1
|
Zhang Y, Zeng Y, Bai H, Zhang W, Xue Z, Hu S, Lu S, Wang N. Depression of Ca V1.2 activation and expression in mast cells ameliorates allergic inflammation diseases. J Pharm Anal 2024; 14:101149. [PMID: 39720622 PMCID: PMC11667708 DOI: 10.1016/j.jpha.2024.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Allergic inflammation is closely related to the activation of mast cells (MCs), which is regulated by its intracellular Ca2+ level, but the intake and effects of the intracellular Ca2+ remain unclear. The Ca2+ influx is controlled by members of Ca2+ channels, among which calcium voltage-gated channel subunit alpha1 C (CaV1.2) is the most robust. This study aimed to reveal the role and underlying mechanism of MC CaV1.2 in allergic inflammation. We found that CaV1.2 participated in MC activation and allergic inflammation. Nimodipine (Nim), as a strong CaV1.2-specific antagonist, ameliorated allergic inflammation in mice. Further, CaV1.2 activation in MC was triggered by phosphatizing at its Ser1928 through protein kinase C (PKC), which calcium/calmodulin-dependent protein kinase II (CaMKII) catalyzed. Overexpression or knockdown of MC CaV1.2 influenced MC activation. Importantly, CaV1.2 expression in MC had detrimental effects, while its deficiency ameliorated allergic pulmonary inflammation. Results provide novel insights into CaV1.2 function and a potential drug target for controlling allergic inflammation.
Collapse
Affiliation(s)
- Yongjing Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yingnan Zeng
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Haoyun Bai
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Wen Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhuoyin Xue
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shiling Hu
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shemin Lu
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Nan Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, 710061, China
| |
Collapse
|
2
|
Hou W, Yin S, Li P, Zhang L, Chen T, Qin D, Mustafa AU, Liu C, Song M, Qiu C, Xiong X, Wang J. Aberrant splicing of Ca V1.2 calcium channel induced by decreased Rbfox1 enhances arterial constriction during diabetic hyperglycemia. Cell Mol Life Sci 2024; 81:164. [PMID: 38575795 PMCID: PMC10995029 DOI: 10.1007/s00018-024-05198-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 04/06/2024]
Abstract
Diabetic hyperglycemia induces dysfunctions of arterial smooth muscle, leading to diabetic vascular complications. The CaV1.2 calcium channel is one primary pathway for Ca2+ influx, which initiates vasoconstriction. However, the long-term regulation mechanism(s) for vascular CaV1.2 functions under hyperglycemic condition remains unknown. Here, Sprague-Dawley rats fed with high-fat diet in combination with low dose streptozotocin and Goto-Kakizaki (GK) rats were used as diabetic models. Isolated mesenteric arteries (MAs) and vascular smooth muscle cells (VSMCs) from rat models were used to assess K+-induced arterial constriction and CaV1.2 channel functions using vascular myograph and whole-cell patch clamp, respectively. K+-induced vasoconstriction is persistently enhanced in the MAs from diabetic rats, and CaV1.2 alternative spliced exon 9* is increased, while exon 33 is decreased in rat diabetic arteries. Furthermore, CaV1.2 channels exhibit hyperpolarized current-voltage and activation curve in VSMCs from diabetic rats, which facilitates the channel function. Unexpectedly, the application of glycated serum (GS), mimicking advanced glycation end-products (AGEs), but not glucose, downregulates the expression of the splicing factor Rbfox1 in VSMCs. Moreover, GS application or Rbfox1 knockdown dynamically regulates alternative exons 9* and 33, leading to facilitated functions of CaV1.2 channels in VSMCs and MAs. Notably, GS increases K+-induced intracellular calcium concentration of VSMCs and the vasoconstriction of MAs. These results reveal that AGEs, not glucose, long-termly regulates CaV1.2 alternative splicing events by decreasing Rbfox1 expression, thereby enhancing channel functions and increasing vasoconstriction under diabetic hyperglycemia. This study identifies the specific molecular mechanism for enhanced vasoconstriction under hyperglycemia, providing a potential target for managing diabetic vascular complications.
Collapse
Affiliation(s)
- Wei Hou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China
| | - Shumin Yin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Pengpeng Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ludan Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tiange Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dongxia Qin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Atta Ul Mustafa
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Caijie Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Miaomiao Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Qiu
- Nanjing Comprehensive Stroke Center, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqing Xiong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China.
| | - Juejin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China.
- The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu, China.
| |
Collapse
|
3
|
Li P, Qin D, Chen T, Hou W, Song X, Yin S, Song M, Fernando WCHA, Chen X, Sun Y, Wang J. Dysregulated Rbfox2 produces aberrant splicing of Ca V1.2 calcium channel in diabetes-induced cardiac hypertrophy. Cardiovasc Diabetol 2023; 22:168. [PMID: 37415128 PMCID: PMC10324275 DOI: 10.1186/s12933-023-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023] Open
Abstract
BACKGROUND L-type Ca2+ channel CaV1.2 is essential for cardiomyocyte excitation, contraction and gene transcription in the heart, and abnormal functions of cardiac CaV1.2 channels are presented in diabetic cardiomyopathy. However, the underlying mechanisms are largely unclear. The functions of CaV1.2 channels are subtly modulated by splicing factor-mediated alternative splicing (AS), but whether and how CaV1.2 channels are alternatively spliced in diabetic heart remains unknown. METHODS Diabetic rat models were established by using high-fat diet in combination with low dose streptozotocin. Cardiac function and morphology were assessed by echocardiography and HE staining, respectively. Isolated neonatal rat ventricular myocytes (NRVMs) were used as a cell-based model. Cardiac CaV1.2 channel functions were measured by whole-cell patch clamp, and intracellular Ca2+ concentration was monitored by using Fluo-4 AM. RESULTS We find that diabetic rats develop diastolic dysfunction and cardiac hypertrophy accompanied by an increased CaV1.2 channel with alternative exon 9* (CaV1.2E9*), but unchanged that with alternative exon 8/8a or exon 33. The splicing factor Rbfox2 expression is also increased in diabetic heart, presumably because of dominate-negative (DN) isoform. Unexpectedly, high glucose cannot induce the aberrant expressions of CaV1.2 exon 9* and Rbfox2. But glycated serum (GS), the mimic of advanced glycation end-products (AGEs), upregulates CaV1.2E9* channels proportion and downregulates Rbfox2 expression in NRVMs. By whole-cell patch clamp, we find GS application hyperpolarizes the current-voltage curve and