1
|
Soltanieh SK, Khastar S, Kaur I, Kumar A, Bansal J, Fateh A, Nathiya D, Husseen B, Rajabivahid M, Dehghani-Ghorbi M, Akhavan-Sigari R. Long Non-Coding RNAs in Non-Alcoholic Fatty Liver Disease; Friends or Foes? Cell Biochem Biophys 2025; 83:279-294. [PMID: 39377981 DOI: 10.1007/s12013-024-01555-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 01/03/2025]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a range of conditions that start with the accumulation of fat in the liver (hepatic steatosis) and can progress to more severe stages like steatohepatitis (NASH) and fibrosis without drinking alcohol. Environmental and genetic variables both contribute to MAFLD's development, with various biological processes and mediators involved at every phase. Long non-coding RNAs (lncRNAs) are a class of RNA molecules that are not translated into protein and are over 200 nucleotides long. They can impact genes that encode protein by controlling transcriptional and post-transcriptional procedures. Dysregulation of lncRNA has been connected to several liver diseases, including MAFLD. Recent research has linked lncRNAs to MAFLD pathology in both patients and animal models. However, the roles of most lncRNAs in MAFLD pathology are still not well recognized. This review provides a comprehensive catalog of recently reported lncRNAs in the pathogenesis of MAFLD and summarizes the current knowledge of lncRNAs usage as therapeutic strategies in MAFLD, the most common liver disease. Collectively, lncRNA's targeting could potentially offer a therapeutic approach by modulating MAFLD.
Collapse
Affiliation(s)
| | - Sahar Khastar
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka-560069, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh-247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand-831001, India
| | - Jaya Bansal
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges, Jhanjeri, Mohali, 140307, Punjab, India
| | - Ata Fateh
- Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
| | - Mansour Rajabivahid
- Department of Internal Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Mahmoud Dehghani-Ghorbi
- Hematology-Oncology Department, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University Warsaw, Warszawa, Poland
| |
Collapse
|
2
|
Ren L, Chang YF, Jiang SH, Li XH, Cheng HP. DNA methylation modification in Idiopathic pulmonary fibrosis. Front Cell Dev Biol 2024; 12:1416325. [PMID: 38915445 PMCID: PMC11194555 DOI: 10.3389/fcell.2024.1416325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/22/2024] [Indexed: 06/26/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and irreversible interstitial lung disease with a prognosis worse than lung cancer. It is a fatal lung disease with largely unknown etiology and pathogenesis, and no effective therapeutic drugs render its treatment largely unsuccessful. With continuous in-depth research efforts, the epigenetic mechanisms in IPF pathogenesis have been further discovered and concerned. As a widely studied mechanism of epigenetic modification, DNA methylation is primarily facilitated by DNA methyltransferases (DNMTs), resulting in the addition of a methyl group to the fifth carbon position of the cytosine base, leading to the formation of 5-methylcytosine (5-mC). Dysregulation of DNA methylation is intricately associated with the advancement of respiratory disorders. Recently, the role of DNA methylation in IPF pathogenesis has also received considerable attention. DNA methylation patterns include methylation modification and demethylation modification and regulate a range of essential biological functions through gene expression regulation. The Ten-Eleven-Translocation (TET) family of DNA dioxygenases is crucial in facilitating active DNA demethylation through the enzymatic conversion of the modified genomic base 5-mC to 5-hydroxymethylcytosine (5-hmC). TET2, a member of TET proteins, is involved in lung inflammation, and its protein expression is downregulated in the lungs and alveolar epithelial type II cells of IPF patients. This review summarizes the current knowledge of pathologic features and DNA methylation mechanisms of pulmonary fibrosis, focusing on the critical roles of abnormal DNA methylation patterns, DNMTs, and TET proteins in impacting IPF pathogenesis. Researching DNA methylation will enchance comprehension of the fundamental mechanisms involved in IPF pathology and provide novel diagnostic biomarkers and therapeutic targets for pulmonary fibrosis based on the studies involving epigenetic mechanisms.
Collapse
Affiliation(s)
- Lu Ren
- Clinical Nursing Teaching and Research Section, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yan-Fen Chang
- Medicine School, Zhengzhou University of Industrial Technology, Zhengzhou, China
| | - Shi-He Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Hong Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hai-Peng Cheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Wierzbicka A, Semik-Gurgul E, Świątkiewicz M, Szmatoła T, Steg A, Oczkowicz M. Changes in DNA Methylation and mRNA Expression in Lung Tissue after Long-Term Supplementation with an Increased Dose of Cholecalciferol. Int J Mol Sci 2023; 25:464. [PMID: 38203636 PMCID: PMC10778667 DOI: 10.3390/ijms25010464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
Maintaining an appropriate concentration of vitamin D is essential for the proper functioning of the body, regardless of age. Nowadays, there are more and more indications that vitamin D supplementation at higher than standard doses may show protective and therapeutic effects. Our study identified differences in the body's response to long-term supplementation with cholecalciferol at an increased dose. Two groups of pigs were used in the experiment. The first group received a standard dose of cholecalciferol (grower, 2000 IU/kg feed, and finisher, 1500 IU/kg feed), and the second group received an increased dose (grower, 3000 IU/kg feed, and finisher, 2500 IU/kg feed). After slaughter, lung samples were collected and used for RRBS and mRNA sequencing. Analysis of the methylation results showed that 2349 CpG sites had significantly altered methylation patterns and 1116 (47.51%) identified DMSs (Differentially Methylated Sites) were related to genes and their regulatory sites. The mRNA sequencing results showed a significant change in the expression of 195 genes. The integrated analysis identified eleven genes with DNA methylation and mRNA expression differences between the analyzed groups. The results of this study suggested that an increased vitamin D intake may be helpful for the prevention of lung cancer and pulmonary fibrosis. These actions may stem from the influence of vitamin D on the expression of genes associated with collagen production, such as SHMT1, UGT1A6, and ITIH2.The anti-cancer properties of vitamin D are also supported by changes in KLHL3 and TTPA gene expression.
Collapse
Affiliation(s)
- Alicja Wierzbicka
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Ewelina Semik-Gurgul
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Małgorzata Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland;
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
- Center for Experimental and Innovative Medicine, The University of Agriculture in Kraków, Rędzina 1c, 30-248 Kraków, Poland
| | - Anna Steg
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| | - Maria Oczkowicz
- Department of Animal Molecular Biology, National Research Institute of Animal Production, ul. Krakowska 1, 32-083 Balice, Poland; (A.W.); (E.S.-G.); (T.S.); (A.S.)
| |
Collapse
|
4
|
Zhao Y, Du L, Sun J, Wang X, Cong Z, Chen S, Wang F, Li Z. Exosomal miR-218 derived from mesenchymal stem cells inhibits endothelial-to-mesenchymal transition by epigenetically modulating of BMP2 in pulmonary fibrosis. Cell Biol Toxicol 2023; 39:2919-2936. [PMID: 37247103 DOI: 10.1007/s10565-023-09810-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 05/08/2023] [Indexed: 05/30/2023]
Abstract
Endothelial-to-mesenchymal transition (EndMT), the process by which endothelial cells lose their characteristics and acquire mesenchymal phenotypes, participates in the pathogenic mechanism of idiopathic pulmonary fibrosis. Recently, exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC-Exos) has been introduced as a promising treatment in organ fibrosis. This study aimed to explore the effects as well as the molecular mechanism for hucMSC-Exo in pulmonary fibrosis. The intravenous administration of hucMSC-Exos alleviated bleomycin-induced pulmonary fibrosis in vivo. Moreover, hucMSC-Exos elevated miR-218 expression and restored endothelial properties weakened by TGF-β in endothelial cells. Knockdown of miR-218 partially abrogated the inhibition effect of hucMSC-Exos on EndMT. Our mechanistic study further demonstrated that MeCP2 was the direct target of miR-218. Overexpressing MeCP2 aggravated EndMT and caused increased CpG islands methylation at BMP2 promoter, which lead to BMP2 post-transcriptional gene silence. Transfection of miR-218 mimic increased BMP2 expression as well, which was downregulated by overexpression of MeCP2. Taken together, these findings indicate exosomal miR-218 derived from hucMSCs may possess anti-fibrotic properties and inhibit EndMT through MeCP2/BMP2 pathway, providing a new avenue of preventive application in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuhao Zhao
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China
| | - Lei Du
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China
| | - Jiali Sun
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China
| | - Xuelian Wang
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China
| | - Zhilei Cong
- Department of Emergency, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Shuyan Chen
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China.
| | - Fei Wang
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China.
| | - Zhen Li
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, 200092, China.
| |
Collapse
|
5
|
Sharma G, Pund S, Govindan R, Nissa MU, Biswas D, Middha S, Ganguly K, Anand MP, Banerjee R, Srivastava S. A Proteomics Investigation of Cigarette Smoke Exposed Wistar Rats Revealed Improved Anti-Inflammatory Effects of the Cysteamine Nanoemulsions Delivered via Inhalation. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:338-360. [PMID: 37581495 DOI: 10.1089/omi.2023.0074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Cigarette smoking is the major cause of chronic inflammatory diseases such as chronic obstructive pulmonary disease (COPD). It is paramount to develop pharmacological interventions and delivery strategies against the cigarette smoke (CS) associated oxidative stress in COPD. This study in Wistar rats examined cysteamine in nanoemulsions to counteract the CS distressed microenvironment. In vivo, 28 days of CS and 15 days of cysteamine nanoemulsions treatment starting on 29th day consisting of oral and inhalation routes were established in Wistar rats. In addition, we conducted inflammatory and epithelial-to-mesenchymal transition (EMT) studies in vitro in human bronchial epithelial cell lines (BEAS2B) using 5% CS extract. Inflammatory and anti-inflammatory markers, such as tumor necrosis factor-alpha (TNF-α), interleukin (IL)-6, IL-1β, IL-8, IL-10, and IL-13, have been quantified in bronchoalveolar lavage fluid (BALF) to evaluate the effects of the cysteamine nanoemulsions in normalizing the diseased condition. Histopathological analysis of the alveoli and the trachea showed the distorted, lung parenchyma and ciliated epithelial barrier, respectively. To obtain mechanistic insights into the CS COPD rat model, "shotgun" proteomics of the lung tissues have been carried out using high-resolution mass spectrometry wherein genes such as ABI1, PPP3CA, PSMA2, FBLN5, ACTG1, CSNK2A1, and ECM1 exhibited significant differences across all the groups. Pathway analysis showed autophagy, signaling by receptor tyrosine kinase, cytokine signaling in immune system, extracellular matrix organization, and hemostasis, as the major contributing pathways across all the studied groups. This work offers new preclinical findings on how cysteamine taken orally or inhaled can combat CS-induced oxidative stress.
