1
|
Papaioannou P, Wallace MJ, Malhotra N, Mohler PJ, El Refaey M. Biochemical Structure and Function of TRAPP Complexes in the Cardiac System. JACC Basic Transl Sci 2023; 8:1599-1612. [PMID: 38205348 PMCID: PMC10774597 DOI: 10.1016/j.jacbts.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/14/2023] [Indexed: 01/12/2024]
Abstract
Trafficking protein particle (TRAPP) is well reported to play a role in the trafficking of protein products within the Golgi and endoplasmic reticulum. Dysfunction in TRAPP has been associated with disorders in the nervous and cardiovascular systems, but the majority of literature focuses on TRAPP function in the nervous system solely. Here, we highlight the known pathways of TRAPP and hypothesize potential impacts of TRAPP dysfunction on the cardiovascular system, particularly the role of TRAPP as a guanine-nucleotide exchange factor for Rab1 and Rab11. We also review the various cardiovascular phenotypes associated with changes in TRAPP complexes and their subunits.
Collapse
Affiliation(s)
- Peter Papaioannou
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Michael J. Wallace
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Nipun Malhotra
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Peter J. Mohler
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Mona El Refaey
- Frick Center for Heart Failure and Arrhythmia Research, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| |
Collapse
|
2
|
Transcriptomic Profile of Genes Regulating the Structural Organization of Porcine Atrial Cardiomyocytes during Primary In Vitro Culture. Genes (Basel) 2022; 13:genes13071205. [PMID: 35885988 PMCID: PMC9319992 DOI: 10.3390/genes13071205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/02/2022] [Accepted: 07/04/2022] [Indexed: 02/04/2023] Open
Abstract
Numerous cardiovascular diseases (CVD) eventually lead to severe myocardial dysfunction, which is the most common cause of death worldwide. A better understanding of underlying molecular mechanisms of cardiovascular pathologies seems to be crucial to develop effective therapeutic options. Therefore, a worthwhile endeavor is a detailed molecular characterization of cells extracted from the myocardium. A transcriptomic profile of atrial cardiomyocytes during long-term primary cell culture revealed the expression patterns depending on the duration of the culture and the heart segment of origin (right atrial appendage and right atrium). Differentially expressed genes (DEGs) were classified as involved in ontological groups such as: “cellular component assembly”, “cellular component organization”, “cellular component biogenesis”, and “cytoskeleton organization”. Transcriptomic profiling allowed us to indicate the increased expression of COL5A2, COL8A1, and COL12A1, encoding different collagen subunits, pivotal in cardiac extracellular matrix (ECM) structure. Conversely, genes important for cellular architecture, such as ABLIM1, TMOD1, XIRP1, and PHACTR1, were downregulated during in vitro culture. The culture conditions may create a favorable environment for reconstruction of the ECM structures, whereas they may be suboptimal for expression of some pivotal transcripts responsible for the formation of intracellular structures.
Collapse
|
3
|
Plasma proteome of brain-dead organ donors predicts heart transplant outcome. J Heart Lung Transplant 2021; 41:311-324. [DOI: 10.1016/j.healun.2021.11.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/13/2022] Open
|
4
|
Valkov N, Das A, Tucker NR, Li G, Salvador AM, Chaffin MD, Pereira De Oliveira Junior G, Kur I, Gokulnath P, Ziegler O, Yeri A, Lu S, Khamesra A, Xiao C, Rodosthenous R, Srinivasan S, Toxavidis V, Tigges J, Laurent LC, Momma S, Kitchen R, Ellinor P, Ghiran I, Das S. SnRNA sequencing defines signaling by RBC-derived extracellular vesicles in the murine heart. Life Sci Alliance 2021; 4:4/12/e202101048. [PMID: 34663679 PMCID: PMC8548207 DOI: 10.26508/lsa.202101048] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 12/21/2022] Open
Abstract
In a unique model of fluorescent based mapping of EV recipient cells, RBC-EVs were found to signal to cardiac cells and regulate gene expression in a model of ischemic heart failure. Extracellular vesicles (EVs) mediate intercellular signaling by transferring their cargo to recipient cells, but the functional consequences of signaling are not fully appreciated. RBC-derived EVs are abundant in circulation and have been implicated in regulating immune responses. Here, we use a transgenic mouse model for fluorescence-based mapping of RBC-EV recipient cells to assess the role of this intercellular signaling mechanism in heart disease. Using fluorescent-based mapping, we detected an increase in RBC-EV–targeted cardiomyocytes in a murine model of ischemic heart failure. Single cell nuclear RNA sequencing of the heart revealed a complex landscape of cardiac cells targeted by RBC-EVs, with enrichment of genes implicated in cell proliferation and stress signaling pathways compared with non-targeted cells. Correspondingly, cardiomyocytes targeted by RBC-EVs more frequently express cellular markers of DNA synthesis, suggesting the functional significance of EV-mediated signaling. In conclusion, our mouse model for mapping of EV-recipient cells reveals a complex cellular network of RBC-EV–mediated intercellular communication in ischemic heart failure and suggests a functional role for this mode of intercellular signaling.
Collapse
Affiliation(s)
- Nedyalka Valkov
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Avash Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Nathan R Tucker
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA.,Masonic Medical Research Institute, Utica, NY, USA
| | - Guoping Li
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ane M Salvador
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Mark D Chaffin
- Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | | | - Ivan Kur
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Priyanka Gokulnath
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Olivia Ziegler
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ashish Yeri
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Shulin Lu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Aushee Khamesra
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Chunyang Xiao
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Srimeenakshi Srinivasan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | | | - John Tigges
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louise C Laurent
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Stefan Momma
- Institute of Neurology (Edinger Institute), University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Robert Kitchen
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Ionita Ghiran
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Saumya Das
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
5
|
Wu Q, Tian JH, He YX, Huang YY, Huang YQ, Zhang GP, Luo JD, Xue Q, Yu XY, Liu YH. Zonisamide alleviates cardiac hypertrophy in rats by increasing Hrd1 expression and inhibiting endoplasmic reticulum stress. Acta Pharmacol Sin 2021; 42:1587-1597. [PMID: 33495518 PMCID: PMC8463597 DOI: 10.1038/s41401-020-00585-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/17/2020] [Indexed: 02/02/2023] Open
Abstract
Antiepileptic drug zonisamide has been shown to be curative for Parkinson's disease (PD) through increasing HMG-CoA reductase degradation protein 1 (Hrd1) level and mitigating endoplasmic reticulum (ER) stress. Hrd1 is an ER-transmembrane E3 ubiquitin ligase, which is involved in cardiac dysfunction and cardiac hypertrophy in a mouse model of pressure overload. In this study, we investigated whether zonisamide alleviated cardiac hypertrophy in rats by increasing Hrd1 expression and inhibiting ER stress. The beneficial effects of zonisamide were assessed in two experimental models of cardiac hypertrophy: in rats subjected to abdominal aorta constriction (AAC) and treated with zonisamide (14, 28, 56 mg · kg-1 · d-1, i.g.) for 6 weeks as well as in neonatal rat cardiomyocytes (NRCMs) co-treated with Ang II (10 μM) and zonisamide (0.3 μM). Echocardiography analysis revealed that zonsiamide treatment significantly improved cardiac function in AAC rats. We found that zonsiamide treatment significantly attenuated cardiac hypertrophy and fibrosis, and suppressed apoptosis and ER stress in the hearts of AAC rats and in Ang II-treated NRCMs. Importantly, zonisamide markedly increased the expression of Hrd1 in the hearts of AAC rats and in Ang II-treated NRCMs. Furthermore, we demonstrated that zonisamide accelerated ER-associated protein degradation (ERAD) in Ang II-treated NRCMs; knockdown of Hrd1 abrogated the inhibitory effects of zonisamide on ER stress and cardiac hypertrophy. Taken together, our results demonstrate that zonisamide is effective in preserving heart structure and function in the experimental models of pathological cardiac hypertrophy. Zonisamide increases Hrd1 expression, thus preventing cardiac hypertrophy and improving the cardiac function of AAC rats.
