51
|
Deng X, Zhou X, Deng Y, Liu F, Feng X, Yin Q, Gu Y, Shi S, Xu M. Thrombin Induces CCL2 Expression in Human Lung Fibroblasts via p300 Mediated Histone Acetylation and NF-KappaB Activation. J Cell Biochem 2017; 118:4012-4019. [PMID: 28407300 DOI: 10.1002/jcb.26057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/11/2017] [Indexed: 02/05/2023]
Abstract
Thrombin has been shown to play a key role in lung diseases such as pulmonary fibrosis via the induction of fibrotic cytokine- chemokine (CC motif) ligand-2 (CCL2) expression. We previously reported that transcription factor nuclear factor-κB (NF-κB) is responsible for thrombin-induced CCL2 expression in human lung fibroblasts (HLFs). Here, we extended our study to investigate the epigenetic regulation mechanism for thrombin-induced CCL2 expression in HLFs. HLFs were cultured in F-12 medium. CCL2 protein and mRNA levels were detected by ELISA and quantitative real-time PCR, respectively. Histone, histone acetyltransferases, and NF-κB binding to CCL2 promoter were detected by ChIP assay. NF-κB activation was detected by Western blotting. We revealed that increased binding of histone acetyltransferase p300 and acetylated histone H3 and H4 to CCL2 promoter are responsible for thrombin induced CCL2 expression in HLF cells. In addition, p300 inhibition attenuates both thrombin induced-CCL2 expression and histone H3 and H4 acetylation in HLFs, suggesting that p300 is involved in thrombin-induced CCL2 expression via hyperacetylating histone H3 and H4. Our data further showed that p300 also regulates CCL2 expression via interaction with NF-κB p65, as depletion of p300 inhibits both NF-κB p65 activation and its binding to CCL2 promoter. The findings strongly suggest that epigenetic dysregulation and the interaction between histone acetyltransferase and transcription factor may be responsible for thrombin induced-CCL2 expression in HLFs. Increased understanding of the epigenetic mechanisms of CCL2 regulation may provide opportunities for identifying novel molecular targets for therapeutic purposes. J. Cell. Biochem. 118: 4012-4019, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xiaoling Deng
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Xiaoqiong Zhou
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Yan Deng
- Department of Respiratory Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, Guangdong Province, People's Republic of China
| | - Fan Liu
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Xiaofan Feng
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Qi Yin
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Yinzhen Gu
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Songlin Shi
- Department of Basic Medical Science, Xiamen University Medical College, Xiamen, 361102, Fujian Province, People's Republic of China
| | - Mingyan Xu
- Department of Oral Biology and Biomaterial, Xiamen Stomatological Research Institute, Xiamen Medical College, Xiamen, 361000, Fujian Province, People's Republic of China
| |
Collapse
|
52
|
Gasparini S, Villa F, Molfetta L, Repaci E, Castagnola P, Quarto R, Giannoni P. Exposure to reversine affects the chondrocyte morphology and phenotype in vitro. J Tissue Eng Regen Med 2017; 12:e1337-e1348. [PMID: 28714568 DOI: 10.1002/term.2515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 07/05/2017] [Accepted: 07/11/2017] [Indexed: 11/11/2022]
Abstract
Articular chondrocytes derived from osteoarthritic tissues (OA HAC) show a severely reduced chondrogenic commitment. This impairment undermines their use for tissue-engineered cartilage repair, which relies on cell proliferation and growth to meet therapeutic needs, but also on efficient cell plasticity to recover the chondrogenic phenotype. Reversine (Rev), a 2,6-disubstituted purine inhibitor of spindle-assembly checkpoints, was described to convert differentiated mesenchymal cells to their undifferentiated precursors. We hypothesized that Rev exposure could divert OA HAC to more plastic cells, re-boosting their subsequent commitment. HAC were enzymatically released from OA cartilage specimens, expanded for 2 weeks and treated with 5 μm Rev in dimethylsulphoxide (DMSO) or with DMSO alone for 6 days. Cell growth was assessed using the AlamarBlueTM assay. Cytoskeletal structure, endoproliferation and caspase-3-immunopositivity were assayed by epifluorescence microscopy. The OA HAC chondrogenic performance was evaluated by quantitative reverse transcription-polymerase chain reaction (RT-PCR) for glyceraldehyde-3-phosphate dehydrogenase, Sox9, Aggrecan (Agg), type II collagen (Col2), Ki67, cyclinD1, transforming growth factor-β1 (TGF-β1), -2 and -3, interleukin-1β (IL-1β) and -6 , SMAD3 and -7, and vascular endothelial growth factor. Rev-treated OA HAC recovered polygonal morphology and reduced Ki67 expression and proliferation. Cell-cycle impairment accounted for altered cytoskeletal organization, endoproliferation and apoptosis, whereas a compensatory mechanism sustained the increased cyclinD1 transcript levels. Sox9, Agg and TGFs were overexpressed, but not Col2. IL transcripts were massively downregulated. These events were dose-related and transient. Overall, in spite of a higher Rev-induced transcriptional activity for extracellular matrix components and in spite of a Rev-treated cell phenotype closer to that of the three-dimensional native articular chondrocyte, Rev effects seem unleashed from a full regained chondrogenic potential.