Collapse
Affiliation(s)
- Gautam Sharma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Swati Pund
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Biobay, Ahmedabad, India
| | - Rajkumar Govindan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- Department of Biomedical Engineering, Hajim School of Engineering & Applied Sciences, University of Rochester, Rochester, New York, USA
| | - Mehar Un Nissa
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Deeptarup Biswas
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sanniya Middha
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Koustav Ganguly
- Unit of Integrative Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
6
|
Basta MD, Petruk S, Mazo A, Walker JL. Fibrosis-the tale of H3K27 histone methyltransferases and demethylases. Front Cell Dev Biol 2023; 11:1193344. [PMID: 37476157 PMCID: PMC10354294 DOI: 10.3389/fcell.2023.1193344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023] Open
Abstract
Fibrosis, or excessive scarring, is characterized by the emergence of alpha-smooth muscle actin (αSMA)-expressing myofibroblasts and the excessive accumulation of fibrotic extracellular matrix (ECM). Currently, there is a lack of effective treatment options for fibrosis, highlighting an unmet need to identify new therapeutic targets. The acquisition of a fibrotic phenotype is associated with changes in chromatin structure, a key determinant of gene transcription activation and repression. The major repressive histone mark, H3K27me3, has been linked to dynamic changes in gene expression in fibrosis through alterations in chromatin structure. H3K27-specific homologous histone methylase (HMT) enzymes, Enhancer of zeste 1 and 2 (EZH1, EZH2), which are the alternative subunits of the Polycomb Repressive Complex 2 (PRC2) and demethylase (KDM) enzymes, Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX), and Lysine demethylase 6B (KDM6B), are responsible for regulating methylation status of H3K27me3. In this review, we explore how these key enzymes regulate chromatin structure to alter gene expression in fibrosis, highlighting them as attractive targets for the treatment of fibrosis.
Collapse
Affiliation(s)
- Morgan D. Basta
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Svetlana Petruk
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexander Mazo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Janice L. Walker
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Ophthalmology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
7
|
Duan J, Zhong B, Fan Z, Zhang H, Xu M, Zhang X, Sanders YY. DNA methylation in pulmonary fibrosis and lung cancer. Expert Rev Respir Med 2022; 16:519-528. [PMID: 35673969 DOI: 10.1080/17476348.2022.2085091] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Juan Duan
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Baiyun Zhong
- Department of Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhihua Fan
- Xiangya Medical school of Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Xiangya Medical school of Central South University, Changsha, Hunan, China
| | - Mengmeng Xu
- Xiangya Medical school of Central South University, Changsha, Hunan, China
| | - Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Y Sanders
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, 901 19 Street South, BMRII Room 408, Birmingham, AL 35294, USA
| |
Collapse
|
8
|
Xue T, Qiu X, Liu H, Gan C, Tan Z, Xie Y, Wang Y, Ye T. Epigenetic regulation in fibrosis progress. Pharmacol Res 2021; 173:105910. [PMID: 34562602 DOI: 10.1016/j.phrs.2021.105910] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/23/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Fibrosis, a common process of chronic inflammatory diseases, is defined as a repair response disorder when organs undergo continuous damage, ultimately leading to scar formation and functional failure. Around the world, fibrotic diseases cause high mortality, unfortunately, with limited treatment means in clinical practice. With the development and application of deep sequencing technology, comprehensively exploring the epigenetic mechanism in fibrosis has been allowed. Extensive remodeling of epigenetics controlling various cells phenotype and molecular mechanisms involved in fibrogenesis was subsequently verified. In this review, we summarize the regulatory mechanisms of DNA methylation, histone modification, noncoding RNAs (ncRNAs) and N6-methyladenosine (m6A) modification in organ fibrosis, focusing on heart, liver, lung and kidney. Additionally, we emphasize the diversity of epigenetics in the cellular and molecular mechanisms related to fibrosis. Finally, the potential and prospect of targeted therapy for fibrosis based on epigenetic is discussed.
Collapse
Affiliation(s)
- Taixiong Xue
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Qiu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyao Liu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cailing Gan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zui Tan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuting Xie
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuxi Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.
| | - Tinghong Ye
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
9
|
Abstract
Rett syndrome (RTT) is a severe X-linked neurodevelopmental disorder characterized by neurodevelopmental regression between 6 and 18 months of life and associated with multi-system comorbidities. Caused mainly by pathogenic variants in the MECP2 (methyl CpG binding protein 2) gene, it is the second leading genetic cause of intellectual disability in girls after Down syndrome. RTT affects not only neurological function but also a wide array of non-neurological organs. RTT-related disorders involve abnormalities of the respiratory, cardiovascular, digestive, metabolic, skeletal, endocrine, muscular, and urinary systems and immune response. Here, we review the different aspects of RTT affecting the main peripheral groups of organs and sometimes occurring independently of nervous system defects.
Collapse
Affiliation(s)
- Emilie Borloz
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385, Marseille, France
| | - Laurent Villard
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385, Marseille, France
| | - Jean-Christophe Roux
- Aix Marseille Univ, INSERM, MMG, U1251, Faculté de médecine Timone, 13385, Marseille, France
| |
Collapse
|
10
|
Alcaraz J, Ikemori R, Llorente A, Díaz-Valdivia N, Reguart N, Vizoso M. Epigenetic Reprogramming of Tumor-Associated Fibroblasts in Lung Cancer: Therapeutic Opportunities. Cancers (Basel) 2021; 13:cancers13153782. [PMID: 34359678 PMCID: PMC8345093 DOI: 10.3390/cancers13153782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Lung cancer is the leading cause of cancer death among both men and women, partly due to limited therapy responses. New avenues of knowledge are indicating that lung cancer cells do not form a tumor in isolation but rather obtain essential support from their surrounding host tissue rich in altered fibroblasts. Notably, there is growing evidence that tumor progression and even the current limited responses to therapies could be prevented by rescuing the normal behavior of fibroblasts, which are critical housekeepers of normal tissue function. For this purpose, it is key to improve our understanding of the molecular mechanisms driving the pathologic alterations of fibroblasts in cancer. This work provides a comprehensive review of the main molecular mechanisms involved in fibroblast transformation based on epigenetic reprogramming, and summarizes emerging therapeutic approaches to prevent or overcome the pathologic effects of tumor-associated fibroblasts. Abstract Lung cancer is the leading cause of cancer-related death worldwide. The desmoplastic stroma of lung cancer and other solid tumors is rich in tumor-associated fibroblasts (TAFs) exhibiting an activated/myofibroblast-like phenotype. There is growing awareness that TAFs support key steps of tumor progression and are epigenetically reprogrammed compared to healthy fibroblasts. Although the mechanisms underlying such epigenetic reprogramming are incompletely understood, there is increasing evidence that they involve interactions with either cancer cells, pro-fibrotic cytokines such as TGF-β, the stiffening of the surrounding extracellular matrix, smoking cigarette particles and other environmental cues. These aberrant interactions elicit a global DNA hypomethylation and a selective transcriptional repression through hypermethylation of the TGF-β transcription factor SMAD3 in lung TAFs. Likewise, similar DNA methylation changes have been reported in TAFs from other cancer types, as well as histone core modifications and altered microRNA expression. In this review we summarize the evidence of the epigenetic reprogramming of TAFs, how this reprogramming contributes to the acquisition and maintenance of a tumor-promoting phenotype, and how it provides novel venues for therapeutic intervention, with a special focus on lung TAFs.
Collapse
Affiliation(s)
- Jordi Alcaraz
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain; (R.I.); (A.L.); (N.D.-V.)
- Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain;
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Spain
- Correspondence: (J.A.); (M.V.)
| | - Rafael Ikemori
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain; (R.I.); (A.L.); (N.D.-V.)
| | - Alejandro Llorente
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain; (R.I.); (A.L.); (N.D.-V.)
| | - Natalia Díaz-Valdivia
- Unit of Biophysics and Bioengineering, Department of Biomedicine, School of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain; (R.I.); (A.L.); (N.D.-V.)
| | - Noemí Reguart
- Thoracic Oncology Unit, Hospital Clinic Barcelona, 08036 Barcelona, Spain;
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Miguel Vizoso
- Division of Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
- Correspondence: (J.A.); (M.V.)
| |
Collapse
|
11
|
Suri GS, Kaur G, Jha CK, Tiwari M. Understanding idiopathic pulmonary fibrosis - Clinical features, molecular mechanism and therapies. Exp Gerontol 2021; 153:111473. [PMID: 34274426 DOI: 10.1016/j.exger.2021.111473] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 10/20/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic lung fibrosing disease with high prevalence that has a prognosis worse than many cancers. There has been a recent influx of new observations aimed at explaining the mechanisms responsible for the initiation and progression of pulmonary fibrosis. However, despite this, the pathogenesis of the disease is largely unclear. Recent progress has been made in the characterization of specific pathologic and clinical features that have enhanced the understanding of pathologically activated molecular pathways during the onset and progression of IPF. This review highlights several of the advances that have been made and focus on the pathobiology of IPF. The work also details the different factors that are responsible for the disposition of the disease - these may be internal factors such as cellular mechanisms and genetic alterations, or they may be external factors from the environment. The changes that primarily occur in epithelial cells and fibroblasts that lead to the activation of profibrotic pathways are discussed in depth. Finally, a complete repertoire of the treatment therapies that have been used in the past as well as future medications and therapies is provided.
Collapse
|
12
|
Li X, Yang Y, Chen S, Zhou J, Li J, Cheng Y. Epigenetics-based therapeutics for myocardial fibrosis. Life Sci 2021; 271:119186. [PMID: 33577852 DOI: 10.1016/j.lfs.2021.119186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 02/07/2023]
Abstract
Myocardial fibrosis (MF) is a reactive remodeling process in response to myocardial injury. It is mainly manifested by the proliferation of cardiac muscle fibroblasts and secreting extracellular matrix (ECM) proteins to replace damaged tissue. However, the excessive production and deposition of extracellular matrix, and the rising proportion of type I and type III collagen lead to pathological fibrotic remodeling, thereby facilitating the development of cardiac dysfunction and eventually causing heart failure with heightened mortality. Currently, the molecular mechanisms of MF are still not fully understood. With the development of epigenetics, it is found that epigenetics controls the transcription of pro-fibrotic genes in MF by DNA methylation, histone modification and noncoding RNAs. In this review, we summarize and discuss the research progress of the mechanisms underlying MF from the perspective of epigenetics, including the newest m6A modification and crosstalk between different epigenetics in MF. We also offer a succinct overview of promising molecules targeting epigenetic regulators, which may provide novel therapeutic strategies against MF.