Collapse
Affiliation(s)
- Qian Wu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jia-Hui Tian
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yong-Xiang He
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yong-Yin Huang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Qing Huang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Gui-Ping Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jian-Dong Luo
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qin Xue
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xi-Yong Yu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Ying-Hua Liu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
6
|
Jasso KR, Kamba TK, Zimmerman AD, Bansal R, Engle SE, Everett T, Wu CH, Kulaga H, Reed RR, Berbari NF, McIntyre JC. An N-terminal fusion allele to study melanin concentrating hormone receptor 1. Genesis 2021; 59:e23438. [PMID: 34124835 DOI: 10.1002/dvg.23438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/10/2022]
Abstract
Cilia on neurons play critical roles in both the development and function of the central nervous system (CNS). While it remains challenging to elucidate the precise roles for neuronal cilia, it is clear that a subset of G-protein-coupled receptors (GPCRs) preferentially localize to the cilia membrane. Further, ciliary GPCR signaling has been implicated in regulating a variety of behaviors. Melanin concentrating hormone receptor 1 (MCHR1), is a GPCR expressed centrally in rodents known to be enriched in cilia. Here we have used MCHR1 as a model ciliary GPCR to develop a strategy to fluorescently tag receptors expressed from the endogenous locus in vivo. Using CRISPR/Cas9, we inserted the coding sequence of the fluorescent protein mCherry into the N-terminus of Mchr1. Analysis of the fusion protein (mCherry MCHR1) revealed its localization to neuronal cilia in the CNS, across multiple developmental time points and in various regions of the adult brain. Our approach simultaneously produced fortuitous in/dels altering the Mchr1 start codon resulting in a new MCHR1 knockout line. Functional studies using electrophysiology show a significant alteration of synaptic strength in MCHR1 knockout mice. A reduction in strength is also detected in mice homozygous for the mCherry insertion, suggesting that while the strategy is useful for monitoring the receptor, activity could be altered. However, both lines should aid in studies of MCHR1 function and contribute to our understanding of MCHR1 signaling in the brain. Additionally, this approach could be expanded to aid in the study of other ciliary GPCRs.
Collapse
Affiliation(s)
- Kalene R Jasso
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA.,Graduate Program in Biomedical Sciences, Neuroscience Concentration, University of Florida, Gainesville, Florida, USA
| | - Tisianna K Kamba
- Graduate Program in Biomedical Sciences, Neuroscience Concentration, University of Florida, Gainesville, Florida, USA
| | - Arthur D Zimmerman
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Ruchi Bansal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Staci E Engle
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Thomas Everett
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Chang-Hung Wu
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| | - Heather Kulaga
- Department of Molecular Genetics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Randal R Reed
- Department of Molecular Genetics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicolas F Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana, USA
| | - Jeremy C McIntyre
- Department of Neuroscience and Center for Smell and Taste, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Tian JH, Wu Q, He YX, Shen QY, Rekep M, Zhang GP, Luo JD, Xue Q, Liu YH. Zonisamide, an antiepileptic drug, alleviates diabetic cardiomyopathy by inhibiting endoplasmic reticulum stress. Acta Pharmacol Sin 2021; 42:393-403. [PMID: 32647341 PMCID: PMC8026994 DOI: 10.1038/s41401-020-0461-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/11/2020] [Indexed: 02/06/2023] Open
Abstract
Endoplasmic reticulum stress (ER stress) plays a key role in the development of cardiac hypertrophy and diabetic cardiomyopathy (DCM). Zonisamide (ZNS) was originally developed as an antiepileptic drug. Studies have shown that ZNS suppresses ER stress-induced neuronal cell damage in the experimental models of Parkinson's disease. Herein, we investigated whether ZNS improved DCM by attenuating ER stress-induced apoptosis. C57BL/6J mice were fed with high-fat diet (HFD) and intraperitoneally injected with low-dose streptozotocin (STZ) to induce type 2 diabetes mellitus (T2DM), and then treated with ZNS (40 mg·kg-1·d-1, i.g.) for 16 weeks. We showed that ZNS administration slightly ameliorated the blood glucose levels, but significantly alleviated diabetes-induced cardiac dysfunction and hypertrophy. Furthermore, ZNS administration significantly inhibited the Bax and caspase-3 activity, upregulated Bcl-2 activity, and decreased the proportion of TUNEL-positive cells in heart tissues. We analyzed the hallmarks of ER stress in heart tissues, and revealed that ZNS administration significantly decreased the protein levels of GRP78, XBP-1s, ATF6, PERK, ATF4, and CHOP, and elevated Hrd1 protein. In high glucose (HG)-treated primary cardiomyocytes, application of ZNS (3 μM) significantly alleviated HG-induced cardiomyocyte hypertrophy and apoptosis. ZNS application also suppressed activated ER stress in HG-treated cardiomyocytes. Moreover, preapplication of the specific ER stress inducer tunicamycin (10 ng/mL) eliminated the protective effects of ZNS against HG-induced cardiac hypertrophy and ER stress-mediated apoptosis. Our findings suggest that ZNS improves the cardiac diastolic function in diabetic mice and prevents T2DM-induced cardiac hypertrophy by attenuating ER stress-mediated apoptosis.
Collapse
Affiliation(s)
- Jia-Hui Tian
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qian Wu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yong-Xiang He
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qi-Ying Shen
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mubarak Rekep
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Gui-Ping Zhang
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jian-Dong Luo
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qin Xue
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Ying-Hua Liu
- Department of Pharmacology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
8
|
Bain CR, Ziemann M, Kaspi A, Khan AW, Taylor R, Trahair H, Khurana I, Kaipananickal H, Wallace S, El-Osta A, Myles PS, Bozaoglu K. DNA methylation patterns from peripheral blood separate coronary artery disease patients with and without heart failure. ESC Heart Fail 2020; 7:2468-2478. [PMID: 32618141 PMCID: PMC7524212 DOI: 10.1002/ehf2.12810] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/11/2020] [Accepted: 05/14/2020] [Indexed: 01/01/2023] Open
Abstract
Aims Natriuretic peptides are useful for diagnosis and prognostication of heart failure of any cause. Now, research aims to discover novel biomarkers that will more specifically define the heart failure phenotype. DNA methylation plays a critical role in the development of cardiovascular disease with the potential to predict fundamental pathogenic processes. There is a lack of data relating DNA methylation in heart failure that specifically focuses on patients with severe multi‐vessel coronary artery disease. To begin to address this, we conducted a pilot study uniquely exploring the utility of powerful whole‐genome methyl‐binding domain‐capture sequencing in a cohort of cardiac surgery patients, matched for the severity of their coronary artery disease, aiming to identify candidate peripheral blood DNA methylation markers of ischaemic cardiomyopathy and heart failure. Methods and results We recruited a cohort of 20 male patients presenting for coronary artery bypass graft surgery with phenotypic extremes of heart failure but who otherwise share a similar coronary ischaemic burden, age, sex, and ethnicity. Methylation profiling in patient blood samples was performed using methyl‐binding domain‐capture sequencing. Differentially methylated regions were validated using targeted bisulfite sequencing. Gene set enrichment analysis was performed to identify differences in methylation at or near gene promoters in certain known Reactome pathways. We detected 567 188 methylation peaks of which our general linear model identified 68 significantly differentially methylated regions in heart failure with a false discovery rate <0.05. Of these regions, 48 occurred within gene bodies and 25 were located near enhancer elements, some within coding genes and some in non‐coding genes. Gene set enrichment analyses identified 103 significantly enriched gene sets (false discovery rate <0.05) in heart failure. Validation analysis of regions with the strongest differential methylation data was performed for two genes: HDAC9 and the uncharacterized miRNA gene MIR3675. Genes of particular interest as novel candidate markers of the heart failure phenotype with reduced methylation were HDAC9, JARID2, and GREM1 and with increased methylation PDSS2. Conclusions We demonstrate the utility of methyl‐binding domain‐capture sequencing to evaluate peripheral blood DNA methylation markers in a cohort of cardiac surgical patients with severe multi‐vessel coronary artery disease and phenotypic extremes of heart failure. The differential methylation status of specific coding genes identified are candidates for larger longitudinal studies. We have further demonstrated the value and feasibility of examining DNA methylation during the perioperative period to highlight biological pathways and processes contributing to complex phenotypes.