Collapse
Affiliation(s)
- S Gasparini
- Stem Cell Laboratory, Department of Experimental Medicine (Di.Me.S.), University of Genova, Advanced Biotechnology Centre, Genova, Italy
| | - F Villa
- Stem Cell Laboratory, Department of Experimental Medicine (Di.Me.S.), University of Genova, Advanced Biotechnology Centre, Genova, Italy
| | - L Molfetta
- Orthopedic Unit, Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Infant Sciences (Di.N.O.G.M.I), University of Genova, Genova, Italy
| | - E Repaci
- Stem Cell Laboratory, Department of Experimental Medicine (Di.Me.S.), University of Genova, Advanced Biotechnology Centre, Genova, Italy
| | | | - R Quarto
- Stem Cell Laboratory, Department of Experimental Medicine (Di.Me.S.), University of Genova, Advanced Biotechnology Centre, Genova, Italy
| | - P Giannoni
- Stem Cell Laboratory, Department of Experimental Medicine (Di.Me.S.), University of Genova, Advanced Biotechnology Centre, Genova, Italy
| |
Collapse
|
53
|
Walczyk M, Paradowska-Gorycka A, Olesinska M. Epigenetics: The Future Direction in Systemic Sclerosis. Scand J Immunol 2017; 86:427-435. [DOI: 10.1111/sji.12595] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/16/2017] [Indexed: 12/14/2022]
Affiliation(s)
- M. Walczyk
- Department of Connective Tissue Diseases; National Institute of Geriatrics, Rheumatology and Rehabilitation; Warsaw Poland
| | - A. Paradowska-Gorycka
- Department of Biochemistry and Molecular Biology; National Institute of Geriatrics, Rheumatology and Rehabilitation; Warsaw Poland
| | - M. Olesinska
- Department of Connective Tissue Diseases; National Institute of Geriatrics, Rheumatology and Rehabilitation; Warsaw Poland
| |
Collapse
|
54
|
Lafyatis R, Mantero JC, Gordon J, Kishore N, Carns M, Dittrich H, Spiera R, Simms RW, Varga J. Inhibition of β-Catenin Signaling in the Skin Rescues Cutaneous Adipogenesis in Systemic Sclerosis: A Randomized, Double-Blind, Placebo-Controlled Trial of C-82. J Invest Dermatol 2017; 137:2473-2483. [PMID: 28807667 DOI: 10.1016/j.jid.2017.06.032] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 05/31/2017] [Accepted: 06/18/2017] [Indexed: 12/13/2022]
Abstract
Several studies have suggested that Wnts might contribute to skin fibrosis in systemic sclerosis (SSc) by affecting the differentiation of pluripotent dermal cells. We tested C-82, a therapeutic that inhibits canonical Wnt signaling by blocking the interaction of the protein CBP with β-Catenin and inhibiting Wnt-activated genes. We used a trial design formulating C-82 for topical application and conducting a placebo-controlled, double-blinded clinical trial in which patients with diffuse cutaneous SSc were treated with C-82 or placebo on opposite forearms. C-82- compared with placebo-treated forearms did not show any clinical effect. Skin biopsies performed before and after treatment showed a very weak trend toward improvement in the C-82-treated skin of biomarkers of local skin disease, THBS1 and COMP. However, on microarray analysis C-82 treatment strongly up-regulated two clusters of genes that correlate negatively with the severity of SSc skin disease. These clusters are highly associated with metabolism and one gene, PLIN2, expressed only by sebocytes and subcutaneous fat cells. These changes in gene expression strongly support a role for Wnts in differentiation of pluripotent cells into profibrotic fibroblasts and the potential for C-82 with longer treatment to promote fat regeneration in SSc skin.
Collapse
Affiliation(s)
- Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Julio C Mantero
- Division of Rheumatology, Boston University Medical Center, Boston, Massachusetts, USA
| | - Jessica Gordon
- Rheumatology Division, Hospital for Special Surgery, New York, New York, USA
| | - Nina Kishore
- Division of Rheumatology, Boston University Medical Center, Boston, Massachusetts, USA
| | - Mary Carns
- Rheumatology Division, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Howard Dittrich
- Abboud CV Research Center, University of Iowa, Carver College of Medicine, Iowa City, Iowa, USA
| | - Robert Spiera
- Rheumatology Division, Hospital for Special Surgery, New York, New York, USA
| | - Robert W Simms
- Division of Rheumatology, Boston University Medical Center, Boston, Massachusetts, USA
| | - John Varga
- Rheumatology Division, Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
55
|
Ayers NB, Sun CM, Chen SY. Transforming growth factor-β signaling in systemic sclerosis. J Biomed Res 2017; 32:3-12. [PMID: 29353817 PMCID: PMC5956255 DOI: 10.7555/jbr.31.20170034] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Systemic sclerosis (SSc) is a complex, multiorgan autoimmune disease of unknown etiology. Manifestation of the disease results from an interaction of three key pathologic features including irregularities of the antigen-specific immune system and the non-specific immune system, resulting in autoantibody production, vascular endothelial activation of small blood vessels, and tissue fibrosis as a result of fibroblast dysfunction. Given the heterogeneity of clinical presentation of the disease, a lack of universal models has impeded adequate testing of potential therapies for SSc. Regardless, recent research has elucidated the roles of various ubiquitous molecular mechanisms that contribute to the clinical manifestation of the disease. Transforming growth factor β (TGF-β) has been identified as a regulator of pathological fibrogenesis in SSc. Various processes, including cell growth, apoptosis, cell differentiation, and extracellular matrix synthesis are regulated by TGF-β, a type of cytokine secreted by macrophages and many other cell types. Understanding the essential role TGF-β pathways play in the pathology of systemic sclerosis could provide a potential outlet for treatment and a better understanding of this severe disease.
