1
|
Meng Q, Bao D, Liu S, Huang J, Guo M, Dai B, Ding L, Xie S, Meng M, Lv C, He W, Luo H, Zhu H. ADAM Metallopeptidase domain 19 promotes skin fibrosis in systemic sclerosis via neuregulin-1. Mol Med 2024; 30:269. [PMID: 39716051 DOI: 10.1186/s10020-024-01047-8] [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: 09/08/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND ADAM19 (ADAM Metallopeptidase Domain 19) is known to be involved in extracellular matrix (ECM) remodeling, yet its specific function in systemic sclerosis (SSc) fibrosis remains unclear. OBJECTIVES This study sought to clarify the role and underlying mechanism of ADAM19 in SSc skin fibrosis. METHODS The expression of ADAM19 was assessed in skin tissues of SSc and wound healing using publicly available transcriptome datasets. This analysis was further validated through real-time PCR, western blot, and immunostaining in our SSc cohort, as well as in a mouse model of hypochlorite (HOCl)-induced fibrosis. To downregulate the expression of ADAM19, ADAM19 siRNA was employed. The influence of ADAM19 on fibroblast transcriptomics was examined using bulk RNA-seq. Data analysis and visualization were conducted using R packages, including edgeR, limma, clusterProfiler, ggplot2, gseaplot2, and complexheatmap. RESULTS ADAM19 exhibited a significant upregulation in skin tissues of SSc patients, as well as in wound healing and a HOCl-induced fibrosis mouse model. Additionally, there was a notable positive correlation between ADAM19 and fibrosis-related genes, local skin score, Modified Rodnan skin score, skin thickness progression rate, and the presence of ARA antibodies in SSc patients. Furthermore, ADAM19 levels were markedly elevated in SSc primary dermal fibroblasts and TGF-β-stimulated healthy controls primary dermal fibroblasts. The downregulation of ADAM19 resulted in the repression of TGF-β-induced ECM deposition and fibroblast activation. ADAM19 was identified as a mediator for the shedding of neuregulin-1 (NRG1) in fibroblasts, a pro-fibrotic cytokine that must be cleaved to exert its function. CONCLUSION ADAM19 plays a role in TGF-β-induced ECM deposition and fibroblast activation by mediating the shedding of NRG1, ultimately contributing to the development of skin fibrosis in SSc.
Collapse
Affiliation(s)
- Qiming Meng
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Ding Bao
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Sijia Liu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jing Huang
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Muyao Guo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Bingying Dai
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Liqing Ding
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Shasha Xie
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Meng Meng
- Department of Pathology, Xiangya Hospital, Changsha, 410008, P.R. China
| | - Chunliu Lv
- Department of Breast Tumor Plastic Surgery, The Affiliated Cancer Hospital of Xiangya School of Medicine, Hunan Cancer Hospital, Central South University, 283 Tongzipo Road, Changsha, Hunan, 410013, P.R. China
| | - Weijia He
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Hui Luo
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- Provincial Clinical Research Center for Rheumatic and Immunologic Diseases, Xiangya Hospital, Changsha, Hunan, 410008, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
| |
Collapse
|
2
|
Sanz-Nogués C, Keane AJ, Creane M, Hynes SO, Chen X, Lyons CJ, Horan E, Elliman SJ, Goljanek-Whysall K, O’Brien T. Mesenchymal stromal cell transplantation ameliorates fibrosis and microRNA dysregulation in skeletal muscle ischemia. Stem Cells 2024; 42:976-991. [PMID: 39283740 PMCID: PMC11541228 DOI: 10.1093/stmcls/sxae058] [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: 04/12/2024] [Accepted: 08/23/2024] [Indexed: 11/08/2024]
Abstract
Peripheral arterial disease (PAD) is associated with lower-extremity muscle wasting. Hallmark features of PAD-associated skeletal muscle pathology include loss of skeletal muscle mass, reduced strength and physical performance, increased inflammation, fibrosis, and adipocyte infiltration. At the molecular level, skeletal muscle ischemia has also been associated with gene and microRNA (miRNA) dysregulation. Mesenchymal stromal cells (MSCs) have been shown to enhance muscle regeneration and improve muscle function in various skeletal muscle injuries. This study aimed to evaluate the effects of intramuscularly delivered human umbilical cord-derived MSCs (hUC-MSCs) on skeletal muscle ischemia. Herein, we report an hUC-MSC-mediated amelioration of ischemia-induced skeletal muscle atrophy and function via enhancement of myofiber regeneration, reduction of tissue inflammation, adipocyte accumulation, and tissue fibrosis. These changes were observed in the absence of cell-mediated enhancement of blood flow recovery as measured by laser Doppler imaging. Furthermore, reduced tissue fibrosis in the hUC-MSC-treated group was associated with upregulation of miR-1, miR-133a, and miR-29b and downregulation of targeted pro-fibrotic genes such as Col1a1 and Fn1. Our results support the use of hUC-MSCs as a novel approach to reduce fibrosis and promote skeletal muscle regeneration after ischemic injury in patients with PAD.
Collapse
Affiliation(s)
- Clara Sanz-Nogués
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Alan J Keane
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Michael Creane
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Sean O Hynes
- Discipline of Pathology, University of Galway, Galway, Ireland
- Division of Anatomic Pathology, University Hospital Galway, Galway, Ireland
| | - Xizhe Chen
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Caomhán J Lyons
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Emma Horan
- Orbsen Therapeutics Ltd., Galway, Ireland
| | | | - Katarzyna Goljanek-Whysall
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Timothy O’Brien
- Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
- CÚRAM SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
3
|
Tang J, Luo Y, Wang Q, Wu J, Wei Y. Stimuli-Responsive Delivery Systems for Intervertebral Disc Degeneration. Int J Nanomedicine 2024; 19:4735-4757. [PMID: 38813390 PMCID: PMC11135562 DOI: 10.2147/ijn.s463939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/13/2024] [Indexed: 05/31/2024] Open
Abstract
As a major cause of low back pain, intervertebral disc degeneration is an increasingly prevalent chronic disease worldwide that leads to huge annual financial losses. The intervertebral disc consists of the inner nucleus pulposus, outer annulus fibrosus, and sandwiched cartilage endplates. All these factors collectively participate in maintaining the structure and physiological functions of the disc. During the unavoidable degeneration stage, the degenerated discs are surrounded by a harsh microenvironment characterized by acidic, oxidative, inflammatory, and chaotic cytokine expression. Loss of stem cell markers, imbalance of the extracellular matrix, increase in inflammation, sensory hyperinnervation, and vascularization have been considered as the reasons for the progression of intervertebral disc degeneration. The current treatment approaches include conservative therapy and surgery, both of which have drawbacks. Novel stimuli-responsive delivery systems are more promising future therapeutic options than traditional treatments. By combining bioactive agents with specially designed hydrogels, scaffolds, microspheres, and nanoparticles, novel stimuli-responsive delivery systems can realize the targeted and sustained release of drugs, which can both reduce systematic adverse effects and maximize therapeutic efficacy. Trigger factors are categorized into internal (pH, reactive oxygen species, enzymes, etc.) and external stimuli (photo, ultrasound, magnetic, etc.) based on their intrinsic properties. This review systematically summarizes novel stimuli-responsive delivery systems for intervertebral disc degeneration, shedding new light on intervertebral disc therapy.
Collapse
Affiliation(s)
- Jianing Tang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yuexin Luo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qirui Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Juntao Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- First Clinic School, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yulong Wei
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
4
|
Rajabi S, Saberi S, Najafipour H, Askaripour M, Rajizadeh MA, Shahraki S, Kazeminia S. Interaction of estradiol and renin-angiotensin system with microRNAs-21 and -29 in renal fibrosis: focus on TGF-β/smad signaling pathway. Mol Biol Rep 2024; 51:137. [PMID: 38236310 DOI: 10.1007/s11033-023-09127-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024]
Abstract
Kidney fibrosis is one of the complications of chronic kidney disease (CKD (and contributes to end-stage renal disease which requires dialysis and kidney transplantation. Several signaling pathways such as renin-angiotensin system (RAS), microRNAs (miRNAs) and transforming growth factor-β1 (TGF-β1)/Smad have a prominent role in pathophysiology and progression of renal fibrosis. Activation of classical RAS, the elevation of angiotensin II (Ang II) production and overexpression of AT1R, develop renal fibrosis via TGF-β/Smad pathway. While the non-classical RAS arm, Ang 1-7/AT2R, MasR reveals an anti-fibrotic effect via antagonizing Ang II. This review focused on studies illustrating the interaction of RAS with sexual female hormone estradiol and miRNAs in the progression of renal fibrosis with more emphasis on the TGF-β signaling pathway. MiRNAs, especially miRNA-21 and miRNA-29 showed regulatory effects in renal fibrosis. Also, 17β-estradiol (E2) is a renoprotective hormone that improved renal fibrosis. Beneficial effects of ACE inhibitors and ARBs are reported in the prevention of renal fibrosis in patients. Future studies are also merited to delineate the new therapy strategies such as miRNAs targeting, combination therapy of E2 or HRT, ACEis, and ARBs with miRNAs mimics and antagomirs in CKD to provide a new therapeutic approach for kidney patients.
Collapse
Affiliation(s)
- Soodeh Rajabi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shadan Saberi
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Askaripour
- Department of Physiology, School of Medicine, Bam University of Medical Sciences, Bam, Iran.
| | - Mohammad Amin Rajizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sarieh Shahraki
- Department of Physiology and Pharmacology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Sara Kazeminia
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
5
|
Iijima H, Wang K, D'Amico E, Tang WY, Rogers RJ, Jakicic JM, Ambrosio F. Exercise-primed extracellular vesicles improve cell-matrix adhesion and chondrocyte health. RESEARCH SQUARE 2023:rs.3.rs-2958821. [PMID: 37333349 PMCID: PMC10274961 DOI: 10.21203/rs.3.rs-2958821/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Extracellular vesicles (EVs) have been suggested to transmit the health-promoting effects of exercise throughout the body. Yet, the mechanisms by which beneficial information is transmitted from extracellular vesicles to recipient cells are poorly understood, precluding a holistic understanding of how exercise promotes cellular and tissue health. In this study, using articular cartilage as a model, we introduced a network medicine paradigm to simulate how exercise facilitates communication between circulating EVs and chondrocytes, the cells resident in articular cartilage. Using the archived small RNA-seq data of EV before and after aerobic exercise, microRNA regulatory network analysis based on network propagation inferred that circulating EVs activated by aerobic exercise perturb chondrocyte-matrix interactions and downstream cellular aging processes. Building on the mechanistic framework identified through computational analyses, follow up experimental studies interrogated the direct influence of exercise on EV-mediated chondrocyte-matrix interactions. We found that pathogenic matrix signaling in chondrocytes was abrogated in the presence of exercise-primed EVs, restoring a more youthful phenotype, as determined by chondrocyte morphological profiling and evaluation of chondrogenicity. Epigenetic reprograming of the gene encoding the longevity protein, α-Klotho, mediated these effects. These studies provide mechanistic evidence that exercise transduces rejuvenation signals to circulating EVs, endowing EVs with the capacity to ameliorate cellular health even in the presence of an unfavorable microenvironmental signals.
Collapse
Affiliation(s)
- Hirotaka Iijima
- Institute for Advanced Research, Nagoya University, Nagoya, Japan
- Biomedical and Health Informatics Unit, Graduate School of Medicine, Nagoya University, Nagoya, Japan
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Kai Wang
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| | - Ella D'Amico
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA
| | - Renee J Rogers
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - John M Jakicic
- Department of Internal Medicine, Division of Physical Activity and Weight Management, University of Kansas Medical Center, Kansas City, KS
| | - Fabrisia Ambrosio
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA
- Discovery Center for Musculoskeletal Recovery, Schoen Adams Research Institute at Spaulding, Boston, MA
- Department of Physical Medicine & Rehabilitation, Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Che J, Su Z, Yang W, Xu L, Li Y, Wang H, Zhou W. Tumor-suppressor p53 specifically binds to miR-29c-3p and reduces ADAM12 expression in hepatocellular carcinoma. Dig Liver Dis 2023; 55:412-421. [PMID: 35853821 DOI: 10.1016/j.dld.2022.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an extremely aggressive malignant tumor associated with high migratory and invasive potential. The present study intends to explore regulatory mechanism of p53/microRNA (miR)-29c-3p/A disintegrin and metalloproteinase 12 (ADAM12) axis in HCC based on clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology. METHODS Putative miR-29c-3p binding sites on ADAM12 3'UTR were verified by a luciferase assay. The binding affinity of p53 to miR-29c-3p was assessed based on CRISPR/Cas9 technology to construct a p53 knockout (p53-/-) HCCLM3 cell line. Furthermore, the effect of p53/miR-29c-3p/ADAM12 was assessed on maligant phenotypes in vitro and tumor formation and metastasis in nude mice. RESULTS ADAM12 was highly expressed but miR-29c-3p was poorly expressed in HCC. miR-29c-3p inhibited migratory and invasive abilities of HCC cells by targeting ADAM12 expression. p53 was found to target and upregulate miR-29c-3p, thus downregulating ADAM12 and conferring inhibitory effect on HCC cell activities. Moreover, ADAM12 knockout or p53 overexpression reduced HCC tumor formation and metastasis, which were reversed by further silencing of miR-29c-3p. CONCLUSION The identification of the p53/miR-29c-3p/ADAM12 axis in migration and invasion of HCC may potentially further our understanding of mechanisms underpinning HCC, and also bear translational value as novel molecular targets.