Collapse
Affiliation(s)
- Chris R Bain
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Anaesthesiology and Perioperative Medicine, The Alfred Hospital and Monash University, The Alfred Centre, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Mark Ziemann
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, Melbourne, VIC, Australia
| | - Antony Kaspi
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Rachael Taylor
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Hugh Trahair
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Ishant Khurana
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Harikrishnan Kaipananickal
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Department of Clinical Pathology, University of Melbourne, Melbourne, VIC, Australia
| | - Sophie Wallace
- Department of Anaesthesiology and Perioperative Medicine, The Alfred Hospital and Monash University, The Alfred Centre, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Assam El-Osta
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia.,Epigenetics in Human Health and Disease Laboratory, Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Shatin, Hong Kong SAR.,Faculty of Health, Department of Technology, Biomedical Laboratory Science, University College Copenhagen, Copenhagen, Denmark
| | - Paul S Myles
- Department of Anaesthesiology and Perioperative Medicine, The Alfred Hospital and Monash University, The Alfred Centre, Level 6, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, Australia
| | - Kiymet Bozaoglu
- Baker IDI Heart and Diabetes Institute, Melbourne, VIC, Australia.,Murdoch Children's Research Institute and Department of Paediatrics, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Melgari D, Barbier C, Dilanian G, Rücker-Martin C, Doisne N, Coulombe A, Hatem SN, Balse E. Microtubule polymerization state and clathrin-dependent internalization regulate dynamics of cardiac potassium channel: Microtubule and clathrin control of K V1.5 channel. J Mol Cell Cardiol 2020; 144:127-139. [PMID: 32445844 DOI: 10.1016/j.yjmcc.2020.05.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/19/2022]
Abstract
Ion channel trafficking powerfully influences cardiac electrical activity as it regulates the number of available channels at the plasma membrane. Studies have largely focused on identifying the molecular determinants of the trafficking of the atria-specific KV1.5 channel, the molecular basis of the ultra-rapid delayed rectifier current IKur. Besides, regulated KV1.5 channel recycling upon changes in homeostatic state and mechanical constraints in native cardiomyocytes has been well documented. Here, using cutting-edge imaging in live myocytes, we investigated the dynamics of this channel in the plasma membrane. We demonstrate that the clathrin pathway is a major regulator of the functional expression of KV1.5 channels in atrial myocytes, with the microtubule network as the prominent organizer of KV1.5 transport within the membrane. Both clathrin blockade and microtubule disruption result in channel clusterization with reduced membrane mobility and internalization, whereas disassembly of the actin cytoskeleton does not. Mobile KV1.5 channels are associated with the microtubule plus-end tracking protein EB1 whereas static KV1.5 clusters are associated with stable acetylated microtubules. In human biopsies from patients in atrial fibrillation associated with atrial remodeling, drastic modifications in the trafficking balance occurs together with alteration in microtubule polymerization state resulting in modest reduced endocytosis and increased recycling. Consequently, hallmark of atrial KV1.5 dynamics within the membrane is clathrin- and microtubule- dependent. During atrial remodeling, predominance of anterograde trafficking activity over retrograde trafficking could result in accumulation ok KV1.5 channels in the plasma membrane.
Collapse
Affiliation(s)
- Dario Melgari
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Camille Barbier
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Gilles Dilanian
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | | | - Nicolas Doisne
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Alain Coulombe
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France
| | - Stéphane N Hatem
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France; Institut de Cardiologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Elise Balse
- INSERM UMRS1166, ICAN - Institute of CardioMetabolism and Nutrition, Sorbonne Université, Paris, France.
| |
Collapse
|
10
|
Okabe K, Matsushima S, Ikeda S, Ikeda M, Ishikita A, Tadokoro T, Enzan N, Yamamoto T, Sada M, Deguchi H, Shinohara K, Ide T, Tsutsui H. DPP (Dipeptidyl Peptidase)-4 Inhibitor Attenuates Ang II (Angiotensin II)-Induced Cardiac Hypertrophy via GLP (Glucagon-Like Peptide)-1-Dependent Suppression of Nox (Nicotinamide Adenine Dinucleotide Phosphate Oxidase) 4-HDAC (Histone Deacetylase) 4 Pathway. Hypertension 2020; 75:991-1001. [PMID: 32160098 DOI: 10.1161/hypertensionaha.119.14400] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nox4 (NADPH [Nicotinamide adenine dinucleotide phosphate] oxidase 4) is a major source of oxidative stress and is intimately involved in cardiac hypertrophy. DPP (Dipeptidyl peptidase)-4 inhibitor has been reported to regulate Nox4 expression in adipose tissues. However, its effects on Nox4 in cardiac hypertrophy are still unclear. We investigated whether DPP-4 inhibitor could ameliorate cardiac hypertrophy by regulating Nox4 and its downstream targets. Ang II (Angiotensin II; 1.44 mg/kg per day) or saline was continuously infused into C57BL/6J mice with or without teneligliptin (a DPP-4 inhibitor, 30 mg/kg per day) in the drinking water for 1 week. Teneligliptin significantly suppressed plasma DPP-4 activity without any significant changing aortic blood pressure or metabolic parameters such as blood glucose and insulin levels. It attenuated Ang II-induced increases in left ventricular wall thickness and the ratio of heart weight to body weight. It also significantly suppressed Ang II-induced increases in Nox4 mRNA, 4-hydroxy-2-nonenal, and phosphorylation of HDAC4 (histone deacetylase 4), a downstream target of Nox4 and a crucial suppressor of cardiac hypertrophy, in the heart. Exendin-3 (150 pmol/kg per minute), a GLP-1 (glucagon-like peptide 1) receptor antagonist, abrogated these inhibitory effects of teneligliptin on Nox4, 4-hydroxy-2-nonenal, phosphorylation of HDAC4, and cardiac hypertrophy. In cultured neonatal cardiomyocytes, exendin-4 (100 nmol/L, 24 hours), a GLP-1 receptor agonist, ameliorated Ang II-induced cardiomyocyte hypertrophy and decreased in Nox4, 4-hydroxy-2-nonenal, and phosphorylation of HDAC4. Furthermore, exendin-4 prevented Ang II-induced decrease in nuclear HDAC4 in cardiomyocytes. In conclusion, GLP-1 receptor stimulation by DPP-4 inhibitor can attenuate Ang II-induced cardiac hypertrophy by suppressing of the Nox4-HDAC4 axis in cardiomyocytes.
Collapse
Affiliation(s)
- Kosuke Okabe
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Kyushu University Hospital, Fukuoka, Japan (S.M.)
| | - Soichiro Ikeda
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Masataka Ikeda
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Akihito Ishikita
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Tomonori Tadokoro
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Nobuyuki Enzan
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Taishi Yamamoto
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Masashi Sada
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Hiroko Deguchi
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| | - Keisuke Shinohara
- Department of Experimental and Clinical Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Japan (K.S., T.I.)
| | - Tomomi Ide
- Department of Experimental and Clinical Cardiovascular Medicine, Graduate School of Medical Sciences, Kyushu University, Japan (K.S., T.I.)
| | - Hiroyuki Tsutsui
- From the Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan (K.O., S.I., M.I., A.I., T.T., N.E., T.Y., M.S., H.D., H.T.)
