1
|
Mohamed HE, Askar ME, Shaheen MA, Salama AE, Idris RA, Younis NN. Infliximab substantially re-silenced Wnt/β-catenin signaling and ameliorated doxorubicin-induced cardiomyopathy in rats. J Biochem Mol Toxicol 2023; 37:e23312. [PMID: 36636964 DOI: 10.1002/jbt.23312] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 11/19/2022] [Accepted: 01/05/2023] [Indexed: 01/14/2023]
Abstract
The release of inflammatory cytokines, namely tumor necrosis factor-α (TNF-α), plays an important role in the pathogenesis of cardiomyopathy. TNF-α increases in plasma and in myocardium of heart failure patients. We aimed to investigate the role of TNF-α inhibitor (infliximab; IFX) in regulating dilated cardiomyopathy (DCM) induced in rats. DCM was induced in rats by doxorubicin (DOX; 3.5 mg. kg-1 , i.p) twice weekly for 3 weeks (21 mg. kg-1 cumulative dose). DCM rats were treated with RPL (1 mg. kg-1 orally, daily), IFX (5 mg. kg-1 ; i.p. once) or their combination for 4 weeks starting next day of last DOX dose. Echocardiography was conducted followed by a collection of blood and left ventricle (LV) for biochemical and histological investigations. DCM rats revealed deteriorated cardiac function (increased CK-MB activity, LVIDs, LVIDd, ESV, and EDV, while decreased EF% and FS%), hypertrophy (increased HW/TL, β-MHC, and α-actin), inflammation (increased IL-1β, IL-6, and TNF-α). The activation of Wnt/β-catenin along with increased gene expression of RAS components (RENIN, ACE, and AT1) were evident. LV architecture also revealed abnormalities and some degree of fibrosis. Treatment with RPL and/or IFX suppressed TNF-α and consequently improved most of these parameters suppressing Wnt/β-catenin/RAS axis. Combined RPL and IFX treatment was the best among all treatments. In conclusion, Wnt/β-catenin/RAS axis is implicated in DOX-induced cardiomyopathy. The upstream TNF-α was proved for the first time in-vivo to stimulate this axis where its inhibition by RPL or IFX prevented DCM. Targeting this axis at two points using RPL and IFX showed better therapeutic efficacy.
Collapse
Affiliation(s)
- Hoda E Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mervat E Askar
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Shaheen
- Department of Histology and Cell Biology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Alaa E Salama
- Department of Cardiology, Faculty of Human Medicine, Zagazig University, Zagazig, Egypt
| | - Reham A Idris
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Nahla N Younis
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
2
|
Helfenberger KE, Argentino GF, Benzo Y, Herrera LM, Finocchietto P, Poderoso C. Angiotensin II Regulates Mitochondrial mTOR Pathway Activity Dependent on Acyl-CoA Synthetase 4 in Adrenocortical Cells. Endocrinology 2022; 163:6763139. [PMID: 36256598 DOI: 10.1210/endocr/bqac170] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 11/19/2022]
Abstract
Two well-known protein complexes in mammalian cells, mTOR type 1 and type 2 (mTORC1/2) are involved in several cellular processes such as protein synthesis, cell proliferation, and commonly dysregulated in cancer. An acyl-CoA synthetase type 4 (ACSL4) is one of the most recently mTORC1/2 regulators described, in breast cancer cells. The expression of ACSL4 is hormone-regulated in adrenocortical cells and required for steroid biosynthesis. mTORC1/2 have been reported to be crucial in the proliferation of human adrenocortical tumor cells H295R and interestingly reported at several subcellular locations, which has brought cell biology to the vanguard of the mTOR signaling field. In the present work, we study the regulation of mTORC1/2 activation by angiotensin II (Ang II)-the trophic hormone for adrenocortical cells-the subcellular localization of mTORC1/2 signaling proteins and the role of ACSL4 in the regulation of this pathway, in H295R cells. Ang II promotes activation by phosphorylation of mTORC1/2 pathway proteins in a time-dependent manner. Mitochondrial pools of ribosomal protein S6, protein kinase B (Akt) in threonine 308, and serine 473 and Rictor are phosphorylated and activated. Glycogen synthase kinase type 3 (GSK3) is phosphorylated and inactivated in mitochondria, favoring mTORC1 activation. Epidermal growth factor, a classic mTORC1/2 activator, promoted unique activation kinetics of mTORC1/2 pathway, except for Akt phosphorylation. Here, we demonstrate that ACSL4 is necessary for mTORC1/2 effectors phosphorylation and H295R proliferation, triggered by Ang II. Ang II promotes activation of mitochondrial mTORC1/2 signaling proteins, through ACSL4, with a direct effect on adrenocortical cellular proliferation.