Collapse
Affiliation(s)
- Jinhui Che
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China
| | - Zhan Su
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China
| | - Weizhong Yang
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China
| | - Lu Xu
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China
| | - Yunjiu Li
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China
| | - Haihong Wang
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China.
| | - Wuyuan Zhou
- Department of Hepatopancreatobiliary Surgery, Xuzhou City Cancer Hospital, Xuzhou 221000, PR China.
| |
Collapse
|
7
|
Wu J, Zhao X, Xiao C, Xiong G, Ye X, Li L, Fang Y, Chen H, Yang W, Du X. The role of lung macrophages in chronic obstructive pulmonary disease. Respir Med 2022; 205:107035. [PMID: 36343504 DOI: 10.1016/j.rmed.2022.107035] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) as a common, preventable and treatable chronic respiratory disease in clinic, gets continuous deterioration and we can't take effective intervention at present. Lung macrophages (LMs) are closely related to the occurrence and development of COPD, but the specific mechanism is not completely clear. In this review we will focus on the role of LMs and potential avenues for therapeutic targeting for LMs in COPD.
Collapse
Affiliation(s)
- Jianli Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xia Zhao
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Chuang Xiao
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China
| | - Guosheng Xiong
- Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiulin Ye
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Lin Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Yan Fang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hong Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Weimin Yang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
| | - Xiaohua Du
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
8
|
Graziosi A, Sita G, Corrieri C, Angelini S, d’Emmanuele di Villa Bianca R, Mitidieri E, Sorrentino R, Hrelia P, Morroni F. Effects of Subtoxic Concentrations of Atrazine, Cypermethrin, and Vinclozolin on microRNA-Mediated PI3K/Akt/mTOR Signaling in SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms232314538. [PMID: 36498866 PMCID: PMC9737829 DOI: 10.3390/ijms232314538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
Endocrine-disrupting chemicals (EDCs) are different natural and synthetic chemicals that may interfere with several mechanisms of the endocrine system producing adverse developmental, metabolic, reproductive, and neurological effects in both human beings and wildlife. Among pesticides, numerous chemicals have been identified as EDCs. MicroRNAs (miRNAs) can regulate gene expression, making fine adjustments in mRNA abundance and regulating proteostasis. We hypothesized that exposure to low doses of atrazine, cypermethrin, and vinclozolin may lead to effects on miRNA expression in SH-SY5Y cells. In particular, the exposure of SH-SY5Y cells to subtoxic concentrations of vinclozolin is able to downregulate miR-29b-3p expression leading to the increase in the related gene expression of ADAM12 and CDK6, which may promote a pro-oncogenic response through the activation of the PI3K/Akt/mTOR pathway and counteracting p53 activity. A better understanding of the molecular mechanisms of EDCs could provide important insight into their role in human disease.
Collapse
Affiliation(s)
- Agnese Graziosi
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Giulia Sita
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Camilla Corrieri
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Sabrina Angelini
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | | | - Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Study of Naples—Federico II, via Montesano 49, 80131 Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine and Surgery, University of Study of Naples—Federico II, via Pansini 5, 80131 Naples, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
- Correspondence: ; Tel.: +39-051-209-1798
| | - Fabiana Morroni
- Department of Pharmacy and BioTechnology—FaBiT, Alma Mater Studiorum—University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
9
|
The Role of miR-29 Family in TGF-β Driven Fibrosis in Glaucomatous Optic Neuropathy. Int J Mol Sci 2022; 23:ijms231810216. [PMID: 36142127 PMCID: PMC9499597 DOI: 10.3390/ijms231810216] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Primary open angle glaucoma (POAG), a chronic optic neuropathy, remains the leading cause of irreversible blindness worldwide. It is driven in part by the pro-fibrotic cytokine transforming growth factor beta (TGF-β) and leads to extracellular matrix remodelling at the lamina cribrosa of the optic nerve head. Despite an array of medical and surgical treatments targeting the only known modifiable risk factor, raised intraocular pressure, many patients still progress and develop significant visual field loss and eventual blindness. The search for alternative treatment strategies targeting the underlying fibrotic transformation in the optic nerve head and trabecular meshwork in glaucoma is ongoing. MicroRNAs are small non-coding RNAs known to regulate post-transcriptional gene expression. Extensive research has been undertaken to uncover the complex role of miRNAs in gene expression and miRNA dysregulation in fibrotic disease. MiR-29 is a family of miRNAs which are strongly anti-fibrotic in their effects on the TGF-β signalling pathway and the regulation of extracellular matrix production and deposition. In this review, we discuss the anti-fibrotic effects of miR-29 and the role of miR-29 in ocular pathology and in the development of glaucomatous optic neuropathy. A better understanding of the role of miR-29 in POAG may aid in developing diagnostic and therapeutic strategies in glaucoma.
Collapse
|
10
|
Baßler K, Fujii W, Kapellos TS, Dudkin E, Reusch N, Horne A, Reiz B, Luecken MD, Osei-Sarpong C, Warnat-Herresthal S, Bonaguro L, Schulte-Schrepping J, Wagner A, Günther P, Pizarro C, Schreiber T, Knoll R, Holsten L, Kröger C, De Domenico E, Becker M, Händler K, Wohnhaas CT, Baumgartner F, Köhler M, Theis H, Kraut M, Wadsworth MH, Hughes TK, Ferreira HJ, Hinkley E, Kaltheuner IH, Geyer M, Thiele C, Shalek AK, Feißt A, Thomas D, Dickten H, Beyer M, Baum P, Yosef N, Aschenbrenner AC, Ulas T, Hasenauer J, Theis FJ, Skowasch D, Schultze JL. Alveolar macrophages in early stage COPD show functional deviations with properties of impaired immune activation. Front Immunol 2022; 13:917232. [PMID: 35979364 PMCID: PMC9377018 DOI: 10.3389/fimmu.2022.917232] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 06/24/2022] [Indexed: 12/13/2022] Open
Abstract
Despite its high prevalence, the cellular and molecular mechanisms of chronic obstructive pulmonary disease (COPD) are far from being understood. Here, we determine disease-related changes in cellular and molecular compositions within the alveolar space and peripheral blood of a cohort of COPD patients and controls. Myeloid cells were the largest cellular compartment in the alveolar space with invading monocytes and proliferating macrophages elevated in COPD. Modeling cell-to-cell communication, signaling pathway usage, and transcription factor binding predicts TGF-β1 to be a major upstream regulator of transcriptional changes in alveolar macrophages of COPD patients. Functionally, macrophages in COPD showed reduced antigen presentation capacity, accumulation of cholesteryl ester, reduced cellular chemotaxis, and mitochondrial dysfunction, reminiscent of impaired immune activation.
Collapse
Affiliation(s)
- Kevin Baßler
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Wataru Fujii
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Theodore S. Kapellos
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Erika Dudkin
- Computational Life Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Neuherberg, Germany
| | - Nico Reusch
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Ari Horne
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | | | - Malte D. Luecken
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Collins Osei-Sarpong
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
| | - Stefanie Warnat-Herresthal
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jonas Schulte-Schrepping
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Allon Wagner
- Department of electrical engineering and computer science, University of California, Berkeley, CA, United States
- Center for computational biology, University of California, Berkeley, CA, United States
| | - Patrick Günther
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
| | - Carmen Pizarro
- Department of Internal Medicine II, University Hospital Bonn, Section of Pneumology, Bonn, Germany
| | - Tina Schreiber
- Department of Internal Medicine II, University Hospital Bonn, Section of Pneumology, Bonn, Germany
| | - Rainer Knoll
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Prevention, Aging & Systems Immunology, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Lisa Holsten
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Charlotte Kröger
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- Prevention, Aging & Systems Immunology, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Elena De Domenico
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Matthias Becker
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kristian Händler
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | | | | | - Heidi Theis
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Michael Kraut
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Marc H. Wadsworth
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of MIT and Harvard; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Travis K. Hughes
- Institute of Structural Biology, University Hospital, University of Bonn, Bonn, Germany
| | - Humberto J. Ferreira
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
| | - Emily Hinkley
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ines H. Kaltheuner
- Institute of Structural Biology, University Hospital, University of Bonn, Bonn, Germany
| | - Matthias Geyer
- Biochemistry & Cell Biology of Lipids, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Christoph Thiele
- University Clinics for Radiology, University Hospital Bonn, Bonn, Germany
| | - Alex K. Shalek
- Institute for Medical Engineering & Science, Department of Chemistry, and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Broad Institute of MIT and Harvard; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Andreas Feißt
- University Clinics for Radiology, University Hospital Bonn, Bonn, Germany
| | - Daniel Thomas
- University Clinics for Radiology, University Hospital Bonn, Bonn, Germany
| | | | - Marc Beyer
- Immunogenomics & Neurodegeneration, German Center for Neurodegenerative Diseases and the University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Patrick Baum
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Nir Yosef
- Department of electrical engineering and computer science, University of California, Berkeley, CA, United States
- Center for computational biology, University of California, Berkeley, CA, United States
- Chan-Zuckerberg Biohub, San Francisco, CA, United States
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
| | - Anna C. Aschenbrenner
- Prevention, Aging & Systems Immunology, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, United States
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, Netherlands
| | - Thomas Ulas
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jan Hasenauer
- Computational Life Sciences, Life & Medical Sciences (LIMES) Institute, University of Bonn, Neuherberg, Germany
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Fabian J. Theis
- Helmholtz Zentrum München - German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany, Department of Mathematics, Technical University of Munich, Munich, Germany
| | - Dirk Skowasch
- Department of Internal Medicine II, University Hospital Bonn, Section of Pneumology, Bonn, Germany
| | - Joachim L. Schultze
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE) and the University of Bonn, Bonn, Germany
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
11
|
Qiu Z, Zhong Z, Zhang Y, Tan H, Deng B, Meng G. Human umbilical cord mesenchymal stem cell-derived exosomal miR-335-5p attenuates the inflammation and tubular epithelial-myofibroblast transdifferentiation of renal tubular epithelial cells by reducing ADAM19 protein levels. Stem Cell Res Ther 2022; 13:373. [PMID: 35902972 PMCID: PMC9330665 DOI: 10.1186/s13287-022-03071-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Renal tubular epithelial-myofibroblast transdifferentiation (EMT) plays a key role in the regulation of renal fibrosis. Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs) play a crucial role in alleviating renal fibrosis and injury. Additionally, hucMSC-derived exosomes contain numerous microRNAs (miRNAs). However, it is unclear whether mesenchymal stem cells can regulate the transforming growth factor (TGF)-β1-induced EMT of human renal tubular epithelial cells (RTECs) through exosomal miRNAs. METHOD HK-2, a human RTEC line, was co-treated with TGF-β1 and hucMSC-derived exosomes. Additionally, TGF-β1-treated HK-2 cells were transfected with a miR-335-5p mimic and disintegrin and metalloproteinase domain-containing protein 19 (ADAM19)-overexpression plasmid. miR-335-5p expression and ADAM19 protein and inflammation levels were measured via quantitative reverse transcription polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assays, respectively. RESULTS TGF-β1 treatment changed the shape of HK-2 cells from a cobblestone morphology to a long spindle shape, accompanied by an increase in interleukin (IL)-6, tumor necrosis factor-α, IL-1β, collagen I, collagen III, α-smooth muscle actin, vimentin, and N-cadherin protein levels, whereas E-cadherin protein levels were reduced in these HK-2 cells, suggesting that TGF-β1 treatment induced the inflammation and EMT of HK-2 cells. HucMSC-exosomes improved the inflammation and EMT phenotype of TGF-β1-induced HK-2 cells by transferring miR-335-5p. miR-335-5p was found to bind the ADAM19 3'-untranslated region to reduce ADAM19 protein levels. Additionally, miR-335-5p improved the inflammation and EMT phenotype of HK-2 cells by reducing ADAM19 protein levels with TGF-β1 induction. CONCLUSIONS HucMSC-derived exosomal miR-335-5p attenuates the inflammation and EMT of HK-2 cells by reducing ADAM19 protein levels upon TGF-β1 induction. This study provides a potential therapeutic strategy and identifies targets for clinically treating renal fibrosis.