Collapse
Affiliation(s)
- Nolan B Ayers
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Chen-Ming Sun
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| | - Shi-You Chen
- Department of Physiology & Pharmacology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
56
|
Tsou PS, Sawalha AH. Unfolding the pathogenesis of scleroderma through genomics and epigenomics. J Autoimmun 2017; 83:73-94. [PMID: 28526340 DOI: 10.1016/j.jaut.2017.05.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 05/09/2017] [Indexed: 12/12/2022]
Abstract
With unknown etiology, scleroderma (SSc) is a multifaceted disease characterized by immune activation, vascular complications, and excessive fibrosis in internal organs. Genetic studies, including candidate gene association studies, genome-wide association studies, and whole-exome sequencing have supported the notion that while genetic susceptibility to SSc appears to be modest, SSc patients are genetically predisposed to this disease. The strongest genetic association for SSc lies within the MHC region, with loci in HLA-DRB1, HLA-DQB1, HLA-DPB1, and HLA-DOA1 being the most replicated. The non-HLA genes associated with SSc are involved in various functions, with the most robust associations including genes for B and T cell activation and innate immunity. Other pathways include genes involved in extracellular matrix deposition, cytokines, and autophagy. Among these genes, IRF5, STAT4, and CD247 were replicated most frequently while SNPs rs35677470 in DNASE1L3, rs5029939 in TNFAIP3, and rs7574685 in STAT4 have the strongest associations with SSc. In addition to genetic predisposition, it became clear that environmental factors and epigenetic influences also contribute to the development of SSc. Epigenetics, which refers to studies that focus on heritable phenotypes resulting from changes in chromatin structure without affecting the DNA sequence, is one of the most rapidly expanding fields in biomedical research. Indeed extensive epigenetic changes have been described in SSc. Alteration in enzymes and mediators involved in DNA methylation and histone modification, as well as dysregulated non-coding RNA levels all contribute to fibrosis, immune dysregulation, and impaired angiogenesis in this disease. Genes that are affected by epigenetic dysregulation include ones involved in autoimmunity, T cell function and regulation, TGFβ pathway, Wnt pathway, extracellular matrix, and transcription factors governing fibrosis and angiogenesis. In this review, we provide a comprehensive overview of the current findings of SSc genetic susceptibility, followed by an extensive description and a systematic review of epigenetic research that has been carried out to date in SSc. We also summarize the therapeutic potential of drugs that affect epigenetic mechanisms, and outline the future prospective of genomics and epigenomics research in SSc.
Collapse
Affiliation(s)
- Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
57
|
Activation and overexpression of Sirt1 attenuates lung fibrosis via P300. Biochem Biophys Res Commun 2017; 486:1021-1026. [DOI: 10.1016/j.bbrc.2017.03.155] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 03/28/2017] [Indexed: 01/03/2023]
|
58
|
Wei J, Zhu H, Lord G, Bhattachayya M, Jones BM, Allaway G, Biswal SS, Korman B, Marangoni RG, Tourtellotte WG, Varga J. Nrf2 exerts cell-autonomous antifibrotic effects: compromised function in systemic sclerosis and therapeutic rescue with a novel heterocyclic chalcone derivative. Transl Res 2017; 183:71-86.e1. [PMID: 28027929 PMCID: PMC7205471 DOI: 10.1016/j.trsl.2016.12.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/26/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) governs antioxidant, innate immune and cytoprotective responses and its deregulation is prominent in chronic inflammatory conditions. To examine the hypothesis that Nrf2 might be implicated in systemic sclerosis (SSc), we investigated its expression, activity, and mechanism of action in SSc patient samples and mouse models of fibrosis and evaluated the effects of a novel pharmacologic Nrf2 agonist. We found that both expression and activity of Nrf2 were significantly reduced in SSc patient skin biopsies and showed negative correlation with inflammatory gene expression. In skin fibroblasts, Nrf2 mitigated fibrotic responses by blocking canonical transforming growth factor-β (TGF-β)-Smad signaling, whereas silencing Nrf2 resulted in constitutively elevated collagen synthesis, spontaneous myofibroblast differentiation, and enhanced TGF-ß responses. Bleomycin treatment of Nrf2-null mice resulted in exaggerated fibrosis. In wild-type mice, treatment with a novel pharmacologic Nrf2 agonist 2-trifluoromethyl-2'-methoxychalcone prevented dermal fibrosis induced by TGF-β. These findings are the first to identify Nrf2 as a cell-intrinsic antifibrotic factor with key roles in maintaining extracellular matrix homeostasis and a pathogenic role in SSc. Pharmacologic reactivation of Nrf2, therefore, represents a novel therapeutic strategy toward effective treatment of fibrosis in SSc.
Collapse
Affiliation(s)
- Jun Wei
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill.
| | - Hongyan Zhu
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill
| | - Gabriel Lord
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill
| | - Mitra Bhattachayya
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill
| | | | | | - Shyam S Biswal
- Department of Environmental Health Sciences, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, Md
| | - Benjamin Korman
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill
| | | | - Warren G Tourtellotte
- Department of Pathology, Feinberg School of Medicine, Chicago, Ill; Department of Neurology, Feinberg School of Medicine, Chicago, Ill
| | - John Varga
- Northwestern Scleroderma Program, Feinberg School of Medicine, Chicago, Ill.
| |
Collapse
|
59
|
Sayad A, Ghafouri-Fard S, Omrani MD, Noroozi R, Taheri M. Myxovirus resistance protein A (MxA) polymorphism is associated with IFNβ response in Iranian multiple sclerosis patients. Neurol Sci 2017; 38:1093-1099. [PMID: 28386647 DOI: 10.1007/s10072-017-2935-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 03/23/2017] [Indexed: 12/01/2022]
Abstract
Multiple sclerosis (MS) is a heterogeneous immune-related demyelinating disorder of central nervous system with several genetic and environmental factors contributing in its pathogenesis or patients' response to therapies. Myxovirus resistance protein A (MxA) is among the genes which are induced by IFNβ and are involved in the MS pathogenesis and/or response to IFNβ. In the present case-control study, we evaluated the association between three SNPs at nt -123 (A or C, rs17000900), nt -88 (G or T, rs2071430), and nt +20 (A or C, rs464138) and MS risk as well as treatment response in a population of Iranian MS patients including 146 IFNβ responders and 85 non-responders as well as 180 healthy controls. The AGA (-123, -88, +20) haplotype was more frequent in controls compared with MS cases (P = 0.038, OR (95% CI) = 1.77 (1.03-3.02)). Of particular note, the frequency of rs464138 AA genotype was significantly higher in responders compared with non-responders. However, the allele and genotype frequencies of other SNPs were not significantly different among patient subtypes or between patients and controls. Besides, we have demonstrated that CGC, ATA, and AGA (-123, -88, +20) haplotypes were significantly associated with IFNβ response in MS patients. As SNPs on MxA promoter region might participate in MS patients' response to IFNβ, prior patients genotyping may increase the rate of responsiveness and help in individualized selection of treatment options.