| |
Collapse
|
11
|
Geng L, Wang S, Zhang F, Xiong K, Huang J, Zhao T, Shi D, Lv F, Li L, Liang D, Cui Y, Liu Y, Xie D, Chen YH. SNX17 (Sorting Nexin 17) Mediates Atrial Fibrillation Onset Through Endocytic Trafficking of the Kv1.5 (Potassium Voltage-Gated Channel Subfamily A Member 5) Channel. Circ Arrhythm Electrophysiol 2020; 12:e007097. [PMID: 30939909 DOI: 10.1161/circep.118.007097] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Kv1.5 (Potassium voltage-gated channel subfamily A member 5) has been regarded as a promising target of interventions for atrial fibrillation (AF). SNX17 (sorting nexin 17), a member of the SNXs (sorting nexin family), regulates the intracellular trafficking of membrane proteins through its FERM (four-point-one, ezrin, radixin, moesin) domain. However, whether SNX17 regulates the trafficking process of Kv1.5 remains unknown. METHODS A SNX17 knockout rat line was generated to test the role of SNX17 in atrial electrophysiology. The protein expression of SNX17 and membrane ion channels was detected by Western blotting. Electrophysiology changes in the atrial tissue and myocytes were analyzed by optical mapping and patch clamp, respectively. Acetylcholine and electrical stimulation were used to induce AF, and ECG recording was adopted to assess the influence of SNX17 deficiency on AF susceptibility. The spatial relationship between Kv1.5 and SNX17 was evaluated by immunostaining and confocal scanning, and the functional region of SNX17 regulating Kv1.5 trafficking was identified using plasmids with truncated SNX17 domains. RESULTS Embryonic death occurred in homozygous SNX17 knockout rats. SNX17 heterozygous rats survived, and the level of the SNX17 protein in the atrium was decreased by ≈50%. SNX17 deficiency increased the membrane expression of Kv1.5 and atria-specific ultrarapid delayed rectifier outward potassium current ( IKur) density, resulting in a shortened action potential duration, and eventually contributing to AF susceptibility. Mechanistically, SNX17 facilitated the endocytic sorting of Kv1.5 from the plasma membrane to early endosomes via the FERM domain. CONCLUSIONS SNX17 mediates susceptibility to AF by regulating endocytic sorting of the Kv1.5 channel through the FERM domain. SNX17 could be a potential target for the development of new drugs for AF.
Collapse
Affiliation(s)
- Li Geng
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Shuo Wang
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Fulei Zhang
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Ke Xiong
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Jian Huang
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Tingting Zhao
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Dan Shi
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Fei Lv
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Li Li
- Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Dandan Liang
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Yingyu Cui
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Yi Liu
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| | - Duanyang Xie
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,School of Life Science and Technology (D.X.), Tongji University, Shanghai, China
| | - Yi-Han Chen
- Key Laboratory of Arrhythmias of the Ministry of Education of East Hospital (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China.,Institute of Medical Genetics (L.G., S.W., F.Z., K.X., J.H., T.Z., D.S., F.L., L.L., D.L., Y.C., Y.L., D.X., Y.-H.C.), Tongji University, Shanghai, China
| |
Collapse
|
12
|
Irvin MR, Sitlani CM, Floyd JS, Psaty BM, Bis JC, Wiggins KL, Whitsel EA, Sturmer T, Stewart J, Raffield L, Sun F, Liu CT, Xu H, Cupples AL, Tanner RM, Rossing P, Smith A, Zilhão NR, Launer LJ, Noordam R, Rotter JI, Yao J, Li X, Guo X, Limdi N, Sundaresan A, Lange L, Correa A, Stott DJ, Ford I, Jukema JW, Gudnason V, Mook-Kanamori DO, Trompet S, Palmas W, Warren HR, Hellwege JN, Giri A, O'donnell C, Hung AM, Edwards TL, Ahluwalia TS, Arnett DK, Avery CL. Genome-Wide Association Study of Apparent Treatment-Resistant Hypertension in the CHARGE Consortium: The CHARGE Pharmacogenetics Working Group. Am J Hypertens 2019; 32:1146-1153. [PMID: 31545351 PMCID: PMC6856621 DOI: 10.1093/ajh/hpz150] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Only a handful of genetic discovery efforts in apparent treatment-resistant hypertension (aTRH) have been described. METHODS We conducted a case-control genome-wide association study of aTRH among persons treated for hypertension, using data from 10 cohorts of European ancestry (EA) and 5 cohorts of African ancestry (AA). Cases were treated with 3 different antihypertensive medication classes and had blood pressure (BP) above goal (systolic BP ≥ 140 mm Hg and/or diastolic BP ≥ 90 mm Hg) or 4 or more medication classes regardless of BP control (nEA = 931, nAA = 228). Both a normotensive control group and a treatment-responsive control group were considered in separate analyses. Normotensive controls were untreated (nEA = 14,210, nAA = 2,480) and had systolic BP/diastolic BP < 140/90 mm Hg. Treatment-responsive controls (nEA = 5,266, nAA = 1,817) had BP at goal (<140/90 mm Hg), while treated with one antihypertensive medication class. Individual cohorts used logistic regression with adjustment for age, sex, study site, and principal components for ancestry to examine the association of single-nucleotide polymorphisms with case-control status. Inverse variance-weighted fixed-effects meta-analyses were carried out using METAL. RESULTS The known hypertension locus, CASZ1, was a top finding among EAs (P = 1.1 × 10-8) and in the race-combined analysis (P = 1.5 × 10-9) using the normotensive control group (rs12046278, odds ratio = 0.71 (95% confidence interval: 0.6-0.8)). Single-nucleotide polymorphisms in this locus were robustly replicated in the Million Veterans Program (MVP) study in consideration of a treatment-responsive control group. There were no statistically significant findings for the discovery analyses including treatment-responsive controls. CONCLUSION This genomic discovery effort for aTRH identified CASZ1 as an aTRH risk locus.
Collapse
Affiliation(s)
- Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Epidemiology, University of Washington, Seattle, Washington, USA
- Department of Health Services, University of Washington, Seattle, Washington, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, Washington, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Eric A Whitsel
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Til Sturmer
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - James Stewart
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Laura Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Fangui Sun
- Department of Biostatistics, Boston University, Boston, Maryland, USA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University, Boston, Maryland, USA
| | - Hanfei Xu
- Department of Biostatistics, Boston University, Boston, Maryland, USA
| | | | - Rikki M Tanner
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | | - Albert Smith
- Icelandic Heart Association, Kopavogur, Iceland
- University of Iceland, Reykjavik, Iceland
| | | | - Lenore J Launer
- Laboratory of Epidemiology and Population Science, Intramural Research Program, National Institute on Aging, Bethesda, Maryland, USA
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Jie Yao
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Xiaohui Li
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Departments of Pediatrics and Medicine, LABioMed at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Nita Limdi
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Aishwarya Sundaresan
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Leslie Lange
- Department of Medicine, University of Colorado–Denver, Aurora, Colorado, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Ian Ford
- Robertson Center for Biostatistics, University of Glasgow, Glasgow, UK
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vilmundur Gudnason
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis O Mook-Kanamori
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Stella Trompet
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - Walter Palmas
- Department of Medicine, Columbia University Medical Center, New York, New York, USA
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, UK
| | - Jacklyn N Hellwege
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, Tennessee, USA
- Division of Genetic Medicine, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ayush Giri
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, Tennessee, USA
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt Genetics Institute, Vanderbilt Epidemiology Center, Institute for Medicine and Public Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Christopher O'donnell
- VA Boston Health Care System, Boston, Massachusetts, USA
- Section of Cardiology and Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Adriana M Hung
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, Tennessee, USA
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Todd L Edwards
- Biomedical Laboratory Research and Development, Tennessee Valley Healthcare System (626)/Vanderbilt University, Nashville, Tennessee, USA
- Division of Epidemiology, Department of Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tarunveer S Ahluwalia
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Donna K Arnett
- Deans Office, School of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Christy L Avery
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Lu D, Wang J, Li J, Guan F, Zhang X, Dong W, Liu N, Gao S, Zhang L. Meox1 accelerates myocardial hypertrophic decompensation through Gata4. Cardiovasc Res 2019; 114:300-311. [PMID: 29155983 DOI: 10.1093/cvr/cvx222] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 11/15/2017] [Indexed: 12/22/2022] Open
Abstract
Aims Pathological hypertrophy is the result of gene network regulation, which ultimately leads to adverse cardiac remodelling and heart failure (HF) and is accompanied by the reactivation of a 'foetal gene programme'. The Mesenchyme homeobox 1 (Meox1) gene is one of the foetal programme genes. Meox1 may play a role in embryonic development, but its regulation of pathological hypertrophy is not known. Therefore, this study investigated the effect of Meox1 on pathological hypertrophy, including familial and pressure overload-induced hypertrophy, and its potential mechanism of action. Methods and results Meox1 expression was markedly down-regulated in the wild-type adult mouse heart with age, and expression was up-regulated in heart tissues from familial dilated cardiomyopathy (FDCM) mice of the cTnTR141W strain, familial hypertrophic cardiomyopathy (FHCM) mice of the cTnTR92Q strain, pressure overload-induced HF mice, and hypertrophic cardiomyopathy (HCM) patients. Echocardiography, histopathology, and hypertrophic molecular markers consistently demonstrated that Meox1 overexpression exacerbated the phenotypes in FHCM and in mice with thoracic aorta constriction (TAC), and that Meox1 knockdown improved the pathological changes. Gata4 was identified as a potential downstream target of Meox1 using digital gene expression (DGE) profiling, real-time PCR, and bioinformatics analysis. Promoter activity data and chromatin immunoprecipitation (ChIP) and Gata4 knockdown analyses indicated that Meox1 acted via activation of Gata4 transcription. Conclusion Meox1 accelerated decompensation via the downstream target Gata4, at least in part directly. Meox1 and other foetal programme genes form a highly interconnected network, which offers multiple therapeutic entry points to dampen the aberrant expression of foetal genes and pathological hypertrophy.
