1
|
Dubey N, Verma A, Goyal A, Vishwakarma V, Bhatiya J, Arya DS, Yadav HN. The role of endothelin and its receptors in cardiomyopathy: From molecular mechanisms to therapeutic insights. Pathol Res Pract 2025; 269:155932. [PMID: 40174273 DOI: 10.1016/j.prp.2025.155932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Cardiomyopathy is an anatomical and pathologic condition that is related to the cardiac muscle or left ventricular failure. A diverse range of illnesses known as cardiomyopathies often result in progressive heart failure with high morbidity and death rates. Primary cardiomyopathies are hereditary, mixed, or adopted. Secondary cardiomyopathies are infiltrative, harmful, or pathogenic. The activation of many paracrine, autocrine, and neuroendocrine factors is closely linked to pathological left ventricular (LV) deformation. After the myocardial injury, in the context of higher LV wall pressure and haemodynamic disturbance, these variables are raised. New therapy techniques have been focused on these novel targets after recent studies revealed that endothelin, nitric oxide or cytokines may be implicated in the remodelling process. Vasoconstrictive peptide endothelin-1 (ET-1) is mostly generated in the endothelium and works by binding to the ETA- and ETB-endothelin receptors (ET-Rs). The expression of both ET-Rs is widespread in cardiac tissues. Heart failure, pulmonary arterial hypertension, hypertension, cardiomyopathy, and coronary artery disease are just a few of the cardiovascular disorders for which the endothelin system has been shown to play a crucial role over the years. The occurrence, pathogenesis, and natural history of endothelin antagonists in cardiomyopathies are currently not well understood, and specific aspects of their treatment responses have not received comprehensive attention. Therefore, in this study, we address the variable degrees of success that have been achieved in treating cardiomyopathy using endothelin-targeting treatments, such as endothelin receptor antagonists.
Collapse
Affiliation(s)
- Nandini Dubey
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Aanchal Verma
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Vishal Vishwakarma
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Jagriti Bhatiya
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
2
|
Tangmahakul N, Rungsipipat A, Surachetpong SD. Investigation of pulmonary artery and circulating endothelin-1 expression in dogs with pulmonary hypertension secondary to myxomatous mitral valve disease. Vet World 2024; 17:2144-2151. [PMID: 39507783 PMCID: PMC11536742 DOI: 10.14202/vetworld.2024.2144-2151] [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: 05/15/2024] [Accepted: 08/26/2024] [Indexed: 11/08/2024] Open
Abstract
Background and Aim Pulmonary hypertension (PH) is a condition characterized by abnormally elevated pressure in the pulmonary vasculature. It is a common complication of myxomatous mitral valve disease (MMVD) in dogs. Several vasoactive substances, including endothelin-1 (ET-1), have been suggested to contribute to pathological changes in the pulmonary arteries of patients with PH. This study aimed to examine the local and systemic expression of ET-1 in dogs with PH secondary to MMVD. Materials and Methods Lung tissues were collected from 20 client-owned dogs during the first stage of the study and divided into three groups: normal dogs (n = 5), MMVD dogs (n = 8), and MMVD+PH dogs (n = 7). The expression of ET-1 and endothelin A receptor (ETAR) in the pulmonary arteries was determined using immunohistochemistry. Blood samples were collected from 61 client-owned dogs for the second stage of the study and divided into three groups: normal (n = 22), MMVD (n = 20), and MMVD+PH (n = 19). Plasma ET-1 concentration was measured using a sandwich enzyme-linked immunosorbent assay. Results There was no difference in ET-1 and ETAR expression in the pulmonary arteries among the three groups. Similarly, there was no difference in the plasma ET-1 concentration between the groups. In addition, no correlation was found between the immunohistochemical expression of ET-1 and ETAR and the thickness of the pulmonary arteries or between the plasma ET-1 level and echocardiographic variables. Conclusion The lack of difference in the expression of ET-1 and ETAR in the pulmonary arteries and in the circulating ET-1 concentration among the studied groups suggests that ET-1 may not be related to the pathological development of PH secondary to MMVD in dogs. Due to the small sample size in this study, further research is needed to confirm these findings.
Collapse
Affiliation(s)
- Nattawan Tangmahakul
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Henri-Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Anudep Rungsipipat
- Center of Excellence for Companion Animal Cancer (CE-CAC), Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Henri-Dunant Road, Pathumwan, Bangkok 10330, Thailand
| | - Sirilak Disatian Surachetpong
- Department of Veterinary Medicine, Faculty of Veterinary Science, Chulalongkorn University, Henri-Dunant Road, Pathumwan, Bangkok 10330, Thailand
| |
Collapse
|
3
|
Shi X, Li P, Herb M, Liu H, Wang M, Wang X, Feng Y, van Beers T, Xia N, Li H, Prokosch V. Pathological high intraocular pressure induces glial cell reactive proliferation contributing to neuroinflammation of the blood-retinal barrier via the NOX2/ET-1 axis-controlled ERK1/2 pathway. J Neuroinflammation 2024; 21:105. [PMID: 38649885 PMCID: PMC11034147 DOI: 10.1186/s12974-024-03075-x] [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: 01/02/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND NADPH oxidase (NOX), a primary source of endothelial reactive oxygen species (ROS), is considered a key event in disrupting the integrity of the blood-retinal barrier. Abnormalities in neurovascular-coupled immune signaling herald the loss of ganglion cells in glaucoma. Persistent microglia-driven inflammation and cellular innate immune system dysregulation often lead to deteriorating retinal degeneration. However, the crosstalk between NOX and the retinal immune environment remains unresolved. Here, we investigate the interaction between oxidative stress and neuroinflammation in glaucoma by genetic defects of NOX2 or its regulation via gp91ds-tat. METHODS Ex vivo cultures of retinal explants from wildtype C57BL/6J and Nox2 -/- mice were subjected to normal and high hydrostatic pressure (Pressure 60 mmHg) for 24 h. In vivo, high intraocular pressure (H-IOP) was induced in C57BL/6J mice for two weeks. Both Pressure 60 mmHg retinas and H-IOP mice were treated with either gp91ds-tat (a NOX2-specific inhibitor). Proteomic analysis was performed on control, H-IOP, and treatment with gp91ds-tat retinas to identify differentially expressed proteins (DEPs). The study also evaluated various glaucoma phenotypes, including IOP, retinal ganglion cell (RGC) functionality, and optic nerve (ON) degeneration. The superoxide (O2-) levels assay, blood-retinal barrier degradation, gliosis, neuroinflammation, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative PCR were performed in this study. RESULTS We found that NOX2-specific deletion or activity inhibition effectively attenuated retinal oxidative stress, immune dysregulation, the internal blood-retinal barrier (iBRB) injury, neurovascular unit (NVU) dysfunction, RGC loss, and ON axonal degeneration following H-IOP. Mechanistically, we unveiled for the first time that NOX2-dependent ROS-driven pro-inflammatory signaling, where NOX2/ROS induces endothelium-derived endothelin-1 (ET-1) overexpression, which activates the ERK1/2 signaling pathway and mediates the shift of microglia activation to a pro-inflammatory M1 phenotype, thereby triggering a neuroinflammatory outburst. CONCLUSIONS Collectively, we demonstrate for the first time that NOX2 deletion or gp91ds-tat inhibition attenuates iBRB injury and NVU dysfunction to rescue glaucomatous RGC loss and ON axon degeneration, which is associated with inhibition of the ET-1/ERK1/2-transduced shift of microglial cell activation toward a pro-inflammatory M1 phenotype, highlighting NOX2 as a potential target for novel neuroprotective therapies in glaucoma management.
Collapse
Affiliation(s)
- Xin Shi
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Panpan Li
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital of Cologne, University of Cologne, Goldenfelsstr. 19-21, 50935, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Maoren Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Xiaosha Wang
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Yuan Feng
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Tim van Beers
- Institut I für Anatomie, Universitätsklinikum Köln (AöR), Cologne, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131, Mainz, Germany
| | - Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany.
| |
Collapse
|
4
|
Zou R, Shi W, Ceylan AF, Dong M, Zhang M, Zou Z, Peng B, Dong F, Turdi S, Lin J, Zhang Y, Wang G, Fan X, Ren J. Cardiomyocyte-specific deletion of endothelin receptor A (ET A) obliterates cardiac aging through regulation of mitophagy and ferroptosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166958. [PMID: 37963542 DOI: 10.1016/j.bbadis.2023.166958] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
Advanced aging evokes unfavorable changes in the heart including cardiac remodeling and contractile dysfunction although the underlying mechanism remains elusive. This study was conducted to evaluate the role of endothelin-1 (ET-1) in the pathogenesis of cardiac aging and mechanism involved. Echocardiographic and cardiomyocyte mechanical properties were determined in young (5-6 mo) and aged (26-28 mo) wild-type (WT) and cardiomyocyte-specific ETA receptor knockout (ETAKO) mice. GSEA enrichment identified differentially expressed genes associated with mitochondrial respiration, mitochondrial protein processing and mitochondrial depolarization in cardiac aging. Aging elevated plasma levels of ET-1, Ang II and suppressed serum Fe2+, evoked cardiac remodeling (hypertrophy and interstitial fibrosis), contractile defects (fractional shortening, ejection fraction, cardiomyocyte peak shortening, maximal velocity of shortening/relengthening and prolonged relengthening) and intracellular Ca2+ mishandling (dampened intracellular Ca2+ release and prolonged decay), the effects with the exception of plasma AngII, ET-1 and Fe2+ were mitigated by ETAKO. Advanced age facilitated O2- production, carbonyl protein damage, cardiac hypertrophy (GATA4, ANP, NFATc3), ER stress, ferroptosis, compromised autophagy (LC3B, Beclin-1, Atg7, Atg5 and p62) and mitophagy (parkin and FUNDC1), and deranged intracellular Ca2+ proteins (SERCA2a and phospholamban), the effects of which were reversed by ETA ablation. ET-1 provoked ferroptosis in vitro, the response was nullified by the ETA receptor antagonist BQ123 and mitophagy inducer CsA. ETA but not ETB receptor antagonism reconciled cardiac aging, which was abrogated by inhibition of mitophagy and ferroptosis. These findings collectively denote promises of targeting ETA, mitophagy and ferroptosis in the management of aging-associated cardiac remodeling and contractile defect.
Collapse
Affiliation(s)
- Rongjun Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China
| | - Wanting Shi
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, Guangdong, China
| | - Asli F Ceylan
- Ankara Yildirim Beyazit University, Faculty of Medicine, Department of Medical Pharmacology, Bilkent, Ankara, Turkey
| | - Maolong Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Miao Zhang
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Zengxiao Zou
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Bo Peng
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA
| | - Subat Turdi
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Jie Lin
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Ge Wang
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China.
| | - Xiaoping Fan
- Department of Cardiovascular Surgery, Guangdong Provincial Hospital of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong, China; The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou 510120, Guangdong, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
5
|
Docosahexaenoic Acid Attenuates Radiation-Induced Myocardial Fibrosis by Inhibiting the p38/ET-1 Pathway in Cardiomyocytes. Int J Radiat Oncol Biol Phys 2023; 115:1229-1243. [PMID: 36529557 DOI: 10.1016/j.ijrobp.2022.11.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/24/2022] [Accepted: 11/01/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE Radiation-induced myocardial fibrosis (RIMF) is a severe delayed complication of thoracic irradiation (IR). Endothelin-1 (ET-1) is critical in cardiac fibroblast activation, and docosahexaenoic acid (DHA) is protective against various cardiac diseases. This study aimed to explore the roles of ET-1 in RIMF and the potential of DHA in preventing RIMF. METHODS AND MATERIALS Hematoxylin and eosin, sirius red, and Masson trichrome staining were carried out to evaluate the histopathologic conditions in mouse models. Enzyme-linked immunosorbent assays were used to detect the concentration of ET-1 in serum and cell supernatants. Western blotting, immunofluorescence, and immunohistochemistry were used to assess the protein levels. The phenotypic alterations of cardiac fibroblasts were evaluated by cell proliferation/migration assays and α-smooth muscle actin (α-SMA) detection. RESULTS Radiation increased ET-1 expression and secretion by increasing p38 phosphorylation in cardiomyocytes, and ET-1 markedly promoted the activation of cardiac fibroblasts, which were characterized by enhanced fibroblast proliferation, migration, and α-SMA expression. Cardiomyocyte-derived ET-1 mediated radiation-induced fibroblast activation by targeting the PI3K-AKT and MEK-ERK pathways in fibroblasts. DHA suppressed ET-1 levels by blocking p38 signaling in cardiomyocytes and significantly attenuated the activation of cardiac fibroblasts induced by the IR/ET-1 axis. Importantly, DHA decreased collagen deposition and α-SMA expression, alleviating cardiac fibrosis caused by radiation in mouse models. CONCLUSIONS Our findings demonstrate that radiation facilitates cardiac fibroblast activation by enhancing p38/ET-1 signaling in cardiomyocytes, revealing the IR/p38/ET-1 regulatory axis in RIMF for the first time. DHA effectively inhibits fibroblast activation by targeting p38/ET-1 and can be recognized as a promising protective agent against RIMF.