Collapse
Affiliation(s)
- Katia E Helfenberger
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Giuliana F Argentino
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Yanina Benzo
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Lucía M Herrera
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Paola Finocchietto
- Laboratorio del Metabolismo del Oxígeno. Hospital de Clínicas "José de San Martín," Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| | - Cecilia Poderoso
- Departamento de Bioquímica Humana, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Buenos Aires-CONICET, Buenos Aires C1121ABG, Argentina
| |
Collapse
|
3
|
Czepiel M, Diviani D, Jaźwa-Kusior A, Tkacz K, Rolski F, Smolenski RT, Siedlar M, Eriksson U, Kania G, Błyszczuk P. Angiotensin II receptor 1 controls profibrotic Wnt/β-catenin signalling in experimental autoimmune myocarditis. Cardiovasc Res 2022; 118:573-584. [PMID: 33576779 PMCID: PMC8803091 DOI: 10.1093/cvr/cvab039] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 02/11/2021] [Indexed: 12/15/2022] Open
Abstract
AIMS Angiotensin (Ang) II signalling has been suggested to promote cardiac fibrosis in inflammatory heart diseases; however, the underlying mechanisms remain obscure. Using Agtr1a-/- mice with genetic deletion of angiotensin receptor type 1 (ATR1) and the experimental autoimmune myocarditis (EAM) model, we aimed to elucidate the role of Ang II-ATR1 pathway in development of heart-specific autoimmunity and post-inflammatory fibrosis. METHODS AND RESULTS EAM was induced in wild-type (WT) and Agtr1a-/- mice by subcutaneous injections with alpha myosin heavy chain peptide emulsified in complete Freund's adjuvant. Agtr1a-/- mice developed myocarditis to a similar extent as WT controls at day 21 but showed reduced fibrosis and better systolic function at day 40. Crisscross bone marrow chimaera experiments proved that ATR1 signalling in the bone marrow compartment was critical for cardiac fibrosis. Heart infiltrating, bone-marrow-derived cells produced Ang II, but lack of ATR1 in these cells reduced transforming growth factor beta (TGF-β)-mediated fibrotic responses. At the molecular level, Agtr1a-/- heart-inflammatory cells showed impaired TGF-β-mediated phosphorylation of Smad2 and TAK1. In WT cells, TGF-β induced formation of RhoA-GTP and RhoA-A-kinase anchoring protein-Lbc (AKAP-Lbc) complex. In Agtr1a-/- cells, stabilization of RhoA-GTP and interaction of RhoA with AKAP-Lbc were largely impaired. Furthermore, in contrast to WT cells, Agtr1a-/- cells stimulated with TGF-β failed to activate canonical Wnt pathway indicated by suppressed activity of glycogen synthase kinase-3 (GSK-3)β and nuclear β-catenin translocation and showed reduced expression of Wnts. In line with these in vitro findings, β-catenin was detected in inflammatory regions of hearts of WT, but not Agtr1a-/- mice and expression of canonical Wnt1 and Wnt10b were lower in Agtr1a-/- hearts. CONCLUSION Ang II-ATR1 signalling is critical for development of post-inflammatory fibrotic remodelling and dilated cardiomyopathy. Our data underpin the importance of Ang II-ATR1 in effective TGF-β downstream signalling response including activation of profibrotic Wnt/β-catenin pathway.