window currents of cardiac CaV1.2 channels. Moreover, GS treatment raises K+-triggered intracellular Ca2+ concentration ([Ca2+]i), enlarges cell surface area of NRVMs and induces hypertrophic genes transcription. Consistently, siRNA-mediated knockdown of Rbfox2 in NRVMs upregulates CaV1.2E9* channel, shifts CaV1.2 window currents to hyperpolarization, increases [Ca2+]i and induces cardiomyocyte hypertrophy. CONCLUSIONS AGEs, not glucose, dysregulates Rbfox2 which thereby increases CaV1.2E9* channels and hyperpolarizes channel window currents. These make the channels open at greater negative potentials and lead to increased [Ca2+]i in cardiomyocytes, and finally induce cardiomyocyte hypertrophy in diabetes. Our work elucidates the underlying mechanisms for CaV1.2 channel regulation in diabetic heart, and targeting Rbfox2 to reset the aberrantly spliced CaV1.2 channel might be a promising therapeutic approach in diabetes-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Pengpeng Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Dongxia Qin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Tiange Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wei Hou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xinyu Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Shumin Yin
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Miaomiao Song
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - W C Hewith A Fernando
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaojie Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yu Sun
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Juejin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| |
Collapse
|
4
|
Gao Y, Su L, Wei Y, Tan S, Hu Z, Tao Z, Kovalik JP, Soong TW, Zhang J, Pu J, Ye L. Ascorbic acid induces MLC2v protein expression and promotes ventricular-like cardiomyocyte subtype in human induced pluripotent stem cells derived cardiomyocytes. Theranostics 2023; 13:3872-3896. [PMID: 37441603 PMCID: PMC10334833 DOI: 10.7150/thno.80801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/09/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction: The potentially unlimited number of cardiomyocyte (CMs) derived from human induced pluripotent stem cells (hiPSCs) in vitro facilitates high throughput applications like cell transplantation for myocardial repair, disease modelling, and cardiotoxicity testing during drug development. Despite promising progress in these areas, a major disadvantage that limits the use of hiPSC derived CMs (hiPSC-CMs) is their immaturity. Methods: Three hiPSC lines (PCBC-hiPSC, DP3-hiPSCs, and MLC2v-mEGFP hiPSC) were differentiated into CMs (PCBC-CMs, DP3-CMs, and MLC2v-CMs, respectively) with or without retinoic acid (RA). hiPSC-CMs were either maintained up to day 30 of contraction (D30C), or D60C, or purified using lactate acid and used for experiments. Purified hiPSC-CMs were cultured in basal maturation medium (BMM) or BMM supplemented with ascorbic acid (AA) for 14 days. The AA treated and non-treated hiPSC-CMs were characterized for sarcomeric proteins (MLC2v, TNNI3, and MYH7), ion channel proteins (Kir2.1, Nav1.5, Cav1.2, SERCA2a, and RyR), mitochondrial membrane potential, metabolomics, and action potential. Bobcat339, a selective and potent inhibitor of DNA demethylation, was used to determine whether AA promoted hiPSC-CM maturation through modulating DNA demethylation. Results: AA significantly increased MLC2v expression in PCBC-CMs, DP3-CMs, MLC2v-CMs, and RA induced atrial-like PCBC-CMs. AA treatment significantly increased mitochondrial mass, membrane potential, and amino acid and fatty acid metabolism in PCBC-CMs. Patch clamp studies showed that AA treatment induced PCBC-CMs and DP3-CMs adaptation to a ventricular-like phenotype. Bobcat339 inhibited MLC2v protein expression in AA treated PCBC-CMs and DP3-CMs. DNA demethylation inhibition was also associated with reduced TET1 and TET2 protein expressions and reduced accumulation of the oxidative product, 5 hmC, in both PCBC-CMs and DP3-CMs, in the presence of AA. Conclusions: Ascorbic acid induced MLC2v protein expression and promoted ventricular-like CM subtype in hiPSC-CMs. The effect of AA on hiPSC-CM was attenuated with inhibition of TET1/TET2 mediated DNA demethylation.
Collapse
Affiliation(s)
- Yu Gao
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Liping Su
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Yuhua Wei
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shihua Tan
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Zhenyu Hu
- Department of Physiology, National University of Singapore, Singapore
- Cardiovascular Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore
| | - Zhonghao Tao
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
- Department of Thoracic and Cardiovascular Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jean-Paul Kovalik
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS, Singapore
| | - Tuck Wah Soong
- Department of Physiology, National University of Singapore, Singapore
- Cardiovascular Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore
| | - Jianyi Zhang
- Department of Biomedical Engineering, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| |
Collapse
|
5
|
Marasco LE, Kornblihtt AR. The physiology of alternative splicing. Nat Rev Mol Cell Biol 2023; 24:242-254. [PMID: 36229538 DOI: 10.1038/s41580-022-00545-z] [Citation(s) in RCA: 201] [Impact Index Per Article: 100.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2022] [Indexed: 11/09/2022]
Abstract
Alternative splicing is a substantial contributor to the high complexity of transcriptomes of multicellular eukaryotes. In this Review, we discuss the accumulated evidence that most of this complexity is reflected at the protein level and fundamentally shapes the physiology and pathology of organisms. This notion is supported not only by genome-wide analyses but, mainly, by detailed studies showing that global and gene-specific modulations of alternative splicing regulate highly diverse processes such as tissue-specific and species-specific cell differentiation, thermal regulation, neuron self-avoidance, infrared sensing, the Warburg effect, maintenance of telomere length, cancer and autism spectrum disorders (ASD). We also discuss how mastering the control of alternative splicing paved the way to clinically approved therapies for hereditary diseases.
Collapse
Affiliation(s)
- Luciano E Marasco
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Moleculary Celular and CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Alberto R Kornblihtt
- Universidad de Buenos Aires (UBA), Facultad de Ciencias Exactas y Naturales, Departamento de Fisiología, Biología Moleculary Celular and CONICET-UBA, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE), Buenos Aires, Argentina.
| |
Collapse
|
6
|
Ferron L, Zamponi GW. The road to the brain in Timothy syndrome is paved with enhanced CaV1.2 activation gating. J Gen Physiol 2022; 154:213558. [PMID: 36264243 PMCID: PMC9587385 DOI: 10.1085/jgp.202213272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Specific gating effects of Timothy syndrome CaV1.2 channel mutations determine cardiovascular versus nervous system deficits.
