1
|
Qi B, Xie Z, Shen D, Song Y, Liu S, Wang Q, Zhou J, Ge J. Blocking Na V1.8 regulates atrial fibrillation inducibility and cardiac conduction after myocardial infarction. BMC Cardiovasc Disord 2024; 24:605. [PMID: 39472780 PMCID: PMC11520513 DOI: 10.1186/s12872-024-04261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/14/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND The role of NaV1.8 impacts in atrial fibrillation susceptibility after myocardial infarction remains only partially understood. We studied the effect of blocking NaV1.8 in the cardiac ganglionated plexi (GP) on the atrial fibrillation inducibility and cardiac conduction in the myocardial infarction model. METHODS Eighteen male beagles were randomly enrolled. Left anterior descending coronary artery was ligated to created myocardial infarction model. Four weeks after surgery, NaV1.8 blocker A-803,467 (n = 9) or DMSO (n = 9, control) was injected into the four cardiac major GPs. Sinus rate, ventricular rate during atrial fibrillation, PR interval, atrial effective refractory period, atrial fibrillation duration and the cumulative window of atrial vulnerability were measured before and 60 min after A-803,467 injection. RESULTS Administration of A-803,467 significantly increased sinus rate, shortened PR interval and increased ventricular rate during atrial fibrillation compared to control. A-803,467 also significantly shortened atrial effective refractory period, prolonged atrial fibrillation duration and increased the cumulative window of atrial vulnerability. A-803,467 suppressed the slowing of heart rate response to high-frequency electrical stimulation of the anterior right GP, which was used as the surrogate marker for GP function. Double staining of ChAT and NaV1.8 demonstrated colocalization of ChAT and NaV1.8 in canine GPs. CONCLUSIONS Blocking NaV1.8 in the cardiac GP may modulate atrial fibrillation inducibility and cardiac conduction after myocardial infarction, and the underlying mechanism may be associated with the regulation of the neural activity of the cardiac GP.
Collapse
Affiliation(s)
- Baozhen Qi
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
- Department of Cardiology, Zhongshan Hospital (Xiamen), Fudan University, 668 Jinhu Road, Xiamen, 361015, China
| | - Zhonglei Xie
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Dongli Shen
- Division of Cardiology, Department of Medicine, the Affiliated People's Hospital of Jiangsu University, Zhenjiang, 212002, Jiangsu, China
| | - Yu Song
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| | - Shaowen Liu
- Department of Cardiology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Qibing Wang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China.
| | - Jingmin Zhou
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, National Clinical Research Center for Interventional Medicine, Fudan University, Shanghai, 200032, China
| |
Collapse
|
2
|
Balachandran S, Prada-Medina CA, Mensah MA, Kakar N, Nagel I, Pozojevic J, Audain E, Hitz MP, Kircher M, Sreenivasan VKA, Spielmann M. STIGMA: Single-cell tissue-specific gene prioritization using machine learning. Am J Hum Genet 2024; 111:338-349. [PMID: 38228144 PMCID: PMC10870135 DOI: 10.1016/j.ajhg.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/18/2024] Open
Abstract
Clinical exome and genome sequencing have revolutionized the understanding of human disease genetics. Yet many genes remain functionally uncharacterized, complicating the establishment of causal disease links for genetic variants. While several scoring methods have been devised to prioritize these candidate genes, these methods fall short of capturing the expression heterogeneity across cell subpopulations within tissues. Here, we introduce single-cell tissue-specific gene prioritization using machine learning (STIGMA), an approach that leverages single-cell RNA-seq (scRNA-seq) data to prioritize candidate genes associated with rare congenital diseases. STIGMA prioritizes genes by learning the temporal dynamics of gene expression across cell types during healthy organogenesis. To assess the efficacy of our framework, we applied STIGMA to mouse limb and human fetal heart scRNA-seq datasets. In a cohort of individuals with congenital limb malformation, STIGMA prioritized 469 variants in 345 genes, with UBA2 as a notable example. For congenital heart defects, we detected 34 genes harboring nonsynonymous de novo variants (nsDNVs) in two or more individuals from a set of 7,958 individuals, including the ortholog of Prdm1, which is associated with hypoplastic left ventricle and hypoplastic aortic arch. Overall, our findings demonstrate that STIGMA effectively prioritizes tissue-specific candidate genes by utilizing single-cell transcriptome data. The ability to capture the heterogeneity of gene expression across cell populations makes STIGMA a powerful tool for the discovery of disease-associated genes and facilitates the identification of causal variants underlying human genetic disorders.
Collapse
Affiliation(s)
- Saranya Balachandran
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany
| | - Cesar A Prada-Medina
- Human Molecular Genetics Group, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Martin A Mensah
- Institut für Medizinische Genetik und Humangenetik, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; BIH Charité Digital Clinician Scientist Program, BIH Biomedical Innovation Academy, Anna-Louisa-Karsch-Strasse 2, 10178 Berlin, Germany; RG Development & Disease, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Naseebullah Kakar
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany; Department of Biotechnology, BUITEMS, Quetta, Pakistan
| | - Inga Nagel
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany
| | - Jelena Pozojevic
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany
| | - Enrique Audain
- Institute of Medical Genetics, Carl von Ossietzky University, 26129 Oldenburg, Germany; DZHK e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck; Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Marc-Phillip Hitz
- Institute of Medical Genetics, Carl von Ossietzky University, 26129 Oldenburg, Germany; DZHK e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck; Department of Congenital Heart Disease and Pediatric Cardiology, University Hospital of Schleswig-Holstein, 24105 Kiel, Germany
| | - Martin Kircher
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany
| | - Varun K A Sreenivasan
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany.
| | - Malte Spielmann
- Institute of Human Genetics, University Hospital Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Germany; Human Molecular Genetics Group, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; DZHK e.V. (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck.
