151
|
|
152
|
Fu Y, Wang C, Zhang D, Xin Y, Li J, Zhang Y, Chu X. Increased TRPC6 expression is associated with tubular epithelial cell proliferation and inflammation in diabetic nephropathy. Mol Immunol 2018; 94:75-81. [DOI: 10.1016/j.molimm.2017.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/12/2017] [Accepted: 12/15/2017] [Indexed: 01/09/2023]
|
153
|
The role of fibroblast growth factor 23 and Klotho in uremic cardiomyopathy. Curr Opin Nephrol Hypertens 2018; 25:314-24. [PMID: 27219043 DOI: 10.1097/mnh.0000000000000231] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW In chronic kidney disease (CKD), multiple factors contribute to the development of cardiac hypertrophy by directly targeting the heart or indirectly by inducing systemic changes such as hypertension, anemia, and inflammation. Furthermore, disturbances in phosphate metabolism have been identified as nonclassical risk factors for cardiovascular mortality in these patients. With declining kidney function, the physiologic regulators of phosphate homeostasis undergo changes in their activity as well as their circulating levels, thus potentially contributing to cardiac hypertrophy once they are out of balance. Recently, two of these phosphate regulators, fibroblast growth factor 23 (FGF23) and Klotho, have been shown to affect cardiac remodeling, thereby unveiling a novel pathomechanism of cardiac hypertrophy in CKD. Here we discuss the potential direct versus indirect effects of FGF23 and the soluble form of Klotho on the heart, and their crosstalk in the regulation of cardiac hypertrophy. RECENT FINDINGS In models of CKD, FGF23 can directly target cardiac myocytes via FGF receptor 4 and induce cardiac hypertrophy in a blood pressure-independent manner. Soluble Klotho may directly target the heart via an unknown receptor thereby protecting the myocardium from pathologic stress stimuli that are associated with CKD, such as uremic toxins or FGF23. SUMMARY Elevated serum levels of FGF23 and reduced serum levels of soluble Klotho contribute to uremic cardiomyopathy in a synergistic manner.
Collapse
|
154
|
Han X, Ross J, Kolumam G, Pi M, Sonoda J, King G, Quarles LD. Cardiovascular Effects of Renal Distal Tubule Deletion of the FGF Receptor 1 Gene. J Am Soc Nephrol 2018; 29:69-80. [PMID: 28993502 PMCID: PMC5748915 DOI: 10.1681/asn.2017040412] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/03/2017] [Indexed: 01/11/2023] Open
Abstract
The bone-derived hormone fibroblast growth factor-23 (FGF-23) activates complexes composed of FGF receptors (FGFRs), including FGFR1, and α-Klotho in the kidney distal tubule (DT), leading to increased sodium retention and hypertension. However, the role of FGFR1 in regulating renal processes linked to hypertension is unclear. Here, we investigated the effects of selective FGFR1 loss in the DT. Conditional knockout (cKO) of FGFR1 in the DT (FGFR1DT-cKO mice) resulted in left ventricular hypertrophy (LVH) and decreased kidney expression of α-Klotho in association with enhanced BP, decreased expression of angiotensin converting enzyme 2, and increased expression of the Na+-K+-2Cl- cotransporter. Notably, recombinant FGF-23 administration similarly decreased the kidney expression of α-Klotho and induced LVH in mice. Pharmacologic activation of FGFR1 with a monoclonal anti-FGFR1 antibody (R1MAb1) normalized BP and significantly attenuated LVH in the Hyp mouse model of excess FGF-23, but did not induce a response in FGFR1DT-cKO mice. The hearts of FGFR1DT-cKO mice showed increased expression of the transient receptor potential cation channel, subfamily C, member 6 (TRPC6), consistent with cardiac effects of soluble Klotho deficiency. Moreover, administration of recombinant soluble Klotho lowered BP in the Hyp mice. Thus, FGFR1 in the DT regulates systemic hemodynamic responses opposite to those predicted by the actions of FGF-23. These cardiovascular effects appear to be mediated by paracrine FGF control of kidney FGFR1 and subsequent regulation of soluble Klotho and TRPC6. FGFR1 in the kidney may provide a new molecular target for treating hypertension.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2
- Animals
- Antibodies, Monoclonal/pharmacology
- Blood Pressure/drug effects
- Blood Pressure/genetics
- Female
- Fibroblast Growth Factor-23
- Fibroblast Growth Factors/pharmacology
- Glucuronidase/genetics
- Glucuronidase/metabolism
- Hypertension/genetics
- Hypertrophy, Left Ventricular/genetics
- Immunologic Factors/pharmacology
- Kidney Tubules, Distal
- Klotho Proteins
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardium/metabolism
- Peptidyl-Dipeptidase A/genetics
- Peptidyl-Dipeptidase A/metabolism
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/immunology
- Recombinant Proteins/pharmacology
- Sodium-Potassium-Chloride Symporters/genetics
- Sodium-Potassium-Chloride Symporters/metabolism
- TRPC Cation Channels/genetics
- TRPC Cation Channels/metabolism
- TRPC6 Cation Channel
Collapse
Affiliation(s)
- Xiaobin Han
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jed Ross
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Ganesh Kolumam
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Min Pi
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Junichiro Sonoda
- Department of Molecular Biology and Biomedical Imaging, Genentech, South San Francisco, California; and
| | - Gwendalyn King
- Department of Neurobiology, University of Alabama in Birmingham, Birmingham, Alabama
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee;
| |
Collapse
|
155
|
Ohba T, Watanabe H, Murakami M, Iino K, Adachi T, Baba Y, Kurosaki T, Ono K, Ito H. Stromal interaction molecule 1 haploinsufficiency causes maladaptive response to pressure overload. PLoS One 2017; 12:e0187950. [PMID: 29145451 PMCID: PMC5690472 DOI: 10.1371/journal.pone.0187950] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/14/2017] [Indexed: 11/18/2022] Open
Abstract
Stromal interaction molecule 1 (STIM1), an endo/sarcoplasmic reticulum Ca2+ sensor, has been shown to control a Ca2+-dependent signal that promotes cardiac hypertrophy. However, whether STIM1 has adaptive role that helps to protect against cardiac overload stress remains unknown. We hypothesized that STIM1 deficiency causes a maladaptive response to pressure overload stress. We investigated STIM1 heterozygous KO (STIM1+/–) mice hearts, in which STIM1 protein levels decreased to 27% of wild-type (WT) with no compensatory increase in STIM2. Under stress-free conditions, no significant differences were observed in electrocardiographic and echocardiographic parameters or blood pressure between STIM1+/–and WT mice. However, when STIM1+/–mice were subjected to transverse aortic constriction (TAC), STIM1+/–mice had a higher mortality rate than WT mice. The TAC-induced increase in the heart weight to body weight ratio (mean mg/g ± standard error of the mean) was significantly inhibited in STIM1+/–mice (WT sham, 4.12 ± 0.14; WT TAC, 6.23 ± 0.40; STIM1+/–sham, 4.53 ± 0.16; STIM1+/–TAC, 4.63 ± 0.08). Reverse transcription-polymerase chain reaction analysis of the left ventricles of TAC-treated STIM1+/–mice showed inhibited induction of cardiac fetal genes, including those encoding brain and atrial natriuretic proteins. Western blot analysis showed upregulated expression of transient receptor potential channel 1 (TRPC1) in TAC-treated WT mice, but suppressed expression in TAC-treated STIM1+/–mice. Taken together, the hearts of STIM1 haploinsufficient mice had a superficial resemblance to the WT phenotype under stress-free conditions; however, STIM1 haploinsufficient mice showed a maladaptive response to cardiac pressure overload.
Collapse
Affiliation(s)
- Takayoshi Ohba
- Department of Cell Physiology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroyuki Watanabe
- Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
- * E-mail:
| | - Manabu Murakami
- Department of Pharmacology, Hirosaki University, Graduate School of Medicine, Aomori, Japan
| | - Kenji Iino
- Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| | - Takeshi Adachi
- Department of Cell Physiology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yoshihiro Baba
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory for Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Kyoichi Ono
- Department of Cell Physiology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroshi Ito
- Department of Cardiovascular and Respiratory Medicine, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
156
|
Adding dimension to cellular mechanotransduction: Advances in biomedical engineering of multiaxial cell-stretch systems and their application to cardiovascular biomechanics and mechano-signaling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [DOI: 10.1016/j.pbiomolbio.2017.06.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
157
|
Nikolova-Krstevski V, Wagner S, Yu ZY, Cox CD, Cvetkovska J, Hill AP, Huttner IG, Benson V, Werdich AA, MacRae C, Feneley MP, Friedrich O, Martinac B, Fatkin D. Endocardial TRPC-6 Channels Act as Atrial Mechanosensors and Load-Dependent Modulators of Endocardial/Myocardial Cross-Talk. ACTA ACUST UNITED AC 2017; 2:575-590. [PMID: 30062171 PMCID: PMC6058914 DOI: 10.1016/j.jacbts.2017.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/05/2017] [Accepted: 05/22/2017] [Indexed: 12/01/2022]
Abstract
Mechanoelectrical feedback may increase arrhythmia susceptibility, but the molecular mechanisms are incompletely understood. This study showed that mechanical stretch altered the localization, protein levels, and function of the cation-selective transient receptor potential channel (TRPC)-6 in atrial endocardial cells in humans, pigs, and mice. In endocardial/myocardial cross-talk studies, addition of media from porcine atrial endocardium (AE) cells altered the calcium (Ca2+) transient characteristics of human-induced pluripotent stem cell-derived cardiomyocytes. These changes did not occur with media from stretched AE cells. Our data suggested that endocardial TRPC-6-dependent paracrine signaling may modulate myocardial Ca2+ homeostasis under basal conditions and protect against stretch-induced atrial arrhythmias.
