the pathology of SSc. Furthermore, epigenetic-based interventions to treat SSc patients were discussed.
Collapse
Affiliation(s)
- Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Soheila Sobhani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Gharibdoost
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Fausther M, Lavoie EG, Goree JR, Dranoff JA. An Elf2-like transcription factor acts as repressor of the mouse ecto-5'-nucleotidase gene expression in hepatic myofibroblasts. Purinergic Signal 2017; 13:417-428. [PMID: 28667437 PMCID: PMC5714833 DOI: 10.1007/s11302-017-9570-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 05/12/2017] [Indexed: 01/20/2023] Open
Abstract
Hepatic fibrosis represents a pathological wound healing and tissue repair process triggered in response to chronic liver injury. A heterogeneous population of activated non-parenchymal liver cells, known as liver myofibroblasts, functions as the effector cells in hepatic fibrosis. Upon activation, liver myofibroblasts become fibrogenic, acquiring contractile properties and increasing collagen production capacity, while developing enhanced sensitivity to endogenous molecules and factors released in the local microenvironment. Hepatic extracellular adenosine is a bioactive small molecule, increasingly recognized as an important regulator of liver myofibroblast functions, and an important mediator in the pathogenesis of liver fibrosis overall. Remarkably, ecto-5'-nucleotidase/Nt5e/Cd73 enzyme, which accounts for the dominant adenosine-generating activity in the extracellular medium, is expressed by activated liver myofibroblasts. However, the molecular signals regulating Nt5e gene expression in liver myofibroblasts remain poorly understood. Here, we show that activated mouse liver myofibroblasts express Nt5e gene products and characterize the putative Nt5e minimal promoter in the mouse species. We describe the existence of an enhancer sequence upstream of the mouse Nt5e minimal promoter and establish that the mouse Nt5e minimal promoter transcriptional activity is negatively regulated by an Elf2-like Ets-related transcription factor in activated mouse liver myofibroblasts.
Collapse
Affiliation(s)
- Michel Fausther
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR, 72205, USA.
- Research Service, Central Arkansas Veterans Administration Health System, Little Rock, AR, 72205, USA.
| | - Elise G Lavoie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR, 72205, USA
- Research Service, Central Arkansas Veterans Administration Health System, Little Rock, AR, 72205, USA
| | - Jessica R Goree
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR, 72205, USA
- Research Service, Central Arkansas Veterans Administration Health System, Little Rock, AR, 72205, USA
| | - Jonathan A Dranoff
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR, 72205, USA
- Research Service, Central Arkansas Veterans Administration Health System, Little Rock, AR, 72205, USA
| |
Collapse
|
15
|
Stawski L, Marden G, Trojanowska M. The Activation of Human Dermal Microvascular Cells by Poly(I:C), Lipopolysaccharide, Imiquimod, and ODN2395 Is Mediated by the Fli1/FOXO3A Pathway. THE JOURNAL OF IMMUNOLOGY 2017; 200:248-259. [PMID: 29141862 DOI: 10.4049/jimmunol.1601968] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 10/17/2017] [Indexed: 01/15/2023]
Abstract
Endothelial cell (EC) dysfunction has been associated with inflammatory and autoimmune diseases; however, the factors contributing to this dysfunction have not been fully explored. Because activation of TLRs has been implicated in autoimmune diseases, the goal of this study was to determine the effects of TLR ligands on EC function. Human dermal microvascular ECs (HDMECs) treated with TLR3 [Poly(I:C)], TLR4 (LPS), and TLR7 (imiquimod) agonists showed decreased proliferation and a reduced total number of branching tubules in three-dimensional human dermal organoid ex vivo culture. In contrast, the TLR9 ligand class C, ODN2395, increased angiogenesis. The antiproliferative effects of TLR3, TLR4, and TLR7 ligands correlated with significant downregulation of a key regulator of vascular homeostasis, Fli1, whereas TLR9 increased Fli1 levels. Furthermore, Poly(I:C) and LPS induced endothelial to mesenchymal transition that was reversed by the pretreatment with TGF-β neutralizing Ab or re-expression of Fli1. We showed that Fli1 was required for the HDMEC proliferation by transcriptionally repressing FOXO3A. In contrast to TLR9, which suppressed activation of the FOXO3A pathway, TLR3, TLR4, and TLR7 ligands activated FOXO3A as indicated by decreased phosphorylation and increased nuclear accumulation. The inverse correlation between Fli1 and FOXO3A was also observed in the vasculature of scleroderma patients. This work revealed opposing effects of TLR9 and TLR3, TLR4, and TLR7 on the key angiogenic pathways, Fli1 and FOXO3A. Our results provide a mechanistic insight into the regulation of angiogenesis by TLRs and confirm a central role of Fli1 in regulating vascular homeostasis.
Collapse
Affiliation(s)
- Lukasz Stawski
- Section of Rheumatology, School of Medicine, Boston University, Boston, MA 02118
| | - Grace Marden
- Section of Rheumatology, School of Medicine, Boston University, Boston, MA 02118
| | - Maria Trojanowska
- Section of Rheumatology, School of Medicine, Boston University, Boston, MA 02118
| |
Collapse
|
16
|
Looney AP, Han R, Stawski L, Marden G, Iwamoto M, Trojanowska M. Synergistic Role of Endothelial ERG and FLI1 in Mediating Pulmonary Vascular Homeostasis. Am J Respir Cell Mol Biol 2017; 57:121-131. [PMID: 28248553 PMCID: PMC5516275 DOI: 10.1165/rcmb.2016-0200oc] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 02/02/2017] [Indexed: 01/09/2023] Open
Abstract
Endothelial cell (EC) activation underlies many vascular diseases, including pulmonary arterial hypertension (PAH). Several members of the E-twenty six (ETS) family of transcription factors are important regulators of the gene network governing endothelial homeostasis, and their aberrant expression is associated with pathological angiogenesis. The goal of this study was to determine whether deficiencies of the ETS family member, Friend leukemia integration 1 transcription factor (FLI1), and its closest homolog, ETS-related gene (ERG), are associated with PAH. We found that endothelial ERG was significantly reduced in the lung samples from patients with PAH, as well as in chronically hypoxic mice. Functional studies revealed that depletion of ERG or FLI1 in human pulmonary ECs led to increased expression of inflammatory genes, including IFN genes, whereas genes regulating endothelial homeostasis and cell-cell adhesion were down-regulated. Simultaneous knockdown of both ERG and FLI1 had synergistic or additive effects on the expression of these genes, suggesting that ERG and FLI1 coregulate at least a subset of their target genes. Functionally, knockdown of ERG and FLI1 induced cell monolayer permeability with a potency similar to that of vascular endothelial growth factor. Notably, stimulation of ECs with Toll-like receptor 3 ligand poly(I:C) suppressed ERG expression and induced ERG dissociation from the IFNB1 promoter, while promoting signal transducers and activators of transcription 1 (STAT1) recruitment. Consistent with the up-regulation of inflammatory genes seen in vitro, Erg and Fli1 double-heterozygote mice showed increased immune cell infiltration and expression of cytokines in the lung. In conclusion, loss of ERG and FLI1 might contribute to the pathogenesis of vascular lung complications through the induction of inflammation.