| |
Collapse
|
3
|
Cui CP, Xiong X, Zhao JX, Fu DH, Zhang Y, Ma PB, Wu D, Li BY. Piezo1 channel activation facilitates baroreflex afferent neurotransmission with subsequent blood pressure reduction in control and hypertension rats. Acta Pharmacol Sin 2024; 45:76-86. [PMID: 37670136 PMCID: PMC10770313 DOI: 10.1038/s41401-023-01154-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/12/2023] [Indexed: 09/07/2023] Open
Abstract
Mechanosensitive cation channels such as Piezo1 and Piezo2 are activated by mechanical force like a starched wall of the aorta while blood pressure (BP) rising, which helps to elucidate the underlying mechanism of mechanotransduction of baroreceptor endings. In this study we investigated how Piezo1 channel activation-mediated gender- and afferent-specific BP regulation in rats. We established high-fat diet and fructose drink-induced hypertension model rats (HFD-HTN) and deoxycorticosterone (DOCA)-sensitive hypertension model rats. We showed that the expression levels of Piezo1 and Piezo2 were significantly up-regulated in left ventricle of HFD and DOCA hypertensive rats, whereas the down-regulation of Piezo1 was likely to be compensated by Piezo2 up-regulation in the aorta. Likewise, down-regulated Piezo1 was observed in the nodose ganglion (NG), while up-regulated Piezo2 was found in the nucleus tractus solitarius (NTS), which might synergistically reduce the excitatory neurotransmitter release from the presynaptic membrane. Notably, microinjection of Yoda1 (0.025-2.5 mg/ml) into the NG concentration-dependently reduced BP in both hypertensive rat models as well as in control rats with similar EC50; the effect of Yoda1 was abolished by microinjection of a Piezo1 antagonist GsMTx4 (1.0 μM). Functional analysis in an in vitro aortic arch preparation showed that instantaneous firing frequency of single Ah-fiber of aortic depressor nerve was dramatically increased by Yoda1 (0.03-1.0 μM) and blocked by GsMTx4 (1.0 μM). Moreover, spontaneous synaptic currents recorded from identified 2nd-order Ah-type baroreceptive neurons in the NTS was also facilitated over 100% by Yoda1 (1.0 μM) and completely blocked by GsMTx4 (3.0 μM). These results demonstrate that Piezo1 expressed on Ah-type baroreceptor and baroreceptive neurons in the NG and NTS plays a key role in a sexual-dimorphic BP regulation under physiological and hypertensive condition through facilitation of baroreflex afferent neurotransmission, which is presumably collaborated by Piezo2 expression at different level of baroreflex afferent pathway via compensatory and synergistic mechanisms.
Collapse
Affiliation(s)
- Chang-Peng Cui
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Xiong
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jia-Xin Zhao
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Dong-Hong Fu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yan Zhang
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Peng-Bo Ma
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Di Wu
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
- Department of Pharmacy, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Bai-Yan Li
- Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
4
|
Dormer A, Narayanan M, Schentag J, Achinko D, Norman E, Kerrigan J, Jay G, Heydorn W. A Review of the Therapeutic Targeting of SCN9A and Nav1.7 for Pain Relief in Current Human Clinical Trials. J Pain Res 2023; 16:1487-1498. [PMID: 37168847 PMCID: PMC10166096 DOI: 10.2147/jpr.s388896] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/14/2023] [Indexed: 05/13/2023] Open
Abstract
Introduction There is a great need to find alternative treatments for chronic pain which have become a healthcare problem. We discuss current therapeutic targeting Nav1.7. Areas Covered Nav1.7 is a sodium ion channel protein that is associated with several human pain genetic syndromes. It has been found that mutations associated with Nav1.7 lead to the loss of the ability to perceive pain in individuals that are otherwise normal. Several therapeutic interventions are presently undergoing preclinical and research using the methodology of damping Nav1.7 expressions as a methodology to decrease the sensation of pain leading to analgesia. Expert Opinion It is our strong belief that there is a viable future in the targeting of protein of Nav1.7 for the relief of chronic pain in humans. The review will look at the genomics associated with SCN1A and proteomic of Nav1.7 as a foundation to explain the mechanism of the therapeutic interventions targeting Nav1.7, the human disease that are associated with Nav1.7, and the current development of treatment for chronic pain whether in preclinical or clinical trials targeting Nav1.7 expressions. The development of therapeutic antagonists targeting Nav1.7 could be a viable alternative to the current treatments which have led to the opioid crisis. Therefore, Nav1.7 targeted treatment has a major clinical significance that will have positive consequences as it relates to chronic pain interventions.
Collapse
Affiliation(s)
- Anton Dormer
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
- Correspondence: Anton Dormer, Research and Development, PepVax, Inc, 8720 Georgia Ave #1000, Silver Spring, MD, 20910, USA, Email
| | | | - Jerome Schentag
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Daniel Achinko
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - Elton Norman
- Research and Development, Pepvax, Inc, Silver Spring, MD, USA
| | - James Kerrigan
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | - Gary Jay
- Research and Development, Navintus, Inc, Princeton, NJ, USA
| | | |
Collapse
|
5
|
Chidiac C, Xue Y, Muniz Moreno MDM, Bakr Rasheed AA, Lorentz R, Birling MC, Gaveriaux-Ruff C, Herault Y. The Human SCN10A G1662S Point Mutation Established in Mice Impacts on Mechanical, Heat, and Cool Sensitivity. Front Pharmacol 2021; 12:780132. [PMID: 34925037 PMCID: PMC8671994 DOI: 10.3389/fphar.2021.780132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 11/13/2022] Open
Abstract
The voltage-gated sodium channel NAV1.8 is expressed in primary nociceptive neurons and is involved in pain transmission. Mutations in the SCN10A gene (encoding NAV1.8 channel) have been identified in patients with idiopathic painful small fiber neuropathy (SFN) including the SCN10AG1662S gain-of-function mutation. However, the role of this mutation in pain sensation remains unknown. We have generated the first mouse model for the G1662S mutation by using homologous recombination in embryonic stem cells. The corresponding Scn10aG1663S mouse line has been analyzed for Scn10a expression, intraepidermal nerve fiber density (IENFD), and nociception using behavioral tests for thermal and mechanical sensitivity. The Scn10aG1663S mutants had a similar Scn10a expression level in dorsal root ganglia (DRG) to their wild-type littermates and showed normal IENFD in hindpaw skin. Mutant mice were more sensitive to touch than wild types in the von Frey test. In addition, sexual dimorphism was observed for several pain tests, pointing to the relevance of performing the phenotypical assessment in both sexes. Female homozygous mutants tended to be more sensitive to cooling stimuli in the acetone test. For heat sensitivity, male homozygous mutants showed shorter latencies to radiant heat in the Hargreaves test while homozygous females had longer latencies in the tail flick test. In addition, mutant males displayed a shorter reaction latency on the 54°C hot plate. Collectively, Scn10aG1663S mutant mice show a moderate but consistent increased sensitivity in behavioral tests of nociception. This altered nociception found in Scn10aG1663S mice demonstrates that the corresponding G1662 mutation of SCN10A found in SFN patients with pain contributes to their pain symptoms.