Collapse
Key Words
- AE, atrial endocardium
- AF, atrial fibrillation
- APB, aminoethoxydiphenyl borate
- Ab, antibody
- CM, cardiomyocyte
- Ca2+, calcium
- Dil-Ac-LDL, dil acetylated−low-density lipoprotein
- ET, endothelin
- HUVEC, human umbilical vein endothelial cell
- OAG, 1-oleoyl-2-acetyl-sn-glycerol
- TAC, thoracic aortic constriction
- TRPC, transient receptor potential channel
- Tet, tetanus toxin
- [Ca2+]i, intracellular global Ca2+
- atrial endocardium
- endothelium
- iPS, induced pluripotent stem
- mechanical stretch
- transient receptor potential channels
Collapse
Affiliation(s)
- Vesna Nikolova-Krstevski
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Soeren Wagner
- Department of Anesthesiology, University Clinic Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Ze Yan Yu
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Charles D Cox
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Jasmina Cvetkovska
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia
| | - Adam P Hill
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Inken G Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Victoria Benson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andreas A Werdich
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Calum MacRae
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Michael P Feneley
- St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia.,Cardiac Physiology and Transplantation Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Cardiology Department, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
| | - Oliver Friedrich
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Boris Martinac
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Kensington, New South Wales, Australia.,Cardiology Department, St. Vincent's Hospital, Darlinghurst, New South Wales, Australia
| |
Collapse
|
158
|
Qu Z, Wang Y, Li X, Wu L, Wang Y. TRPC6 expression in neurons is differentially regulated by NR2A- and NR2B-containing NMDA receptors. J Neurochem 2017; 143:282-293. [PMID: 28902407 DOI: 10.1111/jnc.14215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 08/30/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The expression of transient receptor potential canonical 6 (TRPC6) in central nervous system (CNS) is important for neuronal functions and certain neural disorders. However, the regulatory mechanism of TRPC6 expression in neurons is still obscure. In this study, we show that TRPC6 expression in the primary cultured cortical neurons is bidirectionally regulated by glutamate. Activation of NR2A-containing NMDARs induces TRPC6 transcription through a calcineurin-dependent pathway. In contrast, activation of NR2B-containing NMDARs causes TRPC6 degradation through calpain. Thus, TRPC6 expression in neurons is regulated by glutamate in a bidirectional manner that is dependent on NR2A and NR2B.
Collapse
Affiliation(s)
- Zhongwei Qu
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,The Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Xia Li
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Lin Wu
- Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| | - Yizheng Wang
- Laboratory of Neural Signal Transduction, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China.,Center of Cognition and Brain Science, Beijing Institute of Medical Sciences, Beijing, China
| |
Collapse
|
159
|
Thiel G, Rubil S, Lesch A, Guethlein LA, Rössler OG. Transient receptor potential TRPM3 channels: Pharmacology, signaling, and biological functions. Pharmacol Res 2017; 124:92-99. [DOI: 10.1016/j.phrs.2017.07.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/12/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022]
|
160
|
Dewenter M, von der Lieth A, Katus HA, Backs J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ Res 2017; 121:1000-1020. [DOI: 10.1161/circresaha.117.310355] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium (Ca
2+
) is a universal regulator of various cellular functions. In cardiomyocytes, Ca
2+
is the central element of excitation–contraction coupling, but also impacts diverse signaling cascades and influences the regulation of gene expression, referred to as excitation–transcription coupling. Disturbances in cellular Ca
2+
-handling and alterations in Ca
2+
-dependent gene expression patterns are pivotal characteristics of failing cardiomyocytes, with several excitation–transcription coupling pathways shown to be critically involved in structural and functional remodeling processes. Thus, targeting Ca
2+
-dependent transcriptional pathways might offer broad therapeutic potential. In this article, we (1) review cytosolic and nuclear Ca
2+
dynamics in cardiomyocytes with respect to their impact on Ca
2+
-dependent signaling, (2) give an overview on Ca
2+
-dependent transcriptional pathways in cardiomyocytes, and (3) discuss implications of excitation–transcription coupling in the diseased heart.
Collapse
Affiliation(s)
- Matthias Dewenter
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Albert von der Lieth
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Hugo A. Katus
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| | - Johannes Backs
- From the Department of Molecular Cardiology and Epigenetics (M.D., A.v.d.L., J.B.) and Department of Cardiology (H.A.K.), Heidelberg University, Germany; and DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (M.D., A.v.d.L., H.A.K., J.B.)
| |
Collapse
|
161
|
Abstract
This chapter offers a brief introduction of the functions of TRPC channels in non-neuronal systems. We focus on three major organs of which the research on TRPC channels have been most focused on: kidney, heart, and lung. The chapter highlights on cellular functions and signaling pathways mediated by TRPC channels. It also summarizes several inherited diseases in humans that are related to or caused by TRPC channel mutations and malfunction. A better understanding of TRPC channels functions and the importance of TRPC channels in health and disease should lead to new insights and discovery of new therapeutic approaches for intractable disease.
Collapse
|
162
|
Numaga-Tomita T, Oda S, Shimauchi T, Nishimura A, Mangmool S, Nishida M. TRPC3 Channels in Cardiac Fibrosis. Front Cardiovasc Med 2017; 4:56. [PMID: 28936433 PMCID: PMC5594069 DOI: 10.3389/fcvm.2017.00056] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/21/2017] [Indexed: 01/18/2023] Open
Abstract
Cardiac stiffness, caused by interstitial fibrosis due to deposition of extracellular matrix proteins, is thought as a major clinical outcome of heart failure with preserved ejection fraction (HFpEF). Canonical transient receptor potential (TRPC) subfamily proteins are components of Ca2+-permeable non-selective cation channels activated by receptor stimulation and mechanical stress, and have been attracted attention as a key mediator of maladaptive cardiac remodeling. How TRPC-mediated local Ca2+ influx encodes a specific signal to induce maladaptive cardiac remodeling has been long obscure, but our recent studies suggest a pathophysiological significance of channel activity-independent function of TRPC proteins for amplifying redox signaling in heart. This review introduces the current understanding of the physiological and pathophysiological roles of TRPCs, especially focuses on the role of TRPC3 as a positive regulator of reactive oxygen species (PRROS) in heart. We have revealed that TRPC3 stabilizes NADPH oxidase 2 (Nox2), a membrane-bound reactive oxygen species (ROS)-generating enzyme, by forming stable protein complex with Nox2, which leads to amplification of mechanical stress-induced ROS signaling in cardiomyocytes, resulting in induction of fibrotic responses in cardiomyocytes and cardiac fibroblasts. Thus, the TRPC3 function as PRROS will offer a new therapeutic strategy for the prevention or treatment of HFpEF.
Collapse
Affiliation(s)
- Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Sayaka Oda
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Tsukasa Shimauchi
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiyuki Nishimura
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan
| | - Supachoke Mangmool
- Faculty of Pharmacy, Department of Pharmacology, Mahidol University, Bangkok, Thailand
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, Graduate University for Advanced Studies (SOKENDAI), Okazaki, Japan.,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan.,Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency, Kawaguchi, Japan
| |
Collapse
|
163
|
Hagmann H, Mangold N, Rinschen MM, Koenig T, Kunzelmann K, Schermer B, Benzing T, Brinkkoetter PT. Proline-dependent and basophilic kinases phosphorylate human TRPC6 at serine 14 to control channel activity through increased membrane expression. FASEB J 2017; 32:208-219. [PMID: 28877958 DOI: 10.1096/fj.201700309r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/21/2017] [Indexed: 01/01/2023]
Abstract
Signaling via the transient receptor potential (TRP) ion channel C6 plays a pivotal role in hereditary and sporadic glomerular kidney disease. Several studies have identified gain-of-function mutations of TRPC6 and report induced expression and enhanced channel activity of TRPC6 in association with glomerular diseases. Interfering with TRPC6 activity may open novel therapeutic pathways. TRPC6 channel activity is controlled by protein expression and stability as well as intracellular trafficking. Identification of regulatory phosphorylation sites in TRPC6 and corresponding protein kinases is essential to understand the regulation of TRPC6 activity and may result in future therapeutic strategies. In this study, an unbiased phosphoproteomic screen of human TRPC6 identified several novel serine phosphorylation sites. The phosphorylation site at serine 14 of TRPC6 is embedded in a basophilic kinase motif that is highly conserved across species. We confirmed serine 14 as a target of MAPKs and proline-directed kinases like cyclin-dependent kinase 5 (Cdk5) in cell-based as well as in vitro kinase assays and quantitative phosphoproteomic analysis of TRPC6. Phosphorylation of TRPC6 at serine 14 enhances channel conductance by boosting membrane expression of TRPC6, whereas protein stability and multimerization of TRPC6 are not altered, making serine 14 phosphorylation a potential drug target to interfere with TRPC6 channel activity.-Hagmann, H., Mangold, N., Rinschen, M. M., Koenig, T., Kunzelmann, K., Schermer, B., Benzing, T., Brinkkoetter, P. T. Proline-dependent and basophilic kinases phosphorylate human TRPC6 at serine 14 to control channel activity through increased membrane expression.
Collapse
Affiliation(s)
- Henning Hagmann
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Nicole Mangold
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Markus M Rinschen
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Tim Koenig
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute for Genetics Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; and
| | - Karl Kunzelmann
- Department of Physiology, University Regensburg, Regensburg, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Systems Biology of Ageing Cologne (Sybacol), University of Cologne, Cologne, Germany
| | - Paul T Brinkkoetter
- Department II of Internal Medicine, Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany;
| |
Collapse
|
164
|
Xiao X, Liu HX, Shen K, Cao W, Li XQ. Canonical Transient Receptor Potential Channels and Their Link with Cardio/Cerebro-Vascular Diseases. Biomol Ther (Seoul) 2017; 25:471-481. [PMID: 28274093 PMCID: PMC5590790 DOI: 10.4062/biomolther.2016.096] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 12/04/2016] [Accepted: 12/27/2016] [Indexed: 12/29/2022] Open
Abstract
The canonical transient receptor potential channels (TRPCs) constitute a series of nonselective cation channels with variable degrees of Ca2+ selectivity. TRPCs consist of seven mammalian members, TRPC1, TRPC2, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7, which are further divided into four subtypes, TRPC1, TRPC2, TRPC4/5, and TRPC3/6/7. These channels take charge of various essential cell functions such as contraction, relaxation, proliferation, and dysfunction. This review, organized into seven main sections, will provide an overview of current knowledge about the underlying pathogenesis of TRPCs in cardio/cerebrovascular diseases, including hypertension, pulmonary arterial hypertension, cardiac hypertrophy, atherosclerosis, arrhythmia, and cerebrovascular ischemia reperfusion injury. Collectively, TRPCs could become a group of drug targets with important physiological functions for the therapy of human cardio/cerebro-vascular diseases.