Collapse
Affiliation(s)
- Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishilu, Beijing 100037, China
| | - Jing Li
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Ning Liu
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Shan Gao
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, NHFPC, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Building 5, Panjiayuan Nanli, Chaoyang District, Beijing 100021, China
| |
Collapse
|
14
|
Grassini DR, da Silva J, Hall TE, Baillie GJ, Simons C, Parton RG, Hogan BM, Smith KA. Myosin Vb is required for correct trafficking of N-cadherin and cardiac chamber ballooning. Dev Dyn 2019; 248:284-295. [PMID: 30801852 DOI: 10.1002/dvdy.19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 01/29/2019] [Accepted: 01/30/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND During heart morphogenesis, the cardiac chambers undergo ballooning: a process involving regionalized elongation of cardiomyocytes. Cardiomyocyte shape changes require reorganization of the actin cytoskeleton; however, the genetic regulation of this process is not well understood. RESULTS From a forward genetic screen, we identified the zebrafish uq 23ks mutant which manifests chamber ballooning defects. Whole-genome sequencing-mapping identified a truncating mutation in the gene, myo5b. myo5b encodes an atypical myosin required for endosome recycling and, consistent with this, increased vesicles were observed in myo5b mutant cardiomyocytes. Expression of RFP-Rab11a (a recycling endosome marker) confirmed increased recycling endosomes in cardiomyocytes of myo5b mutants. To investigate potential cargo of MyoVb-associated vesicles, we examined the adherens junction protein, N-cadherin. N-cadherin appeared mispatterned at cell junctions, and an increase in the number of intracellular particles was also apparent. Co-localization with RFP-Rab11a confirmed increased N-cadherin-positive recycling endosomes, demonstrating N-cadherin trafficking is perturbed in myo5b mutants. Finally, phalloidin staining showed disorganized F-actin in myo5b cardiomyocytes, suggesting the cytoskeleton fails to remodel, obstructing chamber ballooning. CONCLUSIONS MyoVb is required for cardiomyocyte endosomal recycling and appropriate N-cadherin localization during the onset of chamber ballooning. Cardiomyocytes lacking MyoVb are unable to reorganize their actin cytoskeleton, resulting in failed chamber ballooning. Developmental Dynamics 248:284-295, 2019. © 2019 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Daniela R Grassini
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Jason da Silva
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Thomas E Hall
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Gregory J Baillie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Cas Simons
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia.,Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Benjamin M Hogan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Kelly A Smith
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
15
|
Chen K, Rekep M, Wei W, Wu Q, Xue Q, Li S, Tian J, Yi Q, Zhang G, Zhang G, Xiao Q, Luo J, Liu Y. Quercetin Prevents In Vivo and In Vitro Myocardial Hypertrophy Through the Proteasome-GSK-3 Pathway. Cardiovasc Drugs Ther 2019; 32:5-21. [PMID: 29435775 DOI: 10.1007/s10557-018-6771-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE Quercetin, a flavonoid, has been reported to ameliorate cardiovascular diseases, such as cardiac hypertrophy. However, the mechanism is not completely understood. In this study, a mechanism related to proteasome-glycogen synthesis kinase 3 (GSK-3) was elucidated in rats and primary neonatal cardiomyocytes. METHODS Rats were subjected to sham or constriction of abdominal aorta surgery groups and treated with or without quercetin for 8 weeks. Angiotensin II (Ang II)-induced primary cardiomyocytes were cultured with quercetin treatment or not for 48 h. Echocardiography, real-time RT-PCR, histology, immunofluorescence, and Western blotting were conducted. Proteasome activities were also detected using a fluorescent peptide substrate. RESULTS Echocardiography showed that quercetin prevented constriction of abdominal aorta-induced cardiac hypertrophy and improved the cardiac diastolic function. In addition, quercetin also significantly reduced the Ang II-induced hypertrophic surface area and atrial natriuretic factor (ANF) mRNA level in primary cardiomyocytes. Proteasome activities were obviously inhibited in the quercetin-treated group both in vivo and in vitro. Quercetin also decreased the levels of proteasome subunit beta type (PSMB) 1, PSMB2, and PSMB5 of the 20S proteasome as well as the levels of proteasome regulatory particle (Rpt) 1 and Rpt4 of the 19S proteasome. In particular, the PSMB5 level in the nucleus was reduced after quercetin treatment. Furthermore, phosphorylated GSK-3α/β (inactivation of GSK-3) was decreased, which means that GSK-3 activity was increased. The phosphorylation levels of upstream AKT (PKB (protein kinase B)) and liver kinase B1/AMP activated protein kinase (LKB1/AMPKα) and those of downstream extracellular signal-regulated kinase (ERK), histone H3, β-catenin, and GATA binding protein 4 (GATA4) were reduced after quercetin treatment, while hypertrophy was reversed after treatment with the GSK-3 inhibitor. CONCLUSION In summary, quercetin prevents cardiac hypertrophy, which is related to proteasome inhibition and activation of GSK-3α/β. Upstream (AKT, LKB1/AMPKα) and downstream hypertrophic factors, such as ERK, histone H3, β-catenin, and GATA4, may also be involved.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Disease Models, Animal
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Hypertrophy, Left Ventricular/prevention & control
- Male
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Proteasome Endopeptidase Complex/drug effects
- Proteasome Endopeptidase Complex/metabolism
- Proteasome Inhibitors/pharmacology
- Quercetin/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Ventricular Function, Left/drug effects
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Kuixiang Chen
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
- Medical College of Jiaying University, Meizhou, 514031, China
| | - Mubarak Rekep
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wei Wei
- Key Laboratory of State Administration of Traditional Chinese Medicine of China, Department of Pathophysiology, School of Medicine, Institute of Brain Research, Jinan University, Guangzhou, 510632, China
| | - Qian Wu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qin Xue
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Sujuan Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiahui Tian
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Quan Yi
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Genshui Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Guiping Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qing Xiao
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiandong Luo
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yinghua Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Key Laboratory of Molecular Clinical Pharmacology and Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
16
|
Conrad R, Stölting G, Hendriks J, Ruello G, Kortzak D, Jordan N, Gensch T, Hidalgo P. Rapid Turnover of the Cardiac L-Type Ca V1.2 Channel by Endocytic Recycling Regulates Its Cell Surface Availability. iScience 2018; 7:1-15. [PMID: 30267672 PMCID: PMC6135870 DOI: 10.1016/j.isci.2018.08.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 07/18/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Calcium entry through CaV1.2 L-type calcium channels regulates cardiac contractility. Here, we study the impact of exocytic and post-endocytic trafficking on cell surface channel abundance in cardiomyocytes. Single-molecule localization and confocal microscopy reveal an intracellular CaV1.2 pool tightly associated with microtubules from the perinuclear region to the cell periphery, and with actin filaments at the cell cortex. Channels newly inserted into the plasma membrane become internalized with an average time constant of 7.5 min and are sorted out to the Rab11a-recycling compartment. CaV1.2 recycling suffices for maintaining stable L-type current amplitudes over 20 hr independent of de novo channel transport along microtubules. Disruption of the actin cytoskeleton re-routes CaV1.2 from recycling toward lysosomal degradation. We identify endocytic recycling as essential for the homeostatic regulation of voltage-dependent calcium influx into cardiomyocytes. This mechanism provides the basis for a dynamic adjustment of the channel's surface availability and thus, of heart's contraction.
