1
|
Watts KM, Nichols W, Richardson WJ. Computational screen for sex-specific drug effects in a cardiac fibroblast signaling network model. Sci Rep 2023; 13:17068. [PMID: 37816826 PMCID: PMC10564891 DOI: 10.1038/s41598-023-44440-9] [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: 07/25/2023] [Accepted: 10/08/2023] [Indexed: 10/12/2023] Open
Abstract
Heart disease is the leading cause of death in both men and women. Cardiac fibrosis is the uncontrolled accumulation of extracellular matrix proteins, which can exacerbate the progression of heart failure, and there are currently no drugs approved specifically to target matrix accumulation in the heart. Computational signaling network models (SNMs) can be used to facilitate discovery of novel drug targets. However, the vast majority of SNMs are not sex-specific and/or are developed and validated using data skewed towards male in vitro and in vivo samples. Biological sex is an important consideration in cardiovascular health and drug development. In this study, we integrate a cardiac fibroblast SNM with estrogen signaling pathways to create sex-specific SNMs. The sex-specific SNMs demonstrated high validation accuracy compared to in vitro experimental studies in the literature while also elucidating how estrogen signaling can modulate the effect of fibrotic cytokines via multi-pathway interactions. Further, perturbation analysis and drug screening uncovered several drug compounds predicted to generate divergent fibrotic responses in male vs. female conditions, which warrant further study in the pursuit of sex-specific treatment recommendations for cardiac fibrosis. Future model development and validation will require more generation of sex-specific data to further enhance modeling capabilities for clinically relevant sex-specific predictions of cardiac fibrosis and treatment.
Collapse
Affiliation(s)
- Kelsey M Watts
- Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA.
| | - Wesley Nichols
- Department of Bioengineering, Clemson University, Clemson, SC, 29634, USA
| | - William J Richardson
- Department of Chemical Engineering, University of Arkansas, Fayetteville, AR, 72701, USA
| |
Collapse
|
2
|
Watts KM, Nichols W, Richardson WJ. Computational Screen for Sex-Specific Drug Effects in a Cardiac Fibroblast Network Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536523. [PMID: 37090681 PMCID: PMC10120687 DOI: 10.1101/2023.04.11.536523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Heart disease is the leading cause of death in both men and women. Cardiac fibrosis is the uncontrolled accumulation of extracellular matrix proteins which can exacerbate the progression of heart failure, and there are currently no drugs approved specifically to target matrix accumulation in the heart. Computational signaling network models (SNMs) can be used to facilitate discovery of novel drug targets. However, the vast majority of SNMs are not sex-specific and/or are developed and validated using data skewed towards male in vitro and in vivo samples. Biological sex is an important consideration in cardiovascular health and drug development. In this study, we integrate a previously constructed cardiac fibroblast SNM with estrogen signaling pathways to create sex-specific SNMs. The sex-specific SNMs maintained previously high validation when compared to in vitro experimental studies in the literature. A sex-specific perturbation analysis and drug screen uncovered several potential pathways that warrant further study in the pursuit of sex-specific treatment recommendations for cardiac fibrosis.