Collapse
Affiliation(s)
- Xiong Xiao
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Hui-Xia Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.,Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Kuo Shen
- Cadet Brigade, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Cao
- Department of Natural Medicine & Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Qiang Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
165
|
Thiel G, Lesch A, Rubil S, Backes TM, Rössler OG. Regulation of Gene Transcription Following Stimulation of Transient Receptor Potential (TRP) Channels. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:167-189. [PMID: 29305012 DOI: 10.1016/bs.ircmb.2017.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Transient receptor potential (TRP) channels belong to a heterogeneous superfamily of cation channels that are involved in the regulation of numerous biological functions, including regulation of Ca2+ and glucose homeostasis, tumorigenesis, temperature, and pain sensation. To understand the functions of TRP channels, their associated intracellular signaling pathways and molecular targets have to be identified on the cellular level. Stimulation of TRP channels frequently induces an influx of Ca2+ ions into the cells and the subsequent activation of protein kinases. These intracellular signal transduction pathways ultimately induce changes in the gene expression pattern of the cells. Here, we review the effects of TRPC6, TRPM3, and TRPV1 channel stimulation on the activation of the stimulus-responsive transcription factors AP-1, CREB, Egr-1, Elk-1, and NFAT. Following activation, these transcription factors induce the transcription of delayed response genes. We propose that many biological functions of TRP channels can be explained by the activation of stimulus-responsive transcription factors and their delayed response genes. The proteins encoded by those delayed response genes may be responsible for the biochemical and physiological changes following TRP channel activation.
Collapse
Affiliation(s)
- Gerald Thiel
- Saarland University Medical Faculty, Homburg, Germany.
| | - Andrea Lesch
- Saarland University Medical Faculty, Homburg, Germany
| | - Sandra Rubil
- Saarland University Medical Faculty, Homburg, Germany
| | | | | |
Collapse
|
166
|
Oda S, Numaga-Tomita T, Kitajima N, Toyama T, Harada E, Shimauchi T, Nishimura A, Ishikawa T, Kumagai Y, Birnbaumer L, Nishida M. TRPC6 counteracts TRPC3-Nox2 protein complex leading to attenuation of hyperglycemia-induced heart failure in mice. Sci Rep 2017; 7:7511. [PMID: 28790356 PMCID: PMC5548754 DOI: 10.1038/s41598-017-07903-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/04/2017] [Indexed: 12/19/2022] Open
Abstract
Excess production of reactive oxygen species (ROS) caused by hyperglycemia is a major risk factor for heart failure. We previously reported that transient receptor potential canonical 3 (TRPC3) channel mediates pressure overload-induced maladaptive cardiac fibrosis by forming stably functional complex with NADPH oxidase 2 (Nox2). Although TRPC3 has been long suggested to form hetero-multimer channels with TRPC6 and function as diacylglycerol-activated cation channels coordinately, the role of TRPC6 in heart is still obscure. We here demonstrated that deletion of TRPC6 had no impact on pressure overload-induced heart failure despite inhibiting interstitial fibrosis in mice. TRPC6-deficient mouse hearts 1 week after transverse aortic constriction showed comparable increases in fibrotic gene expressions and ROS production but promoted inductions of inflammatory cytokines, compared to wild type hearts. Treatment of TRPC6-deficient mice with streptozotocin caused severe reduction of cardiac contractility with enhancing urinary and cardiac lipid peroxide levels, compared to wild type and TRPC3-deficient mice. Knockdown of TRPC6, but not TRPC3, enhanced basal expression levels of cytokines in rat cardiomyocytes. TRPC6 could interact with Nox2, but the abundance of TRPC6 was inversely correlated with that of Nox2. These results strongly suggest that Nox2 destabilization through disrupting TRPC3-Nox2 complex underlies attenuation of hyperglycemia-induced heart failure by TRPC6.
Collapse
Affiliation(s)
- Sayaka Oda
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi, 444-8787, Japan
| | - Takuro Numaga-Tomita
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi, 444-8787, Japan
| | - Naoyuki Kitajima
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Takashi Toyama
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.,Environmental Biology Laboratory, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Eri Harada
- Ajinomoto Co. Inc., Tokyo, 104-8315, Japan.,EA Pharma Co., Ltd., Tokyo, 104-0042, Japan
| | - Tsukasa Shimauchi
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Akiyuki Nishimura
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan.,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi, 444-8787, Japan
| | - Tatsuya Ishikawa
- Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.,Ajinomoto Co. Inc., Tokyo, 104-8315, Japan.,EA Pharma Co., Ltd., Tokyo, 104-0042, Japan
| | - Yoshito Kumagai
- Environmental Biology Laboratory, Faculty of Medicine and Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, 305-8575, Japan
| | - Lutz Birnbaumer
- Laboratory of Neuroscience, NIEHS, NIH, Research Triangle Park, NC, 27709, USA.,Institute for Biomedical Research (BIOMED), Catholic University of Argentina, C1107AFF, Buenos, Aires, Argentina
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, National Institute for Physiological Sciences (Okazaki Institute for Integrative Bioscience), National Institutes of Natural Sciences, Aichi, 444-8787, Japan. .,Department of Physiological Sciences, SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi, 444-8787, Japan. .,Department of Translational Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan. .,PRESTO, JST, 4-1-8 Honcho, Kawaguchi, Saitama, 332-0012, Japan.
| |
Collapse
|
167
|
Li S, Pan Y, Ke R, Xie X, Zhai C, Shi W, Wang J, Yan X, Chai L, Wang Q, Zhang Q, Su X, Yang L, Gao L, Li M. Inhibition of phosphodiesterase-5 suppresses calcineurin/NFAT- mediated TRPC6 expression in pulmonary artery smooth muscle cells. Sci Rep 2017; 7:6088. [PMID: 28729555 PMCID: PMC5519690 DOI: 10.1038/s41598-017-06350-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 06/13/2017] [Indexed: 02/07/2023] Open
Abstract
The up-regulation of transient receptor potential channel 6 (TRPC6) has been found to contribute to the proliferation of pulmonary artery smooth muscle cells (PASMCs), and inhibition of phosphodiesterase-5 (PDE5) has been shown to suppress TRPC6 expression in PASMCs. However, the molecular mechanisms underlying the up-regulation of TRPC6 expression and PDE5 modulation of TRPC6 expression in PASMCs remain largely unclear. The aim of this study is to address these issues. Endothelin-1 (ET-1) dose and time-dependently up-regulated TRPC6 expression in primary cultured rat PASMCs, and this was accompanied with the activation of calcineurin and subsequent translocation of NFATc4 to the nucleus. Further study indicated that inhibition of calcineurin by cyclosporine A or knockdown of NFATc4 using small interfering RNA suppressed ET-1-induced TRPC6 up-regulation. In addition, luciferase reporter assay showed that NFATc4 directly regulated the expression of TRPC6 in PASMCs. Inhibition of PDE5 by sildenafil suppressed ET-1-induced activation of calcineurin/NFATc4 signaling pathway and consequent TRPC6 up-regulation in PASMCs, while these inhibitory effects of sildenafil were abolished by PKG inhibitor Rp-8Br-cGMPs. Taken together, our study indicates that ET-1 stimulates TRPC6 expression by activation of calcineurin/NFATc4 signaling pathway, and inhibition of PDE5 suppresses calcineurin/NFATc4- mediated TRPC6 expression in PASMCs in a cGMP-PKG-dependent manner.
Collapse
Affiliation(s)
- Shaojun Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Yilin Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Rui Ke
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Cui Zhai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Wenhua Shi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Jian Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Xin Yan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Limin Chai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qingting Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qianqian Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Xiaofan Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Li Gao
- Division of Allergy and Clinical Immunology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, 21224, USA
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| |
Collapse
|
168
|
Can the Drosophila model help in paving the way for translational medicine in heart failure? Biochem Soc Trans 2017; 44:1549-1560. [PMID: 27911738 DOI: 10.1042/bst20160017c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 01/09/2023]
Abstract
Chronic heart failure is a common consequence of various heart diseases. Mechanical force is known to play a key role in heart failure development through regulating cardiomyocyte hypertrophy. In order to understand the complex disease mechanism, this article discussed a multi-disciplinary approach that may aid the illustration of heart failure molecular process.
Collapse
|
169
|
Role of TRPC3 and TRPC6 channels in the myocardial response to stretch: Linking physiology and pathophysiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [PMID: 28645743 DOI: 10.1016/j.pbiomolbio.2017.06.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Transient receptor potential (TRP) channels constitute a large family of versatile multi-signal transducers. In particular, TRP canonical (TRPC) channels are known as receptor-operated, non-selective cation channels. TRPC3 and TRPC6, two members in the TRPC family, are highly expressed in the heart, and participate in the pathogenesis of cardiac hypertrophy and heart failure as a pathological response to chronic mechanical stress. In the pathological response, myocardial stretch increases intracellular Ca2+ levels and activates nuclear factor of activated T cells to induce cardiac hypertrophy. Recent studies have revealed that TRPC3 and TRPC6 also contribute to the physiological stretch-induced slow force response (SFR), a slow increase in the Ca2+ transient and twitch force during stretch. In the physiological response, a stretch-induced increase in intracellular Ca2+ mediated by TRPC3 and TRPC6 causes the SFR. We here overview experimental evidence of the involvement of TRPC3 and TRPC6 in cardiac physiology and pathophysiology in response to stretch.
Collapse
|
170
|
Ahmad AA, Streiff M, Hunter C, Hu Q, Sachse FB. Physiological and pathophysiological role of transient receptor potential canonical channels in cardiac myocytes. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [PMID: 28629808 DOI: 10.1016/j.pbiomolbio.2017.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Transient receptor potential canonical (TRPC) channels constitute a family of seven Ca2+ permeable ion channels, named TRPC1 to 7. These channels are abundantly expressed in the mammalian heart, yet mechanisms underlying activation of TRPC channels and their precise role in cardiac physiology remain poorly understood. In this review, we perused original literature regarding TRPC channels in cardiomyocytes. We first reviewed studies on TRPC channel assembly and sub-cellular localization across multiple species and cell types. Our review indicates that TRPC localization in cardiac cells is still a topic of controversy. We then examined common molecular biology tools used to infer on location and physiological roles of TRPC channels in the heart. We subsequently reviewed pharmacological tools used to modulate TRPC activity in both cardiac and non-cardiac cells. Suggested physiological roles in the heart include modulation of heart rate and sensing of mechanical strain. We examined studies on the contribution of TRPC to cardiac pathophysiology, mainly hypertrophic signaling. Several TRPC channels, particularly TRPC1, 3 and 6 were proposed to play a crucial role in hypertrophic signaling. Finally, we discussed gaps in our understanding of the location and physiological role of TRPC channels in cardiomyocytes. Closing these gaps will be crucial to gain a full understanding of the role of TRPC channels in cardiac pathophysiology and to further explore these channels as targets for treatments for cardiac diseases, in particular, hypertrophy.