Collapse
Affiliation(s)
- Xuping Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Ying Yang
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Sixuan Chen
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jiuyao Zhou
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Jingyan Li
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Yuanyuan Cheng
- School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| |
Collapse
|
13
|
Ganguly N, Chakrabarti S. Role of long non‑coding RNAs and related epigenetic mechanisms in liver fibrosis (Review). Int J Mol Med 2021; 47:23. [PMID: 33495817 PMCID: PMC7846421 DOI: 10.3892/ijmm.2021.4856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023] Open
Abstract
Liver fibrosis is one of the major liver pathologies affecting patients worldwide. It results from an improper tissue repair process following liver injury or inflammation. If left untreated, it ultimately leads to liver cirrhosis and liver failure. Long non‑coding RNAs (lncRNAs) have been implicated in a wide variety of diseases. They can regulate gene expression and modulate signaling. Some of the lncRNAs promote, while others inhibit liver fibrosis. Similarly, other epigenetic processes, such as methylation and acetylation regulate gene transcription and can modulate gene expression. Notably, there are several regulatory associations of lncRNAs with other epigenetic processes. A major mechanism of action of long non‑coding RNAs is to competitively bind to their target microRNAs (miRNAs or miRs), which in turn affects miRNA availability and bioactivity. In the present review, the role of lncRNAs and related epigenetic processes contributing to liver fibrosis is discussed. Finally, various potential therapeutic approaches targeting lncRNAs and related epigenetic processes, which are being considered as possible future treatment targets for liver fibrosis are identified.
Collapse
Affiliation(s)
- Niladri Ganguly
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
14
|
Li D, Yang E, Zhao J, Zhang H. Association between MeCP2 and Smad7 in the pathogenesis and development of pathological scars. J Plast Surg Hand Surg 2021; 55:284-293. [PMID: 33475023 DOI: 10.1080/2000656x.2021.1874399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
To explore the relationship between methylated binding protein 2 (MeCP2) and mothers against decapentaplegic homolog 7 (Smad7) in the pathogenesis and development of pathological scars. Immunohistochemistry, Western blot and real-time polymerase chain reaction (RT-PCR) were used to detect the expression of MeCP2 in different types of human scars and hypertrophic scars at different growth times. The methylation status of Smad7 gene promoter in different scar tissues was determined by methylation-specific PCR. After transfection with MeCP2-siRNA (small interfering RNA) in human keloid fibroblasts, MTT assay was used to assess the proliferation activity of keloid fibroblasts, while RT-PCR and Western blot assays were used to detect the expression levels of MeCP2, transforming growth factor-β1 (TGF-β1), α-smooth muscle actin (α-SMA), phospho-Smad2 (p-Smad2) and Smad7. MeCP2 was mainly expressed in the nucleus of fibroblasts. The mRNA and protein levels of MeCP2 were significantly higher in keloids than in hypertrophic scars, normal scars and normal skin (p<.05). The expression level of MeCP2 in hypertrophic scars during the growth period of <6 months was markedly higher than that of >6 months (p<.05). The methylation level of Smad7 was significantly higher in keloids compared to normal skin. After MeCP2 silencing, the proliferation rate of human keloid fibroblasts was decreased, the mRNA and protein levels of Smad7 were increased, and the expression levels of TGF-β1, α-SMA and p-Smad2 were decreased (p<.05). MeCP2 and Smad7 play an important role in formation of pathological scars. During keloid formation, MeCP2 weakens the inhibitory effect of Smad7 on p-Smad2/3 by downregulating the expression of Smad7, which in turn promotes fibrosis and scar hyperplasia.
Collapse
Affiliation(s)
- Dan Li
- Department of Plastic and Burn Surgery, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - E Yang
- Department of Plastic and Burn Surgery, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Juan Zhao
- Department of Plastic and Burn Surgery, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| | - Hengshu Zhang
- Department of Plastic and Burn Surgery, Chongqing Medical University First Affiliated Hospital, Chongqing, China
| |
Collapse
|
15
|
Emerging physiological and pathological roles of MeCP2 in non-neurological systems. Arch Biochem Biophys 2021; 700:108768. [PMID: 33485848 DOI: 10.1016/j.abb.2021.108768] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 01/08/2021] [Accepted: 01/14/2021] [Indexed: 02/08/2023]
Abstract
Numerous neurological and non-neurological disorders are associated with dysfunction of epigenetic modulators, and methyl CpG binding protein 2 (MeCP2) is one of such proteins. Initially identified as a transcriptional repressor, MeCP2 specifically binds to methylated DNA, and mutations of MeCP2 have been shown to cause Rett syndrome (RTT), a severe neurological disorder. Recently, accumulating evidence suggests that ubiquitously expressed MeCP2 also plays a central role in non-neurological disorders including cardiac dysfunction, liver injury, respiratory disorders, urological dysfunction, adipose tissue metabolism disorders, movement abnormality and inflammatory responses in a DNA methylation dependent or independent manner. Despite significant progresses in our understanding of MeCP2 over the last few decades, there is still a considerable knowledge gap to translate the in vitro and in vivo experimental findings into therapeutic interventions. In this review, we provide a synopsis of the role of MeCP2 in the pathophysiology of non-neurological disorders, MeCP2-based research directions and therapeutic strategies for non-neurological disorders are also discussed.
Collapse
|
16
|
Wang Y, Zhang L, Wu GR, Zhou Q, Yue H, Rao LZ, Yuan T, Mo B, Wang FX, Chen LM, Sun F, Song J, Xiong F, Zhang S, Yu Q, Yang P, Xu Y, Zhao J, Zhang H, Xiong W, Wang CY. MBD2 serves as a viable target against pulmonary fibrosis by inhibiting macrophage M2 program. SCIENCE ADVANCES 2021; 7:eabb6075. [PMID: 33277324 PMCID: PMC7775789 DOI: 10.1126/sciadv.abb6075] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 10/29/2020] [Indexed: 05/22/2023]
Abstract
Despite past extensive studies, the mechanisms underlying pulmonary fibrosis (PF) still remain poorly understood. Here, we demonstrated that lungs originating from different types of patients with PF, including coronavirus disease 2019, systemic sclerosis-associated interstitial lung disease, and idiopathic PF, and from mice following bleomycin (BLM)-induced PF are characterized by the altered methyl-CpG-binding domain 2 (MBD2) expression in macrophages. Depletion of Mbd2 in macrophages protected mice against BLM-induced PF. Mbd2 deficiency significantly attenuated transforming growth factor-β1 (TGF-β1) production and reduced M2 macrophage accumulation in the lung following BLM induction. Mechanistically, Mbd2 selectively bound to the Ship promoter in macrophages, by which it repressed Ship expression and enhanced PI3K/Akt signaling to promote the macrophage M2 program. Therefore, intratracheal administration of liposomes loaded with Mbd2 siRNA protected mice from BLM-induced lung injuries and fibrosis. Together, our data support the possibility that MBD2 could be a viable target against PF in clinical settings.
Collapse
Affiliation(s)
- Yi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Lei Zhang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Guo-Rao Wu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Qing Zhou
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Huihui Yue
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Li-Zong Rao
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, 212 Renmin Road, Guilin 541000, China
| | - Ting Yuan
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, 212 Renmin Road, Guilin 541000, China
| | - Biwen Mo
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, 212 Renmin Road, Guilin 541000, China
| | - Fa-Xi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Long-Min Chen
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Fei Sun
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Jia Song
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Fei Xiong
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Shu Zhang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Qilin Yu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Ping Yang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Yongjian Xu
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Jianping Zhao
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China
| | - Huilan Zhang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China.
| | - Weining Xiong
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China.
- Department of Respiratory Medicine, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, 639 Zhizaoju Lu, Shanghai 200011, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, 1095 Jiefang Ave., Wuhan 430030, China.
| |
Collapse
|
17
|
Bartczak K, Białas AJ, Kotecki MJ, Górski P, Piotrowski WJ. More than a Genetic Code: Epigenetics of Lung Fibrosis. Mol Diagn Ther 2020; 24:665-681. [PMID: 32926347 PMCID: PMC7677145 DOI: 10.1007/s40291-020-00490-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
At the end of the last century, genetic studies reported that genetic information is not transmitted solely by DNA, but is also transmitted by other mechanisms, named as epigenetics. The well-described epigenetic mechanisms include DNA methylation, biochemical modifications of histones, and microRNAs. The role of altered epigenetics in the biology of various fibrotic diseases is well-established, and recent advances demonstrate its importance in the pathogenesis of pulmonary fibrosis-predominantly referring to idiopathic pulmonary fibrosis, the most lethal of the interstitial lung diseases. The deficiency in effective medications suggests an urgent need to better understand the underlying pathobiology. This review summarizes the current knowledge concerning epigenetic changes in pulmonary fibrosis and associations of these changes with several cellular pathways of known significance in its pathogenesis. It also designates the most promising substances for further research that may bring us closer to new therapeutic options.
Collapse
Affiliation(s)
- Krystian Bartczak
- Department of Pneumology and Allergology, The Medical University of Lodz, Kopcińskiego 22, 90-153, Lodz, Poland.
| | - Adam J Białas
- Department of Pathobiology of Respiratory Diseases, The Medical University of Lodz, Lodz, Poland
| | - Mateusz J Kotecki
- Department of Pneumology and Allergology, The Medical University of Lodz, Kopcińskiego 22, 90-153, Lodz, Poland
| | - Paweł Górski
- Department of Pneumology and Allergology, The Medical University of Lodz, Kopcińskiego 22, 90-153, Lodz, Poland
| | - Wojciech J Piotrowski
- Department of Pneumology and Allergology, The Medical University of Lodz, Kopcińskiego 22, 90-153, Lodz, Poland
| |
Collapse
|
18
|
Casanova EL, Baeza-Velasco C, Buchanan CB, Casanova MF. The Relationship between Autism and Ehlers-Danlos Syndromes/Hypermobility Spectrum Disorders. J Pers Med 2020; 10:E260. [PMID: 33271870 PMCID: PMC7711487 DOI: 10.3390/jpm10040260] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 12/12/2022] Open
Abstract
Considerable interest has arisen concerning the relationship between hereditary connective tissue disorders such as the Ehlers-Danlos syndromes (EDS)/hypermobility spectrum disorders (HSD) and autism, both in terms of their comorbidity as well as co-occurrence within the same families. This paper reviews our current state of knowledge, as well as highlighting unanswered questions concerning this remarkable patient group, which we hope will attract further scientific interest in coming years. In particular, patients themselves are demanding more research into this growing area of interest, although science has been slow to answer that call. Here, we address the overlap between these two spectrum conditions, including neurobehavioral, psychiatric, and neurological commonalities, shared peripheral neuropathies and neuropathologies, and similar autonomic and immune dysregulation. Together, these data highlight the potential relatedness of these two conditions and suggest that EDS/HSD may represent a subtype of autism.