Collapse
Affiliation(s)
- Laurent Ferron
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Canada
| |
Collapse
|
7
|
Progressive aortic stiffness in aging C57Bl/6 mice displays altered contractile behaviour and extracellular matrix changes. Commun Biol 2022; 5:605. [PMID: 35710942 PMCID: PMC9203497 DOI: 10.1038/s42003-022-03563-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/02/2022] [Indexed: 12/26/2022] Open
Abstract
Aortic stiffness is a hallmark of cardiovascular disease, but its pathophysiology remains incompletely understood. This study presents an in-dept characterization of aortic aging in male C57Bl/6 mice (2–24 months). Cardiovascular measurements include echocardiography, blood pressure measurement, and ex vivo organ chamber experiments. In vivo and ex vivo aortic stiffness increases with age, and precede the development of cardiac hypertrophy and peripheral blood pressure alterations. Contraction-independent stiffening (due to extracellular matrix changes) is pressure-dependent. Contraction-dependent aortic stiffening develops through heightened α1-adrenergic contractility, aberrant voltage-gated calcium channel function, and altered vascular smooth muscle cell calcium handling. Endothelial dysfunction is limited to a modest decrease in sensitivity to acetylcholine-induced relaxation with age. Our findings demonstrate that progressive arterial stiffening in C57Bl/6 mice precedes associated cardiovascular disease. Aortic aging is due to changes in extracellular matrix and vascular smooth muscle cell signalling, and not to altered endothelial function. A 24-month aging study in male C57Bl/6 mice reveals that aortic aging precedes cardiovascular disease and is due to changes in the extracellular matrix and vascular smooth muscle cell signaling.
Collapse
|
8
|
Diminished Rbfox1 increases vascular constriction by dynamically regulating alternative splicing of CaV1.2 calcium channel in hypertension. Clin Sci (Lond) 2022; 136:803-817. [PMID: 35543237 DOI: 10.1042/cs20220226] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Calcium influx from depolarized CaV1.2 calcium channels triggers the contraction of vascular smooth muscle cells (VSMCs), which is important for maintaining vascular myogenic tone and blood pressure. The function of CaV1.2 channel can be subtly modulated by alternative splicing (AS), and its aberrant splicing involves in the pathogenesis of multiple cardiovascular diseases. The RNA binding protein Rbfox1 is reported to regulate the AS events of CaV1.2 channel in the neuronal development, but its potential roles in vascular CaV1.2 channels and vasoconstriction remain undefined. Here, we detect Rbfox1 is expressed in rat vascular smooth muscles. Moreover, the protein level of Rbfox1 is dramatically decreased in the hypertensive small arteries from spontaneously hypertensive rats in comparison to normotensive ones from Wistar-Kyoto rats. In VSMCs, Rbfox1 could dynamically regulate the AS of CaV1.2 exons 9* and 33. By whole-cell patch clamp, we identify knockdown of Rbfox1 induces the hyperpolarization of CaV1.2 current-voltage relationship curve in VSMCs. Furthermore, siRNA-mediated knockdown of Rbfox1 increases the K+-induced constriction of rat mesenteric arteries. In summary, our results indicate Rbfox1 modulates vascular constriction by dynamically regulating CaV1.2 alternative exons 9* and 33. Therefore, our work elucidates the underlying mechanisms for CaV1.2 channels regulation and provides a potential therapeutic target for hypertension.
Collapse
|
9
|
Navakkode S, Zhai J, Wong YP, Li G, Soong TW. Enhanced long-term potentiation and impaired learning in mice lacking alternative exon 33 of Ca V1.2 calcium channel. Transl Psychiatry 2022; 12:1. [PMID: 35013113 PMCID: PMC8748671 DOI: 10.1038/s41398-021-01683-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/25/2021] [Accepted: 09/13/2021] [Indexed: 12/14/2022] Open
Abstract
The CACNA1C (calcium voltage-gated channel subunit alpha 1 C) gene that encodes the CaV1.2 channel is a prominent risk gene for neuropsychiatric and neurodegenerative disorders with cognitive and social impairments like schizophrenia, bipolar disorders, depression and autistic spectrum disorders (ASD). We have shown previously that mice with exon 33 deleted from CaV1.2 channel (CaV1.2-exon 33-/-) displayed increased CaV1.2 current density and single channel open probability in cardiomyocytes, and were prone to develop arrhythmia. As Ca2+ entry through CaV1.2 channels activates gene transcription in response to synaptic activity, we were intrigued to explore the possible role of Cav1.2Δ33 channels in synaptic plasticity and behaviour. Homozygous deletion of alternative exon 33 resulted in enhanced long-term potentiation (LTP), and lack of long- term depression (LTD), which did not correlate with enhanced learning. Exon 33 deletion also led to a decrease in social dominance, sociability and social novelty. Our findings shed light on the effect of gain-of-function of CaV1.2Δ33 signalling on synaptic plasticity and behaviour and provides evidence for a link between CaV1.2 and distinct cognitive and social behaviours associated with phenotypic features of psychiatric disorders like schizophrenia, bipolar disorder and ASD.
Collapse
Affiliation(s)
- Sheeja Navakkode
- grid.4280.e0000 0001 2180 6431Department of Physiology, National University of Singapore, Singapore, Singapore ,grid.59025.3b0000 0001 2224 0361Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jing Zhai
- grid.4280.e0000 0001 2180 6431Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Yuk Peng Wong
- grid.4280.e0000 0001 2180 6431Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Guang Li
- grid.4280.e0000 0001 2180 6431Department of Physiology, National University of Singapore, Singapore, Singapore ,grid.410578.f0000 0001 1114 4286Present Address: Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan China
| | - Tuck Wah Soong
- Department of Physiology, National University of Singapore, Singapore, Singapore. .,Healthy Longevity Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
10
|
Montañés-Agudo P, Casini S, Aufiero S, Ernault AC, van der Made I, Pinto YM, Remme CA, Creemers EE. Inhibition of minor intron splicing reduces Na+ and Ca2+ channel expression and function in cardiomyocytes. J Cell Sci 2021; 135:273616. [PMID: 34859816 PMCID: PMC8767276 DOI: 10.1242/jcs.259191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/26/2021] [Indexed: 12/04/2022] Open
Abstract
Eukaryotic genomes contain a tiny subset of ‘minor class’ introns with unique sequence elements that require their own splicing machinery. These minor introns are present in certain gene families with specific functions, such as voltage-gated Na+ and voltage-gated Ca2+ channels. Removal of minor introns by the minor spliceosome has been proposed as a post-transcriptional regulatory layer, which remains unexplored in the heart. Here, we investigate whether the minor spliceosome regulates electrophysiological properties of cardiomyocytes by knocking down the essential minor spliceosome small nuclear snRNA component U6atac in neonatal rat ventricular myocytes. Loss of U6atac led to robust minor intron retention within Scn5a and Cacna1c, resulting in reduced protein levels of Nav1.5 and Cav1.2 channels. Functional consequences were studied through patch-clamp analysis, and revealed reduced Na+ and L-type Ca2+ currents after loss of U6atac. In conclusion, minor intron splicing modulates voltage-dependent ion channel expression and function in cardiomyocytes. This may be of particular relevance in situations in which minor splicing activity changes, such as in genetic diseases affecting minor spliceosome components, or in acquired diseases in which minor spliceosome components are dysregulated, such as heart failure. Summary: Knockdown of minor spliceosome component U6atac in cardiomyocytes reveals that expression of the Na+ channel Scn5a and the L-type Ca2+ channel Cacna1c critically depend on minor intron splicing.