Collapse
Affiliation(s)
- Arezou Sayad
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran
| | - Rezvan Noroozi
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran. .,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran. .,Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, 8th Floor, SBUMS Bldg., Next to Ayatollah Taleghani Hospital, Evin, Tehran, 198396-3113, Iran.
| |
Collapse
|
60
|
Angiolilli C, Baeten DL, Radstake TR, Reedquist KA. The acetyl code in rheumatoid arthritis and other rheumatic diseases. Epigenomics 2017; 9:447-461. [DOI: 10.2217/epi-2016-0136] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Growing evidence supports the idea that aberrancies in epigenetic processes contribute to the onset and progression of human immune-mediated inflammatory diseases, such as rheumatoid arthritis (RA). Epigenetic regulators of histone tail modifications play a role in chromatin accessibility and transcriptional responses to inflammatory stimuli. Among these, histone deacetylases (HDACs) regulate the acetylation status of histones and nonhistone proteins, essential for immune responses. Broad-spectrum HDAC inhibitors are well-known anti-inflammatory agents and reduce disease severity in animal models of arthritis; however, selective HDAC inhibitors remain poorly studied. In this review, we describe emerging findings regarding the aberrant acetyl code in RA and other rheumatic disorders which may help identify not only novel diagnostic and prognostic clinical biomarkers for RA, but also new targets for epigenetic pharmacological applications.
Collapse
Affiliation(s)
- Chiara Angiolilli
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Amsterdam Rheumatology & Immunology Center, Department of Clinical Immunology & Rheumatology, Department of Experimental Immunology Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Dominique L Baeten
- Amsterdam Rheumatology & Immunology Center, Department of Clinical Immunology & Rheumatology, Department of Experimental Immunology Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timothy R Radstake
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kris A Reedquist
- Laboratory of Translational Immunology & Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
61
|
Ghosh AK, Rai R, Flevaris P, Vaughan DE. Epigenetics in Reactive and Reparative Cardiac Fibrogenesis: The Promise of Epigenetic Therapy. J Cell Physiol 2017; 232:1941-1956. [PMID: 27883184 DOI: 10.1002/jcp.25699] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/20/2022]
Abstract
Epigenetic changes play a pivotal role in the development of a wide spectrum of human diseases including cardiovascular diseases, cancer, diabetes, and intellectual disabilities. Cardiac fibrogenesis is a common pathophysiological process seen during chronic and stress-induced accelerated cardiac aging. While adequate production of extracellular matrix (ECM) proteins is necessary for post-injury wound healing, excessive synthesis and accumulation of extracellular matrix protein in the stressed or injured hearts causes decreased or loss of lusitropy that leads to cardiac failure. This self-perpetuating deposition of collagen and other matrix proteins eventually alter cellular homeostasis; impair tissue elasticity and leads to multi-organ failure, as seen during pathogenesis of cardiovascular diseases, chronic kidney diseases, cirrhosis, idiopathic pulmonary fibrosis, and scleroderma. In the last 25 years, multiple studies have investigated the molecular basis of organ fibrosis and highlighted its multi-factorial genetic, epigenetic, and environmental regulation. In this minireview, we focus on five major epigenetic regulators and discuss their central role in cardiac fibrogenesis. Additionally, we compare and contrast the epigenetic regulation of hypertension-induced reactive fibrogenesis and myocardial infarction-induced reparative or replacement cardiac fibrogenesis. As microRNAs-one of the major epigenetic regulators-circulate in plasma, we also advocate their potential diagnostic role in cardiac fibrosis. Lastly, we discuss the evolution of novel epigenetic-regulating drugs and predict their clinical role in the suppression of pathological cardiac remodeling, cardiac aging, and heart failure. J. Cell. Physiol. 232: 1941-1956, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Rahul Rai
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Panagiotis Flevaris
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Douglas E Vaughan
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
62
|
Sun J, Wang Y, Cui W, Lou Y, Sun G, Zhang D, Miao L. Role of Epigenetic Histone Modifications in Diabetic Kidney Disease Involving Renal Fibrosis. J Diabetes Res 2017; 2017:7242384. [PMID: 28695133 PMCID: PMC5485509 DOI: 10.1155/2017/7242384] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/14/2017] [Indexed: 12/18/2022] Open
Abstract
One of the commonest causes of end-stage renal disease is diabetic kidney disease (DKD). Renal fibrosis, characterized by the accumulation of extracellular matrix (ECM) proteins in glomerular basement membranes and the tubulointerstitium, is the final manifestation of DKD. The TGF-β pathway triggers epithelial-to-mesenchymal transition (EMT), which plays a key role in the accumulation of ECM proteins in DKD. DCCT/EDIC studies have shown that DKD often persists and progresses despite glycemic control in diabetes once DKD sets in due to prior exposure to hyperglycemia called "metabolic memory." These imply that epigenetic factors modulate kidney gene expression. There is evidence to suggest that in diabetes and hyperglycemia, epigenetic histone modifications have a significant effect in modulating renal fibrotic and ECM gene expression induced by TGF-β1, as well as its downstream profibrotic genes. Histone modifications are also implicated in renal fibrosis through its ability to regulate the EMT process triggered by TGF-β signaling. In view of this, efforts are being made to develop HAT, HDAC, and HMT inhibitors to delay, stop, or even reverse DKD. In this review, we outline the latest advances that are being made to regulate histone modifications involved in DKD.