Collapse
Affiliation(s)
- Azmi A Ahmad
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA; Bioengineering Department, University of Utah, Salt Lake City, USA
| | - Molly Streiff
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA; Bioengineering Department, University of Utah, Salt Lake City, USA
| | - Chris Hunter
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA
| | - Qinghua Hu
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA
| | - Frank B Sachse
- Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, Salt Lake City, USA; Bioengineering Department, University of Utah, Salt Lake City, USA.
| |
Collapse
|
171
|
Alonso-Carbajo L, Kecskes M, Jacobs G, Pironet A, Syam N, Talavera K, Vennekens R. Muscling in on TRP channels in vascular smooth muscle cells and cardiomyocytes. Cell Calcium 2017; 66:48-61. [PMID: 28807149 DOI: 10.1016/j.ceca.2017.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 02/07/2023]
Abstract
The human TRP protein family comprises a family of 27 cation channels with diverse permeation and gating properties. The common theme is that they are very important regulators of intracellular Ca2+ signaling in diverse cell types, either by providing a Ca2+ influx pathway, or by depolarising the membrane potential, which on one hand triggers the activation of voltage-gated Ca2+ channels, and on the other limits the driving force for Ca2+ entry. Here we focus on the role of these TRP channels in vascular smooth muscle and cardiac striated muscle. We give an overview of highlights from the recent literature, and highlight the important and diverse roles of TRP channels in the pathophysiology of the cardiovascular system. The discovery of the superfamily of Transient Receptor Potential (TRP) channels has significantly enhanced our knowledge of multiple signal transduction mechanisms in cardiac muscle and vascular smooth muscle cells (VSMC). In recent years, multiple studies have provided evidence for the involvement of these channels, not only in the regulation of contraction, but also in cell proliferation and remodeling in pathological conditions. The mammalian family of TRP cation channels is composed by 28 genes which can be divided into 6 subfamilies groups based on sequence similarity: TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipins), TRPV (Vanilloid), TRPP (Policystin) and TRPA (Ankyrin-rich protein). Functional TRP channels are believed to form four-unit complexes in the plasma, each of them expressed with six transmembrane domain and intracellular N and C termini. Here we review the current knowledge on the expression of TRP channels in both muscle types, and discuss their functional properties and role in physiological and pathophysiological processes.
Collapse
Affiliation(s)
- Lucía Alonso-Carbajo
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Miklos Kecskes
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Griet Jacobs
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Andy Pironet
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Ninda Syam
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Karel Talavera
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| | - Rudi Vennekens
- Laboratory of Ion Channel Research, TRP Research Platform Leuven (TRPLe), Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
172
|
Gopalakrishnan K, More AS, Hankins GD, Nanovskaya TN, Kumar S. Postnatal Cardiovascular Consequences in the Offspring of Pregnant Rats Exposed to Smoking and Smoking Cessation Pharmacotherapies. Reprod Sci 2017; 24:919-933. [PMID: 27733658 PMCID: PMC5933098 DOI: 10.1177/1933719116673199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Approximately 20% of pregnant women smoke despite intentions to quit. Smoking cessation drugs, such as nicotine replacement therapy (NRT) and bupropion, are recommended treatments. Adverse cardiovascular outcomes in offspring have raised concerns about NRT's safety during pregnancy. However, the effect of bupropion is unknown. Using a rat model, we determined whether NRT and bupropion interventions during pregnancy are safer than continued smoking on offspring's cardiovascular function. Male offspring of controls and dams exposed to cigarette smoke (1.6 packs/day, inhalation), nicotine (2 mg/kg/d subcutaneously), and bupropion (13 mg/kg twice daily orally) were assessed for fetoplacental weight, cardiac function, blood pressure, and vascular reactivity. Fetoplacental weights were decreased and spontaneous beating and intracellular calcium in neonatal cardiomyocytes were increased in smoking, nicotine, and bupropion offspring; however, these effects were more accentuated in smoking followed by nicotine and bupropion offspring. Increased heart rate and decreased cardiac output, stroke volume, and left ventricular percent posterior wall thickening were observed in smoking, nicotine, and bupropion offspring. The left ventricular mass was reduced in smoking and nicotine but not in bupropion offspring. Blood pressure was higher with decreased endothelium-dependent relaxation and exaggerated vascular contraction to angiotensin II in smoking and nicotine offspring, with more pronounced dysfunctions in smoking than nicotine offspring. Maternal bupropion did not impact offspring's blood pressure, endothelium-dependent relaxation, and vascular contraction. In conclusion, maternal nicotine intervention adversely affects offspring's cardiovascular outcomes, albeit less severely than continued smoking. However, bupropion causes cardiac derangement in offspring but does not adversely affect blood pressure and vascular function.
Collapse
Affiliation(s)
- Kathirvel Gopalakrishnan
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Amar S. More
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Gary D. Hankins
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Tatiana N. Nanovskaya
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Sathish Kumar
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
173
|
Major contribution of the 3/6/7 class of TRPC channels to myocardial ischemia/reperfusion and cellular hypoxia/reoxygenation injuries. Proc Natl Acad Sci U S A 2017; 114:E4582-E4591. [PMID: 28526717 DOI: 10.1073/pnas.1621384114] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The injury phase after myocardial infarcts occurs during reperfusion and is a consequence of calcium release from internal stores combined with calcium entry, leading to cell death by apoptopic and necrotic processes. The mechanism(s) by which calcium enters cells has(ve) not been identified. Here, we identify canonical transient receptor potential channels (TRPC) 3 and 6 as the cation channels through which most of the damaging calcium enters cells to trigger their death, and we describe mechanisms activated during the injury phase. Working in vitro with H9c2 cardiomyoblasts subjected to 9-h hypoxia followed by 6-h reoxygenation (H/R), and analyzing changes occurring in areas-at-risk (AARs) of murine hearts subjected to a 30-min ischemia followed by 24-h reperfusion (I/R) protocol, we found: (i) that blocking TRPC with SKF96365 significantly ameliorated damage induced by H/R, including development of the mitochondrial permeability transition and proapoptotic changes in Bcl2/BAX ratios; and (ii) that AAR tissues had increased TUNEL+ cells, augmented Bcl2/BAX ratios, and increased p(S240)NFATc3, p(S473)AKT, p(S9)GSK3β, and TRPC3 and -6 proteins, consistent with activation of a positive-feedback loop in which calcium entering through TRPCs activates calcineurin-mediated NFATc3-directed transcription of TRPC genes, leading to more Ca2+ entry. All these changes were markedly reduced in mice lacking TRPC3, -6, and -7. The changes caused by I/R in AAR tissues were matched by those seen after H/R in cardiomyoblasts in all aspects except for p-AKT and p-GSK3β, which were decreased after H/R in cardiomyoblasts instead of increased. TRPC should be promising targets for pharmacologic intervention after cardiac infarcts.
Collapse
|
174
|
Weber EW, Muller WA. Roles of transient receptor potential channels in regulation of vascular and epithelial barriers. Tissue Barriers 2017; 5:e1331722. [PMID: 28581893 DOI: 10.1080/21688370.2017.1331722] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transient receptor potential (TRP) channels are a ubiquitously expressed multi-family group of cation channels that are critical to signaling events in many tissues. Their roles have been documented in many physiologic and pathologic conditions. Nevertheless, direct studies of their roles in maintain barrier function in endothelial and epithelia are relatively infrequent. This seems somewhat surprising considering that calcium ion concentrations are known to regulate barrier function. This short review provides an introduction to TRP channels and reviews some of the work in which investigators directly studied the role of TRP channels in endothelial permeability to electric current, solute, or leukocytes during the inflammatory response.
Collapse
Affiliation(s)
- Evan W Weber
- a Stanford Cancer Institute, Stanford University School of Medicine, Lokey Stem Cell Research Building , Stanford , CA , USA
| | - William A Muller
- b Northwestern University , Feinberg School of Medicine , Chicago , IL , USA
| |
Collapse
|
175
|
Peana D, Domeier TL. Cardiomyocyte Ca 2+ homeostasis as a therapeutic target in heart failure with reduced and preserved ejection fraction. Curr Opin Pharmacol 2017; 33:17-26. [PMID: 28437711 DOI: 10.1016/j.coph.2017.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 03/22/2017] [Indexed: 12/28/2022]
Abstract
Heart failure is a highly prevalent syndrome of multiple etiologies and associated comorbidities, and aberrant intracellular Ca2+ homeostasis is a hallmark finding in heart failure patients. The cyclical changes in Ca2+ concentration within cardiomyocytes control cycles of cardiac contraction and relaxation, and dysregulation of Ca2+ handling processes leads to systolic dysfunction, diastolic dysfunction, and adverse remodeling. For this reason, greater understanding of Ca2+ handling mechanisms in heart failure is critical for selection of appropriate treatment strategies. In this review, we summarize the mechanisms of altered Ca2+ handling in two subsets of heart failure, heart failure with reduced ejection fraction and heart failure with preserved ejection fraction, and outline current and experimental treatments that target cardiomyocyte Ca2+ handling processes.