Collapse
MESH Headings
- Angiotensin II/metabolism
- Animals
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- Autoimmunity
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cells, Cultured
- Disease Models, Animal
- Fibrosis
- Inflammation Mediators/metabolism
- Lymphocyte Activation
- Mice, Inbred BALB C
- Mice, Knockout
- Myocarditis/genetics
- Myocarditis/immunology
- Myocarditis/metabolism
- Myocarditis/pathology
- Myocytes, Cardiac/immunology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Wnt Proteins/genetics
- Wnt Proteins/metabolism
- Wnt Signaling Pathway
- Wnt1 Protein/genetics
- Wnt1 Protein/metabolism
- beta Catenin/genetics
- beta Catenin/metabolism
- Mice
Collapse
Affiliation(s)
- Marcin Czepiel
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, 30-663, Cracow, Poland
| | - Dario Diviani
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, 1005, Lausanne, Switzerland
| | - Agnieszka Jaźwa-Kusior
- Department of Medical Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Cracow, Poland
| | - Karolina Tkacz
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, 30-663, Cracow, Poland
| | - Filip Rolski
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, 30-663, Cracow, Poland
| | - Ryszard T Smolenski
- Department of Biochemistry, Medical University of Gdansk, M. Skłodowskiej-Curie 3a, 80-210, Gdansk, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, 30-663, Cracow, Poland
| | - Urs Eriksson
- Cardioimmunology, Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland, GZO—Zurich Regional Health Center, Spitalstrasse 66, 8620, Wetzikon, Switzerland
| | - Gabriela Kania
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, Wielicka 265, 30-663, Cracow, Poland
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Wagistrasse 14, 8952 Schlieren, Switzerland
| |
Collapse
|
4
|
Rebolledo DL, Acuña MJ, Brandan E. Role of Matricellular CCN Proteins in Skeletal Muscle: Focus on CCN2/CTGF and Its Regulation by Vasoactive Peptides. Int J Mol Sci 2021; 22:5234. [PMID: 34063397 PMCID: PMC8156781 DOI: 10.3390/ijms22105234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/08/2023] Open
Abstract
The Cellular Communication Network (CCN) family of matricellular proteins comprises six proteins that share conserved structural features and play numerous biological roles. These proteins can interact with several receptors or soluble proteins, regulating cell signaling pathways in various tissues under physiological and pathological conditions. In the skeletal muscle of mammals, most of the six CCN family members are expressed during embryonic development or in adulthood. Their roles during the adult stage are related to the regulation of muscle mass and regeneration, maintaining vascularization, and the modulation of skeletal muscle fibrosis. This work reviews the CCNs proteins' role in skeletal muscle physiology and disease, focusing on skeletal muscle fibrosis and its regulation by Connective Tissue Growth factor (CCN2/CTGF). Furthermore, we review evidence on the modulation of fibrosis and CCN2/CTGF by the renin-angiotensin system and the kallikrein-kinin system of vasoactive peptides.
Collapse
Affiliation(s)
- Daniela L. Rebolledo
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6213515, Chile
| | - María José Acuña
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, Santiago 8370854, Chile
| | - Enrique Brandan
- Centro de Envejecimiento y Regeneración, CARE Chile UC, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Fundación Ciencia & Vida, Santiago 7810000, Chile
| |
Collapse
|
5
|
Abstract
The Wnt signaling pathway regulates physiological processes such as cell proliferation and differentiation, cell fate decisions, and stem cell maintenance and, thus, plays essential roles in embryonic development, but also in adult tissue homeostasis and repair. The Wnt signaling pathway has been associated with heart development and repair and has been shown to be crucially involved in proliferation and differentiation of progenitor cells into cardiomyocytes. The investigation of the role of the Wnt signaling pathway and the regulation of its expression/activity in atrial fibrillation has only just begun. The present minireview (I) provides original data regarding the expression of Wnt signaling components in atrial tissue of patients with atrial fibrillation or sinus rhythm and (II) summarizes the current state of knowledge of the regulation of Wnt signaling components' expression/activity and the contribution of the various levels of the Wnt signal transduction pathway to the processes of the development, maintenance, and progression of atrial fibrillation.