Collapse
Affiliation(s)
- Agnieszka P. Looney
- Boston University School of Medicine, Arthritis Center/Rheumatology, Boston, Massachusetts; and
| | - Rong Han
- Boston University School of Medicine, Arthritis Center/Rheumatology, Boston, Massachusetts; and
| | - Lukasz Stawski
- Boston University School of Medicine, Arthritis Center/Rheumatology, Boston, Massachusetts; and
| | - Grace Marden
- Boston University School of Medicine, Arthritis Center/Rheumatology, Boston, Massachusetts; and
| | - Masahiro Iwamoto
- Orthopaedic Research, The Children’s Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Maria Trojanowska
- Boston University School of Medicine, Arthritis Center/Rheumatology, Boston, Massachusetts; and
| |
Collapse
|
17
|
Yang Y, Kim HJ, Woo KJ, Cho D, Bang SI. Lipo-PGE1 suppresses collagen production in human dermal fibroblasts via the ERK/Ets-1 signaling pathway. PLoS One 2017. [PMID: 28644845 PMCID: PMC5482458 DOI: 10.1371/journal.pone.0179614] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dysregulation of collagen production contributes to various pathological processes, including tissue fibrosis as well as impaired wound healing. Lipo-prostaglandin E1 (Lipo-PGE1), a lipid microsphere-incorporated prostaglandin E1, is used as a vasodilator for the treatment of peripheral vascular diseases. Lipo-PGE1 was recently shown to enhance human dermal fibroblast (HDF) migration and in vivo wound healing. No published study has characterized the role of Lipo-PGE1 in collagen regulation in HDFs. Here, we investigated the cellular signaling mechanism by which Lipo-PGE1 regulates collagen in HDFs. Collagen production was evaluated by the Sircol collagen assay, Western blot analysis of type I collagen and real time PCR. Unexpectedly, Lipo-PGE1 decreased mRNA expression of collagen 1A1, 1A2, and 3A1. Lipo-PGE1 markedly inhibited type I collagen and total soluble collagen production. In addition, Lipo-PGE1 inhibited transforming growth factor-β-induced collagen expression via Smad2 phosphorylation. To further investigate whether extracellular signal-regulated kinase (ERK)/Ets-1 signaling, a crucial pathway in collagen regulation, is involved in Lipo-PGE1-inhibited collagen production, cells were pretreated with an ERK-specific inhibitor, PD98059, prior to the addition of Lipo-PGE1. Lipo-PGE1-inhibited collagen mRNA expression and total soluble collagen production were recovered by pretreatment with PD98059. Moreover, Lipo-PGE1 directly induced the phosphorylation of ERK. Furthermore, silencing of Ets-1 recovered Lipo-PGE1-inhibited collagen production and PD98059 blocked Lipo-PGE1-enhanced Ets-1 expression. The present study reveals an important role for Lipo-PGE1 as a negative regulator of collagen gene expression and production via ERK/Ets-1 signaling. These results suggest that Lipo-PGE1 could potentially be a therapeutic target in diseases with deregulated collagen turnover.
Collapse
Affiliation(s)
- Yoolhee Yang
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Jung Kim
- Institute of Women's Life Medical Science, Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, Korea
| | - Kyong-Je Woo
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Daeho Cho
- Department of Life Systems, Sookmyung Women's University, Seoul, Korea
| | - Sa Ik Bang
- Department of Plastic Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
18
|
Ahmadi M, Gharibi T, Dolati S, Rostamzadeh D, Aslani S, Baradaran B, Younesi V, Yousefi M. Epigenetic modifications and epigenetic based medication implementations of autoimmune diseases. Biomed Pharmacother 2017; 87:596-608. [DOI: 10.1016/j.biopha.2016.12.072] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023] Open
|
19
|
Fan X, Xie J, Tian J. Reducing Cardiac Fibrosis: Na/K-ATPase Signaling Complex as a Novel Target. ACTA ACUST UNITED AC 2017; 6. [PMID: 29034264 DOI: 10.4172/2329-6607.1000204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is a common pathological process in cardiac disease and may lead to heart failure. It can also cause sudden death even in those without cardiac symptoms. Tissue fibrosis can be categorized into two categories: replacement fibrosis (also called reparative fibrosis) and reactive fibrosis. In replacement fibrosis, infiltration of inflammatory cells and accumulation of Extracellular Matrix (ECM) proteins are the initial steps in forming scarlike fibrotic tissue after acute cardiac injury and cardiac cell necrosis. Reactive fibrosis can be formed in response to hormonal change and pressure or volume overload. Experimental studies in animals have identified important pathways such as the Renin-Angiotensin-Aldosterone System (RAAS) and the endothelin pathway that contribute to fibrosis formation. Despite the fact that clinical trials using RAAS inhibitors as therapies for reducing cardiac fibrosis and improving cardiac function have been promising, heart failure is still the leading cause of deaths in the United States. Intensive efforts have been made to find novel targets and to develop new treatments for cardiac fibrosis and heart failure in the past few decades. The Na/K-ATPase, a canonical ion transporter, has been shown to also function as a signal transducer and prolonged activation of Na/K-ATPase signaling has been found to promote the formation of cardiac fibrosis. Novel tools that block the activation of Na/K-ATPase signaling have been developed and have shown promise in reducing cardiac fibrosis. This review will discuss the recent development of novel molecular targets, focusing on the Na/K-ATPase signaling complex as a therapeutic target in treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- X Fan
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Xie
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Tian
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| |
Collapse
|
20
|
Asano Y. Recent advances in animal models of systemic sclerosis. J Dermatol 2017; 43:19-28. [PMID: 26782003 DOI: 10.1111/1346-8138.13185] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 12/19/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem connective tissue disease characterized by the three cardinal pathological features, comprising aberrant immune activation, vasculopathy and tissue fibrosis, with unknown etiology. Although many inducible and genetic animal models mimicking the selected aspects of SSc have been well documented, the lack of models encompassing the full clinical manifestations hindered the development and preclinical testing of therapies against this disease. Under this situation, three new genetic animal models have recently been established, such as Fra2 transgenic mice, urokinase-type plasminogen activator receptor deficient mice and Klf5(+/-) ;Fli1(+/-) mice, all of which recapitulate the pathological cascade of SSc. The former two murine models demonstrate endothelial cell apoptosis and capillary loss followed by tissue fibrosis, whereas the immune systems show no remarkable abnormality. Klf5(+/-) ;Fli1(+/-) mice develop immune activation, vasculopathy and tissue fibrosis in this sequence, eventually resulting in the development of dermal fibrosis, interstitial lung disease and pulmonary vascular involvement resembling those of SSc. Because Krueppel-like factor (KLF)5 and Friend leukemia integration 1 transcription factor (Fli1) are the transcription factors epigenetically suppressed in SSc dermal fibroblasts, the reproduction of SSc manifestations in Klf5(+/-) ;Fli1(+/-) mice supports the canonical idea that environmental influences play a central role in the development of SSc in genetically predisposed individuals. These new animal models offer important clues for the better understanding of the underlying molecular mechanisms of SSc pathology and the identification of potential molecular targets for the treatment of this incurable disease.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
21
|
Lou N, Lennard Richard ML, Yu J, Kindy M, Zhang XK. The Fli-1 transcription factor is a critical regulator for controlling the expression of chemokine C-X-C motif ligand 2 (CXCL2). Mol Immunol 2016; 81:59-66. [PMID: 27889620 DOI: 10.1016/j.molimm.2016.11.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/19/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Abstract
Mammalian cells produce inflammatory cytokines and chemokines in response to innate immune signals and their expression is tightly regulated. Chemokine (C-X-C motif) ligand 2 (CXCL2), also known as macrophage inflammatory protein 2-alpha (MIP2-alpha), is an inflammatory chemokine belonging to the CXC chemokine family. CXCL2 is chemotactic for neutrophils and elevated expression of CXCL2 is associated with many inflammatory and autoimmune diseases. The Fli-1 gene belongs to the large Ets transcription factor family, whose members regulate a wide variety of cellular functions including the immune response. In this study, we demonstrate that endothelial cells transfected with Fli-1 specific siRNA produce significantly less CXCL2 compared to cells transfected with control siRNA after stimulation by the Toll-like receptor (TLR) 4 ligands, lipopolysaccharide (LPS) and tumor necrosis factor alpha (TNF-α). The production of CXCL2 in endothelial cells stimulated with LPS stimulation is dose-dependent. We found that Fli-1 binds to the CXCL2 promoter as established by Chromatin immunoprecipitation (ChIP) assay. Transient transfection assays show that Fli-1 drives transcription from the CXCL2 promoter in a dose-dependent manner and Fli-1 regulates the expression of CXCL2 largely by directly binding to the promoter. Targeted knockdown and transient transfection experiments suggest that both Fli-1 and the p65 subunit of NF-κB affect the activation of CXCL2 in an additive manner. These results indicate that Fli-1 is a novel, critical transcription factor that regulates the expression of the inflammatory chemokine CXCL2.