Collapse
Affiliation(s)
- Celeste Chidiac
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Yaping Xue
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Maria Del Mar Muniz Moreno
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Ameer Abu Bakr Rasheed
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Romain Lorentz
- CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| | - Marie-Christine Birling
- CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| | - Claire Gaveriaux-Ruff
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France
| | - Yann Herault
- CNRS, INSERM Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, Illkirch, France.,CNRS, INSERM, PHENOMIN-Institut Clinique de la Souris, Université de Strasbourg, Illkirch, France
| |
Collapse
|
6
|
Qi B, Dai S, Song Y, Shen D, Li F, Wei L, Zhang C, Nie Z, Lin J, Cai L, Ge J. Blockade of Na V1.8 Increases the Susceptibility to Ventricular Arrhythmias During Acute Myocardial Infarction. Front Cardiovasc Med 2021; 8:708279. [PMID: 34409080 PMCID: PMC8365037 DOI: 10.3389/fcvm.2021.708279] [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: 05/11/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022] Open
Abstract
SCN10A/NaV1.8 may be associated with a lower risk of ventricular fibrillation in the setting of acute myocardial infarction (AMI), but if and by which mechanism NaV1.8 impacts on ventricular electrophysiology is still a matter of debate. The purpose of this study was to elucidate the contribution of NaV1.8 in ganglionated plexi (GP) to ventricular arrhythmias in the AMI model. Twenty beagles were randomized to either the A-803467 group (n = 10) or the control group (n = 10). NaV1.8 blocker (A-803467, 1 μmol/0.5 mL per GP) or DMSO (0.5 mL per GP) was injected into four major GPs. Ventricular effective refractory period, APD90, ventricular fibrillation threshold, and the incidence of ventricular arrhythmias were measured 1 h after left anterior descending coronary artery occlusion. A-803467 significantly shortened ventricular effective refractory period, APD90, and ventricular fibrillation threshold compared to control. In the A-803467 group, the incidence of ventricular arrhythmias was significantly higher compared to control. A-803467 suppressed the slowing of heart rate response to high-frequency electrical stimulation of the anterior right GP, suggesting that A-803467 could inhibit GP activity. SCN10A/NaV1.8 was readily detected in GPs, but was not validated in ventricles by quantitative RT-PCR, western blot and immunohistochemistry. While SCN10A/NaV1.8 is detectible in canine GPs but not in ventricles, blockade of NaV1.8 in GP increases the incidence of ventricular arrhythmias in AMI hearts. Our study shows for the first time an influence of SCN10A/NaV1.8 on the regulation of ventricular arrhythmogenesis via modulating GP activity in the AMI model.
Collapse
Affiliation(s)
- Baozhen Qi
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Shimo Dai
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Yu Song
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Dongli Shen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Fuhai Li
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Lanfang Wei
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Chunyu Zhang
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Zhenning Nie
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Jiaxiong Lin
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| | - Lidong Cai
- Department of Cardiology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Disease, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Contribution of Multiple Inherited Variants to Autism Spectrum Disorder (ASD) in a Family with 3 Affected Siblings. Genes (Basel) 2021; 12:genes12071053. [PMID: 34356069 PMCID: PMC8303619 DOI: 10.3390/genes12071053] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/21/2021] [Accepted: 07/07/2021] [Indexed: 12/13/2022] Open
Abstract
Autism Spectrum Disorder (ASD) is the most common neurodevelopmental disorder in children and shows high heritability. However, how inherited variants contribute to ASD in multiplex families remains unclear. Using whole-genome sequencing (WGS) in a family with three affected children, we identified multiple inherited DNA variants in ASD-associated genes and pathways (RELN, SHANK2, DLG1, SCN10A, KMT2C and ASH1L). All are shared among the three children, except ASH1L, which is only present in the most severely affected child. The compound heterozygous variants in RELN, and the maternally inherited variant in SHANK2, are considered to be major risk factors for ASD in this family. Both genes are involved in neuron activities, including synaptic functions and the GABAergic neurotransmission system, which are highly associated with ASD pathogenesis. DLG1 is also involved in synapse functions, and KMT2C and ASH1L are involved in chromatin organization. Our data suggest that multiple inherited rare variants, each with a subthreshold and/or variable effect, may converge to certain pathways and contribute quantitatively and additively, or alternatively act via a 2nd-hit or multiple-hits to render pathogenicity of ASD in this family. Additionally, this multiple-hits model further supports the quantitative trait hypothesis of a complex genetic, multifactorial etiology for the development of ASDs.