Collapse
Affiliation(s)
- Deborah Peana
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Timothy L Domeier
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
176
|
Chen J, Li Z, Hatcher JT, Chen QH, Chen L, Wurster RD, Chan SL, Cheng Z. Deletion of TRPC6 Attenuates NMDA Receptor-Mediated Ca 2+ Entry and Ca 2+-Induced Neurotoxicity Following Cerebral Ischemia and Oxygen-Glucose Deprivation. Front Neurosci 2017; 11:138. [PMID: 28400714 PMCID: PMC5368256 DOI: 10.3389/fnins.2017.00138] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/06/2017] [Indexed: 12/21/2022] Open
Abstract
Transient receptor potential canonical 6 (TRPC6) channels are permeable to Na+ and Ca2+ and are widely expressed in the brain. In this study, the role of TRPC6 was investigated following ischemia/reperfusion (I/R) and oxygen-glucose deprivation (OGD). We found that TRPC6 expression was increased in wild-type (WT) mice cortical neurons following I/R and in primary neurons with OGD, and that deletion of TRPC6 reduced the I/R-induced brain infarct in mice and the OGD- /neurotoxin-induced neuronal death. Using live-cell imaging to examine intracellular Ca2+ levels ([Ca2+]i), we found that OGD induced a significant higher increase in glutamate-evoked Ca2+ influx compared to untreated control and such an increase was reduced by TRPC6 deletion. Enhancement of TRPC6 expression using AdCMV-TRPC6-GFP infection in WT neurons increased [Ca2+]i in response to glutamate application compared to AdCMV-GFP control. Inhibition of N-methyl-d-aspartic acid receptor (NMDAR) with MK801 decreased TRPC6-dependent increase of [Ca2+]i in TRPC6 infected cells, indicating that such a Ca2+ influx was NMDAR dependent. Furthermore, TRPC6-dependent Ca2+ influx was blunted by blockade of Na+ entry in TRPC6 infected cells. Finally, OGD-enhanced Ca2+ influx was reduced, but not completely blocked, in the presence of voltage-dependent Na+ channel blocker tetrodotoxin (TTX) and dl-α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) blocker CNQX. Altogether, we concluded that I/R-induced brain damage was, in part, due to upregulation of TRPC6 in cortical neurons. We postulate that overexpression of TRPC6 following I/R may induce neuronal death partially through TRPC6-dependent Na+ entry which activated NMDAR, thus leading to a damaging Ca2+ overload. These findings may provide a potential target for future intervention in stroke-induced brain damage.
Collapse
Affiliation(s)
- Jin Chen
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Zhaozhong Li
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Jeffery T Hatcher
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Qing-Hui Chen
- Department of Kinesiology and Integrative Physiology, Michigan Technological University Houghton, MI, USA
| | - Li Chen
- Department of Clinical Laboratory, The First Central Hospital of Tianjin Tianjin, China
| | - Robert D Wurster
- Department of Cellular and Molecular Physiology, Stritch School of Medicine, Loyola University Maywood, IL, USA
| | - Sic L Chan
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida Orlando, FL, USA
| | - Zixi Cheng
- Division of Neuroscience, Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlando, FL, USA; Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central FloridaOrlando, FL, USA
| |
Collapse
|
177
|
Effect of lithium on ventricular remodelling in infarcted rats via the Akt/mTOR signalling pathways. Biosci Rep 2017; 37:BSR20160257. [PMID: 28115595 PMCID: PMC5469250 DOI: 10.1042/bsr20160257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/23/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Activation of phosphoinositide 3-kinase (PI3K)/Akt signalling is the molecular pathway driving physiological hypertrophy. As lithium, a PI3K agonist, is highly toxic at regular doses, we assessed the effect of lithium at a lower dose on ventricular hypertrophy after myocardial infarction (MI). Male Wistar rats after induction of MI were randomized to either vehicle or lithium (1 mmol/kg per day) for 4 weeks. The dose of lithium led to a mean serum level of 0.39 mM, substantially lower than the therapeutic concentrations (0.8–1.2 mM). Infarction in the vehicle was characterized by pathological hypertrophy in the remote zone; histologically, by increased cardiomyocyte sizes, interstitial fibrosis and left ventricular dilatation; functionally, by impaired cardiac contractility; and molecularly, by an increase of p-extracellular-signal-regulated kinase (ERK) levels, nuclear factor of activated T cells (NFAT) activity, GATA4 expression and foetal gene expressions. Lithium administration mitigated pathological remodelling. Furthermore, lithium caused increased phosphorylation of eukaryotic initiation factor 4E binding protein 1 (p-4E-BP1), the downstream target of mammalian target of rapamycin (mTOR). Blockade of the Akt and mTOR signalling pathway with deguelin and rapamycin resulted in markedly diminished levels of p-4E-BP1, but not ERK. The present study demonstrated that chronic lithium treatment at low doses mitigates pathological hypertrophy through an Akt/mTOR dependent pathway.
Collapse
|
178
|
The TRPM4 channel is functionally important for the beneficial cardiac remodeling induced by endurance training. J Muscle Res Cell Motil 2017; 38:3-16. [DOI: 10.1007/s10974-017-9466-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 02/02/2017] [Indexed: 10/20/2022]
|
179
|
Murakami K, Osanai T, Tanaka M, Nishizaki K, Kinjo T, Tanno T, Ishida Y, Suzuki A, Endo T, Tomita H, Okumura K. Enhanced transient receptor potential channel-mediated Ca2+influx in the cells with phospholipase C-δ1 overexpression: its possible role in coronary artery spasm. Fundam Clin Pharmacol 2017; 31:383-391. [DOI: 10.1111/fcp.12269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 01/17/2017] [Indexed: 02/04/2023]
Affiliation(s)
- Kazuo Murakami
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Tomohiro Osanai
- Department of Nursing; Hirosaki University Graduate School of Health Science; 66-1 Hon-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Makoto Tanaka
- Department of Hypertension and Stroke Internal Medicine; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Kimitaka Nishizaki
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Takahiko Kinjo
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Tomohiro Tanno
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Yuji Ishida
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Akiko Suzuki
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Tomohide Endo
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Hirofumi Tomita
- Department of Cardiology; Hirosaki University Graduate School of Medicine; 5 Zaifu-cho Hirosaki Hirosaki Aomori Prefecture Japan
| | - Ken Okumura
- Division of Cardiology; Saiseikai Kumamoto Hospital; 5-3-1 Chikami Minamiku Kumamoto Kumamoto Kumamoto Prefecture Japan
| |
Collapse
|
180
|
Parra V, Rothermel BA. Calcineurin signaling in the heart: The importance of time and place. J Mol Cell Cardiol 2017; 103:121-136. [PMID: 28007541 PMCID: PMC5778886 DOI: 10.1016/j.yjmcc.2016.12.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/12/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022]
Abstract
The calcium-activated protein phosphatase, calcineurin, lies at the intersection of protein phosphorylation and calcium signaling cascades, where it provides an essential nodal point for coordination between these two fundamental modes of intracellular communication. In excitatory cells, such as neurons and cardiomyocytes, that experience rapid and frequent changes in cytoplasmic calcium, calcineurin protein levels are exceptionally high, suggesting that these cells require high levels of calcineurin activity. Yet, it is widely recognized that excessive activation of calcineurin in the heart contributes to pathological hypertrophic remodeling and the progression to failure. How does a calcium activated enzyme function in the calcium-rich environment of the continuously contracting heart without pathological consequences? This review will discuss the wide range of calcineurin substrates relevant to cardiovascular health and the mechanisms calcineurin uses to find and act on appropriate substrates in the appropriate location while potentially avoiding others. Fundamental differences in calcineurin signaling in neonatal verses adult cardiomyocytes will be addressed as well as the importance of maintaining heterogeneity in calcineurin activity across the myocardium. Finally, we will discuss how circadian oscillations in calcineurin activity may facilitate integration with other essential but conflicting processes, allowing a healthy heart to reap the benefits of calcineurin signaling while avoiding the detrimental consequences of sustained calcineurin activity that can culminate in heart failure.
Collapse
Affiliation(s)
- Valentina Parra
- Advanced Centre for Chronic Disease (ACCDiS), Facultad Ciencias Quimicas y Farmaceuticas, Universidad de Chile, Santiago,Chile; Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Quimicas y Farmaceuticas, Universidad de Chie, Santiago, Chile
| | - Beverly A Rothermel
- Department of Internal Medicine (Cardiology Division), University of Texas Southwestern Medical Centre, Dallas, TX, USA; Department of Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
181
|
Groschner K, Shrestha N, Fameli N. Cardiovascular and Hemostatic Disorders: SOCE in Cardiovascular Cells: Emerging Targets for Therapeutic Intervention. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:473-503. [PMID: 28900929 DOI: 10.1007/978-3-319-57732-6_24] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The discovery of the store-operated Ca2+ entry (SOCE) phenomenon is tightly associated with its recognition as a pathway of high (patho)physiological significance in the cardiovascular system. Early on, SOCE has been investigated primarily in non-excitable cell types, and the vascular endothelium received particular attention, while a role of SOCE in excitable cells, specifically cardiac myocytes and pacemakers, was initially ignored and remains largely enigmatic even to date. With the recent gain in knowledge on the molecular components of SOCE as well as their cellular organization within nanodomains, potential tissue/cell type-dependent heterogeneity of the SOCE machinery along with high specificity of linkage to downstream signaling pathways emerged for cardiovascular cells. The basis of precise decoding of cellular Ca2+ signals was recently uncovered to involve correct spatiotemporal organization of signaling components, and even minor disturbances in these assemblies trigger cardiovascular pathologies. With this chapter, we wish to provide an overview on current concepts of cellular organization of SOCE signaling complexes in cardiovascular cells with particular focus on the spatiotemporal aspects of coupling to downstream signaling and the potential disturbance of these mechanisms by pathogenic factors. The significance of these mechanistic concepts for the development of novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Klaus Groschner
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria.
| | - Niroj Shrestha
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
| | - Nicola Fameli
- Institute of Biophysics, Medical University of Graz, Neue Stiftingtalstrasse 6/4, 8010, Graz, Austria
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
182
|
Eder P. Cardiac Remodeling and Disease: SOCE and TRPC Signaling in Cardiac Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:505-521. [DOI: 10.1007/978-3-319-57732-6_25] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
183
|
Harada E, Mizuno Y, Kugimiya F, Shono M, Maeda H, Yano N, Kuwahara K, Yasue H. B-Type Natriuretic Peptide in Heart Failure With Preserved Ejection Fraction ― Relevance to Age-Related Left Ventricular Modeling in Japanese ―. Circ J 2017; 81:1006-1013. [DOI: 10.1253/circj.cj-16-1282] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Eisaku Harada
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Yuji Mizuno
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Fumihito Kugimiya
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Makoto Shono
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Hiroyuki Maeda
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Naotsugu Yano
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine, Shinshu University School of Medicine
| | - Hirofumi Yasue
- Division of Cardiovascular Medicine, Kumamoto Kinoh Hospital, Kumamoto Aging Research Institute
| |
Collapse
|
184
|
Dalton GD, Xie J, An SW, Huang CL. New Insights into the Mechanism of Action of Soluble Klotho. Front Endocrinol (Lausanne) 2017; 8:323. [PMID: 29250031 PMCID: PMC5715364 DOI: 10.3389/fendo.2017.00323] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/02/2017] [Indexed: 12/31/2022] Open
Abstract
The klotho gene encodes a type I single-pass transmembrane protein that contains a large extracellular domain, a membrane spanning segment, and a short intracellular domain. Klotho protein exists in several forms including the full-length membrane form (mKl) and a soluble circulating form [soluble klotho (sKl)]. mKl complexes with fibroblast growth factor receptors to form coreceptors for FGF23, which allows it to participate in FGF23-mediated signal transduction and regulation of phosphate and calcium homeostasis. sKl is present in the blood, urine, and cerebrospinal fluid where it performs a multitude of functions including regulation of ion channels/transporters and growth factor signaling. How sKl exerts these pleiotropic functions is poorly understood. One hurdle in understanding sKl's mechanism of action as a "hormone" has been the inability to identify a receptor that mediates its effects. In the body, the kidneys are a major source of sKl and sKl levels decline during renal disease. sKl deficiency in chronic kidney disease makes the heart susceptible to stress-induced injury. Here, we summarize the current knowledge of mKl's mechanism of action, the mechanistic basis of sKl's protective, FGF23-independent effects on the heart, and provide new insights into the mechanism of action of sKl focusing on recent findings that sKl binds sialogangliosides in membrane lipid rafts to regulate growth factor signaling.
