1
|
Lian P, Li L, Lu R, Zhang B, Wazir J, Gu C, Ma B, Pu W, Cao W, Huang Z, Su Z, Wang H. S1PR3-driven positive feedback loop sustains STAT3 activation and keratinocyte hyperproliferation in psoriasis. Cell Death Dis 2025; 16:31. [PMID: 39833165 PMCID: PMC11746942 DOI: 10.1038/s41419-025-07358-w] [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: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Psoriasis is a chronic inflammatory skin disorder characterized by hyperproliferation of keratinocytes and persistent inflammation. Although persistent activation of signal transducer and activator of transcription 3 (STAT3) is implicated in its pathogenesis, the mechanisms underlying the sustained STAT3 activation remain poorly understood. Here, we identify sphingosine-1-phosphate receptor 3 (S1PR3) as a critical regulator of STAT3 activation and psoriasis pathogenesis, orchestrating a self-amplifying circuit that sustains keratinocyte hyperproliferation and chronic inflammation. S1PR3 expression is markedly elevated in psoriatic lesions and correlates with disease severity. Using genetic and pharmacological approaches, we reveal a novel S1PR3-Src-STAT3 signaling axis that drives both early and prolonged STAT3 activation in keratinocytes. Mechanistically, S1PR3 operates through Gαi/PKA-mediated Src activation, enhancing STAT3 phosphorylation and subsequent transcriptional activity. Importantly, we reveal a previously unrecognized positive feedback loop wherein activated STAT3 directly upregulates S1PR3 expression, perpetuating inflammation and hyperproliferation. Genetic deletion of S1pr3 in mice or pharmacological inhibition of S1PR3 significantly attenuates psoriasis-like skin inflammation, decreasing epidermal hyperplasia, dermal angiogenesis, and inflammatory mediator production. These findings provide new insights into the molecular mechanisms underlying psoriasis and identify S1PR3 as a promising therapeutic target. Our study suggests that disrupting the S1PR3-STAT3 feedback loop may offer a novel strategy for treating psoriasis and potentially other chronic inflammatory diseases driven by persistent STAT3 activation.
Collapse
Affiliation(s)
- Panpan Lian
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Li Li
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Renwei Lu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Bin Zhang
- Central Laboratory, Nanjing Chest Hospital, Affiliated Nanjing Brain Hospital, Nanjing Medical University, 210029, Nanjing, P.R. China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Chaode Gu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Bojie Ma
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Wenyuan Pu
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Wangsen Cao
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China.
| | - Zhonglan Su
- Department of Dermatology, First Affiliated Hospital, Nanjing Medical University, 210029, Nanjing, P.R. China.
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210093, Nanjing, P.R. China.
| |
Collapse
|
2
|
Zheng H, Yang L, Huang H, Lin Y, Chen L. Morroniside improves AngII-induced cardiac fibroblast proliferation, migration, and extracellular matrix deposition by blocking p38/JNK signaling pathway through the downregulation of KLF5. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6611-6621. [PMID: 38472369 PMCID: PMC11422283 DOI: 10.1007/s00210-024-03039-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Myocardial fibrosis (MF), which is an inevitable pathological manifestation of many cardiovascular diseases in the terminal stage, often contributes to severe cardiac dysfunction and sudden death. Morroniside (MOR) is the main active component of Cornus officinalis with a variety of biological activities. This study was designed to explore the efficacy of MOR in MF and to investigate its pharmacological mechanism. The viability of MOR-treated human cardiac fibroblast (HCF) cells with or without Angiotensin II (AngII) induction was assessed with Cell Counting Kit-8 (CCK-8). The migration of AngII-induced HCF cells was appraised with a transwell assay. Gelatin zymography analysis was adopted to evaluate the activities of MMP2 and MMP9, while immunofluorescence assay was applied for the estimation of Collagen I and Collagen III. By means of western blot, the expressions of migration-, fibrosis-, and p38/c-Jun N-terminal kinase (JNK) signal pathway-related proteins were resolved. The transfection efficacy of oe-Kruppel-like factor 5 (KLF5) was examined with reverse transcription-quantitative PCR (RT-qPCR) and western blot. In this study, it was found that MOR treatment inhibited AngII-induced hyperproliferation, migration, and fibrosis of HCF cells, accompanied with decreased activities of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), connective tissue growth factor (CTGF), Fibronectin, and α-SMA, which were all reversed by KLF5 overexpression. Collectively, MOR exerted protective effects on MF by blocking p38/JNK signal pathway through the downregulation of KLF5.
Collapse
Affiliation(s)
- Haotian Zheng
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China
| | - Linxin Yang
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Ultraphonic Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Huashang Huang
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China
| | - Yazhou Lin
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China.
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China.
| | - Lin Chen
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China.
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China.
| |
Collapse
|
3
|
Tsare EPG, Klapa MI, Moschonas NK. Protein-protein interaction network-based integration of GWAS and functional data for blood pressure regulation analysis. Hum Genomics 2024; 18:15. [PMID: 38326862 PMCID: PMC11465932 DOI: 10.1186/s40246-023-00565-6] [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: 08/08/2023] [Accepted: 11/12/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND It is valuable to analyze the genome-wide association studies (GWAS) data for a complex disease phenotype in the context of the protein-protein interaction (PPI) network, as the related pathophysiology results from the function of interacting polyprotein pathways. The analysis may include the design and curation of a phenotype-specific GWAS meta-database incorporating genotypic and eQTL data linking to PPI and other biological datasets, and the development of systematic workflows for PPI network-based data integration toward protein and pathway prioritization. Here, we pursued this analysis for blood pressure (BP) regulation. METHODS The relational scheme of the implemented in Microsoft SQL Server BP-GWAS meta-database enabled the combined storage of: GWAS data and attributes mined from GWAS Catalog and the literature, Ensembl-defined SNP-transcript associations, and GTEx eQTL data. The BP-protein interactome was reconstructed from the PICKLE PPI meta-database, extending the GWAS-deduced network with the shortest paths connecting all GWAS-proteins into one component. The shortest-path intermediates were considered as BP-related. For protein prioritization, we combined a new integrated GWAS-based scoring scheme with two network-based criteria: one considering the protein role in the reconstructed by shortest-path (RbSP) interactome and one novel promoting the common neighbors of GWAS-prioritized proteins. Prioritized proteins were ranked by the number of satisfied criteria. RESULTS The meta-database includes 6687 variants linked with 1167 BP-associated protein-coding genes. The GWAS-deduced PPI network includes 1065 proteins, with 672 forming a connected component. The RbSP interactome contains 1443 additional, network-deduced proteins and indicated that essentially all BP-GWAS proteins are at most second neighbors. The prioritized BP-protein set was derived from the union of the most BP-significant by any of the GWAS-based or the network-based criteria. It included 335 proteins, with ~ 2/3 deduced from the BP PPI network extension and 126 prioritized by at least two criteria. ESR1 was the only protein satisfying all three criteria, followed in the top-10 by INSR, PTN11, CDK6, CSK, NOS3, SH2B3, ATP2B1, FES and FINC, satisfying two. Pathway analysis of the RbSP interactome revealed numerous bioprocesses, which are indeed functionally supported as BP-associated, extending our understanding about BP regulation. CONCLUSIONS The implemented workflow could be used for other multifactorial diseases.