Collapse
Affiliation(s)
- Emily L. Casanova
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29615, USA;
| | - Carolina Baeza-Velasco
- Laboratory of Psychopathology and Health Processes, University of Paris, 92100 Boulogne Billancourt, France;
- Department of Emergency Psychiatry and Acute Care, CHU Montpellier, 34000 Montpellier, France
| | | | - Manuel F. Casanova
- School of Medicine Greenville, University of South Carolina, Greenville, SC 29615, USA;
- Department of Psychiatry and Behavioral Sciences, University of Louisville, Louisville, KY 40292, USA
| |
Collapse
|
19
|
Ramahi A, Altorok N, Kahaleh B. Epigenetics and systemic sclerosis: An answer to disease onset and evolution? Eur J Rheumatol 2020; 7:S147-S156. [PMID: 32697935 PMCID: PMC7647676 DOI: 10.5152/eurjrheum.2020.19112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/06/2020] [Indexed: 12/22/2022] Open
Abstract
There is growing evidence that implicates epigenetic modification in the pathogenesis of systemic sclerosis (SSc). The complexity of epigenetic regulation and its dynamic nature complicate the investigation of its role in the disease. We will review the current literature for factors that link epigenetics to SSc by discussing DNA methylation, histone acetylation and methylation, and non-coding RNAs (ncRNAs), particularly microRNA changes in endothelial cells, fibroblasts (FBs), and lymphocytes. These three cell types are significantly involved in the early stages and throughout the course of the disease and are particularly vulnerable to epigenetic regulation. The pathogenesis of SSc is likely related to modifications of the epigenome by environmental signals in individuals with a specific genetic makeup. The epigenome is an attractive therapeutic target; however, successful epigenetics-based treatments require a better understanding of the molecular mechanisms controlling the epigenome and its alteration in the disease.
Collapse
Affiliation(s)
- Ahmad Ramahi
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| | - Nezam Altorok
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| | - Bashar Kahaleh
- Division of Rheumatology and Immunology, Department of Internal Medicine, University of Toledo Medical Center, Toledo, OH, USA
| |
Collapse
|
20
|
Research Advances on DNA Methylation in Idiopathic Pulmonary Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1255:73-81. [PMID: 32949391 DOI: 10.1007/978-981-15-4494-1_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic complex lung disease with no specific treatment and poor prognosis, characterized by the pulmonary progressive fibrosis and dysfunctions that lead to respiratory failure. Several factors may impact the progress of IPF, including age, cigarette smoking, and dusts, of which genetic and epigenetic factors mainly contribute to lung tissue fibrosis. DNA methylation is one of epigenetic processes that occur in many diseases and regulate chromosomal and extrachromosomal DNA functions in response to environmental exposures. The methylation plays pivotal roles in regulation of gene expression to facilitate the formation of fibroblastic foci and lung fibrosis. This chapter will describe alterations and effects of the DNA methylation on gene expression, the potential application of DNA methylation as a biomarker, and significance as therapeutic targets. Those understanding will provide us new insight into the treatment and prognosis of IPF.
Collapse
|
21
|
Abstract
Fibrosis is characterized by aberrant myofibroblast accumulation and excessive extracellular matrix deposition, which leads to organ failure and significantly contributes to mortality worldwide. Exosomes, which are extracellular nanovesicles with a diameter of 30-100 nm that are secreted into the extracellular space by various types of cells, facilitate intercellular communication by delivering different cargos such as proteins, mRNAs and microRNAs. Growing evidence indicates that exosomes play an important role in various fibrotic diseases. A deeper understanding of the effects of exosomes in fibrosis may help in exploring new diagnostic and therapeutic targets. In this review, we summarize recent findings on exosomes in fibrotic diseases, with a special focus on exosomal cargo dysregulation and their potential diagnostic and therapeutic value in fibrosis.
Collapse
Affiliation(s)
- Xi-Ji Qin
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jia-Xiang Zhang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Rui-Lan Wang
- Department of Critical Care Medicine, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Li X, Li X, He S, Zhao M. MeCP2-421-mediated RPE epithelial-mesenchymal transition and its relevance to the pathogenesis of proliferative vitreoretinopathy. J Cell Mol Med 2020; 24:9420-9427. [PMID: 32638535 PMCID: PMC7417696 DOI: 10.1111/jcmm.15602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 06/13/2020] [Indexed: 12/17/2022] Open
Abstract
Proliferative vitreoretinopathy (PVR) is a blinding eye disease. Epithelial‐mesenchymal transition (EMT) of RPE cells plays an important role in the pathogenesis of PVR. In the current study, we sought to investigate the role of the methyl‐CpG‐binding protein 2 (MeCP2), especially P‐MeCP2‐421 in the pathogenesis of PVR. The expressions of P‐MeCP2‐421, P‐MeCP2‐80, PPAR‐γ and the double labelling of P‐MeCP2‐421 with α‐SMA, cytokeratin, TGF‐β and PPAR‐γ in human PVR membranes were analysed by immunohistochemistry. The effect of knocking down MeCP2 using siRNA on the expressions of α‐SMA, phospho‐Smad2/3, collagen I, fibronectin and PPAR‐γ; the expression of α‐SMA stimulated by recombinant MeCP2 in ARPE‐19; and the effect of TGF‐β and 5‐AZA treatment on PPAR‐γ expression were analysed by Western blot. Chromatin immunoprecipitation was used to determine the binding of MeCP2 to TGF‐β. Our results showed that P‐MeCP2‐421 was highly expressed in PVR membranes and was double labelled with α‐SMA, cytokeratin and TGF‐β, knocking down MeCP2 inhibited the activation of Smad2/3 and the expression of collagen I and fibronectin induced by TGF‐β. TGF‐β inhibited the expression of PPAR‐γ, silence of MeCP2 by siRNA or using MeCP2 inhibitor (5‐AZA) increased the expression of PPAR‐γ. α‐SMA was up‐regulated by the treatment of recombinant MeCP2. Importantly, we found that MeCP2 bound to TGF‐β as demonstrated by Chip assay. The results suggest that MeCP2 especially P‐MeCP2‐421 may play a significant role in the pathogenesis of PVR and targeting MeCP2 may be a potential therapeutic approach for the treatment of PVR.
Collapse
Affiliation(s)
- Xiaohua Li
- Henan Provincial People's Hospital, Zhengzhou, China.,Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, China.,People's Hospital of Zhengzhou University, Zhengzhou, China.,People's Hospital of Henan University, Zhengzhou, China
| | - Xue Li
- Henan Provincial People's Hospital, Zhengzhou, China.,Henan Eye Hospital, Henan Eye Institute, Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, China.,People's Hospital of Zhengzhou University, Zhengzhou, China.,People's Hospital of Henan University, Zhengzhou, China
| | - Shikun He
- Ophthalmology Optometry Centre, Peking University People's Hospital, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China.,Departments of Pathology, USC Roski Eye Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mingwei Zhao
- Ophthalmology Optometry Centre, Peking University People's Hospital, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, Beijing, China
| |
Collapse
|
23
|
Xiang Z, Zhou Q, Hu M, Sanders YY. MeCP2 epigenetically regulates alpha-smooth muscle actin in human lung fibroblasts. J Cell Biochem 2020; 121:3616-3625. [PMID: 32115750 DOI: 10.1002/jcb.29655] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/18/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND A critical feature for fibroblasts differentiation into myofibroblasts is the expression of alpha-smooth muscle actin (α-SMA) during the tissue injury and repair process. The epigenetic mechanism, DNA methylation, is involved in regulating α-SMA expression. It is not clear how methyl-CpG-binding protein 2 (MeCP2) interacts with CpG-rich region in α-SMA, and if the CpG methylation status would affect MeCP2 binding and regulation of α-SMA expression. METHODS The association of MeCP2 with α-SMA CpG rich region were examined by chromatin immunoprecipitation (ChIP) assays in primary fibroblasts from idiopathic pulmonary fibrosis (IPF) and non-IPF control individuals, and in the lung fibroblasts treated with profibrotic cytokine transforming growth factor β1 (TGF-β1). The regulation of α-SMA by MeCP2 was examined by knocking down MeCP2 with small interfering RNA (siRNA). To explore the effects of the DNA methylation status of the CpG rich region on α-SMA expression, the cells were treated with DNA methyltransferase inhibitor, 5'-azacytidine (5'-aza). The expression of α-SMA was examined by Western blot and quantitative polymerase chain reaction, the association with MeCP2 was assessed by ChIP assays, and the methylation status was checked by bisulfate sequencing. RESULTS The human lung fibroblasts with increased α-SMA showed an enriched association of MeCP2, while knockdown MeCP2 by siRNA reduced α-SMA upregulation by TGF-β1. The 5'-Aza-treated cells have decreased α-SMA expression with reduced MeCP2 association. However, bisulfite sequencing revealed that most CpG sites are unmethylated despite the different expression levels of α-SMA after being treated by TGF-β1 or 5'-aza. CONCLUSION Our data indicate that the methyl-binding protein MeCP2 is critical for α-SMA expression in human lung myofibroblast, and the DNA methylation status at the CpG rich region of α-SMA is not a determinative factor for its inducible expression.
Collapse
Affiliation(s)
- Zheyi Xiang
- Laboratory of Clinical Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.,Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Qingxian Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Min Hu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yan Y Sanders
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
24
|
Zhu Z, Hou Q, Li M, Fu X. Molecular mechanism of myofibroblast formation and strategies for clinical drugs treatments in hypertrophic scars. J Cell Physiol 2019; 235:4109-4119. [PMID: 31612497 DOI: 10.1002/jcp.29302] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/30/2019] [Indexed: 12/14/2022]
Abstract
Hypertrophic scars (HTS) commonly occurred after burn and trauma. It was characterized by the excessive deposition of extracellular matrix with the inadequate remodeling, which could result in severe physiological and psychological problems. However, the effective available prevention and treatment measures were still limited. The main pathological feature of HTS was the excessive formation of myofibroblasts, and they persist in the repaired tissue. To better understand the mechanics of this process, this review focused on the characteristics and formation of myofibroblasts, the main effector cells in HTS. We summarized the present theories and opinions on myofibroblasts formation from the perspective of related signaling pathways and epigenetic regulation, such as DNA methylation, miRNA/lncRNA/ceRNA action, histone modification, and so forth for a better understanding on the development of HTS. This information might assist in developing effective experimental and clinical treatment strategies. Additionally, we also summarized currently known clinical strategies for HTS treatment, including traditional drugs, molecular medicine, stem cells, and exosomes.
Collapse
Affiliation(s)
- Ziying Zhu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Qian Hou
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Meirong Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China.,Central Laboratory, Trauma Treatment Center, Central Laboratory, Chinese PLA General Hospital Hainan Branch, Sanya, China
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Zhang N, Liu K, Wang K, Zhou C, Wang H, Che S, Liu Z, Yang H. Dust induces lung fibrosis through dysregulated DNA methylation. ENVIRONMENTAL TOXICOLOGY 2019; 34:728-741. [PMID: 30815999 DOI: 10.1002/tox.22739] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/28/2019] [Accepted: 02/07/2019] [Indexed: 06/09/2023]
Abstract
Pneumoconiosis is a serious occupational disease that often occurs to coal workers with no early diagnosis and effective treatment at present. Diffuse pulmonary fibrosis is the major pathological change of pneumoconiosis, and its mechanism is still unclear. Epigenetics is involved in the development of many diseases, and it is closely associated with fibrosis. In this study, we investigated whether DNA methylation contributes to the pathogenesis of pulmonary fibrosis in pneumoconiosis. By exposure to coal dust or silica dust, we established the models of coal worker's pneumoconiosis (CWP), which showed an increased expression of COL-I, COL-III. We further found that DNMT1, DNMT3a, DNMT3b, MBD2, MeCP2 protein expression changed. Pretreatment with DNMT inhibitor 5-aza-dC reduced expression of COL-I, COL-III, and reduced pulmonary fibrosis. In summary, our results showed that DNA methylation contributes to dust-induced pulmonary fibrosis and that it may serve as a theoretical basis for testing DNA methyltransferase inhibitors in the treatment of CWP.