Collapse
Affiliation(s)
- Ning Lou
- Jinan Central Hospital, Shandong University, Jinan, Shangdong, China; Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Mara L Lennard Richard
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Jin Yu
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Mark Kindy
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, United States
| | - Xian K Zhang
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, United States.
| |
Collapse
|
22
|
Haller ST, Yan Y, Drummond CA, Xie J, Tian J, Kennedy DJ, Shilova VY, Xie Z, Liu J, Cooper CJ, Malhotra D, Shapiro JI, Fedorova OV, Bagrov AY. Rapamycin Attenuates Cardiac Fibrosis in Experimental Uremic Cardiomyopathy by Reducing Marinobufagenin Levels and Inhibiting Downstream Pro-Fibrotic Signaling. J Am Heart Assoc 2016; 5:JAHA.116.004106. [PMID: 27694325 PMCID: PMC5121507 DOI: 10.1161/jaha.116.004106] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Experimental uremic cardiomyopathy causes cardiac fibrosis and is causally related to the increased circulating levels of the cardiotonic steroid, marinobufagenin (MBG), which signals through Na/K-ATPase. Rapamycin is an inhibitor of the serine/threonine kinase mammalian target of rapamycin (mTOR) implicated in the progression of many different forms of renal disease. Given that Na/K-ATPase signaling is known to stimulate the mTOR system, we speculated that the ameliorative effects of rapamycin might influence this pathway. METHODS AND RESULTS Biosynthesis of MBG by cultured human JEG-3 cells is initiated by CYP27A1, which is also a target for rapamycin. It was demonstrated that 1 μmol/L of rapamycin inhibited production of MBG in human JEG-2 cells. Male Sprague-Dawley rats were subjected to either partial nephrectomy (PNx), infusion of MBG, and/or infusion of rapamycin through osmotic minipumps. PNx animals showed marked increase in plasma MBG levels (1025±60 vs 377±53 pmol/L; P<0.01), systolic blood pressure (169±1 vs 111±1 mm Hg; P<0.01), and cardiac fibrosis compared to controls. Plasma MBG levels were significantly decreased in PNx-rapamycin animals compared to PNx (373±46 vs 1025±60 pmol/L; P<0.01), and cardiac fibrosis was substantially attenuated by rapamycin treatment. CONCLUSIONS Rapamycin treatment in combination with MBG infusion significantly attenuated cardiac fibrosis. Our results suggest that rapamycin may have a dual effect on cardiac fibrosis through (1) mTOR inhibition and (2) inhibiting MBG-mediated profibrotic signaling and provide support for beneficial effect of a novel therapy for uremic cardiomyopathy.
Collapse
Affiliation(s)
- Steven T Haller
- University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Yanling Yan
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | | | - Joe Xie
- University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Jiang Tian
- University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - David J Kennedy
- University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Victoria Y Shilova
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD
| | - Zijian Xie
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | | | - Deepak Malhotra
- University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV
| | - Olga V Fedorova
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD
| | - Alexei Y Bagrov
- Laboratory of Cardiovascular Science, National Institute on Aging, Baltimore, MD
| |
Collapse
|
23
|
Lennard Richard ML, Brandon D, Lou N, Sato S, Caldwell T, Nowling TK, Gilkeson G, Zhang XK. Acetylation impacts Fli-1-driven regulation of granulocyte colony stimulating factor. Eur J Immunol 2016; 46:2322-2332. [PMID: 27431361 DOI: 10.1002/eji.201646315] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/22/2016] [Accepted: 07/15/2016] [Indexed: 11/07/2022]
Abstract
Fli-1 has emerged as a critical regulator of inflammatory mediators, including MCP-1, CCL5, and IL-6. The cytokine, granulocyte colony stimulating factor (G-CSF) regulates neutrophil precursor maturation and survival, and activates mature neutrophils. Previously, a significant decrease in neutrophil infiltration into the kidneys of Fli-1+/- lupus-prone mice was observed. In this study, a significant decrease in G-CSF protein expression was detected in stimulated murine and human endothelial cells when expression of Fli-1 was inhibited. The murine G-CSF promoter contains numerous putative Fli-1 binding sites and several regions within the proximal promoter are significantly enriched for Fli-1 binding. Transient transfection assays indicate that Fli-1 drives transcription from the G-CSF promoter and mutation of the Fli-1 DNA binding domain resulted in a 94% loss of transcriptional activation. Mutation of a known acetylation site, led to a significant increase in G-CSF promoter activation. The histone acetyltransferases p300/CBP and p300/CBP associated factor (PCAF) significantly decrease Fli-1 specific activation of the G-CSF promoter. Thus, acetylation appears to be an important mechanism behind Fli-1 driven activation of the G-CSF promoter. These results further support the theory that Fli-1 plays a major role in the regulation of several inflammatory mediators, ultimately affecting inflammatory disease pathogenesis.
Collapse
Affiliation(s)
- Mara L Lennard Richard
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Danielle Brandon
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Ning Lou
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA.,Jinan Central Hospital, Shandong University, Jinan, Shangdong, China
| | - Shuzo Sato
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Tomika Caldwell
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Gary Gilkeson
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA.,Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA
| | - Xian K Zhang
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, USA. .,Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
24
|
Han R, Pacifici M, Iwamoto M, Trojanowska M. Endothelial Erg expression is required for embryogenesis and vascular integrity. Organogenesis 2016; 11:75-86. [PMID: 26061019 DOI: 10.1080/15476278.2015.1031435] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Members of the ETS family of transcription factors are involved in several developmental processes including endothelial cell specification and blood vessel formation, but their exact roles remain unclear. The family member Erg is highly expressed in endothelial cells as compared to other developing cell types including chondrocytes, hematopoietic cells and mesodermal cells. To study the specific roles ERG plays in endothelial cell specification and function during early embryogenesis, we conditionally ablated it by mating ErgloxP/loxP and Tie2-Cre mice. We found that mutant embryos died by mid-gestation and that angiogenesis and vascular integrity were highly compromised. Our study reveals that ERG has essential and cell autonomous roles in endothelial cell development and blood vessel maintenance.
Collapse
Affiliation(s)
- Rong Han
- a Arthritis Center; Boston University School of Medicine ; Boston , MA USA
| | | | | | | |
Collapse
|
25
|
Marinobufagenin-induced vascular fibrosis is a likely target for mineralocorticoid antagonists. J Hypertens 2016; 33:1602-10. [PMID: 26136067 DOI: 10.1097/hjh.0000000000000591] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Endogenous cardiotonic steroids, including marinobufagenin (MBG), stimulate vascular synthesis of collagen. Because mineralocorticoid antagonists competitively antagonize effect of cardiotonic steroids on the Na/K-ATPase, we hypothesized that spironolactone would reverse the profibrotic effects of MBG. METHODS Experiment 1: Explants of thoracic aortae and aortic vascular smooth muscle cells from Wistar rats were cultured for 24 h in the presence of vehicle or MBG (100 nmol/l) with or without canrenone (10 μmol/l), an active metabolite of spironolactone. Experiment 2: In 16 patients (56 ± 2 years) with resistant hypertension on a combined (lisinopril/amlodipine/hydrochlorothiazide) therapy, we determined arterial pressure, pulse wave velocity, plasma MBG, and erythrocyte Na/K-ATPase before and 6 months after addition of placebo (n = 8) or spironolactone (50 mg/day; n = 8) to the therapy. RESULTS In rat aortic explants and in vascular smooth muscle cells, pretreatment with MBG resulted in a two-fold rise in collagen-1, and a marked reduction in the sensitivity of the aortic rings to the vasorelaxant effect of sodium nitroprusside following endothelin-1-induced constriction (EC50 = 480 ± 67 vs. 23 ± 3 nmol/l in vehicle-treated rings; P < 0.01). Canrenone blocked effects of MBG on collagen synthesis and restored sensitivity of vascular rings to sodium nitroprusside (EC50 = 17 ± 1 nmol/l). Resistant hypertension patients exhibited elevated plasma MBG (0.42 ± 0.07 vs. 0.24 ± 0.03 nmol/l; P = 0.01) and reduced Na/K-ATPase activity (1.9 ± 0.15 vs. 2.8 ± 0.2 μmol Pi/ml per h, P < 0.01) vs. seven healthy individuals. Six-month administration of spironolactone, unlike placebo treatment, was associated with a decrease in pulse wave velocity and arterial pressure, and with restoration of Na/K-ATPase activity in the presence of unchanged MBG levels. CONCLUSION MBG-induced vascular fibrosis is a likely target for spironolactone.