Collapse
Affiliation(s)
- Zhenhua Qiu
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China.
| | - Zhihui Zhong
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Yuehan Zhang
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Haoling Tan
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Bo Deng
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Guohuang Meng
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| |
Collapse
|
12
|
Guo F, Tang C, Huang B, Gu L, Zhou J, Mo Z, Liu C, Liu Y. LncRNA H19 Drives Proliferation of Cardiac Fibroblasts and Collagen Production via Suppression of the miR-29a-3p/miR-29b-3p-VEGFA/TGF-β Axis. Mol Cells 2022; 45:122-133. [PMID: 34887365 PMCID: PMC8926865 DOI: 10.14348/molcells.2021.0066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 11/27/2022] Open
Abstract
The aim of this study was to investigating whether lncRNA H19 promotes myocardial fibrosis by suppressing the miR-29a-3p/miR-29b-3p-VEGFA/TGF-β axis. Patients with atrial fibrillation (AF) and healthy volunteers were included in the study, and their biochemical parameters were collected. In addition, pcDNA3.1-H19, si-H19, and miR-29a/b-3p mimic/inhibitor were transfected into cardiac fibroblasts (CFs), and proliferation of CFs was detected by MTT assay. Expression of H19 and miR-29a/b-3p were detected using real-time quantitative polymerase chain reaction, and expression of α-smooth muscle actin (α-SMA), collagen I, collagen II, matrix metalloproteinase-2 (MMP-2), and elastin were measured by western blot analysis. The dual luciferase reporter gene assay was carried out to detect the sponging relationship between H19 and miR-29a/b-3p in CFs. Compared with healthy volunteers, the level of plasma H19 was significantly elevated in patients with AF, while miR-29a-3p and miR-29b-3p were markedly depressed (P < 0.05). Serum expression of lncRNA H19 was negatively correlated with the expression of miR-29a-3p and miR-29b-3p among patients with AF (rs = -0.337, rs = -0.236). Moreover, up-regulation of H19 expression and down-regulation of miR-29a/b-3p expression facilitated proliferation and synthesis of extracellular matrix (ECM)-related proteins. SB431542 and si-VEGFA are able to reverse the promotion of miR-29a/b-3p on proliferation of CFs and ECM-related protein synthesis. The findings of the present study suggest that H19 promoted CF proliferation and collagen synthesis by suppressing the miR-29a-3p/miR-29b-3p-VEGFA/TGF-β axis, and provide support for a potential new direction for the treatment of AF.
Collapse
Affiliation(s)
- Feng Guo
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Chengchun Tang
- Department of Cardiology, Zhongda Hospital Southeast University, Nanjing 210009, China
| | - Bo Huang
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Lifei Gu
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Jun Zhou
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Zongyang Mo
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Chang Liu
- Department of Cardiology, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Yuqing Liu
- Department of Emergency, Naval Characteristic Medical Center Affiliated to Shanghai, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
13
|
Horita M, Hsu SN, Raper A, Farquharson C, Stephen LA. miR-29b inhibits TGF-β1-induced cell proliferation in articular chondrocytes. Biochem Biophys Rep 2022; 29:101216. [PMID: 35128082 PMCID: PMC8800026 DOI: 10.1016/j.bbrep.2022.101216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 01/20/2022] [Indexed: 10/25/2022] Open
|
14
|
Guo X, Sunil C, Adeyanju O, Parker A, Huang S, Ikebe M, Tucker TA, Idell S, Qian G. PD-L1 mediates lung fibroblast to myofibroblast transition through Smad3 and β-catenin signaling pathways. Sci Rep 2022; 12:3053. [PMID: 35197539 PMCID: PMC8866514 DOI: 10.1038/s41598-022-07044-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/10/2022] [Indexed: 12/11/2022] Open
Abstract
Programmed death ligand-1 (PD-L1) is an immune checkpoint protein that has been linked with idiopathic pulmonary fibrosis (IPF) and fibroblast to myofibroblast transition (FMT). However, it remains largely unclear how PD-L1 mediates this process. We found significantly increased PD-L1 in the lungs of idiopathic pulmonary fibrosis patients and mice with pulmonary fibrosis induced by bleomycin and TGF-β. In primary human lung fibroblasts (HLFs), TGF-β induced PD-L1 expression that is dependent on both Smad3 and p38 pathways. PD-L1 knockdown using siRNA significantly attenuated TGF-β-induced expression of myofibroblast markers α-SMA, collagen-1, and fibronectin in normal and IPF HLFs. Further, we found that PD-L1 interacts with Smad3, and TGF-β induces their interaction. Interestingly, PD-L1 knockdown reduced α-SMA reporter activity induced by TGF-β in HLFs, suggesting that PD-L1 might act as a co-factor of Smad3 to promote target gene expression. TGF-β treatment also phosphorylates GSK3β and upregulates β-catenin protein levels. Inhibiting β-catenin signaling with the pharmaceutical inhibitor ICG001 significantly attenuated TGF-β-induced FMT. PD-L1 knockdown also attenuated TGF-β-induced GSK3β phosphorylation/inhibition and β-catenin upregulation, implicating GSK3β/β-catenin signaling in PD-L1-mediated FMT. Collectively, our findings demonstrate that fibroblast PD-L1 may promote pulmonary fibrosis through both Smad3 and β-catenin signaling and may represent a novel interventional target for IPF.
Collapse
Affiliation(s)
- Xia Guo
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
| | - Christudas Sunil
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
| | - Oluwaseun Adeyanju
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
| | - Andrew Parker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
| | - Steven Huang
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine at the University of Michigan, Ann Arbor, USA
| | - Mitsuo Ikebe
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
- The Texas Lung Injury Institute, Tyler, TX, USA
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA
- The Texas Lung Injury Institute, Tyler, TX, USA
| | - Guoqing Qian
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA.
| |
Collapse
|
15
|
ADAM and ADAMTS disintegrin and metalloproteinases as major factors and molecular targets in vascular malfunction and disease. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2022; 94:255-363. [PMID: 35659374 PMCID: PMC9231755 DOI: 10.1016/bs.apha.2021.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A Disintegrin and Metalloproteinase (ADAM) and A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) are two closely related families of proteolytic enzymes. ADAMs are largely membrane-bound enzymes that act as molecular scissors or sheddases of membrane-bound proteins, growth factors, cytokines, receptors and ligands, whereas ADAMTS are mainly secreted enzymes. ADAMs have a pro-domain, and a metalloproteinase, disintegrin, cysteine-rich and transmembrane domain. Similarly, ADAMTS family members have a pro-domain, and a metalloproteinase, disintegrin, and cysteine-rich domain, but instead of a transmembrane domain they have thrombospondin motifs. Most ADAMs and ADAMTS are activated by pro-protein convertases, and can be regulated by G-protein coupled receptor agonists, Ca2+ ionophores and protein kinase C. Activated ADAMs and ADAMTS participate in numerous vascular processes including angiogenesis, vascular smooth muscle cell proliferation and migration, vascular cell apoptosis, cell survival, tissue repair, and wound healing. ADAMs and ADAMTS also play a role in vascular malfunction and cardiovascular diseases such as hypertension, atherosclerosis, coronary artery disease, myocardial infarction, heart failure, peripheral artery disease, and vascular aneurysm. Decreased ADAMTS13 is involved in thrombotic thrombocytopenic purpura and microangiopathies. The activity of ADAMs and ADAMTS can be regulated by endogenous tissue inhibitors of metalloproteinases and other synthetic small molecule inhibitors. ADAMs and ADAMTS can be used as diagnostic biomarkers and molecular targets in cardiovascular disease, and modulators of ADAMs and ADAMTS activity may provide potential new approaches for the management of cardiovascular disorders.
Collapse
|
16
|
Xu X, Hong P, Wang Z, Tang Z, Li K. MicroRNAs in Transforming Growth Factor-Beta Signaling Pathway Associated With Fibrosis Involving Different Systems of the Human Body. Front Mol Biosci 2021; 8:707461. [PMID: 34381815 PMCID: PMC8350386 DOI: 10.3389/fmolb.2021.707461] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis, a major cause of morbidity and mortality, is a histopathological manifestation of many chronic inflammatory diseases affecting different systems of the human body. Two types of transforming growth factor beta (TGF-β) signaling pathways regulate fibrosis: the canonical TGF-β signaling pathway, represented by SMAD-2 and SMAD-3, and the noncanonical pathway, which functions without SMAD-2/3 participation and currently includes TGF-β/mitogen-activated protein kinases, TGF-β/SMAD-1/5, TGF-β/phosphatidylinositol-3-kinase/Akt, TGF-β/Janus kinase/signal transducer and activator of transcription protein-3, and TGF-β/rho-associated coiled-coil containing kinase signaling pathways. MicroRNA (miRNA), a type of non-coding single-stranded small RNA, comprises approximately 22 nucleotides encoded by endogenous genes, which can regulate physiological and pathological processes in fibrotic diseases, particularly affecting organs such as the liver, the kidney, the lungs, and the heart. The aim of this review is to introduce the characteristics of the canonical and non-canonical TGF-β signaling pathways and to classify miRNAs with regulatory effects on these two pathways based on the influenced organ. Further, we aim to summarize the limitations of the current research of the mechanisms of fibrosis, provide insights into possible future research directions, and propose therapeutic options for fibrosis.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Pengyu Hong
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhefu Wang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Zhangui Tang
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| | - Kun Li
- Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital and School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
17
|
Therapeutic effect of mesenchymal stem cells on histopathological, immunohistochemical, and molecular analysis in second-grade burn model. Stem Cell Res Ther 2021; 12:308. [PMID: 34051875 PMCID: PMC8164255 DOI: 10.1186/s13287-021-02365-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/30/2021] [Indexed: 12/12/2022] Open
Abstract
Background and aim Deleterious cutaneous tissue damages could result from exposure to thermal trauma, which could be ameliorated structurally and functionally through therapy via the most multipotent progenitor bone marrow mesenchymal stem cells (BM-MSCs). This study aimed to induce burns and examine the effect of BM-MSCs during a short and long period of therapy. Material and methods Ninety albino rats were divided into three groups: group I (control); group II (burn model), the animals were exposed to the preheated aluminum bar at 100°C for 15 s; and group III (the burned animals subcutaneously injected with BM-MSCs (2×106 cells/ ml)); they were clinically observed and sacrificed at different short and long time intervals, and skin samples were collected for histopathological and immunohistochemical examination and analysis of different wound healing mediators via quantitative polymerase chain reaction (qPCR). Results Subcutaneous injection of BM-MSCs resulted in the decrease of the wound contraction rate; the wound having a pinpoint appearance and regular arrangement of the epidermal layer with thin stratum corneum; decrease in the area percentages of ADAMs10 expression; significant downregulation of transforming growth factor-β (TGF-β), interleukin-6 (IL-6), tumor necrotic factor-α (TNF-α), metalloproteinase-9 (MMP-9), and microRNA-21; and marked upregulation of heat shock protein-90α (HSP-90α) especially in late stages. Conclusion BM-MSCs exhibited a powerful healing property through regulating the mediators of wound healing and restoring the normal skin structures, reducing the scar formation and the wound size.
Collapse
|
18
|
Wang J, Nie W, Xie X, Bai M, Ma Y, Jin L, Xiao L, Shi P, Yang Y, Jose PA, Armando I, Chen J, Lin W, Han F. MicroRNA-874-3p/ADAM (A Disintegrin and Metalloprotease) 19 Mediates Macrophage Activation and Renal Fibrosis After Acute Kidney Injury. Hypertension 2021; 77:1613-1626. [PMID: 33775119 DOI: 10.1161/hypertensionaha.120.16900] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Junni Wang
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Wanyun Nie
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Xishao Xie
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Mengqiu Bai
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China (M.B., P.S., W.L.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Yanhong Ma
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Lini Jin
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Liang Xiao
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Peng Shi
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China (M.B., P.S., W.L.)
| | - Yi Yang
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Pedro A Jose
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC (P.A.J., I.A.)
| | - Ines Armando
- Department of Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC (P.A.J., I.A.)
| | - Jianghua Chen
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Weiqiang Lin
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China (M.B., P.S., W.L.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| | - Fei Han
- Kidney Disease Center, The First Affiliated Hospital (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Institute of Nephrology, Zhejiang University, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.).,Key Laboratory of Kidney Disease Prevention and Control Technology, Zhejiang Province, Hangzhou, China (J.W., W.N., X.X., M.B., Y.M., L.J., L.X., Y.Y., J.C., W.L., F.H.)
| |
Collapse
|
19
|
Horita M, Farquharson C, Stephen LA. The role of miR-29 family in disease. J Cell Biochem 2021; 122:696-715. [PMID: 33529442 PMCID: PMC8603934 DOI: 10.1002/jcb.29896] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/05/2021] [Accepted: 01/10/2021] [Indexed: 02/06/2023]
Abstract
MicroRNAs are small noncoding RNAs that can bind to the target sites in the 3’‐untranslated region of messenger RNA to regulate posttranscriptional gene expression. Increasing evidence has identified the miR‐29 family, consisting of miR‐29a, miR‐29b‐1, miR‐29b‐2, and miR‐29c, as key regulators of a number of biological processes. Moreover, their abnormal expression contributes to the etiology of numerous diseases. In the current review, we aimed to summarize the differential expression patterns and functional roles of the miR‐29 family in the etiology of diseases including osteoarthritis, osteoporosis, cardiorenal, and immune disease. Furthermore, we highlight the therapeutic potential of targeting members of miR‐29 family in these diseases. We present miR‐29s as promoters of osteoblast differentiation and apoptosis but suppressors of chondrogenic and osteoclast differentiation, fibrosis, and T cell differentiation, with clear avenues for therapeutic manipulation. Further research will be crucial to identify the precise mechanism of miR‐29 family in these diseases and their full potential in therapeutics.