Collapse
|
6
|
Anti-inflammatory effects of endothelin receptor blockade in left atrial tissue of spontaneously hypertensive rats. IJC HEART & VASCULATURE 2022; 42:101088. [PMID: 35879971 PMCID: PMC9307454 DOI: 10.1016/j.ijcha.2022.101088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022]
|
7
|
Zheng SL, Luo QB, Suo SK, Zhao YQ, Chi CF, Wang B. Preparation, Identification, Molecular Docking Study and Protective Function on HUVECs of Novel ACE Inhibitory Peptides from Protein Hydrolysate of Skipjack Tuna Muscle. Mar Drugs 2022; 20:md20030176. [PMID: 35323475 PMCID: PMC8954214 DOI: 10.3390/md20030176] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 02/25/2022] [Indexed: 02/07/2023] Open
Abstract
To prepare bioactive peptides with high angiotensin-I-converting enzyme (ACE)-inhibitory (ACEi) activity, Alcalase was selected from five kinds of protease for hydrolyzing Skipjack tuna (Katsuwonus pelamis) muscle, and its best hydrolysis conditions were optimized using single factor and response surface experiments. Then, the high ACEi protein hydrolysate (TMPH) of skipjack tuna muscle was prepared using Alcalase under the optimum conditions of enzyme dose 2.3%, enzymolysis temperature 56.2 °C, and pH 9.4, and its ACEi activity reached 72.71% at 1.0 mg/mL. Subsequently, six novel ACEi peptides were prepared from TMPH using ultrafiltration and chromatography methods and were identified as Ser-Pro (SP), Val-Asp-Arg-Tyr-Phe (VDRYF), Val-His-Gly-Val-Val (VHGVV), Tyr-Glu (YE), Phe-Glu-Met (FEM), and Phe-Trp-Arg-Val (FWRV), with molecular weights of 202.3, 698.9, 509.7, 310.4, 425.6, and 606.8 Da, respectively. SP and VDRYF displayed noticeable ACEi activity, with IC50 values of 0.06 ± 0.01 and 0.28 ± 0.03 mg/mL, respectively. Molecular docking analysis illustrated that the high ACEi activity of SP and VDRYF was attributed to effective interaction with the active sites/pockets of ACE by hydrogen bonding, electrostatic force, and hydrophobic interaction. Furthermore, SP and VDRYF could significantly up-regulate nitric oxide (NO) production and down-regulate endothelin-1 (ET-1) secretion in HUVECs after 24 h treatment, but also abolish the negative effect of 0.5 μM norepinephrine (NE) on the generation of NO and ET-1. Therefore, ACEi peptides derived from skipjack tuna (K. pelamis) muscle, especially SP and VDRYF, are beneficial components for functional food against hypertension and cardiovascular diseases.
Collapse
Affiliation(s)
- Shuo-Lei Zheng
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (S.-L.Z.); (S.-K.S.); (Y.-Q.Z.)
| | - Qian-Bin Luo
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, School of Marine Science and Technology, Zhejiang Ocean University, Zhoushan 316022, China;
| | - Shi-Kun Suo
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (S.-L.Z.); (S.-K.S.); (Y.-Q.Z.)
| | - Yu-Qin Zhao
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (S.-L.Z.); (S.-K.S.); (Y.-Q.Z.)
| | - Chang-Feng Chi
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, School of Marine Science and Technology, Zhejiang Ocean University, Zhoushan 316022, China;
- Correspondence: (C.-F.C.); (B.W.); Tel./Fax: +86-580-255-4818 (C.-F.C.); +86-580-255-4781 (B.W.)
| | - Bin Wang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China; (S.-L.Z.); (S.-K.S.); (Y.-Q.Z.)
- Correspondence: (C.-F.C.); (B.W.); Tel./Fax: +86-580-255-4818 (C.-F.C.); +86-580-255-4781 (B.W.)
| |
Collapse
|
8
|
Bourque K, Hawey C, Jones-Tabah J, Pétrin D, Martin RD, Ling Sun Y, Hébert TE. Measuring hypertrophy in neonatal rat primary cardiomyocytes and human iPSC-derived cardiomyocytes. Methods 2021; 203:447-464. [PMID: 34933120 DOI: 10.1016/j.ymeth.2021.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 12/14/2022] Open
Abstract
In the heart, left ventricular hypertrophy is initially an adaptive mechanism that increases wall thickness to preserve normal cardiac output and function in the face of coronary artery disease or hypertension. Cardiac hypertrophy develops in response to pressure and volume overload but can also be seen in inherited cardiomyopathies. As the wall thickens, it becomes stiffer impairing the distribution of oxygenated blood to the rest of the body. With complex cellular signalling and transcriptional networks involved in the establishment of the hypertrophic state, several model systems have been developed to better understand the molecular drivers of disease. Immortalized cardiomyocyte cell lines, primary rodent and larger animal models have all helped understand the pathological mechanisms underlying cardiac hypertrophy. Induced pluripotent stem cell-derived cardiomyocytes are also used and have the additional benefit of providing access to human samples with direct disease relevance as when generated from patients suffering from hypertrophic cardiomyopathies. Here, we briefly review in vitro and in vivo model systems that have been used to model hypertrophy and provide detailed methods to isolate primary neonatal rat cardiomyocytes as well as to generate cardiomyocytes from human iPSCs. We also describe how to model hypertrophy in a "dish" using gene expression analysis and immunofluorescence combined with automated high-content imaging.
Collapse
Affiliation(s)
- Kyla Bourque
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Cara Hawey
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jace Jones-Tabah
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Ryan D Martin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Yi Ling Sun
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
9
|
Han Z, Bi S, Xu Y, Dong X, Mei L, Lin H, Li X. Cholecystokinin Expression in the Development of Myocardial Hypertrophy. SCANNING 2021; 2021:8231559. [PMID: 34497680 PMCID: PMC8405328 DOI: 10.1155/2021/8231559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/06/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Expression of cholecystokinin is found in myocardial tissues as a gastrointestinal hormone and may be involved in cardiovascular regulation. However, it is unclear whether there is an increase in cholecystokinin expression in myocardial hypertrophy progression induced by abdominal aortic constriction. The study is aimed at exploring the relationship between cholecystokinin expression and myocardial hypertrophy. METHODS We randomly divided the 70 Sprague-Dawley rats into two groups: the sham operation group and the abdominal aortic constriction group. The hearts of rats were measured by echocardiography, and myocardial tissues and blood were collected at 4 weeks, 8 weeks, and 12 weeks after surgery. Morphological changes were assessed by microscopy. The cholecystokinin expression was evaluated by immunochemistry, Western blotting, quantitative real-time polymerase chain reaction, and enzyme-linked immunosorbent assay. RESULTS The relative protein levels of cholecystokinin were significantly increased in the abdominal aortic constriction groups compared with the corresponding sham operation groups at 8 weeks and 12 weeks. The cholecystokinin mRNA in the abdominal aortic constriction groups was significantly higher than the time-matched sham operation groups. Changes in the left ventricular wall thickness were positively correlated with the relative protein levels of cholecystokinin and the mRNA of cholecystokinin. CONCLUSIONS The development of myocardial hypertrophy can affect the cholecystokinin expression of myocardial tissues.
Collapse
Affiliation(s)
- Zhongshu Han
- Department of Cardiology, Harbin Medical University Fourth Hospital, Harbin 150086, China
| | - Sheng Bi
- Department of Critical Care Medicine, Affiliated Qiqihar Hospital, Southern Medical University (The First Hospital of Qiqihar), Qiqihar 161005, China
| | - Yongsheng Xu
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Xiaoying Dong
- Department of Cardiology, Harbin Medical University Fourth Hospital, Harbin 150086, China
| | - Lixia Mei
- Department of Ultrasonic Medicine, Affiliated Qiqihar Hospital, Southern Medical University (The First Hospital of Qiqihar), Qiqihar 161005, China
| | - Hailong Lin
- Department of Cardiology, Central Hospital of Dalian, Dalian 116003, China
| | - Xueqi Li
- Department of Cardiology, Harbin Medical University Fourth Hospital, Harbin 150086, China
| |
Collapse
|
10
|
Duan Y, Qi D, Liu Y, Song Y, Wang X, Jiao S, Li H, Gonzalez FJ, Qi Y, Xu Q, Du J, Qu A. Deficiency of peroxisome proliferator-activated receptor α attenuates apoptosis and promotes migration of vascular smooth muscle cells. Biochem Biophys Rep 2021; 27:101091. [PMID: 34381883 PMCID: PMC8339143 DOI: 10.1016/j.bbrep.2021.101091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR) α is widely expressed in the vasculature and has pleiotropic and lipid-lowering independent effects, but its role in the growth and function of vascular smooth muscle cells (VSMCs) during vascular pathophysiology is still unclear. Herein, VSMC-specific PPARα-deficient mice (Ppara ΔSMC) were generated by Cre-LoxP site-specific recombinase technology and VSMCs were isolated from mice aorta. PPARα deficiency attenuated VSMC apoptosis induced by angiotensin (Ang) II and hydrogen peroxide, and increased the migration of Ang II-challenged cells.
Collapse
Key Words
- Ang II, angiotensin II
- Angiotensin II
- EC, endothelial cell
- ECM, extracellular matrix
- ERK, extracellular signal-regulated kinase
- MAPK, mitogen-activated protein kinase
- MCP-1, monocyte chemoattractant protein-1
- PCR, polymerase chain reaction
- PPAR, peroxisome proliferator-activated receptor
- PPARα
- SM22α, smooth muscle 22α
- TGF, tumor growth factor
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- VSMC, vascular smooth muscle cell
- Vascular remodeling
- Vascular smooth muscle cell
Collapse
Affiliation(s)
- Yan Duan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Dan Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Ye Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Yanting Song
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Xia Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Shiyu Jiao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| | - Huihua Li
- Department of Nutrition and Food Hygiene, School of Public Health, Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yongfen Qi
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Qingbo Xu
- School of Cardiovascular Medicine and Sciences, King' s College of London, London, UK
| | - Jie Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China.,Beijing Anzhen Hospital of Capital Medical University and Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Aijuan Qu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University; Key Laboratory of Remodeling-Related Cardiovascular Diseases, Ministry of Education; Beijing, China
| |
Collapse
|
11
|
Rajendran R, Sharma VK, Nandeesha H, Ananthakrishnan R, Vinod KV, Subramanian SK. Assessment of vascular function in complete glycaemic spectrum. Clin Exp Hypertens 2021; 43:436-442. [PMID: 33703968 DOI: 10.1080/10641963.2021.1896729] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Purpose: The present study was conceived to delineate the point of vascular dysfunction along the glycemic spectrum (normoglycemic individuals with no family history of diabetes, normoglycemic individuals with family history of diabetes, prediabetic individuals, and diabetic individuals).Materials and Methods: In this cross-sectional comparative study, we enrolled 252 participants of both gender in the age group of 30-50 years. They were classified based on their family history of diabetes and glycemic status into four groups along the glycemic spectrum as mentioned above. We measured flow-mediated dilation (FMD) from brachial artery and vascular function biomarkers such as enthothelin-1 (ET-1), von Willbrand Factor (vWF), Vascular Endothelial Growth Factor (VEGF) to assess the vascular function. The comparison of data between groups were done using One Way ANOVA/Kruskal-Wallis followed by post-hoc analysis using LSD/Mann-Whitney U Test depending on the normality of the data. Spearman correlation was done between vascular function and plasma glucose levels to identify its relationship. Linear regression was carried out to identify the factors influencing the FMD across the glycemic spectrum.Results: We observed that vascular function negatively correlated with blood glucose levels. However, endothelin-1 and vWF derangement was there even in normoglycemic first degree relatives of diabetes (FDRD) and the derangement increased in prediabetes and diabetes. Physiological dysfunction in terms of decreased flow-mediated dilation starts from prediabetes only. VEGF derangement is found only in diabetic individuals.Conclusion: Vascular dysfunction is found even in normoglycemic FDRD and the derangement increased and compounded with the advancement of disease.