Collapse
Affiliation(s)
- Na Zhang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Keliang Liu
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Kai Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Ci Zhou
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Hejing Wang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Shuangshuang Che
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Zhihong Liu
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Huifang Yang
- Department of Occupational and Environmental Health, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
26
|
Barcena-Varela M, Colyn L, Fernandez-Barrena MG. Epigenetic Mechanisms in Hepatic Stellate Cell Activation During Liver Fibrosis and Carcinogenesis. Int J Mol Sci 2019; 20:E2507. [PMID: 31117267 PMCID: PMC6566358 DOI: 10.3390/ijms20102507] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/17/2019] [Accepted: 05/19/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is an essential component of chronic liver disease (CLD) and hepatocarcinogenesis. The fibrotic stroma is a consequence of sustained liver damage combined with exacerbated extracellular matrix (ECM) accumulation. In this context, activation of hepatic stellate cells (HSCs) plays a key role in both initiation and perpetuation of fibrogenesis. These cells suffer profound remodeling of gene expression in this process. This review is focused on the epigenetic alterations participating in the transdifferentiation of HSCs from the quiescent to activated state. Recent advances in the field of DNA methylation and post-translational modifications (PTM) of histones (acetylation and methylation) patterns are discussed here, together with altered expression and activity of epigenetic remodelers. We also consider recent advances in translational approaches, including the use of epigenetic marks as biomarkers and the promising antifibrotic properties of epigenetic drugs that are currently being used in patients.
Collapse
Affiliation(s)
| | - Leticia Colyn
- Hepatology Program, CIMA, University of Navarra, 31180 Pamplona, Spain.
| | - Maite G Fernandez-Barrena
- Hepatology Program, CIMA, University of Navarra, 31180 Pamplona, Spain.
- CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Instituto de Investigaciones Sanitarias de Navarra-IdiSNA, 31180 Pamplona, Spain.
| |
Collapse
|
27
|
Maria ATJ, Partouche L, Goulabchand R, Rivière S, Rozier P, Bourgier C, Le Quellec A, Morel J, Noël D, Guilpain P. Intriguing Relationships Between Cancer and Systemic Sclerosis: Role of the Immune System and Other Contributors. Front Immunol 2019; 9:3112. [PMID: 30687318 PMCID: PMC6335319 DOI: 10.3389/fimmu.2018.03112] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/17/2018] [Indexed: 01/09/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disorder, characterized by multisystem involvement, vasculopathy, and fibrosis. An increased risk of malignancy is observed in SSc (including breast and lung cancers), and in a subgroup of patients with specific autoantibodies (i.e., anti-RNA polymerase III and related autoantibodies), SSc could be a paraneoplastic syndrome and might be directly related to an immune response against cancer. Herein, we reviewed the literature, focusing on the most recent articles, and shed light onto the potential relationship between cancer and scleroderma regarding temporal and immunological dimensions.
Collapse
Affiliation(s)
- Alexandre Thibault Jacques Maria
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Léo Partouche
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Radjiv Goulabchand
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Sophie Rivière
- Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Pauline Rozier
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Céline Bourgier
- Medical School, Montpellier University, Montpellier, France.,Department of Radiation Oncology, INSERM U1194/IRCM, ICM-Val d'Aurelle, Montpellier, France
| | - Alain Le Quellec
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France
| | - Jacques Morel
- Medical School, Montpellier University, Montpellier, France.,Department of Rheumatology, Lapeyronie Hospital, Montpellier, France
| | - Danièle Noël
- IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| | - Philippe Guilpain
- Medical School, Montpellier University, Montpellier, France.,Department of Internal Medicine-Multiorganic Diseases, Local Referral Center for Auto-immune Diseases, Saint-Eloi Hospital, Montpellier University, Montpellier, France.,IRMB, INSERM, CHU Montpellier, Montpellier University, Montpellier, France
| |
Collapse
|
28
|
Li Z, Song S, Zha S, Wang C, Chen S, Wang F. MeCP2 promotes endothelial-to-mesenchymal transition in human endothelial cells by downregulating BMP7 expression. Exp Cell Res 2018; 375:82-89. [PMID: 30597142 DOI: 10.1016/j.yexcr.2018.12.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 12/21/2018] [Accepted: 12/27/2018] [Indexed: 01/03/2023]
Abstract
Endothelial-to-mesenchymal transition (EndMT) plays a pivotal role in the development of organ fibrosis and can be induced by TGF-β. It is characterized by the loss of endothelial cell markers and the acquisition of mesenchymal markers. In this study, we found that methyl-CpG binding protein 2 (MeCP2) was increased in TGF-β-induced EndMT, and silencing of MeCP2 inhibited EndMT induction. Viral overexpression of MeCP2 in vitro promoted EndMT and suppressed the expression of bone morphogenic protein-7 (BMP7). The methylation of CpG islands in BMP7 promoter was increased in MeCP2-overexpressing endothelial cells and Chromatin immunoprecipitation assay showed the direct binding of MeCP2 at the BMP7 promoter. In summary, our results suggest that MeCP2 promotes EndMT by epigenetically silencing BMP7 in endothelial cells and MeCP2 may be a target for diseases driven by EndMT.
Collapse
Affiliation(s)
- Zhen Li
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, China.
| | - Shuai Song
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siyuan Zha
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, China
| | - Chunli Wang
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, China
| | - Shuyan Chen
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, China.
| | - Fei Wang
- Department of Geriatrics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kong Jiang Road, Shanghai, China.
| |
Collapse
|
29
|
Bijkerk R, Trimpert C, van Solingen C, de Bruin RG, Florijn BW, Kooijman S, van den Berg R, van der Veer EP, Bredewold EOW, Rensen PCN, Rabelink TJ, Humphreys BD, Deen PMT, van Zonneveld AJ. MicroRNA-132 controls water homeostasis through regulating MECP2-mediated vasopressin synthesis. Am J Physiol Renal Physiol 2018; 315:F1129-F1138. [DOI: 10.1152/ajprenal.00087.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fine-tuning of the body’s water balance is regulated by vasopressin (AVP), which induces the expression and apical membrane insertion of aquaporin-2 water channels and subsequent water reabsorption in the kidney. Here we demonstrate that silencing of microRNA-132 (miR-132) in mice causes severe weight loss due to acute diuresis coinciding with increased plasma osmolality, reduced renal total and plasma membrane expression of aquaporin-2, and abrogated increase in AVP levels. Infusion with synthetic AVP fully reversed the antagomir-132-induced diuresis, and low-dose intracerebroventricular administration of antagomir-132 similarly caused acute diuresis. Central and intracerebroventricular antagomir-132 injection both decreased hypothalamic AVP mRNA levels. At the molecular level, antagomir-132 increased the in vivo and in vitro mRNA expression of methyl-CpG-binding protein-2 (MECP2), which is a miR-132 target and which blocks AVP gene expression by binding its enhancer region. In line with this, treatment of hypothalamic N6 cells with a high-salt solution increased its miR-132 levels, whereas it attenuated endogenous Mecp2 mRNA levels. In conclusion, we identified miR-132 as a first miRNA regulating the osmotic balance by regulating the hypothalamic AVP gene mRNA expression.
Collapse
Affiliation(s)
- Roel Bijkerk
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Renal Division, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Christiane Trimpert
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Coen van Solingen
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Medical Center, New York, New York
| | - Ruben G. de Bruin
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Barend W. Florijn
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Sander Kooijman
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Rosa van den Berg
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric P. van der Veer
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Edwin O. W. Bredewold
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Patrick C. N. Rensen
- Department of Internal Medicine (Endocrinology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Benjamin D. Humphreys
- Renal Division, Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Renal Division, Washington University School of Medicine, St. Louis, Missouri
| | - Peter M. T. Deen
- Department of Physiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
He Y, Tsou PS, Khanna D, Sawalha AH. Methyl-CpG-binding protein 2 mediates antifibrotic effects in scleroderma fibroblasts. Ann Rheum Dis 2018; 77:1208-1218. [PMID: 29760157 PMCID: PMC7297461 DOI: 10.1136/annrheumdis-2018-213022] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/19/2018] [Accepted: 04/21/2018] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Emerging evidence supports a role for epigenetic regulation in the pathogenesis of scleroderma (SSc). We aimed to assess the role of methyl-CpG-binding protein 2 (MeCP2), a key epigenetic regulator, in fibroblast activation and fibrosis in SSc. METHODS Dermal fibroblasts were isolated from patients with diffuse cutaneous SSc (dcSSc) and from healthy controls. MeCP2 expression was measured by qPCR and western blot. Myofibroblast differentiation was evaluated by gel contraction assay in vitro. Fibroblast proliferation was analysed by ki67 immunofluorescence staining. A wound healing assay in vitro was used to determine fibroblast migration rates. RNA-seq was performed with and without MeCP2 knockdown in dcSSc to identify MeCP2-regulated genes. The expression of MeCP2 and its targets were modulated by siRNA or plasmid. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) using anti-MeCP2 antibody was performed to assess MeCP2 binding sites within MeCP2-regulated genes. RESULTS Elevated expression of MeCP2 was detected in dcSSc fibroblasts compared with normal fibroblasts. Overexpressing MeCP2 in normal fibroblasts suppressed myofibroblast differentiation, fibroblast proliferation and fibroblast migration. RNA-seq in MeCP2-deficient dcSSc fibroblasts identified MeCP2-regulated genes involved in fibrosis, including PLAU, NID2 and ADA. Plasminogen activator urokinase (PLAU) overexpression in dcSSc fibroblasts reduced myofibroblast differentiation and fibroblast migration, while nidogen-2 (NID2) knockdown promoted myofibroblast differentiation and fibroblast migration. Adenosine deaminase (ADA) depletion in dcSSc fibroblasts inhibited cell migration rates. Taken together, antifibrotic effects of MeCP2 were mediated, at least partly, through modulating PLAU, NID2 and ADA. ChIP-seq further showed that MeCP2 directly binds regulatory sequences in NID2 and PLAU gene loci. CONCLUSIONS This study demonstrates a novel role for MeCP2 in skin fibrosis and identifies MeCP2-regulated genes associated with fibroblast migration, myofibroblast differentiation and extracellular matrix degradation, which can be potentially targeted for therapy in SSc.