Collapse
Affiliation(s)
- Carmen Wolke
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Elmer Antileo
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| | - Uwe Lendeckel
- Institute of Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Greifswald D-17475, Germany
| |
Collapse
|
6
|
Ge W, Hou C, Zhang W, Guo X, Gao P, Song X, Gao R, Liu Y, Guo W, Li B, Zhao H, Wang J. Mep1a contributes to Ang II-induced cardiac remodeling by promoting cardiac hypertrophy, fibrosis and inflammation. J Mol Cell Cardiol 2020; 152:52-68. [PMID: 33301800 DOI: 10.1016/j.yjmcc.2020.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 11/20/2020] [Accepted: 11/26/2020] [Indexed: 01/12/2023]
Abstract
Pathological cardiac remodeling, characterized by excessive deposition of extracellular matrix proteins and cardiac hypertrophy, leads to the development of heart failure. Meprin α (Mep1a), a zinc metalloprotease, previously reported to participate in the regulation of inflammatory response and fibrosis, may also contribute to cardiac remodeling, although whether and how it participates in this process remains unknown. Here, in this work, we investigated the role of Mep1a in pathological cardiac remodeling, as well as the effects of the Mep1a inhibitor actinonin on cardiac remodeling-associated phenotypes. We found that Mep1a deficiency or chemical inhibition both significantly alleviated TAC- and Ang II-induced cardiac remodeling and dysfunction. Mep1a deletion and blocking both attenuated TAC- and Ang II-induced heart enlargement and increases in the thickness of the left ventricle anterior and posterior walls, and reduced expression of pro-hypertrophic markers, including atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and myosin heavy chain beta (β-MHC). In addition, Mep1a deletion and blocking significantly inhibited TAC- and Ang II-induced cardiac fibroblast activation and production of extracellular matrix (ECM). Moreover, in Mep1a-/- mice and treatment with actinonin significantly reduced Ang II-induced infiltration of macrophages and proinflammatory cytokines. Notably, we found that in vitro, Mep1a is expressed in cardiac myocytes and fibroblasts and that Mep1a deletion or chemical inhibition both markedly suppressed Ang II-induced hypertrophy of rat or mouse cardiac myocytes and activation of rat or mouse cardiac fibroblasts. In addition, blocking Mep1a in macrophages reduced Ang II-induced expression of interleukin (IL)-6 and IL-1β, strongly suggesting that Mep1a participates in cardiac remodeling processes through regulation of inflammatory cytokine expression. Mechanism studies revealed that Mep1a mediated ERK1/2 activation in cardiac myocytes, fibroblasts and macrophages and contributed to cardiac remodeling. In light of our findings that blocking Mep1a can ameliorate cardiac remodeling via inhibition of cardiac hypertrophy, fibrosis, and inflammation, Mep1a may therefore serve as a strong potential candidate for therapeutic targeting to prevent cardiac remodeling.
Collapse
Affiliation(s)
- Weipeng Ge
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Cuiliu Hou
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Wei Zhang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Pan Gao
- Department of Geriatrics, Southwest Hospital, The First Affiliate Hospital to Army Medical University, Chongqing, China
| | - Xiaomin Song
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Ran Gao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Ying Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Physiology, Peking Union Medical College, Beijing, China
| | - Wenjun Guo
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Bolun Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China
| | - Hongmei Zhao
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China.
| | - Jing Wang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Institute of Basic Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, China.