Collapse
Affiliation(s)
- Kelsey M Watts
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | - Wesley Nichols
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | - William J Richardson
- Department of Chemical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
3
|
Miao S, Wang L, Guan S, Gu T, Wang H, Shangguan W, Wang W, Liu Y, Liang X. Integrated whole transcriptome analysis for the crucial regulators and functional pathways related to cardiac fibrosis in rats. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:5413-5429. [PMID: 36896551 DOI: 10.3934/mbe.2023250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
BACKGROUND Cardiac fibrosis has gradually gained significance in the field of cardiovascular disease; however, its specific pathogenesis remains unclear. This study aims to establish the regulatory networks based on whole-transcriptome RNA sequencing analyses and reveal the underlying mechanisms of cardiac fibrosis. METHODS An experimental model of myocardial fibrosis was induced using the chronic intermittent hypoxia (CIH) method. Expression profiles of long non-coding RNA (lncRNA), microRNA (miRNA), and messenger RNA (mRNA) were acquired from right atrial tissue samples of rats. Differentially expressed RNAs (DERs) were identified, and functional enrichment analysis was performed. Moreover, a protein-protein interaction (PPI) network and competitive endogenous RNA (ceRNA) regulatory network that are related to cardiac fibrosis were constructed, and the relevant regulatory factors and functional pathways were identified. Finally, the crucial regulators were validated using qRT-PCR. RESULTS DERs, including 268 lncRNAs, 20 miRNAs, and 436 mRNAs, were screened. Further, 18 relevant biological processes, such as "chromosome segregation, " and 6 KEGG signaling pathways, such as "cell cycle, " were significantly enriched. The regulatory relationship of miRNA-mRNA-KEGG pathways showed eight overlapping disease pathways, including "pathways in cancer." In addition, crucial regulatory factors, such as Arnt2, WNT2B, GNG7, LOC100909750, Cyp1a1, E2F1, BIRC5, and LPAR4, were identified and verified to be closely related to cardiac fibrosis. CONCLUSION This study identified the crucial regulators and related functional pathways in cardiac fibrosis by integrating the whole transcriptome analysis in rats, which might provide novel insights into the pathogenesis of cardiac fibrosis.
Collapse
Affiliation(s)
- Shuai Miao
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Lijun Wang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Siyu Guan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Tianshu Gu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Hualing Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wenfeng Shangguan
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Weiding Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yu Liu
- Taikang Ningbo Hospital, Ningbo 315100, Zhejiang, China
| | - Xue Liang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular disease, Department of Cardiology, Tianjin Institute of Cardiology, the Second Hospital of Tianjin Medical University, Tianjin 300211, China
| |
Collapse
|
4
|
Omar IS, Mat Adenan NA, Godoy A, Teo IH, Gunasagran Y, Chung I. Aberrant upregulation of CDK1 contributes to medroxyprogesterone acetate (MPA) resistance in cancer-associated fibroblasts of the endometrium. Biochem Biophys Res Commun 2022; 628:133-140. [PMID: 36084551 DOI: 10.1016/j.bbrc.2022.08.088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 08/26/2022] [Accepted: 08/27/2022] [Indexed: 01/04/2023]
Abstract
The response to medroxyprogesterone acetate (MPA) decreases as endometrial disease progresses from the benign to malignancy. In a mouse model, progesterone receptor (PR) expression in normal fibroblasts is accountable for the MPA's inhibitory effects in cancer cells. However, it is still unclear, if and how, fibroblasts from human tumors respond to MPA. In this study, three benign-associated fibroblasts (BAFs) and four cancer-associated fibroblasts (CAFs) were isolated from human benign and cancerous endometrial tissues, respectively, to examine MPA activation on PR signaling. PR-B protein expression were heterogeneously expressed in both CAFs and BAFs, despite a lower mRNA expression in the former. In a luciferase reporter assay, MPA treatment stimulated some PR DNA-binding activity in BAFs but not in CAFs. Yet, activation of PR target gene was generally more pronounced in MPA-treated CAFs compared to BAFs. Cyclin-dependent kinase 1 (CDK1) was exclusively upregulated by 10 nM MPA in CAFs (5.1-fold vs. 1.1-fold in BAFs, P < 0.05), leading to a higher CDK1 protein expression. Subsequently in a dose-response study, CAFs showed an average of ∼20% higher cell viability when compared to BAFs, indicative of drug resistance to MPA. MPA resistance was also observed in EC-CAFs co-culture, when MPA-treated cells showed greater tumor spheroid formation than in EC-BAFs co-culture (2-fold, P < 0.01). The increased cell viability observed in CAFs was reversed with mifepristone (RU486), a PR antagonist which suppressed MPA-induced CDK1 expression. This indicates that MPA-induced abnormal upregulation of CDK1 may contribute to the enhanced CAFs cell proliferation, suggesting a new mechanism of MPA resistance within endometrial cancer microenvironment.