Collapse
Affiliation(s)
- Rajathi Rajendran
- Tutor, Department of Physiology, AIIMS, Mangalagiri, Andhra Pradesh, India
| | - Vivek Kumar Sharma
- Department of Physiology, All India Institute of Medical Sciences, Rajkot, India
| | - Hanumanthappa Nandeesha
- Department of Biochemistry, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Ramesh Ananthakrishnan
- Department Department of Radiology, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | - Kolar Vishwanath Vinod
- Department of Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research, Pondicherry, India
| | | |
Collapse
|
12
|
Cluntun AA, Badolia R, Lettlova S, Parnell KM, Shankar TS, Diakos NA, Olson KA, Taleb I, Tatum SM, Berg JA, Cunningham CN, Van Ry T, Bott AJ, Krokidi AT, Fogarty S, Skedros S, Swiatek WI, Yu X, Luo B, Merx S, Navankasattusas S, Cox JE, Ducker GS, Holland WL, McKellar SH, Rutter J, Drakos SG. The pyruvate-lactate axis modulates cardiac hypertrophy and heart failure. Cell Metab 2021; 33:629-648.e10. [PMID: 33333007 PMCID: PMC7933116 DOI: 10.1016/j.cmet.2020.12.003] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 10/12/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022]
Abstract
The metabolic rewiring of cardiomyocytes is a widely accepted hallmark of heart failure (HF). These metabolic changes include a decrease in mitochondrial pyruvate oxidation and an increased export of lactate. We identify the mitochondrial pyruvate carrier (MPC) and the cellular lactate exporter monocarboxylate transporter 4 (MCT4) as pivotal nodes in this metabolic axis. We observed that cardiac assist device-induced myocardial recovery in chronic HF patients was coincident with increased myocardial expression of the MPC. Moreover, the genetic ablation of the MPC in cultured cardiomyocytes and in adult murine hearts was sufficient to induce hypertrophy and HF. Conversely, MPC overexpression attenuated drug-induced hypertrophy in a cell-autonomous manner. We also introduced a novel, highly potent MCT4 inhibitor that mitigated hypertrophy in cultured cardiomyocytes and in mice. Together, we find that alteration of the pyruvate-lactate axis is a fundamental and early feature of cardiac hypertrophy and failure.
Collapse
Affiliation(s)
- Ahmad A Cluntun
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Rachit Badolia
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sandra Lettlova
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - K Mark Parnell
- Vettore Biosciences, 1700 Owens Street Suite 515, San Francisco, CA 94158, USA
| | - Thirupura S Shankar
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Nikolaos A Diakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Kristofor A Olson
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Iosif Taleb
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sean M Tatum
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Jordan A Berg
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Corey N Cunningham
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Tyler Van Ry
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Metabolomics, Proteomics and Mass Spectrometry Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Alex J Bott
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Aspasia Thodou Krokidi
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Sarah Fogarty
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Sophia Skedros
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Wojciech I Swiatek
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - Xuejing Yu
- University of Utah, School of Medicine, Salt Lake City, UT 84132, USA; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, UT 84132, USA
| | - Bai Luo
- Drug Discovery Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Shannon Merx
- Vettore Biosciences, 1700 Owens Street Suite 515, San Francisco, CA 94158, USA
| | - Sutip Navankasattusas
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Metabolomics, Proteomics and Mass Spectrometry Core Facility, University of Utah, Salt Lake City, UT 84112, USA
| | - Gregory S Ducker
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT 84112, USA
| | - Stephen H McKellar
- University of Utah, School of Medicine, Salt Lake City, UT 84132, USA; Division of Cardiothoracic Surgery, Department of Surgery, Salt Lake City, UT 84132, USA; U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake VA (Veterans Affairs) Health Care System, Salt Lake City, UT, USA
| | - Jared Rutter
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84132, USA; Howard Hughes Medical Institute, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | - Stavros G Drakos
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, UT 84112, USA; U.T.A.H. (Utah Transplant Affiliated Hospitals) Cardiac Transplant Program: University of Utah Healthcare and School of Medicine, Intermountain Medical Center, Salt Lake VA (Veterans Affairs) Health Care System, Salt Lake City, UT, USA.
| |
Collapse
|
13
|
Nomura S, Komuro I. Precision medicine for heart failure based on molecular mechanisms: The 2019 ISHR Research Achievement Award Lecture. J Mol Cell Cardiol 2021; 152:29-39. [PMID: 33275937 DOI: 10.1016/j.yjmcc.2020.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Heart failure is a leading cause of death, and the number of patients with heart failure continues to increase worldwide. To realize precision medicine for heart failure, its underlying molecular mechanisms must be elucidated. In this review summarizing the "The Research Achievement Award Lecture" of the 2019 XXIII ISHR World Congress held in Beijing, China, we would like to introduce our approaches for investigating the molecular mechanisms of cardiac hypertrophy, development, and failure, as well as discuss future perspectives.
Collapse
Affiliation(s)
- Seitaro Nomura
- Department of Cardiovascular Medicine, The University of Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo, Japan.
| |
Collapse
|
14
|
Corrêa JWDN, Prado CM, Riul ME, Araújo AV, Rossi MA, Bendhack LM. Reversion of cardiovascular remodelling in renovascular hypertensive 2K-1C rats by renin-angiotensin system inhibitors. Clin Exp Pharmacol Physiol 2020; 47:1965-1977. [PMID: 32688435 DOI: 10.1111/1440-1681.13384] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/13/2020] [Accepted: 07/14/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVES Evaluate whether the RAS dual blockade would induce additional beneficial effects on cardiovascular remodelling when compared to monotherapy in renal hypertensive two kidneys-one clip (2K-1C) rats. METHODS Hypertensive 2K-1C and normotensive (2K) rats were treated for 14 days with submaximal doses of losartan (LOS), enalapril (ENA), losartan plus enalapril (LOS + ENA) or vehicle (water). Blood pressure and some parameters of cardiovascular remodelling were evaluated. RESULTS Systolic blood pressure (SBP) was higher in 2K-1C (209 ± 3 mm Hg, P < .05) than in 2K (113 ± 1 mm Hg) rats. There was an additional effect in 2K-1C treated with LOS + ENA (153 ± 9 mm Hg) on lowering SBP when compared to LOS (184 ± 12 mm Hg) or ENA (177 ± 9 mm Hg). None of the treatments had effect on SBP in 2K rats. In 2K-1C, cardiomyocyte hypertrophy was reduced by all treatments, although the cardiac hypertrophy indexes remained unchanged. 2K-1C aortas presented medial thickening that was partially reduced by the treatments. Intimal hyperplasia observed in 2K-1C (15.56 ± 0.89 µm vs 8.24 ± 0.80 µm) was reversed by ENA (9.52 ± 0.45 µm) or LOS + ENA (8.17 ± 0.53 µm). Collagen deposition was increased in 2K-1C hearts (1.77 ± 0.16 vs 1.28 ± 0.09) and aortas (8.1 ± 0.6 vs 5.2 ± 0.2). Treatment with LOS reduced (1.12 ± 0.14%) and ENA (0.81 ± 0.11%) or LOS + ENA (0.86 ± 0.11%) additionally diminished collagen only in 2K-1C hearts. CONCLUSIONS Submaximal doses of ACEi and/or ARB have inhibitory actions on cardiac remodelling and vascular hypertrophy not entirely dependent on their effects on blood pressure normalization in renovascular hypertensive rats. Combined therapy produced additional reduction in blood pressure than monotherapy despite a similar inhibition on cardiovascular remodelling.
Collapse
Affiliation(s)
| | - Cibele Maria Prado
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Maria Elena Riul
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Alice Valença Araújo
- Nucleus of Public Health, Academic Center of Vitória (CAV), Federal University of Pernambuco (UFPE), Vitória de Santo Antão, Brazil
| | - Marcos Antonio Rossi
- Department of Pathology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Lusiane Maria Bendhack
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|
15
|
Swiatlowska P, Sanchez-Alonso JL, Mansfield C, Scaini D, Korchev Y, Novak P, Gorelik J. Short-term angiotensin II treatment regulates cardiac nanomechanics via microtubule modifications. NANOSCALE 2020; 12:16315-16329. [PMID: 32720664 DOI: 10.1039/d0nr02474k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Mechanical properties of single myocytes contribute to the whole heart performance, but the measurement of mechanics in living cells at high resolution with minimal force interaction remains challenging. Angiotensin II (AngII) is a peptide hormone that regulates a number of physiological functions, including heart performance. It has also been shown to contribute to cell mechanics by inducing cell stiffening. Using non-contact high-resolution Scanning Ion Conductance Microscopy (SICM), we determine simultaneously cell topography and membrane transverse Young's modulus (YM) by a constant pressure application through a nanopipette. While applying pressure, the vertical position is recorded and a deformation map is generated from which YM can be calculated and corrected for the uneven geometry. High resolution of this method also allows studying specific membrane subdomains, such as Z-grooves and crests. We found that short-term AngII treatment reduces the transversal YM in isolated adult rat cardiomyocytes acting via an AT1 receptor. Blocking either a TGF-β1 receptor or Rho kinase abolishes this effect. Analysis of the cytoskeleton showed that AngII depletes microtubules by decreasing long-lived detyrosinated and acetylated microtubule populations. Interestingly, in the failing cardiomyocytes, which are stiffer than controls, the short-term AngII treatment also reduces the YM, thus normalizing the mechanical state of cells. This suggests that the short-term softening effect of AngII on cardiac cells is opposite to the well-characterized long-term hypertrophic effect. In conclusion, we generate a precise nanoscale indication map of location-specific transverse cortical YM within the cell and this can substantially advance our understanding of cellular mechanics in a physiological environment, for example in isolated cardiac myocytes.
Collapse
Affiliation(s)
- Pamela Swiatlowska
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Jose L Sanchez-Alonso
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Catherine Mansfield
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| | - Denis Scaini
- Department of Medicine, Imperial College London, London, UK and International School for Advanced Studies, Trieste, Italy
| | - Yuri Korchev
- Department of Medicine, Imperial College London, London, UK and Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Pavel Novak
- Department of Medicine, Imperial College London, London, UK and National University of Science and Technology, MISiS, Leninskiy prospect 4, Moscow, 119991, Russia
| | - Julia Gorelik
- Department of Cardiovascular Sciences, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
16
|
Li P, Schmidt IM, Sabbisetti V, Tio MC, Opotowsky AR, Waikar SS. Plasma Endothelin-1 and Risk of Death and Hospitalization in Patients Undergoing Maintenance Hemodialysis. Clin J Am Soc Nephrol 2020; 15:784-793. [PMID: 32381583 PMCID: PMC7274287 DOI: 10.2215/cjn.11130919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 03/19/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVES Endothelin-1 is a potent endothelium-derived vasoconstrictor peptide implicated in the pathogenesis of hypertension, congestive heart failure, and inflammation, all of which are critical pathophysiologic features of CKD. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS To test the hypothesis that plasma endothelin-1 levels are associated with increased risks of mortality and hospitalization in patients with chronic kidney failure, we measured plasma endothelin-1 levels in a prospective cohort of 794 individuals receiving maintenance hemodialysis. The primary outcomes were time to death and time to hospitalization. RESULTS The median plasma endothelin-1 level was 2.02 (interquartile range, 1.57-2.71) pg/ml. During a median follow-up period of 28 (interquartile range, 21-29) months, 253 individuals (32%) died and 643 individuals (81%) were hospitalized at least once. In multivariable models adjusted for demographic, clinical, and laboratory variables, individuals in the highest quartile of plasma endothelin-1 had a 2.44-fold higher risk of death (hazard ratio, 2.44; 95% confidence interval, 1.61 to 3.70) and a 1.54-fold higher risk of hospitalization (hazard ratio, 1.54; 95% confidence interval, 1.19 to 1.99) compared with individuals in the lowest quartile. The Harrell C-statistic of the fully adjusted model increased from 0.73 to 0.74 after addition of natural log-transformed plasma endothelin-1 (P<0.001) for all-cause mortality, and increased from 0.608 to 0.614 after addition of natural log-transformed plasma endothelin-1 (P=0.002) for hospitalization. CONCLUSIONS Higher plasma endothelin-1 is associated with adverse clinical events in patients receiving hemodialysis independent of previously described risk factors. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2020_05_15_CJN11130919.mp3.