| |
Collapse
|
7
|
Li Y, Zhu X, Liu X, Du A, Yu B. miR-200a mediates protection of thymosin β-4 in cardiac microvascular endothelial cells as a novel mechanism under hypoxia-reoxygenation injury. J Cell Biochem 2019; 120:19098-19106. [PMID: 31265170 DOI: 10.1002/jcb.29237] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Thymosin β-4 (Tβ4) is a ubiquitous protein, which has been suggested to regulate multiple cell signal pathways and a variety of cellular functions. However, the role Tβ4 plays in the cardiac microvascular endothelial cells (CMECs) under myocardial ischemia/reperfusion injury is currently unknown. Here we investigated the effects of Tβ4 on hypoxia/reoxygenation (H/R) induced CMECs injury and its potential molecular mechanism. Cultured CMECs were positively identified by flow cytometry using antibody against CD31 and VWF/Factor VIII, which are constitutively expressed on the surface of CMECs. Then the reduced level of Tβ4 was detected in H/R-CMECs by a real-time quantitative polymerase chain reaction. To determine the effects of Tβ4 on H/R-CMECs, we transfected the overexpression or silence vector of Tβ4 into CMECs under H/R condition. Our results indicated that H/R treatment could reduce proliferation, increased apoptosis, adhesion, and reactive oxygen species (ROS) production in CMECs, which were attenuated by Tβ4 overexpression or aggravated by Tβ4 silencing, implying Tβ4 is able to promote CMECs against H/R-induced cell injury. Furthermore, the microRNA-200a (miR-200a) level was also increased by Tβ4 in H/R-CMECs or reduced by Tβ4 small interfering RNA. To investigated the mechanism of protective effects of Tβ4 on CMECs injury, the miR-200a inhibitor was transfected into H/R-CMECs. The results indicated that inhibition of miR-200a inversed the protection of Tβ4 on H/R-CMECs, specifically including cell proliferation, cell adhesion, cell apoptosis, and ROS production, as well as nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation. In conclusion, our results determined that Tβ4 attenuated H/R-induced CMECs injury by miR-200a-Nrf2 signaling.
Collapse
Affiliation(s)
- Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiaolong Zhu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xiping Liu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Bo Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, PR China
| |
Collapse
|
8
|
Relationship of serum Wnt1-inducible signaling pathway protein 1 levels with coronary artery disease and its severity. Coron Artery Dis 2019; 30:542-546. [PMID: 30998609 DOI: 10.1097/mca.0000000000000745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the association of Wnt1-inducible signaling pathway protein 1 (WISP1) concentrations in circulation with the presence and severity of coronary artery disease (CAD). PARTICIPANTS AND METHODS A total of 120 consecutive participants who underwent coronary angiography between May 2017 and July 2018 at our center were enrolled. Participants were divided into two groups based on the presence of CAD. Serum WISP1 levels were measured using enzyme-linked immunosorbent assay. Univariate and multivariate analyses were used to determine the association between variables and the presence of CAD. RESULTS The average age of the study population was 59.8 years, 66.7% were male, and 58.3% were positive for CAD. Serum WISP1 levels were significantly higher in patients with CAD than non-CAD group (339.8 vs. 322.4 pg/ml, P = 0.012). Moreover, a stepwise increase in serum WISP1 levels was observed with the number of diseased vessels (zero-vessel, one-vessel, two-vessel, and three-vessel disease: 322.4, 324.7, 345.4, and 392.1 pg/ml, respectively, P < 0.001) or Gensini score (r = 0.376, P < 0.001). Importantly, serum WISP1 levels were positively associated with the presence of CAD ( β = 1.011, 95% confidence interval: 1.001-1.021, P = 0.026). This association persisted after adjusting for age, sex, hypertension, type 2 diabetes mellitus, hypercholesterolemia, smoking, and high-sensitivity C-reactive protein ( β = 1.011, 95% confidence interval: 1.000-1.021, P = 0.047). In addition, serum WISP1 concentrations were positively correlated with BMI (r = 0.212, P = 0.020), insulin (r = 0.237, P = 0.009), and homeostatic model assessment for insulin resistance (r = 0.223, P = 0.014). CONCLUSION We demonstrated for the first time that serum WISP1 concentration is associated with the presence and severity of CAD.