Collapse
Affiliation(s)
- Rachel Conrad
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Gabriel Stölting
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Johnny Hendriks
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Giovanna Ruello
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Daniel Kortzak
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Nadine Jordan
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Thomas Gensch
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Patricia Hidalgo
- Institute of Complex Systems 4, Zelluläre Biophysik, Forschungszentrum Jülich, 52425 Jülich, Germany; Institute of Biochemistry, Heinrich-Heine University, Düsseldorf, Germany.
| |
Collapse
|
17
|
Zhao X, Zhang W, Ji W. MYO5A inhibition by miR-145 acts as a predictive marker of occult neck lymph node metastasis in human laryngeal squamous cell carcinoma. Onco Targets Ther 2018; 11:3619-3635. [PMID: 29950866 PMCID: PMC6016585 DOI: 10.2147/ott.s164597] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION Each year, ~50,000 patients worldwide die of laryngeal squamous cell carcinoma (LSCC) because of its highly metastatic properties. However, its pathogenic mechanisms are still unclear, and in particular, the prediction of metastasis remains elusive. This study aimed to define the role of microRNA-145 (miR-145) in LSCC progression. We also aimed to elucidate the clinical significance of the miR-145/MYO5A pathway, especially the predictive function of MYO5A in neck lymph node metastasis. MATERIALS AND METHODS MYO5A and miR-145 expression was analyzed in 132 patients with LSCC, and associations between their expression and clinicopathological features were evaluated. We validated the regulatory relationship between miR-145b and MYO5A by dual luciferase reporter assay. The role of the miR-145/MYO5A pathway in proliferation, metastasis, and apoptosis was examined in vitro. The predictive functions of MYO5A in neck lymph node metastasis and prognosis were defined according to patient follow-up. RESULTS Our results showed downregulation of miR-145 in LSCC, which was negatively correlated with MYO5A suppression of LSCC progression and metastasis. MiR-145 directly regulated MYO5A expression in vitro and suppressed LSCC proliferation and invasion while promoting apoptosis by inhibiting MYO5A. CONCLUSION Notably, overexpression of serum MYO5A in LSCC predicted cervical nodal occult metastasis and poor prognosis, providing an effective indicator for predicting neck lymph node metastasis and assessing LSCC prognosis.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of Otorhinolaryngology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wei Zhang
- Department of Endocrinology, Shengjing Hospital, China Medical University, Shenyang, China
| | - Wenyue Ji
- Department of Otorhinolaryngology, Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
18
|
Sabour D, Machado RSR, Pinto JP, Rohani S, Sahito RGA, Hescheler J, Futschik ME, Sachinidis A. Parallel Genome-wide Profiling of Coding and Non-coding RNAs to Identify Novel Regulatory Elements in Embryonic and Maturated Heart. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:158-173. [PMID: 30195755 PMCID: PMC6023836 DOI: 10.1016/j.omtn.2018.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 04/30/2018] [Accepted: 04/30/2018] [Indexed: 12/18/2022]
Abstract
Heart development is a complex process, tightly regulated by numerous molecular mechanisms. Key components of the regulatory network underlying heart development are transcription factors (TFs) and microRNAs (miRNAs), yet limited investigation of the role of miRNAs in heart development has taken place. Here, we report the first parallel genome-wide profiling of polyadenylated RNAs and miRNAs in a developing murine heart. These data enable us to identify dynamic activation or repression of numerous biological processes and signaling pathways. More than 200 miRNAs and 25 long non-coding RNAs were differentially expressed during embryonic heart development compared to the mature heart; most of these had not been previously associated with cardiogenesis. Integrative analysis of expression data and potential regulatory interactions suggested 28 miRNAs as novel regulators of embryonic heart development, representing a considerable expansion of the current repertoire of known cardiac miRNAs. To facilitate follow-up investigations, we constructed HeartMiR (http://heartmir.sysbiolab.eu), an open access database and interactive visualization tool for the study of gene regulation by miRNAs during heart development.
Collapse
Affiliation(s)
- Davood Sabour
- University of Cologne (UKK), Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany; Department of Genetics, Faculty of Medicine, Babol University of Medical Sciences, 47134 Babol, Iran
| | - Rui S R Machado
- Systems Biology and Bioinformatics Laboratory (SysBioLab), Center for Biomedical Research (CBMR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - José P Pinto
- Systems Biology and Bioinformatics Laboratory (SysBioLab), Center for Biomedical Research (CBMR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal
| | - Susan Rohani
- University of Cologne (UKK), Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Raja G A Sahito
- University of Cologne (UKK), Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Jürgen Hescheler
- University of Cologne (UKK), Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Matthias E Futschik
- Systems Biology and Bioinformatics Laboratory (SysBioLab), Center for Biomedical Research (CBMR), University of Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; Centre of Marine Sciences (CCMAR), University of Algarve, 8005-139 Faro, Portugal; School of Biomedical Sciences, Faculty of Medicine and Dentistry, Institute of Translational and Stratified Medicine (ITSMED), University of Plymouth, Plymouth PL6 8BU, UK.
| | - Agapios Sachinidis
- University of Cologne (UKK), Institute of Neurophysiology and Center for Molecular Medicine Cologne (CMMC), Robert-Koch-Str. 39, 50931 Cologne, Germany.
| |
Collapse
|
19
|
Howarth FC, Qureshi MA, Jayaprakash P, Parekh K, Oz M, Dobrzynski H, Adrian TE. The Pattern of mRNA Expression Is Changed in Sinoatrial Node from Goto-Kakizaki Type 2 Diabetic Rat Heart. J Diabetes Res 2018; 2018:8454078. [PMID: 30246030 PMCID: PMC6139199 DOI: 10.1155/2018/8454078] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/16/2018] [Accepted: 08/12/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND In vivo experiments in Goto-Kakizaki (GK) type 2 diabetic rats have demonstrated reductions in heart rate from a young age. The expression of genes encoding more than 70 proteins that are associated with the generation and conduction of electrical activity in the GK sinoatrial node (SAN) have been evaluated to further clarify the molecular basis of the low heart rate. MATERIALS AND METHODS Heart rate and expression of genes were evaluated with an extracellular electrode and real-time RT-PCR, respectively. Rats aged 12-13 months were employed in these experiments. RESULTS Isolated spontaneous heart rate was reduced in GK heart (161 ± 12 bpm) compared to controls (229 ± 11 bpm). There were many differences in expression of mRNA, and some of these differences were of particular interest. Compared to control SAN, expression of some genes were downregulated in GK-SAN: gap junction, Gja1 (Cx43), Gja5 (Cx40), Gjc1 (Cx45), and Gjd3 (Cx31.9); cell membrane transport, Trpc1 (TRPC1) and Trpc6 (TRPC6); hyperpolarization-activated cyclic nucleotide-gated channels, Hcn1 (HCN1) and Hcn4 (HCN4); calcium channels, Cacna1d (Cav1.3), Cacna1g (Cav3.1), Cacna1h (Cav3.2), Cacna2d1 (Cavα2δ1), Cacna2d3 (Cavα2δ3), and Cacng4 (Cav γ 4); and potassium channels, Kcna2 (Kv1.2), Kcna4 (Kv1.4), Kcna5 (Kv1.5), Kcnb1 (Kv2.1), Kcnd3 (Kv4.3), Kcnj2 (Kir2.1), Kcnk1 (TWIK1), Kcnk5 (K2P5.1), Kcnk6 (TWIK2), and Kcnn2 (SK2) whilst others were upregulated in GK-SAN: Ryr2 (RYR2) and Nppb (BNP). CONCLUSIONS This study provides new insight into the changing expression of genes in the sinoatrial node of diabetic heart.