Collapse
|
8
|
Proteomic analysis of bone marrow-derived mesenchymal stem cell extracellular vesicles from healthy donors: implications for proliferation, angiogenesis, Wnt signaling, and the basement membrane. Stem Cell Res Ther 2021; 12:328. [PMID: 34090527 PMCID: PMC8180068 DOI: 10.1186/s13287-021-02405-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023] Open
Abstract
Background Bone marrow-derived mesenchymal stem cells (BM-MSCs) have shown therapeutic potential in various in vitro and in vivo studies in cutaneous wound healing. Furthermore, there are ubiquitous studies highlighting the pro-regenerative effects of BM-MSC extracellular vesicles (BM-MSC EVs). The similarities and differences in BM-MSC EV cargo among potential healthy donors are not well understood. Variation in EV protein cargo is important to understand, as it may be useful in identifying potential therapeutic applications in clinical trials. We hypothesized that the donors would share both important similarities and differences in cargo relating to cell proliferation, angiogenesis, Wnt signaling, and basement membrane formation—processes shown to be critical for effective cutaneous wound healing. Methods We harvested BM-MSC EVs from four healthy human donors who underwent strict screening for whole bone marrow donation and further Good Manufacturing Practices-grade cell culture expansion for candidate usage in clinical trials. BM-MSC EV protein cargo was determined via mass spectrometry and Proteome Discoverer software. Corresponding proteomic networks were analyzed via the UniProt Consortium and STRING consortium databases. Results More than 3000 proteins were identified in each of the donors, sharing > 600 proteins among all donors. Despite inter-donor variation in protein identities, there were striking similarities in numbers of proteins per biological functional category. In terms of biologic function, the proteins were most associated with transport of ions and proteins, transcription, and the cell cycle, relating to cell proliferation. The donors shared essential cargo relating to angiogenesis, Wnt signaling, and basement membrane formation—essential processes in modulating cutaneous wound repair. Conclusions Healthy donors of BM-MSC EVs contain important similarities and differences among protein cargo that may play important roles in their pro-regenerative functions. Further studies are needed to correlate proteomic signatures to functional outcomes in cutaneous repair.
Collapse
|
9
|
Man JCK, Bosada FM, Scholman KT, Offerhaus JA, Walsh R, van Duijvenboden K, van Eif VWW, Bezzina CR, Verkerk AO, Boukens BJ, Barnett P, Christoffels VM. Variant Intronic Enhancer Controls SCN10A-short Expression and Heart Conduction. Circulation 2021; 144:229-242. [PMID: 33910361 DOI: 10.1161/circulationaha.121.054083] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Genetic variants in SCN10A, encoding the neuronal voltage-gated sodium channel NaV1.8, are strongly associated with atrial fibrillation, Brugada syndrome, cardiac conduction velocities, and heart rate. The cardiac function of SCN10A has not been resolved, however, and diverging mechanisms have been proposed. Here, we investigated the cardiac expression of SCN10A and the function of a variant-sensitive intronic enhancer previously linked to the regulation of SCN5A, encoding the major essential cardiac sodium channel NaV1.5. METHODS The expression of SCN10A was investigated in mouse and human hearts. With the use of CRISPR/Cas9 genome editing, the mouse intronic enhancer was disrupted, and mutant mice were characterized by transcriptomic and electrophysiological analyses. The association of genetic variants at SCN5A-SCN10A enhancer regions and gene expression were evaluated by genome-wide association studies single-nucleotide polymorphism mapping and expression quantitative trait loci analysis. RESULTS We found that cardiomyocytes of the atria, sinoatrial node, and ventricular conduction system express a short transcript comprising the last 7 exons of the gene (Scn10a-short). Transcription occurs from an intronic enhancer-promoter complex, whereas full-length Scn10a transcript was undetectable in the human and mouse heart. Expression quantitative trait loci analysis revealed that the genetic variants in linkage disequilibrium with genetic variant rs6801957 in the intronic enhancer associate with SCN10A transcript levels in the heart. Genetic modification of the enhancer in the mouse genome led to reduced cardiac Scn10a-short expression in atria and ventricles, reduced cardiac sodium current in atrial cardiomyocytes, atrial conduction slowing and arrhythmia, whereas the expression of Scn5a, the presumed enhancer target gene, remained unaffected. In patch-clamp transfection experiments, expression of Scn10a-short-encoded NaV1.8-short increased NaV1.5-mediated sodium current. We propose that noncoding genetic variation modulates transcriptional regulation of Scn10a-short in cardiomyocytes that impacts NaV1.5-mediated sodium current and heart rhythm. CONCLUSIONS Genetic variants in and around SCN10A modulate enhancer function and expression of a cardiac-specific SCN10A-short transcript. We propose that noncoding genetic variation modulates transcriptional regulation of a functional C-terminal portion of NaV1.8 in cardiomyocytes that impacts on NaV1.5 function, cardiac conduction velocities, and arrhythmia susceptibility.
Collapse
Affiliation(s)
- Joyce C K Man
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Fernanda M Bosada
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Koen T Scholman
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Joost A Offerhaus
- Department of Experimental Cardiology (J.A.O., R.W., C.R.B., A.O.V., B.J.B.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Roddy Walsh
- Department of Experimental Cardiology (J.A.O., R.W., C.R.B., A.O.V., B.J.B.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Karel van Duijvenboden
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Vincent W W van Eif
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology (J.A.O., R.W., C.R.B., A.O.V., B.J.B.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Arie O Verkerk
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Department of Experimental Cardiology (J.A.O., R.W., C.R.B., A.O.V., B.J.B.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Bastiaan J Boukens
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Department of Experimental Cardiology (J.A.O., R.W., C.R.B., A.O.V., B.J.B.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Phil Barnett
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands.,Amsterdam Cardiovascular Sciences, Amsterdam Reproduction and Development (J.C.K.M., F.M.B., K.T.S., K.v.D., V.W.W.v.E., A.O.V., B.J.B., P.B., V.M.C.), Amsterdam UMC, University of Amsterdam, location AMC, The Netherlands
| |
Collapse
|
10
|
Ahmadirad N, Fathollahi Y, Janahmadi M, Ghasemi Z, Shojaei A, Rezaei M, Barkley V, Mirnajafi-Zadeh J. The role of α adrenergic receptors in mediating the inhibitory effect of electrical brain stimulation on epileptiform activity in rat hippocampal slices. Brain Res 2021; 1765:147492. [PMID: 33887250 DOI: 10.1016/j.brainres.2021.147492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/06/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
The Inhibitory effect of electrical low-frequency stimulation (LFS) on neuronal excitability and seizure occurrence has been indicated in experimental models, but the precise mechanism has not established. This investigation was intended to figure out the role of α1 and α2 adrenergic receptors in LFS' inhibitory effect on neuronal excitability. Epileptiform activity induced in an in vitro rat hippocampal slice preparation by high K+ ACSF and LFS (900 square wave pulses at 1 Hz) was administered at the beginning of epileptiform activity to the Schaffer collaterals. In CA1 pyramidal neurons, the electrophysiological properties were measured at the baseline, before high K+ ACSF washout, and at 15 min after high K+ ACSF washout using whole-cell, patch-clamp recording. Results indicated that after high K+ ACSF washout, prazosine (10 µM; α1 adrenergic receptor antagonist) and yohimbine (5 µM; α2 adrenergic receptor antagonist) suppressed the LFS' effect of reducing rheobase current and utilization time following depolarizing ramp current, the latency to the first spike following a depolarizing current and latency to the first rebound action potential following hyperpolarizing current pulses. Thus, it may be proposed that LFS' inhibitory action on the neuronal hyperexcitability, in some way, is mediated by α1 and α2 adrenergic receptors.