Collapse
Affiliation(s)
- Evridiki-Pandora G Tsare
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece
| | - Maria I Klapa
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| | - Nicholas K Moschonas
- Department of General Biology, School of Medicine, University of Patras, Patras, Greece.
- Metabolic Engineering and Systems Biology Laboratory, Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas (FORTH/ICE-HT), Patras, Greece.
| |
Collapse
|
4
|
Role of c-Src and reactive oxygen species in cardiovascular diseases. Mol Genet Genomics 2023; 298:315-328. [PMID: 36700976 DOI: 10.1007/s00438-023-01992-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
Oxidative stress, caused by the over production of oxidants or inactivity of antioxidants, can modulate the redox state of several target proteins such as tyrosine kinases, mitogen-activated protein kinases and tyrosine phosphatases. c-Src is one such non-receptor tyrosine kinase which activates NADPH oxidases (Noxs) in response to various growth factors and shear stress. Interaction between c-Src and Noxs is influenced by cell type and primary messengers such as angiotensin II, which binds to G-protein coupled receptor and activates the intracellular signaling cascade. c-Src stimulated activation of Noxs results in elevated release of intracellular and extracellular reactive oxygen species (ROS). These ROS species disturb vascular homeostasis and cause cardiac hypertrophy, coronary artery disease, atherosclerosis and hypertension. Interaction between c-Src and ROS in the pathobiology of cardiac fibrosis is hypothesized to be influenced by cell type and stimuli. c-Src and ROS have a bidirectional relationship, thus increased ROS levels due to c-Src mediated activation of Noxs can further activate c-Src by promoting the oxidation and sulfenylation of critical cysteine residues. This review highlights the role of c-Src and ROS in mediating downstream signaling pathways underlying cardiovascular diseases. Furthermore, due to the central role of c-Src in activation of various signaling proteins involved in differentiation, migration, proliferation, and cytoskeletal reorganization of vascular cells, it is presented as therapeutic target for treating cardiovascular diseases except cardiac fibrosis.
Collapse
|
5
|
Shu J, Shi J, Gu Y, Deng L, Zhao C, Wu C, Zhao J, Wang H, Jin L. Levocarnitine regulates the growth of angiotensin II-induced myocardial fibrosis cells via TIMP-1. Open Life Sci 2023; 18:20220554. [PMID: 36816804 PMCID: PMC9922061 DOI: 10.1515/biol-2022-0554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/03/2022] [Accepted: 11/30/2022] [Indexed: 02/11/2023] Open
Abstract
This study aimed to explore the effects of tissue inhibitor of metalloproteinases-1 (TIMP-1) on levocarnitine (LC)-mediated regulation of angiotensin II (AngII)-induced myocardial fibrosis (MF) and its underlying mechanisms. H9C2 cells were treated with AngII for 24 h to induce fibrosis. The cells were then treated with LC or transfected with TIMP-1-OE plasmid/si‑TIMP-1. Cell apoptosis, viability, migration, and related gene expression were analyzed. AngII treatment significantly upregulated Axl, α-SMA, and MMP3 expression (P < 0.05) and downregulated STAT4 and TIMP1 expression (P < 0.05) relative to the control levels. After transfection, cells with TIMP-1 overexpression/knockdown were successfully established. Compared with that of the control, AngII significantly inhibited cell viability and cell migration while promoting cell apoptosis (P < 0.05). LC and TIMP-1-OE transfection further suppressed cell viability and migration induced by Ang II and upregulated apoptosis, whereas si-TIMP-1 had the opposite effect. Furthermore, LC and TIMP-1-OE transfection downregulated Axl, AT1R, α-SMA, collagen III, Bcl-2, and MMP3 expression caused by AngII and upregulated caspase 3, p53, and STAT4 expression, whereas si-TIMP-1 had the opposite effect. TIMP-1 is therefore a potential therapeutic target for delaying MF progression.
Collapse
Affiliation(s)
- Jin Shu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Jue Shi
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Yiwen Gu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Lei Deng
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Chen Zhao
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Chun Wu
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Jiachen Zhao
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| | - Haiya Wang
- Department of Gerontology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200023, China
| | - Li Jin
- Department of Gerontology, Shibei Hospital of Jing’an District, Shanghai, 200443, China
| |
Collapse
|
6
|
Xu M, Bermea KC, Ayati M, Kim HB, Yang X, Medina A, Fu Z, Heravi A, Zhang X, Na CH, Everett AD, Gabrielson K, Foster DB, Paolocci N, Murphy AM, Ramirez-Correa GA. Alteration in tyrosine phosphorylation of cardiac proteome and EGFR pathway contribute to hypertrophic cardiomyopathy. Commun Biol 2022; 5:1251. [PMID: 36380187 PMCID: PMC9666710 DOI: 10.1038/s42003-022-04021-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Alterations of serine/threonine phosphorylation of the cardiac proteome are a hallmark of heart failure. However, the contribution of tyrosine phosphorylation (pTyr) to the pathogenesis of cardiac hypertrophy remains unclear. We use global mapping to discover and quantify site-specific pTyr in two cardiac hypertrophic mouse models, i.e., cardiac overexpression of ErbB2 (TgErbB2) and α myosin heavy chain R403Q (R403Q-αMyHC Tg), compared to control hearts. From this, there are significant phosphoproteomic alterations in TgErbB2 mice in right ventricular cardiomyopathy, hypertrophic cardiomyopathy (HCM), and dilated cardiomyopathy (DCM) pathways. On the other hand, R403Q-αMyHC Tg mice indicated that the EGFR1 pathway is central for cardiac hypertrophy, along with angiopoietin, ErbB, growth hormone, and chemokine signaling pathways activation. Surprisingly, most myofilament proteins have downregulation of pTyr rather than upregulation. Kinase-substrate enrichment analysis (KSEA) shows a marked downregulation of MAPK pathway activity downstream of k-Ras in TgErbB2 mice and activation of EGFR, focal adhesion, PDGFR, and actin cytoskeleton pathways. In vivo ErbB2 inhibition by AG-825 decreases cardiomyocyte disarray. Serine/threonine and tyrosine phosphoproteome confirm the above-described pathways and the effectiveness of AG-825 Treatment. Thus, altered pTyr may play a regulatory role in cardiac hypertrophic models.