Collapse
Affiliation(s)
- Ye He
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Dinesh Khanna
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Amr H Sawalha
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
31
|
Liu F, Zhang J, Qian J, Wu G, Ma Z. Emodin alleviates CCl4‑induced liver fibrosis by suppressing epithelial‑mesenchymal transition and transforming growth factor‑β1 in rats. Mol Med Rep 2018; 18:3262-3270. [PMID: 30066878 PMCID: PMC6102702 DOI: 10.3892/mmr.2018.9324] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/12/2018] [Indexed: 12/20/2022] Open
Abstract
Liver fibrosis is a chronic disease that exhibits a complicated pathophysiology. It is characterized by the deposition of the extracellular matrix. Emodin, an active constituent isolated from rhubarb, has antibacterial, immunosuppressive and anti-inflammatory effects. In the present study, the mechanism through which emodin alleviates liver fibrosis in rats was investigated. A rat model of liver fibrosis was generated by administering CCl4 via subcutaneous injection twice a week for 12 weeks. Emodin or sodium carboxymethylcellulose (CMC), as the vehicle, were intragastrically administered daily. After 12 weeks, the liver function index was examined by blood analysis, histopathological scores of fibrosis was determined by hematoxylin and eosin staining and level of collagen deposition was examined by Masson staining. In addition, protein and RNA samples were collected for further analysis. The results of the present study revealed that emodin significantly reduced the liver function index and level of collagen deposition in a dose-dependent manner. Furthermore, emodin reduced the expression of transforming growth factor-β1 (TGF-β1) and the phosphorylation levels of mothers against decapentaplegic homolog 2/3, and inhibited the CCl4-induced downregulation of E-cadherin and upregulation of the mesenchymal markers, fibronectin and vimentin. The expression levels of TGF-β1, Snail family transcriptional repressor (Snail) 2, Snail, twist-related protein 1 and zinc finger E-box-binding homeobox (ZEB)1 and 2 mRNA were significantly decreased in emodin-treated groups compared with the untreated control. Collectively, the results of the present study suggested that emodin may exert antifibrotic effects via the suppression of TGF-β1 signaling and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Feng Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jing Zhang
- Department of Nursing Center, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Jianmin Qian
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Gang Wu
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| | - Zhenyu Ma
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, P.R. China
| |
Collapse
|
32
|
Vella S, Conaldi PG, Cova E, Meloni F, Liotta R, Cuzzocrea S, Martino L, Bertani A, Luca A, Vitulo P. Lung resident mesenchymal cells isolated from patients with the Bronchiolitis Obliterans Syndrome display a deregulated epigenetic profile. Sci Rep 2018; 8:11167. [PMID: 30042393 PMCID: PMC6057887 DOI: 10.1038/s41598-018-29504-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 07/09/2018] [Indexed: 12/13/2022] Open
Abstract
Bronchiolitis Obliterans Syndrome is the major determinant of the graft function loss after lung transplantation, but its pathogenesis is still incompletely understood and currently available therapeutic strategies are poorly effective. A deeper understanding of its pathogenic mechanisms is crucial for the development of new strategies to prevent and treat this devastating complication. In this study, we focused on the mesenchymal stromal cells, recently recognized as BOS key effectors, and our primary aim was to identify their epigenetic determinants, such as histone modifications and non-coding RNA regulation, which could contribute to their differentiation in myofibroblasts. Interestingly, we identified a deregulated expression of histone deacetylases and methyltransferases, and a microRNA-epigenetic regulatory network, which could represent novel targets for anti-fibrotic therapy. We validated our results in vitro, in a cell model of fibrogenesis, confirming the epigenetic involvement in this process and paving the way for a new application for epigenetic drugs.
Collapse
Affiliation(s)
- Serena Vella
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy.
- Anemocyte S.r.l, Gerenzano, Italy.
| | - Pier Giulio Conaldi
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Emanuela Cova
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Federica Meloni
- Department of Respiratory Diseases, IRCCS San Matteo Foundation and University of Pavia, Pavia, Italy
| | - Rosa Liotta
- Department of Diagnostic and Therapeutic Services, Pathology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Lavinia Martino
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Alessandro Bertani
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Angelo Luca
- Department of Diagnostic and Therapeutic Services, Radiology Service, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Patrizio Vitulo
- Department for the Treatment and Study of Cardiothoracic Diseases and Cardiothoracic Transplantation, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| |
Collapse
|
33
|
Shin JM, Um JY, Lee SA, Park IH, Lee SH, Lee HM. Effect of MeCP2 on TGF-β1-induced Extracellular Matrix Production in Nasal Polyp-derived Fibroblasts. Am J Rhinol Allergy 2018; 32:228-235. [DOI: 10.1177/1945892418770291] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Purpose Methyl-CpG-binding protein 2 (MeCP2), known as a transcriptional regulator, has been suggested to play an important role in myofibroblast differentiation in the lung. The purpose of this study was to investigate the role of MeCP2 in transforming growth factor (TGF)- β1-induced myofibroblast differentiation and extracellular matrix (ECM) production in nasal polyp-derived fibroblasts (NPDFs). Methods To identify the expression of MeCP2 in nasal polyp tissues, immunohistochemistry staining and Western blot were performed. TGF- β1-induced NPDFs were treated with 5-azacytidine, a DNA methylation inhibitor, and the expression levels of α-SMA and fibronectin were determined by semiquantitative reverse transcription polymerase chain reaction, immunofluorescent staining, and Western blotting. The total soluble collagen was analyzed by the Sircol collagen assay. MeCP2 silenced by MeCP2-specific small interference ( si) RNA was verified by Western blot. Results The expression levels of MeCP2 increased in nasal polyp tissues compared to normal inferior turbinate tissues. 5-Azacytidine significantly inhibited the expression of α-SMA and fibronectin mRNA in a dose-dependent manner. In addition, 5-azacytidine suppressed collagen production and the expression of MeCP2 in the same manner. The expression levels of a-SMA and collagen production were significantly blocked by MeCP2 silencing in TGF- β1-induced NPDFs. Conclusions Our data suggest that MeCP2 plays an essential role in TGF- β1-induced myofibroblast differentiation and ECM production in NPDFs.
Collapse
Affiliation(s)
- Jae-Min Shin
- Department of Otorhinolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Ji-Young Um
- Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
| | - Seoung-Ae Lee
- Institute for Korea University Medical Devices Support Center, Korea University College of Medicine, Seoul, South Korea
| | - Il-Ho Park
- Department of Otorhinolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Soo-Hyung Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
| | - Heung-Man Lee
- Department of Otorhinolaryngology—Head and Neck Surgery, Korea University College of Medicine, Seoul, South Korea
- Division of Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Seoul, South Korea
- Institute for Korea University Medical Devices Support Center, Korea University College of Medicine, Seoul, South Korea
| |
Collapse
|
34
|
Aloperine Protects Mice against Bleomycin-induced Pulmonary Fibrosis by Attenuating Fibroblast Proliferation and Differentiation. Sci Rep 2018; 8:6265. [PMID: 29674691 PMCID: PMC5908909 DOI: 10.1038/s41598-018-24565-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 04/06/2018] [Indexed: 02/06/2023] Open
Abstract
Aloperine is a quinolizidine alkaloid extracted from Sophora alopecuroides. It has been proven to alleviate oxidative stress and effectively promote tumor cell apoptosis in mice. Herein, we investigated whether aloperine could also mediate its protective effects on bleomycin (BLM)-induced pulmonary fibrosis. Pathological staining, western blot, RT-PCR and flow cytometry were used to evaluate the impact of aloperine on the development of pulmonary fibrosis. The effect of aloperine on fibroblast proliferation, differentiation and related signaling pathways were next investigated to demonstrate the underlying mechanisms. In the present report, we showed that aloperine provided protection for mice against BLM-induced pulmonary fibrosis as manifested by the attenuated lung injury and reduced fibrosis along with alleviated fibroblast proliferation and differentiation. Additionally, we provided in vitro evidence revealing that aloperine inhibited cellular proliferation in PDGF-BB-stimulated mouse lung fibroblasts by repressed PI3K/AKT/mTOR signaling and fibroblast to myofibroblast differentiation by repressed TGF-β/Smad signaling. Overall, our data showed that aloperine could protect the mice against BLM-induced pulmonary fibrosis by attenuated fibroblast proliferation and differentiation, which indicated that aloperine may be therapeutically beneficial for IPF patients.
Collapse
|
35
|
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
|
36
|
Pathogenesis of Lethal Aspiration Pneumonia in Mecp2-null Mouse Model for Rett Syndrome. Sci Rep 2017; 7:12032. [PMID: 28931890 PMCID: PMC5607245 DOI: 10.1038/s41598-017-12293-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 09/06/2017] [Indexed: 11/09/2022] Open
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder mainly caused by mutations in the gene encoding the transcriptional regulator Methyl-CpG-binding protein 2 (MeCP2), located on the X chromosome. Many RTT patients have breathing abnormalities, such as apnea and breathing irregularity, and respiratory infection is the most common cause of death in these individuals. Previous studies showed that MeCP2 is highly expressed in the lung, but its role in pulmonary function remains unknown. In this study, we found that MeCP2 deficiency affects pulmonary gene expression and structures. We also found that Mecp2-null mice, which also have breathing problems, often exhibit inflammatory lung injury. These injuries occurred in specific sites in the lung lobes. In addition, polarizable foreign materials were identified in the injured lungs of Mecp2-null mice. These results indicated that aspiration might be a cause of inflammatory lung injury in Mecp2-null mice. On the other hand, MeCP2 deficiency affected the expression of several neuromodulator genes in the lower brainstem. Among them, neuropeptide substance P (SP) immunostaining was reduced in Mecp2-null brainstem. These findings suggest that alteration of SP expression in brainstem may be involved in autonomic dysregulation, and may be one of the causes of aspiration in Mecp2-null mice.