Collapse
Affiliation(s)
- Pablo Montañés-Agudo
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Simona Casini
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Simona Aufiero
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands.,Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Auriane C Ernault
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Ingeborg van der Made
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Yigal M Pinto
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Carol Ann Remme
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - Esther E Creemers
- Departments of Experimental Cardiology, Biostatistics and Bioinformatics, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| |
Collapse
|
11
|
Hofer NT, Pinggera A, Nikonishyna YV, Tuluc P, Fritz EM, Obermair GJ, Striessnig J. Stabilization of negative activation voltages of Cav1.3 L-Type Ca 2+-channels by alternative splicing. Channels (Austin) 2021; 15:38-52. [PMID: 33380256 PMCID: PMC7781618 DOI: 10.1080/19336950.2020.1859260] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 11/30/2022] Open
Abstract
-->Low voltage-activated Cav1.3 L-type Ca2+-channels are key regulators of neuronal excitability controlling neuronal development and different types of learning and memory. Their physiological functions are enabled by their negative activation voltage-range, which allows Cav1.3 to be active at subthreshold voltages. Alternative splicing in the C-terminus of their pore-forming α1-subunits gives rise to C-terminal long (Cav1.3L) and short (Cav1.3S) splice variants allowing Cav1.3S to activate at even more negative voltages than Cav1.3L. We discovered that inclusion of exons 8b, 11, and 32 in Cav1.3S further shifts activation (-3 to -4 mV) and inactivation (-4 to -6 mV) to more negative voltages as revealed by functional characterization in tsA-201 cells. We found transcripts of these exons in mouse chromaffin cells, the cochlea, and the brain. Our data further suggest that Cav1.3-containing exons 11 and 32 constitute a significant part of native channels in the brain. We therefore investigated the effect of these splice variants on human disease variants. Splicing did not prevent the gating defects of the previously reported human pathogenic variant S652L, which further shifted the voltage-dependence of activation of exon 11-containing channels by more than -12 mV. In contrast, we found no evidence for gating changes of the CACNA1D missense variant R498L, located in exon 11, which has recently been identified in a patient with an epileptic syndrome. Our data demonstrate that alternative splicing outside the C-terminus involving exons 11 and 32 contributes to channel fine-tuning by stabilizing negative activation and inactivation gating properties of wild-type and mutant Cav1.3 channels.
Collapse
Affiliation(s)
- Nadja T. Hofer
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Yuliia V. Nikonishyna
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Petronel Tuluc
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Eva M. Fritz
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| | - Gerald J. Obermair
- Institute of Physiology, Medical University Innsbruck, Innsbruck, Austria
- Division Physiology, Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Jörg Striessnig
- Department of Pharmacology and Toxicology, Centre for Molecular Biosciences, University of Innsbruck, Austria
| |
Collapse
|
12
|
Alternative Splicing in Cardiovascular Disease-A Survey of Recent Findings. Genes (Basel) 2021; 12:genes12091457. [PMID: 34573439 PMCID: PMC8469243 DOI: 10.3390/genes12091457] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
Alternative splicing, a driver of posttranscriptional variance, differs from canonical splicing by arranging the introns and exons of an immature pre-mRNA transcript in a multitude of different ways. Although alternative splicing was discovered almost half a century ago, estimates of the proportion of genes that undergo alternative splicing have risen drastically over the last two decades. Deep sequencing methods and novel bioinformatic algorithms have led to new insights into the prevalence of spliced variants, tissue-specific splicing patterns and the significance of alternative splicing in development and disease. Thus far, the role of alternative splicing has been uncovered in areas ranging from heart development, the response to myocardial infarction to cardiac structural disease. Circular RNAs, a product of alternative back-splicing, were initially discovered in 1976, but landmark publications have only recently identified their regulatory role, tissue-specific expression, and transcriptomic abundance, spurring a renewed interest in the topic. The aim of this review is to provide a brief insight into some of the available findings on the role of alternative splicing in cardiovascular disease, with a focus on atherosclerosis, myocardial infarction, heart failure, dilated cardiomyopathy and circular RNAs in myocardial infarction.
Collapse
|
13
|
Arslanova A, Shafaattalab S, Lin E, Barszczewski T, Hove-Madsen L, Tibbits GF. Investigating inherited arrhythmias using hiPSC-derived cardiomyocytes. Methods 2021; 203:542-557. [PMID: 34197925 DOI: 10.1016/j.ymeth.2021.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 10/21/2022] Open
Abstract
Fundamental to the functional behavior of cardiac muscle is that the cardiomyocytes are integrated as a functional syncytium. Disrupted electrical activity in the cardiac tissue can lead to serious complications including cardiac arrhythmias. Therefore, it is important to study electrophysiological properties of the cardiac tissue. With advancements in stem cell research, protocols for the production of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) have been established, providing great potential in modelling cardiac arrhythmias and drug testing. The hiPSC-CM model can be used in conjunction with electrophysiology-based platforms to examine the electrical activity of the cardiac tissue. Techniques for determining the myocardial electrical activity include multielectrode arrays (MEAs), optical mapping (OM), and patch clamping. These techniques provide critical approaches to investigate cardiac electrical abnormalities that underlie arrhythmias.