Collapse
Affiliation(s)
- Mingguo Xu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Pediatrics, The Third People’s Hospital of Longgang District, Shenzhen, 518115 China
| | - Kevin C. Bermea
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Marzieh Ayati
- grid.449717.80000 0004 5374 269XDeparment of Computer Science/College of Engineering and Computer Science, University of Texas Rio Grande Valley School of Medicine, Edinburgh, Texas USA
| | - Han Byeol Kim
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xiaomei Yang
- grid.27255.370000 0004 1761 1174Department of Anesthesiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Andres Medina
- Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| | - Zongming Fu
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Amir Heravi
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Xinyu Zhang
- grid.27255.370000 0004 1761 1174Department of Cardiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Ji’nan, China
| | - Chan Hyun Na
- grid.21107.350000 0001 2171 9311Department of Neurology/Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Biological Chemistry/McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Allen D. Everett
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Kathleen Gabrielson
- grid.21107.350000 0001 2171 9311Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - D. Brian Foster
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Nazareno Paolocci
- grid.21107.350000 0001 2171 9311Department of Medicine/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.5608.b0000 0004 1757 3470Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Anne M. Murphy
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Genaro A. Ramirez-Correa
- grid.21107.350000 0001 2171 9311Department of Pediatrics/Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,Department of Molecular Science/UT Health Rio Grande Valley, McAllen, TX USA
| |
Collapse
|
7
|
Truncation of the N-terminus of cardiac troponin I initiates adaptive remodeling of the myocardial proteosome via phosphorylation of mechano-sensitive signaling pathways. Mol Cell Biochem 2022; 477:1803-1815. [PMID: 35316461 DOI: 10.1007/s11010-022-04414-3] [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: 10/15/2021] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
The cardiac isoform of troponin I has a unique N-terminal extension (~ 1-30 amino acids), which contributes to the modulation of cardiac contraction and relaxation. Hearts of various species including humans produce a truncated variant of cardiac troponin I (cTnI-ND) deleting the first ~ 30 amino acids as an adaption in pathophysiological conditions. In this study, we investigated the impact of cTnI-ND chronic expression in transgenic mouse hearts compared to wildtype (WT) controls (biological n = 8 in each group). We aimed to determine the global phosphorylation effects of cTnI-ND on the cardiac proteome, thereby determining the signaling pathways that have an impact on cardiac function. The samples were digested and isobarically labeled and equally mixed for relative quantification via nanoLC-MS/MS. The peptides were then enriched for phospho-peptides and bioinformatic analysis was done with Ingenuity Pathway Analysis (IPA). We found approximately 77% replacement of the endogenous intact cTnI with cTnI-ND in the transgenic mouse hearts with 1674 phospho-proteins and 2971 non-modified proteins. There were 73 significantly altered phospho-proteins; bioinformatic analysis identified the top canonical pathways as associated with integrin, protein kinase A, RhoA, and actin cytoskeleton signaling. Among the 73 phospho-proteins compared to controls cTnI-ND hearts demonstrated a significant decrease in paxillin and YAP1, which are known to play a role in cell mechano-sensing pathways. Our data indicate that cTnI-ND modifications in the sarcomere are sufficient to initiate changes in the phospho-signaling profile that may underly the chronic-adaptive response associated with cTnI cleavage in response to stressors by modifying mechano-sensitive signaling pathways.
Collapse
|
8
|
Chen Y, Huang M, Yan Y, He D. Tranilast inhibits angiotensin II-induced myocardial fibrosis through S100A11/ transforming growth factor-β (TGF-β1)/Smad axis. Bioengineered 2021; 12:8447-8456. [PMID: 34663163 PMCID: PMC8806955 DOI: 10.1080/21655979.2021.1982322] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tranilast has an ameliorative effect on myocardial fibrosis (MF), but the specific mechanism has not been studied. S100A11 is a key regulator of collagen expression in MF. In this paper, we will study the regulatory roles of Tranilast and S100A11 in MF. After the introduction of angiotensin II (AngII) to Human cardiac fibroblasts (HCF), Tranilast was administered. CCK-8 kit was used to detect cell viability. Wound Healing assay detected cell migration, and Western blot was used to detect the expression of migration-related proteins and proteins related to extracellular matrix synthesis. The expression of α-SMA was detected by immunofluorescence (IF). The expression of S100A11 was detected by qPCR and Western blot, and then S100A11 was overexpressed by cell transfection technology, so as to explore the mechanism by which Tranilast regulated MF. In addition, the expression of TGF-β1/Smad pathway related proteins was detected by Western blot. Tranilast inhibited Ang II–induced over-proliferation, migration and fibrosis of human cardiac fibroblasts (HCF), and simultaneously significantly decreased S100A11 expression was observed. Overexpression of S100A11 reversed the inhibition of Tranilast on AngII–induced over-proliferation, migration, and fibrosis in HCF, accompanied by activation of the TGF-β1/Smad pathway. Overall, Tranilast inhibits angiotensin II-induced myocardial fibrosis through S100A11/TGF-β1/Smad axis.
Collapse
Affiliation(s)
- Youquan Chen
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Ming Huang
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Yi Yan
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| | - Dequan He
- Department of Cardiology, Third Affiliated Hospital of Guangzhou Medical University, Guangdong, China
| |
Collapse
|
9
|
Lino CA, de Bortoli Teixeira L, Capelupe Simões S, de Oliveira Silva T, Diniz GP, da Costa-Neto CM, Barreto-Chaves MLM. Beta-arrestin 2 mediates cardiac hypertrophy induced by thyroid hormones via AT1R. J Cell Physiol 2021; 236:4640-4654. [PMID: 33345322 DOI: 10.1002/jcp.30187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/30/2022]
Abstract
We have previously reported that angiotensin II receptor type 1 (AT1R) contributes to the hypertrophic effects of thyroid hormones (TH) in cardiac cells. Even though evidence indicates crosstalks between TH and AT1R, the underlying mechanisms are poorly understood. Beta-arrestin (ARRB) signaling has been described as noncanonical signal transduction pathway that exerts important effects in the cardiovascular system through G-protein-coupled receptors, as AT1R. Herein, we investigated the contribution of ARRB signaling in TH-induced cardiomyocyte hypertrophy. Primary cardiomyocyte cultures were treated with Triiodothyronine (T3) to induce cell hypertrophy. T3 rapidly activates extracellular signal-regulated kinase 1/2 (ERK1/2) signaling, which was partially inhibited by AT1R blockade. Also, ERK1/2 inhibition attenuated the hypertrophic effects of T3. ARRB2 was upregulated by T3, and small interfering RNA assays revealed the role of ARRB2-but not ARRB1-on ERK1/2 activation and cardiomyocyte hypertrophy. Corroborating these findings, the ARRB2-overexpressed cells showed increased expression of hypertrophic markers, which were attenuated by ERK1/2 inhibition. Immunocytochemistry and immunoprecipitation assays revealed the increased expression of nuclear AT1R after T3 stimulation and the increased interaction of AT1R/ARRB2. The inhibition of endocytosis also attenuated the T3 effects on cardiac cells. Our results evidence the contribution of ARRB2 on ERK1/2 activation and cardiomyocyte hypertrophy induced by T3 via AT1R.