Collapse
|
37
|
Zhang X, Hu M, Lyu X, Li C, Thannickal VJ, Sanders YY. DNA methylation regulated gene expression in organ fibrosis. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2389-2397. [PMID: 28501566 PMCID: PMC5567836 DOI: 10.1016/j.bbadis.2017.05.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 01/05/2023]
Abstract
DNA methylation is a major epigenetic mechanism to regulate gene expression. Epigenetic regulation, including DNA methylation, histone modifications and RNA interference, results in heritable changes in gene expression independent of alterations in DNA sequence. Epigenetic regulation often occurs in response to aging and environment stimuli, including exposures and diet. Studies have shown that DNA methylation is critical in the pathogenesis of fibrosis involving multiple organ systems, contributing to significant morbidity and mortality. Aberrant DNA methylation can silence or activate gene expression patterns that drive the fibrosis process. Fibrosis is a pathological wound healing process in response to chronic injury. It is characterized by excessive extracellular matrix production and accumulation, which eventually affects organ architecture and results in organ failure. Fibrosis can affect a wide range of organs, including the heart and lungs, and have limited therapeutic options. DNA methylation, like other epigenetic process, is reversible, therefore regarded as attractive therapeutic interventions. Although epigenetic mechanisms are highly interactive and often reinforcing, this review discusses DNA methylation-dependent mechanisms in the pathogenesis of organ fibrosis, with focus on cardiac and pulmonary fibrosis. We discuss specific pro- and anti-fibrotic genes and pathways regulated by DNA methylation in organ fibrosis; we further highlight the potential benefits and side-effects of epigenetic therapies in fibrotic disorders.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Min Hu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xing Lyu
- Laboratory of Clinical Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chun Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Victor J Thannickal
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yan Y Sanders
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
38
|
Abstract
Ischemic heart disease(IHD) is the leading cause of death worldwide. Despite the development of continuously improving therapeutic strategies, morbidity and mortality of patients with IHD remain relatively high. Exosomes are a subpopulation of vesicles that are universally recognized as major mediators in intercellular communication. Numerous preclinical studies have shown that these tiny vesicles were protective in IHD, through such actions as alleviating myocardial ischemia-reperfusion injury, promoting angiogenesis, inhibiting fibrosis, and facilitating cardiac regeneration. Our review focused on these beneficial exosome-mediated processes. In addition, we discuss in detail how to fully exploit the therapeutic potentials of exosomes in the field of IHD. Topics include identifying robust sources of exosomes, loading protective agents into exosomes, developing heart-specific exosomes, optimizing isolation methods, and translating the cardioprotective effects of exosomes into clinical practice. Finally, both the advantages and disadvantages of utilizing exosomes in clinical settings are addressed.
Collapse
Affiliation(s)
- Gui-Hao Chen
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jun Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
39
|
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
|
40
|
Zeybel M, Luli S, Sabater L, Hardy T, Oakley F, Leslie J, Page A, Moran Salvador E, Sharkey V, Tsukamoto H, Chu DCK, Singh US, Ponzoni M, Perri P, Di Paolo D, Mendivil EJ, Mann J, Mann DA. A Proof-of-Concept for Epigenetic Therapy of Tissue Fibrosis: Inhibition of Liver Fibrosis Progression by 3-Deazaneplanocin A. Mol Ther 2017; 25:218-231. [PMID: 28129116 PMCID: PMC5363305 DOI: 10.1016/j.ymthe.2016.10.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 02/08/2023] Open
Abstract
The progression of fibrosis in chronic liver disease is dependent upon hepatic stellate cells (HSCs) transdifferentiating to a myofibroblast-like phenotype. This pivotal process is controlled by enzymes that regulate histone methylation and chromatin structure, which may be targets for developing anti-fibrotics. There is limited pre-clinical experimental support for the potential to therapeutically manipulate epigenetic regulators in fibrosis. In order to learn if epigenetic treatment can halt the progression of pre-established liver fibrosis, we treated mice with the histone methyltransferase inhibitor 3-deazaneplanocin A (DZNep) in a naked form or by selectively targeting HSC-derived myofibroblasts via an antibody-liposome-DZNep targeting vehicle. We discovered that DZNep treatment inhibited multiple histone methylation modifications, indicative of a broader specificity than previously reported. This broad epigenetic repression was associated with the suppression of fibrosis progression as assessed both histologically and biochemically. The anti-fibrotic effect of DZNep was reproduced when the drug was selectively targeted to HSC-derived myofibroblasts. Therefore, the in vivo modulation of HSC histone methylation is sufficient to halt progression of fibrosis in the context of continuous liver damage. This discovery and our novel HSC-targeting vehicle, which avoids the unwanted effects of epigenetic drugs on parenchymal liver cells, represents an important proof-of-concept for epigenetic treatment of liver fibrosis.
Collapse
Affiliation(s)
- Müjdat Zeybel
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; School of Medicine, Koc University, 34450 Istanbul, Turkey
| | - Saimir Luli
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Laura Sabater
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Timothy Hardy
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Fiona Oakley
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Jack Leslie
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Agata Page
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Eva Moran Salvador
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Victoria Sharkey
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Hidekazu Tsukamoto
- Southern California Research Center for ALPD and Cirrhosis, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Veterans Affairs, Greater Los Angeles Healthcare System, Los Angeles, CA 90033, USA
| | - David C K Chu
- The University of Georgia College of Pharmacy, Athens, GA 30602, USA
| | - Uma Sharan Singh
- The University of Georgia College of Pharmacy, Athens, GA 30602, USA
| | - Mirco Ponzoni
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Patrizia Perri
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Daniela Di Paolo
- Experimental Therapy Unit, Laboratory of Oncology, Istituto Giannina Gaslini, 16148 Genova, Italy
| | - Edgar J Mendivil
- Department of Molecular Biology and Genomics, Institute for Molecular Biology and Gene Therapy, University of Guadalajara, 44100 Guadalajara, Mexico
| | - Jelena Mann
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Derek A Mann
- Institute of Cellular Medicine, Faculty of Medical Sciences, 4(th) Floor, William Leech Building, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
41
|
Wang Y, Chen C, Deng Z, Bian E, Huang C, Lei T, Lv X, Liu L, Li J. Repression of TSC1/TSC2 mediated by MeCP2 regulates human embryo lung fibroblast cell differentiation and proliferation. Int J Biol Macromol 2016; 96:578-588. [PMID: 28041914 DOI: 10.1016/j.ijbiomac.2016.12.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
Pulmonary fibrosis (PF) is a severe inflammatory disease with limited effective treatments. It is known that the transdifferentiation of human embryo lung fibroblast (HELF) cells from pulmonary fibroblasts into myofibroblasts, contributes to the progression of pulmonary fibrogenesis. The tuberous sclerosis proteins TSC1 and TSC2 are two key signaling factors which can suppress cell growth and proliferation. However, the roles of TSC1 and TSC2 in lung fibroblast are unclear. Here, we developed a PF model with bleomycin (BLM) in mice and conducted several simulation experiments in HELF cells. Our study shows that the expression of TSC1 and TSC2 in fibrotic mice lung was reduced and stimulation of HELF cells with TGF-β1 resulted in a down-regulation of TSC1 and TSC2. In addition, overexpression of TSC1 or TSC2 decreased cell proliferation and differentiation. Furthermore, we found that reduced expression of TSC1 and TSC2 caused by TGF-β1 is associated with the promoter methylation status of TSC1 and TSC2. MeCP2, controls an epigenetic pathway that promotes myofibroblast transdifferentiation and fibrosis. We found that expression of TSC1 and TSC2 can be repressed by MeCP2, which regulates HELF cell differentiation and proliferation as myofibroblasts and lead to PF ultimately.
Collapse
Affiliation(s)
- Yuanyuan Wang
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Chen Chen
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Hefei Binghu Hospital, 3200 Changsha Road, Hefei 230000, Anhui, China
| | - Ziyu Deng
- Department of Scientific and Educational, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Erbao Bian
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Cheng Huang
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ting Lei
- Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Xiongwen Lv
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Liping Liu
- Department of Pharmacology, The Second Hospital of Anhui Medical University, 678 Furong Road, Hefei 230601, Anhui, China
| | - Jun Li
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
42
|
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
|
43
|
Increased Global DNA Methylation and Decreased TGFβ1 Promoter Methylation in Glaucomatous Lamina Cribrosa Cells. J Glaucoma 2016; 25:e834-e842. [DOI: 10.1097/ijg.0000000000000453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Dowson C, O'Reilly S. DNA methylation in fibrosis. Eur J Cell Biol 2016; 95:323-30. [PMID: 27346523 DOI: 10.1016/j.ejcb.2016.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/10/2016] [Accepted: 06/12/2016] [Indexed: 02/07/2023] Open
Abstract
Fibrosis is characterised by an exuberant wound healing response and the major cell type responsible is the myofibroblast. The myofibroblast is typified by excessive ECM production and contractile activity and is demarcated by alpha-smooth muscle actin expression. What has recently come to light is that the activation of the fibroblast to myofibroblast may be under epigenetic control, specifically methylation. Methylation of DNA is a conserved mechanism to precisely regulate gene expression in a specific context. Hypermethylation leads to gene repression and hypomethylation results in gene induction. Methylation abnormalities have recently been uncovered in fibrosis, both organ specific and widespread fibrosis. The fact that these methylation changes are rapid and reversible lends themselves amenable to therapeutic intervention. This review considers the role of methylation in fibrosis and the activation of the myofibroblasts and how this could be targeted for fibrosis. Fibrosis is of course currently intractable to therapeutics and is a leading cause of morbidity and mortality and is an urgent unmet clinical need.
Collapse
Affiliation(s)
- Christopher Dowson
- Cell Biology Group, Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle Upon Tyne NE1 3HY, United Kingdom
| | - Steven O'Reilly
- Cell Biology Group, Faculty of Health and Life Sciences, Northumbria University, Ellison Building, Newcastle Upon Tyne NE1 3HY, United Kingdom.
| |
Collapse
|
45
|
Kyle SM, Saha PK, Brown HM, Chan LC, Justice MJ. MeCP2 co-ordinates liver lipid metabolism with the NCoR1/HDAC3 corepressor complex. Hum Mol Genet 2016; 25:3029-3041. [PMID: 27288453 PMCID: PMC5181597 DOI: 10.1093/hmg/ddw156] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/02/2016] [Accepted: 05/18/2016] [Indexed: 01/11/2023] Open
Abstract
Rett syndrome (RTT; OMIM 312750), a progressive neurological disorder, is caused by mutations in methyl-CpG-binding protein 2 (MECP2; OMIM 300005), a ubiquitously expressed factor. A genetic suppressor screen designed to identify therapeutic targets surprisingly revealed that downregulation of the cholesterol biosynthesis pathway improves neurological phenotypes in Mecp2 mutant mice. Here, we show that MeCP2 plays a direct role in regulating lipid metabolism. Mecp2 deletion in mice results in a host of severe metabolic defects caused by lipid accumulation, including insulin resistance, fatty liver, perturbed energy utilization, and adipose inflammation by macrophage infiltration. We show that MeCP2 regulates lipid homeostasis by anchoring the repressor complex containing NCoR1 and HDAC3 to its lipogenesis targets in hepatocytes. Consistently, we find that liver targeted deletion of Mecp2 causes fatty liver disease and dyslipidemia similar to HDAC3 liver-specific deletion. These findings position MeCP2 as a novel component in metabolic homeostasis. Rett syndrome patients also show signs of peripheral dyslipidemia; thus, together these data suggest that RTT should be classified as a neurological disorder with systemic metabolic components. We previously showed that treatment of Mecp2 mice with statin drugs alleviated motor symptoms and improved health and longevity. Lipid metabolism is a highly treatable target; therefore, our results shed light on new metabolic pathways for treatment of Rett syndrome.