Collapse
Affiliation(s)
- Ping Li
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Nephrology, State Key Laboratory of Kidney Disease, National Clinical Research Center for Kidney Disease, Chinese PLA General Hospital, Beijing, China
| | - Insa M. Schmidt
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Boston University Medical Center, Boston, Massachusetts
| | - Venkata Sabbisetti
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Maria Clarissa Tio
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alexander R. Opotowsky
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Cardiology, Boston Children’s Hospital, Boston, Massachusetts
| | - Sushrut S. Waikar
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
- Boston University Medical Center, Boston, Massachusetts
| |
Collapse
|
17
|
Leipziger J, Praetorius H. Renal Autocrine and Paracrine Signaling: A Story of Self-protection. Physiol Rev 2020; 100:1229-1289. [PMID: 31999508 DOI: 10.1152/physrev.00014.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Autocrine and paracrine signaling in the kidney adds an extra level of diversity and complexity to renal physiology. The extensive scientific production on the topic precludes easy understanding of the fundamental purpose of the vast number of molecules and systems that influence the renal function. This systematic review provides the broader pen strokes for a collected image of renal paracrine signaling. First, we recapitulate the essence of each paracrine system one by one. Thereafter the single components are merged into an overarching physiological concept. The presented survey shows that despite the diversity in the web of paracrine factors, the collected effect on renal function may not be complicated after all. In essence, paracrine activation provides an intelligent system that perceives minor perturbations and reacts with a coordinated and integrated tissue response that relieves the work load from the renal epithelia and favors diuresis and natriuresis. We suggest that the overall function of paracrine signaling is reno-protection and argue that renal paracrine signaling and self-regulation are two sides of the same coin. Thus local paracrine signaling is an intrinsic function of the kidney, and the overall renal effect of changes in blood pressure, volume load, and systemic hormones will always be tinted by its paracrine status.
Collapse
Affiliation(s)
- Jens Leipziger
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| | - Helle Praetorius
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; and Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark
| |
Collapse
|
18
|
Caires A, Convento MB, Castino B, Leme AM, Pessoa EDA, Aragão A, Schor N, Borges FT. Antioxidant effect of endothelin-1 receptor antagonist protects the rat kidney against chronic injury induced by hypertension and hyperglycemia. J Bras Nefrol 2019; 41:451-461. [PMID: 31508666 PMCID: PMC6979570 DOI: 10.1590/2175-8239-jbn-2018-0162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 05/27/2019] [Indexed: 11/22/2022] Open
Abstract
ABSTRACT Hypertension and Diabetes mellitus are the two main causes of chronic kidney disease that culminate in the final stage of kidney disease. Since these two risk factors are common and can overlap, new approaches to prevent or treat them are needed. Macitentan (MAC) is a new non-selective antagonist of the endothelin-1 (ET-1) receptor. This study aimed to evaluate the effect of chronic blockade of ET-1 receptor with MAC on the alteration of renal function observed in hypertensive and hyperglycemic animals. Genetically hypertensive rats were divided into control hypertensive (HT-CTL) group, hypertensive and hyperglycemic (HT+DIAB) group, and hypertensive and hyperglycemic group that received 25 mg/kg macitentan (HT-DIAB+MAC25) via gavage for 60 days. Kidney function and parameters associated with oxidative and nitrosative stress were evaluated. Immunohistochemistry for neutrophil gelatinase-associated lipocalin (NGAL), ET-1, and catalase in the renal cortex was performed. The HT+DIAB group showed a decrease in kidney function and an increase in NGAL expression in the renal cortex, as well as an increase in oxidative stress. MAC treatment was associated with attenuated ET-1 and NGAL production and increases in antioxidant defense (catalase expression) and nitric oxide production. In addition, MAC prevented an increase in oxidant injury (as measured by urinary hydroperoxide and lipid peroxidation), thus improving renal function. Our results suggest that the antioxidant effect of the ET-1 receptor antagonist MAC is involved in the improvement of kidney function observed in hypertensive and hyperglycemic rats.
Collapse
|
19
|
Lee CY, Lee J, Seo HH, Shin S, Kim SW, Lee S, Lim S, Hwang KC. TAK733 attenuates adrenergic receptor-mediated cardiomyocyte hypertrophy via inhibiting ErkThr188 phosphorylation. Clin Hemorheol Microcirc 2019; 72:179-187. [PMID: 30714951 DOI: 10.3233/ch-180476] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cardiac hypertrophy is an important risk factor for heart failure. The MEK-ERK axis has been reported as a major regulator in controlling cardiac hypertrophy. TAK733 is a potent and selective MEK inhibitor that suppresses cell growth in a broad range of cell lines. OBJECTIVE Therefore, we aimed to investigate the anti-hypertrophic effect of TAK733 in cardiomyocytes. METHODS Cardiomyocyte hypertrophy was induced with norepinephrine (NE) or phenylepinephrine (PE) using H9c2 cells. To confirm the cardiomyocyte hypertrophy, cell size and protein synthesis were measured and hypertrophy-related gene expression was estimated by reverse transcription polymerase chain reaction. To identify the signaling pathway involved, immunoblot analysis were performed. RESULTS We observed that NE activated MEK-ERK signaling and increased ANP and BNP expression, resulting in cardiomyocyte hypertrophy. TAK733 significantly reduced cardiomyocyte hypertrophy by regulating NE-induced ERK1/2 and ERKThr188 activation, hypertrophy marker expression, and cardiomyocyte hypertrophy through depression of MEK activity. In addition, we examined that PE-induced cardiomyocyte hypertrophy was also attenuated by TAK733. CONCLUSIONS Here, we report that TAK733 suppressed NE- or PE-induced cardiomyocyte hypertrophy by repressing a crucial component of cardiac hypertrophy-related pathways. These results suggest that TAK733 may be a useful therapeutics for cardiac hypertrophy and warrants further in vivo studies.
Collapse
Affiliation(s)
- Chang Youn Lee
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, Republic of Korea
| | - Jiyun Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyang-Hee Seo
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Sunhye Shin
- Department of Integrated Omics for Biomedical Sciences, Yonsei University, Seoul, Republic of Korea
| | - Sang Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| | - Ki-Chul Hwang
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung, Gangwon-do, Republic of Korea
| |
Collapse
|
20
|
Yeves AM, Ennis IL. Na +/H + exchanger and cardiac hypertrophy. HIPERTENSION Y RIESGO VASCULAR 2019; 37:22-32. [PMID: 31601481 DOI: 10.1016/j.hipert.2019.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/16/2019] [Accepted: 09/03/2019] [Indexed: 12/31/2022]
Abstract
Reactive cardiac hypertrophy (CH) is an increase in heart mass in response to hemodynamic overload. Exercise-induced CH emerges as an adaptive response with improved cardiac function, in contrast to pathological CH that represents a risk factor for cardiovascular health. The Na+/H+ exchanger (NHE-1) is a membrane transporter that not only regulates intracellular pH but also intracellular Na+ concentration. In the scenario of cardiovascular diseases, myocardial NHE-1 is activated by a variety of stimuli, such as neurohumoral factors and mechanical stress, leading to intracellular Na+ overload and activation of prohypertrophic cascades. NHE-1 hyperactivity is intimately linked to heart diseases, including ischemia-reperfusion injury, maladaptive CH and heart failure. In this review, we will present evidence to support that the NHE-1 hyperactivity constitutes a "switch on/off" for the pathological phenotype during CH development. We will also discuss some classical and novel strategies to avoid NHE-1 hyperactivity, and that are therefore worthwhile to improve cardiovascular health.
Collapse
Affiliation(s)
- A M Yeves
- Centro de Investigaciones Cardiovasculares "Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata - CONICET, Calle 60 y 120, 1900 La Plata, Argentina
| | - I L Ennis
- Centro de Investigaciones Cardiovasculares "Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata - CONICET, Calle 60 y 120, 1900 La Plata, Argentina.
| |
Collapse
|
21
|
Cdc42 activation by endothelin regulates neural crest cell migration in the cardiac outflow tract. Dev Dyn 2019; 248:795-812. [DOI: 10.1002/dvdy.75] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
|
22
|
Dowrick JM, Tran K, Loiselle DS, Nielsen PMF, Taberner AJ, Han J, Ward M. The slow force response to stretch: Controversy and contradictions. Acta Physiol (Oxf) 2019; 226:e13250. [PMID: 30614655 DOI: 10.1111/apha.13250] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/20/2018] [Accepted: 01/02/2019] [Indexed: 12/19/2022]
Abstract
When exposed to an abrupt stretch, cardiac muscle exhibits biphasic active force enhancement. The initial, instantaneous, force enhancement is well explained by the Frank-Starling mechanism. However, the cellular mechanisms associated with the second, slower phase remain contentious. This review explores hypotheses regarding this "slow force response" with the intention of clarifying some apparent contradictions in the literature. The review is partitioned into three sections. The first section considers pathways that modify the intracellular calcium handling to address the role of the sarcoplasmic reticulum in the mechanism underlying the slow force response. The second section focuses on extracellular calcium fluxes and explores the identity and contribution of the stretch-activated, non-specific, cation channels as well as signalling cascades associated with G-protein coupled receptors. The final section introduces promising candidates for the mechanosensor(s) responsible for detecting the stretch perturbation.
Collapse
Affiliation(s)
- Jarrah M. Dowrick
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Denis S. Loiselle
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Physiology University of Auckland Auckland New Zealand
| | - Poul M. F. Nielsen
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - Andrew J. Taberner
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
- Department of Engineering Science University of Auckland Auckland New Zealand
| | - June‐Chiew Han
- Auckland Bioengineering Institute University of Auckland Auckland New Zealand
| | - Marie‐Louise Ward
- Department of Physiology University of Auckland Auckland New Zealand
| |
Collapse
|
23
|
Miyauchi T, Sakai S. Endothelin and the heart in health and diseases. Peptides 2019; 111:77-88. [PMID: 30352269 DOI: 10.1016/j.peptides.2018.10.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/03/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022]
Abstract
Endothelin-1 (ET-1), a 21-amino acid peptide, was initially identified in 1988 as a potent vasoconstrictor and pressor substance isolated from the culture supernatant of porcine aortic endothelial cells. From human genomic DNA analysis, two other family peptides, ET-2 and ET-3, were found. They showed different effects and distribution, suggesting that each peptide may play separate roles in different organs. In the heart, ET-1 also causes positive inotropic and chronotropic responses and hypertrophic activity of the cardiomyocytes. ETs act via activation of two receptor subtypes, ETA and ETB receptors, both of which are coupled to various GTP-binding proteins depending on cell types. Endogenous ET-1 may be involved in progression of various cardiovascular diseases. ET antagonists are currently used clinically in the treatment for patients with pulmonary hypertension, and are considered to have further target diseases as heart failure, cardiac hypertrophy and other cardiac diseases, renal diseases, systemic hypertension, and cerebral vasospasm.
Collapse
Affiliation(s)
- Takashi Miyauchi
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Japan.
| | - Satoshi Sakai
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Japan
| |
Collapse
|
24
|
Mechanical stresses induce paracrine β-2 microglobulin from cardiomyocytes to activate cardiac fibroblasts through epidermal growth factor receptor. Clin Sci (Lond) 2018; 132:1855-1874. [PMID: 30072448 DOI: 10.1042/cs20180486] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 11/17/2022]
Abstract
By employing a proteomic analysis on supernatant of mechanically stretched cardiomyocytes, we found that stretch induced a significantly high level of β-2 microglobulin (β2M), a non-glycosylated protein, which is related to inflammatory diseases but rarely known in cardiovascular diseases. The present data showed that serum β2M level was increased in patients with hypertension and further increased in patients with chronic heart failure (HF) as compared with control group, and the high level of serum β2M level correlated to cardiac dysfunction in these patients. In pressure overload mice model by transverse aortic constriction (TAC), β2M levels in serum and heart tissue increased progressively in a time-dependent manner. Exogenous β2M showed pro-fibrotic effects in cultured cardiac fibroblasts but few effects in cardiomyocytes. Adeno-associated virus 9 (AAV9)-mediated knockdown of β2M significantly reduced cardiac β2M level and inhibited myocardial fibrosis and cardiac dysfunction but not cardiac hypertrophy at 4 weeks after TAC. In vitro, mechanical stretch induced the rapid secretion of β2M mainly from cardiomyocytes by activation of extracellular-regulated protein kinase (ERK). Conditional medium (CM) from mechanically stretched cardiomyocytes activated cultured cardiac fibroblasts, and the effect was partly abolished by CM from β2M-knockdown cardiomyocytes. In vivo, knockdown of β2M inhibited the increase in phosphorylation of epidermal growth factor receptor (EGFR) induced by TAC. In cultured cardiac fibroblasts, inhibition of EGFR significantly attenuated the β2M-induced the activation of EGFR and pro-fibrotic responses. The present study suggests that β2M is a paracrine pro-fibrotic mediator and associated with cardiac dysfunction in response to pressure overload.