Collapse
Affiliation(s)
- Masahiro Horita
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| | - Colin Farquharson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| | - Louise A Stephen
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, Scotland, UK
| |
Collapse
|
20
|
Gao QY, Zhang HF, Chen ZT, Li YW, Wang SH, Wen ZZ, Xie Y, Mai JT, Wang JF, Chen YX. Construction and Analysis of a ceRNA Network in Cardiac Fibroblast During Fibrosis Based on in vivo and in vitro Data. Front Genet 2021; 11:503256. [PMID: 33552116 PMCID: PMC7859616 DOI: 10.3389/fgene.2020.503256] [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: 10/15/2019] [Accepted: 12/29/2020] [Indexed: 11/24/2022] Open
Abstract
Aims Activation of cardiac fibroblasts (CF) is crucial to cardiac fibrosis. We constructed a cardiac fibroblast-related competing endogenous RNA (ceRNA) network. Potential functions related to fibrosis of “hub genes” in this ceRNA network were explored. Materials and Methods The Gene Expression Omnibus database was searched for eligible datasets. Differentially expressed messenger (m)RNA (DE-mRNA) and long non-coding (lnc)RNA (DE-lncRNA) were identified. microRNA was predicted and validated. A predicted ceRNA network was constructed and visualized by Cytoscape, and ceRNA crosstalk was validated. A Single Gene Set Enrichment Analysis (SGSEA) was done, and the Comparative Toxicogenomics Database (CTD) was employed to analyze the most closely associated pathways and diseases of DE-mRNA in the ceRNA network. The functions of DE-mRNA and DE-lncRNA in the ceRNA network were validated by small interfering (si)RNA depletion. Results The GSE97358 and GSE116250 datasets (which described differentially expressed genes in human cardiac fibroblasts and failing ventricles, respectively) were used for analyses. Four-hundred-and-twenty DE-mRNA and 39 DE-lncRNA, and 369 DE-mRNA and 93 DE-lncRNA were identified, respectively, in the GSE97358 and GSE116250 datasets. Most of the genes were related to signal transduction, cytokine activity, and cell proliferation. Thirteen DE-mRNA with the same expression tendency were overlapped in the two datasets. Twenty-three candidate microRNAs were predicted and the expression of 11 were different. Only two DE-lncRNA were paired to any one of 11 microRNA. Finally, two mRNA [ADAM metallopeptidase domain 19, (ADAM19) and transforming growth factor beta induced, (TGFBI)], three microRNA (miR-9-5p, miR-124-3p, and miR-153-3p) and two lncRNA (LINC00511 and SNHG15) constituted our ceRNA network. siRNA against LINC00511 increased miR-124-3p and miR-9-5p expression, and decreased ADAM19 and TGFBI expression, whereas siRNA against SNHG15 increased miR-153-3p and decreased ADAM19 expression. ADAM19 and TGFBI were closely related to the TGF-β1 pathway and cardiac fibrosis, as shown by SGSEA and CTD, respectively. Depletion of two mRNA or two lncRNA could alleviate CF activation. Conclusions The CF-specific ceRNA network, including two lncRNA, three miRNA, and two mRNA, played a crucial role during cardiac fibrosis, which provided potential target genes in this field.
Collapse
Affiliation(s)
- Qing-Yuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Zhi-Teng Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Yue-Wei Li
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Shao-Hua Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Zhu-Zhi Wen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Yong Xie
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Jing-Ting Mai
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| | - Yang-Xin Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Laboratory of Cardiac Electrophysiology and Arrhythmia in Guangdong Province, Guangzhou, China
| |
Collapse
|
21
|
Yamazaki T, Mimura I, Tanaka T, Nangaku M. Treatment of Diabetic Kidney Disease: Current and Future. Diabetes Metab J 2021; 45:11-26. [PMID: 33508907 PMCID: PMC7850867 DOI: 10.4093/dmj.2020.0217] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic kidney disease (DKD) is the major cause of end-stage kidney disease. However, only renin-angiotensin system inhibitor with multidisciplinary treatments is effective for DKD. In 2019, sodium-glucose cotransporter 2 (SGLT2) inhibitor showed efficacy against DKD in Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) trial, adding a new treatment option. However, the progression of DKD has not been completely controlled. The patients with transient exposure to hyperglycemia develop diabetic complications, including DKD, even after normalization of their blood glucose. Temporary hyperglycemia causes advanced glycation end product (AGE) accumulations and epigenetic changes as metabolic memory. The drugs that improve metabolic memory are awaited, and AGE inhibitors and histone modification inhibitors are the focus of clinical and basic research. In addition, incretin-related drugs showed a renoprotective ability in many clinical trials, and these trials with renal outcome as their primary endpoint are currently ongoing. Hypoxia-inducible factor prolyl hydroxylase inhibitors recently approved for renal anemia may be renoprotective since they improve tubulointerstitial hypoxia. Furthermore, NF-E2-related factor 2 activators improved the glomerular filtration rate of DKD patients in Bardoxolone Methyl Treatment: Renal Function in chronic kidney disease/Type 2 Diabetes (BEAM) trial and Phase II Study of Bardoxolone Methyl in Patients with Chronic Kidney Disease and Type 2 Diabetes (TSUBAKI) trial. Thus, following SGLT2 inhibitor, numerous novel drugs could be utilized in treating DKD. Future studies are expected to provide new insights.
Collapse
Affiliation(s)
- Tomotaka Yamazaki
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Imari Mimura
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
- Corresponding author: Tetsuhiro Tanaka https://orcid.org/0000-0002-2238-4215 Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan E-mail:
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
22
|
Fan X, Gao Y, Zhang X, Lughmani HY, Kennedy DJ, Haller ST, Pierre SV, Shapiro JI, Tian J. A strategic expression method of miR-29b and its anti-fibrotic effect based on RNA-sequencing analysis. PLoS One 2020; 15:e0244065. [PMID: 33332475 PMCID: PMC7746150 DOI: 10.1371/journal.pone.0244065] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/02/2020] [Indexed: 02/07/2023] Open
Abstract
Tissue fibrosis is a significant health issue associated with organ dysfunction and failure. Increased deposition of collagen and other extracellular matrix (ECM) proteins in the interstitial area is a major process in tissue fibrosis. The microRNA-29 (miR-29) family has been demonstrated as anti-fibrotic microRNAs. Our recent work showed that dysregulation of miR-29 contributes to the formation of cardiac fibrosis in animal models of uremic cardiomyopathy, whereas replenishing miR-29 attenuated cardiac fibrosis in these animals. However, excessive overexpression of miR-29 is a concern because microRNAs usually have multiple targets, which could result in unknown and unexpected side effect. In the current study, we constructed a novel Col1a1-miR-29b vector using collagen 1a1 (Col1a1) promoter, which can strategically express miR-29b-3p (miR-29b) in response to increased collagen synthesis and reach a dynamic balance between collagen and miR-29b. Our experimental results showed that in mouse embryonic fibroblasts (MEF cells) transfected with Col1a1-miR-29b vector, the miR-29b expression is about 1000 times less than that in cells transfected with CMV-miR-29b vector, which uses cytomegalovirus (CMV) as a promoter for miR-29b expression. Moreover, TGF-β treatment increased the miR-29b expression by about 20 times in cells transfected with Col1a1-miR-29b, suggesting a dynamic response to fibrotic stimulation. Western blot using cell lysates and culture media demonstrated that transfection of Col1a1-miR-29b vector significantly reduced TGF-β induced collagen synthesis and secretion, and the effect was as effective as the CMV-miR-29b vector. Using RNA-sequencing analysis, we found that 249 genes were significantly altered (180 upregulated and 69 downregulated, at least 2-fold change and adjusted p-value <0.05) after TGF-β treatment in MEF cells transfected with empty vector. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using GAGE R-package showed that the top 5 upregulated pathways after TGF-β treatment were mostly fibrosis-related, including focal adhesion, ECM reaction, and TGF-β signaling pathways. As expected, transfection of Col1a1-miR-29b or CMV-miR-29b vector partially reversed the activation of these pathways. We also analyzed the expression pattern of the top 100 miR-29b targeting genes in these cells using the RNA-sequencing data. We identified that miR-29b targeted a broad spectrum of ECM genes, but the inhibition effect is mostly moderate. In summary, our work demonstrated that the Col1a1-miR-29b vector can be used as a dynamic regulator of collagen and other ECM protein expression in response to fibrotic stimulation, which could potentially reduce unnecessary side effect due to excessive miR-29b levels while remaining an effective potential therapeutic approach for fibrosis.
Collapse
Affiliation(s)
- Xiaoming Fan
- Department of Medicine, University of Toledo, Toledo, Ohio, United States of America
| | - Yingnyu Gao
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States of America
| | - Xiaolu Zhang
- Department of Medicine, University of Toledo, Toledo, Ohio, United States of America
| | - Haroon Y. Lughmani
- Department of Medicine, University of Toledo, Toledo, Ohio, United States of America
| | - David J. Kennedy
- Department of Medicine, University of Toledo, Toledo, Ohio, United States of America
| | - Steven T. Haller
- Department of Medicine, University of Toledo, Toledo, Ohio, United States of America
| | - Sandrine V. Pierre
- Marshall Institute for Interdisciplinary Research, Marshall University, Huntington, West Virginia, United States of America
| | - Joseph I. Shapiro
- Joan C. Edwards School of Medicine, Department of Biomedical Sciences, Marshall University, Huntington, West Virginia, United States of America
| | - Jiang Tian
- Joan C. Edwards School of Medicine, Department of Biomedical Sciences, Marshall University, Huntington, West Virginia, United States of America
| |
Collapse
|
23
|
Yang X, Ni J, Li Y, Zou L, Guo T, Li Y, Chu L, Zhu Z. LncRNA-RP11 Modulates TGF-β1-Activated Radiation-Induced Lung Injury Through Downregulating microRNA-29a. Dose Response 2020; 18:1559325820949071. [PMID: 33117089 PMCID: PMC7573740 DOI: 10.1177/1559325820949071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Radiation-induced lung injury (RILI) is one of the most serious complications of thoracic radiation and TGF-β1 is a central regulator of RILI. However, the molecular mechanism underlying the fine tuning of TGF-β1 signaling in RILI has not been fully understood. In the current study, differentially expressed long non-coding RNAs (LncRNAs) among human lung fibroblasts cell lines HFL-1 and WI-38 treated with TGF-β1, were identified by microarray and validated by real time PCR. LncRNA-RP11 was found to be the most increased LncRNA and it mediated the promotion of fibrogenic activity in human lung fibroblasts after TGF-β1 treatment. Bioinformatic analysis revealed that TGF-β1 may be associated with the component and structure of extracellular matrix in lung fibroblasts cells, and LncRNA-RP11 was predicted and confirmed to be a competing endogenous RNA by directly binding to miR-29a. Functional experiments investigating the biological role of LncRNA-RP11/miR-29a axis in RILI, were then carried out in human fibroblasts. The results showed that radiation promoted the expression of LncRNA-RP11, but regressed the expression of miR-29a. Furthermore, radiation elevated the expression of various common collagenic proteins, which could be abolished by overexpression of miR-29a.
Collapse
Affiliation(s)
- Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yida Li
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuan Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Li Chu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Mirani Sargazi F, Alidadi A, Taheri H, Heidari Nia M, Sargazi S, Saravani R, Malek Raisi H. Functional miR29a gene polymorphism enhanced the risk of chronic kidney disease in an Iranian population: A preliminary case-control study and bioinformatics analyses. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
25
|
Functional analysis of miRNAs combined with TGF-β1/Smad3 inhibitor in an intrauterine rat adhesion cell model. Mol Cell Biochem 2020; 470:15-28. [PMID: 32447720 DOI: 10.1007/s11010-020-03741-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
In this study, we aimed to study the role of miRNAs in intrauterine adhesion (IUA) disease. An IUA cell model was constructed by TGF-β1. Smad3 inhibitor (SIS3) can inhibit the Smad3 signaling pathway and affect the role of TGF-β1; thus, it was used to identify the role of Smad3 and related miRNAs in IUA. Cell number significantly increased in the TGF-β1 group after 72 h and 96 h, respectively, compared with that in the control group (P < 0.05). However, cell proliferation was significantly decreased in the TGF-β1 + SIS3 group (P < 0.0001). Cell apoptosis was increased in the TGF-β1 + SIS3 group compared with that in the TGF-β1 group. Western Blot (WB) analysis suggested that TGF-β1 treatment could effectively increase the expression of α-SMA, COL1, Smad3, and p-Smad3, which could be inhibited by SIS3 treatment. A total of 235 and 530 differentially expressed miRNAs in the TGF-β1 + SIS3 group were significantly up- and downregulated compared with those in the TGF-β1 group, respectively. These differentially expressed miRNAs were enriched in the MAPK and PI3K-AKT pathways. The ten most differentially expressed miRNAs were selected to verify their expressions using quantitative real-time polymerase chain reaction (qPCR). Furthermore, overexpression of rno-miR-3586-3p and rno-miR-455-5p can promote cell proliferation and exacerbate the IUA pathogenic process. However, overexpression of rno-miR-204-3p and rno-miR-3578 can inhibit cell behavior and IUA progression. The above results can provide detailed information for the understanding of IUA molecular mechanisms.