Collapse
|
26
|
Jinnin M. A possible mechanism of hypercoagulation status in scleroderma. Br J Dermatol 2016; 174:263. [PMID: 26871920 DOI: 10.1111/bjd.14254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- M Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
27
|
Aslani S, Mahmoudi M, Karami J, Jamshidi AR, Malekshahi Z, Nicknam MH. Epigenetic alterations underlying autoimmune diseases. Autoimmunity 2016; 49:69-83. [DOI: 10.3109/08916934.2015.1134511] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
28
|
Tsujino K, Sheppard D. Critical Appraisal of the Utility and Limitations of Animal Models of Scleroderma. Curr Rheumatol Rep 2015; 18:4. [DOI: 10.1007/s11926-015-0553-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
29
|
Abstract
PURPOSE OF REVIEW In this review of the literature from 2014 through mid-2015, we examine new data that shed light on how macrophages and other innate immune cells and signals contribute to inflammation, vascular dysfunction, and fibrosis in scleroderma. RECENT FINDINGS Recent human studies have focused on changes early in scleroderma, and linked macrophages to inflammation in skin and progression of lung disease. Plasmacytoid dendritic cells have been implicated in vascular dysfunction. In mice, several factors have been identified that influence macrophage activation and experimental fibrosis. However, emerging data also suggest that myeloid cells can have differential effects in fibrosis. Sustained signaling through different toll-like receptors can lead to inflammation or fibrosis, and these signals can influence both immune and nonimmune cells. SUMMARY There are many types of innate immune cells that can potentially contribute to scleroderma and will be worth exploring in detail. Experimentally dissecting the roles of macrophages based on ontogeny and activation state, and the innate signaling pathways in the tissue microenvironment, may also lead to better understanding of scleroderma pathogenesis.
Collapse
Affiliation(s)
- Jennifer J Chia
- aWeill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program bImmunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences cAutoimmunity and Inflammation Program dAutoimmunity and Inflammation Program and Department of Pediatric Rheumatology, Hospital for Special Surgery eDepartment of Microbiology and Immunology, Weill Cornell Medical College, New York, USA
| | | |
Collapse
|
30
|
Asano Y. [A Uuifying hypothesis for the pathogenesis of systemic sclerosis based on the deficiency of transcription factor Fli1 - the development of a new animal model of systemic sclerosis -]. ACTA ACUST UNITED AC 2015; 37:475-87. [PMID: 25748132 DOI: 10.2177/jsci.37.475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem connective tissue disease featured by immune abnormalities, vasculopathy and resultant fibrosis of the skin and various internal organs. Although the pathogenesis of SSc remains incompletely elucidated, it is currently accepted that this disease is caused by the complex interplay between hereditary and environmental factors. The deficiency of transcription factor Fli1, which is epigenetically suppressed in SSc dermal fibroblasts, potentially causes SSc-like phenotypical alteration in various cell types such as fibroblasts, endothelial cells, and macrophages, suggesting that Fli1 is a predisposing factor of SSc. KLF5 is another transcription factor which is suppressed in SSc dermal fibroblasts through an epigenetic mechanism. Importantly, double heterozygous mice for Fli1 and KLF5 develop three cardinal features of SSc, including immune abnormalities, vasculopathy and fibrosis. Therefore, these two transcription factors are likely to be critical predisposing factors regulating the development of SSc. Given that potential disease modifying drugs, bosentan and imatinib, reverse the expression and transcriptional activity of Fli1, the studies on the pathological process of double heterozygous mice and the impact of these transcription factors on various cell types may provide a new clue to further understand the pathogenesis of SSc leading to the development of new therapies.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine
| |
Collapse
|
31
|
Epigenetic suppression of Fli1, a potential predisposing factor in the pathogenesis of systemic sclerosis. Int J Biochem Cell Biol 2015; 67:86-91. [DOI: 10.1016/j.biocel.2015.06.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 06/01/2015] [Accepted: 06/02/2015] [Indexed: 11/24/2022]
|
32
|
Rao VH, Rai V, Stoupa S, Agrawal DK. Blockade of Ets-1 attenuates epidermal growth factor-dependent collagen loss in human carotid plaque smooth muscle cells. Am J Physiol Heart Circ Physiol 2015; 309:H1075-86. [PMID: 26254334 DOI: 10.1152/ajpheart.00378.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/27/2015] [Indexed: 12/27/2022]
Abstract
Although degradation of extracellular matrix by matrix metalloproteinases (MMPs) is thought to be involved in symptomatic (S) carotid plaques in atherosclerosis, the mechanisms of MMP expression are poorly understood. Here, we demonstrate that collagen loss in vascular smooth vessel cells (VSMCs) isolated from S plaques was induced by epidermal growth factor (EGF) through the activation of p38-MAPK and JNK-MAPK pathways. Inhibitors of p38-MAPK and JNK-MAPK signaling pathways downregulated the expression of MMP-1 and MMP-9. In addition, we examined whether v-ets erythroblastosis virus E26 oncogene homologue 1 (Ets-1), an important regulator of different genes, is involved in destabilizing S plaques in patients with carotid stenosis. We demonstrate that EGF induces Ets-1 expression and decreases interstitial and basement membrane collagen in vascular smooth muscle cells (VSMCs) from patients with carotid stenosis. Increased expression of MMP-1 and -9 and decreased collagen mRNA transcripts were also found in Ets-1-overexpressed VSMCs. Transfection with both dominant-negative form of Ets-1 and small interfering RNA blocked EGF-induced MMP-1 and -9 expressions and increased the mRNA transcripts for collagen I (α1) and collagen III (α1) in S compared with asymptomatic (AS) carotid plaques. Inhibitors of p38-MAPK (SB202190) and JNK-MAPK (SP600125) signaling pathways decreased the expression of Ets-1, MMP-1, and MMP-9 and increased collagen type I and III expression in EGF-treated VSMCs. This study provides a mechanistic insight into the role of Ets-1 in the plaque destabilization in patients with carotid stenosis involving p38-MAPK and JNK signaling pathways.
Collapse
Affiliation(s)
- Velidi H Rao
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - Vikrant Rai
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - Samantha Stoupa
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| | - Devendra K Agrawal
- Center for Clinical and Translational Science, Creighton University School of Medicine, Omaha, Nebraska
| |
Collapse
|
33
|
De Langhe E, Lories R. Fibrogenesis, novel lessons from animal models. Semin Immunopathol 2015; 37:565-74. [PMID: 26141608 DOI: 10.1007/s00281-015-0510-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 06/16/2015] [Indexed: 12/15/2022]
Abstract
Systemic sclerosis (SSc) is a devastating chronic autoimmune connective tissue disease characterized by vasculopathy, autoimmunity with inflammation, and progressive fibrogenesis. The current paradigm of the pathogenesis of SSc is that of an unknown initial trigger, leading to a complex interaction of immune cells, endothelial cells, and fibroblasts, producing cytokines, growth and angiogenic factors, and resulting in uncontrolled and persistent tissue fibrogenesis by an altered mesenchymal cell compartment. Animal models are of utmost importance to investigate the different steps in the pathogenesis. This review will elaborate on recent findings in established and more recently developed animal models, presenting data on compounds that are in or ready to be translated into clinical trials, or provide interesting new findings in the understanding of the pathophysiology of SSc. We focus on recent findings concerning the vessel-extracellular matrix interaction, the initial triggering aggressor, the concept of autoimmunity and inflammatory changes, the effector cells and their origins, and the complex interaction of the different signaling pathways in fibrogenesis.