Collapse
|
9
|
Gupta S, Li L. The role of Thymosin β4 in angiotensin II-induced cardiomyocytes growth. Expert Opin Biol Ther 2019; 18:105-110. [PMID: 30063846 DOI: 10.1080/14712598.2018.1494718] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Thymosin beta-4 (Tβ4) is an actin sequestering protein and is furthermore involved in diverse biological processes including cell proliferation, differentiation, wound healing, stem- or progenitor cell differentiation, and modulates inflammatory mediators. Tβ4 also attenuates fibrosis. However, the role of Tβ4 in cardiomyocytes hypertrophy is unknown. AREAS COVERED In this review, we will discuss the role of Tβ4 in cardiac remodeling that specifically includes cardiac hypertrophy and fibrosis only. Our review will further cover a new signaling pathway, the wingless and integrated-1 (Wnt) pathway in cardiac remodeling. In rat neonatal and adult cardiomyocytes stimulated with angiotensin II (Ang II), we showed that Tβ4 has the ability to reduce cell sizes, attenuate hypertrophy marker genes expression, along with a panel of WNT-associated gene expressions induced by Ang II. Selected target gene WNT1-inducible-signaling pathway protein 1 (WISP-1) was identified by Tβ4. Data further confirmed that WISP-1 overexpression promoted cardiomyocytes growth and was reversed by Tβ4 pretreatment. EXPERT OPINION Our data suggested that Tβ4 protects cardiomyocytes from hypertrophic response by targeting WISP-1. The new role of Tβ4 in cardiac hypertrophy advances our understanding, and the mechanism of action of Tβ4 may provide a solid foundation for the treatment of cardiac disease.
Collapse
Affiliation(s)
- Sudhiranjan Gupta
- a Department of Medical Physiology , Texas A&M University; Central Texas Veterans Health Care System , Temple , TX , USA
| | - Li Li
- a Department of Medical Physiology , Texas A&M University; Central Texas Veterans Health Care System , Temple , TX , USA
| |
Collapse
|
10
|
Zhang S, Yuan Y, Zhang Z, Guo J, Li J, Zhao K, Qin Y, Qiu C. LncRNA FOXC2-AS1 protects cardiomyocytes from doxorubicin-induced cardiotoxicity through activation of WNT1-inducible signaling pathway protein-1. Biosci Biotechnol Biochem 2018; 83:653-658. [PMID: 30558506 DOI: 10.1080/09168451.2018.1553606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Doxorubicin (Dox) is an anthracycline antibiotic that has been used to treat different cancers. Dox-induced cardiotoxicity is common in clinical practice, while its mechanism is unknown. It has been proved that lncRNA FOXC2-AS1 may promote doxorubicin resistance and WNT1-inducible signaling pathway protein-1 (WISP1) blocks doxorubicin-induced cardiomyocyte death. Our study aimed to investigate the involvement of lncRNA FOXC2-AS1 and WISP1 in doxorubicin-induced cardiotoxicity and to explore their interactions. In our study we observed that FOXC2-AS1 and WISP1 mRNA were downregulated in heart tissues of mice with Dox-induced cardiotoxicity. FOXC2-AS1 and WISP1 mRNA expression were positively correlated in mice with Dox-induced cardiotoxicity but not in healthy mice. Overexpression of FOXC2-AS1 promoted to viability of mice cardiomyocytes under Dox treatment and also increased the expression level of WISP1. In contrast, WISP1 overexpression showed no significant effect on FOXC2-AS1. We therefore conclude that lncRNA FOXC2-AS1 may upregulate WISP1 to protect cardiomyocytes from doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Shenwei Zhang
- a Department of cardiology , the first affiliated hospital of Zhengzhou university , Zhengzhou City , Henan Province , PR. China
| | - Yiqiang Yuan
- b Department of cardiology , The seventh people'S hospital of Zhengzhou , Zhengzhou City , Henan Province , PR. China
| | - Zheng Zhang
- c Department of cardiology , The General Hospital of the PLA Rocket Force , Beijing City , PR. China
| | - Jing Guo
- b Department of cardiology , The seventh people'S hospital of Zhengzhou , Zhengzhou City , Henan Province , PR. China
| | - Jing Li
- b Department of cardiology , The seventh people'S hospital of Zhengzhou , Zhengzhou City , Henan Province , PR. China
| | - Kui Zhao
- b Department of cardiology , The seventh people'S hospital of Zhengzhou , Zhengzhou City , Henan Province , PR. China
| | - Yanping Qin
- b Department of cardiology , The seventh people'S hospital of Zhengzhou , Zhengzhou City , Henan Province , PR. China