Collapse
Affiliation(s)
- Caroline Antunes Lino
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Larissa de Bortoli Teixeira
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sarah Capelupe Simões
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | | - Gabriela Placoná Diniz
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Claudio Miguel da Costa-Neto
- Department of Biochemistry and Immunology, Ribeirao Preto Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | | |
Collapse
|
10
|
Liu S, Luttrell LM, Premont RT, Rockey DC. β-Arrestin2 is a critical component of the GPCR-eNOS signalosome. Proc Natl Acad Sci U S A 2020; 117:11483-11492. [PMID: 32404425 PMCID: PMC7261012 DOI: 10.1073/pnas.1922608117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell nitric oxide (NO) synthase (eNOS), the enzyme responsible for synthesis of NO in endothelial cells, is regulated by complex posttranslational mechanisms. Sinusoidal portal hypertension, a disorder characterized by liver sinusoidal endothelial cell (SEC) injury with resultant reduced eNOS activity and NO production within the liver, has been associated with defects in eNOS protein-protein interactions and posttranslational modifications. We and others have previously identified novel eNOS interactors, including G protein-coupled receptor (GPCR) kinase interactor 1 (GIT1), which we found to play an unexpected stimulatory role in GPCR-mediated eNOS signaling. Here we report that β-arrestin 2 (β-Arr2), a canonical GPCR signaling partner, localizes in SECs with eNOS in a GIT1/eNOS/NO signaling module. Most importantly, we show that β-Arr2 stimulates eNOS activity, and that β-Arr2 expression is reduced and formation of the GIT1/eNOS/NO signaling module is interrupted during liver injury. In β-Arr2-deficient mice, bile duct ligation injury (BDL) led to significantly reduced eNOS activity and to a dramatic increase in portal hypertension compared to BDL in wild-type mice. Overexpression of β-Arr2 in injured or β-Arr2-deficient SECs rescued eNOS function by increasing eNOS complex formation and NO production. We also found that β-Arr2-mediated GIT1/eNOS complex formation is dependent on Erk1/2 and Src, two kinases known to interact with and be activated by β-Arr2 in response to GCPR activation. Our data emphasize that β-Arr2 is an integral component of the GIT1/eNOS/NO signaling pathway and have implications for the pathogenesis of sinusoidal portal hypertension.
Collapse
Affiliation(s)
- Songling Liu
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425
| | - Louis M Luttrell
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425
| | - Richard T Premont
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Don C Rockey
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425;
| |
Collapse
|
11
|
Wu J, You J, Wang X, Wang S, Huang J, Xie Q, Gong B, Ding Z, Ye Y, Wang C, Kang L, Xu R, Li Y, Chen R, Sun A, Yang X, Jiang H, Yang F, Backx PH, Ge J, Zou Y. Left ventricular response in the transition from hypertrophy to failure recapitulates distinct roles of Akt, β-arrestin-2, and CaMKII in mice with aortic regurgitation. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:219. [PMID: 32309366 PMCID: PMC7154424 DOI: 10.21037/atm.2020.01.51] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Background Although aortic regurgitation (AR) is a clinically important condition that is becoming increasingly common, few relevant murine models and mechanistic studies exist for this condition. In this study, we attempted to delineate the pathological and molecular changes and address the roles of some potentially relevant molecules in an animal model of surgically induced AR. Methods AR was induced by puncturing the aortic valve leaflets in C57BL/6J mice under echocardiographic guidance. Results As early as 1 week following AR, the left ventricles (LV) displayed marked impairments in diastolic function and coronary flow reserve (CFR), as well as cardiac hypertrophy and chamber dilatation at both end-systole and end-diastole. LV free wall thickening and cardiomyocyte hypertrophy in LV were observed 2 weeks following of AR while a decline in ejection fraction was not seen until after 4 weeks. Nppa (natriuretic peptide A) and Nppb (natriuretic peptide B) increased over time, in conjunction with prominent Akt activation as well as slight CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation and biphasic changes in β-arrestin-2 expression. Treatment of AR mice with Akt inhibition exacerbated the eccentric hypertrophy, while neither inhibition of CaMKII nor β-arrestin-2 overexpression influenced the response to AR. Conclusions Our structural, functional, molecular and therapeutic analyses reveal that Akt, but not CaMKII or β-arrestin-2, plays a regulatory role in the development of LV remodeling after AR in Mice. These results may shed important light on therapeutic targets for volume overloaded cardiomyopathy.
Collapse
Affiliation(s)
- Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jieyun You
- Department of Cardiovascular Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Jiayuan Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Qihai Xie
- Department of Cardiology, Shanghai Jiading District Central Hospital, Shanghai 201800, China
| | - Baoyong Gong
- Guangdong Laboratory Animal Monitoring Institute, Guangzhou 510663, China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yong Ye
- Department of Cardiovascular Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Cong Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Le Kang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ran Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yang Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ruizhen Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Aijun Sun
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Hong Jiang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Fenghua Yang
- Guangdong Laboratory Animal Monitoring Institute, Guangzhou 510663, China
| | - Peter H Backx
- Department of Biology, York University, Toronto, ON, Canada.,Division of Cardiology, Peter Munk Heart Centre, University Health Network, Toronto, ON, Canada
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Ward M, Iskratsch T. Mix and (mis-)match - The mechanosensing machinery in the changing environment of the developing, healthy adult and diseased heart. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118436. [PMID: 30742931 PMCID: PMC7042712 DOI: 10.1016/j.bbamcr.2019.01.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/07/2019] [Accepted: 01/29/2019] [Indexed: 01/01/2023]
Abstract
The composition and the stiffness of cardiac microenvironment change during development and/or in heart disease. Cardiomyocytes (CMs) and their progenitors sense these changes, which decides over the cell fate and can trigger CM (progenitor) proliferation, differentiation, de-differentiation or death. The field of mechanobiology has seen a constant increase in output that also includes a wealth of new studies specific to cardiac or cardiomyocyte mechanosensing. As a result, mechanosensing and transduction in the heart is increasingly being recognised as a main driver of regulating the heart formation and function. Recent work has for instance focused on measuring the molecular, physical and mechanical changes of the cellular environment - as well as intracellular contributors to the passive stiffness of the heart. On the other hand, a variety of new studies shed light into the molecular machinery that allow the cardiomyocytes to sense these properties. Here we want to discuss the recent work on this topic, but also specifically focus on how the different components are regulated at various stages during development, in health or disease in order to highlight changes that might contribute to disease progression and heart failure.