Collapse
MESH Headings
- Action Potentials
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/physiopathology
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/metabolism
- Diabetic Cardiomyopathies/physiopathology
- Disease Models, Animal
- Gene Expression Regulation
- Heart Rate/genetics
- Isolated Heart Preparation
- Male
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Wistar
- Sinoatrial Node/metabolism
- Sinoatrial Node/physiopathology
Collapse
Affiliation(s)
- F. C. Howarth
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - M. A. Qureshi
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - P. Jayaprakash
- Department of Pharmacology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - K. Parekh
- Department of Physiology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - M. Oz
- Department of Pharmacology, College of Medicine & Health Sciences, UAE University, Al Ain, UAE
| | - H. Dobrzynski
- Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - T. E. Adrian
- Department of Basic Medical Sciences, Mohammed Bin Rashid University of Medicine & Health Sciences, Dubai, UAE
| |
Collapse
|
20
|
The inhibitory effects of levo-tetrahydropalmatine on rat Kv1.5 channels expressed in HEK293 cells. Eur J Pharmacol 2017; 809:105-110. [PMID: 28502629 DOI: 10.1016/j.ejphar.2017.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 05/05/2017] [Accepted: 05/10/2017] [Indexed: 01/07/2023]
Abstract
Levo-tetrahydropalmatine (l-THP) exerts various pharmacological effects on neural and cardiac tissues and K+ channel can be one of its multiple targets. The rapidly activating Kv1.5 channel is expressed in a variety of tissues including atrial cells and hippocampal neurons, and has an essential role in tuning the action potential and excitability in those cells. The aim of current study is to explore whether there are the possible effects of l-THP on Kv1.5 channels expressed in HEK293 cells. Superfusion of l-THP led to a dose-dependent blockage of Kv1.5 currents with an IC50 value of 53.2μM. This blocking effect was substantially attenuated in mutant H452G rather than R476V and R476Y, suggesting a specific binding site in the outer mouth region. In addition, the properties of Kv1.5 channel kinetics were markedly altered by l-THP. Treatment with l-THP resulted in a potential left shift of the inactivation curve, with the half-maximum inactivation potential (V1/2) of 4.5mV in control and -12.8mV in 50μM l-THP. Our data reveal that l-THP can exert an inhibitory effect on the delayed rectifier Kv1.5 channels expressed in HEK293 cells. These lines of evidence provided an insight to understand the possible effects exerted by l-THP on relative tissues.
Collapse
|
21
|
Inhibitory effects of cholinesterase inhibitor donepezil on the Kv1.5 potassium channel. Sci Rep 2017; 7:41509. [PMID: 28198801 PMCID: PMC5304190 DOI: 10.1038/srep41509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 12/21/2016] [Indexed: 11/25/2022] Open
Abstract
Kv1.5 channels carry ultra-rapid delayed rectifier K+ currents in excitable cells, including neurons and cardiac myocytes. In the current study, the effects of cholinesterase inhibitor donepezil on cloned Kv1.5 channels expressed in HEK29 cells were explored using whole-cell recording technique. Exposure to donepezil resulted in a rapid and reversible block of Kv1.5 currents, with an IC50 value of 72.5 μM. The mutant R476V significantly reduced the binding affinity of donepezil to Kv1.5 channels, showing the target site in the outer mouth region. Donepezil produced a significant delay in the duration of activation and deactivation, and mutant R476V potentiated these effects without altering activation curves. In response to slowed deactivation time course, a typical crossover of Kv1.5 tail currents was clearly evident after bath application of donepezil. In addition, both this chemical and mutant R476V accelerated current decay during channel inactivation in a voltage-dependent way, but barely changed the inactivation and recovery curves. The presence of donepezil exhibited the use-dependent block of Kv1.5 currents in response to a series of depolarizing pulses. Our data indicate that donepezil can directly block Kv1.5 channels in its open and closed states.
Collapse
|
22
|
Farquhar RE, Rodrigues E, Hamilton KL. The Role of the Cytoskeleton and Myosin-Vc in the Targeting of KCa3.1 to the Basolateral Membrane of Polarized Epithelial Cells. Front Physiol 2017; 7:639. [PMID: 28101059 PMCID: PMC5209343 DOI: 10.3389/fphys.2016.00639] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 12/06/2016] [Indexed: 12/27/2022] Open
Abstract
Understanding the targeting of KCa3.1 to the basolateral membrane (BLM) of polarized epithelial cells is still emerging. Here, we examined the role of the cytoskeleton (microtubules and microfilaments) and Myosin-Vc (Myo-Vc) in the targeting of KCa3.1 in Fischer rat thyroid epithelial cells. We used a pharmacological approach with immunoblot (for the BLM expression of KCa3.1), Ussing chamber (functional BLM expression of KCa3.1) and siRNA experiments. The actin cytoskeleton inhibitors cytochalasin D (10 μM, 5 h) and latrunculin A (10 μM, 5 h) reduced the targeting of KCa3.1 to the BLM by 88 ± 4 and 70 ± 5%, respectively. Colchicine (10 μM, 5 h) a microtubule inhibitor reduced targeting of KCa3.1 to the BLM by 63 ± 7% and decreased 1-EBIO-stimulated KCa3.1 K+ current by 46 ± 18%, compared with control cells. ML9 (10 μM, 5 h), an inhibitor of myosin light chain kinase, decreased targeting of the channel by 83 ± 2% and reduced K+ current by 54 ± 8% compared to control cells. Inhibiting Myo-V with 2,3-butanedione monoxime (10 mM, 5 h) reduced targeting of the channel to the BLM by 58 ± 5% and decreased the stimulated current of KCa3.1 by 48 ± 12% compared with control cells. Finally, using siRNA for Myo-Vc, we demonstrated that knockdown of Myo-Vc reduced the BLM expression of KCa3.1 by 44 ± 7% and KCa3.1 K+ current by 1.04 ± 0.14 μA compared with control cells. These data suggest that the microtubule and microfilament cytoskeleton and Myo-Vc are critical for the targeting of KCa3.1.
Collapse
Affiliation(s)
- Rachel E Farquhar
- Department of Physiology, Otago School of Medical Sciences, University of Otago Dunedin, New Zealand
| | - Ely Rodrigues
- Department of Medicine, Otago School of Medical Sciences, University of Otago Dunedin, New Zealand
| | - Kirk L Hamilton
- Department of Physiology, Otago School of Medical Sciences, University of Otago Dunedin, New Zealand
| |
Collapse
|
23
|
McDowell GS, Lemire JM, Paré JF, Cammarata G, Lowery LA, Levin M. Conserved roles for cytoskeletal components in determining laterality. Integr Biol (Camb) 2016; 8:267-86. [PMID: 26928161 DOI: 10.1039/c5ib00281h] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consistently-biased left-right (LR) patterning is required for the proper placement of organs including the heart and viscera. The LR axis is especially fascinating as an example of multi-scale pattern formation, since here chiral events at the subcellular level are integrated and amplified into asymmetric transcriptional cascades and ultimately into the anatomical patterning of the entire body. In contrast to the other two body axes, there is considerable controversy about the earliest mechanisms of embryonic laterality. Many molecular components of asymmetry have not been widely tested among phyla with diverse bodyplans, and it is unknown whether parallel (redundant) pathways may exist that could reverse abnormal asymmetry states at specific checkpoints in development. To address conservation of the early steps of LR patterning, we used the Xenopus laevis (frog) embryo to functionally test a number of protein targets known to direct asymmetry in plants, fruit fly, and rodent. Using the same reagents that randomize asymmetry in Arabidopsis, Drosophila, and mouse embryos, we show that manipulation of the microtubule and actin cytoskeleton immediately post-fertilization, but not later, results in laterality defects in Xenopus embryos. Moreover, we observed organ-specific randomization effects and a striking dissociation of organ situs from effects on the expression of left side control genes, which parallel data from Drosophila and mouse. Remarkably, some early manipulations that disrupt laterality of transcriptional asymmetry determinants can be subsequently "rescued" by the embryo, resulting in normal organ situs. These data reveal the existence of novel corrective mechanisms, demonstrate that asymmetric expression of Nodal is not a definitive marker of laterality, and suggest the existence of amplification pathways that connect early cytoskeletal processes to control of organ situs bypassing Nodal. Counter to alternative models of symmetry breaking during neurulation (via ciliary structures absent in many phyla), our data suggest a widely-conserved role for the cytoskeleton in regulating left-right axis formation immediately after fertilization of the egg. The novel mechanisms that rescue organ situs, even after incorrect expression of genes previously considered to be left-side master regulators, suggest LR patterning as a new context in which to explore multi-scale redundancy and integration of patterning from the subcellular structure to the entire bodyplan.