Collapse
Affiliation(s)
- Intan Sofia Omar
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Universiti Malaya Cancer Research Institute, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Noor Azmi Mat Adenan
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Department of Obstetrics and Gynaecology, Ara Damansara and Subang Jaya Medical Center, Ramsay Sime Darby Health Care, 47500, Subang Jaya, Selangor, Malaysia
| | - Alejandro Godoy
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile; Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, United States
| | - Ik Hui Teo
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yogeeta Gunasagran
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ivy Chung
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Universiti Malaya Cancer Research Institute, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
5
|
MacLean J, Pasumarthi KBS. Characterization of primary adult mouse cardiac fibroblast cultures. Can J Physiol Pharmacol 2020; 98:861-869. [PMID: 32721222 DOI: 10.1139/cjpp-2020-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The role of cardiac fibroblasts (CFs) in disease states has been a focus of cardiovascular research over the past decade. Here, we briefly describe methods for isolation and characterization of CFs from adult mouse ventricles. Primary cultures were stained using antibodies for several marker proteins such as α-smooth muscle actin (αSMA), vimentin, and discoidin domain receptor 2 (DDR2) to confirm the identity of CFs or cardiac myofibroblasts (CMFs). Most cells in primary cultures consisted of CFs, with very low frequencies of endothelial cells, cardiomyocytes, and smooth muscle cells. We compared marker expression between cultures that were not passaged (P0) or passaged for few times (P1-3). When compared with P1-3 cultures, P0 cultures consistently displayed a lower percentage of cells positive for αSMA and DDR2, whereas vimentin expression was significantly higher in P0 cultures compared with P1-3 cultures. P0 cells were also smaller in area than P1-3 cells. Further, P1-3 mouse CFs were found to express both β1 and β2 adrenergic receptors (ARs) and β1ARs were more readily detected on the cell surface compared with β2ARs. In summary, mouse CF cultures underwent phenotype conversion into CMFs after passaging, consistent with what is seen with CF cultures from other species.
Collapse
Affiliation(s)
- Jessica MacLean
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Kishore B S Pasumarthi
- Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
6
|
Li Y, Tan W, Ye F, Xue F, Gao S, Huang W, Wang Z. Identification of microRNAs and genes as biomarkers of atrial fibrillation using a bioinformatics approach. J Int Med Res 2019; 47:3580-3589. [PMID: 31218935 PMCID: PMC6726789 DOI: 10.1177/0300060519852235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Objective We aimed to explore potential microRNAs (miRNAs) and target genes related to atrial fibrillation (AF). Methods Data for microarrays GSE70887 and GSE68475, both of which include AF and control groups, were downloaded from the Gene Expression Omnibus database. Differentially expressed miRNAs between AF and control groups were identified within each microarray, and the intersection of these two sets was obtained. These miRNAs were mapped to target genes in the miRNet database. Functional annotation and enrichment analysis of these target genes was performed in the DAVID database. The protein-protein interaction (PPI) network from the STRING database and the miRNA-target-gene network were merged into a PPI-miRNA network using Cytoscape software. Modules of this network containing miRNAs were detected and further analyzed. Results Ten differentially expressed miRNAs and 1520 target genes were identified. Three PPI-miRNA modules were constructed, which contained miR-424, miR-15a, miR-542-3p, and miR-421 as well as their target genes, CDK1, CDK6, and CCND3. Conclusion The identified miRNAs and genes may be related to the pathogenesis of AF. Thus, they may be potential biomarkers for diagnosis and targets for treatment of AF.