Collapse
Affiliation(s)
- Alia Arslanova
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser, University, Burnaby, BC V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z4H4, Canada
| | - Sanam Shafaattalab
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser, University, Burnaby, BC V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z4H4, Canada
| | - Eric Lin
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser, University, Burnaby, BC V5A 1S6, Canada
| | - Tiffany Barszczewski
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser, University, Burnaby, BC V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z4H4, Canada
| | - Leif Hove-Madsen
- Cardiac Rhythm and Contraction Group, IIBB-CSIC, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain; CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona 08041, Spain
| | - Glen F Tibbits
- Molecular Cardiac Physiology Group, Department of Biomedical Physiology and Kinesiology, Simon Fraser, University, Burnaby, BC V5A 1S6, Canada; hiPSC-CM Research Team, British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z4H4, Canada; Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada.
| |
Collapse
|
14
|
Striessnig J. Voltage-Gated Ca 2+-Channel α1-Subunit de novo Missense Mutations: Gain or Loss of Function - Implications for Potential Therapies. Front Synaptic Neurosci 2021; 13:634760. [PMID: 33746731 PMCID: PMC7966529 DOI: 10.3389/fnsyn.2021.634760] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
This review summarizes our current knowledge of human disease-relevant genetic variants within the family of voltage gated Ca2+ channels. Ca2+ channelopathies cover a wide spectrum of diseases including epilepsies, autism spectrum disorders, intellectual disabilities, developmental delay, cerebellar ataxias and degeneration, severe cardiac arrhythmias, sudden cardiac death, eye disease and endocrine disorders such as congential hyperinsulinism and hyperaldosteronism. A special focus will be on the rapidly increasing number of de novo missense mutations identified in the pore-forming α1-subunits with next generation sequencing studies of well-defined patient cohorts. In contrast to likely gene disrupting mutations these can not only cause a channel loss-of-function but can also induce typical functional changes permitting enhanced channel activity and Ca2+ signaling. Such gain-of-function mutations could represent therapeutic targets for mutation-specific therapy of Ca2+-channelopathies with existing or novel Ca2+-channel inhibitors. Moreover, many pathogenic mutations affect positive charges in the voltage sensors with the potential to form gating-pore currents through voltage sensors. If confirmed in functional studies, specific blockers of gating-pore currents could also be of therapeutic interest.
Collapse
Affiliation(s)
- Jörg Striessnig
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
15
|
Lei J, Liu X, Song M, Zhou Y, Fan J, Shen X, Xu X, Kapoor I, Zhu G, Wang (王觉进) J. Aberrant Exon 8/8a Splicing by Downregulated PTBP (Polypyrimidine Tract-Binding Protein) 1 Increases Ca V1.2 Dihydropyridine Resistance to Attenuate Vasodilation. Arterioscler Thromb Vasc Biol 2020; 40:2440-2453. [PMID: 32787518 DOI: 10.1161/atvbaha.120.315010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE Calcium channel blockers, such as dihydropyridines, are commonly used to inhibit enhanced activity of vascular CaV1.2 channels in hypertension. However, patients who are insensitive to such treatments develop calcium channel blocker-resistant hypertension. The function of CaV1.2 channel is diversified by alternative splicing, and the splicing factor PTBP (polypyrimidine tract-binding protein) 1 influences the utilization of mutually exclusive exon 8/8a of the CaV1.2 channel during neuronal development. Nevertheless, whether and how PTBP1 makes a role in the calcium channel blocker sensitivity of vascular CaV1.2 channels, and calcium channel blocker-induced vasodilation remains unknown. Approach and Results: We detected high expression of PTBP1 and, inversely, low expression of exon 8a in CaV1.2 channels (CaV1.2E8a) in rat arteries. In contrast, the opposite expression patterns were observed in brain and heart tissues. In comparison to normotensive rats, the expressions of PTBP1 and CaV1.2E8a channels were dysregulated in mesenteric arteries of hypertensive rats. Notably, PTBP1 expression was significantly downregulated, and CaV1.2E8a channels were aberrantly increased in dihydropyridine-resistant arteries compared with dihydropyridine-sensitive arteries of rats and human. In rat vascular smooth muscle cells, PTBP1 knockdown resulted in shifting of CaV1.2 exon 8 to 8a. Using patch-clamp recordings, we demonstrated a concomitant reduction of sensitivity of CaV1.2 channels to nifedipine, due to the higher expression of CaV1.2E8a isoform. In vascular myography experiments, small interfering RNA-mediated knockdown of PTBP1 attenuated nifedipine-induced vasodilation of rat mesenteric arteries. CONCLUSIONS PTBP1 finely modulates the sensitivities of CaV1.2 channels to dihydropyridine by shifting the utilization of exon 8/8a and resulting in changes of responses in dihydropyridine-induced vasodilation.
Collapse
Affiliation(s)
- Jianzhen Lei
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Xiaoxin Liu
- Shanghai Chest Hospital, Shanghai Jiaotong University, China (X.L.)
| | - Miaomiao Song
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Yingying Zhou
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Jia Fan
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Xiaowei Shen
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (X.S., X.X.)
| | - Xiaohan Xu
- Department of Cardiovascular Surgery, the First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (X.S., X.X.)
| | - Isha Kapoor
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Guoqing Zhu
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| | - Juejin Wang (王觉进)
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (J.L., M.S., Y.Z., J.F., G.Z., J.W.)
| |
Collapse
|
16
|
Regulation of cardiovascular calcium channel activity by post-translational modifications or interacting proteins. Pflugers Arch 2020; 472:653-667. [PMID: 32435990 DOI: 10.1007/s00424-020-02398-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/04/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
Abstract
Voltage-gated calcium channels are the major pathway for Ca2+ influx to initiate the contraction of smooth and cardiac muscles. Alterations of calcium channel function have been implicated in multiple cardiovascular diseases, such as hypertension, atrial fibrillation, and long QT syndrome. Post-translational modifications do expand cardiovascular calcium channel structure and function to affect processes such as channel trafficking or polyubiquitination by two E3 ubiquitin ligases, Ret finger protein 2 (Rfp2) or murine double minute 2 protein (Mdm2). Additionally, biophysical property such as Ca2+-dependent inactivation (CDI) could be altered through binding of calmodulin, or channel activity could be modulated via S-nitrosylation by nitric oxide and phosphorylation by protein kinases or by interacting protein partners, such as galectin-1 and Rem. Understanding how cardiovascular calcium channel function is post-translationally remodeled under distinctive disease conditions will provide better information about calcium channel-related disease mechanisms and improve the development of more selective therapeutic agents for cardiovascular diseases.