Collapse
Affiliation(s)
- Gary S McDowell
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155-4243, USA. and Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Joan M Lemire
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155-4243, USA.
| | - Jean-Francois Paré
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155-4243, USA.
| | | | | | - Michael Levin
- Biology Department, and Center for Regenerative and Developmental Biology, Tufts University, 200 Boston Avenue, Suite 4600, Medford, MA 02155-4243, USA.
| |
Collapse
|
24
|
The inhibitory effects of nifedipine on outward voltage-gated potassium currents in mouse neuroblastoma N2A cells. Pharmacol Rep 2016; 68:631-7. [DOI: 10.1016/j.pharep.2015.12.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/17/2015] [Accepted: 12/18/2015] [Indexed: 11/22/2022]
|
25
|
Chen-Izu Y, Shaw RM, Pitt GS, Yarov-Yarovoy V, Sack JT, Abriel H, Aldrich RW, Belardinelli L, Cannell MB, Catterall WA, Chazin WJ, Chiamvimonvat N, Deschenes I, Grandi E, Hund TJ, Izu LT, Maier LS, Maltsev VA, Marionneau C, Mohler PJ, Rajamani S, Rasmusson RL, Sobie EA, Clancy CE, Bers DM. Na+ channel function, regulation, structure, trafficking and sequestration. J Physiol 2015; 593:1347-60. [PMID: 25772290 DOI: 10.1113/jphysiol.2014.281428] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 11/02/2014] [Indexed: 12/19/2022] Open
Abstract
This paper is the second of a series of three reviews published in this issue resulting from the University of California Davis Cardiovascular Symposium 2014: Systems approach to understanding cardiac excitation-contraction coupling and arrhythmias: Na(+) channel and Na(+) transport. The goal of the symposium was to bring together experts in the field to discuss points of consensus and controversy on the topic of sodium in the heart. The present review focuses on Na(+) channel function and regulation, Na(+) channel structure and function, and Na(+) channel trafficking, sequestration and complexing.
Collapse
Affiliation(s)
- Ye Chen-Izu
- Department of Pharmacology, University of California, Davis, USA; Department of Biomedical Engineering, University of California, Davis, USA; Department of Internal Medicine/Cardiology, University of California, Davis, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Identification of Region-Specific Myocardial Gene Expression Patterns in a Chronic Swine Model of Repaired Tetralogy of Fallot. PLoS One 2015; 10:e0134146. [PMID: 26252659 PMCID: PMC4529093 DOI: 10.1371/journal.pone.0134146] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022] Open
Abstract
Surgical repair of Tetralogy of Fallot (TOF) is highly successful but may be complicated in adulthood by arrhythmias, sudden death, and right ventricular or biventricular dysfunction. To better understand the molecular and cellular mechanisms of these delayed cardiac events, a chronic animal model of postoperative TOF was studied using microarrays to perform cardiac transcriptomic studies. The experimental study included 12 piglets (7 rTOF and 5 controls) that underwent surgery at age 2 months and were further studied after 23 (+/- 1) weeks of postoperative recovery. Two distinct regions (endocardium and epicardium) from both ventricles were analyzed. Expression levels from each localization were compared in order to decipher mechanisms and signaling pathways leading to ventricular dysfunction and arrhythmias in surgically repaired TOF. Several genes were confirmed to participate in ventricular remodeling and cardiac failure and some new candidate genes were described. In particular, these data pointed out FRZB as a heart failure marker. Moreover, calcium handling and contractile function genes (SLN, ACTC1, PLCD4, PLCZ), potential arrhythmia-related genes (MYO5B, KCNA5), and cytoskeleton and cellular organization-related genes (XIRP2, COL8A1, KCNA6) were among the most deregulated genes in rTOF ventricles. To our knowledge, this is the first comprehensive report on global gene expression profiling in the heart of a long-term swine model of repaired TOF.
Collapse
|
27
|
Xiao S, Shaw RM. Cardiomyocyte protein trafficking: Relevance to heart disease and opportunities for therapeutic intervention. Trends Cardiovasc Med 2014; 25:379-89. [PMID: 25649302 DOI: 10.1016/j.tcm.2014.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 12/18/2014] [Accepted: 12/20/2014] [Indexed: 11/30/2022]
Abstract
Cardiomyocytes, the individual contractile units of heart muscle, are long-lived and robust. Given the longevity of these cells, it can be easy to overlook their dynamic intracellular environment that contain rapid protein movements and frequent protein turnover. Critical gene transcription and protein translation occur continuously, as well as trafficking and localization of proteins to specific functional zones of cell membrane. As heart failure becomes an increasingly important clinical entity, growing numbers of investigative teams are examining the cell biology of healthy and diseased cardiomyocytes. In this review, we introduce the major architectural structures and types of protein movements within cardiac cells, and then review recent studies that explore the regulation of such movements. We conclude by introducing current translational directions of the basic studies with a focus on novel areas of therapeutic development.
Collapse
Affiliation(s)
- Shaohua Xiao
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Robin M Shaw
- Heart Institute and Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA; Department of Medicine, University of California Los Angeles, Los Angeles, CA.
| |
Collapse
|
28
|
Dun W, Wright P, Danilo P, Mohler PJ, Boyden PA. SAP97 and cortactin remodeling in arrhythmogenic Purkinje cells. PLoS One 2014; 9:e106830. [PMID: 25184222 PMCID: PMC4153673 DOI: 10.1371/journal.pone.0106830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/01/2014] [Indexed: 11/18/2022] Open
Abstract
Because structural remodeling of several proteins, including ion channels, may underlie the abnormal action potentials of Purkinje cells (PCs) that survive in the 48 hr infarcted zone of the canine heart (IZPCs), we sought to determine the subcellular structure and function of the KV1.5 (KCNA5) protein in single IZPCs. Clustering of the Kv1.5 subunit in axons is regulated by a synapse-associated protein, SAP97, and is linked to an actin-binding protein, cortactin, and an intercellular adhesion molecule, N-cadherin. To understand the functional remodeling of the Kv1.5 channel and its regulation in IZPCs, Kv1.5 currents in PCs were measured as the currents blocked by 10 µM RSD1379 using patch-clamp techniques. Immunocytochemistry and confocal imaging were used for both single and aggregated IZPCs vs normal PCs (NZPCs) to determine the relationship of Kv1.5 with SAP-97, cortactin and N-cadherin. In IZPCs, both the sarcolemma (SL) and intercalated disk (ID) Kv1.5 protein are abundant, and the amount of cytosolic Kv1.5 protein is greatly increased. SAP-97 is also increased at IDs and has notable cytosolic localization suggesting that SAP-97 may regulate the functional expression and stabilization of Kv1.5 channels in IZPCs. Cortactin, which is located with N-cadherin at IDs in NZPCs, remains at IDs but begins to dissociate from N-cadherin, often forming ring structures and colocalizing with Kv1.5 within IZPCs. At the same time, cortactin/Kv1.5 colocalization is increased at the ID, suggesting an ongoing active process of membrane trafficking of the channel protein. Finally, the Kv1.5 current, measured as the RSD1379-sensitive current, at +40 mV did not differ between NZPCs (0.81±0.24 pA/pF, n = 14) and IZPCs (0.83±0.21 pA/pF, n = 13, NS). In conclusion, the subcellular structural remodeling of Kv1.5, SAP97 and cortactin maintained and normalized the function of the Kv1.5 channel in Purkinje cells that survived myocardial infarction.
Collapse
Affiliation(s)
- Wen Dun
- Department of Pharmacology, Center for Molecular Therapeutics, Columbia University, New York, New York, United States of America
| | - Patrick Wright
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Peter Danilo
- Department of Pharmacology, Center for Molecular Therapeutics, Columbia University, New York, New York, United States of America
| | - Peter J. Mohler
- The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Penelope A. Boyden
- Department of Pharmacology, Center for Molecular Therapeutics, Columbia University, New York, New York, United States of America
- * E-mail:
| |
Collapse
|