Collapse
Affiliation(s)
- Ellen De Langhe
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Department of Development and Regeneration, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | | |
Collapse
|
34
|
Taniguchi T, Asano Y, Akamata K, Noda S, Takahashi T, Ichimura Y, Toyama T, Trojanowska M, Sato S. Fibrosis, vascular activation, and immune abnormalities resembling systemic sclerosis in bleomycin-treated Fli-1-haploinsufficient mice. Arthritis Rheumatol 2015; 67:517-26. [PMID: 25385187 DOI: 10.1002/art.38948] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 11/04/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Fli-1, a potential predisposing factor for systemic sclerosis (SSc), is constitutively down-regulated in the lesional skin of patients with SSc by an epigenetic mechanism. To investigate the impact of Fli-1 deficiency on the induction of an SSc phenotype in various cell types, we generated bleomycin-induced skin fibrosis in Fli-1(+/-) mice and investigated the molecular mechanisms underlying its phenotypic alterations. METHODS Messenger RNA (mRNA) levels and protein expression of target molecules were examined by quantitative reverse transcription-polymerase chain reaction and immunostaining. Transforming growth factor β (TGFβ) bioassay was used to evaluate the activation of latent TGFβ. The binding of Fli-1 to the target gene promoters was assessed with chromatin immunoprecipitation. RESULTS Bleomycin induced more severe dermal fibrosis in Fli-1(+/-) mice than in wild-type mice. Fli-1 haploinsufficiency activated dermal fibroblasts via the up-regulation of αvβ3 and αvβ5 integrins and activation of latent TGFβ. Dermal fibrosis in Fli-1(+/-) mice was also attributable to endothelial-to-mesenchymal transition, which is directly induced by Fli-1 deficiency and amplified by bleomycin. Th2/Th17-skewed inflammation and increased infiltration of mast cells and macrophages were seen, partly due to the altered expression of cell adhesion molecules in endothelial cells as well as the induction of the skin chemokines. Fli-1(+/-) mouse macrophages preferentially differentiated into an M2 phenotype upon stimulation with interleukin-4 (IL-4) or IL-13. CONCLUSION Our findings provide strong evidence for the fundamental role of Fli-1 deficiency in inducing SSc-like phenotypic alterations in dermal fibroblasts, endothelial cells, and macrophages in a manner consistent with human disease.
Collapse
|
35
|
Sato S, Lennard Richard M, Brandon D, Jones Buie JN, Oates JC, Gilkeson GS, Zhang XK. A critical role of the transcription factor fli-1 in murine lupus development by regulation of interleukin-6 expression. Arthritis Rheumatol 2015; 66:3436-44. [PMID: 25155007 DOI: 10.1002/art.38818] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 08/05/2014] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The Fli-1 transcription factor is implicated in the pathogenesis of systemic lupus erythematosus (SLE), both in humans and in animal models. Dysregulation of interleukin-6 (IL-6) is also associated with SLE. The purpose of this study was to investigate whether Fli-1 directly regulates the expression of IL-6. METHODS Sera were collected from wild-type and Fli-1-heterozygous (Fli-1(+/-) ) MRL/lpr mice, and the concentration of IL-6 was measured by enzyme-linked immunosorbent assay (ELISA). Expression of IL-6 in the kidney was measured by real-time polymerase chain reaction analysis. T cells were isolated from wild-type and Fli-1(+/-) MRL/lpr mice and stimulated with CD3/CD28 beads, and the concentration of IL-6 in the supernatants was measured by ELISA. MS1 endothelial cells were transfected with Fli-1 and control small interfering RNA, and the production of IL-6 was compared after lipopolysaccharide stimulation. A chromatin immunoprecipitation (ChIP) assay was performed to determine whether Fli-1 binds to the IL-6 promoter region. Transient transfections with the NIH3T3 cell line were performed to examine whether Fli-1 regulates the expression of IL-6. RESULTS Fli-1(+/-) MRL/lpr mice had significantly decreased IL-6 levels in sera and reduced expression of IL-6 in kidneys as compared to their wild-type littermates. T cells isolated from Fli-1(+/-) MRL/lpr mice produced less IL-6 than did those from wild-type mice. Inhibiting the expression of Fli-1 in endothelial cells resulted in reduced production of IL-6. The ChIP assay revealed direct binding of Fli-1 to 3 regions within the IL-6 promoter. Fli-1 activated transcription from the IL-6 promoter in a dose-dependent manner. CONCLUSION The direct regulation of IL-6 expression by Fli-1 represents one possible mechanism for the protective effect of decreased Fli-1 expression in lupus.
Collapse
Affiliation(s)
- Shuzo Sato
- Medical University of South Carolina, Charleston
| | | | | | | | | | | | | |
Collapse
|
36
|
Beishline K, Azizkhan-Clifford J. Sp1 and the 'hallmarks of cancer'. FEBS J 2015; 282:224-58. [PMID: 25393971 DOI: 10.1111/febs.13148] [Citation(s) in RCA: 396] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 09/26/2014] [Accepted: 11/10/2014] [Indexed: 12/19/2022]
Abstract
For many years, transcription factor Sp1 was viewed as a basal transcription factor and relegated to a role in the regulation of so-called housekeeping genes. Identification of Sp1's role in recruiting the general transcription machinery in the absence of a TATA box increased its importance in gene regulation, particularly in light of recent estimates that the majority of mammalian genes lack a TATA box. In this review, we briefly consider the history of Sp1, the founding member of the Sp family of transcription factors. We review the evidence suggesting that Sp1 is highly regulated by post-translational modifications that positively and negatively affect the activity of Sp1 on a wide array of genes. Sp1 is over-expressed in many cancers and is associated with poor prognosis. Targeting Sp1 in cancer treatment has been suggested; however, our review of the literature on the role of Sp1 in the regulation of genes that contribute to the 'hallmarks of cancer' illustrates the extreme complexity of Sp1 functions. Sp1 both activates and suppresses the expression of a number of essential oncogenes and tumor suppressors, as well as genes involved in essential cellular functions, including proliferation, differentiation, the DNA damage response, apoptosis, senescence and angiogenesis. Sp1 is also implicated in inflammation and genomic instability, as well as epigenetic silencing. Given the apparently opposing effects of Sp1, a more complete understanding of the function of Sp1 in cancer is required to validate its potential as a therapeutic target.
Collapse
Affiliation(s)
- Kate Beishline
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
37
|
Chan ESL, Liu H, Fernandez P, Luna A, Perez-Aso M, Bujor AM, Trojanowska M, Cronstein BN. Adenosine A(2A) receptors promote collagen production by a Fli1- and CTGF-mediated mechanism. Arthritis Res Ther 2014; 15:R58. [PMID: 23663495 PMCID: PMC4060252 DOI: 10.1186/ar4229] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 02/25/2013] [Accepted: 05/11/2013] [Indexed: 12/13/2022] Open
Abstract
Introduction Adenosine, acting through the A2A receptor, promotes tissue matrix production in the skin and the liver and induces the development of dermal fibrosis and cirrhosis in murine models. Since expression of A2A receptors is increased in scleroderma fibroblasts, we examined the mechanisms by which the A2A receptor produces its fibrogenic effects. Methods The effects of A2A receptor ligation on the expression of the transcription factor, Fli1, a constitutive repressor for the synthesis of matrix proteins, such as collagen, is studied in dermal fibroblasts. Fli1 is also known to repress the transcription of CTGF/CCN2, and the effects of A2A receptor stimulation on CTGF and TGF-β1 expression are also examined. Results A2A receptor occupancy suppresses the expression of Fli1 by dermal fibroblasts. A2A receptor activation induces the secretion of CTGF by dermal fibroblasts, and neutralization of CTGF abrogates the A2A receptor-mediated enhancement of collagen type I production. A2AR activation, however, resulted in a decrease in TGF-β1 protein release. Conclusions Our results suggest that Fli1 and CTGF are important mediators of the fibrogenic actions of adenosine and the use of small molecules such as adenosine A2A receptor antagonists may be useful in the therapy of dermal fibrosis in diseases such as scleroderma.