Collapse
Affiliation(s)
- George D. Dalton
- Department of Medicine, Division of Gastroenterology, Duke University Medical Center, Durham, NC, United States
| | - Jian Xie
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Sung-Wan An
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Chou-Long Huang
- Department of Internal Medicine, Division of Nephrology and Hypertension, University of Iowa Carver College of Medicine, Iowa City, IA, United States
- *Correspondence: Chou-Long Huang,
| |
Collapse
|
185
|
Kirschmer N, Bandleon S, von Ehrlich-Treuenstätt V, Hartmann S, Schaaf A, Lamprecht AK, Miranda-Laferte E, Langsenlehner T, Ritter O, Eder P. TRPC4α and TRPC4β Similarly Affect Neonatal Cardiomyocyte Survival during Chronic GPCR Stimulation. PLoS One 2016; 11:e0168446. [PMID: 27992507 PMCID: PMC5167390 DOI: 10.1371/journal.pone.0168446] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 12/01/2016] [Indexed: 11/19/2022] Open
Abstract
The Transient Receptor Potential Channel Subunit 4 (TRPC4) has been considered as a crucial Ca2+ component in cardiomyocytes promoting structural and functional remodeling in the course of pathological cardiac hypertrophy. TRPC4 assembles as homo or hetero-tetramer in the plasma membrane, allowing a non-selective Na+ and Ca2+ influx. Gαq protein-coupled receptor (GPCR) stimulation is known to increase TRPC4 channel activity and a TRPC4-mediated Ca2+ influx which has been regarded as ideal Ca2+ source for calcineurin and subsequent nuclear factor of activated T-cells (NFAT) activation. Functional properties of TRPC4 are also based on the expression of the TRPC4 splice variants TRPC4α and TRPC4β. Aim of the present study was to analyze cytosolic Ca2+ signals, signaling, hypertrophy and vitality of cardiomyocytes in dependence on the expression level of either TRPC4α or TRPC4β. The analysis of Ca2+ transients in neonatal rat cardiomyocytes (NRCs) showed that TRPC4α and TRPC4β affected Ca2+ cycling in beating cardiomyocytes with both splice variants inducing an elevation of the Ca2+ transient amplitude at baseline and TRPC4β increasing the Ca2+ peak during angiotensin II (Ang II) stimulation. NRCs infected with TRPC4β (Ad-C4β) also responded with a sustained Ca2+ influx when treated with Ang II under non-pacing conditions. Consistent with the Ca2+ data, NRCs infected with TRPC4α (Ad-C4α) showed an elevated calcineurin/NFAT activity and a baseline hypertrophic phenotype but did not further develop hypertrophy during chronic Ang II/phenylephrine stimulation. Down-regulation of endogenous TRPC4α reversed these effects, resulting in less hypertrophy of NRCs at baseline but a markedly increased hypertrophic enlargement after chronic agonist stimulation. Ad-C4β NRCs did not exhibit baseline calcineurin/NFAT activity or hypertrophy but responded with an increased calcineurin/NFAT activity after GPCR stimulation. However, this effect was not translated into an increased propensity towards hypertrophy but rather less hypertrophy during GPCR stimulation. Further analyses revealed that, although hypertrophy was preserved in Ad-C4α NRCs and even attenuated in Ad-C4β NRCs, cardiomyocytes had an increased apoptosis rate and thus were less viable after chronic GPCR stimulation. These findings suggest that TRPC4α and TRPC4β differentially affect Ca2+ signals, calcineurin/NFAT signaling and hypertrophy but similarly impair cardiomyocyte viability during GPCR stimulation.
Collapse
Affiliation(s)
- Nadine Kirschmer
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Sandra Bandleon
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Viktor von Ehrlich-Treuenstätt
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Sonja Hartmann
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, United States of America
| | - Alice Schaaf
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Anna-Karina Lamprecht
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
| | | | - Tanja Langsenlehner
- Department of Therapeutic Radiology and Oncology, Medical University of Graz, Graz, Austria
| | - Oliver Ritter
- Department of Cardiology and Pulmology, Brandenburg Medical School, University Hospital Brandenburg, Brandenburg, Germany
| | - Petra Eder
- Department of Internal Medicine I, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center Würzburg, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
186
|
TRPC3 positively regulates reactive oxygen species driving maladaptive cardiac remodeling. Sci Rep 2016; 6:37001. [PMID: 27833156 PMCID: PMC5105134 DOI: 10.1038/srep37001] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 10/20/2016] [Indexed: 01/28/2023] Open
Abstract
Reactive oxygen species (ROS) produced by NADPH oxidase 2 (Nox2) function as key mediators of mechanotransduction during both physiological adaptation to mechanical load and maladaptive remodeling of the heart. This is despite low levels of cardiac Nox2 expression. The mechanism underlying the transition from adaptation to maladaptation remains obscure, however. We demonstrate that transient receptor potential canonical 3 (TRPC3), a Ca2+-permeable channel, acts as a positive regulator of ROS (PRROS) in cardiomyocytes, and specifically regulates pressure overload-induced maladaptive cardiac remodeling in mice. TRPC3 physically interacts with Nox2 at specific C-terminal sites, thereby protecting Nox2 from proteasome-dependent degradation and amplifying Ca2+-dependent Nox2 activation through TRPC3-mediated background Ca2+ entry. Nox2 also stabilizes TRPC3 proteins to enhance TRPC3 channel activity. Expression of TRPC3 C-terminal polypeptide abolished TRPC3-regulated ROS production by disrupting TRPC3-Nox2 interaction, without affecting TRPC3-mediated Ca2+ influx. The novel TRPC3 function as a PRROS provides a mechanistic explanation for how diastolic Ca2+ influx specifically encodes signals to induce ROS-mediated maladaptive remodeling and offers new therapeutic possibilities.
Collapse
|
187
|
Morine KJ, Paruchuri V, Qiao X, Aronovitz M, Huggins GS, DeNofrio D, Kiernan MS, Karas RH, Kapur NK. Endoglin selectively modulates transient receptor potential channel expression in left and right heart failure. Cardiovasc Pathol 2016; 25:478-482. [PMID: 27614169 PMCID: PMC5443561 DOI: 10.1016/j.carpath.2016.08.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Transient receptor potential (TRP) channels are broadly expressed cation channels that mediate diverse physiological stimuli and include canonical (TRPC), melastatin (TRPM), and vanilloid (TRPV) subtypes. Recent studies have implicated a role for TRPC6 channels as an important component of signaling via the cytokine, transforming growth factor beta 1 (TGFβ1) in right (RV) or left ventricular (LV) failure. Endoglin (Eng) is a transmembrane glycoprotein that promotes TRPC6 expression and TGFβ1 activity. No studies have defined biventricular expression of all TRP channel family members in heart failure. HYPOTHESIS We hypothesized that heart failure is associated with distinct patterns of TRP channel expression in the LV and RV. METHODS Paired viable LV and RV free wall tissue was obtained from human subjects with end-stage heart failure (n=12) referred for cardiac transplantation or biventricular assist device implantation. Paired LV and RV samples from human subjects without heart failure served as controls (n=3). To explore a functional role for Eng as a regulator of TRP expression in response to RV or LV pressure overload, wild-type (Eng+/+) and Eng haploinsufficient (Eng+/-) mice were exposed to thoracic aortic (TAC) or pulmonary arterial (PAC) constriction for 8weeks. Biventricular tissue was analyzed by real-time polymerase chain reaction. RESULTS Compared to nonfailing human LV and RV samples, mRNA levels of TRPC1, 3, 4, 6, and TRPV-2 were increased and TRPM2, 3, and 8 were decreased in failing LV and RV samples. TRPC1 and 6 levels were higher in failing RV compared to failing LV samples. After TAC, murine LV levels of TPRC1 and 6 were increased in both Eng+/+ and Eng+/- mice compared to sham controls. LV levels of TRPC4, TRPM3 and 7, TRPV2 and 4 were increased in Eng+/+, not in Eng+/- mice after TAC. After PAC, all TRP channel family members were increased in the RV, but not LV, of Eng+/+ compared to sham controls. In contrast to Eng+/+, PAC did not increase RV or LV levels of TRP channels in Eng+/- mice. CONCLUSIONS This is the first study to demonstrate that TRP channels exhibit distinct profiles of expression in the LV and RV of patients with heart failure and in murine models of univentricular pressure overload. We further introduce that the TGFβ1 coreceptor Eng selectively regulates expression of multiple TRP channels in the setting of LV or RV pressure overload.