Collapse
Affiliation(s)
- Jing Sun
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Yangwei Wang
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Wenpeng Cui
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Yan Lou
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Guangdong Sun
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Dongmei Zhang
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
| | - Lining Miao
- Department of Nephrology, Second Hospital of Jilin University, Changchun 130041, China
- *Lining Miao:
| |
Collapse
|
63
|
Lafyatis R. Editorial: Epigenetics in Systemic Sclerosis. Arthritis Rheumatol 2016; 68:2841-2844. [DOI: 10.1002/art.39830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 07/26/2016] [Indexed: 02/02/2023]
Affiliation(s)
- Robert Lafyatis
- University of Pittsburgh Medical Center; Pittsburgh Pennsylvania
| |
Collapse
|
64
|
Wang C, Yang S, Huang J, Chen S, Li Y, Li Q. Activation of corticotropin releasing factor receptors up regulates collagen production by hepatic stellate cells via promoting p300 expression. Biol Chem 2016; 397:437-44. [PMID: 26756093 DOI: 10.1515/hsz-2015-0233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 12/30/2015] [Indexed: 12/20/2022]
Abstract
Liver fibrosis is characterized with the over expression and excessive accumulation of extracellular matrix proteins, including collagens. The causative factors in the over production of collagens are not fully understood. This study aims to test a hypothesis that activation of corticotropin releasing factor receptors up regulates the expression of collagen in hepatic stellate cells. In this study, human hepatic stellate cell line, LX-2 cells were cultured. Expression of collagens by LX-2 cells was assessed by real time RT-PCR, Western blotting. The results showed that, upon exposure to urocortin in the culture, LX-2 cells (a human hepatic stellate cell line) increased the expression of collagen IV (Col4) markedly. The exposure to urocortin also enhanced the levels of pTip60, H3K9, RNA polymerase II and forkhead box protein 3 at the collagen promoter locus as well as increase in the expression of Col4 mRNA and protein in the cells. Blocking p300 efficiently suppressed the urocortin-induced Col4 expression in LX-2 cells and unveiled an apoptosis-inducing effect of urocortin. In conclusion, activation of CRF receptors is capable of enforcing the production of Col4 by LX-2 cells via up regulating the p300 pathway, which may contribute to the development of liver fibrosis.
Collapse
|
65
|
Sandoval J, Pereda J, Pérez S, Finamor I, Vallet-Sánchez A, Rodríguez JL, Franco L, Sastre J, López-Rodas G. Epigenetic Regulation of Early- and Late-Response Genes in Acute Pancreatitis. THE JOURNAL OF IMMUNOLOGY 2016; 197:4137-4150. [PMID: 27798150 DOI: 10.4049/jimmunol.1502378] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 09/16/2016] [Indexed: 12/12/2022]
Abstract
Chromatin remodeling seems to regulate the patterns of proinflammatory genes. Our aim was to provide new insights into the epigenetic mechanisms that control transcriptional activation of early- and late-response genes in initiation and development of severe acute pancreatitis as a model of acute inflammation. Chromatin changes were studied by chromatin immunoprecipitation analysis, nucleosome positioning, and determination of histone modifications in promoters of proinflammatory genes in vivo in the course of taurocholate-induced necrotizing pancreatitis in rats and in vitro in rat pancreatic AR42J acinar cells stimulated with taurocholate or TNF-α. Here we show that the upregulation of early and late inflammatory genes rely on histone acetylation associated with recruitment of histone acetyltransferase CBP. Chromatin remodeling of early genes during the inflammatory response in vivo is characterized by a rapid and transient increase in H3K14ac, H3K27ac, and H4K5ac as well as by recruitment of chromatin-remodeling complex containing BRG-1. Chromatin remodeling in late genes is characterized by a late and marked increase in histone methylation, particularly in H3K4. JNK and p38 MAPK drive the recruitment of transcription factors and the subsequent upregulation of early and late inflammatory genes, which is associated with nuclear translocation of the early gene Egr-1 In conclusion, specific and strictly ordered epigenetic markers such as histone acetylation and methylation, as well as recruitment of BRG-1-containing remodeling complex are associated with the upregulation of both early and late proinflammatory genes in acute pancreatitis. Our findings highlight the importance of epigenetic regulatory mechanisms in the control of the inflammatory cascade.
Collapse
Affiliation(s)
- Juan Sandoval
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Valencia, 46100 Valencia, Spain
| | - Javier Pereda
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain; and
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain; and
| | - Isabela Finamor
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain; and
| | - Azahara Vallet-Sánchez
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Valencia, 46100 Valencia, Spain.,Institute of Health Research INCLIVA (Foundation for Research of the Clinic Hospital of the "Comunidad Valenciana"), 46010 Valencia, Spain
| | - José Luis Rodríguez
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Valencia, 46100 Valencia, Spain
| | - Luis Franco
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Valencia, 46100 Valencia, Spain.,Institute of Health Research INCLIVA (Foundation for Research of the Clinic Hospital of the "Comunidad Valenciana"), 46010 Valencia, Spain
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Valencia, Spain; and
| | - Gerardo López-Rodas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Valencia, 46100 Valencia, Spain.,Institute of Health Research INCLIVA (Foundation for Research of the Clinic Hospital of the "Comunidad Valenciana"), 46010 Valencia, Spain
| |
Collapse
|
66
|
Epigenetics in fibrosis. Mol Aspects Med 2016; 54:89-102. [PMID: 27720780 DOI: 10.1016/j.mam.2016.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022]
Abstract
Fibrosis is a common and important disease. It is a pathological state due to excessive scar formation mediated by an increase in activated fibroblasts that express alpha smooth muscle actin and copious amounts of extracellular matrix molecules. Epigenetics is an area of research that encompasses three main mechanisms: methylation, histone modifications to the tails of histones and also non-coding RNAs including long and short non-coding RNAs. These three mechanisms all seek to regulate gene expression without a change in the underlying DNA sequence. In recent years an explosion of research, aided by deep sequencing technology becoming available, has demonstrated a role for epigenetics in fibrosis, either organ specific like lung fibrosis or more widespread as in systemic sclerosis. While the great majority of epigenetic work in fibrosis is centered on histone codes, more recently the non-coding RNAs have been examined in greater detail. It is known that one modification can affect the other and cross-talk among all three adds a new layer of complexity. This review aims to examine the role of epigenetics in fibrosis, evaluating all three mechanisms, and to suggest possible areas where epigenetics could be targeted therapeutically.