Collapse
|
17
|
Misra C, Bangru S, Lin F, Lam K, Koenig SN, Lubbers ER, Hedhli J, Murphy NP, Parker DJ, Dobrucki LW, Cooper TA, Tajkhorshid E, Mohler PJ, Kalsotra A. Aberrant Expression of a Non-muscle RBFOX2 Isoform Triggers Cardiac Conduction Defects in Myotonic Dystrophy. Dev Cell 2020; 52:748-763.e6. [PMID: 32109384 PMCID: PMC7098852 DOI: 10.1016/j.devcel.2020.01.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/20/2022]
Abstract
Myotonic dystrophy type 1 (DM1) is a multisystemic genetic disorder caused by the CTG repeat expansion in the 3'-untranslated region of DMPK gene. Heart dysfunctions occur in ∼80% of DM1 patients and are the second leading cause of DM1-related deaths. Herein, we report that upregulation of a non-muscle splice isoform of RNA-binding protein RBFOX2 in DM1 heart tissue-due to altered splicing factor and microRNA activities-induces cardiac conduction defects in DM1 individuals. Mice engineered to express the non-muscle RBFOX240 isoform in heart via tetracycline-inducible transgenesis, or CRISPR/Cas9-mediated genome editing, reproduced DM1-related cardiac conduction delay and spontaneous episodes of arrhythmia. Further, by integrating RNA binding with cardiac transcriptome datasets from DM1 patients and mice expressing the non-muscle RBFOX2 isoform, we identified RBFOX240-driven splicing defects in voltage-gated sodium and potassium channels, which alter their electrophysiological properties. Thus, our results uncover a trans-dominant role for an aberrantly expressed RBFOX240 isoform in DM1 cardiac pathogenesis.
Collapse
Affiliation(s)
- Chaitali Misra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Sushant Bangru
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Feikai Lin
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Kin Lam
- Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Sara N Koenig
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Ellen R Lubbers
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jamila Hedhli
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Nathaniel P Murphy
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Darren J Parker
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Lawrence W Dobrucki
- Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Thomas A Cooper
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Emad Tajkhorshid
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Department of Physics, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Department of Bioengineering, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Centers for Macromolecular Modeling, Bioinformatics and Experimental Molecular Imaging at Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA
| | - Peter J Mohler
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Auinash Kalsotra
- Department of Biochemistry, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Urbana, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
18
|
Gandini MA, Souza IA, Raval D, Xu J, Pan YX, Zamponi GW. Differential regulation of Cav2.2 channel exon 37 variants by alternatively spliced μ-opioid receptors. Mol Brain 2019; 12:98. [PMID: 31775826 PMCID: PMC6880636 DOI: 10.1186/s13041-019-0524-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022] Open
Abstract
We have examined the regulation of mutually exclusive Cav2.2 exon 37a and b variants by the mouse μ-opioid receptor (mMOR) C-terminal splice variants 1, 1C and 1O in tsA-201 cells. Electrophysiological analyses revealed that both channel isoforms exhibit DAMGO-induced voltage-dependent (Gβγ-mediated) inhibition and its recovery by voltage pre-pulses, as well as a voltage-independent component. However, the two channel isoforms differ in their relative extent of voltage-dependent and independent inhibition, with Cav2.2-37b showing significantly more voltage-dependent inhibition upon activation of the three mMOR receptors studied. In addition, coexpression of either mMOR1 or mMOR1C results in an agonist-independent reduction in the peak current density of Cav2.2-37a channels, whereas the peak current density of Cav2.2-37b does not appear to be affected. Interestingly, this decrease is not due to an effect on channel expression at the plasma membrane, as demonstrated by biotinylation experiments. We further examined the mechanism underlying the agonist-independent modulation of Cav2.2-37a by mMOR1C. Incubation of cells with pertussis toxin did not affect the mMOR1C mediated inhibition of Cav2.2-37a currents, indicating a lack of involvement of Gi/o signaling. However, when a Src tyrosine kinase inhibitor was applied, the effect of mMOR1C was lost. Moreover, when we recorded currents using a Cav2.2-37a mutant in which tyrosine 1747 was replaced with phenylalanine (Y1747F), the agonist independent effects of mMOR1C were abolished. Altogether our findings show that Cav2.2-37a and Cav2.2-37b isoforms are subject to differential regulation by C-terminal splice variants of mMORs, and that constitutive mMOR1C activity and downstream tyrosine kinase activity exert a selective inhibition of the Cav2.2-37a splice variant, an N-type channel isoform that is highly enriched in nociceptors. Our study provides new insights into the roles of the MOR full-length C-terminal variants in modulating Cav2.2 channel isoform activities.
Collapse
Affiliation(s)
- Maria A Gandini
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Ivana A Souza
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Dvij Raval
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jin Xu
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Ying-Xian Pan
- Department of Neurology and the Molecular Pharmacology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Gerald W Zamponi
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
19
|
Wang J, Li G, Yu D, Wong YP, Yong TF, Liang MC, Liao P, Foo R, Hoppe UC, Soong TW. Characterization of Ca V1.2 exon 33 heterozygous knockout mice and negative correlation between Rbfox1 and Ca V1.2 exon 33 expressions in human heart failure. Channels (Austin) 2019; 12:51-57. [PMID: 28949795 PMCID: PMC5774182 DOI: 10.1080/19336950.2017.1381805] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recently, we reported that homozygous deletion of alternative exon 33 of CaV1.2 calcium channel in the mouse resulted in ventricular arrhythmias arising from increased CaV1.2Δ33ICaL current density in the cardiomyocytes. We wondered whether heterozygous deletion of exon 33 might produce cardiac phenotype in a dose-dependent manner, and whether the expression levels of RNA splicing factors known to regulate alternative splicing of exon 33 might change in human heart failure. Unexpectedly, we found that exon 33+/− cardiomyocytes showed similar CaV1.2 channel properties as wild-type cardiomyocyte, even though CaV1.2Δ33 channels exhibit a gain-in-function. In human hearts, we found that the mRNA level of splicing factor Rbfox1, but not Rbfox2, was downregulated in dilated cardiomyopathy, and CACNA1C mRNA level was dramatically decreased in the both of dilated and ischemic cardiomyopathy. These data imply Rbfox1 may be involved in the development of cardiomyopathies via regulating the alternative splicing of CaV1.2 exon 33. (149 words)
Collapse
Affiliation(s)
- Juejin Wang
- a Department of Physiology , National University of Singapore , Singapore.,b Department of Physiology , Nanjing Medical University , Nanjing , P.R. China
| | - Guang Li
- a Department of Physiology , National University of Singapore , Singapore.,c Key Laboratory of Medical Electrophysiology , Ministry of Education, Institute of Cardiovascular Research, Southwest Medical University , Luzhou , P.R. China
| | - Dejie Yu
- a Department of Physiology , National University of Singapore , Singapore
| | - Yuk Peng Wong
- a Department of Physiology , National University of Singapore , Singapore