Collapse
|
38
|
Scheiber MN, Watson PM, Rumboldt T, Stanley C, Wilson RC, Findlay VJ, Anderson PE, Watson DK. FLI1 expression is correlated with breast cancer cellular growth, migration, and invasion and altered gene expression. Neoplasia 2014; 16:801-13. [PMID: 25379017 PMCID: PMC4212256 DOI: 10.1016/j.neo.2014.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 08/15/2014] [Indexed: 12/21/2022] Open
Abstract
ETS factors have been shown to be dysregulated in breast cancer. ETS factors control the expression of genes involved in many biological processes, such as cellular proliferation, differentiation, and apoptosis. FLI1 is an ETS protein aberrantly expressed in retrovirus-induced hematological tumors, but limited attention has been directed towards elucidating the role of FLI1 in epithelial-derived cancers. Using data mining, we show that loss of FLI1 expression is associated with shorter survival and more aggressive phenotypes of breast cancer. Gain and loss of function cellular studies indicate the inhibitory effect of FLI1 expression on cellular growth, migration, and invasion. Using Fli1 mutant mice and both a transgenic murine breast cancer model and an orthotopic injection of syngeneic tumor cells indicates that reduced Fli1 contributes to accelerated tumor growth. Global expression analysis and RNA-Seq data from an invasive human breast cancer cell line with over expression of either FLI1 and another ETS gene, PDEF, shows changes in several cellular pathways associated with cancer, such as the cytokine-cytokine receptor interaction and PI3K-Akt signaling pathways. This study demonstrates a novel role for FLI1 in epithelial cells. In addition, these results reveal that FLI1 down-regulation in breast cancer may promote tumor progression.
Collapse
Key Words
- Ad-FLI1, Ad-GFP-FLI1
- EMT, Epithelial-mesenchymal transition
- ER, Estrogen receptor
- FLI1, Friend leukemia virus integration 1
- GAPDH, Glyceraldehyde-3-phosphate dehydrogenase
- GEO, Gene Expression Omnibus
- GOBO, Gene expression-based Outcome for Breast cancer Online
- IDC, Invasive ductal carcinoma
- IHC, Immunohistochemistry
- ILC, Invasive lobular carcinoma
- N, Normal Breast Tissue
- PDEF, Prostate-derived ETS factor
- PyVT, FVB/N-Tg(MMTV-PyVT)634Mul/J
- Rb, Retinoblastoma
- T, Tumor
- uPA, Urokinase plasminogen activator
Collapse
Affiliation(s)
- Melissa N Scheiber
- Department of Pathology and Laboratory Medicine, The James E. Clyburn Research Center, Medical University of South Carolina, 68 President Street, Charleston, SC 29425
| | - Patricia M Watson
- Department of Medicine, Division of Hematology/Oncology, The James E. Clyburn Research Center, Medical University of South Carolina, 68 President Street, Charleston, SC 29425
| | - Tihana Rumboldt
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Children's Hospital, 171 Ashley Avenue, Charleston, SC 29425
| | - Connor Stanley
- Department of Computer Science, College of Charleston, Charleston, SC 29424
| | - Robert C Wilson
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, The James E. Clyburn Research Center, Medical University of South Carolina, 68 President Street, Charleston, SC 29425
| | - Victoria J Findlay
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Walton Research Building, 39 Sabin Street, Charleston, SC 29425
| | - Paul E Anderson
- Department of Computer Science, College of Charleston, Charleston, SC 29424
| | - Dennis K Watson
- Department of Pathology and Laboratory Medicine, The James E. Clyburn Research Center, Medical University of South Carolina, 68 President Street, Charleston, SC 29425
| |
Collapse
|
39
|
Abstract
The skin is composed of a variety of cell types expressing specific molecules and possessing different properties that facilitate the complex interactions and intercellular communication essential for maintaining the structural integrity of the skin. Importantly, a single mutation in one of these molecules can disrupt the entire organization and function of these essential networks, leading to cell separation, blistering, and other striking phenotypes observed in inherited skin diseases. Over the past several decades, the genetic basis of many monogenic skin diseases has been elucidated using classical genetic techniques. Importantly, the findings from these studies has shed light onto the many classes of molecules and essential genetic as well as molecular interactions that lend the skin its rigid, yet flexible properties. With the advent of the human genome project, next-generation sequencing techniques, as well as several other recently developed methods, tremendous progress has been made in dissecting the genetic architecture of complex, non-Mendelian skin diseases.
Collapse
Affiliation(s)
- Gina M DeStefano
- Department of Genetics and Development, Columbia University, New York, New York 10032
| | - Angela M Christiano
- Department of Genetics and Development, Columbia University, New York, New York 10032 Department of Dermatology, Columbia University, New York, New York 10032
| |
Collapse
|
40
|
Kudo H, Jinnin M, Asano Y, Trojanowska M, Nakayama W, Inoue K, Honda N, Kajihara I, Makino K, Fukushima S, Ihn H. Decreased interleukin-20 expression in scleroderma skin contributes to cutaneous fibrosis. Arthritis Rheumatol 2014; 66:1636-47. [PMID: 24470401 DOI: 10.1002/art.38380] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Accepted: 01/21/2014] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To clarify the role of interleukin-20 (IL-20) in the regulatory mechanism of extracellular matrix expression and to determine the contribution of IL-20 to the phenotype of systemic sclerosis (SSc). METHODS Protein and messenger RNA (mRNA) levels of collagen, Fli-1, IL-20, and IL-20 receptor (IL-20R) were analyzed using polymerase chain reaction (PCR) array, immunoblotting, immunohistochemical staining, enzyme-linked immunosorbent assay, and real-time PCR. RESULTS PCR array revealed that IL-20 decreased gene expression of α2(I) collagen (0.03-fold), Smad3 (0.02-fold), and endoglin (0.05-fold) in cultured normal dermal fibroblasts. Fli-1 protein expression was induced by IL-20 (~2-fold). The inhibition of collagen by IL-20, the induction of Fli-1 by IL-20, and the reduction of Smad3 and endoglin by IL-20 were also observed in SSc fibroblasts. Serum IL-20 levels were reduced only slightly in SSc patients but were significantly decreased in patients with scleroderma spectrum disorders (the prodromal stage of SSc) compared with those in normal subjects (111.3 pg/ml versus 180.4 pg/ml; P < 0.05). On the other hand, IL-20 mRNA expression in SSc skin was decreased compared with that in normal skin (P < 0.05), which may result in the induction of collagen synthesis in SSc dermal fibroblasts. IL-20R was expressed in normal and SSc fibroblasts. Moreover, IL-20 supplementation by injection into the skin reversed skin fibrosis induced by bleomycin in mice (~0.5-fold). CONCLUSION IL-20 reduces basal collagen transcription via Fli-1 induction, while down-regulation of Smad3 and endoglin may cancel the effect of transforming growth factor β in SSc fibroblasts. To confirm the therapeutic value of IL-20 and IL-20R, their function and expression in vivo should be further studied.
Collapse
|
41
|
Lennard Richard ML, Nowling TK, Brandon D, Watson DK, Zhang XK. Fli-1 controls transcription from the MCP-1 gene promoter, which may provide a novel mechanism for chemokine and cytokine activation. Mol Immunol 2014; 63:566-73. [PMID: 25108845 DOI: 10.1016/j.molimm.2014.07.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/29/2014] [Accepted: 07/10/2014] [Indexed: 12/14/2022]
Abstract
Regulation of proinflammatory cytokines and chemokines is a primary role of the innate immune response. MCP-1 is a chemokine that recruits immune cells to sites of inflammation. Expression of MCP-1 is reduced in primary kidney endothelial cells from mice with a heterozygous knockout of the Fli-1 transcription factor. Fli-1 is a member of the Ets family of transcription factors, which are evolutionarily conserved across several organisms including Drosophilla, Xenopus, mouse and human. Ets family members bind DNA through a consensus sequence GGAA/T, or Ets binding site (EBS). Fli-1 binds to EBSs within the endogenous MCP-1 promoter by ChIP assay. In this study, transient transfection assays indicate that the Fli-1 gene actively promotes transcription from the MCP-1 gene promoter in a dose-dependent manner. Mutation of the DNA binding domain of Fli-1 demonstrated that Fli-1 activates transcription of MCP-1 both directly, by binding to the promoter, and indirectly, likely through interactions with other transcription factors. Another Ets transcription factor, Ets-1, was also tested, but failed to promote transcription. While Ets-1 failed to drive transcription independently, a weak synergistic activation of the MCP-1 promoter was observed between Ets-1 and Fli-1. In addition, Fli-1 and the NFκB family member p65 were found to interact synergistically to activate transcription from the MCP-1 promoter, while Sp1 and p50 inhibit this interaction. Deletion studies identified that EBSs in the distal and proximal MCP-1 promoter are critical for Fli-1 activation from the MCP-1 promoter. Together, these results demonstrate that Fli-1 is a novel regulator of the proinflammatory chemokine MCP-1, that interacts with other transcription factors to form a complex transcriptional mechanism for the activation of MCP-1 and mediation of the inflammatory response.