| | - Chunguang Qiu
- a Department of cardiology , the first affiliated hospital of Zhengzhou university , Zhengzhou City , Henan Province , PR. China
| |
Collapse
|
11
|
Ziegler T, Hinkel R, Kupatt C. Induced pluripotent stem cell derived cardiac models: effects of Thymosin β4. Expert Opin Biol Ther 2018; 18:111-120. [PMID: 30063852 DOI: 10.1080/14712598.2018.1473370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION The establishment of induced pluripotent stem cells (iPSCs) and cardiomyocytes differentiated from them generated a new platform to study pathophysiological processes and to generate drug screening platforms and iPSC-derived tissues as therapeutic agents. Although major advances have been made in iPSC-reprogramming, cardiac differentiation and EHT production, reprogramming efficiency and the maturity of iPSC-CMs need to be further improved. AREAS COVERED In this review, the authors summarize the current state of the field of iPSC research, the methodology of cardiac differentiation of iPSCs, the use of iPSC-CMs as disease models and toxicity screening platforms, and the potential of EHTs as therapeutic agents. The authors furthermore highlight the mechanisms by which Thymosin β4 might enhance the production of iPSC-CMs and EHTs to improve their maturity and performance. EXPERT OPINION iPSCs derived cardiomyocytes and EHTs represent a still young research field with many problems and pitfalls that need to be resolved to realize the full potential of iPSC-CMs and EHTs. Given that Thymosin β4 directly enhances cardiac differentiation while also promoting angiogenic sprouting and vessel maturation, Tβ4 might be of particular interest as a novel agent in tackling the difficulty of iPSC-CMs and engineered heart tissue grafts.
Collapse
Affiliation(s)
- Tilman Ziegler
- a I. Medizinische Klinik & Poliklinik, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany.,b DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance , Munich , German
| | - Rabea Hinkel
- a I. Medizinische Klinik & Poliklinik, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany.,b DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance , Munich , German.,c Institute for Cardiovascular Prevention , Ludwig Maximilians University of Munich , Munich , Germany
| | - Christian Kupatt
- a I. Medizinische Klinik & Poliklinik, Klinikum rechts der Isar , Technical University of Munich , Munich , Germany.,b DZHK (German Center for Cardiovascular Research), partner site Munich Heart Alliance , Munich , German
| |
Collapse
|
12
|
An essential role for Wnt/β-catenin signaling in mediating hypertensive heart disease. Sci Rep 2018; 8:8996. [PMID: 29895976 PMCID: PMC5997634 DOI: 10.1038/s41598-018-27064-2] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/24/2018] [Indexed: 12/11/2022] Open
Abstract
Activation of the renin-angiotensin system (RAS) is associated with hypertension and heart disease. However, how RAS activation causes cardiac lesions remains elusive. Here we report the involvement of Wnt/β-catenin signaling in this process. In rats with chronic infusion of angiotensin II (Ang II), eight Wnt ligands were induced and β-catenin activated in both cardiomyocytes and cardiac fibroblasts. Blockade of Wnt/β-catenin signaling by small molecule inhibitor ICG-001 restrained Ang II-induced cardiac hypertrophy by normalizing heart size and inhibiting hypertrophic marker genes. ICG-001 also attenuated myocardial fibrosis and inhibited α-smooth muscle actin, fibronectin and collagen I expression. These changes were accompanied by a reduced expression of atrial natriuretic peptide and B-type natriuretic peptide. Interestingly, ICG-001 also lowered blood pressure induced by Ang II. In vitro, Ang II induced multiple Wnt ligands and activated β-catenin in rat primary cardiomyocytes and fibroblasts. ICG-001 inhibited myocyte hypertrophy and Snail1, c-Myc and atrial natriuretic peptide expression, and abolished the fibrogenic effect of Ang II in cardiac fibroblasts. Finally, recombinant Wnt3a was sufficient to induce cardiomyocyte injury and fibroblast activation in vitro. Taken together, these results illustrate an essential role for Wnt/β-catenin in mediating hypertension, cardiac hypertrophy and myocardial fibrosis. Therefore, blockade of this pathway may be a novel strategy for ameliorating hypertensive heart disease.