Collapse
Key Words
- cm, cardiomyocytes
- hcm, hypertrophic cardiomyopathy
- dcm, dilated cardiomyopathy
- icm, idiopathic cardiomyopathy
- myh, myosin heavy chain
- tnnt, troponin t
- tnni, troponin i
- afm, atomic force microscope
- mre, magnetic resonance elastography
- swe, ultrasound cardiac shear-wave elastography
- lv, left ventricle
- lox, lysyl oxidase
- loxl, lysyl oxidase like protein
- lh, lysyl hydroxylase
- lys, lysin
- lccs, lysald-derived collagen crosslinks
- hlccs, hylald-derived collagen crosslinks
- pka, protein kinase a
- pkc, protein kinase c
- vash1, vasohibin-1
- svbp, small vasohibin binding protein
- tcp, tubulin carboxypeptidase
- ttl, tubulin tyrosine ligase
- mrtf, myocardin-related transcription factor
- gap, gtpase activating protein
- gef, guanine nucleotide exchange factor
Collapse
Affiliation(s)
- Matthew Ward
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom
| | - Thomas Iskratsch
- Division of Bioengineering, School of Engineering and Materials Science & Institute for Bioengineering, Queen Mary University of London, United Kingdom.
| |
Collapse
|
13
|
Yang L, He J, Xia G, Yang J, Tang Q, Yang Y, Deng J. Crim1 suppresses left ventricular hypertrophy. Biomed Rep 2019; 1:1-5. [PMID: 31258903 DOI: 10.3892/br.2019.1214] [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: 11/02/2018] [Accepted: 05/02/2019] [Indexed: 11/06/2022] Open
Abstract
Left ventricular hypertrophy is a leading cause of heart failure and sudden death. Cysteine-rich transmembrane bone morphogenetic protein regulator 1 (Crim1) is expressed at a high level in the heart and has a regulatory role in heart development. The present study aimed to test the hypothesis that Crim1 can have an inhibitory function on ventricular hypertrophy. Rat primary ventricular myocytes were stretched to induce myocyte hypertrophy, and treated with telmisartan or infected with Crim1-expressing recombinant adenovirus (Ad-Crim1). Rat ventricular hypertrophy was induced by abdominal aortic coarctation (AAC), and treated either with telmisartan or myocardial injection of Ad-Crim1 or empty adenovirus vector. The results showed that the expression of Crim1 decreased in the hypertrophic ventricle. The inhibition of angiotensin receptor type 1 (AT1R) by telmisartan in vitro and in vivo significantly increased the expression of Crim1 in the left ventricle. The overexpression of Crim1 by infection with Ad-Crim1 significantly inhibited stretch-induced ventricular myocyte hypertrophy in vitro. The overexpression of Crim1 by gavage with AT1R inhibitor telmisartan or myocardial injection of Ad-Crim1 markedly suppressed AAC-induced left ventricular hypertrophy in vivo. These results suggest that Crim1 has a suppressive function on ventricular hypertrophy and provides a novel therapeutic target for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Long Yang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Jionghong He
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Guiling Xia
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Jun Yang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qian Tang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Yongyao Yang
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Jiusheng Deng
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
14
|
Turu G, Balla A, Hunyady L. The Role of β-Arrestin Proteins in Organization of Signaling and Regulation of the AT1 Angiotensin Receptor. Front Endocrinol (Lausanne) 2019; 10:519. [PMID: 31447777 PMCID: PMC6691095 DOI: 10.3389/fendo.2019.00519] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/15/2019] [Indexed: 12/30/2022] Open
Abstract
AT1 angiotensin receptor plays important physiological and pathophysiological roles in the cardiovascular system. Renin-angiotensin system represents a target system for drugs acting at different levels. The main effects of ATR1 stimulation involve activation of Gq proteins and subsequent IP3, DAG, and calcium signaling. It has become evident in recent years that besides the well-known G protein pathways, AT1R also activates a parallel signaling pathway through β-arrestins. β-arrestins were originally described as proteins that desensitize G protein-coupled receptors, but they can also mediate receptor internalization and G protein-independent signaling. AT1R is one of the most studied receptors, which was used to unravel the newly recognized β-arrestin-mediated pathways. β-arrestin-mediated signaling has become one of the most studied topics in recent years in molecular pharmacology and the modulation of these pathways of the AT1R might offer new therapeutic opportunities in the near future. In this paper, we review the recent advances in the field of β-arrestin signaling of the AT1R, emphasizing its role in cardiovascular regulation and heart failure.
Collapse
Affiliation(s)
- Gábor Turu
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
| | - András Balla
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Hunyady
- Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- MTA-SE Laboratory of Molecular Physiology, Semmelweis University, Hungarian Academy of Sciences, Budapest, Hungary
- *Correspondence: László Hunyady
| |
Collapse
|
15
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 693] [Impact Index Per Article: 99.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
16
|
Wang Y, Jin L, Song Y, Zhang M, Shan D, Liu Y, Fang M, Lv F, Xiao RP, Zhang Y. β-arrestin 2 mediates cardiac ischemia-reperfusion injury via inhibiting GPCR-independent cell survival signalling. Cardiovasc Res 2018; 113:1615-1626. [PMID: 29016703 DOI: 10.1093/cvr/cvx147] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 08/03/2017] [Indexed: 01/14/2023] Open
Abstract
Aims Ischemic heart disease is a leading cause of morbidity and mortality worldwide. Although timely restoration of coronary blood flow (reperfusion) is the most effective therapeutics of myocardial infarction, reperfusion causes further cardiac damage, i.e. ischemia-reperfusion (I/R) injury. β-arrestins (Arrbs) have been traditionally defined as negative regulators of G protein-coupled receptor (GPCR) signalling, but recent studies have shown that they are essential for G protein-independent, GPCR-mediated biased signalling. Several ligands have been reported to be cardioprotective via Arrbs dependent pathway. However, it is unclear whether Arrbs exert receptor-independent physiological or pathological functions in the heart. Here, we sought to determine whether and how Arrbs play a role in regulating cardiomyocyte viability and myocardial remodelling following I/R injury. Methods and results The expression of β-arrestin 2 (Arrb2), but not β-arrestin 1 (Arrb1), is upregulated in rat hearts subjected to I/R injury, or in cultured neonatal rat cardiomyocytes treated with hypoxia-reoxygenation (H/R) injury. Deficiency of Arrb2 in cultured neonatal rat cardiomyocytes alleviates H/R-induced cardiomyocyte death and Arrb2-/- mice are resistant to myocardial damage caused by I/R injury. In contrast, upregulation of Arrb2 triggers cardiomyocyte death and exaggerates I/R (or H/R)-induced detrimental effects. Mechanically, Arrb2 induces cardiomyocyte death by interacting with the p85 subunit of PI3K, and negatively regulating the formation of p85-PI3K/CaV3 survival complex, thus blocking activation of PI3K-Akt-GSK3β cell survival signalling pathway. Conclusion We define an upregulation of Arrb2 as a pathogenic factor in cardiac I/R injury, and also reveal a novel GPCR-independent mechanism of Arrb2-mediated cell death signalling in the heart.