Collapse
Affiliation(s)
- Kevin J Morine
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Vikram Paruchuri
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Xiaoying Qiao
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Mark Aronovitz
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Gordon S Huggins
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - David DeNofrio
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Michael S Kiernan
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Richard H Karas
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA
| | - Navin K Kapur
- Molecular Cardiology Research Institute and Division of Cardiology, Department of Medicine, Tufts Medical Center, 800 Washington Street, Boston, MA 02111, USA.
| |
Collapse
|
188
|
Epand RM, So V, Jennings W, Khadka B, Gupta RS, Lemaire M. Diacylglycerol Kinase-ε: Properties and Biological Roles. Front Cell Dev Biol 2016; 4:112. [PMID: 27803897 PMCID: PMC5067486 DOI: 10.3389/fcell.2016.00112] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 09/27/2016] [Indexed: 12/20/2022] Open
Abstract
In mammals there are at least 10 isoforms of diacylglycerol kinases (DGK). All catalyze the phosphorylation of diacylglycerol (DAG) to phosphatidic acid (PA). Among DGK isoforms, DGKε has several unique features. It is the only DGK isoform with specificity for a particular species of DAG, i.e., 1-stearoyl-2-arachidonoyl glycerol. The smallest of all known DGK isoforms, DGKε, is also the only DGK devoid of a regulatory domain. DGKε is the only DGK isoform that has a hydrophobic segment that is predicted to form a transmembrane helix. As the only membrane-bound, constitutively active DGK isoform with exquisite specificity for particular molecular species of DAG, the functional overlap between DGKε and other DGKs is predicted to be minimal. DGKε exhibits specificity for DAG containing the same acyl chains as those found in the lipid intermediates of the phosphatidylinositol-cycle. It has also been shown that DGKε affects the acyl chain composition of phosphatidylinositol in whole cells. It is thus likely that DGKε is responsible for catalyzing one step in the phosphatidylinositol-cycle. Steps of this cycle take place in both the plasma membrane and the endoplasmic reticulum membrane. DGKε is likely present in both of these membranes. DGKε is the only DGK isoform that is associated with a human disease. Indeed, recessive loss-of-function mutations in DGKε cause atypical hemolytic-uremic syndrome (aHUS). This condition is characterized by thrombosis in the small vessels of the kidney. It causes acute renal insufficiency in infancy and most patients develop end-stage renal failure before adulthood. Disease pathophysiology is poorly understood and there is no therapy. There are also data suggesting that DGKε may play a role in epilepsy and Huntington disease. Thus, DGKε has many unique molecular and biochemical properties when compared to all other DGK isoforms. DGKε homologs also contain a number of conserved sequence features that are distinctive characteristics of either the rodents or specific groups of primate homologs. How cells, tissues and organisms harness DGKε's catalytic prowess remains unclear. The discovery of DGKε's role in causing aHUS will hopefully boost efforts to unravel the mechanisms by which DGKε dysfunction causes disease.
Collapse
Affiliation(s)
- Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Vincent So
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences CentreHamilton, ON, Canada; Nephrology Division and Cell Biology Program, Hospital for Sick ChildrenToronto, ON, Canada
| | - William Jennings
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Bijendra Khadka
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Radhey S Gupta
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Mathieu Lemaire
- Nephrology Division and Cell Biology Program, Hospital for Sick ChildrenToronto, ON, Canada; Department of Biochemistry, University of TorontoToronto, ON, Canada; Institute of Medicine, University of TorontoToronto, ON, Canada
| |
Collapse
|
189
|
Epand RM, So V, Jennings W, Khadka B, Gupta RS, Lemaire M. Diacylglycerol Kinase-ε: Properties and Biological Roles. Front Cell Dev Biol 2016. [PMID: 27803897 DOI: 10.3389/fcell.2016.00112)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
In mammals there are at least 10 isoforms of diacylglycerol kinases (DGK). All catalyze the phosphorylation of diacylglycerol (DAG) to phosphatidic acid (PA). Among DGK isoforms, DGKε has several unique features. It is the only DGK isoform with specificity for a particular species of DAG, i.e., 1-stearoyl-2-arachidonoyl glycerol. The smallest of all known DGK isoforms, DGKε, is also the only DGK devoid of a regulatory domain. DGKε is the only DGK isoform that has a hydrophobic segment that is predicted to form a transmembrane helix. As the only membrane-bound, constitutively active DGK isoform with exquisite specificity for particular molecular species of DAG, the functional overlap between DGKε and other DGKs is predicted to be minimal. DGKε exhibits specificity for DAG containing the same acyl chains as those found in the lipid intermediates of the phosphatidylinositol-cycle. It has also been shown that DGKε affects the acyl chain composition of phosphatidylinositol in whole cells. It is thus likely that DGKε is responsible for catalyzing one step in the phosphatidylinositol-cycle. Steps of this cycle take place in both the plasma membrane and the endoplasmic reticulum membrane. DGKε is likely present in both of these membranes. DGKε is the only DGK isoform that is associated with a human disease. Indeed, recessive loss-of-function mutations in DGKε cause atypical hemolytic-uremic syndrome (aHUS). This condition is characterized by thrombosis in the small vessels of the kidney. It causes acute renal insufficiency in infancy and most patients develop end-stage renal failure before adulthood. Disease pathophysiology is poorly understood and there is no therapy. There are also data suggesting that DGKε may play a role in epilepsy and Huntington disease. Thus, DGKε has many unique molecular and biochemical properties when compared to all other DGK isoforms. DGKε homologs also contain a number of conserved sequence features that are distinctive characteristics of either the rodents or specific groups of primate homologs. How cells, tissues and organisms harness DGKε's catalytic prowess remains unclear. The discovery of DGKε's role in causing aHUS will hopefully boost efforts to unravel the mechanisms by which DGKε dysfunction causes disease.
Collapse
Affiliation(s)
- Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Vincent So
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences CentreHamilton, ON, Canada; Nephrology Division and Cell Biology Program, Hospital for Sick ChildrenToronto, ON, Canada
| | - William Jennings
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Bijendra Khadka
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Radhey S Gupta
- Department of Biochemistry and Biomedical Sciences, McMaster University Health Sciences Centre Hamilton, ON, Canada
| | - Mathieu Lemaire
- Nephrology Division and Cell Biology Program, Hospital for Sick ChildrenToronto, ON, Canada; Department of Biochemistry, University of TorontoToronto, ON, Canada; Institute of Medicine, University of TorontoToronto, ON, Canada
| |
Collapse
|
190
|
Resistance to pathologic cardiac hypertrophy and reduced expression of CaV1.2 in Trpc3-depleted mice. Mol Cell Biochem 2016; 421:55-65. [DOI: 10.1007/s11010-016-2784-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 08/05/2016] [Indexed: 11/26/2022]
|
191
|
Foster DB, Liu T, Kammers K, O'Meally R, Yang N, Papanicolaou KN, Talbot CC, Cole RN, O'Rourke B. Integrated Omic Analysis of a Guinea Pig Model of Heart Failure and Sudden Cardiac Death. J Proteome Res 2016; 15:3009-28. [PMID: 27399916 DOI: 10.1021/acs.jproteome.6b00149] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Here, we examine key regulatory pathways underlying the transition from compensated hypertrophy (HYP) to decompensated heart failure (HF) and sudden cardiac death (SCD) in a guinea pig pressure-overload model by integrated multiome analysis. Relative protein abundances from sham-operated HYP and HF hearts were assessed by iTRAQ LC-MS/MS. Metabolites were quantified by LC-MS/MS or GC-MS. Transcriptome profiles were obtained using mRNA microarrays. The guinea pig HF proteome exhibited classic biosignatures of cardiac HYP, left ventricular dysfunction, fibrosis, inflammation, and extravasation. Fatty acid metabolism, mitochondrial transcription/translation factors, antioxidant enzymes, and other mitochondrial procsses, were downregulated in HF but not HYP. Proteins upregulated in HF implicate extracellular matrix remodeling, cytoskeletal remodeling, and acute phase inflammation markers. Among metabolites, acylcarnitines were downregulated in HYP and fatty acids accumulated in HF. The correlation of transcript and protein changes in HF was weak (R(2) = 0.23), suggesting post-transcriptional gene regulation in HF. Proteome/metabolome integration indicated metabolic bottlenecks in fatty acyl-CoA processing by carnitine palmitoyl transferase (CPT1B) as well as TCA cycle inhibition. On the basis of these findings, we present a model of cardiac decompensation involving impaired nuclear integration of Ca(2+) and cyclic nucleotide signals that are coupled to mitochondrial metabolic and antioxidant defects through the CREB/PGC1α transcriptional axis.
Collapse
Affiliation(s)
- D Brian Foster
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Ting Liu
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Kai Kammers
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health , Baltimore, Maryland 21205, United States
| | - Robert O'Meally
- Proteomics Core Facility, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Ni Yang
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Kyriakos N Papanicolaou
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Robert N Cole
- Proteomics Core Facility, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| | - Brian O'Rourke
- Division of Cardiology, Johns Hopkins University School of Medicine , Baltimore, Maryland 21205, United States
| |
Collapse
|
192
|
Soni H, Adebiyi A. TRPC6 channel activation promotes neonatal glomerular mesangial cell apoptosis via calcineurin/NFAT and FasL/Fas signaling pathways. Sci Rep 2016; 6:29041. [PMID: 27383564 PMCID: PMC4935859 DOI: 10.1038/srep29041] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/14/2016] [Indexed: 02/06/2023] Open
Abstract
Glomerular mesangial cell (GMC) proliferation and death are involved in the pathogenesis of glomerular disorders. The mechanisms that control GMC survival are poorly understood, but may include signal transduction pathways that are modulated by changes in intracellular Ca2+ ([Ca2+]i) concentration. In this study, we investigated whether activation of the canonical transient receptor potential (TRPC) 6 channels and successive [Ca2+]i elevation alter neonatal GMC survival. Hyperforin (HF)-induced TRPC6 channel activation increased [Ca2+]i concentration, inhibited proliferation, and triggered apoptotic cell death in primary neonatal pig GMCs. HF-induced neonatal GMC apoptosis was not associated with oxidative stress. However, HF-induced TRPC6 channel activation stimulated nuclear translocation of the nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1). HF also increased cell death surface receptor Fas ligand (FasL) level and caspase-8 activity in the cells; effects mitigated by [Ca2+]i chelator BAPTA, calcineurin/NFAT inhibitor VIVIT, and TRPC6 channel knockdown. Accordingly, HF-induced neonatal GMC apoptosis was attenuated by BAPTA, VIVIT, Fas blocking antibody, and a caspase-3/7 inhibitor. These findings suggest that TRPC6 channel-dependent [Ca2+]i elevation and the ensuing induction of the calcineurin/NFAT, FasL/Fas, and caspase signaling cascades promote neonatal pig GMC apoptosis.