Collapse
Affiliation(s)
- Stephanie M Kyle
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, ON M5G 0A4, Canada.,Department of Molecular and Human Genetics
| | - Pradip K Saha
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Lawrence C Chan
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | - Monica J Justice
- Genetics and Genome Biology Program, The Hospital for Sick Children, The Peter Gilgan Centre for Research and Learning, Toronto, ON M5G 0A4, Canada .,Department of Molecular and Human Genetics.,Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
46
|
SCHWARTZ DAVIDA. IDIOPATHIC PULMONARY FIBROSIS IS A COMPLEX GENETIC DISORDER. TRANSACTIONS OF THE AMERICAN CLINICAL AND CLIMATOLOGICAL ASSOCIATION 2016; 127:34-45. [PMID: 28066036 PMCID: PMC5216513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a complex, heterogeneous genetic disorder that is associated with rare and common sequence variants in many genes (MUC5B, SFTPC, SFTPA2, RTEL1, TERT, and hTR), 11 novel loci, and multiple emerging epigenetic and transcriptional profiles. In the past 5 years, we have found that: 1) genetic risk variants play major and similar roles in the development of both familial and sporadic fibrotic idiopathic interstitial pneumonia, accounting for up to 35% of the risk of idiopathic interstitial pneumonia (a disease that was previously thought to be idiopathic); 2) a promoter variant in MUC5B rs35705950 is the strongest risk factor for the development of IIP and IPF; however, rs35705950 has a low penetrance; and 3) IPF is a complex genetic disease with 11 independent loci contributing to the development of this disease, pronounced changes in DNA methylation, and transcriptional subtypes. In aggregate, these findings suggest that IPF is a heterogeneous disease and that genetic and molecular subtypes of IPF will provide essential clues to disease pathogenesis, prognosis, treatment, and survival, all of which remain major problems in understanding and treating patients with IPF. Although the basic biological mechanisms involved in IPF are emerging, the disease is heterogeneous pathologically and the final common pathways of fibrogenesis are not well understood. These observations lead us to postulate that the etiology and severity/extent of this complex condition will best be understood through an integrated approach that accounts for inherited factors, epigenetic marks, and dynamic changes in the transcriptome.
Collapse
|
47
|
Toedebusch RG, Ruegsegger GN, Braselton JF, Heese AJ, Hofheins JC, Childs TE, Thyfault JP, Booth FW. AMPK agonist AICAR delays the initial decline in lifetime-apex V̇o2 peak, while voluntary wheel running fails to delay its initial decline in female rats. Physiol Genomics 2015; 48:101-15. [PMID: 26578698 DOI: 10.1152/physiolgenomics.00078.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/11/2015] [Indexed: 11/22/2022] Open
Abstract
There has never been an outcome measure for human health more important than peak oxygen consumption (V̇o2 peak), yet little is known regarding the molecular triggers for its lifetime decline with aging. We examined the ability of physical activity or 5 wk of 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR) administration to delay the initial aging-induced decline in lifetime-apex V̇o2 peak and potential underlying molecular mechanisms. Experiment 1 consisted of female rats with (RUN) and without (NO RUN) running wheels, while experiment 2 consisted of female nonrunning rats getting the AMPK agonist AICAR (0.5 mg/g/day) subcutaneously for 5 wk beginning at 17 wk of age. All rats underwent frequent, weekly or biweekly V̇o2 peak tests beginning at 10 wk of age. In experiment 1, lifetime-apex V̇o2 peak occurred at 19 wk of age in both RUN and NO RUN and decreased thereafter. V̇o2 peak measured across experiment 1 was ∼25% higher in RUN than in NO RUN. In experiment 2, AICAR delayed the chronological age observed in experiment 1 by 1 wk, from 19 wk to 20 wk of age. RUN and NO RUN showed different skeletal muscle transcriptomic profiles both pre- and postapex. Additionally, growth and development pathways are differentially regulated between RUN and NO RUN. Angiomotin mRNA was downregulated postapex in RUN and NO RUN. Furthermore, strong significant correlations to V̇o2 peak and trends for decreased protein concentration supports angiomotin's potential importance in our model. Contrary to our primary hypothesis, wheel running was not sufficient to delay the chronological age of lifetime-apex V̇o2 peak decline, whereas AICAR delayed it 1 wk.
Collapse
Affiliation(s)
- Ryan G Toedebusch
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | | | - Joshua F Braselton
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Alexander J Heese
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John C Hofheins
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - Tom E Childs
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas; and
| | - Frank W Booth
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri; Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
48
|
He S, Barron E, Ishikawa K, Nazari Khanamiri H, Spee C, Zhou P, Kase S, Wang Z, Dustin LD, Hinton DR. Inhibition of DNA Methylation and Methyl-CpG-Binding Protein 2 Suppresses RPE Transdifferentiation: Relevance to Proliferative Vitreoretinopathy. Invest Ophthalmol Vis Sci 2015; 56:5579-89. [PMID: 26305530 DOI: 10.1167/iovs.14-16258] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE The purpose of this study was to evaluate expression of methyl-CpG-binding protein 2 (MeCP2) in epiretinal membranes from patients with proliferative vitreoretinopathy (PVR) and to investigate effects of inhibition of MeCP2 and DNA methylation on transforming growth factor (TGF)-β-induced retinal pigment epithelial (RPE) cell transdifferentiation. METHODS Expression of MeCP2 and its colocalization with cytokeratin and α-smooth muscle actin (α-SMA) in surgically excised PVR membranes was studied using immunohistochemistry. The effects of 5-AZA-2'-deoxycytidine (5-AZA-dC) on human RPE cell migration and viability were evaluated using a modified Boyden chamber assay and the colorimetric 3-(4,5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide (MTT) assay. Expression of RASAL1 mRNA and its promoter region methylation were evaluated by real-time PCR and methylation-specific PCR. Effects of 5-AZA-dC on expression of α-SMA, fibronectin (FN), and TGF-β receptor 2 (TGF-β R2) and Smad2/3 phosphorylation were analyzed by Western blotting. Effect of short interfering RNA (siRNA) knock-down of MeCP2 on expression of α-SMA and FN induced by TGFβ was determined. RESULTS MeCP2 was abundantly expressed in cells within PVR membranes where it was double labeled with cells positive for cytokeratin and α-SMA. 5-AZA-dC inhibited expression of MeCP2 and suppressed RASAL1 gene methylation while increasing expression of the RASAL1 gene. Treatment with 5-AZA-dC significantly suppressed the expression of α-SMA, FN, TGF-β R2 and phosphorylation of Smad2/3 and inhibited RPE cell migration. TGF-β induced expression of α-SMA, and FN was suppressed by knock-down of MeCP2. CONCLUSIONS MeCP2 and DNA methylation regulate RPE transdifferentiation and may be involved in the pathogenesis of PVR.
Collapse
Affiliation(s)
- Shikun He
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 2Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, Unit
| | - Ernesto Barron
- Doheny Eye Institute, Los Angeles, California, United States
| | | | - Hossein Nazari Khanamiri
- Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Chris Spee
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - Peng Zhou
- Doheny Eye Institute, Los Angeles, California, United States
| | - Satoru Kase
- Doheny Eye Institute, Los Angeles, California, United States
| | - Zhuoshi Wang
- Doheny Eye Institute, Los Angeles, California, United States
| | - Laurie Diane Dustin
- Department of Preventive Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States
| | - David R Hinton
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California, United States 2Department of Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, California, Unit
| |
Collapse
|
49
|
Huan C, Yang T, Liang J, Xie T, Cheng L, Liu N, Kurkciyan A, Monterrosa Mena J, Wang C, Dai H, Noble PW, Jiang D. Methylation-mediated BMPER expression in fibroblast activation in vitro and lung fibrosis in mice in vivo. Sci Rep 2015; 5:14910. [PMID: 26442443 PMCID: PMC4595647 DOI: 10.1038/srep14910] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease. Although the pathogenesis is poorly understood, evidence suggests that genetic and epigenetic alterations, such as DNA methylation, may play a key role. Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-β (TGF-β) superfamily and are important regulators in IPF. Here we identified BMP endothelial cell precursor-derived regulator (BMPER) as a key regulator of fibroblast activation. BMPER is a secreted glycoprotein that binds directly to BMPs and may regulate TGF-β/BMP signaling, but its role in lung fibrosis is not clear. BMPER is highly expressed in human IPF lung fibroblasts compared to normal lung fibroblasts. Demethylation agent 5′-azacytidine decreased BMPER expression in fibroblasts, and attenuated the invasion and migration of IPF lung fibroblasts. Furthermore, siRNA-mediated reduction of BMPER in the human lung fibroblasts impaired cell migration and invasion. 5′-azacytidine treatment additionally regulated BMPER expression and reduced lung fibrosis in mice in vivo. These findings demonstrate that methylation of specific genes in fibroblasts may offer a new therapeutic strategy for IPF by modulating fibroblast activation.
Collapse
Affiliation(s)
- Caijuan Huan
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China.,Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ting Yang
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China
| | - Jiurong Liang
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ting Xie
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Luis Cheng
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Ningshan Liu
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Adrianne Kurkciyan
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | | | - Chen Wang
- China-Japan Friendship Hospital, Beijing, China
| | - Huaping Dai
- Department of Respiratory and Critical Care Medicine, Beijing Key Laboratory of Respiratory and Pulmonary Circulation Disorders, Beijing Chao-Yang Hospital-Beijing Institute of Respiratory Medicine, Capital Medical University, Beijing 100020, China
| | - Paul W Noble
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| | - Dianhua Jiang
- Cedars-Sinai Medical Center, Department of Medicine, Los Angeles, CA 90048, USA
| |
Collapse
|
50
|
Neary R, Watson CJ, Baugh JA. Epigenetics and the overhealing wound: the role of DNA methylation in fibrosis. FIBROGENESIS & TISSUE REPAIR 2015; 8:18. [PMID: 26435749 PMCID: PMC4591063 DOI: 10.1186/s13069-015-0035-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 09/04/2015] [Indexed: 12/20/2022]
Abstract
Fibrosis is a progressive and potentially fatal process that can occur in numerous organ systems. Characterised by the excessive deposition of extracellular matrix proteins such as collagens and fibronectin, fibrosis affects normal tissue architecture and impedes organ function. Although a considerable amount of research has focused on the mechanisms underlying disease pathogenesis, current therapeutic options do not directly target the pro-fibrotic process. As a result, there is a clear unmet clinical need to develop new agents. Novel findings implicate a role for epigenetic modifications contributing to the progression of fibrosis by alteration of gene expression profiles. This review will focus on DNA methylation; its association with fibroblast differentiation and activation and the consequent buildup of fibrotic scar tissue. The potential use of therapies that modulate this epigenetic pathway for the treatment of fibrosis in several organ systems is also discussed.
Collapse
Affiliation(s)
- Roisin Neary
- UCD School of Medicine and Medical Science, Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4 Ireland
| | - Chris J Watson
- UCD School of Medicine and Medical Science, Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4 Ireland
| | - John A Baugh
- UCD School of Medicine and Medical Science, Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4 Ireland
| |
Collapse
|