Collapse
Affiliation(s)
- Yimei Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Li Jin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Ying Song
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Mao Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Dan Shan
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Yuli Liu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Meng Fang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Fengxiang Lv
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui-Ping Xiao
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| |
Collapse
|
17
|
Abstract
β-arrestin1 (or arrestin2) and β-arrestin2 (or arrestin3) are ubiquitously expressed cytosolic adaptor proteins that were originally discovered for their inhibitory role in G protein-coupled receptor (GPCR) signaling through heterotrimeric G proteins. However, further biochemical characterization revealed that β-arrestins do not just "block" the activated GPCRs, but trigger endocytosis and kinase activation leading to specific signaling pathways that can be localized on endosomes. The signaling pathways initiated by β-arrestins were also found to be independent of G protein activation by GPCRs. The discovery of ligands that blocked G protein activation but promoted β-arrestin binding, or vice-versa, suggested the exciting possibility of selectively activating intracellular signaling pathways. In addition, it is becoming increasingly evident that β-arrestin-dependent signaling is extremely diverse and provokes distinct cellular responses through different GPCRs even when the same effector kinase is involved. In this review, we summarize various signaling pathways mediated by β-arrestins and highlight the physiologic effects of β-arrestin-dependent signaling.
Collapse
|
18
|
Luckey SW, Haines CD, Konhilas JP, Luczak ED, Messmer-Kratzsch A, Leinwand LA. Cyclin D2 is a critical mediator of exercise-induced cardiac hypertrophy. Exp Biol Med (Maywood) 2017; 242:1820-1830. [PMID: 28901173 PMCID: PMC5714145 DOI: 10.1177/1535370217731503] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/23/2017] [Indexed: 01/19/2023] Open
Abstract
A number of signaling pathways underlying pathological cardiac hypertrophy have been identified. However, few studies have probed the functional significance of these signaling pathways in the context of exercise or physiological pathways. Exercise studies were performed on females from six different genetic mouse models that have been shown to exhibit alterations in pathological cardiac adaptation and hypertrophy. These include mice expressing constitutively active glycogen synthase kinase-3β (GSK-3βS9A), an inhibitor of CaMK II (AC3-I), both GSK-3βS9A and AC3-I (GSK-3βS9A/AC3-I), constitutively active Akt (myrAkt), mice deficient in MAPK/ERK kinase kinase-1 (MEKK1-/-), and mice deficient in cyclin D2 (cyclin D2-/-). Voluntary wheel running performance was similar to NTG littermates for five of the mouse lines. Exercise induced significant cardiac growth in all mouse models except the cyclin D2-/- mice. Cardiac function was not impacted in the cyclin D2-/- mice and studies using a phospho-antibody array identified six proteins with increased phosphorylation (greater than 150%) and nine proteins with decreased phosphorylation (greater than 33% decrease) in the hearts of exercised cyclin D2-/- mice compared to exercised NTG littermate controls. Our results demonstrate that unlike the other hypertrophic signaling molecules tested here, cyclin D2 is an important regulator of both pathologic and physiological hypertrophy. Impact statement This research is relevant as the hypertrophic signaling pathways tested here have only been characterized for their role in pathological hypertrophy, and not in the context of exercise or physiological hypertrophy. By using the same transgenic mouse lines utilized in previous studies, our findings provide a novel and important understanding for the role of these signaling pathways in physiological hypertrophy. We found that alterations in the signaling pathways tested here had no impact on exercise performance. Exercise induced cardiac growth in all of the transgenic mice except for the mice deficient in cyclin D2. In the cyclin D2 null mice, cardiac function was not impacted even though the hypertrophic response was blunted and a number of signaling pathways are differentially regulated by exercise. These data provide the field with an understanding that cyclin D2 is a key mediator of physiological hypertrophy.
Collapse
Affiliation(s)
- Stephen W Luckey
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Biology Department, Seattle University, Seattle, WA 98122, USA
| | - Chris D Haines
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| | - John P Konhilas
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Sarver Molecular Cardiovascular Research Program, Department of Physiology, University of Arizona, Tucson, AZ 85724, USA
| | - Elizabeth D Luczak
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
- Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Antke Messmer-Kratzsch
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| | - Leslie A Leinwand
- Department of Molecular, Cellular and Developmental Biology and BioFrontiers Institute University of Colorado at Boulder, Boulder, CO 80309, USA
| |
Collapse
|
19
|
Blaustein MP. How does pressure overload cause cardiac hypertrophy and dysfunction? High-ouabain affinity cardiac Na + pumps are crucial. Am J Physiol Heart Circ Physiol 2017; 313:H919-H930. [PMID: 28733446 PMCID: PMC5792198 DOI: 10.1152/ajpheart.00131.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/17/2022]
Abstract
Left ventricular hypertrophy is frequently observed in hypertensive patients and is believed to be due to the pressure overload and cardiomyocyte stretch. Three recent reports on mice with genetically engineered Na+ pumps, however, have demonstrated that cardiac ouabain-sensitive α2-Na+ pumps play a key role in the pathogenesis of transaortic constriction-induced hypertrophy. Hypertrophy was delayed/attenuated in mice with mutant, ouabain-resistant α2-Na+ pumps and in mice with cardiac-selective knockout or transgenic overexpression of α2-Na+ pumps. The latter, seemingly paradoxical, findings can be explained by comparing the numbers of available (ouabain-free) high-affinity (α2) ouabain-binding sites in wild-type, knockout, and transgenic hearts. Conversely, hypertrophy was accelerated in α2-ouabain-resistant (R) mice in which the normally ouabain-resistant α1-Na+ pumps were mutated to an ouabain-sensitive (S) form (α1S/Sα2R/R or "SWAP" vs. wild-type or α1R/R α2S/S mice). Furthermore, transaortic constriction-induced hypertrophy in SWAP mice was prevented/reversed by immunoneutralizing circulating endogenous ouabain (EO). These findings show that EO and its receptor, ouabain-sensitive α2, are critical factors in pressure overload-induced cardiac hypertrophy. This complements reports linking elevated plasma EO to hypertension, cardiac hypertrophy, and failure in humans and elucidates the underappreciated role of the EO-Na+ pump pathway in cardiovascular disease.