Collapse
|
26
|
Delić D, Wiech F, Urquhart R, Gabrielyan O, Rieber K, Rolser M, Tsuprykov O, Hasan AA, Krämer BK, Baum P, Köhler A, Gantner F, Mark M, Hocher B, Klein T. Linagliptin and telmisartan induced effects on renal and urinary exosomal miRNA expression in rats with 5/6 nephrectomy. Sci Rep 2020; 10:3373. [PMID: 32099009 PMCID: PMC7042229 DOI: 10.1038/s41598-020-60336-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 02/11/2020] [Indexed: 11/10/2022] Open
Abstract
Dipeptidyl peptidase 4 inhibitors and angiotensin II receptor blockers attenuate chronic kidney disease progression in experimental diabetic and non-diabetic nephropathy in a blood pressure and glucose independent manner, but the exact molecular mechanisms remain unclear. MicroRNAs (miRNAs) are short, non-coding RNA species that are important post-transcriptional regulators of gene expression and play an important role in the pathogenesis of nephropathy. miRNAs are present in urine in a remarkably stable form, packaged in extracellular vesicles. Here, we investigated linagliptin and telmisartan induced effects on renal and urinary exosomal miRNA expression in 5/6 nephrectomized rats. In the present study, renal miRNA profiling was conducted using the Nanostring nCounter technology and mRNA profiling using RNA sequencing from the following groups of rats: sham operated plus placebo; 5/6 nephrectomy plus placebo; 5/6 nephrectomy plus telmisartan; and 5/6 nephrectomy plus linagliptin. TaqMan Array miRNA Cards were used to evaluate which of the deregulated miRNAs in the kidney are present in urinary exosomes. In kidneys from 5/6 nephrectomized rats, the expression of 13 miRNAs was significantly increased (>1.5-fold, P < 0.05), whereas the expression of 7 miRNAs was significantly decreased (>1.5-fold, P < 0.05). Most of the deregulated miRNA species are implicated in endothelial-to-mesenchymal transition and inflammatory processes. Both telmisartan and linagliptin suppressed the induction of pro-fibrotic miRNAs, such as miR-199a-3p, and restored levels of anti-fibrotic miR-29c. In conclusion, the linagliptin and telmisartan-induced restorative effects on miR-29c expression were reflected in urinary exosomes, suggesting that miRNA profiling of urinary exosomes might be used as a biomarker for CKD progression and monitoring of treatment effects.
Collapse
Affiliation(s)
- Denis Delić
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany.
| | - Franziska Wiech
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Richard Urquhart
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Ogsen Gabrielyan
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Kathrin Rieber
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Marcel Rolser
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Oleg Tsuprykov
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Ahmed A Hasan
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.,Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Baum
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Andreas Köhler
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Florian Gantner
- Boehringer Ingelheim Pharma GmbH & Co. KG, Translational Medicine & Clinical Pharmacology, Biberach, Germany
| | - Michael Mark
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardiometabolic Diseases Research, Biberach, Germany
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany.
| | - Thomas Klein
- Boehringer Ingelheim Pharma GmbH & Co. KG, Cardiometabolic Diseases Research, Biberach, Germany
| |
Collapse
|
27
|
Maurer S, Kopp HG, Salih HR, Kropp KN. Modulation of Immune Responses by Platelet-Derived ADAM10. Front Immunol 2020; 11:44. [PMID: 32117229 PMCID: PMC7012935 DOI: 10.3389/fimmu.2020.00044] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 01/09/2020] [Indexed: 12/15/2022] Open
Abstract
Platelets have a crucial function in maintaining hemostasis. However, beyond their role in coagulation and thrombus formation, platelets have been implicated to affect various pathophysiological conditions such as infectious diseases, autoimmune disorders, and cancer. It is well-established that platelets aid local cancer growth by providing growth factors or contributing to cancer angiogenesis. In addition, they promote metastasis, among others by facilitation of tumor cell-extravasation and epithelial-to-mesenchymal-like transition as well as protecting metastasizing cancer cells from immunosurveillance. A variety of membrane-bound and soluble platelet-derived factors are involved in these processes, and many aspects of platelet biology in both health and disease are regulated by platelet-associated metalloproteinases and their inhibitors. Platelets synthesize (i) members of the matrix metalloproteinase (MMP) family and also inhibitors of MMPs such as members of the "tissue inhibitor of metalloproteinases" (TIMP) family as well as (ii) members of the "a disintegrin and metalloproteinase" (ADAM) family including ADAM10. Notably, platelet-associated metalloproteinase activity not only influences functions of platelets themselves: platelets can also induce expression and/or release of metalloproteinases e.g., in leukocytes or cancer cells, and ADAMs are emerging as important components by which platelets directly affect other cell types and function. This review outlines the function of metalloproteinases in platelet biology with a focus on ADAM10 and discusses the role of platelet-derived metalloproteinases in the interaction of platelets with components of the immune system and/or cancer cells.
Collapse
Affiliation(s)
- Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany.,Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Hans-Georg Kopp
- Departments of Molecular Oncology and Thoracic Oncology, Robert-Bosch-Hospital Stuttgart, Stuttgart, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany.,DFG Cluster of Excellence 2180 'Image-guided and Functional Instructed Tumor Therapy' (IFIT), University of Tuebingen, Tubingen, Germany
| | - Korbinian N Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, Mainz, Germany
| |
Collapse
|
28
|
MicroRNA-29a Mitigates Subacromial Bursa Fibrosis in Rotator Cuff Lesion with Shoulder Stiffness. Int J Mol Sci 2019; 20:ijms20225742. [PMID: 31731750 PMCID: PMC6888443 DOI: 10.3390/ijms20225742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/11/2019] [Accepted: 11/12/2019] [Indexed: 02/08/2023] Open
Abstract
Rotator cuff lesion with shoulder stiffness is a major cause of shoulder pain and motionlessness. Subacromial bursa fibrosis is a prominent pathological feature of the shoulder disorder. MicroRNA-29a (miR-29a) regulates fibrosis in various tissues; however, the miR-29a action to subacromial bursa fibrosis remains elusive. Here, we reveal that subacromial synovium in patients with rotator cuff tear with shoulder stiffness showed severe fibrosis, hypertrophy, and hyperangiogenesis histopathology along with significant increases in fibrotic matrices collagen (COL) 1A1, 3A1, and 4A1 and inflammatory cytokines, whereas miR-29a expression was downregulated. Supraspinatus and infraspinatus tenotomy-injured shoulders in transgenic mice overexpressing miR-29a showed mild swelling, vascularization, fibrosis, and regular gait profiles as compared to severe rotator cuff damage in wild-type mice. Treatment with miR-29a precursor compromised COL3A1 production and hypervascularization in injured shoulders. In vitro, gain of miR-29a function attenuated COL3A1 expression through binding to the 3’-untranslated region (3′-UTR) of COL3A1 in inflamed tenocytes, whereas silencing miR-29a increased the matrix expression. Taken together, miR-29a loss is correlated with subacromial bursa inflammation and fibrosis in rotator cuff tear with shoulder stiffness. miR-29a repressed subacromial bursa fibrosis through directly targeting COL3A1 mRNA, improving rotator cuff integrity and shoulder function. Collective analysis offers a new insight into the molecular mechanism underlying rotator cuff tear with shoulder stiffness. This study also highlights the remedial potential of miR-29a precursor for alleviating the shoulder disorder.
Collapse
|
29
|
Naciri I, Laisné M, Ferry L, Bourmaud M, Gupta N, Di Carlo S, Huna A, Martin N, Peduto L, Bernard D, Kirsh O, Defossez PA. Genetic screens reveal mechanisms for the transcriptional regulation of tissue-specific genes in normal cells and tumors. Nucleic Acids Res 2019; 47:3407-3421. [PMID: 30753595 PMCID: PMC6468300 DOI: 10.1093/nar/gkz080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
The proper tissue-specific regulation of gene expression is essential for development and homeostasis in metazoans. However, the illegitimate expression of normally tissue-restricted genes—like testis- or placenta-specific genes—is frequently observed in tumors; this promotes transformation, but also allows immunotherapy. Two important questions are: how is the expression of these genes controlled in healthy cells? And how is this altered in cancer? To address these questions, we used an unbiased approach to test the ability of 350 distinct genetic or epigenetic perturbations to induce the illegitimate expression of over 40 tissue-restricted genes in primary human cells. We find that almost all of these genes are remarkably resistant to reactivation by a single alteration in signaling pathways or chromatin regulation. However, a few genes differ and are more readily activated; one is the placenta-expressed gene ADAM12, which promotes invasion. Using cellular systems, an animal model, and bioinformatics, we find that a non-canonical but druggable TGF-β/KAT2A/TAK1 axis controls ADAM12 induction in normal and cancer cells. More broadly, our data show that illegitimate gene expression in cancer is an heterogeneous phenomenon, with a few genes activatable by simple events, and most genes likely requiring a combination of events to become reactivated.
Collapse
Affiliation(s)
- Ikrame Naciri
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Marthe Laisné
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Laure Ferry
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Morgane Bourmaud
- INSERM U1132 and USPC Paris-Diderot, Hôpital Lariboisière, Paris, France
| | - Nikhil Gupta
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Selene Di Carlo
- Unité Stroma, Inflammation & Tissue Repair, Institut Pasteur, 75724 Paris, France; INSERM U1224, 75724 Paris, France
| | - Anda Huna
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Lucie Peduto
- Unité Stroma, Inflammation & Tissue Repair, Institut Pasteur, 75724 Paris, France; INSERM U1224, 75724 Paris, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Olivier Kirsh
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Pierre-Antoine Defossez
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| |
Collapse
|
30
|
Abstract
As one type of the most common endogenous short noncoding RNAs (ncRNAs), microRNAs (miRNAs) act as posttranscriptional regulators of gene expression and have great potential biological functions in the physiological and pathological processes of various diseases. The role of miRNAs in renal fibrosis has also attracted great attention in the previous 20 years, and new therapeutic strategies targeting miRNAs appear to be promising. Some researchers have previously reviewed the roles of miRNA in renal fibrosis disease, but numerous studies have emerged over the recent 5 years. It is necessary to update and summarize research progress in miRNAs in renal fibrosis. Thus, in this review, we summarize progress in miRNA-mediated renal fibrosis over the last 5 years and evaluate the biological functions of some miRNAs in different stages of renal fibrosis. Furthermore, we also expound the recent clinical applications of these miRNAs to provide new insights into the treatment of renal fibrosis disease.
Collapse
Affiliation(s)
- Youling Fan
- Department of Anesthesiology, The First People's Hospital of Kashgar, Xinjiang Province, China.,Department of Anesthesiology, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Hongtao Chen
- Department of Anesthesiology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Zhenxing Huang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Hong Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Province, China
| | - Jun Zhou
- Department of Anesthesiology, The third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
31
|
Srivastava SP, Hedayat AF, Kanasaki K, Goodwin JE. microRNA Crosstalk Influences Epithelial-to-Mesenchymal, Endothelial-to-Mesenchymal, and Macrophage-to-Mesenchymal Transitions in the Kidney. Front Pharmacol 2019; 10:904. [PMID: 31474862 PMCID: PMC6707424 DOI: 10.3389/fphar.2019.00904] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022] Open
Abstract
microRNAs (miRNAs) are small, non-coding nucleotides that regulate diverse biological processes. Altered microRNA biosynthesis or regulation contributes to pathological processes including kidney fibrosis. Kidney fibrosis is characterized by deposition of excess extracellular matrix (ECM), which is caused by infiltration of immune cells, inflammatory cells, altered chemokines, and cytokines as well as activation and accumulation of fibroblasts in the kidney. These activated fibroblasts can arise from epithelial cells via epithelial-to-mesenchymal transition (EMT), from bone marrow-derived M2 phenotype macrophages via macrophage-to-mesenchymal transition (MMT), from endothelial cells via endothelial-to-mesenchymal transition (EndMT), from resident fibroblasts, and from bone marrow-derived monocytes and play a crucial role in fibrotic events. Disrupted microRNA biosynthesis and aberrant regulation contribute to the activation of mesenchymal programs in the kidney. miR-29 regulates the interaction between dipeptidyl peptidase-4 (DPP-4) and integrin β1 and the associated active transforming growth factor β (TGFβ) and pro-EndMT signaling; however, miR-let-7 targets transforming growth factor β receptors (TGFβRs) to inhibit TGFβ signaling. N-acetyl-seryl-aspartyl-lysyl-proline (AcSDKP) is an endogenous anti-fibrotic peptide, which is associated with fibroblast growth factor receptor 1 (FGFR1) phosphorylation and subsequently responsible for the production of miR-let-7. miR-29 and miR-let-7 family clusters participate in crosstalk mechanisms, which are crucial for endothelial cell homeostasis. The physiological level of AcSDKP is vital for the activation of anti-fibrotic mechanisms including restoration of anti-fibrotic microRNA crosstalk and suppression of profibrotic signaling by mitigating DPP-4-associated mesenchymal activation in the epithelial cells, endothelial cells, and M2 phenotype macrophages. The present review highlights recent advancements in the understanding of both the role of microRNAs in the development of kidney disease and their potential as novel therapeutic targets for fibrotic disease states.