Collapse
Affiliation(s)
- Nooshin Ahmadirad
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yaghoub Fathollahi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahyar Janahmadi
- Department of Physiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Ghasemi
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Amir Shojaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mahmoud Rezaei
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Victoria Barkley
- Krembil Research Institute, University Health Network, Toronto, Canada
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Institute for Brain Sciences and Cognition, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
11
|
Xue Y, Chidiac C, Herault Y, Gaveriaux-Ruff C. Pain behavior in SCN9A (Nav1.7) and SCN10A (Nav1.8) mutant rodent models. Neurosci Lett 2021; 753:135844. [PMID: 33775738 DOI: 10.1016/j.neulet.2021.135844] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 11/18/2022]
Abstract
The two voltage gated sodium channels Nav1.7 and Nav1.8 are expressed in the peripheral nervous system and involved in various pain conditions including inflammatory and neuropathic pain. Rodent models bearing deletions or mutations of the corresponding genes, Scn9a and Scn10a, were created in order to understand the role of these channels in the pathophysiological mechanism underlying pain symptoms. This review summarizes the pain behavior profiles reported in Scn9a and Scn10a rodent models. The complete loss-of-function or knockout (KO) of Scn9a or Scn10a and the conditional KO (cKO) of Scn9a in specific cell populations were shown to decrease sensitivity to various pain stimuli. The Possum mutant mice bearing a dominant hypermorphic mutation in Scn10a revealed higher sensitivity to noxious stimuli. Several gain-of-function mutations were identified in patients with painful small fiber neuropathy. Future knowledge obtained from preclinical models bearing these mutations will allow understanding how these mutations affect pain. In addition, the review gives perspectives for creating models that better mimic patients' pain symptoms in view to developing novel analgesic strategies.
Collapse
Affiliation(s)
- Yaping Xue
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| | - Celeste Chidiac
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| | - Yann Herault
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France.
| | - Claire Gaveriaux-Ruff
- Université de Strasbourg, CNRS, INSERM, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC) Translational Medicine and Neurogenetics Department, Illkirch, France
| |
Collapse
|
12
|
Bennett DL, Clark AJ, Huang J, Waxman SG, Dib-Hajj SD. The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev 2019; 99:1079-1151. [DOI: 10.1152/physrev.00052.2017] [Citation(s) in RCA: 256] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Acute pain signaling has a key protective role and is highly evolutionarily conserved. Chronic pain, however, is maladaptive, occurring as a consequence of injury and disease, and is associated with sensitization of the somatosensory nervous system. Primary sensory neurons are involved in both of these processes, and the recent advances in understanding sensory transduction and human genetics are the focus of this review. Voltage-gated sodium channels (VGSCs) are important determinants of sensory neuron excitability: they are essential for the initial transduction of sensory stimuli, the electrogenesis of the action potential, and neurotransmitter release from sensory neuron terminals. Nav1.1, Nav1.6, Nav1.7, Nav1.8, and Nav1.9 are all expressed by adult sensory neurons. The biophysical characteristics of these channels, as well as their unique expression patterns within subtypes of sensory neurons, define their functional role in pain signaling. Changes in the expression of VGSCs, as well as posttranslational modifications, contribute to the sensitization of sensory neurons in chronic pain states. Furthermore, gene variants in Nav1.7, Nav1.8, and Nav1.9 have now been linked to human Mendelian pain disorders and more recently to common pain disorders such as small-fiber neuropathy. Chronic pain affects one in five of the general population. Given the poor efficacy of current analgesics, the selective expression of particular VGSCs in sensory neurons makes these attractive targets for drug discovery. The increasing availability of gene sequencing, combined with structural modeling and electrophysiological analysis of gene variants, also provides the opportunity to better target existing therapies in a personalized manner.
Collapse
Affiliation(s)
- David L. Bennett
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Alex J. Clark
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Jianying Huang
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Stephen G. Waxman
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| | - Sulayman D. Dib-Hajj
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, Connecticut; and Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
13
|
Zhang F, Zhang C, Xu X, Zhang Y, Gong X, Yang Z, Zhang H, Tang D, Liang S, Liu Z. Naja atra venom peptide reduces pain by selectively blocking the voltage-gated sodium channel Nav1.8. J Biol Chem 2019; 294:7324-7334. [PMID: 30804211 DOI: 10.1074/jbc.ra118.007370] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 02/20/2019] [Indexed: 01/14/2023] Open
Abstract
The voltage-gated sodium channel Nav1.8 is preferentially expressed in peripheral nociceptive neurons and contributes to inflammatory and neuropathic pain. Therefore, Nav1.8 has emerged as one of the most promising analgesic targets for pain relief. Using large-scale screening of various animal-derived toxins and venoms for Nav1.8 inhibitors, here we identified μ-EPTX-Na1a, a 62-residue three-finger peptide from the venom of the Chinese cobra (Naja atra), as a potent inhibitor of Nav1.8, exhibiting high selectivity over other voltage-gated sodium channel subtypes. Using whole-cell voltage-clamp recordings, we observed that purified μ-EPTX-Na1a blocked the Nav1.8 current. This blockade was associated with a depolarizing shift of activation and repolarizing shift of inactivation, a mechanism distinct from that of any other gating modifier toxin identified to date. In rodent models of inflammatory and neuropathic pain, μ-EPTX-Na1a alleviated nociceptive behaviors more potently than did morphine, indicating that μ-EPTX-Na1a has a potent analgesic effect. μ-EPTX-Na1a displayed no evident cytotoxicity and cardiotoxicity and produced no obvious adverse responses in mice even at a dose 30-fold higher than that producing a significant analgesic effect. Our study establishes μ-EPTX-Na1a as a promising lead for the development of Nav1.8-targeting analgesics to manage pain.