Collapse
Affiliation(s)
- Yingyuan Li
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Wulin Tan
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Fang Ye
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Faling Xue
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Shaowei Gao
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Wenqi Huang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Zhongxing Wang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| |
Collapse
|
7
|
Su Y, Li Q, Zheng Z, Wei X, Hou P. Integrative bioinformatics analysis of miRNA and mRNA expression profiles and identification of associated miRNA-mRNA network in aortic dissection. Medicine (Baltimore) 2019; 98:e16013. [PMID: 31192949 PMCID: PMC6587623 DOI: 10.1097/md.0000000000016013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Aortic dissection (AD) is one of the most lethal cardiovascular diseases. The aim of this study was to identify core genes and pathways revealing pathogenesis in AD. METHODS We screened differentially expressed mRNAs and miRNAs using mRNA and miRNA expression profile data of AD from Gene Expression Omnibus. Then functional and pathway enrichment analyses of differential expression genes (DEGs) was performed utilizing the database for annotation, visualization, and integrated discovery (DAVID). Target genes with differential expression miRNAs (DEMIs) were predicted using the miRWalk database, and the intersection between these predictions and DEGs was selected as differentially expressed miRNA-target genes. In addition, a protein-protein interaction (PPI) network and miRNA-mRNA regulatory network were constructed. RESULTS In total, 130 DEGs and 47 DEMIs were identified from mRNA and miRNA microarray, respectively, and 45 DEGs were DEMI-target genes. The PPI and miRNA-mRNA network included 79 node genes and 74 node genes, respectively, while 23 hub genes and 2 hub miRNAs were identified. The DEGs, PPI and modules differential expression miRNA-target genes were all mainly enriched in cell cycle, cell proliferation and cell apoptosis signaling pathways. CONCLUSION Taken above, the study reveals some candidate genes and pathways potentially involving molecular mechanisms of AD. These findings provide a new insight for research and treatment of AD.
Collapse
|
8
|
Zhuang Y, Li T, Zhuang Y, Li Z, Yang W, Huang Q, Li D, Wu H, Zhang G, Yang T, Zhan L, Pan Z, Lu Y. Involvement of lncR-30245 in Myocardial Infarction-Induced Cardiac Fibrosis Through Peroxisome Proliferator-Activated Receptor-γ-Mediated Connective Tissue Growth Factor Signalling Pathway. Can J Cardiol 2019; 35:480-489. [PMID: 30935639 DOI: 10.1016/j.cjca.2019.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are emerging as important mediators of cardiac pathophysiology. The aim of the present study is to investigate the effects of lncR-30245, an lncRNA, on cardiac fibrogenesis and the underlying mechanism. METHODS Myocardial infarction (MI) and transforming growth factor (TGF)-β1 were used to induce fibrotic phenotypes. Cardiac fibrosis was detected by Masson's trichrome staining. Cardiac function was evaluated by echocardiography. Western blot, quantitative reverse transcription-polymerase chain reaction, and pharmacological approaches were used to investigate the role of lncR-30245 in cardiac fibrogenesis. RESULTS Expression of lncR-30245 was significantly increased in MI hearts and TGF-β1-treated cardiac fibroblasts (CFs). LncR-30245 was mainly located in the cytoplasm. Overexpression of lncR-30245 promoted collagen production and CF proliferation. Knockdown of lncR-30245 significantly inhibited TGF-β1-induced collagen production and CF proliferation. LncR-30245 overexpression inhibited the antifibrotic role of peroxisome proliferator-activated receptor (PPAR)-γ and increased connective tissue growth factor (CTGF) expression, whereas lncR-30245 knockdown exerted the opposite effects. Rosiglitazone, a PPAR-γ agonist, significantly inhibited lncR-30245-induced CTGF upregulation and collagen production in CFs. In contrast, T0070907, a PPAR-γ antagonist, attenuated the inhibitory effects of lncR-30245 small interfering RNA (siRNA) on TGF-β1-induced CTGF expression and collagen production. LncR-30245 knockdown significantly enhanced ejection fraction and fractional shortening and attenuated cardiac fibrosis in MI mice. CONCLUSION Our study indicates that the lncR-30245/PPAR-γ/CTGF pathway mediates MI-induced cardiac fibrosis and might be a therapeutic target for various cardiac diseases associated with fibrosis.