Collapse
|
25
|
Falk M, Huhn R, Behmenburg F, Ritz-Timme S, Mayer F. Biomechanical stress in myocardial infarctions: can endothelin-1 and growth differentiation factor 15 serve as immunohistochemical markers? Int J Legal Med 2017; 132:509-518. [DOI: 10.1007/s00414-017-1726-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/25/2017] [Indexed: 01/08/2023]
|
26
|
Sun Y, Han M, Shen Z, Huang H, Miao X. Anti-hypertensive and cardioprotective effects of a novel apitherapy formulation via upregulation of peroxisome proliferator-activated receptor-α and -γ in spontaneous hypertensive rats. Saudi J Biol Sci 2017; 25:213-219. [PMID: 29472767 PMCID: PMC5816011 DOI: 10.1016/j.sjbs.2017.10.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 02/01/2023] Open
Abstract
Ventricular remodeling is associated with many heart diseases, and ventricular remodeling induced by hypertension can be fatal independent of hypertension. In this study, we prepared a novel apitherapy formulation, designated Bao-Yuan-Ling (BYL), which contained propolis, royal jelly, and bee venom, to treat spontaneous hypertensive rats (SHRs). We then evaluated the pharmacology of BYL and the potential mechanisms through which BYL affects hypertension and ventricular remodeling. We found that BYL treatment could reduce blood pressure in SHRs. Thereafter, we found that BYL treatment reduced serum levels of angiotensin II, endothelin 1, and transforming growth factor-β and improved the myocardial structure. Moreover, the results of quantitative real-time polymerase chain reaction indicated that BYL treatment could upregulate the mRNA expression of peroxisome proliferator-activated receptor (PPAR)-α and PPAR-γ. Thus, we could conclude that BYL had hypotensive and cardioprotective effects in SHRs, potentially through improvement of myocardial energy metabolism.
Collapse
Affiliation(s)
- Yanru Sun
- College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China.,College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China
| | - Mingfeng Han
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Zhenhuang Shen
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Haibo Huang
- National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| | - Xiaoqing Miao
- College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou 350000, China.,National United Engineering Laboratory of Natural Biological Toxins, Fuzhou 350000, China
| |
Collapse
|
27
|
Wu MY, Li CJ, Hou MF, Chu PY. New Insights into the Role of Inflammation in the Pathogenesis of Atherosclerosis. Int J Mol Sci 2017; 18:2034. [PMID: 28937652 PMCID: PMC5666716 DOI: 10.3390/ijms18102034] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 09/19/2017] [Accepted: 09/19/2017] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipids, smooth muscle cell proliferation, cell apoptosis, necrosis, fibrosis, and local inflammation. Immune and inflammatory responses have significant effects on every phase of atherosclerosis, and increasing evidence shows that immunity plays a more important role in atherosclerosis by tightly regulating its progression. Therefore, understanding the relationship between immune responses and the atherosclerotic microenvironment is extremely important. This article reviews existing knowledge regarding the pathogenesis of immune responses in the atherosclerotic microenvironment, and the immune mechanisms involved in atherosclerosis formation and activation.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| | - Ming-Feng Hou
- Department of Surgery, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Surgery, Kaohsiung Municipal Hsiao Kang Hospital, Kaohsiung 807, Taiwan.
- Division of Breast Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei 242, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
| |
Collapse
|
28
|
Li Q, Yu Q, Na R, Liu B. Etanercept protects rat cardiomyocytes against hypertrophy by regulating inflammatory cytokines secretion and cell apoptosis. Braz J Med Biol Res 2017; 50:e5868. [PMID: 28513772 PMCID: PMC5479384 DOI: 10.1590/1414-431x20175868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 03/27/2017] [Indexed: 01/19/2023] Open
Abstract
We aimed to investigate the effect of etanercept, a tumor necrosis factor-α (TNF-α) inhibitor, on rat cardiomyocyte hypertrophy and its underlying mechanism. Primary neonatal rat cardiomyocytes were isolated from Sprague-Dawley rats. The model of rat cardiomyocyte hypertrophy was induced by endothelin, and then treated with different concentrations of etanercept (1, 10, and 50 μM). After treatment, cell counts, viability and cell apoptosis were evaluated. The mRNA levels of myocardial hypertrophy marker genes, including atrial natriuretic factor (ANF), matrix metalloproteinase (MMP)-9 and MMP-13, were detected by qRT-PCR, and the expressions of apoptosis-related proteins (Bcl-2 and Bax) were measured by western blotting. The protein levels of transforming growth factor-β1 (TGF-β1), interleukin (IL)-1β, IL-6, leukemia inhibitory factor (LIF) and cardiotrophin-1 (CT-1) were determined using enzyme linked immunosorbent assay (ELISA) kits. In the present study, TNF-α level in cardiomyocytes with hypertrophy was significantly enhanced (P<0.05). Compared to the model group, cell number and viability were significantly increased and ratio of apoptotic cells was reduced by etanercept (P<0.05, P<0.01, or P<0.001). In addition, etanercept remarkably reduced the mRNA levels of ANF, MMP-9 and MMP-13, inhibited the expression of Bax, and increased the expression of Bcl-2 compared to the model group (P<0.05). ELISA results further showed that etanercept lowered the levels of IL-1β, IL-6, LIF and CT-1 but not TGF-β1 compared to the model group (P<0.05). Etanercept may protect rat cardiomyocytes from hypertrophy by inhibiting inflammatory cytokines secretion and cell apoptosis.
Collapse
Affiliation(s)
- Q. Li
- Zhejiang Province Hospital of Integrated Traditional Chinese and Western Medicine, Hangzhou, China
| | - Q. Yu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - R. Na
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - B. Liu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
29
|
Gaggin HK, Truong QA, Gandhi PU, Motiwala SR, Belcher AM, Weiner RB, Baggish AL, Januzzi JL. Systematic Evaluation of Endothelin 1 Measurement Relative to Traditional and Modern Biomarkers for Clinical Assessment and Prognosis in Patients With Chronic Systolic Heart Failure: Serial Measurement and Multimarker Testing. Am J Clin Pathol 2017; 147:461-472. [PMID: 28398455 DOI: 10.1093/ajcp/aqx014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES To define the role of single or serial measurement of endothelin 1 (ET-1) for prognostication beyond traditional and modern markers of risk in heart failure (HF). METHODS In total, 115 patients with chronic systolic HF were followed for 10 months. Clinical assessment and ET-1, N-terminal pro-B-type natriuretic peptide (NT-proBNP), highly sensitive troponin I (hsTnI), soluble ST2 (sST2), and galectin 3 were measured at each visit. RESULTS Elevated ET-1 was associated with worse HF, lower right ventricular function, higher pulmonary pressure, and higher left atrial volume index despite similar left ventricular function. ET-1 correlated with angiotensin-converting enzyme inhibitor use. A model containing traditional risk factors, ET-1, NT-proBNP, hsTnI, and sST2 best predicted cardiovascular events, and ET-1 improved reclassification. In an adjusted time-integrated model, percent time spent with ET-1 of 5.90 pg/mL or less was predictive of fewer cardiovascular events (odds ratio, 0.75; 95% confidence interval, 0.62-0.91). ET-1 reduction over time was associated with a lower rate of cardiovascular events compared with increasing or stable ET-1 (24.4% vs 50.0%). CONCLUSIONS ET-1 may be a unique predictor of HF prognosis, complementing other biomarkers in a multimarker profile. Serial measurement of ET-1 may provide additional prognostic information.
Collapse
Affiliation(s)
- Hanna K Gaggin
- Massachusetts General Hospital, Boston
- Baim Institute for Clinical Research, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Heo SY, Ko SC, Kim CS, Oh GW, Ryu B, Qian ZJ, Kim G, Park WS, Choi IW, Phan TTV, Heo SJ, Kang DH, Yi M, Jung WK. A heptameric peptide purified from Spirulina sp. gastrointestinal hydrolysate inhibits angiotensin I-converting enzyme- and angiotensin II-induced vascular dysfunction in human endothelial cells. Int J Mol Med 2017; 39:1072-1082. [PMID: 28393188 PMCID: PMC5403476 DOI: 10.3892/ijmm.2017.2941] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2016] [Accepted: 03/13/2017] [Indexed: 11/06/2022] Open
Abstract
In this study, a marine microalga Spirulina sp.-derived protein was hydrolyzed using gastrointestinal enzymes to produce an angiotensin I (Ang I)-converting enzyme (ACE) inhibitory peptide. Following consecutive purification, the potent ACE inhibitory peptide was composed of 7 amino acids, Thr-Met-Glu-Pro-Gly-Lys-Pro (molecular weight, 759 Da). Analysis using the Lineweaver-Burk plot and molecular modeling suggested that the purified peptide acted as a mixed non-competitive inhibitor of ACE. The inhibitory effects of the peptide against the cellular production of vascular dysfunction-related factors induced by Ang II were also investigated. In human endothelial cells, the Ang II-induced production of nitric oxide and reactive oxygen species was inhibited, and the expression of inducible nitric oxide synthase (iNOS) and endothelin-1 (ET-1) was downregulated when the cells were cultured with the purified peptide. Moreover, the peptide blocked the activation of p38 mitogen-activated protein kinase. These results indicated that this Spirulina sp.-derived peptide warrants further investigation as a potential pharmacological inhibitor of ACE and vascular dysfunction.
Collapse
Affiliation(s)
- Seong-Yeong Heo
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Seok-Chun Ko
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Chang Su Kim
- Department of Orthopedic Surgery, Kosin University Gospel Hospital, Busan 49267, Republic of Korea
| | - Gun-Woo Oh
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Bomi Ryu
- School of Pharmacy, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Zhong-Ji Qian
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, Guangdong 524088, P.R. China
| | - Geunhyung Kim
- Department of Bio-Mechatronic Engineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi 16419, Republic of Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University, School of Medicine, Chuncheon, Gangwon 24341, Republic of Korea
| | - Il-Whan Choi
- Department of Microbiology, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Thi Tuong Vy Phan
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Soo-Jin Heo
- Jeju International Marine Science Center for Research and Education, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Do-Hyung Kang
- Jeju International Marine Science Center for Research and Education, Korea Institute of Ocean Science and Technology (KIOST), Jeju 63349, Republic of Korea
| | - Myunggi Yi
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Republic of Korea
| |
Collapse
|
31
|
Riley CJ, Gavin M. Physiological Changes to the Cardiovascular System at High Altitude and Its Effects on Cardiovascular Disease. High Alt Med Biol 2017; 18:102-113. [PMID: 28294639 DOI: 10.1089/ham.2016.0112] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Riley, Callum James, and Matthew Gavin. Physiological changes to the cardiovascular system at high altitude and its effects on cardiovascular disease. High Alt Med Biol. 18:102-113, 2017.-The physiological changes to the cardiovascular system in response to the high altitude environment are well understood. More recently, we have begun to understand how these changes may affect and cause detriment to cardiovascular disease. In addition to this, the increasing availability of altitude simulation has dramatically improved our understanding of the physiology of high altitude. This has allowed further study on the effect of altitude in those with cardiovascular disease in a safe and controlled environment as well as in healthy individuals. Using a thorough PubMed search, this review aims to integrate recent advances in cardiovascular physiology at altitude with previous understanding, as well as its potential implications on cardiovascular disease. Altogether, it was found that the changes at altitude to cardiovascular physiology are profound enough to have a noteworthy effect on many forms of cardiovascular disease. While often asymptomatic, there is some risk in high altitude exposure for individuals with certain cardiovascular diseases. Although controlled research in patients with cardiovascular disease was largely lacking, meaning firm conclusions cannot be drawn, these risks should be a consideration to both the individual and their physician.
Collapse
Affiliation(s)
| | - Matthew Gavin
- 2 University of Leeds School of Biomedical Sciences , Leeds, United Kingdom
| |
Collapse
|
32
|
Chan EAW, Buckley B, Farraj AK, Thompson LC. The heart as an extravascular target of endothelin-1 in particulate matter-induced cardiac dysfunction. Pharmacol Ther 2016; 165:63-78. [PMID: 27222357 PMCID: PMC6390286 DOI: 10.1016/j.pharmthera.2016.05.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Exposure to particulate matter air pollution has been causally linked to cardiovascular disease in humans. Several broad and overlapping hypotheses describing the biological mechanisms by which particulate matter exposure leads to cardiovascular disease have been explored, although linkage with specific factors or genes remains limited. These hypotheses may or may not also lead to particulate matter-induced cardiac dysfunction. Evidence pointing to autocrine/paracrine signaling systems as modulators of cardiac dysfunction has increased interest in the emerging role of endothelins as mediators of cardiac function following particulate matter exposure. Endothelin-1, a well-described small peptide expressed in the pulmonary and cardiovascular systems, is best known for its ability to constrict blood vessels, although it can also induce extravascular effects. Research on the role of endothelins in the context of air pollution has largely focused on vascular effects, with limited investigation of responses resulting from the direct effects of endothelins on cardiac tissue. This represents a significant knowledge gap in air pollution health effects research, given the abundance of endothelin receptors found on cardiac tissue and the ability of endothelin-1 to modulate cardiac contractility, heart rate, and rhythm. The plausibility of endothelin-1 as a mediator of particulate matter-induced cardiac dysfunction is further supported by the therapeutic utility of certain endothelin receptor antagonists. The present review examines the possibility that endothelin-1 release caused by exposure to PM directly modulates extravascular effects on the heart, deleteriously altering cardiac function.