Collapse
Affiliation(s)
- Fan Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Changxin Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Xunxun Xu
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Yunxiao Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Xue Gong
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Zuqin Yang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Heng Zhang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Dongfang Tang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Songping Liang
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| | - Zhonghua Liu
- From The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081 Hunan, China
| |
Collapse
|
14
|
Increased Resurgent Sodium Currents in Nav1.8 Contribute to Nociceptive Sensory Neuron Hyperexcitability Associated with Peripheral Neuropathies. J Neurosci 2019; 39:1539-1550. [PMID: 30617209 DOI: 10.1523/jneurosci.0468-18.2018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 10/22/2018] [Accepted: 11/25/2018] [Indexed: 11/21/2022] Open
Abstract
Neuropathic pain is a significant public health challenge, yet the underlying mechanisms remain poorly understood. Painful small fiber neuropathy (SFN) may be caused by gain-of-function mutations in Nav1.8, a sodium channel subtype predominantly expressed in peripheral nociceptive neurons. However, it is not clear how Nav1.8 disease mutations induce sensory neuron hyperexcitability. Here we studied two mutations in Nav1.8 associated with hypersensitive sensory neurons: G1662S reported in painful SFN; and T790A, which underlies increased pain behaviors in the Possum transgenic mouse strain. We show that, in male DRG neurons, these mutations, which impair inactivation, significantly increase TTX-resistant resurgent sodium currents mediated by Nav1.8. The G1662S mutation doubled resurgent currents, and the T790A mutation increased them fourfold. These unusual currents are typically evoked during the repolarization phase of action potentials. We show that the T790A mutation greatly enhances DRG neuron excitability by reducing current threshold and increasing firing frequency. Interestingly, the mutation endows DRG neurons with multiple early afterdepolarizations and leads to substantial prolongation of action potential duration. In DRG neurons, siRNA knockdown of sodium channel β4 subunits fails to significantly alter T790A current density but reduces TTX-resistant resurgent currents by 56%. Furthermore, DRG neurons expressing T790A channels exhibited reduced excitability with fewer early afterdepolarizations and narrower action potentials after β4 knockdown. Together, our data demonstrate that open-channel block of TTX-resistant currents, enhanced by gain-of-function mutations in Nav1.8, can make major contributions to the hyperexcitability of nociceptive neurons, likely leading to altered sensory phenotypes including neuropathic pain in SFN.SIGNIFICANCE STATEMENT This work demonstrates that two disease mutations in the voltage-gated sodium channel Nav1.8 that induce nociceptor hyperexcitability increase resurgent currents. Nav1.8 is crucial for pain sensations. Because resurgent currents are evoked during action potential repolarization, they can be crucial regulators of action potential activity. Our data indicate that increased Nav1.8 resurgent currents in DRG neurons greatly prolong action potential duration and enhance repetitive firing. We propose that Nav1.8 open-channel block is a major factor in Nav1.8-associated pain mechanisms and that targeting the molecular mechanism underlying these unique resurgent currents represents a novel therapeutic target for the treatment of aberrant pain sensations.
Collapse
|
15
|
Hoffstaetter LJ, Bagriantsev SN, Gracheva EO. TRPs et al.: a molecular toolkit for thermosensory adaptations. Pflugers Arch 2018; 470:745-759. [PMID: 29484488 PMCID: PMC5945325 DOI: 10.1007/s00424-018-2120-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/03/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
The ability to sense temperature is crucial for the survival of an organism. Temperature influences all biological operations, from rates of metabolic reactions to protein folding, and broad behavioral functions, from feeding to breeding, and other seasonal activities. The evolution of specialized thermosensory adaptations has enabled animals to inhabit extreme temperature niches and to perform specific temperature-dependent behaviors. The function of sensory neurons depends on the participation of various types of ion channels. Each of the channels involved in neuronal excitability, whether through the generation of receptor potential, action potential, or the maintenance of the resting potential have temperature-dependent properties that can tune the neuron's response to temperature stimuli. Since the function of all proteins is affected by temperature, animals need adaptations not only for detecting different temperatures, but also for maintaining sensory ability at different temperatures. A full understanding of the molecular mechanism of thermosensation requires an investigation of all channel types at each step of thermosensory transduction. A fruitful avenue of investigation into how different molecules can contribute to the fine-tuning of temperature sensitivity is to study the specialized adaptations of various species. Given the diversity of molecular participants at each stage of sensory transduction, animals have a toolkit of channels at their disposal to adapt their thermosensitivity to their particular habitats or behavioral circumstances.