Collapse
Affiliation(s)
- Yuting Zhuang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Tingting Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Yanan Zhuang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Zhuoyun Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Wanqi Yang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Qihe Huang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Danyang Li
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Hao Wu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Guiye Zhang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Ti Yang
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Linfeng Zhan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China
| | - Zhenwei Pan
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China.
| | - Yanjie Lu
- Department of Pharmacology, State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang, P. R. China; Northern Translational Medicine Research and Cooperation Center, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, P. R. China.
| |
Collapse
|
9
|
Feridooni T, Pasumarthi KBS. Fractionation of embryonic cardiac progenitor cells and evaluation of their differentiation potential. Differentiation 2018; 105:1-13. [PMID: 30530197 DOI: 10.1016/j.diff.2018.11.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/24/2018] [Accepted: 11/22/2018] [Indexed: 12/19/2022]
Abstract
Mid-gestation mouse ventricles (E11.5) contain a larger number of Nkx2.5+ cardiac progenitor cells (CPCs). The proliferation rates are consistently higher in CPCs compared to myocyte population of developing ventricles. Recent studies suggested that CPCs are an ideal donor cell type for replacing damaged tissue in diseased hearts. Thus, the ability to isolate and expand CPCs from embryos or stem cell cultures could be useful for cell fate studies and regenerative therapies. Since embryonic CPCs possess fewer mitochondria compared to cardiomyocytes, we reasoned that CPCs can be fractionated using a fluorescent mitochondrial membrane potential dye (TMRM) and these cells may retain cardiomyogenic potential even in the absence of cardiomyocytes (CMs). FACS sorting of TMRM stained embryonic ventricular cells indicated that over 99% of cells in TMRM high fraction stained positive for sarcomeric myosin (MF20) and all of them expressed Nkx2.5. Although majority of cells present in TMRM low fraction expressed Nkx2.5, very few cells (~1%) stained positive for MF20. Further culturing of TMRM low cells over a period of 48 h showed a progressive increase in MF20 positive cells. Additional analyses revealed that MF20 negative cells in TMRM low fraction do not express markers for endothelial cells (vWF, CD31) or smooth muscle cells (SM myosin). Treatment of TMRM low cells with known cardiogenic factors DMSO and dynorphin B significantly increased the percentage of MF20+ cells compared to untreated cultures. Collectively, these studies suggest that embryonic CPCs can be separated as a TMRM low fraction and their differentiation potential can be enhanced by exogenous addition of known cardiomyogenic factors.
Collapse
Affiliation(s)
- Tiam Feridooni
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, P.O. Box 15000, Halifax, Nova Scotia, Canada B3H 4R2
| | - Kishore B S Pasumarthi
- Department of Pharmacology, Dalhousie University, Sir Charles Tupper Building, 5850 College Street, P.O. Box 15000, Halifax, Nova Scotia, Canada B3H 4R2.
| |
Collapse
|
10
|
Zhuang Y, Li T, Zhuang Y, Li Z, Yang W, Huang Q, Li D, Wu H, Zhang G, Yang T, Zhan L, Pan Z, Lu Y. WITHDRAWN: Suppression of lncR-30245 alleviates myocardial infarction induced cardiac fibrosis via the PPAR-γ/CTGF pathway. Can J Cardiol 2018. [DOI: 10.1016/j.cjca.2018.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
11
|
Zhang Y, Luo G, Zhang Y, Zhang M, Zhou J, Gao W, Xuan X, Yang X, Yang D, Tian Z, Ni B, Tang J. Critical effects of long non-coding RNA on fibrosis diseases. Exp Mol Med 2018; 50:e428. [PMID: 29350677 PMCID: PMC5799794 DOI: 10.1038/emm.2017.223] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023] Open
Abstract
The expression or dysfunction of long non-coding RNAs (lncRNAs) is closely related to various hereditary diseases, autoimmune diseases, metabolic diseases and tumors. LncRNAs were also recently recognized as functional regulators of fibrosis, which is a secondary process in many of these diseases and a primary pathology in fibrosis diseases. We review the latest findings on lncRNAs in fibrosis diseases of the liver, myocardium, kidney, lung and peritoneum. We also discuss the potential of disease-related lncRNAs as therapeutic targets for the clinical treatment of human fibrosis diseases.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Dermatology, 105th Hospital of PLA, Hefei, China.,Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China.,Graduate School, Bengbu Medical College, Bengbu, China
| | - Gang Luo