Collapse
Affiliation(s)
- Elizabeth A W Chan
- Oak Ridge Institute for Science and Education (ORISE) Fellow at the National Center for Environmental Assessment, U.S. Environmental Protection Agency (EPA), Research Triangle Park, NC, USA
| | - Barbara Buckley
- National Center for Environmental Assessment, U.S. EPA, Research Triangle Park, NC, USA
| | - Aimen K Farraj
- Environmental Public Health Division, U.S. EPA, Research Triangle Park, NC, USA
| | - Leslie C Thompson
- Environmental Public Health Division, U.S. EPA, Research Triangle Park, NC, USA.
| |
Collapse
|
33
|
Meyer MR, Fredette NC, Sharma G, Barton M, Prossnitz ER. GPER is required for the age-dependent upregulation of the myocardial endothelin system. Life Sci 2016; 159:61-65. [PMID: 26880534 PMCID: PMC4983270 DOI: 10.1016/j.lfs.2016.02.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 01/31/2016] [Accepted: 02/10/2016] [Indexed: 02/07/2023]
Abstract
AIMS Cardiac aging is associated with progressive structural changes and functional impairment, such as left ventricular hypertrophy, fibrosis and diastolic dysfunction. Aging also increases myocardial activity of endothelin-1 (ET-1), a multifunctional peptide with growth-promoting and pro-fibrotic activity. Because the G protein-coupled estrogen receptor (GPER) regulates vascular responsiveness to ET-1, we investigated whether GPER also plays a role in the regulation of the myocardial endothelin system with aging. MAIN METHODS Young (4month-old) and aged (24month-old) wild-type and Gper-deficient (Gper(-/-)) mice were studied. Gene expression levels of prepro-ET-1, endothelin converting enzymes ECE-1 and ECE-2, and endothelin ETA and ETB receptors were determined by qPCR in left ventricular myocardium. KEY FINDINGS Aging markedly increased steady-state mRNA expression levels of ECE-1, ECE-2, ETA and ETB receptors (each p<0.001 vs. young mice). Deletion of Gper inhibited the age-dependent increase in ECE-2 and ETB receptor mRNA levels (57% and 40% reduction, respectively, each p<0.01 vs. wild-type mice), whereas gene expression of prepro-ET-1, ECE-1, and the ETA receptor was unaffected in Gper(-/-) mice. SIGNIFICANCE We identified a novel regulatory mechanism through which the endogenous Gper facilitates the age-dependent increase in myocardial expression of ECE-2 and the ETB receptor, which is compatible with an activating role of GPER for the local endothelin system with aging. Targeting GPER signaling by selective antagonists may therefore be considered a new therapeutic approach to reduce age-dependent increased ET-1 activity and the associated development of left ventricular hypertrophy, fibrosis and heart failure.
Collapse
Affiliation(s)
- Matthias R Meyer
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.
| | - Natalie C Fredette
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Geetanjali Sharma
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Matthias Barton
- Molecular Internal Medicine, University of Zürich, Zürich, Switzerland
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.
| |
Collapse
|
34
|
Liu X, Zou C, Yu C, Xie R, Sui M, Mu S, Li L, Zhao S. Original Research: Atorvastatin prevents rat cardiomyocyte hypertrophy induced by parathyroid hormone 1-34 associated with the Ras-ERK signaling. Exp Biol Med (Maywood) 2016; 241:1745-50. [PMID: 27190264 DOI: 10.1177/1535370216649259] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/30/2016] [Indexed: 12/13/2022] Open
Abstract
We investigated the effects of atorvastatin (Ator) on cardiomyocyte hypertrophy (CMH) induced by rat parathyroid hormone 1-34 (PTH1-34) and Ras-extracellular signal regulated protein kinases 1/2 (ERK1/2) signaling. Rat cardiomyocytes were randomly divided into seven groups: normal controls (NC), PTH1-34 (10(-7) mol/L), Ator (10(-5) mol/L), farnesyl transferase inhibitors-276 (FTI-276, 4 × 10(-5) mol/L), PTH1-34 + Ator, PTH1-34 + FTI-276 and PTH1-34 + Ator + mevalonic acid (MVA, 10(-4) mol/L). After treatment, the hypertrophic responses of cardiomyocytes were assessed by measuring cell diameter, detecting protein synthesis, and single-cell protein content. The concentrations of hypertrophic markers such as atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) were measured by ELISA. Protein expressions of ERK1/2, p-ERK1/2 and Ras were detected by western blotting. The results showed that compared with the PTH1-34 group, cellular diameter, 3H-leucine incorporation, single-cell protein content, ANP and BNP concentration decreased by 12.07 µm, 1622 cpm/well, 84.34 pg, 7.13 ng/L and 20.04 µg/L, respectively, and the expressions of Ras and p-ERK1/2 were downregulated in PTH1-34 + Ator group (P < 0.05). Compared to the PTH1-34 + Ator group, the corresponding hypertrophic responses and hypertrophic markers increased by 4.95 µm, 750 cpm/well, 49.08 pg, 3.12 ng/L and 9.35 µg/L, respectively, and the expressions of Ras and p-ERK1/2 were upregulated in the PTH1-34 + Ator + MVA group (P < 0.05). In conclusion, Ator prevents neonatal rat CMH induced by PTH1-34 and Ras-ERK signaling may be involved in this process.
Collapse
Affiliation(s)
- Xiaogang Liu
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Chunbo Zou
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Chengyuan Yu
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Rujuan Xie
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Manshu Sui
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Suhong Mu
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Li Li
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Shilei Zhao
- Department of Nephrology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| |
Collapse
|
35
|
Michel MC, Brunner HR, Foster C, Huo Y. Angiotensin II type 1 receptor antagonists in animal models of vascular, cardiac, metabolic and renal disease. Pharmacol Ther 2016; 164:1-81. [PMID: 27130806 DOI: 10.1016/j.pharmthera.2016.03.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 02/07/2023]
Abstract
We have reviewed the effects of angiotensin II type 1 receptor antagonists (ARBs) in various animal models of hypertension, atherosclerosis, cardiac function, hypertrophy and fibrosis, glucose and lipid metabolism, and renal function and morphology. Those of azilsartan and telmisartan have been included comprehensively whereas those of other ARBs have been included systematically but without intention of completeness. ARBs as a class lower blood pressure in established hypertension and prevent hypertension development in all applicable animal models except those with a markedly suppressed renin-angiotensin system; blood pressure lowering even persists for a considerable time after discontinuation of treatment. This translates into a reduced mortality, particularly in models exhibiting marked hypertension. The retrieved data on vascular, cardiac and renal function and morphology as well as on glucose and lipid metabolism are discussed to address three main questions: 1. Can ARB effects on blood vessels, heart, kidney and metabolic function be explained by blood pressure lowering alone or are they additionally directly related to blockade of the renin-angiotensin system? 2. Are they shared by other inhibitors of the renin-angiotensin system, e.g. angiotensin converting enzyme inhibitors? 3. Are some effects specific for one or more compounds within the ARB class? Taken together these data profile ARBs as a drug class with unique properties that have beneficial effects far beyond those on blood pressure reduction and, in some cases distinct from those of angiotensin converting enzyme inhibitors. The clinical relevance of angiotensin receptor-independent effects of some ARBs remains to be determined.
Collapse
Affiliation(s)
- Martin C Michel
- Dept. Pharmacology, Johannes Gutenberg University, Mainz, Germany; Dept. Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim, Ingelheim, Germany.
| | | | - Carolyn Foster
- Retiree from Dept. of Research Networking, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Yong Huo
- Dept. Cardiology & Heart Center, Peking University First Hospital, Beijing, PR China
| |
Collapse
|
36
|
Saad NS, Floyd K, Ahmed AAE, Mohler PJ, Janssen PML, Elnakish MT. The Effect of Sorafenib, Tadalafil and Macitentan Treatments on Thyroxin-Induced Hemodynamic Changes and Cardiac Abnormalities. PLoS One 2016; 11:e0153694. [PMID: 27082116 PMCID: PMC4833287 DOI: 10.1371/journal.pone.0153694] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/03/2016] [Indexed: 11/18/2022] Open
Abstract
Multikinase inhibitors (e.g. Sorafenib), phosphodiesterase-5 inhibitors (e.g. Tadalafil), and endothelin-1 receptor blockers (e.g. Macitentan) exert influential protection in a variety of animal models of cardiomyopathy; however, their effects on thyroxin-induced cardiomyopathy have never been investigated. The goal of the present study was to assess the functional impact of these drugs on thyroxin-induced hemodynamic changes, cardiac hypertrophy and associated altered responses of the contractile myocardium both in-vivo at the whole heart level and ex-vivo at the cardiac tissue level. Control and thyroxin (500 μg/kg/day)-treated mice with or without 2-week treatments of sorafenib (10 mg/kg/day; I.P), tadalafil (1 mg/kg/day; I.P or 4 mg/kg/day; oral), macitentan (30 and 100 mg/kg/day; oral), and their vehicles were studied. Blood pressure, echocardiography and electrocardiogram were non-invasively evaluated, followed by ex-vivo assessments of isolated multicellular cardiac preparations. Thyroxin increased blood pressure, resulted in cardiac hypertrophy and left ventricular dysfunction in-vivo. Also, it caused contractile abnormalities in right ventricular papillary muscles ex-vivo. None of the drug treatments were able to significantly attenuate theses hemodynamic changes or cardiac abnormalities in thyroxin-treated mice. We show here for the first time that multikinase (raf1/b, VEGFR, PDGFR), phosphodiesterase-5, and endothelin-1 pathways have no major role in thyroxin-induced hemodynamic changes and cardiac abnormalities. In particular, our data show that the involvement of endothelin-1 pathway in thyroxine-induced cardiac hypertrophy/dysfunction seems to be model-dependent and should be carefully interpreted.
Collapse
Affiliation(s)
- Nancy S. Saad
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Kyle Floyd
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Amany A. E. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Peter J. Mohler
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Paul M. L. Janssen
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Mohammad T. Elnakish
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
- * E-mail:
| |
Collapse
|
37
|
Li N, Si B, Ju JF, Zhu M, You F, Wang D, Ren J, Ning YS, Zhang FQ, Dong K, Huang J, Yu WQ, Wang TJ, Qiao B. Nicotine Induces Cardiomyocyte Hypertrophy Through TRPC3-Mediated Ca 2+/NFAT Signalling Pathway. Can J Cardiol 2015; 32:1260.e1-1260.e10. [PMID: 26952156 DOI: 10.1016/j.cjca.2015.12.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 11/11/2015] [Accepted: 12/02/2015] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Nicotine is thought to be an important risk factor for the development of cardiovascular diseases. However, the effects of nicotine on cardiomyocyte hypertrophy are poorly understood. The present study was designed to explore the role of nicotine in cardiomyocyte hypertrophy and its underlying mechanism. METHODS We used primary cardiomyocytes isolated from Wistar rats to examine the effects of nicotine on intracellular Ca2+ mobilization and hypertrophy determined by immunofluorescence, quantitative polymerase chain reaction, and western blot analysis. A luciferase reporter assay was used to examine the activity of NFAT signalling. RESULTS We found that nicotine caused cardiomyocyte hypertrophy, which was accompanied by increased intracellular Ca2+. Nicotine-enhanced intracellular Ca2+ concentration ([Ca2+]i) was significantly abolished by store-operated Ca2+ entry (SOCE) and TRPC inhibitors. Knockdown of TRPC3 significantly decreased nicotine-induced SOCE and hypertrophy. Moreover, calcineurin-nuclear factor of activated T cells (NFAT) is involved in TRPC3-mediated Ca2+ signalling and cardiomyocyte hypertrophy. Notably, upregulation of TRPC3 by nicotine requires TRPC3-mediated Ca2+ influx and calcineurin-NFAT signalling activation. CONCLUSIONS Our findings demonstrate that the prohypertrophic effect of nicotine on cardiomyocytes is dependent on enhanced TRPC3 expression through a calcium-dependent regulatory loop, which could become a potential target for prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Na Li
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Biao Si
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Ji-Feng Ju
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Meng Zhu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng You
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Dong Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jie Ren
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Yan-Song Ning
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Feng-Quan Zhang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Kai Dong
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Jing Huang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Wen-Qian Yu
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China
| | - Tong-Jian Wang
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| | - Bin Qiao
- Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China.
| |
Collapse
|
38
|
Breitling S, Ravindran K, Goldenberg NM, Kuebler WM. The pathophysiology of pulmonary hypertension in left heart disease. Am J Physiol Lung Cell Mol Physiol 2015; 309:L924-41. [DOI: 10.1152/ajplung.00146.2015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/20/2015] [Indexed: 12/17/2022] Open
Abstract
Pulmonary hypertension (PH) is characterized by elevated pulmonary arterial pressure leading to right-sided heart failure and can arise from a wide range of etiologies. The most common cause of PH, termed Group 2 PH, is left-sided heart failure and is commonly known as pulmonary hypertension with left heart disease (PH-LHD). Importantly, while sharing many clinical features with pulmonary arterial hypertension (PAH), PH-LHD differs significantly at the cellular and physiological levels. These fundamental pathophysiological differences largely account for the poor response to PAH therapies experienced by PH-LHD patients. The relatively high prevalence of this disease, coupled with its unique features compared with PAH, signal the importance of an in-depth understanding of the mechanistic details of PH-LHD. The present review will focus on the current state of knowledge regarding the pathomechanisms of PH-LHD, highlighting work carried out both in human trials and in preclinical animal models. Adaptive processes at the alveolocapillary barrier and in the pulmonary circulation, including alterations in alveolar fluid transport, endothelial junctional integrity, and vasoactive mediator secretion will be discussed in detail, highlighting the aspects that impact the response to, and development of, novel therapeutics.