Collapse
Affiliation(s)
| | - Ahmad Fahim Hedayat
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Julie E Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
32
|
Horita M, Nishida K, Hasei J, Furumatsu T, Sakurai M, Onodera Y, Fukuda K, Salter DM, Ozaki T. Involvement of ADAM12 in Chondrocyte Differentiation by Regulation of TGF-β1-Induced IGF-1 and RUNX-2 Expressions. Calcif Tissue Int 2019; 105:97-106. [PMID: 30993375 DOI: 10.1007/s00223-019-00549-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Accepted: 04/10/2019] [Indexed: 01/28/2023]
Abstract
A disintegrin and metalloproteinase 12 (ADAM12) is known to be involved in chondrocyte proliferation and maturation; however, the mechanisms are not fully understood. In this study, expression and localization of ADAM12 during chondrocyte differentiation were examined in the mouse growth plate by immunohistochemistry. Adam12 expression during ATDC5 chondrogenic differentiation was examined by real-time PCR and compared with the expression pattern of type X collagen. The clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 system was used to generate Adam12-knockout (KO) ATDC5 cells. Adam12-KO and Adam12 overexpressing cells were used for analyses of ADAM12 expression with or without TGF-β1 stimulation. ADAM12 was identified predominantly in chondrocytes of the proliferative zone in mouse growth plates by immunohistochemistry. Adam12 was upregulated prior to Col10a1 during chondrogenic differentiation in wild-type ATDC5 cells. In Adam12-KO ATDC5 cells, following initiation of chondrogenic differentiation, we observed a reduction in Igf-1 expression along with an upregulation of hypertrophy-associated Runx2, Col10a1, and type X collagen protein expressions. In ATDC5 wild-type cells, stimulation with TGF-β1 upregulated the expressions of Adam12 and Igf-1 and downregulated the expression of Runx2. In contrast, in Adam12-KO ATDC5 cells, these TGF-β1-induced changes were suppressed. Adam12 overexpression resulted in an upregulation of Igf-1 and downregulation of Runx2 expression in ATDC5 cells. The findings suggest that ADAM12 has important role in the regulation of chondrocyte differentiation, potentially by regulation of TGF-β1-dependent signaling and that targeting of ADAM12 may have a role in management of abnormal chondrocyte differentiation.
Collapse
Affiliation(s)
- Masahiro Horita
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keiichiro Nishida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Joe Hasei
- Department of Sports Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Miwa Sakurai
- Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe (FBRI), Kobe, Japan
| | - Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University Faculty of Medicine, Osaka, Japan
| | - Donald M Salter
- Centre for Genomic and Experimental Medicine, IGMM - University of Edinburgh, Edinburgh, UK
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
33
|
Control of lung myofibroblast transformation by monovalent ion transporters. CURRENT TOPICS IN MEMBRANES 2019. [PMID: 31196603 DOI: 10.1016/bs.ctm.2019.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Abstract
Myofibroblast differentiation is a critical process in the pathogenesis of tissue fibrosis. We focus our mini-review on recent data showing an implication of monovalent ion transporters in fibroblast to myofibroblast transformation of human lung fibroblasts (HLF). In cultured HLF, cardiotonic steroids (CTS) known as potent inhibitors of Na+,K+-ATPase suppress myofibroblast differentiation in parallel with up- and down-regulated expression of cyclooxygenase-2 (COX-2) and TGF-β receptor subunit TGFBR2, respectively. K+-free medium mimics antifibrotic action of CTS indicating a key role of elevated intracellular [Na+]i/[K+]i ratio. Augmented expression of COX-2 is abolished by inhibition of Na+/Ca2+ exchanger. Side-by-side with CTS acting via elevation of the [Na+]i/[K+]i ratio fibroblast to myofibroblast transformation is also suppressed by potent inhibitors of Ca2+-activated chloride channels tannic acid and K+,Cl- cotransporter DIOA. The relative impact of [Formula: see text] -mediated and -independent signaling triggered by elevated [Na+]i/[K+]i ratio and altered intracellular anion handling in transcriptomic changes involved in myofibroblast differentiation should be examined further.
Collapse
|
34
|
A therapeutic approach towards microRNA29 family in vascular diabetic complications: A boon or curse? J Diabetes Metab Disord 2019; 18:243-254. [PMID: 31275895 DOI: 10.1007/s40200-019-00409-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 05/02/2019] [Indexed: 02/06/2023]
Abstract
Diabetes Mellitus (DM) is one of the major metabolic disorders and its severity leads to death. Enhancement in hyperglycaemic conditions of DM gives rise to endothelial impairment in small and large blood vessels contributing towards microvascular and macrovascular complications respectively. The pathogenesis of diabetic complications is associated with interruption of various signal transduction pathways due to epigenetic modifications such as aberrant histone modifications, DNA methylation and expression of miRNAs along with the long non-coding RNAs (lncRNAs). Amongst these epigenetic alterations, modulated expressions of miRNAs confer to apoptosis and endothelial dysfunction of organs that gives rise to vascular complications. In this review, we principally focussed on physiological role of miR29 family in DM and have discussed crosstalk between miR29 family and numerous genes involved in signal transduction pathways of Diabetic vascular complications. Incidences of diabetic retinopathy exploiting the role of miR29 in regulation of EMT process, differential expression patterns of miR29 and promising therapeutic role of miR29 have been discussed. We have summarised the therapeutic role of miR29 in podocyte impairment and how miR29 regulates the expressions of profibrotic, inflammatory and ECM encoding genes in renal fibrosis under diabetic conditions. We have also highlighted impact of miR29 expression patterns in cardiac angiopathy, cardiomyocyte's apoptosis and cardiac fibrosis. Additionally, we have also presented the contradictory actions of miR29 family in amelioration as well as in enhancement of diabetic complications.
Collapse
|
35
|
Non-Coding RNAs as New Therapeutic Targets in the Context of Renal Fibrosis. Int J Mol Sci 2019; 20:ijms20081977. [PMID: 31018516 PMCID: PMC6515288 DOI: 10.3390/ijms20081977] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/15/2022] Open
Abstract
Fibrosis, or tissue scarring, is defined as the excessive, persistent and destructive accumulation of extracellular matrix components in response to chronic tissue injury. Renal fibrosis represents the final stage of most chronic kidney diseases and contributes to the progressive and irreversible decline in kidney function. Limited therapeutic options are available and the molecular mechanisms governing the renal fibrosis process are complex and remain poorly understood. Recently, the role of non-coding RNAs, and in particular microRNAs (miRNAs), has been described in kidney fibrosis. Seminal studies have highlighted their potential importance as new therapeutic targets and innovative diagnostic and/or prognostic biomarkers. This review will summarize recent scientific advances and will discuss potential clinical applications as well as future research directions.
Collapse
|
36
|
Abstract
Myofibroblast activation is a critical process in the pathogenesis of tissue fibrosis accounting for 45% of all deaths. No effective therapies are available for the treatment of fibrotic diseases. We focus our mini-review on recent data showing that cardiotonic steroids (CTS) that are known as potent inhibitors of Na+,K+-ATPase affect myofibroblast differentiation in a cell type-specific manner. In cultured human lung fibroblasts (HLF), epithelial cells, and cancer-associated fibroblasts, CTS blocked myofibroblast differentiation triggered by profibrotic cytokine TGF-β. In contrast, in the absence of TGF-β, CTS augmented myofibroblast differentiation of cultured cardiac fibroblasts. The cell type-specific action of CTS in myofibroblast differentiation is consistent with data obtained in in vivo studies. Thus, infusion of ouabain via osmotic mini-pumps attenuated the development of lung fibrosis in bleomycintreated mice, whereas marinobufagenin stimulated renal and cardiac fibrosis in rats with experimental renal injury. In TGF-β-treated HLF, suppression of myofibroblast differentiation by ouabain is mediated by elevation of the [Na+]i/[K+]i ratio and is accompanied by upregulation of cyclooxygenase COX-2 and downregulation of TGF-β receptor TGFBR2. Augmented expression of COX-2 is abolished by inhibition of Na+/Ca2+ exchanger, suggesting a key role of [Ca2+]i-mediated signaling. What is the relative impact in tissue fibrosis of [Na+]i,[K+]iindependent signaling documented in several types of CTS-treated cells? Do the different conformational transitions of Na+,K+-ATPase α1 subunit in the presence of ouabain and marinobufagenin contribute to their distinct involvement in myofibroblast differentiation? Additional experiments should be done to answer these questions and to develop novel pharmacological approaches for the treatment of fibrosis-related disorders.
Collapse
Affiliation(s)
- Sergei N. Orlov
- Faculty of Biology, Lomonosov Moscow State University, Russian Federation
| | - Jennifer La
- Department of Medicine, The University of Chicago, IL, United States
| | | | - Nickolai O. Dulin
- Department of Medicine, The University of Chicago, IL, United States
| |
Collapse
|
37
|
Feng G, Zha Z, Huang Y, Li J, Wang Y, Ke W, Chen H, Liu L, Song Y, Ge Z. Sustained and Bioresponsive Two-Stage Delivery of Therapeutic miRNA via Polyplex Micelle-Loaded Injectable Hydrogels for Inhibition of Intervertebral Disc Fibrosis. Adv Healthc Mater 2018; 7:e1800623. [PMID: 30296017 DOI: 10.1002/adhm.201800623] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/09/2018] [Indexed: 02/05/2023]
Abstract
Intervertebral disc degeneration (IDD) is frequently caused by gradual pathological changes inside intervertebral discs (IVDs) and progressive fibrosis. MicroRNA-29 (miR-29) family possesses potent fibrosis suppression capability, but their application for treatment of chronic IDD is limited due to lack of suitable local delivery systems. In this report, given various overexpressed matrix metalloproteinases (MMPs) during IDD, injectable MMP-degradable hydrogels encapsulating MMP-responsive polyplex micelles are developed for sustained and bioresponsive delivery of miR-29a into nucleus pulposus cells via a two-stage process. Cationic block copolymers are designed to complex miR-29a, and subsequently mixed with the poly(ethylene glycol) (PEG) gelation precursors and MMP-cleavable peptide cross-linkers for in situ formation of polyplex micelle-encapsulated hydrogels in the diseased IVDs. In the presence of MMPs, the polyplex micelles are first released by MMP cleavage of the hydrogels, and subsequently, MMPs-responsive detachment of PEG shells from polyplex micelles contributes to efficient cellular uptake and endosomal escape. MiR-29a is demonstrated to effectively silence the expression of MMP-2, inhibit the fibrosis process, and reverse IDD in animal models through blocking the β-catenin translocation pathway from the cytoplasm to the nucleus. This two-stage bioresponsive local miRNA delivery system represents a novel and promising strategy for the treatment of chronic IDD.
Collapse
Affiliation(s)
- Ganjun Feng
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Zengshi Zha
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Yong Huang
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Junjie Li
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Yuheng Wang
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Wendong Ke
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| | - Hongying Chen
- Technology Center for Public Research; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Limin Liu
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Yueming Song
- Department of Orthopedic Surgery; West China Hospital; Sichuan University; Chengdu 610041 Sichuan China
| | - Zhishen Ge
- CAS Key Laboratory of Soft Matter Chemistry; Department of Polymer Science and Engineering; University of Science and Technology of China; Hefei 230026 Anhui China
| |
Collapse
|
38
|
Ma Z, Wei Q, Zhang M, Chen JK, Dong Z. Dicer deficiency in proximal tubules exacerbates renal injury and tubulointerstitial fibrosis and upregulates Smad2/3. Am J Physiol Renal Physiol 2018; 315:F1822-F1832. [PMID: 30280598 DOI: 10.1152/ajprenal.00402.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Renal fibrosis is a common pathological feature in chronic kidney disease (CKD), including diabetic kidney disease (DKD) and obstructive nephropathy. Multiple microRNAs have been implicated in the pathogenesis of both DKD and obstructive nephropathy, although the overall role of microRNAs in tubular injury and renal fibrosis in CKD is unclear. Dicer (a key RNase III enzyme for microRNA biogenesis) was specifically ablated from kidney proximal tubules in mice via the Cre-lox system to deplete micoRNAs. Proximal tubular Dicer knockout (PT- Dicer KO) mice and wild-type (WT) littermates were subjected to streptozotocin (STZ) treatment to induce DKD or unilateral ureteral obstruction (UUO) to induce obstructive nephropathy. Renal hypertrophy, renal tubular apoptosis, kidney inflammation, and tubulointerstitial fibrosis were examined. Compared with WT mice, PT- Dicer KO mice showed more severe tubular injury and renal inflammation following STZ treatment. These mice also developed higher levels of tubolointerstitial fibrosis. Meanwhile, PT- Dicer KO mice had a significantly higher Smad2/3 expression in kidneys than WT mice (at 6 mo of age) in both control and STZ-treated mice. Similarly, UUO induced more severe renal injury, inflammation, and interstitial fibrosis in PT- Dicer KO mice than WT. Although we did not detect obvious Smad2/3 expression in sham-operated mice (2-3 mo old), significantly more Smad2/3 was induced in obstructed PT- Dicer KO kidneys. These results supported a protective role of Dicer-dependent microRNA synthesis in renal injury and fibrosis development in CKD, specifically in DKD and obstructive nephropathy. Depletion of Dicer and microRNAs may upregulate Smad2/3-related signaling pathway to enhance the progression of CKD.