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Yi Zhang
- Department of Clinical Laboratory, 150th Hospital of PLA, Luoyang, China
| | - Mengjie Zhang
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Jian Zhou
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Weiwu Gao
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Xiuyun Xuan
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, China
| | - Xia Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Di Yang
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Zhiqiang Tian
- Institute of Immunology, PLA, Third Military Medical University, Chongqing, China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing, China
| | - Jun Tang
- Department of Dermatology, 105th Hospital of PLA, Hefei, China.,Graduate School, Bengbu Medical College, Bengbu, China
| |
Collapse
|
12
|
Wang LX, Yang X, Yue Y, Fan T, Hou J, Chen GX, Liang MY, Wu ZK. Imatinib attenuates cardiac fibrosis by inhibiting platelet-derived growth factor receptors activation in isoproterenol induced model. PLoS One 2017; 12:e0178619. [PMID: 28570599 PMCID: PMC5453565 DOI: 10.1371/journal.pone.0178619] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/16/2017] [Indexed: 11/19/2022] Open
Abstract
Cardiac fibrosis is a significant global health problem with limited treatment choices. Although previous studies have shown that imatinib (IMA) inhibited cardiac fibrosis, the anti-fibrotic mechanisms have not been clearly uncovered. The aim of this study is to evaluate whether IMA attenuates cardiac fibrosis by inhibiting platelet-derived growth factor receptors (PDGFR) on isoproterenol (ISO)-induced mice. Adult male C57BL/6 mice were treated with vehicle or ISO ± IMA for one week. After echocardiography examination, the hearts of mice were used for histopathologic, RT-qPCR, and western blot analyses. We found that the ventricular wall thickness, cardiac hypertrophy, and apoptosis were enhanced following ISO treatment. IMA decreased the left ventricular wall thickness, prevented hypertrophy, and inhibited apoptosis induced by ISO. In addition, IMA attenuated the accumulation of collagens and α-smooth muscle actin (α-SMA) (the markers of fibrosis) caused by ISO treatment. Moreover, the expression of fibrosis related genes, and the phosphorylation of PDGFRs in ISO-treated mice hearts were inhibited by IMA as well. However, IMA did not change the expression of the matrix metalloproteinase-9 (MMP-9) in ISO-treated hearts. Furthermore, IMA reduced the expressions of collagens as well as α-SMA caused by activation of PDGFRα in cardiac fibroblasts. Taken together, our data demonstrate that IMA attenuated the cardiac fibrosis by blocking the phosphorylation of PDGFRs in the ISO-induced mice model. This study indicates that IMA could be a potentially therapeutic option for cardiac fibrosis in clinical application.
Collapse
Affiliation(s)
- Le-Xun Wang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiao Yang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuan Yue
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tian Fan
- Key Laboratory of South China Agricultural Plant Molecular Analysis and Genetic Improvement, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Provincial Key Laboratory of Applied Botany, South China Botanical Garden, Chinese Academy of Sciences, Guangzhou, China
| | - Jian Hou
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guang-Xian Chen
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meng-Ya Liang
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhong-Kai Wu
- Second Department of Cardiac Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Assisted Circulatory Laboratory of Health Ministry, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
13
|
ANO1 inhibits cardiac fibrosis after myocardial infraction via TGF-β/smad3 pathway. Sci Rep 2017; 7:2355. [PMID: 28539652 PMCID: PMC5443797 DOI: 10.1038/s41598-017-02585-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Abstract
As a newly identified factor in calcium-activated chloride channel, ANO1 participates in various physiological processes like proliferation and differentiation, and expresses in human cardiac fibroblasts. In this experiment, we investigated the function of ANO1 in cardiac fibrosis after myocardial infraction (MI) with methods of Western blotting, Quantitative real-time PCR (qRT-PCR), metabolic reduction of 3-(4,5-dimethylthiozol-2-yl)-2, 5-diphenyltetrazo-lium bromide (MTT), immunofluorescence and confocal imaging, and Masson’s trichrome staining. The results showed that the expression of ANO1 significantly increased in neonatal rats’ cardiac fibroblasts after hypoxia and in cardiac tissues after MI. After ANO1 over-expression, cardiac fibrosis was reduced in vitro and in vivo. Moreover, the expression of TGF-β and p-smad3 declined after ANO1over-expression in cardiac fiborblasts. In conclusion, ANO1 inhibits cardiac fibrosis after MI via TGF-β/smad3 pathway in rats.