Collapse
Affiliation(s)
- Siegfried Breitling
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Germany
| | - Krishnan Ravindran
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Neil M. Goldenberg
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Anesthesia, University of Toronto, Toronto, Ontario, Canada
| | - Wolfgang M. Kuebler
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Germany
- Departments of Surgery and Physiology, University of Toronto, Toronto, Ontario, Canada; and
- German Heart Institute Berlin, Berlin, Germany
| |
Collapse
|
39
|
Ding XQ, Ge PC, Liu Z, Jia H, Chen X, An FH, Li LH, Chen ZH, Mao HW, Li ZY, Gu Y, Zhu TB, Li CJ, Wang LS, Ma WZ, Yang ZJ, Jia EZ. Interaction between microRNA expression and classical risk factors in the risk of coronary heart disease. Sci Rep 2015; 5:14925. [PMID: 26446730 PMCID: PMC4597355 DOI: 10.1038/srep14925] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 09/09/2015] [Indexed: 01/07/2023] Open
Abstract
The aim of this study was to identify the synergistic effect of microRNA expression with classical risk factors of coronary heart disease (CHD) and to explore their diagnostic value for coronary stenotic lesions in subjects with CHD. Plasma samples were obtained from 66 subjects with CHD and from 58 control individuals. A quantitative reverse-transcription PCR (RT-qPCR) assay was conducted to confirm the relative expressions of the known CHD-related miRNAs. The severity of coronary atherosclerosis was based on the Gensini scoring system. The expression of miR-125b in plasma of the CHD group was lower than that of the non-CHD group (0.14 ± 0.09 vs. 0.18 ± 0.10, p = 0.055), and the miR-125b levels significantly decreased following an increasing Gensini score (P = 0.037). Spearman correlation analyses indicated the Gensini score was negatively associated with miR-125b (r = −0.215, p = 0.017). Of all the miRNAs, miR-125b showed the lowest AUC (0.405; 95% CI: 0.305 ~ 0.506, p = 0.070). We found several synergistic effects between miR-125b and classical risk factors, such as age, sex, CR, FBG and HDL-C; the proportion of CHD attributable to the interaction of miR-125b and age was as high as 80%. Therefore, miR-125b was shown to play an important role in individual’s susceptibility to developing CHD.
Collapse
Affiliation(s)
- Xiao-Qing Ding
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Peng-Cheng Ge
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Zhe Liu
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Heng Jia
- Kangda school, Nanjing Medical University, Lianyungang 222000, Jiangsu Province, China
| | - Xi Chen
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210029, Jiangsu Province, China
| | - Feng-Hui An
- Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining 835000, Xinjiang, China
| | - Li-Hua Li
- Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining 835000, Xinjiang, China
| | - Zhao-Hong Chen
- Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining 835000, Xinjiang, China
| | - Hong-Wei Mao
- Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining 835000, Xinjiang, China
| | - Zhao-Yang Li
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Yan Gu
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Tie-Bing Zhu
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Chun-Jian Li
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Lian-Sheng Wang
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Wen-Zhu Ma
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Zhi-Jian Yang
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - En-Zhi Jia
- First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| |
Collapse
|
40
|
Quarles EK, Dai DF, Tocchi A, Basisty N, Gitari L, Rabinovitch PS. Quality control systems in cardiac aging. Ageing Res Rev 2015; 23:101-15. [PMID: 25702865 PMCID: PMC4686341 DOI: 10.1016/j.arr.2015.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/02/2015] [Accepted: 02/12/2015] [Indexed: 12/31/2022]
Abstract
Cardiac aging is an intrinsic process that results in impaired cardiac function, along with cellular and molecular changes. These degenerative changes are intimately associated with quality control mechanisms. This review provides a general overview of the clinical and cellular changes which manifest in cardiac aging, and the quality control mechanisms involved in maintaining homeostasis and retarding aging. These mechanisms include autophagy, ubiquitin-mediated turnover, apoptosis, mitochondrial quality control and cardiac matrix homeostasis. Finally, we discuss aging interventions that have been observed to impact cardiac health outcomes. These include caloric restriction, rapamycin, resveratrol, GDF11, mitochondrial antioxidants and cardiolipin-targeted therapeutics. A greater understanding of the quality control mechanisms that promote cardiac homeostasis will help to understand the benefits of these interventions, and hopefully lead to further improved therapeutic modalities.
Collapse
Affiliation(s)
- Ellen K Quarles
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Dao-Fu Dai
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Autumn Tocchi
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Nathan Basisty
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Lemuel Gitari
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Peter S Rabinovitch
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| |
Collapse
|
41
|
Santillo M, Colantuoni A, Mondola P, Guida B, Damiano S. NOX signaling in molecular cardiovascular mechanisms involved in the blood pressure homeostasis. Front Physiol 2015. [PMID: 26217233 PMCID: PMC4493385 DOI: 10.3389/fphys.2015.00194] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Blood pressure homeostasis is maintained by several mechanisms regulating cardiac output, vascular resistances, and blood volume. At cellular levels, reactive oxygen species (ROS) signaling is involved in multiple molecular mechanisms controlling blood pressure. Among ROS producing systems, NADPH oxidases (NOXs), expressed in different cells of the cardiovascular system, are the most important enzymes clearly linked to the development of hypertension. NOXs exert a central role in cardiac mechanosensing, endothelium-dependent relaxation, and Angiotensin-II (Ang-II) redox signaling regulating vascular tone. The central role of NOXs in redox-dependent cardiovascular cell functions renders these enzymes a promising pharmacological target for the treatment of cardiovascular diseases, including hypertension. The aim of the present review is to focus on the physiological role of the cardiovascular NOX-generating ROS in the molecular and cellular mechanisms affecting blood pressure.
Collapse
Affiliation(s)
- Mariarosaria Santillo
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II" Naples, Italy
| | - Antonio Colantuoni
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II" Naples, Italy
| | - Paolo Mondola
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II" Naples, Italy
| | - Bruna Guida
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II" Naples, Italy
| | - Simona Damiano
- Dipartimento di Medicina Clinica e Chirurgia, Università di Napoli "Federico II" Naples, Italy
| |
Collapse
|
42
|
Goetze JP, Johnsen AH, Kistorp C, Gustafsson F, Johnbeck CB, Rehfeld JF. Cardiomyocyte expression and cell-specific processing of procholecystokinin. J Biol Chem 2015; 290:6837-43. [PMID: 25627687 DOI: 10.1074/jbc.m114.622670] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heart muscle cells produce peptide hormones such as natriuretic peptides. Developing hearts also express the gene for the classic intestinal hormone cholecystokinin (CCK) in amounts similar to those in the intestine and brain. However, cardiac expression of peptides other than natriuretic peptides has only been suggested using transcriptional measures or methods, with the post-translational phase of gene expression unaddressed. In this study, we examined the cardiac expression of the CCK gene in adult mammals and its expression at the protein level. Using quantitative PCR, a library of sequence-specific pro-CCK assays, peptide purification, and mass spectrometry, we demonstrate that the mammalian heart expresses pro-CCK in amounts comparable to natriuretic prohormones and processes it to a unique, triple-sulfated, and N-terminally truncated product distinct from intestinal and cerebral CCK peptides. Isoprenaline rapidly stimulated cardiac CCK gene expression in vitro and in vivo, which suggests that the cardiac-specific truncated pro-CCK may have pathophysiological relevance as a new marker of heart failure. The suggestion is confirmed by measurement of plasma from heart failure patients.
Collapse
Affiliation(s)
- Jens P Goetze
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Department of Clinical Medicine, Aarhus University, 8000 Aarhus C, Denmark the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| | - Anders H Johnsen
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| | - Caroline Kistorp
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| | - Finn Gustafsson
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| | - Camilla B Johnbeck
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| | - Jens F Rehfeld
- From the Departments of Clinical Biochemistry and Cardiology, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, the Department of Endocrinology, Herlev Hospital, University of Copenhagen, 2730 Herlev, and
| |
Collapse
|
43
|
Wang X, Guo Z, Ding Z, Khaidakov M, Lin J, Xu Z, Sharma SG, Jiwani S, Mehta JL. Endothelin-1 upregulation mediates aging-related cardiac fibrosis. J Mol Cell Cardiol 2015; 80:101-9. [PMID: 25584774 DOI: 10.1016/j.yjmcc.2015.01.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 12/30/2014] [Accepted: 01/04/2015] [Indexed: 01/30/2023]
Abstract
Endothelin-1 (ET-1) plays a major role in regulating myocardial fibrosis in several pathological conditions, such as hypertension and diabetes. Aging is an independent risk factor for myocardial fibrosis. We hypothesized that ET-1 upregulation may be a basis of enhanced collagen synthesis in the senescent fibroblasts resulting in cardiac fibrosis with aging. To examine this hypothesis, we cultured mouse cardiac fibroblasts to passage-30 (P30). β-Galactosidase activity and several other aging markers were markedly increased in P30 (vs. P3) fibroblasts, indicating that these cells were indeed undergoing senescence. Importantly, ET-1 expression was markedly upregulated in P30 (vs. P3) fibroblasts. Of note, estrogen receptor-α (ER-α), an important negative regulator of ET-1, was downregulated in P30 fibroblasts. We also studied aged (130-weeks old, female) mice hearts, and observed that ET-1 was upregulated and ER-α was downregulated in these hearts (vs. 6-week old mice hearts, female). Similar observations were made in the fibroblasts isolated from aged mice hearts. ET-1 upregulation with aging was also seen in ≈70-year old (vs. ≈30-year old) human heart sections. In concert with ET-1 upregulation, the expression of fibronectin and collagens was found to be markedly increased in P30 cardiac fibroblasts in culture, fibroblasts isolated from the aged mice hearts, and in aged human hearts. Interestingly, inhibition of ET-1 in the senescent P30 fibroblasts by 2 different strategies (the use of siRNA and the use of endothelin converting enzyme inhibitors) markedly suppressed expression of fibrosis signals. Further, treatment with synthetic ET-1 enhanced fibronectin and collagen expression in P3 cardiac fibroblasts. These observations in mice and human hearts suggest that aging-related cardiac fibrosis is, at least partially, dependent on the upregulation of ET-1.