Collapse
Affiliation(s)
- Mordecai P. Blaustein
- Departments of Physiology and Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
20
|
Mapping Heart Development in Flies: Src42A Acts Non-Autonomously to Promote Heart Tube Formation in Drosophila. Vet Sci 2017; 4:vetsci4020023. [PMID: 29056682 PMCID: PMC5606601 DOI: 10.3390/vetsci4020023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 11/27/2016] [Accepted: 12/07/2016] [Indexed: 12/26/2022] Open
Abstract
Congenital heart defects, clinically identified in both small and large animals, are multifactorial and complex. Although heritable factors are known to have a role in cardiovascular disease, the full genetic aetiology remains unclear. Model organism research has proven valuable in providing a deeper understanding of the essential factors in heart development. For example, mouse knock-out studies reveal a role for the Integrin adhesion receptor in cardiac tissue. Recent research in Drosophila melanogaster (the fruit fly), a powerful experimental model, has demonstrated that the link between the extracellular matrix and the cell, mediated by Integrins, is required for multiple aspects of cardiogenesis. Here we test the hypothesis that Integrins signal to the heart cells through Src42A kinase. Using the powerful genetics and cell biology analysis possible in Drosophila, we demonstrate that Src42A acts in early events of heart tube development. Careful examination of mutant heart tissue and genetic interaction data suggests that Src42A’s role is independent of Integrin and the Integrin-related Focal Adhesion Kinase. Rather, Src42A acts non-autonomously by promoting programmed cell death of the amnioserosa, a transient tissue that neighbors the developing heart.
Collapse
|
21
|
Carneiro AP, Fonseca-Alaniz MH, Dallan LAO, Miyakawa AA, Krieger JE. β-arrestin is critical for early shear stress-induced Akt/eNOS activation in human vascular endothelial cells. Biochem Biophys Res Commun 2017; 483:75-81. [PMID: 28062183 DOI: 10.1016/j.bbrc.2017.01.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/03/2017] [Indexed: 01/14/2023]
Abstract
Recent evidence suggests that β-arrestins, which are involved in G protein-coupled receptors desensitization, may influence mechanotransduction. Here, we observed that nitric oxide (NO) production was abrogated in human saphenous vein endothelial cells (SVECs) transfected with siRNA against β-arrestin 1 and 2 subjected to shear stress (SS, 15 dynes/cm2, 10 min). The downregulation of β-arrestins 1/2 in SVECs cells also prevented the SS-induced rise in levels of phosphorylation of Akt and endothelial nitric oxide synthase (eNOS, Serine 1177). Interestingly, immunoprecipitation revealed that β-arrestin interacts with Akt, eNOS and caveolin-1 and these interactions are not influenced by SS. Our data indicate that β-arrestins and Akt/eNOS downstream signaling are required for early SS-induced NO production in SVECs, which is consistent with the idea that β-arrestins and caveolin-1 are part of a pre-assembled complex associated with the cellular mechanotransduction machinery.
Collapse
Affiliation(s)
- Ana Paula Carneiro
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | | | | | - Ayumi Aurea Miyakawa
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Jose Eduardo Krieger
- Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, Brazil.
| |
Collapse
|
22
|
Shradhanjali A, Riehl BD, Kwon IK, Lim JY. Cardiomyocyte stretching for regenerative medicine and hypertrophy study. Tissue Eng Regen Med 2015. [DOI: 10.1007/s13770-015-0010-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
23
|
Major JL, Salih M, Tuana BS. Interplay between the E2F pathway and β-adrenergic signaling in the pathological hypertrophic response of myocardium. J Mol Cell Cardiol 2015; 84:179-90. [PMID: 25944088 DOI: 10.1016/j.yjmcc.2015.04.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 04/15/2015] [Accepted: 04/29/2015] [Indexed: 12/28/2022]
Abstract
The E2F/Pocket protein (Rb) pathway regulates cell growth, differentiation, and death by modulating gene expression. We previously examined this pathway in the myocardium via manipulation of the unique E2F repressor, E2F6, which is believed to repress gene activity independently of Rb. Mice with targeted expression of E2F6 in postnatal myocardium developed dilated cardiomyopathy (DCM) without hypertrophic growth. We assessed the mechanisms of the apparent failure of compensatory hypertrophic growth as well as their response to the β-adrenergic agonist isoproterenol. As early as 2 weeks, E2F6 transgenic (Tg) mice present with dilated thinner left ventricles and significantly reduced ejection fraction and fractional shortening which persists at 6 weeks of age, but with no apparent increase in left ventricle weight: body weight (LVW:BW). E2F6-Tg mice treated with isoproterenol (6.1 mg/kg/day) show double the increase in LVW:BW than their Wt counterparts (32% vs 16%, p-value: 0.007). Western blot analysis revealed the activation of the adrenergic pathway in Tg heart tissue under basal conditions with ~2-fold increase in the level of β2-adrenergic receptors (p-value: 8.9E-05), protein kinase A catalytic subunit (PKA-C) (p-value: 0.0176), activated c-Src tyrosine-protein kinase (p-value: 0.0002), extracellular receptor kinase 2 (ERK2) (p-value: 0.0005), and induction of the anti-apoptotic protein Bcl2 (p-value 0. 0.00001). In contrast, a ~60% decrease in the cardiac growth regulator: AKT1 (p-value 0.0001) and a ~four fold increase in cyclic AMP dependent phosphodiesterase 4D (PDE4D), the negative regulator of PKA activity, were evident in the myocardium of E2F6-Tg mice. The expression of E2F3 was down-regulated by E2F6, but was restored by isoproterenol. Further, Rb expression was down-regulated in Tg mice in response to isoproterenol implying a net activation of the E2F pathway. Thus the unique regulation of E2F activity by E2F6 renders the myocardium hypersensitive to adrenergic stimulus resulting in robust hypertrophic growth. These data reveal a novel interplay between the E2F pathway, β2-adrenergic/PKA/PDE4D, and ERK/c-Src axis in fine tuning the pathological hypertrophic growth response. E2F6 deregulates E2F3 such that pro-hypertrophic growth and survival are enhanced via β2-adrenergic signaling however this response is outweighed by the induction of anti-hypertrophic signals so that left ventricle dilation proceeds without any increase in muscle mass.