Collapse
Affiliation(s)
- Hitesh Soni
- Department of Physiology University of Tennessee Health Science Center, Memphis TN, USA
| | - Adebowale Adebiyi
- Department of Physiology University of Tennessee Health Science Center, Memphis TN, USA
| |
Collapse
|
193
|
Rainer PP, Kass DA. Old dog, new tricks: novel cardiac targets and stress regulation by protein kinase G. Cardiovasc Res 2016; 111:154-62. [PMID: 27297890 DOI: 10.1093/cvr/cvw107] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/18/2016] [Indexed: 12/11/2022] Open
Abstract
The second messenger cyclic guanosine 3'5' monophosphate (cGMP) and its downstream effector protein kinase G (PKG) have been discovered more than 40 years ago. In vessels, PKG1 induces smooth muscle relaxation in response to nitric oxide signalling and thus lowers systemic and pulmonary blood pressure. In platelets, PKG1 stimulation by cGMP inhibits activation and aggregation, and in experimental models of heart failure (HF), PKG1 activation by inhibiting cGMP degradation is protective. The net effect of the above-mentioned signalling is cardiovascular protection. Yet, while modulation of cGMP-PKG has entered clinical practice for treating pulmonary hypertension or erectile dysfunction, translation of promising studies in experimental HF to clinical success has failed thus far. With the advent of new technologies, novel mechanisms of PKG regulation, including mechanosensing, redox regulation, protein quality control, and cGMP degradation, have been discovered. These novel, non-canonical roles of PKG1 may help understand why clinical translation has disappointed thus far. Addressing them appears to be a requisite for future, successful translation of experimental studies to the clinical arena.
Collapse
Affiliation(s)
- Peter P Rainer
- Division of Cardiology, Medical University of Graz, Auenbruggerplatz 15, A-8036 Graz, Austria
| | - David A Kass
- Division of Cardiology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
194
|
Shimizu I, Minamino T. Physiological and pathological cardiac hypertrophy. J Mol Cell Cardiol 2016; 97:245-62. [PMID: 27262674 DOI: 10.1016/j.yjmcc.2016.06.001] [Citation(s) in RCA: 634] [Impact Index Per Article: 79.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/10/2016] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The heart must continuously pump blood to supply the body with oxygen and nutrients. To maintain the high energy consumption required by this role, the heart is equipped with multiple complex biological systems that allow adaptation to changes of systemic demand. The processes of growth (hypertrophy), angiogenesis, and metabolic plasticity are critically involved in maintenance of cardiac homeostasis. Cardiac hypertrophy is classified as physiological when it is associated with normal cardiac function or as pathological when associated with cardiac dysfunction. Physiological hypertrophy of the heart occurs in response to normal growth of children or during pregnancy, as well as in athletes. In contrast, pathological hypertrophy is induced by factors such as prolonged and abnormal hemodynamic stress, due to hypertension, myocardial infarction etc. Pathological hypertrophy is associated with fibrosis, capillary rarefaction, increased production of pro-inflammatory cytokines, and cellular dysfunction (impairment of signaling, suppression of autophagy, and abnormal cardiomyocyte/non-cardiomyocyte interactions), as well as undesirable epigenetic changes, with these complex responses leading to maladaptive cardiac remodeling and heart failure. This review describes the key molecules and cellular responses involved in physiological/pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Division of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
195
|
Vrenken KS, Jalink K, van Leeuwen FN, Middelbeek J. Beyond ion-conduction: Channel-dependent and -independent roles of TRP channels during development and tissue homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1436-46. [DOI: 10.1016/j.bbamcr.2015.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 10/26/2015] [Accepted: 11/11/2015] [Indexed: 01/09/2023]
|
196
|
Salanova Villanueva L, Sánchez González C, Sánchez Tomero JA, Aguilera A, Ortega Junco E. Bone mineral disorder in chronic kidney disease: Klotho and FGF23; cardiovascular implications. Nefrologia 2016; 36:368-75. [PMID: 27118192 DOI: 10.1016/j.nefro.2016.01.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Revised: 12/17/2015] [Accepted: 01/02/2016] [Indexed: 12/30/2022] Open
Abstract
Cardiovascular factors are one of the main causes of morbidity and mortality in patients with chronic kidney disease. Bone mineral metabolism disorders and inflammation are pathological conditions that involve increased cardiovascular risk in chronic kidney disease. The cardiovascular risk involvement of bone mineral metabolism classical biochemical parameters such as phosphorus, calcium, vitamin D and PTH is well known. The newest markers, FGF23 and klotho, could also be implicated in cardiovascular disease.
Collapse
|
197
|
Sabourin J, Bartoli F, Antigny F, Gomez AM, Benitah JP. Transient Receptor Potential Canonical (TRPC)/Orai1-dependent Store-operated Ca2+ Channels: NEW TARGETS OF ALDOSTERONE IN CARDIOMYOCYTES. J Biol Chem 2016; 291:13394-409. [PMID: 27129253 DOI: 10.1074/jbc.m115.693911] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Indexed: 12/31/2022] Open
Abstract
Store-operated Ca(2+) entry (SOCE) has emerged as an important mechanism in cardiac pathology. However, the signals that up-regulate SOCE in the heart remain unexplored. Clinical trials have emphasized the beneficial role of mineralocorticoid receptor (MR) signaling blockade in heart failure and associated arrhythmias. Accumulated evidence suggests that the mineralocorticoid hormone aldosterone, through activation of its receptor, MR, might be a key regulator of Ca(2+) influx in cardiomyocytes. We thus assessed whether and how SOCE involving transient receptor potential canonical (TRPC) and Orai1 channels are regulated by aldosterone/MR in neonatal rat ventricular cardiomyocytes. Molecular screening using qRT-PCR and Western blotting demonstrated that aldosterone treatment for 24 h specifically increased the mRNA and/or protein levels of Orai1, TRPC1, -C4, -C5, and stromal interaction molecule 1 through MR activation. These effects were correlated with a specific enhancement of SOCE activities sensitive to store-operated channel inhibitors (SKF-96365 and BTP2) and to a potent Orai1 blocker (S66) and were prevented by TRPC1, -C4, and Orai1 dominant negative mutants or TRPC5 siRNA. A mechanistic approach showed that up-regulation of serum- and glucocorticoid-regulated kinase 1 mRNA expression by aldosterone is involved in enhanced SOCE. Functionally, 24-h aldosterone-enhanced SOCE is associated with increased diastolic [Ca(2+)]i, which is blunted by store-operated channel inhibitors. Our study provides the first evidence that aldosterone promotes TRPC1-, -C4-, -C5-, and Orai1-mediated SOCE in cardiomyocytes through an MR and serum- and glucocorticoid-regulated kinase 1 pathway.
Collapse
Affiliation(s)
- Jessica Sabourin
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Fiona Bartoli
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Fabrice Antigny
- UMR S999, INSERM, Université Paris-Sud, Université Paris-Saclay, Centre Chirurgical Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - Ana Maria Gomez
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| | - Jean-Pierre Benitah
- From the UMR S1180, INSERM, Université Paris-Sud, Université Paris-Saclay, 92296 Châtenay-Malabry, France and
| |
Collapse
|
198
|
YAO XINGMEI, LIU YUJUN, WANG YUNMAN, WANG HAO, ZHU BINGBING, LIANG YONGPING, YAO WEIGUO, YU HUI, WANG NIANSONG, ZHANG XUEMEI, PENG WEN. Astragaloside IV prevents high glucose-induced podocyte apoptosis via downregulation of TRPC6. Mol Med Rep 2016; 13:5149-56. [DOI: 10.3892/mmr.2016.5167] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 02/15/2016] [Indexed: 12/21/2022] Open
|
199
|
Mallipattu SK, He JC. The podocyte as a direct target for treatment of glomerular disease? Am J Physiol Renal Physiol 2016; 311:F46-51. [PMID: 27097894 DOI: 10.1152/ajprenal.00184.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 04/18/2016] [Indexed: 11/22/2022] Open
Abstract
The Centers for Disease Control and Prevention estimates more than 10% of adults in the United States, over 20 million Americans, have chronic kidney disease (CKD). A failure to maintain the glomerular filtration barrier directly contributes to the onset of CKD. The visceral epithelial cells, podocytes, are integral to the maintenance of this renal filtration barrier. Direct podocyte injury contributes to the onset and progression of glomerular diseases such as minimal change disease (MCD), focal segmental glomerular sclerosis (FSGS), diabetic nephropathy, and HIV-associated nephropathy (HIVAN). Since podocytes are terminally differentiated with minimal capacity to self-replicate, they are extremely sensitive to cellular injury. In the past two decades, our understanding of the mechanism(s) by which podocyte injury occurs has greatly expanded. With this newfound knowledge, therapeutic strategies have shifted to identifying targets directed specifically at the podocyte. Although the systemic effects of these agents are important, their direct effect on the podocyte proves to be essential in ameliorating glomerular disease. In this review, we highlight the mechanisms by which these agents directly target the podocyte independent of its systemic effects.
Collapse
Affiliation(s)
- Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York
| | - John C He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York; and Renal Section, James J. Peters VA Medical Center, New York, New York
| |
Collapse
|
200
|
Zhou Y, Greka A. Calcium-permeable ion channels in the kidney. Am J Physiol Renal Physiol 2016; 310:F1157-67. [PMID: 27029425 DOI: 10.1152/ajprenal.00117.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023] Open
Abstract
Calcium ions (Ca(2+)) are crucial for a variety of cellular functions. The extracellular and intracellular Ca(2+) concentrations are thus tightly regulated to maintain Ca(2+) homeostasis. The kidney, one of the major organs of the excretory system, regulates Ca(2+) homeostasis by filtration and reabsorption. Approximately 60% of the Ca(2+) in plasma is filtered, and 99% of that is reabsorbed by the kidney tubules. Ca(2+) is also a critical signaling molecule in kidney development, in all kidney cellular functions, and in the emergence of kidney diseases. Recently, studies using genetic and molecular biological approaches have identified several Ca(2+)-permeable ion channel families as important regulators of Ca(2+) homeostasis in kidney. These ion channel families include transient receptor potential channels (TRP), voltage-gated calcium channels, and others. In this review, we provide a brief and systematic summary of the expression, function, and pathological contribution for each of these Ca(2+)-permeable ion channels. Moreover, we discuss their potential as future therapeutic targets.
Collapse
Affiliation(s)
- Yiming Zhou
- Department of Medicine and Glom-NExT Center for Glomerular Kidney Disease and Novel Experimental Therapeutics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and
| | - Anna Greka
- Department of Medicine and Glom-NExT Center for Glomerular Kidney Disease and Novel Experimental Therapeutics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; and The Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| |
Collapse
|