Collapse
Affiliation(s)
- Xianwei Wang
- Central Arkansas Veterans Healthcare System, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Key Laboratory of Henan province for Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Zhikun Guo
- Key Laboratory of Henan province for Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zufeng Ding
- Central Arkansas Veterans Healthcare System, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Magomed Khaidakov
- Central Arkansas Veterans Healthcare System, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Juntang Lin
- Key Laboratory of Henan province for Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhenping Xu
- Key Laboratory of Henan province for Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Shree G Sharma
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Shahanawaz Jiwani
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jawahar L Mehta
- Central Arkansas Veterans Healthcare System, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
44
|
Hammad FT, Wheatley AM, Davis G. Bosentan normalizes the GFR response to renal nerve stimulation following reversible unilateral ureteric obstruction in the rat. Physiol Res 2014; 63:713-22. [PMID: 25157662 DOI: 10.33549/physiolres.932667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We investigated the renal response to direct renal nerve stimulation, 2 weeks following reversal of 24-h unilateral (left) ureteric obstruction. Renal nerve stimulation caused a 13-15 % fall in renal blood flow, in 4 groups of anesthetized rats following ureteric obstruction (n=9) or a sham operation (n=7) both with (n=9) and without (n=7) treatment with the mixed ET(A/B) receptor antagonist, bosentan. In the sham-operated rats, renal nerve stimulation did not change glomerular filtration rate but reduced urine flow rate (37+/-3 %, P<0.001), and absolute (38+/-4 %, P<0.001) and fractional (35+/-5 %, P<0.01) sodium excretion. Following unilateral ureteric obstruction, renal nerve stimulation increased glomerular filtration rate by 22+/-3 % (P<0.01), but reduced urine flow rate (14+/-2 %, P<0.001) and fractional sodium excretion (23+/-5 %, P<0.01). Bosentan treatment had no effect on baseline or renal responses to renal nerve stimulation in the sham group but normalized the renal response to renal nerve stimulation in the unilateral ureteric obstruction group. We conclude that 14 days after a 24-h period of unilateral ureteric obstruction there is an increase in GFR in response to direct renal nerve stimulation, which is due, in part, to the actions of endothelin at the time of obstruction.
Collapse
Affiliation(s)
- F T Hammad
- Department of Physiology, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand.
| | | | | |
Collapse
|
45
|
Richard V. [Endothelin: From discovery to pharmacotherapeutic innovations]. Presse Med 2014; 43:742-55. [PMID: 24797866 DOI: 10.1016/j.lpm.2014.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/25/2013] [Accepted: 01/20/2014] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Endothelin (ET) is a major therapeutic target in cardiopulmonary diseases. The purpose of this review is to present the main concepts concerning ET biology, its pathophysiological roles and the major pharmacological and medical advances recently developed around the concept of ET receptor blockade. METHODS Analysis of PubMed database (keywords: endothelin, endothelin receptor antagonists, pulmonary hypertension, etc.), and of abstract originating from recent international meetings. RESULTS ET is a peptide produced by vascular endothelial cells as well as by many other tissues. Both its production and its effects are activated in pathological situations associated with endothelial dysfunction. ET is characterized by a strong tropism toward tissues because of its polarized release, the strong tissue receptor density and high affinity of the receptors for the peptide. ET exerts several vascular effects, including vasoconstriction, proliferation and hypertrophy, as well as non-vascular effects, notably stimulation of cardiac hypertrophy, tissue fibrosis and inflammation. Both vascular and non-vascular effects depend on the stimulation of two receptor subtypes, ETA and ETB. ET receptor antagonists (ERA) demonstrated beneficial effects in many different pre-clinical models of cardiovascular and pulmonary diseases, and constitute a first-line treatment of patients with pulmonary arterial hypertension (PAH). Recently, the targeted search for a novel ERA led to the development of macitentan which, compared to existing ERA, show optimized tissue penetration, increased receptor affinity and in vivo pharmacological efficacy in pre-clinical models, associated with a favorable profile, in terms of hepatic safety and drug interactions. The clinical efficacy of macitentan in the treatment of PAH was recently demonstrated in the SERAPHIN trial, which contrasts with previous PAH trials because of its long duration, the high number of patients enrolled, and its primary endpoint evaluating morbidity/mortality. Results show a significant reduction of the primary composite morbidity/mortality endpoint (taking into account both progression of PAH and death) by 30 and 45% with macitentan 3 and 10mg, respectively, compared to placebo, and confirm on the large scale the favorable tolerance profile, especially at the hepatic level. CONCLUSION The extensive knowledge on the complexity of the ET system allowed the synthesis of a new antagonist optimized, in terms of pharmacological efficacy and safety, which also show promising therapeutic effects in PAH patients, with demonstrated results in a prospective study using a composite primary endpoint of morbidity-mortality.
Collapse
Affiliation(s)
- Vincent Richard
- CHU de Rouen, service de pharmacologie, unité Inserm U1096, UFR médecine pharmacie de Rouen, 76183 Rouen cedex, France.
| |
Collapse
|
46
|
Favero G, Paganelli C, Buffoli B, Rodella LF, Rezzani R. Endothelium and its alterations in cardiovascular diseases: life style intervention. BIOMED RESEARCH INTERNATIONAL 2014; 2014:801896. [PMID: 24719887 PMCID: PMC3955677 DOI: 10.1155/2014/801896] [Citation(s) in RCA: 164] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 01/11/2014] [Indexed: 01/07/2023]
Abstract
The endothelium, which forms the inner cellular lining of blood vessels and lymphatics, is a highly metabolically active organ that is involved in many physiopathological processes, including the control of vasomotor tone, barrier function, leukocyte adhesion, and trafficking and inflammation. In this review, we summarized and described the following: (i) endothelial cell function in physiological conditions and (ii) endothelial cell activation and dysfunction in the main cardiovascular diseases (such as atherosclerosis, and hypertension) and to diabetes, cigarette smoking, and aging physiological process. Finally, we presented the currently available evidence that supports the beneficial effects of physical activity and various dietary compounds on endothelial functions.
Collapse
Affiliation(s)
- Gaia Favero
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Corrado Paganelli
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Barbara Buffoli
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Luigi Fabrizio Rodella
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| | - Rita Rezzani
- Section of Anatomy and Physiopathology, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy
| |
Collapse
|
47
|
Abstract
A series of studies conducted 20 years ago, documenting the cardiac hypertrophy phenotype and its underlying signaling mechanism induced by angiotensin II (Ang II) and mechanical stress, showed a remarkable similarity between the effect of the Gαq agonist and that of mechanical forces on cardiac hypertrophy. Subsequent studies confirmed the involvement of autocrine/paracrine mechanisms, including stretch-induced release of Ang II in load-induced cardiac hypertrophy. Recent studies showed that the Ang II type 1 (AT1) receptor is also directly activated by mechanical forces, suggesting that AT1 receptors play an important role in mediating load-induced cardiac hypertrophy through both ligand- and mechanical stress-dependent mechanisms.
Collapse
Affiliation(s)
- Daniela Zablocki
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark
| | | |
Collapse
|
48
|
Correa MV, Nolly MB, Caldiz CI, de Cingolani GEC, Cingolani HE, Ennis IL. Endogenous endothelin 1 mediates angiotensin II-induced hypertrophy in electrically paced cardiac myocytes through EGFR transactivation, reactive oxygen species and NHE-1. Pflugers Arch 2013; 466:1819-30. [PMID: 24327206 DOI: 10.1007/s00424-013-1413-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 11/26/2013] [Accepted: 11/27/2013] [Indexed: 01/20/2023]
Abstract
Emerging evidence supports a key role for endothelin-1 (ET-1) and the transactivation of the epidermal growth factor receptor (EGFR) in angiotensin II (Ang II) action. We aim to determine the potential role played by endogenous ET-1, EGFR transactivation and redox-dependent sodium hydrogen exchanger-1 (NHE-1) activation in the hypertrophic response to Ang II of cardiac myocytes. Electrically paced adult cat cardiomyocytes were placed in culture and stimulated with 1 nmol l(-1) Ang II or 5 nmol l(-1) ET-1. Ang II increased ~45 % cell surface area (CSA) and ~37 % [(3)H]-phenylalanine incorporation, effects that were blocked not only by losartan (Los) but also by BQ123 (AT1 and ETA receptor antagonists, respectively). Moreover, Ang II significantly increased ET-1 messenger RNA (mRNA) expression. ET-1 similarly increased myocyte CSA and protein synthesis, actions prevented by the reactive oxygen species scavenger MPG or the NHE-1 inhibitor cariporide (carip). ET-1 increased the phosphorylation of the redox-sensitive ERK1/2-p90(RSK) kinases, main activators of the NHE-1. This effect was prevented by MPG and the antagonist of EGFR, AG1478. Ang II, ET-1 and EGF increased myocardial superoxide production (187 ± 9 %, 149 ± 8 % and 163.7 ± 6 % of control, respectively) and AG1478 inhibited these effects. Interestingly, Los inhibited only Ang II whilst BQ123 cancelled both Ang II and ET-1 actions, supporting the sequential and unidirectional activation of AT1, ETA and EGFR. Based on the present evidence, we propose that endogenous ET-1 mediates the hypertrophic response to Ang II by a mechanism that involves EGFR transactivation and redox-dependent activation of the ERK1/2-p90(RSK) and NHE-1 in adult cardiomyocytes.
Collapse
Affiliation(s)
- María V Correa
- Centro de Investigaciones Cardiovasculares, Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120 S/N, La Plata, 1900, Argentina,
| | | | | | | | | | | |
Collapse
|
49
|
Arumugam S, Mito S, Thandavarayan RA, Giridharan VV, Pitchaimani V, Karuppagounder V, Harima M, Nomoto M, Suzuki K, Watanabe K. Mulberry Leaf Diet Protects Against Progression of Experimental Autoimmune Myocarditis to Dilated Cardiomyopathy Via Modulation of Oxidative Stress and MAPK-Mediated Apoptosis. Cardiovasc Ther 2013; 31:352-62. [DOI: 10.1111/1755-5922.12029] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Somasundaram Arumugam
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Sayaka Mito
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Rajarajan A. Thandavarayan
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
- Bristol Heart Institute; University of Bristol, Bristol Royal Infirmary; Bristol UK
| | - Vijayasree V. Giridharan
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Vigneshwaran Pitchaimani
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Vengadeshprabhu Karuppagounder
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Meilei Harima
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Mayumi Nomoto
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| | - Kenji Suzuki
- Department of Gastroenterology; Niigata University Graduate School of Medical and Dental Sciences; Niigata City Japan
| | - Kenichi Watanabe
- Department of Clinical Pharmacology; Faculty of Pharmaceutical Sciences; Niigata University of Pharmacy and Applied Life Sciences; Niigata City Japan
| |
Collapse
|
50
|
Murtaza I, Wang HX, Mushtaq S, Javed Q, Li PF. Interplay of Phosphorylated Apoptosis Repressor with CARD, Casein Kinase-2 and Reactive Oxygen Species in Regulating Endothelin-1-Induced Cardiomyocyte Hypertrophy. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2013; 16:928-35. [PMID: 24106598 PMCID: PMC3786106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 03/11/2013] [Indexed: 11/22/2022]
Abstract
OBJECTIVE(S) The role of the Apoptosis repressor with caspase recruitment domain (ARC) in apoptosis and in certain hypertrophic responses has been previously investigated, but its regulation of Endothelin-1 induced cardiac hypertrophy remains unknown. The present study discusses the inhibitory role of ARC against endothelin-induced hypertrophy. RESULTS In present study Endothelin treated cardiomyocytes were used as a hypertrophic model, that were subsequently treated with adenovirus ARC and its mutant at different multiplicity of infections. Casein-kinase-2 inhibitors were used to produce dephosphorylated ARC and to study its effect on hypertrophy. Hypertrophy was assessed by cell surface area measurement, Atrial-natriuretic-Factor mRNA analysis and total protein assay. Reactive oxygen species analysis was carried out using the dichlorofluorescin-diacetate (DCFH-DA) assay. Over expression of ARC significantly inhibits Endothelin-induced cardiomyocyte hypertrophy. The nonphosphorylated mutant ARC (T149 A) remained unable to control endothelin-induced hypertrophy, suggesting a vital role for ARC phosphorylation in regulation of its activity. Sensitization study has been carried out to check the role of endogenous ARC using casein-kinase inhibitors. Finally, the significant role of ARC in regulating reactive oxygen species -mediated control of endothelin induced hypertrophy has also been assessed. Conclusion : Conclusively, present study showed the vital and potential therapeutic interventional role of ARC in preventing endothelin-1-induced cardiomyocyte hypertrophy. The regulation of hypertrophic pathway by ARC relies on blunting the reactive oxygen species attack. This study further suggests a mediatory role of casein-kinase-2 in Endothelin-induced hypertrophy, mainly through its phosphorylation of ARC.
Collapse
Affiliation(s)
- Iram Murtaza
- Division of Cardiovascular Research, National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, People's Republic of China,Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad, 45320, Islamabad, Pakistan,Corresponding author: Iram Murtaza, Department of Bio-Chemsitry, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad, 45320, Islamabad, Pakistan. Tel: +92-51-90643175; /
| | - Hong-Xia Wang
- Division of Cardiovascular Research, National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, People's Republic of China
| | - Sobia Mushtaq
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad, 45320, Islamabad, Pakistan
| | - Qamar Javed
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University Islamabad, 45320, Islamabad, Pakistan
| | - Pei-Feng Li
- Division of Cardiovascular Research, National Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100080, People's Republic of China
| |
Collapse
|