Collapse
Affiliation(s)
- Zhengwei Ma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| | - Ming Zhang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| | - Jian-Kang Chen
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center , Augusta, Georgia
| |
Collapse
|
39
|
Huang YQ, Huang C, Li J, Zhang B, Feng YQ. The association of miR-29a with proteinuria in essential hypertension. J Hum Hypertens 2018; 32:775-780. [PMID: 30127486 DOI: 10.1038/s41371-018-0097-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/12/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Recently, miRNAs have emerged as new indirect markers of inflammation that are associated with adverse outcomes in cardiovascular disease. The aim of the study was to evaluate the relationship between miR29a and proteinuria in hypertension. Fifty patients with normal albuminuria, fifty patients with micro-albuminuria, and fifty patients with macro-albuminuria were enrolled. The highest levels of miR-29a and transforming growth factor-β1 (TGF-β1) were observed in the macro-albuminuria group, followed by the micro-albuminuria and the normal albuminuria groups. The level of miR-29a was negatively correlated with the glomerular filtration rate, but was positively correlated with C-reactive protein, TGF-β1, and the urinary albumin to creatinine ratio (UACR). Circulating miR-29a was found to be significantly and independently associated with proteinuria. Our findings showed that miR-29a reflects the pathogenesis of hypertensive nephropathy and may serve as a potential non-invasive marker for detecting early stages of hypertensive nephropathy.
Collapse
Affiliation(s)
- Yu-Qing Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Cheng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Jie Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Bin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China.
| | - Ying-Qing Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China.
| |
Collapse
|
40
|
Drummond CA, Fan X, Haller ST, Kennedy DJ, Liu J, Tian J. Na/K-ATPase signaling mediates miR-29b-3p regulation and cardiac fibrosis formation in mice with chronic kidney disease. PLoS One 2018; 13:e0197688. [PMID: 29775473 PMCID: PMC5959191 DOI: 10.1371/journal.pone.0197688] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/07/2018] [Indexed: 01/19/2023] Open
Abstract
The Na/K-ATPase is an important membrane ion transporter and a signaling receptor that is essential for maintaining normal cell function. The current study examined the role of Na/K-ATPase signaling in regulating miR-29b-3p, an anti-fibrotic microRNA, in a mouse chronic kidney disease (CKD) model (5/6th partial nephrectomy or PNx). The results showed that CKD induced significant reduction of miR-29b-3p expression in the heart tissue by activation of Src and NFκB signaling in these animals. To demonstrate the role of Na/K-ATPase signaling, we also performed the PNx surgery on Na/K-ATPase α1 heterozygous (α1+/-) mice, which expresses ~40% less Na/K-ATPase α1 compared to their wild type littermates (WT) and exhibits deficiency in Na/K-ATPase signaling. We found that CKD did not significantly change the miR-29b-3p expression in heart tissue from the α1+/- animals. We also found that CKD failed to activate Src and NFκB signaling in these animals. Using isolated cardiac fibroblasts from α1+/- mice and their WT littermates, we showed that ouabain, a specific Na/K-ATPase ligand, induces decreased miR-29b-3p expression in fibroblasts isolated from WT mice, but had no effect in cells from α1+/- mice. Inhibition of NFκB by Bay11-7082 prevented ouabain-induced miR-29b-3p reduction in WT fibroblasts. To further confirm the in vivo effect of Na/K-ATPase signaling in regulation of miR-29b-3p and cardiac fibrosis in CKD animals, we used pNaKtide, a Src inhibiting peptide derived from the sequence of Na/K-ATPase, to block the activation of Na/K-ATPase signaling. The result showed that pNaKtide injection significantly increased miR-29b-3p expression and mitigated the CKD-induced cardiac fibrosis in these animals. These results clearly demonstrated that Na/K-ATPase signaling is an important mediator in CKD that regulates miR-29b-3p expression and cardiac fibrosis, which provides a novel target for regulation of miR-29b-3p in CKD. We also demonstrate that antagonizing Na/K-ATPase signaling by pNaKtide can reduce organ fibrosis through the stimulation of tissue miR-29b-3p expression.
Collapse
Affiliation(s)
| | - Xiaoming Fan
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - Steven T. Haller
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - David J. Kennedy
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, United States of America
| | - Jiang Tian
- Department of Medicine at the University of Toledo, Toledo, OH, United States of America
- * E-mail:
| |
Collapse
|
41
|
Duong TE, Hagood JS. Epigenetic Regulation of Myofibroblast Phenotypes in Fibrosis. CURRENT PATHOBIOLOGY REPORTS 2018; 6:79-96. [PMID: 30271681 DOI: 10.1007/s40139-018-0155-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Purpose of Review Myofibroblasts are the fundamental drivers of fibrosing disorders; there is great value in better defining epigenetic networks involved in myofibroblast behavior. Complex epigenetic paradigms, which are likely organ and/or disease specific, direct pathologic myofibroblast phenotypes. In this review, we highlight epigenetic regulators and the mechanisms through which they shape myofibroblast phenotype in fibrotic diseases of different organs. Recent Findings Hundreds of genes and their expression contribute to the myofibroblast transcriptional regime influencing myofibroblast phenotype. An increasingly large number of epigenetic modifications have been identified in the regulation of these signaling pathways driving myofibroblast activation and disease progression. Drugs that inhibit or reverse profibrotic epigenetic modifications have shown promise in vitro and in vivo; however, no current epigenetic therapies have been approved to treat fibrosis. Newly described epigenetic mechanisms will be mentioned, along with potential therapeutic targets and innovative strategies to further understand myofibroblast-directed fibrosis. Summary Epigenetic regulators that direct myofibroblast behavior and differentiation into pathologic myofibroblast phenotypes in fibrotic disorders comprise both overlapping and organ-specific epigenetic mechanisms.
Collapse
Affiliation(s)
- Thu Elizabeth Duong
- Division of Pediatric Respiratory Medicine, University of California-San Diego, La Jolla, California.,Division of Respiratory Medicine, Rady Children's Hospital of San Diego, San Diego, California
| | - James S Hagood
- Division of Pediatric Respiratory Medicine, University of California-San Diego, La Jolla, California.,Division of Respiratory Medicine, Rady Children's Hospital of San Diego, San Diego, California
| |
Collapse
|
42
|
Qi H, Liu Y, Li S, Chen Y, Li L, Cao Y, E M, Shi P, Song C, Li B, Sun H. Activation of AMPK Attenuated Cardiac Fibrosis by Inhibiting CDK2 via p21/p27 and miR-29 Family Pathways in Rats. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:277-290. [PMID: 28918029 PMCID: PMC5527157 DOI: 10.1016/j.omtn.2017.07.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 01/22/2023]
Abstract
Cardiac fibrosis is pathological damage associated with nearly all forms of heart disease. AMP-activated protein kinase (AMPK) is an evolutionary conserved energy-sensing enzyme. Emerging evidences indicate that AMPK plays an important role in cardiac fibrosis and cell proliferation. However, less is known about the detailed mechanism of AMPK activation on cardiac fibrosis. In this study, we found the AMPK activation improved the impaired cardiac function of cardiac fibrosis rats and decreased interstitial fibrosis. Further results indicated AMPK activation promoted p21 and p27 and inhibited CDK2 and cyclin E protein expressions both in vivo and in vitro. Moreover, AMPK activation repressed downstream transcription factor hepatocyte nuclear factor 4 alpha (HNF-4α) expression and decreased the binding of HNF-4α to TGF-β1 promoters, which eventually resulted in TGF-β1 downregulation and miR-29 family upregulation. Furthermore, miR-29, in turn, inhibited the progression of cardiac fibrosis through suppressing its target CDK2. Taken together, activation of AMPK, on the one hand, upregulated p21 and p27 expression, further inhibited CDK2 and cyclin E complex, and finally suppressed the progression of cardiac fibrosis, and, on the other hand, repressed HNF-4α expression, further downregulated the activity of TGF-β1 promoter, promoted miR-29 expression, and finally prevented the development of cardiac fibrosis.
Collapse
Affiliation(s)
- Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yan Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Shuzhi Li
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Yunping Chen
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Lei Li
- Department of Orthopedics, Fifth Clinical College of Harbin Medical University, Daqing, Heilongjiang 163316, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Mingyao E
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Chao Song
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China
| | - Baiyan Li
- Department of Pharmacology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, Heilongjiang 163319, China.
| |
Collapse
|
43
|
Jin W, Deng QQ, Chen BY, Lu ZX, Li Q, Zhao HK, Chang P, Yu J, Pei ZH. Inhibitory effects of low decibel infrasound on the cardiac fibroblasts and the involved mechanism. Noise Health 2017; 19:149-153. [PMID: 28615545 PMCID: PMC5501025 DOI: 10.4103/nah.nah_14_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION Infrasound is a mechanical vibration wave with frequency between 0.0001 and 20 Hz. It has been established that infrasound of 120 dB or stronger is dangerous to humans. However, the biological effects of low decibel infrasound are largely unknown. The purpose of this study was to investigate the effects of low decibel infrasound on the cardiac fibroblasts. MATERIALS AND METHODS The cardiac fibroblasts were isolated and cultured from Sprague-Dawley rats. The cultured cells were assigned into the following four groups: control group, angiotensin II (Ang II) group, infrasound group, and Ang II+infrasound group. The cell proliferation and collagen synthesis rates were evaluated by means of [3H]-thymidine and [3H]-proline incorporation, respectively. The levels of TGF-β were determined by enzyme-linked immunosorbent assay. Moreover, RNAi approaches were used for the analysis of the biological functions of miR-29a, and the phosphorylation status of Smad3 was detected using western blotting analysis. RESULTS The results showed that low decibel infrasound significantly alleviated Ang II-induced enhancement of cell proliferation and collagen synthesis. DISCUSSION Compared with the control, Ang II markedly decreased the expression of miR-29a levels and increased the secretion of TGF-β and phosphorylation of Smad3, which was partly reversed by the treatment with low decibel infrasound. Importantly, knockdown of miR-29a diminished the effects of infrasound on the cardiac fibroblasts. In conclusion, low decibel infrasound inhibits Ang II-stimulated cardiac fibroblasts via miR-29a targeting TGF-β/Smad3 signaling.
Collapse
Affiliation(s)
- Wei Jin
- Department of Cardiology, Third Hospital of Nanchang, Nanchang, China
| | - Qin-Qin Deng
- Department of Cardiology, Third Hospital of Nanchang, Nanchang, China
| | - Bao-Ying Chen
- Department of Radiology, Second Affiliated Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhen-Xing Lu
- Department of Cardiology, Third Hospital of Nanchang, Nanchang, China
| | - Qing Li
- Department of Cardiology, Third Hospital of Nanchang, Nanchang, China
| | - Hai-Kang Zhao
- Department of Neurosurgery, Second Affiliated Hospital, Xi'an Medical University, Xi'an, China
| | - Pan Chang
- Central Laboratory, Second Affiliated Hospital, Xi'an Medical University, Xi'an, China
| | - Jun Yu
- Central Laboratory, Second Affiliated Hospital, Xi'an Medical University, Xi'an, China
| | - Zhao-Hui Pei
- Department of Cardiology, Third Hospital of Nanchang, Nanchang, China
| |
Collapse
|
44
|
Fan X, Xie J, Tian J. Reducing Cardiac Fibrosis: Na/K-ATPase Signaling Complex as a Novel Target. ACTA ACUST UNITED AC 2017; 6. [PMID: 29034264 DOI: 10.4172/2329-6607.1000204] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiac fibrosis is a common pathological process in cardiac disease and may lead to heart failure. It can also cause sudden death even in those without cardiac symptoms. Tissue fibrosis can be categorized into two categories: replacement fibrosis (also called reparative fibrosis) and reactive fibrosis. In replacement fibrosis, infiltration of inflammatory cells and accumulation of Extracellular Matrix (ECM) proteins are the initial steps in forming scarlike fibrotic tissue after acute cardiac injury and cardiac cell necrosis. Reactive fibrosis can be formed in response to hormonal change and pressure or volume overload. Experimental studies in animals have identified important pathways such as the Renin-Angiotensin-Aldosterone System (RAAS) and the endothelin pathway that contribute to fibrosis formation. Despite the fact that clinical trials using RAAS inhibitors as therapies for reducing cardiac fibrosis and improving cardiac function have been promising, heart failure is still the leading cause of deaths in the United States. Intensive efforts have been made to find novel targets and to develop new treatments for cardiac fibrosis and heart failure in the past few decades. The Na/K-ATPase, a canonical ion transporter, has been shown to also function as a signal transducer and prolonged activation of Na/K-ATPase signaling has been found to promote the formation of cardiac fibrosis. Novel tools that block the activation of Na/K-ATPase signaling have been developed and have shown promise in reducing cardiac fibrosis. This review will discuss the recent development of novel molecular targets, focusing on the Na/K-ATPase signaling complex as a therapeutic target in treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- X Fan
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Xie
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| | - J Tian
- Department of Medicine, Center for Hypertension and Personalized Medicine, University of Toledo, Ohio 43614, USA
| |
Collapse
|
45
|
Wang F, Li Y, Yang Z, Lu K, Zuo J, Zhou Z. Effect of Low-Intensity Pulsed Ultrasound on a Rat Model of Dentin-Dental Pulp Injury and Repair. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:163-175. [PMID: 27814934 DOI: 10.1016/j.ultrasmedbio.2016.08.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 08/17/2016] [Accepted: 08/18/2016] [Indexed: 06/06/2023]
Abstract
This study investigated histopathologic changes in dental pulp after treatment with low-intensity pulsed ultrasound (LIPUS). Fifty rats were randomly divided into an experimental group (n = 25) and a blank control group (n = 25). In the experimental group, a cavity was prepared in the bilateral maxillary first molars. The upper right first molars were stimulated with LIPUS (30 mW/cm2, 1.5 MHz) for 20 min/d. The cavities prepared in the left teeth were used as experimental controls (i.e., no LIPUS). Five rats in each group were sacrificed at days 1, 3, 5, 7 and 14. Inflammatory response was visible at different time points after cavity preparation, peaking at day 3, after which it gradually weakened. More reparative dentin was found on the LIPUS treatment side. transforming growth factor-β1 expression increased after treatment, peaking at day 5 and returning to normal at day 14 on both sides, but was stronger with LIPUS treatment. SMAD2 and SMAD3 expressions in the dental pulp gradually increased after cavity preparation, especially in the experimental group. LIPUS promoted the repair of dentin-pulp complex injury, to a certain extent and should be investigated further as a potential therapy.