Collapse
Affiliation(s)
- Mara L Lennard Richard
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Tamara K Nowling
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA; Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA
| | - Danielle Brandon
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Dennis K Watson
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA; Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Xian K Zhang
- Department of Medicine, Division of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC 29425, USA; Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403, USA.
| |
Collapse
|
42
|
Lennard Richard ML, Sato S, Suzuki E, Williams S, Nowling TK, Zhang XK. The Fli-1 transcription factor regulates the expression of CCL5/RANTES. THE JOURNAL OF IMMUNOLOGY 2014; 193:2661-8. [PMID: 25098295 DOI: 10.4049/jimmunol.1302779] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The friend leukemia insertion site 1 (Fli-1) transcription factor, an Ets family member, is implicated in the pathogenesis of systemic lupus erythematosus in human patients and murine models of lupus. Lupus-prone mice with reduced Fli-1 expression have significantly less nephritis, prolonged survival, and decreased infiltrating inflammatory cells into the kidney. Inflammatory chemokines, including CCL5, are critical for attracting inflammatory cells. In this study, decreased CCL5 mRNA expression was observed in kidneys of lupus-prone NZM2410 mice with reduced Fli-1 expression. CCL5 protein expression was significantly decreased in endothelial cells transfected with Fli-1-specific small interfering RNA compared with controls. Fli-1 binds to endogenous Ets binding sites in the distal region of the CCL5 promoter. Transient transfection assays demonstrate that Fli-1 drives transcription from the CCL5 promoter in a dose-dependent manner. Both Ets1, another Ets family member, and Fli-1 drive transcription from the CCL5 promoter, although Fli-1 transactivation was significantly stronger. Ets1 acts as a dominant-negative transcription factor for Fli-1, indicating that they may have at least one DNA binding site in common. Systematic deletion of DNA binding sites demonstrates the importance of the sites located within a 225-bp region of the promoter. Mutation of the Fli-1 DNA binding domain significantly reduces transactivation of the CCL5 promoter by Fli-1. We identified a novel regulator of transcription for CCL5. These results suggest that Fli-1 is a novel and critical regulator of proinflammatory chemokines and affects the pathogenesis of disease through the regulation of factors that recruit inflammatory cells to sites of inflammation.
Collapse
Affiliation(s)
- Mara L Lennard Richard
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; and
| | - Shuzo Sato
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; and
| | - Eiji Suzuki
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; and
| | - Sarah Williams
- Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403
| | - Tamara K Nowling
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; and Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403
| | - Xian K Zhang
- Division of Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; and Medical Research Service, Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29403
| |
Collapse
|
43
|
Akamata K, Asano Y, Aozasa N, Noda S, Taniguchi T, Takahashi T, Ichimura Y, Toyama T, Sato S. Bosentan reverses the pro-fibrotic phenotype of systemic sclerosis dermal fibroblasts via increasing DNA binding ability of transcription factor Fli1. Arthritis Res Ther 2014; 16:R86. [PMID: 24708674 PMCID: PMC4060196 DOI: 10.1186/ar4529] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Accepted: 03/21/2014] [Indexed: 02/06/2023] Open
Abstract
Introduction Although the pathogenesis of systemic sclerosis (SSc) still remains unknown, recent studies have demonstrated that endothelins are deeply involved in the developmental process of fibrosis and vasculopathy associated with SSc, and a dual endothelin receptor antagonist, bosentan, has a potential to serve as a disease modifying drug for this disorder. Importantly, endothelin-1 (ET-1) exerts a pro-fibrotic effect on normal dermal fibroblasts and bosentan reverses the pro-fibrotic phenotype of SSc dermal fibroblasts. The purpose of this study was to clarify the details of molecular mechanisms underlying the effects of ET-1 and bosentan on dermal fibroblasts, which have not been well studied. Methods The mRNA levels of target genes and the expression and phosphorylation levels of target proteins were determined by reverse transcription real-time PCR and immunoblotting, respectively. Promoter assays were performed using a sequential deletion of human α2 (I) collagen (COL1A2) promoter. DNA affinity precipitation and chromatin immunoprecipitation were employed to evaluate the DNA binding ability of Fli1. Fli1 protein levels in murine skin were evaluated by immunostaining. Results In normal fibroblasts, ET-1 activated c-Abl and protein kinase C (PKC)-δ and induced Fli1 phosphorylation at threonine 312, leading to the decreased DNA binding of Fli1, a potent repressor of the COL1A2 gene, and the increase in type I collagen expression. On the other hand, bosentan reduced the expression of c-Abl and PKC-δ, the nuclear localization of PKC-δ, and Fli1 phosphorylation, resulting in the increased DNA binding of Fli1 and the suppression of type I collagen expression in SSc fibroblasts. In bleomycin-treated mice, bosentan prevented dermal fibrosis and increased Fli1 expression in lesional dermal fibroblasts. Conclusions ET-1 exerts a potent pro-fibrotic effect on normal fibroblasts by activating “c-Abl - PKC-δ - Fli1” pathway. Bosentan reverses the pro-fibrotic phenotype of SSc fibroblasts and prevents the development of dermal fibrosis in bleomycin-treated mice by blocking this signaling pathway. Although the efficacy of bosentan for dermal and pulmonary fibrosis is limited in SSc, the present observation definitely provides us with a useful clue to further explore the potential of the upcoming new dual endothelin receptor antagonists as disease modifying drugs for SSc.
Collapse
|
44
|
Haller ST, Drummond CA, Yan Y, Liu J, Tian J, Malhotra D, Shapiro JI. Passive immunization against marinobufagenin attenuates renal fibrosis and improves renal function in experimental renal disease. Am J Hypertens 2014; 27:603-9. [PMID: 24014658 DOI: 10.1093/ajh/hpt169] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND We have shown that the cardiotonic steroid marinobufagenin (MBG) is elevated in clinical and experimental renal disease, and significantly contributes to the development of experimental uremic cardiomyopathy induced by removal of five-sixths of the kidney (5/6 nephrectomy; PNx) in the rat. We have demonstrated that both active and passive immunization against MBG with an anti-MBG monoclonal antibody (mAb 3E9) significantly attenuated cardiac fibrosis following PNx. In the present study we sought to determine whether the use of mAb 3E9 could improve renal function following PNx. METHODS Sprague-Dawley rats were treated with either mAb 3E9 or with DigiFab (an affinity-purified anti-digoxin antibody formerly named Digibind) during the fourth week after PNx. Sham-operated animals and PNx animals treated with an IgG antibody served as controls. Plasma, urine, and renal tissue were collected at the completion of the study to determine the effects of antibody treatment on renal function. RESULTS In PNx rats, treatments with mAb 3E9 and DigiFab, respectively, significantly reduced plasma creatinine, improved creatinine clearance, and reduced proteinuria below the values of these three measures in IgG-treated PNx controls. Additionally, treatment with mAb 3E9 and DigiFab significantly reduced renal fibrosis as measured with Western blotting and Sirius red/Fast green staining. CONCLUSIONS Passive immunization against MBG significantly improved renal function and markedly reduced renal fibrosis following the experimental induction of renal disease. The work in the study reported here adds to a growing body of knowledge implicating MBG in the development of chronic renal disease. Passive immunization against cardiotonic steroids may serve as a promising treatment for chronic renal disease.