Collapse
|
67
|
Muta K, Obata Y, Oka S, Abe S, Minami K, Kitamura M, Endo D, Koji T, Nishino T. Curcumin ameliorates nephrosclerosis via suppression of histone acetylation independent of hypertension. Nephrol Dial Transplant 2016; 31:1615-23. [DOI: 10.1093/ndt/gfw036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/31/2016] [Indexed: 01/10/2023] Open
|
68
|
Stratton MS, McKinsey TA. Epigenetic regulation of cardiac fibrosis. J Mol Cell Cardiol 2016; 92:206-13. [PMID: 26876451 PMCID: PMC4987078 DOI: 10.1016/j.yjmcc.2016.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/05/2016] [Accepted: 02/10/2016] [Indexed: 01/01/2023]
Abstract
Fibrosis is defined as excess deposition of extracellular matrix (ECM), resulting in tissue scarring and organ dysfunction. In the heart, fibrosis may be reparative, replacing areas of myocyte loss with a structural scar following infarction, or reactive, which is triggered in the absence of cell death and involves interstitial ECM deposition in response to long-lasting stress. Interstitial fibrosis can increase the passive stiffness of the myocardium, resulting in impaired relaxation and diastolic dysfunction. Additionally, fibrosis can lead to disruption of electrical conduction in the heart, causing arrhythmias, and can limit myocyte oxygen availability and thus exacerbate myocardial ischemia. Here, we review recent studies that have illustrated key roles for epigenetic events in the control of pro-fibrotic gene expression, and highlight the potential of small molecules that target epigenetic regulators as a means of treating fibrotic cardiac diseases.
Collapse
Affiliation(s)
- Matthew S Stratton
- Department of Medicine, Division of Cardiology and Center for Fibrosis Research and Translation, University of Colorado Denver, 12700 E. 19th Ave, Aurora, CO 80045-0508, United States
| | - Timothy A McKinsey
- Department of Medicine, Division of Cardiology and Center for Fibrosis Research and Translation, University of Colorado Denver, 12700 E. 19th Ave, Aurora, CO 80045-0508, United States.
| |
Collapse
|
69
|
Ghosh AK. FAT-free p300 is good for scar-free tissue repair. J Cell Biochem 2015; 115:1486-9. [PMID: 24733520 DOI: 10.1002/jcb.24820] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 11/10/2022]
Abstract
Fibrosis, the deadly pathological manifestation of an abnormal tissue remodeling in any organ due to excessive collagen deposition, is associated with a wide variety of organ failure-related human diseases. Chronic stress or repeated injury in a particular organ induces abnormal molecular signals that lead to super-activation of matrix protein producing fibroblasts, excessive matrix proteins accumulation, loss of physiological tissue architecture or elasticity, and ultimately leading to organ failure. There is no effective therapy for fibrosis. Factor acetyltransferase p300 (FATp300), a major epigenetic regulator that acetylates specific lysines in histones and transcription factors, is essential for elevated collagen synthesis and the levels of FATp300 are significantly elevated in different fibrotic tissues. Pharmacological inhibition of FAT activity of p300 is associated with decreased collagen synthesis by fibroblasts in tissues and amelioration of organ fibrosis. Therefore, FAT-free p300 is superior for physiological tissue repair and must be exploited as a viable therapeutic target against multi-organ fibrosis.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
70
|
Epigenetic mechanisms: An emerging role in pathogenesis and its therapeutic potential in systemic sclerosis. Int J Biochem Cell Biol 2015; 67:92-100. [DOI: 10.1016/j.biocel.2015.05.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 05/23/2015] [Accepted: 05/27/2015] [Indexed: 11/18/2022]
|
71
|
MicroRNAs Regulating Signaling Pathways: Potential Biomarkers in Systemic Sclerosis. GENOMICS PROTEOMICS & BIOINFORMATICS 2015; 13:234-41. [PMID: 26365208 PMCID: PMC4610971 DOI: 10.1016/j.gpb.2015.07.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 07/22/2015] [Accepted: 07/27/2015] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem fibrotic and autoimmune disease. Both genetic and epigenetic elements mediate SSc pathophysiology. This review summarizes the role of one epigenetic element, known as microRNAs (miRNAs), involved in different signaling pathways of SSc pathogenesis. The expression of key components in transforming growth factor-β (TGF-β) signaling pathway has been found to be regulated by miRNAs both upstream and downstream of TGF-β. We are specifically interested in the pathway components upstream of TGF-β, while miRNAs in other signaling pathways have not been extensively studied. The emerging role of miRNAs in vasculopathy of SSc suggests a promising new direction for future investigation. Elucidation of the regulatory role of miRNAs in the expression of signaling factors may facilitate the discovery of novel biomarkers in SSc and improve the understanding and treatment of this disease.