Collapse
|
14
|
Verjans R, van Bilsen M, Schroen B. MiRNA Deregulation in Cardiac Aging and Associated Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:207-263. [PMID: 28838539 DOI: 10.1016/bs.ircmb.2017.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The prevalence of age-related diseases is increasing dramatically, among which cardiac disease represents the leading cause of death. Aging of the heart is characterized by various molecular and cellular hallmarks impairing both cardiomyocytes and noncardiomyocytes, and resulting in functional deteriorations of the cardiac system. The aging process includes desensitization of β-adrenergic receptor (βAR)-signaling and decreased calcium handling, altered growth signaling and cardiac hypertrophy, mitochondrial dysfunction and impaired autophagy, increased programmed cell death, low-grade inflammation of noncanonical inflammatory cells, and increased ECM deposition. MiRNAs play a fundamental role in regulating the processes underlying these detrimental changes in the cardiac system, indicating that MiRNAs are crucially involved in aging. Among others, MiR-34, MiR-146a, and members of the MiR-17-92 cluster, are deregulated during senescence and drive cardiac aging processes. It is therefore suggested that MiRNAs form possible therapeutic targets to stabilize the aged failing myocardium.
Collapse
Affiliation(s)
- Robin Verjans
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marc van Bilsen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Blanche Schroen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
15
|
Krishnan A, K D, Babu P.S S, Jagadeeshan S, Prasad M, Nair SA. Oncogenic Actions of SKP2 Involves Deregulation of CDK1 Turnover Mediated by FOXM1. J Cell Biochem 2016; 118:797-807. [DOI: 10.1002/jcb.25754] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 09/28/2016] [Indexed: 11/06/2022]
Affiliation(s)
- Anand Krishnan
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| | - Dhanya K
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| | - Saneesh Babu P.S
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| | - Sankar Jagadeeshan
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| | - Manu Prasad
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| | - S. Asha Nair
- Cancer Research Program; Rajiv Gandhi Centre for Biotechnology; Thiruvananthapuram India
| |
Collapse
|
16
|
Noncoding RNA as regulators of cardiac fibrosis: current insight and the road ahead. Pflugers Arch 2016; 468:1103-11. [PMID: 26786602 DOI: 10.1007/s00424-016-1792-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/11/2015] [Accepted: 01/07/2016] [Indexed: 12/19/2022]
Abstract
Cardiac fibrosis is an important pathological feature of cardiac remodeling in heart diseases. The molecular mechanisms of cardiac fibrosis are unknown. Genomic analyses estimated that many noncoding DNA regions generate noncoding RNAs (ncRNAs). ncRNAs have emerged as key molecular players in the regulation of gene expression in different biological processes. Recent studies have started to reveal the importance of ncRNAs in heart development and suggest also an involvement in cardiac fibrosis. These molecules are emerging as important regulators of cellular process. Here, we review particularly focuses on the involvement of two large families of ncRNAs, namely microRNAs (miRNAs) and long noncoding RNAs (LncRNAs) in the regulation of cardiac fibrosis. Furthermore, we review the functions and role of ncRNAs in cardiac biology and discuss these reports and the therapeutic potential of ncRNAs for cardiac fibrosis associated with fibroblast activation and proliferation.