Collapse
|
45
|
Findlay VJ, LaRue AC, Turner DP, Watson PM, Watson DK. Understanding the role of ETS-mediated gene regulation in complex biological processes. Adv Cancer Res 2014; 119:1-61. [PMID: 23870508 DOI: 10.1016/b978-0-12-407190-2.00001-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ets factors are members of one of the largest families of evolutionarily conserved transcription factors, regulating critical functions in normal cell homeostasis, which when perturbed contribute to tumor progression. The well-documented alterations in ETS factor expression and function during cancer progression result in pleiotropic effects manifested by the downstream effect on their target genes. Multiple ETS factors bind to the same regulatory sites present on target genes, suggesting redundant or competitive functions. The anti- and prometastatic signatures obtained by examining specific ETS regulatory networks will significantly improve our ability to accurately predict tumor progression and advance our understanding of gene regulation in cancer. Coordination of multiple ETS gene functions also mediates interactions between tumor and stromal cells and thus contributes to the cancer phenotype. As such, these new insights may provide a novel view of the ETS gene family as well as a focal point for studying the complex biological control involved in tumor progression. One of the goals of molecular biology is to elucidate the mechanisms that contribute to the development and progression of cancer. Such an understanding of the molecular basis of cancer will provide new possibilities for: (1) earlier detection, as well as better diagnosis and staging of disease; (2) detection of minimal residual disease recurrences and evaluation of response to therapy; (3) prevention; and (4) novel treatment strategies. Increased understanding of ETS-regulated biological pathways will directly impact these areas.
Collapse
Affiliation(s)
- Victoria J Findlay
- Department of Pathology and Laboratory Medicine, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | |
Collapse
|
46
|
Xie JX, Shapiro AP, Shapiro JI. The Trade-Off between Dietary Salt and Cardiovascular Disease; A Role for Na/K-ATPase Signaling? Front Endocrinol (Lausanne) 2014; 5:97. [PMID: 25101054 PMCID: PMC4101451 DOI: 10.3389/fendo.2014.00097] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 06/07/2014] [Indexed: 12/12/2022] Open
Abstract
It has been postulated for some time that endogenous digitalis-like substances, also called cardiotonic steroids (CTS), exist, and that these substances are involved in sodium handling. Within the past 20 years, these substances have been unequivocally identified and measurements of circulating and tissue concentrations have been made. More recently, it has been identified that CTS also mediate signal transduction through the Na/K-ATPase, and consequently been implicated in profibrotic pathways. This review will discuss the mechanism of CTS in renal sodium handling and a potential "trade-off" effect from their role in inducing tissue fibrosis.
Collapse
Affiliation(s)
- Joe X. Xie
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Anna Pearl Shapiro
- Department of Medicine, University of Toledo College of Medicine, Toledo, OH, USA
| | - Joseph Isaac Shapiro
- Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
- *Correspondence: Joseph Isaac Shapiro, Department of Medicine, Joan C. Edwards School of Medicine, Marshall University, 1600 Medical Center Drive Suite 3408, Huntington, WV 25701, USA e-mail:
| |
Collapse
|
47
|
|
48
|
Ghosh AK, Quaggin SE, Vaughan DE. Molecular basis of organ fibrosis: potential therapeutic approaches. Exp Biol Med (Maywood) 2013; 238:461-81. [PMID: 23856899 DOI: 10.1177/1535370213489441] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Fibrosis, a non-physiological wound healing in multiple organs, is associated with end-stage pathological symptoms of a wide variety of vascular injury and inflammation related diseases. In response to chemical, immunological and physical insults, the body's defense system and matrix synthetic machinery respond to healing the wound and maintain tissue homeostasis. However, uncontrolled wound healing leads to scarring or fibrosis, a pathological condition characterized by excessive synthesis and accumulation of extracellular matrix proteins, loss of tissue homeostasis and organ failure. Understanding the actual cause of pathological wound healing and identification of igniter(s) of fibrogenesis would be helpful to design novel therapeutic approaches to control pathological wound healing and to prevent fibrosis related morbidity and mortality. In this article, we review the significance of a few key cytokines (TGF-β, IFN-γ, IL-10) transcriptional activators (Sp1, Egr-1, Smad3), repressors (Smad7, Fli-1, PPAR-γ, p53, Klotho) and epigenetic modulators (acetyltransferase, methyltransferases, deacetylases, microRNAs) involved in major matrix protein collagen synthesis under pathological stage of wound healing, and the potentiality of these regulators as therapeutic targets for fibrosis treatment. The significance of endothelial to mesenchymal transition (EndMT) and senescence, two newly emerged fields in fibrosis research, has also been discussed.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute & Division of Nephrology, Northwestern University, Chicago, IL, USA.
| | | | | |
Collapse
|
49
|
Ponticos M, Smith BD. Extracellular matrix synthesis in vascular disease: hypertension, and atherosclerosis. J Biomed Res 2013; 28:25-39. [PMID: 24474961 PMCID: PMC3904172 DOI: 10.7555/jbr.27.20130064] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 05/28/2013] [Accepted: 06/18/2013] [Indexed: 12/18/2022] Open
Abstract
Extracellular matrix (ECM) within the vascular network provides both a structural and regulatory role. The ECM is a dynamic composite of multiple proteins that form structures connecting cells within the network. Blood vessels are distended by blood pressure and, therefore, require ECM components with elasticity yet with enough tensile strength to resist rupture. The ECM is involved in conducting mechanical signals to cells. Most importantly, ECM regulates cellular function through chemical signaling by controlling activation and bioavailability of the growth factors. Cells respond to ECM by remodeling their microenvironment which becomes dysregulated in vascular diseases such hypertension, restenosis and atherosclerosis. This review examines the cellular and ECM components of vessels, with specific emphasis on the regulation of collagen type I and implications in vascular disease.
Collapse
Affiliation(s)
- Markella Ponticos
- Centre for Rheumatology & Connective Tissue Diseases, Division of Medicine-Inflammation, Royal Free & University College Medical School, University College London, London NW3 2PF, UK
| | - Barbara D Smith
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
50
|
Asano Y, Trojanowska M. Fli1 represses transcription of the human α2(I) collagen gene by recruitment of the HDAC1/p300 complex. PLoS One 2013; 8:e74930. [PMID: 24058639 PMCID: PMC3772867 DOI: 10.1371/journal.pone.0074930] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/07/2013] [Indexed: 12/28/2022] Open
Abstract
Fli1, a member of the Ets transcription factor family, is a key repressor of the human α2(I) collagen (COL1A2) gene. Although our previous studies have delineated that TGF-β induces displacement of Fli1 from the COL1A2 promoter through sequential post-translational modifications, the detailed mechanism by which Fli1 functions as a potent transcriptional repressor of the COL1A2 gene has not been fully investigated. To address this issue, we carried out a series of experiments especially focusing on protein-protein interaction and epigenetic transcriptional regulation. The combination of tandem affinity purification and mass spectrometry identified HDAC1 as a Fli1 interacting protein. Under quiescent conditions, HDAC1 induced deacetylation of Fli1 resulting in an increase of Fli1 DNA binding ability and p300 enhanced this process by promoting the formation of a Fli1-HDAC1-p300 complex. TGF-β-induced phosphorylation of Fli1 at threonine 312 led to disassembly of this protein complex. In quiescent dermal fibroblasts Fli1, HDAC1, and p300 occupied the −404 to −237 region, including the Fli1 binding site, of the COL1A2 promoter. TGF-β induced Fli1 and HDAC1 dissociation from the COL1A2 promoter, while promoting Ets1 and p300 recruitment. Furthermore, acetylation levels of histone H3 around the Fli1 binding site in the COL1A2 promoter inversely correlated with the DNA occupancy of Fli1 and HDAC1, while positively correlating with that of Ets1 and p300. In the functional studies, HDAC1 overexpression magnified the inhibitory effect of Fli1 on the COL1A2 promoter. Moreover, pharmacological blockade of HDAC1 by entinostat enhanced collagen production in dermal fibroblasts. Collectively, these results indicate that under quiescent conditions Fli1 recruits HDAC1/p300 to the COL1A2 promoter and suppresses the expression of the COL1A2 gene by chromatin remodeling through histone deacetylation. TGF-β-dependent phosphorylation of Fli1 at threonine 312 is a critical step regulating the remodeling of the Fli1 transcription repressor complex, leading to transcriptional activation of the COL1A2 gene.
Collapse
Affiliation(s)
- Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
- * E-mail:
| | - Maria Trojanowska
- Arthritis Center, Boston University School of Medicine, Boston, Massachusetts, United States of America
| |
Collapse
|