Collapse
|
72
|
Marie I, Gehanno JF. Environmental risk factors of systemic sclerosis. Semin Immunopathol 2015; 37:463-73. [DOI: 10.1007/s00281-015-0507-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/16/2015] [Indexed: 12/16/2022]
|
73
|
Wei J, Ghosh AK, Chu H, Fang F, Hinchcliff ME, Wang J, Marangoni RG, Varga J. The Histone Deacetylase Sirtuin 1 Is Reduced in Systemic Sclerosis and Abrogates Fibrotic Responses by Targeting Transforming Growth Factor β Signaling. Arthritis Rheumatol 2015; 67:1323-34. [PMID: 25707573 DOI: 10.1002/art.39061] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 01/29/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Persistent fibroblast activation underlies skin fibrosis in systemic sclerosis (SSc), but the transcriptional and epigenetic mechanisms controlling this process are not well understood. In view of the potent influence of acetylation status governing tissue fibrosis, we undertook this study to investigate the expression of the antiaging deacetylase enzyme sirtuin 1 (SIRT1) in SSc and its effects on fibrotic responses in vitro and in vivo. METHODS Tissue expression of SIRTs was interrogated from publicly available genome-wide expression data sets and by immunohistochemistry. The effects of SIRT1 on modulating fibrotic responses, as well as the underlying mechanisms, were examined in human and mouse fibroblasts in culture and in an experimental fibrosis model in the mouse. RESULTS Analysis of transcriptome data revealed a selective reduction of SIRT1 messenger RNA (mRNA) levels in SSc skin biopsy samples as well as a negative correlation of SIRT1 mRNA with the skin score. Cellular SIRT1 levels were suppressed in normal fibroblasts exposed to hypoxia or platelet-derived growth factor and were constitutively down-regulated in SSc fibroblasts. Activation of SIRT1 attenuated fibrotic responses in skin fibroblasts and skin organ cultures, while genetic or pharmacologic inhibition of SIRT1 had profibrotic effects. The antifibrotic effects of SIRT1 were due in part to decreased expression and function of the acetyltransferase p300. In mice, experimentally induced skin fibrosis was accompanied by reduced SIRT1 expression in lesional tissue fibroblasts, and both fibrosis and loss of SIRT1 in these mice were mitigated by treatment with a SIRT1 activator. CONCLUSION SIRT1 has antifibrotic effects, and its reduced tissue expression in patients with SSc might have a direct causal role in progression of fibrosis. Pharmacologic modulation of SIRT1 in these patients therefore might represent a potential treatment strategy.
Collapse
Affiliation(s)
- Jun Wei
- Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | | | | | | | | | | | | |
Collapse
|
74
|
Abstract
Systemic sclerosis is a complex autoimmune disease characterized by a chronic and frequently progressive course and by extensive patient-to-patient variability. Like other autoimmune diseases, systemic sclerosis occurs more frequently in women, with a peak of onset in the fifth decade of life. The exact cause of systemic sclerosis remains elusive but is likely to involve environmental factors in a genetically primed individual. Pathogenesis is dominated by vascular changes; evidence of autoimmunity with distinct autoantibodies and activation of both innate and adaptive immunity; and fibrosis of the skin and visceral organs that results in irreversible scarring and organ failure. Intractable progression of vascular and fibrotic organ damage accounts for the chronic morbidity and high mortality. Early and accurate diagnosis and classification might improve patient outcomes. Screening strategies facilitate timely recognition of life-threatening complications and initiation of targeted therapies to halt their progression. Effective treatments of organ-based complications are now within reach. Discovery of biomarkers - including autoantibodies that identify patient subsets at high risk for particular disease complications or rapid progression - is a research priority. Understanding the key pathogenetic pathways, cell types and mediators underlying disease manifestations opens the door for the development of targeted therapies with true disease-modifying potential. For an illustrated summary of this Primer, visit: http://go.nature.com/lchkcA.
Collapse
|
75
|
Xu H, Wu X, Qin H, Tian W, Chen J, Sun L, Fang M, Xu Y. Myocardin-Related Transcription Factor A Epigenetically Regulates Renal Fibrosis in Diabetic Nephropathy. J Am Soc Nephrol 2014; 26:1648-60. [PMID: 25349198 DOI: 10.1681/asn.2014070678] [Citation(s) in RCA: 101] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 09/09/2014] [Indexed: 11/03/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most common complications associated with diabetes and characterized by renal microvascular injury along with accelerated synthesis of extracellular matrix proteins causing tubulointerstitial fibrosis. Production of type I collagen, the major component of extracellular matrix, is augmented during renal fibrosis after chronic exposure to hyperglycemia. However, the transcriptional modulator responsible for the epigenetic manipulation leading to induction of type I collagen genes is not clearly defined. We show here that tubulointerstitial fibrosis as a result of DN was diminished in myocardin-related transcription factor A (MRTF-A) -deficient mice. In cultured renal tubular epithelial cells and the kidneys of mice with DN, MRTF-A was induced by glucose and synergized with glucose to activate collagen transcription. Notably, MRTF-A silencing led to the disappearance of prominent histone modifications indicative of transcriptional activation, including acetylated histone H3K18/K27 and trimethylated histone H3K4. Detailed analysis revealed that MRTF-A recruited p300, a histone acetyltransferase, and WD repeat-containing protein 5 (WDR5), a key component of the histone H3K4 methyltransferase complex, to the collagen promoters and engaged these proteins in transcriptional activation. Estradiol suppressed collagen production by dampening the expression and binding activity of MRTF-A and interfering with the interaction between p300 and WDR5 in renal epithelial cells. Therefore, targeting the MRTF-A-associated epigenetic machinery might yield interventional strategies against DN-associated renal fibrosis.