Collapse
|
13
|
Drum CL, Tan WKY, Chan SP, Pakkiri LS, Chong JPC, Liew OW, Ng TP, Ling LH, Sim D, Leong KTG, Yeo DPS, Ong HY, Jaufeerally F, Wong RCC, Chai P, Low AF, Davidsson P, Liljeblad M, Söderling AS, Gan LM, Bhat RV, Purnamawati K, Lam CSP, Richards AM. Thymosin Beta-4 Is Elevated in Women With Heart Failure With Preserved Ejection Fraction. J Am Heart Assoc 2017; 6:JAHA.117.005586. [PMID: 28611096 PMCID: PMC5669175 DOI: 10.1161/jaha.117.005586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Thymosin beta-4 (TB4) is an X-linked gene product with cardioprotective properties. Little is known about plasma concentration of TB4 in heart failure (HF), and its relationship with other cardiovascular biomarkers. We sought to evaluate circulating TB4 in HF patients with preserved (HFpEF) or reduced (HFrEF) ejection fraction compared to non-HF controls. METHODS AND RESULTS TB4 was measured using a liquid chromatography and mass spectrometry assay in age- and sex-matched HFpEF (n=219), HFrEF (n=219) patients, and controls (n=219) from a prospective nationwide study. Additionally, a 92-marker multiplex proximity extension assay was measured to identify biomarker covariates. Compared with controls, plasma TB4 was elevated in HFpEF (985 [421-1723] ng/mL versus 1401 [720-2379] ng/mL, P<0.001), but not in HFrEF (1106 [556-1955] ng/mL, P=0.642). Stratifying by sex, only women (1623 [1040-2625] ng/mL versus 942 [386-1891] ng/mL, P<0.001), but not men (1238.5 [586-1967] ng/mL versus 1004 [451-1538] ng/mL, P=1.0), had significantly elevated TB4 in the setting of HFpEF. Adjusted for New York Heart Association class, N-terminal pro B-type natriuretic peptide, age, and myocardial infarction, hazard ratio to all-cause mortality is significantly higher in women with elevated TB4 (1.668, P=0.036), but not in men (0.791, P=0.456) with HF. TB4 is strongly correlated with a cluster of 7 markers from the proximity extension assay panel, which are either X-linked, regulated by sex hormones, or involved with NF-κB signaling. CONCLUSIONS We show that plasma TB4 is elevated in women with HFpEF and has prognostic information. Because TB4 can preserve EF in animal studies of cardiac injury, the relation of endogenous, circulating TB4 to X chromosome biology and differential outcomes in female heart disease warrants further study.
Collapse
Affiliation(s)
- Chester L Drum
- Cardiovascular Research Institute, National University Health System, Singapore .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Warren K Y Tan
- Cardiovascular Research Institute, National University Health System, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore
| | - Siew-Pang Chan
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Mathematics & Statistics, College of Science, Health & Engineering, La Trobe University, Melbourne, Australia
| | | | - Jenny P C Chong
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Oi-Wah Liew
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Tze-Pin Ng
- Cardiovascular Research Institute, National University Health System, Singapore.,Department of Psychological Medicine, National University of Singapore, Singapore
| | - Lieng-Hsi Ling
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,National University Heart Centre Singapore, Singapore
| | - David Sim
- National Heart Centre Singapore, Singapore.,Duke-NUS Medical School, Singapore
| | | | | | - Hean-Yee Ong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Cardiology, Khoo Teck Puat Hospital, Singapore
| | - Fazlur Jaufeerally
- Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | | | - Ping Chai
- National University Heart Centre Singapore, Singapore
| | - Adrian F Low
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,National University Heart Centre Singapore, Singapore
| | - Pia Davidsson
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Mathias Liljeblad
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Ann-Sofi Söderling
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Li-Ming Gan
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Ratan V Bhat
- Innovative Medicines & Early Development, Cardiovascular & Metabolic Diseases iMed, AstraZeneca R&D, Gothenburg, Sweden
| | - Kristy Purnamawati
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore
| | - Carolyn S P Lam
- National Heart Centre Singapore, Singapore.,Duke-NUS Medical School, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, National University Health System, Singapore .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Christchurch Heart Institute, University of Otago, New Zealand
| |
Collapse
|