Collapse
Affiliation(s)
- Lydia J Hoffstaetter
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, 06520-8026, USA.
| |
Collapse
|
16
|
Abou Ziki MD, Seidelmann SB, Smith E, Atteya G, Jiang Y, Fernandes RG, Marieb MA, Akar JG, Mani A. Deleterious protein-altering mutations in the SCN10A voltage-gated sodium channel gene are associated with prolonged QT. Clin Genet 2018; 93:741-751. [PMID: 28407228 PMCID: PMC5640462 DOI: 10.1111/cge.13036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/27/2017] [Accepted: 04/09/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Long QT syndrome (LQT) is a pro-arrhythmogenic condition with life-threatening complications. Fifteen genes have been associated with congenital LQT, however, the genetic causes remain unknown in more than 20% of cases. MATERIALS AND METHODS Eighteen patients with history of palpitations, pre-syncope, syncope and prolonged QT were referred to the Yale Cardiovascular Genetics Program. All subjects underwent whole-exome sequencing (WES) followed by confirmatory Sanger sequencing. Mutation burden analysis was carried out using WES data from 16 subjects with no identifiable cause of LQT. RESULTS Deleterious and novel SCN10A mutations were identified in 3 of the 16 patients (19%) with idiopathic LQT. These included 2 frameshifts and 1 missense variants (p.G810fs, p.R1259Q, and p.P1877fs). Further analysis identified 2 damaging SCN10A mutations with allele frequencies of approximately 0.2% (p.R14L and p.R1268Q) in 2 independent cases. None of the SCN10A mutation carriers had mutations in known arrhythmia genes. Damaging SCN10A mutations (p.R209H and p.R485C) were also identified in the 2 subjects on QT prolonging medications. CONCLUSION Our findings implicate SCN10A in LQT. The presence of frameshift mutations suggests loss-of-function as the underlying disease mechanism. The common association with atrial fibrillation suggests a unique mechanism of disease for this LQT gene.
Collapse
Affiliation(s)
- Maen D. Abou Ziki
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02115
| | - Emily Smith
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Gourg Atteya
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Yuexin Jiang
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Rodolfo Gil Fernandes
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Mark A. Marieb
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Joseph G. Akar
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
| | - Arya Mani
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510
- Deparetment of Genetics, Yale University School of Medicine, New Haven, CT, 06510
| |
Collapse
|
17
|
Abstract
Of somatosensory modalities cold is one of the more ambiguous percepts, evoking the pleasant sensation of cooling, the stinging bite of cold pain, and welcome relief from chronic pain. Moreover, unlike the precipitous thermal thresholds for heat activation of thermosensitive afferent neurons, thresholds for cold fibers are across a range of cool to cold temperatures that spans over 30°C. Until recently, how cold produces this myriad of biologic effects was unknown. However, recent advances in our understanding of cold mechanisms at the behavioral, physiologic, and cellular level have begun to provide insights into this sensory modality. The identification of a number of ion channels that either serve as the principal detectors of a cold stimulus in the peripheral nervous system, or are part of a differential expression pattern of channels that maintain cell excitability in the cold, endows select neurons with properties that are amenable to electric signaling in the cold. This chapter highlights the current understanding of the molecules involved in cold transduction in the mammalian peripheral nervous system, as well as presenting a hypothetic model to account for the broad range of cold thermal thresholds and distinct functions of cold fibers in perception, pain, and analgesia.
Collapse
Affiliation(s)
- David D McKemy
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States.
| |
Collapse
|
18
|
Ghasemi Z, Naderi N, Shojaei A, Raoufy MR, Ahmadirad N, Mirnajafi-Zadeh J. Effect of Low-Frequency Electrical Stimulation on the High-K+-Induced Neuronal Hyperexcitability in Rat Hippocampal Slices. Neuroscience 2018; 369:87-96. [DOI: 10.1016/j.neuroscience.2017.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 12/23/2022]
|
19
|
Yu L, Wang M, Hu D, Huang B, Zhou L, Zhou X, Wang Z, Wang S, Jiang H. Blocking the Nav1.8 channel in the left stellate ganglion suppresses ventricular arrhythmia induced by acute ischemia in a canine model. Sci Rep 2017; 7:534. [PMID: 28373696 PMCID: PMC5428783 DOI: 10.1038/s41598-017-00642-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 03/07/2017] [Indexed: 01/01/2023] Open
Abstract
Left stellate ganglion (LSG) hyperactivity promotes ischemia induced ventricular arrhythmia (VA). Blocking the Nav1.8 channel decreases neuron activity. Therefore, the present study aimed to investigate whether blocking the Nav1.8 channel with its specific blocker A-803467 in the LSG reduces sympathetic activity and exerts anti-arrhythmic effects. Forty canines were divided into dimethylsulfoxide (DMSO) group and 10 mM, 15 mM, and 20 mM A-803467 groups. A volume of 0.1 ml of A-803467 or DMSO was injected into the LSG. The ventricular electrophysiological parameters, LSG function were measured before and 30 min after the injection. VA was assessed for 60 min after ischemia and then LSG tissues were collected for molecular biological experiments. Compared with DMSO, concentration-dependent prolonged action potential duration and effective refractory period, decreased LSG function were identified after A-803467 treatment. Moreover, the severity of ischemia induced VA was decreased in A-803467 groups. Furthermore, decreased nerve growth factor, decreased c-fos and increased sympathetic neuron apoptosis were found in the LSG after A-803467 injection. In conclusion, blocking the Nav1.8 channel could significantly attenuate ischemia-induced VA, primarily by suppressing LSG activity.