| | - Tan Fong Yong
- a Department of Physiology , National University of Singapore , Singapore
| | - Mui Cheng Liang
- a Department of Physiology , National University of Singapore , Singapore
| | - Ping Liao
- d National Neuroscience Institute , Singapore
| | - Roger Foo
- e Genome Institute of Singapore , Singapore
| | - Uta C Hoppe
- f Department of Internal Medicine II , Paracelsus Medical University , Salzburg , Austria
| | - Tuck Wah Soong
- a Department of Physiology , National University of Singapore , Singapore.,d National Neuroscience Institute , Singapore.,g Graduate School for Integrative Sciences and Engineering , National University of Singapore , Singapore.,h Neurobiology/Ageing Programme, National University of Singapore , Singapore
| |
Collapse
|
20
|
Tan Y, Fei D, He X, Dai J, Xu R, Xu X, Wu J, Li B. L-type voltage-gated calcium channels in stem cells and tissue engineering. Cell Prolif 2019; 52:e12623. [PMID: 31115100 PMCID: PMC6669004 DOI: 10.1111/cpr.12623] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/26/2019] [Accepted: 03/29/2019] [Indexed: 12/22/2022] Open
Abstract
L-type voltage-gated calcium ion channels (L-VGCCs) have been demonstrated to be the mediator of several significant intracellular activities in excitable cells, such as neurons, chromaffin cells and myocytes. Recently, an increasing number of studies have investigated the function of L-VGCCs in non-excitable cells, particularly stem cells. However, there appear to be no systematic reviews of the relationship between L-VGCCs and stem cells, and filling this gap is prescient considering the contribution of L-VGCCs to the proliferation and differentiation of several types of stem cells. This review will discuss the possible involvement of L-VGCCs in stem cells, mainly focusing on osteogenesis mediated by mesenchymal stem cells (MSCs) from different tissues and neurogenesis mediated by neural stem/progenitor cells (NSCs). Additionally, advanced applications that use these channels as the target for tissue engineering, which may offer the hope of tissue regeneration in the future, will also be explored.
Collapse
Affiliation(s)
- Yi‐zhou Tan
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Dong‐dong Fei
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Xiao‐ning He
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| | - Ji‐min Dai
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Rong‐chen Xu
- Doctoral students of eight-year programThe Fourth Military Medical UniversityXi’anChina
| | - Xin‐yue Xu
- Department of Periodontology, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Jun‐jie Wu
- Department of Orthodontics, State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, School of StomatologyThe Fourth Military Medical UniversityXi’anChina
| | - Bei Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Center for Tissue EngineeringFourth Military Medical UniversityXi’anChina
| |
Collapse
|
21
|
Bartels P, Yu D, Huang H, Hu Z, Herzig S, Soong TW. Alternative Splicing at N Terminus and Domain I Modulates Ca V1.2 Inactivation and Surface Expression. Biophys J 2019; 114:2095-2106. [PMID: 29742403 DOI: 10.1016/j.bpj.2018.03.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 03/12/2018] [Accepted: 03/27/2018] [Indexed: 10/17/2022] Open
Abstract
The CaV1.2 L-type calcium channel is a key conduit for Ca2+ influx to initiate excitation-contraction coupling for contraction of the heart and vasoconstriction of the arteries and for altering membrane excitability in neurons. Its α1C pore-forming subunit is known to undergo extensive alternative splicing to produce many CaV1.2 isoforms that differ in their electrophysiological and pharmacological properties. Here, we examined the structure-function relationship of human CaV1.2 with respect to the inclusion or exclusion of mutually exclusive exons of the N-terminus exons 1/1a and IS6 segment exons 8/8a. These exons showed tissue selectivity in their expression patterns: heart variant 1a/8a, one smooth-muscle variant 1/8, and a brain isoform 1/8a. Overall, the 1/8a, when coexpressed with CaVβ2a, displayed a significant and distinct shift in voltage-dependent activation and inactivation and inactivation kinetics as compared to the other three splice variants. Further analysis showed a clear additive effect of the hyperpolarization shift in V1/2inact of CaV1.2 channels containing exon 1 in combination with 8a. However, this additive effect was less distinct for V1/2act. However, the measured effects were β-subunit-dependent when comparing CaVβ2a with CaVβ3 coexpression. Notably, calcium-dependent inactivation mediated by local Ca2+-sensing via the N-lobe of calmodulin was significantly enhanced in exon-1-containing CaV1.2 as compared to exon-1a-containing CaV1.2 channels. At the cellular level, the current densities of the 1/8a or 1/8 variants were significantly larger than the 1a/8a and 1a/8 variants when coexpressed either with CaVβ2a or CaVβ3 subunit. This finding correlated well with a higher channel surface expression for the exon 1-CaV1.2 isoform that we quantified by protein surface-expression levels or by gating currents. Our data also provided a deeper molecular understanding of the altered biophysical properties of alternatively spliced human CaV1.2 channels by directly comparing unitary single-channel events with macroscopic whole-cell currents.
Collapse
Affiliation(s)
- Peter Bartels
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Dejie Yu
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Hua Huang
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Zhenyu Hu
- Department of Physiology, National University of Singapore, Singapore, Singapore
| | - Stefan Herzig
- Department of Pharmacology, University of Cologne, Cologne, Germany
| | - Tuck Wah Soong
- Department of Physiology, National University of Singapore, Singapore, Singapore; NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore; Neurobiology/Ageing Programme, National University of Singapore, Singapore, Singapore; National Neuroscience Institute, Singapore, Singapore.
| |
Collapse
|
22
|
LRP5 controls cardiac QT interval by modulating the metabolic homeostasis of L-type calcium channel. Int J Cardiol 2019; 275:120-128. [PMID: 30309679 DOI: 10.1016/j.ijcard.2018.06.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 06/04/2018] [Accepted: 06/06/2018] [Indexed: 01/04/2023]
Abstract
BACKGROUND Low-density lipoprotein receptor-related protein 5 (LRP5) has been intensively studied as a co-receptor for β-catenin-dependent Wnt signaling. Emerging evidences have demonstrated β-catenin-independent functions of LRP5. However, the biological role of LRP5 in the mammalian heart is largely unknown. METHODS AND RESULTS Conditional cardiac-specific Lrp5 knockout (Lrp5-CKO) mice were generated by crossing Lrp5flox/flox mice with αMHC/MerCreMer mice. Lrp5-CKO mice consistently displayed normal cardiac structure and function. Telemetric electrocardiogram recordings revealed a short QT interval in Lrp5-CKO mice, which was tightly linked to the striking abbreviation of action potential duration (APD) in ventricular myocytes. The analysis of whole-cell currents indicated that a reduction in activity and protein expression of L-type calcium channel (LTCC), rather than other ion channels, contributed to the abnormality in APD. Furthermore, we showed that Lrp5 ablation induced a significant convergence of CaV1.2α1c proteins to the endoplasmic reticulum. Consequently, increased proteasomal degradation of these proteins was observed, which was independent of the Wnt/β-catenin signaling pathway. CONCLUSIONS LRP5 directly modulates the degradation of LTCC to control cardiac QT interval. These findings provide compelling evidence for the potential role of LRPs in cardiac electrophysiology.