Collapse
Affiliation(s)
- Fei Wang
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Yueheng Li
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Zhengyan Yang
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Keke Lu
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Jing Zuo
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China
| | - Zhi Zhou
- The College of Stomatology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory for Oral Diseases and Biomedical Science, Chongqing, China; and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, People's Republic of China.
| |
Collapse
|
46
|
Zhu Y, Li Z, Wang Y, Li L, Wang D, Zhang W, Liu L, Jiang H, Yang J, Cheng J. Overexpression of miR-29b reduces collagen biosynthesis by inhibiting heat shock protein 47 during skin wound healing. Transl Res 2016; 178:38-53.e6. [PMID: 27477081 DOI: 10.1016/j.trsl.2016.07.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 07/04/2016] [Accepted: 07/06/2016] [Indexed: 12/26/2022]
Abstract
Skin scar formation is characterized by excessive synthesis and aberrant deposition of collagens during wound healing. MicroRNAs are endogenous gene regulators critically involved in diverse biological events including skin scar formation and hold considerable promise as therapeutic targets. However, the detailed molecular mechanisms responsible for collagen production during skin wound repair and scar formation remain incompletely known. Here our data revealed that significant downregulation of miR-29b and upregulation of heat shock protein 47 (HSP47) were observed during wound healing in both excisional and burn wound models and also detected in facial skin scar as compared to adjacent healthy skin. HSP47, a specific chaperon for collagen production and secretion, was identified as a novel and direct post-transcriptional target of miR-29b in skin fibroblasts via bioinformatics prediction and experimental validation. Moreover, the regulatory functions of miR-29b in collagen biosynthesis are partially achieved through modulating HSP47 expression in skin fibroblasts. Furthermore, the profibrotic growth factor TGF-β1 inhibited miR-29b transcription by activating TGF-β/Smads signaling and in turn depressed HSP47 and enhanced collagen 1 production. In contrast, the proinflammatory cytokines IL-1β and TNF-α significantly induced miR-29b transcription via activating NF-κB signaling but had no significant effect on HSP47 and collagen production in skin fibroblasts. Importantly, local delivery of miR-29b lentiviral particles inhibited HSP47 expression and collagen biosynthesis as well as suppressed angiogenesis, thus reducing scar formation in an excisional wound splinting model. Collectively, our data reveal that miR-29b can reduce collagen biosynthesis during skin wound healing likely via post-transcriptional inhibition of HSP47 expression. These findings also suggest that therapeutic targeting of miR-29b/HSP47 might be a viable alternative strategy to prevent or reduce scar formation.
Collapse
Affiliation(s)
- Yumin Zhu
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China PRC
| | - Zhongwu Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Nanjing Medical University, Nanjing, China PRC
| | - Yanling Wang
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China PRC
| | - Lin Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Tongji University, Shanghai, China PRC
| | - Dongmiao Wang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Nanjing Medical University, Nanjing, China PRC
| | - Wei Zhang
- Department of Oral Pathology School of Stomatology, Nanjing Medical University, Nanjing, China PRC
| | - Laikui Liu
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China PRC; Department of Oral Pathology School of Stomatology, Nanjing Medical University, Nanjing, China PRC
| | - Hongbing Jiang
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China PRC; Department of Oral and Maxillofacial Surgery, School of Stomatology, Nanjing Medical University, Nanjing, China PRC
| | - Jianrong Yang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Nanjing Medical University, Nanjing, China PRC.
| | - Jie Cheng
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China PRC; Department of Oral and Maxillofacial Surgery, School of Stomatology, Nanjing Medical University, Nanjing, China PRC.
| |
Collapse
|
47
|
Drummond CA, Crotty Alexander LE, Haller ST, Fan X, Xie JX, Kennedy DJ, Liu J, Yan Y, Hernandez DA, Mathew DP, Cooper CJ, Shapiro JI, Tian J. Cigarette smoking causes epigenetic changes associated with cardiorenal fibrosis. Physiol Genomics 2016; 48:950-960. [PMID: 27789733 DOI: 10.1152/physiolgenomics.00070.2016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 10/22/2016] [Indexed: 12/14/2022] Open
Abstract
Clinical studies indicate that smoking combustible cigarettes promotes progression of renal and cardiac injury, leading to functional decline in the setting of chronic kidney disease (CKD). However, basic studies using in vivo small animal models that mimic clinical pathology of CKD are lacking. To address this issue, we evaluated renal and cardiac injury progression and functional changes induced by 4 wk of daily combustible cigarette smoke exposure in the 5/6th partial nephrectomy (PNx) CKD model. Molecular evaluations revealed that cigarette smoke significantly (P < 0.05) decreased renal and cardiac expression of the antifibrotic microRNA miR-29b-3 and increased expression of molecular fibrosis markers. In terms of cardiac and renal organ structure and function, exposure to cigarette smoke led to significantly increased systolic blood pressure, cardiac hypertrophy, cardiac and renal fibrosis, and decreased renal function. These data indicate that decreased expression of miR-29b-3p is a novel mechanism wherein cigarette smoke promotes accelerated cardiac and renal tissue injury in CKD. (155 words).
Collapse
Affiliation(s)
- Christopher A Drummond
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio;
| | - Laura E Crotty Alexander
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System and Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego Health Sciences, San Diego, California; and
| | - Steven T Haller
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| | - Xiaoming Fan
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| | - Jeffrey X Xie
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| | - David J Kennedy
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| | - Jiang Liu
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Yanling Yan
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Dawn-Alita Hernandez
- Division of Pulmonary Medicine (Critical Care and Sleep Medicine), University of Toledo, Toledo, Ohio
| | - Denzil P Mathew
- Pulmonary Critical Care Section, Veterans Affairs San Diego Healthcare System and Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego Health Sciences, San Diego, California; and
| | - Christopher J Cooper
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia
| | - Jiang Tian
- College of Medicine and Life Sciences, Department of Medicine, Division of Cardiovascular Medicine and Center for Hypertension and Personalized Medicine, University of Toledo, Toledo, Ohio
| |
Collapse
|
48
|
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
|
49
|
Yang C, Zheng SD, Wu HJ, Chen SJ. Regulatory Mechanisms of the Molecular Pathways in Fibrosis Induced by MicroRNAs. Chin Med J (Engl) 2016; 129:2365-72. [PMID: 27647197 PMCID: PMC5040024 DOI: 10.4103/0366-6999.190677] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE MicroRNAs (miRNAs or miRs) play critical roles in the fibrotic process in different organs. We summarized the latest research progress on the roles and mechanisms of miRNAs in the regulation of the molecular signaling pathways involved in fibrosis. DATA SOURCES Papers published in English from January 2010 to August 2015 were selected from the PubMed and Web of Science databases using the search terms "microRNA", "miR", "transforming growth factor β", "tgf β", "mitogen-activated protein kinase", "mapk", "integrin", "p38", "c-Jun NH2-terminal kinase", "jnk", "extracellular signal-regulated kinase", "erk", and "fibrosis". STUDY SELECTION Articles were obtained and reviewed to analyze the regulatory effects of miRNAs on molecular signaling pathways involved in the fibrosis. RESULTS Recent evidence has shown that miRNAs are involved in regulating fibrosis by targeting different substrates in the molecular processes that drive fibrosis, such as immune cell sensitization, effector cell activation, and extracellular matrix remodeling. Moreover, several important molecular signaling pathways involve in fibrosis, such as the transforming growth factor-beta (TGF-β) pathway, mitogen-activated protein kinase (MAPK) pathways, and the integrin pathway are regulated by miRNAs. Third, regulation of the fibrotic pathways induced by miRNAs is found in many other tissues in addition to the heart, lung, liver, and kidney. Interestingly, the actions of many drugs on the human body are also induced by miRNAs. It is encouraging that the fibrotic process can be blocked or reversed by targeting specific miRNAs and their signaling pathways, thereby protecting the structures and functions of different organs. CONCLUSIONS miRNAs not only regulate molecular signaling pathways in fibrosis but also serve as potential targets of novel therapeutic interventions for fibrosing diseases.
Collapse
Affiliation(s)
- Cui Yang
- Department of Cardiology, Huairou Hospital of Traditional Chinese Medicine, Beijing 101400, China
| | - Si-Dao Zheng
- Department of Cardiology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing 100039, China
| | - Hong-Jin Wu
- Department of Cardiology, Beijing Haidian Hospital, Haidian Section of Peking University Third Hospital, Beijing 100191, China
| | - Shao-Jun Chen
- Department of Cardiology, Huairou Hospital of Traditional Chinese Medicine, Beijing 101400, China
| |
Collapse
|
50
|
Hahn JM, McFarland KL, Combs KA, Supp DM. Partial epithelial-mesenchymal transition in keloid scars: regulation of keloid keratinocyte gene expression by transforming growth factor-β1. BURNS & TRAUMA 2016; 4:30. [PMID: 27574697 PMCID: PMC4994224 DOI: 10.1186/s41038-016-0055-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/04/2016] [Indexed: 12/17/2022]
Abstract
Background Keloids are an extreme form of abnormal scarring that result from a pathological fibroproliferative wound healing process. The molecular mechanisms driving keloid pathology remain incompletely understood, hindering development of targeted, effective therapies. Recent studies in our laboratory demonstrated that keloid keratinocytes exhibit adhesion abnormalities and display a transcriptional signature reminiscent of cells undergoing epithelial-mesenchymal transition (EMT), suggesting a role for EMT in keloid pathology. In the current study, we further define the EMT-like phenotype of keloid scars and investigate regulation of EMT-related genes in keloid. Methods Primary keratinocytes from keloid scar and normal skin were cultured in the presence or absence of transforming growth factor beta 1 (TGF-β1) +/− inhibitors of TGF-β1 and downstream signaling pathways. Gene expression was measured using quantitative polymerase chain reaction. Migration was analyzed using an in vitro wound healing assay. Proteins in keloid scar and normal skin sections were localized by immunohistochemistry. Statistical analyses utilized SigmaPlot (SyStat Software, San Jose, CA) or SAS® (SAS Institute, Cary, NC). Results In keloid and normal keratinocytes, TGF-β1 regulated expression of EMT-related genes, including hyaluronan synthase 2, vimentin, cadherin-11, wingless-type MMTV integration site family, member 5A, frizzled 7, ADAM metallopeptidase domain 19, and interleukin-6. Inhibition of canonical TGF-β1 signaling in keloid keratinocytes significantly inhibited expression of these genes, and TGF-β1 stimulation of normal keratinocytes increased their expression. The inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway or the p38 mitogen-activated protein kinase pathway attenuated TGF-β1-induced expression of subsets of these genes. Migration of keloid keratinocytes, previously shown to be increased compared with normal keratinocytes, was significantly reduced by inhibition of TGF-β1 or ERK1/2 signaling. Biomarkers of EMT, including reduced E-cadherin and increased active β-catenin, were observed in keloid epidermis in vivo. However, evidence of basement membrane breakdown in keloid scar was not observed. Conclusions The results suggest that keloid keratinocytes exist in an EMT-like metastable state, similar to activated keratinocytes in healing wounds. The EMT-like gene expression pattern of keloid keratinocytes is regulated by canonical and non-canonical TGF-β1 signaling pathways. Therefore, interventions targeting TGF-β1-regulated EMT-like gene expression in keloid keratinocytes may serve to suppress keloid scarring. Electronic supplementary material The online version of this article (doi:10.1186/s41038-016-0055-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jennifer M Hahn
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kevin L McFarland
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Kelly A Combs
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA
| | - Dorothy M Supp
- Research Department, Shriners Hospitals for Children - Cincinnati, Cincinnati, OH USA ; Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH USA
| |
Collapse
|