Collapse
Affiliation(s)
- Jennifer L Major
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Maysoon Salih
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| | - Balwant S Tuana
- Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada; University of Ottawa Heart Institute, 40 Ruskin Street, Ottawa, Ontario K1Y 4W7, Canada.
| |
Collapse
|
24
|
Hamzeh MT, Sridhara R, Alexander LD. Cyclic stretch-induced TGF-β1 and fibronectin expression is mediated by β1-integrin through c-Src- and STAT3-dependent pathways in renal epithelial cells. Am J Physiol Renal Physiol 2014; 308:F425-36. [PMID: 25477471 DOI: 10.1152/ajprenal.00589.2014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Extracellular matrix (ECM) proteins, including fibronectin, may contribute to the early development and progression of renal interstitial fibrosis associated with chronic renal disease. Recent studies showed that β1-integrin is associated with the development of renal fibrosis in a murine model of unilateral ureteral obstruction (UUO). However, the molecular events responsible for β1-integrin-mediated signaling, following UUO, have yet to be determined. In this study, we investigated the mechanism by which mechanical stretch, an in vitro model for chronic obstructive nephropathy, regulates fibronectin and transforming growth factor-β1 (TGF-β1) expression in cultured human proximal tubular epithelium (HK-2) cells. Mechanical stretch upregulated fibronectin and TGF-β1 expression and activated signal transducer and transcription factor 3 (STAT3) in a time-dependent manner. Stretch-induced fibronectin and TGF-β1 were suppressed by a STAT3 inhibitor, S3I-201, and by small interfering RNA (siRNA) targeting human STAT3 (STAT3 siRNA). Similarly, fibronectin and TGF-β1 expression and STAT3 activation induced by mechanical stretch were suppressed by the Src family kinase inhibitor PP2 and by transfection of HK-2 cells with a dominant-negative mutant of c-Src (DN-Src), whereas PP3, an inactive analog of PP2, had no significant effect. Furthermore, mechanical stretch resulted in increased β1-integrin mRNA and protein levels in HK-2 cells. Furthermore, neutralizing antibody against β1-integrin and silencing of β1-integrin expression with siRNAs resulted in decreased c-Src and STAT3 activation and TGF-β1 and fibronectin expression evoked by mechanical stretch. This work demonstrates, for the first time, a role for β1-integrin in stretch-induced renal fibrosis through the activation of c-Src and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Mona T Hamzeh
- Department of Biology, Division of Natural Sciences, University of Michigan-Dearborn, Dearborn, Michigan
| | - Rashmi Sridhara
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| | - Larry D Alexander
- Midwestern University, Arizona College of Osteopathic Medicine, Department of Physiology, Glendale, Arizona; and
| |
Collapse
|
25
|
Wang JH, Su F, Wang S, Lu XC, Zhang SH, Chen D, Chen NN, Zhong JQ. CXCR6 deficiency attenuates pressure overload-induced monocytes migration and cardiac fibrosis through downregulating TNF-α-dependent MMP9 pathway. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:6514-6523. [PMID: 25400729 PMCID: PMC4230124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 09/13/2014] [Indexed: 06/04/2023]
Abstract
An immerging role of TNF-α in collagen synthesis and cardiac fibrosis implies the significance of TNF-α production in the development of myocardial remodeling. Our previous study showed a reduction of TNF-α and attenuated cardiac remodeling in CXCR6 knockout (KO) mice after ischemia/reperfusion injury. However, the potential mechanism of TNF-α-mediated cardiac fibrosis with pressure overload has not been well elucidated. In the present study, we aim to investigate the role of CXCR6 in TNF-α release and myocardial remodeling in response to pressure overload. Pressure overload was performed by constriction of transverse aorta (TAC) surgery on CXCR6 KO mice and C57 wild-type (WT) counterparts. At 6 weeks after TAC, cardiac remodeling was assessed by echocardiography, cardiac TNF-α release and its type I receptor (TNFRI), were detected by ELISA and western blot, collagen genes Col1a1 (type I) and Col3a1 (type III) were examined by real-time PCR. Compared with CXCR6 WT mice, CXCR6 KO mice exhibited less cardiac dysfunction, reduced expression of TNFRI, Col1a1 and Col3a. In vitro, we confirmed that CXCR6 deficiency led to reduced homing and infiltration of CD11b(+) monocytes, which contributed to attenuated TNF-α release in myocardium. Furthermore, TNFRI antagonist pretreatment blocked AT1 receptor signaling and NOX4 expression, reduced collagen synthesis, and blunted the activity of MMP9 in CXCR6 WT mice after TAC, but these were not observed in CXCR6 KO mice. In the present work, we propose a mechanism that CXCR6 is essential for pressure overload-mediated myocardial recruitment of monocytes, which contributes to cardiac fibrosis through TNF-α-dependent MMP9 activation and collagen synthesis.
Collapse
MESH Headings
- Animals
- CD11b Antigen/metabolism
- Cells, Cultured
- Chemotaxis, Leukocyte
- Collagen Type I/genetics
- Collagen Type I/metabolism
- Collagen Type I, alpha 1 Chain
- Collagen Type III/genetics
- Collagen Type III/metabolism
- Disease Models, Animal
- Down-Regulation
- Enzyme Activation
- Fibrosis
- Heart Diseases/genetics
- Heart Diseases/immunology
- Heart Diseases/metabolism
- Heart Diseases/pathology
- Male
- Matrix Metalloproteinase 9/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/immunology
- Monocytes/metabolism
- Myocardium/immunology
- Myocardium/metabolism
- Myocardium/pathology
- NADPH Oxidase 4
- NADPH Oxidases/metabolism
- Receptors, CXCR/deficiency
- Receptors, CXCR/genetics
- Receptors, CXCR6
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/metabolism
- Ventricular Remodeling
Collapse
Affiliation(s)
- Jia-Hong Wang
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University250012, China
- Department of Cardiology, Yangpu Hospital Affiliated to Tongji UniversityShanghai 200090, China
| | - Feng Su
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University250012, China
- Department of Cardiology, Yangpu Hospital Affiliated to Tongji UniversityShanghai 200090, China
| | - Shijun Wang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan UniversityShanghai 200032, China
| | - Xian-Cheng Lu
- Department of Cardiology, Gongli HospitalShanghai 200135, China
| | - Shao-Heng Zhang
- Department of Cardiology, Yangpu Hospital Affiliated to Tongji UniversityShanghai 200090, China
| | - De Chen
- Department of Cardiology, Yangpu Hospital Affiliated to Tongji UniversityShanghai 200090, China
| | - Nan-Nan Chen
- Department of Cardiology, Yangpu Hospital Affiliated to Tongji UniversityShanghai 200090, China
| | - Jing-Quan Zhong
- Department of Cardiology, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital of Shandong University250012, China
| |
Collapse
|
26
|
Wu J, You J, Wang S, Zhang L, Gong H, Zou Y. Insights Into the Activation and Inhibition of Angiotensin II Type 1 Receptor in the Mechanically Loaded Heart. Circ J 2014; 78:1283-9. [DOI: 10.1253/circj.cj-14-0470] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| | - Jieyun You
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| | - Li Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University
| |
Collapse
|