Collapse
|
23
|
|
24
|
Zhang Q, Chen J, Qin Y, Wang J, Zhou L. Mutations in voltage-gated L-type calcium channel: implications in cardiac arrhythmia. Channels (Austin) 2018; 12:201-218. [PMID: 30027834 PMCID: PMC6104696 DOI: 10.1080/19336950.2018.1499368] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/08/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023] Open
Abstract
The voltage-gated L-type calcium channel (LTCC) is essential for multiple cellular processes. In the heart, calcium influx through LTCC plays an important role in cardiac electrical excitation. Mutations in LTCC genes, including CACNA1C, CACNA1D, CACNB2 and CACNA2D, will induce the dysfunctions of calcium channels, which result in the abnormal excitations of cardiomyocytes, and finally lead to cardiac arrhythmias. Nevertheless, the newly found mutations in LTCC and their functions are continuously being elucidated. This review summarizes recent findings on the mutations of LTCC, which are associated with long QT syndromes, Timothy syndromes, Brugada syndromes, short QT syndromes, and some other cardiac arrhythmias. Indeed, we describe the gain/loss-of-functions of these mutations in LTCC, which can give an explanation for the phenotypes of cardiac arrhythmias. Moreover, we present several challenges in the field at present, and propose some diagnostic or therapeutic approaches to these mutation-associated cardiac diseases in the future.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, the Second Affiliated Hospital of Nantong University, Nantong First Hospital, Nantong, Jiangsu, China
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Chen
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Qin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Alternative Splicing of L-type Ca V1.2 Calcium Channels: Implications in Cardiovascular Diseases. Genes (Basel) 2017; 8:genes8120344. [PMID: 29186814 PMCID: PMC5748662 DOI: 10.3390/genes8120344] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/09/2017] [Accepted: 11/21/2017] [Indexed: 01/28/2023] Open
Abstract
L-type CaV1.2 calcium channels are the major pathway for Ca2+ influx to initiate the contraction of smooth and cardiac muscles. Alteration of CaV1.2 channel function has been implicated in multiple cardiovascular diseases, such as hypertension and cardiac hypertrophy. Alternative splicing is a post-transcriptional mechanism that expands CaV1.2 channel structures to modify function, pharmacological and biophysical property such as calcium/voltage-dependent inactivation (C/VDI), or to influence its post-translational modulation by interacting proteins such as Galectin-1. Alternative splicing has generated functionally diverse CaV1.2 isoforms that can be developmentally regulated in the heart, or under pathophysiological conditions such as in heart failure. More importantly, alternative splicing of certain exons of CaV1.2 has been reported to be regulated by splicing factors such as RNA-binding Fox-1 homolog 1/2 (Rbfox 1/2), polypyrimidine tract-binding protein (PTBP1) and RNA-binding motif protein 20 (RBM20). Understanding how CaV1.2 channel function is remodelled in disease will provide better information to guide the development of more targeted approaches to discover therapeutic agents for cardiovascular diseases.
Collapse
|
26
|
Zhou Y, Fan J, Zhu H, Ji L, Fan W, Kapoor I, Wang Y, Wang Y, Zhu G, Wang J. Aberrant Splicing Induced by Dysregulated Rbfox2 Produces Enhanced Function of Ca V1.2 Calcium Channel and Vascular Myogenic Tone in Hypertension. Hypertension 2017; 70:1183-1192. [PMID: 28993448 DOI: 10.1161/hypertensionaha.117.09301] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 08/26/2017] [Accepted: 09/11/2017] [Indexed: 01/12/2023]
Abstract
Calcium influx from activated voltage-gated calcium channel CaV1.2 in vascular smooth muscle cells is indispensable for maintaining myogenic tone and blood pressure. The function of CaV1.2 channel can be optimized by alternative splicing, one of post-transcriptional modification mechanisms. The splicing factor Rbfox2 is known to regulate the CaV1.2 pre-mRNA alternative splicing events during neuronal development. However, Rbfox2's roles in modulating the key function of vascular CaV1.2 channel and in the pathogenesis of hypertension remain elusive. Here, we report that the proportion of CaV1.2 channels with alternative exon 9* is increased by 10.3%, whereas that with alternative exon 33 is decreased by 10.5% in hypertensive arteries. Surprisingly, the expression level of Rbfox2 is increased ≈3-folds, presumably because of the upregulation of a dominant-negative isoform of Rbfox2. In vascular smooth muscle cells, we find that knockdown of Rbfox2 dynamically increases alternative exon 9*, whereas decreases exon 33 inclusion of CaV1.2 channels. By patch-clamp studies, we show that diminished Rbfox2-induced alternative splicing shifts the steady-state activation and inactivation curves of vascular CaV1.2 calcium channel to hyperpolarization, which makes the window current potential to more negative. Moreover, siRNA-mediated knockdown of Rbfox2 increases the pressure-induced vascular myogenic tone of rat mesenteric artery. Taken together, our data indicate that Rbfox2 modulates the functions of vascular CaV1.2 calcium channel by dynamically regulating the expressions of alternative exons 9* and 33, which in turn affects the vascular myogenic tone. Therefore, our work suggests a key role for Rbfox2 in hypertension, which provides a rational basis for designing antihypertensive therapies.
Collapse
Affiliation(s)
- Yingying Zhou
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Jia Fan
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Huayuan Zhu
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Li Ji
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Wenyong Fan
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Isha Kapoor
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Yue Wang
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Yuan Wang
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Guoqing Zhu
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.)
| | - Juejin Wang
- From the Key Laboratory of Cardiovascular Disease and Molecular Intervention, Department of Physiology, Nanjing Medical University, Jiangsu, China (Y.Z., J.F., L.J., W.F., Yue Wang, Yuan Wang, G.Z., J.W.); Department of Hematology, First Affiliated Hospital of Nanjing Medical University, Jiangsu, China (H.Z.); and Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, OH (I.K.).
| |
Collapse
|