Collapse
Affiliation(s)
- Huihui Xu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and
| | - Xiaoyan Wu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing, China
| | - Hao Qin
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and
| | - Wenfang Tian
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and
| | - Junliang Chen
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and School of Basic Medical Sciences, Jiangnan University, Wuxi, China
| | - Lina Sun
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and Department of Pathology and Pathophysiology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China; and
| | - Mingming Fang
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and Department of Medicine and Nursing, Jiangsu Jiankang Vocational University, Nanjing, China
| | - Yong Xu
- State Key Laboratory of Reproductive Medicine, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Pathophysiology and
| |
Collapse
|
76
|
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease of unclear aetiology. A multitude of genetic studies, ranging from candidate-gene studies to genome-wide association studies, have identified a large number of genetic susceptibility factors for SSc and its clinical phenotypes, but the contribution of these factors to disease susceptibility is only modest. However, in an endeavour to explore how the environment might affect genetic susceptibility, epigenetic research into SSc is rapidly expanding. Orchestrated by environmental factors, epigenetic modifications can drive genetically predisposed individuals to develop autoimmunity, and are thought to represent the crossroads between the environment and genetics in SSc. Therefore, in addition to providing a comprehensive description of the current understanding of genetic susceptibility underlying SSc, this Review describes the involvement of epigenetic phenomena, including DNA methylation patterns, histone modifications and microRNAs, in SSc.
Collapse
|
77
|
van Loosdregt J, Coffer PJ. Post-translational modification networks regulating FOXP3 function. Trends Immunol 2014; 35:368-78. [PMID: 25047417 DOI: 10.1016/j.it.2014.06.005] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 06/23/2014] [Accepted: 06/23/2014] [Indexed: 01/01/2023]
Abstract
Forkhead box (FOX)P3 is a requisite transcription factor for the development and maintenance of immunosuppressive function of regulatory T (Treg) cells, and therefore for immune homeostasis. Post-translational modifications (PTMs) can transiently alter the functionality of transcription factors, and recent evidence reveals that FOXP3 can be regulated by various PTMs including acetylation, ubiquitination, and phosphorylation. Here, we review the current understanding of how these modifications control FOXP3, including regulation of DNA binding, transactivation potential, and proteasomal degradation. We place these findings in the context of the biology of Treg cells, and discuss both limitations in translating biochemical findings into in vivo functions and the opportunities presented by a better understanding of the molecular mechanisms that can transiently control FOXP3 activity in response to environmental cues.
Collapse
Affiliation(s)
- Jorg van Loosdregt
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Cell Biology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul J Coffer
- Division of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands; Department of Cell Biology, University Medical Center Utrecht, Utrecht, The Netherlands; Memorial Sloan-Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
78
|
Influence of Egr-1 in cardiac tissue-derived mesenchymal stem cells in response to glucose variations. BIOMED RESEARCH INTERNATIONAL 2014; 2014:254793. [PMID: 24967343 PMCID: PMC4054710 DOI: 10.1155/2014/254793] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 05/06/2014] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cells (MSCs) represent a promising cell population for cell therapy and regenerative medicine applications. However, how variations in glucose are perceived by MSC pool is still unclear. Since, glucose metabolism is cell type and tissue dependent, this must be considered when MSCs are derived from alternative sources such as the heart. The zinc finger transcription factor Egr-1 is an important early response gene, likely to play a key role in the glucose-induced response. Our aim was to investigate how short-term changes in in vitro glucose concentrations affect multipotent cardiac tissue-derived MSCs (cMSCs) in a mouse model of Egr-1 KO (Egr-1−/−). Results showed that loss of Egr-1 does not significantly influence cMSC proliferation. In contrast, responses to glucose variations were observed in wt but not in Egr-1−/− cMSCs by clonogenic assay. Phenotype analysis by RT-PCR showed that cMSCs Egr-1−/− lost the ability to regulate the glucose transporters GLUT-1 and GLUT-4 and, as expected, the Egr-1 target genes VEGF, TGFβ-1, and p300. Acetylated protein levels of H3 histone were impaired in Egr-1−/− compared to wt cMSCs. We propose that Egr-1 acts as immediate glucose biological sensor in cMSCs after a short period of stimuli, likely inducing epigenetic modifications.
Collapse
|
79
|
Chen S, Puthanveetil P, Feng B, Matkovich SJ, Dorn GW, Chakrabarti S. Cardiac miR-133a overexpression prevents early cardiac fibrosis in diabetes. J Cell Mol Med 2014; 18:415-21. [PMID: 24428157 PMCID: PMC3955148 DOI: 10.1111/jcmm.12218] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 11/27/2013] [Indexed: 01/14/2023] Open
Abstract
Diabetic cardiomyopathy is a cascade of complex events leading to eventual failure of the heart and cardiac fibrosis being considered as one of its major causes. miR-133a is one of the most abundantly expressed microRNAs in the heart. We investigated the role of miR-133a during severe hyperglycaemia. And, our aim was to find out what role miR-133a plays during diabetes-induced cardiac fibrosis. We saw a drastic decrease in miR-133a expression in the hearts of streptozotocin-induced diabetic animals, as measured by RT-qPCR. This decrease was accompanied by an increase in the transcriptional co-activator EP300 mRNA and major markers of fibrosis [transforming growth factor-β1, connective tissue growth factor, fibronectin (FN1) and COL4A1]; in addition, focal cardiac fibrosis assessed by Masson's trichome stain was increased. Interestingly, in diabetic mice with cardiac-specific miR-133aa overexpression, cardiac fibrosis was significantly decreased, as observed by RT-qPCR and immunoblotting of COL4A1, ELISA for FN1 and microscopic examination. Furthermore, Cardiac miR-133a overexpression prevented ERK1/2 and SMAD-2 phosphorylation. These findings show that miR-133a could be a potential therapeutic target for diabetes-induced cardiac fibrosis and related cardiac dysfunction.
Collapse
Affiliation(s)
- Shali Chen
- Department of Pathology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | | | | | | | | | | |
Collapse
|
80
|
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
|
81
|
|