Collapse
|
17
|
Tao H, Yang JJ, Shi KH. Non-coding RNAs as direct and indirect modulators of epigenetic mechanism regulation of cardiac fibrosis. Expert Opin Ther Targets 2015; 19:707-16. [PMID: 25652534 DOI: 10.1517/14728222.2014.1001740] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Cardiac fibroblast activation is a pivotal cellular event in cardiac fibrosis. Numerous studies have indicated that epigenetic modifications control cardiac fibroblast activation. Greater knowledge of the role of epigenetic modifications could improve understanding of the cardiac fibrosis pathogenesis. AREAS COVERED The aim of this review is to describe the present knowledge about the important role of non-coding RNA (ncRNA) transcripts in epigenetic gene regulation in cardiac fibrosis and looks ahead on new perspectives of epigenetic modification research. Furthermore, we will discuss examples of ncRNAs that interact with histone modification or DNA methylation to regulate gene expression. EXPERT OPINION MicroRNAs (miRNAs) and long ncRNAs (lncRNAs) modulate several important aspects of function. Recently, some studies continue to find novel pathways, including the important role of ncRNA transcripts in epigenetic gene regulation. Targeting the miRNAs and lncRNAs can be a promising direction in cardiac fibrosis treatment. We discuss new perspectives of ncRNAs that interact with histone modification or DNA methylation to regulate gene expression, others that are targets of these epigenetic mechanisms. The emerging recognition of the diverse functions of ncRNAs in regulating gene expression by epigenetic mechanisms suggests that they may represent new targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hui Tao
- The Second Hospital of Anhui Medical University, Department of Cardiothoracic Surgery , Fu Rong Road, Hefei 230601, Anhui Province , China +86 551 63869531 ; +86 551 63869531 ;
| | | | | |
Collapse
|
18
|
Gao Y, Chu M, Hong J, Shang J, Xu D. Hypoxia induces cardiac fibroblast proliferation and phenotypic switch: a role for caveolae and caveolin-1/PTEN mediated pathway. J Thorac Dis 2014; 6:1458-68. [PMID: 25364523 DOI: 10.3978/j.issn.2072-1439.2014.08.31] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/05/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cardiac fibrosis following myocardial infarction (MI) results in heart failure. Caveolin-1, the main structural protein of caveolae, regulates signal transduction pathways controlling cell proliferation and apoptosis. Meanwhile, low phosphatase and tensin homolog (PTEN) activity enhances the PI3K/Akt signal pathway to induce cell proliferation. But whether caveolin-1 and PTEN activation regulates cardiac fibroblast proliferation and contributes to cardiac fibrosis from ischemic injury is incompletely understood. This study investigates whether hypoxia inducing cardiac fibroblast proliferation and phenotypic switch is caveolin-dependent. METHODS We used in vitro and in vivo models of ischemic injury, immunohistochemical staining, and cell proliferation assays to address this hypothesis. RESULTS We found that MI induced collagen deposition and cardiac dysfunction. After MI, mice displayed reduced caveolin-1 and PTEN expression and increased α-smooth muscle actin (α-SMA) expression in the infarct zone. Qualitative and quantitative analyses indicated that caveolin-1 expression was lowest at 7 days after MI, accompanied by increased collagen deposition and attenuated cardiac function. We cultured cardiac fibroblasts of mice were in hypoxia or normoxia conditions for 12, 24 and 48 hours. At all the time points, caveolin-1 and PTEN expression were gradually reduced, whereas, α-SMA was gradually increased. We also observed that cell viability was increased at 12 and 24 h after hypoxia then lightly decreased at 48 h. Additionally, disruption of caveolae with methyl-β-cyclodextrin (MβCD) enhanced p-Akt and α-SMA expression and fibroblast proliferation and phenotypic switch. CONCLUSIONS These findings suggest a key role for caveolae, perhaps through the caveolin-1/PTEN signaling pathway, in cardiac fibroblast proliferation and phenotypic switch under hypoxia.
Collapse
Affiliation(s)
- Yao Gao
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Ming Chu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jian Hong
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Jingping Shang
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Di Xu
- Department of Geriatrics, First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|