Collapse
Affiliation(s)
- Lilei Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Dan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China.,Masonic Medical Research Laboratory, 2150 Bleecker Street, Utica, New York, 13501-1787, USA
| | - Bing Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Liping Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xiaoya Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Zhuo Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Songyun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
20
|
Association of SCN10A Polymorphisms with the Recurrence of Atrial Fibrillation after Catheter Ablation in a Chinese Han Population. Sci Rep 2017; 7:44003. [PMID: 28281580 PMCID: PMC5345091 DOI: 10.1038/srep44003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 02/02/2017] [Indexed: 11/08/2022] Open
Abstract
The nonsynonymous SCN10A single nucleotide polymorphism (SNP) rs6795970 has been reported to associate with PR interval and atrial fibrillation (AF) and in strong linkage disequilibrium (LD) with the AF-associated SNP rs6800541. In this study, we investigated whether rs6795970 polymorphisms are associated with AF recurrence after catheter ablation. A total of 502 consecutive patients with AF who underwent catheter ablation were included. AF recurrence was defined as a documented episode of any atrial arrhythmias lasting ≥30 s after a blanking period of 3 months. AF recurrence was observed between 3 and 12 months after catheter ablation in 24.5% of the patients. There was a significant difference in the allele distribution (p = 7.86 × 10−5) and genotype distribution (p = 1.42 × 10−5) of rs6795970 between the AF recurrence and no recurrence groups. In a multivariate analysis, we identified the following independent predictors of AF recurrence: the rs6795970 genotypes in an additive model (OR 0.36, 95%CI 0.22~0.60, p = 7.04 × 10−5), a history of AF ≥36 months (OR 3.57, 95%CI 2.26~5.63, p = 4.33 × 10−8) and left atrial diameter (LAD) ≥40 mm (OR 1.85, 95%CI 1.08~3.19, p = 0.026). These data suggest that genetic variation in SCN10A may play an important role in predicting AF recurrence after catheter ablation in the Chinese Han population.
Collapse
|
21
|
Voltage-gated sodium channels and pain-related disorders. Clin Sci (Lond) 2016; 130:2257-2265. [DOI: 10.1042/cs20160041] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 09/15/2016] [Indexed: 11/17/2022]
Abstract
Voltage-gated sodium channels (VGSCs) are heteromeric transmembrane protein complexes. Nine homologous members, SCN1A–11A, make up the VGSC gene family. Sodium channel isoforms display a wide range of kinetic properties endowing different neuronal types with distinctly varied firing properties. Among the VGSCs isoforms, Nav1.7, Nav1.8 and Nav1.9 are preferentially expressed in the peripheral nervous system. These isoforms are known to be crucial in the conduction of nociceptive stimuli with mutations in these channels thought to be the underlying cause of a variety of heritable pain disorders. This review provides an overview of the current literature concerning the role of VGSCs in the generation of pain and heritable pain disorders.
Collapse
|
22
|
Volland C, Bremer S, Hellenkamp K, Hartmann N, Dybkova N, Khadjeh S, Kutschenko A, Liebetanz D, Wagner S, Unsöld B, Didié M, Toischer K, Sossalla S, Hasenfuß G, Seidler T. Enhanced cardiac TBC1D10C expression lowers heart rate and enhances exercise capacity and survival. Sci Rep 2016; 6:33853. [PMID: 27667030 PMCID: PMC5036039 DOI: 10.1038/srep33853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 09/05/2016] [Indexed: 11/21/2022] Open
Abstract
TBC1D10C is a protein previously demonstrated to bind and inhibit Ras and Calcineurin. In cardiomyocytes, also CaMKII is inhibited and all three targeted enzymes are known to promote maladaptive cardiomyocyte hypertrophy. Here, in accordance with lack of Calcineurin inhibition in vivo, we did not observe a relevant anti-hypertrophic effect despite inhibition of Ras and CaMKII. However, cardiomyocyte-specific TBC1D10C overexpressing transgenic mice exhibited enhanced longevity. Ejection fraction and exercise capacity were enhanced in transgenic mice, but shortening of isolated cardiomyocytes was not increased. This suggests longevity resulted from enhanced cardiac performance but independent of cardiomyocyte contractile force. In further search for mechanisms, a transcriptome-wide analysis revealed expressional changes in several genes pertinent to control of heart rate (HR) including Hcn4, Scn10a, Sema3a and Cacna2d2. Indeed, telemetric holter recordings demonstrated slower atrial conduction and significantly lower HR. Pharmacological reduction of HR was previously demonstrated to enhance survival in mice. Thus, in addition to inhibition of stress signaling, TBC1D10C economizes generation of cardiac output via HR reduction, enhancing exercise capacity and survival. TBC1D10C may be a new target for HR reduction and longevity.
Collapse
Affiliation(s)
- Cornelia Volland
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Sebastian Bremer
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Kristian Hellenkamp
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Nico Hartmann
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Nataliya Dybkova
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Sara Khadjeh
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Anna Kutschenko
- Department of Clinical Neurophysiology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - David Liebetanz
- Department of Clinical Neurophysiology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Bernhard Unsöld
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
- Department of Internal Medicine II, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Michael Didié
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
- Institute of Pharmacology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Karl Toischer
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Samuel Sossalla
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Gerd Hasenfuß
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| | - Tim Seidler
- Department of Cardiology and Pulmonology, Georg-August-University, Robert-Koch Str. 40, 37075 Göttingen, Germany
| |
Collapse
|
23
|
Kochunov P, Ganjgahi H, Winkler A, Kelly S, Shukla DK, Du X, Jahanshad N, Rowland L, Sampath H, Patel B, O'Donnell P, Xie Z, Paciga SA, Schubert CR, Chen J, Zhang G, Thompson PM, Nichols TE, Hong LE. Heterochronicity of white matter development and aging explains regional patient control differences in schizophrenia. Hum Brain Mapp 2016; 37:4673-4688. [PMID: 27477775 DOI: 10.1002/hbm.23336] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 07/21/2016] [Accepted: 07/24/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Altered brain connectivity is implicated in the development and clinical burden of schizophrenia. Relative to matched controls, schizophrenia patients show (1) a global and regional reduction in the integrity of the brain's white matter (WM), assessed using diffusion tensor imaging (DTI) fractional anisotropy (FA), and (2) accelerated age-related decline in FA values. In the largest mega-analysis to date, we tested if differences in the trajectories of WM tract development influenced patient-control differences in FA. We also assessed if specific tracts showed exacerbated decline with aging. METHODS Three cohorts of schizophrenia patients (total n = 177) and controls (total n = 249; age = 18-61 years) were ascertained with three 3T Siemens MRI scanners. Whole-brain and regional FA values were extracted using ENIGMA-DTI protocols. Statistics were evaluated using mega- and meta-analyses to detect effects of diagnosis and age-by-diagnosis interactions. RESULTS In mega-analysis of whole-brain averaged FA, schizophrenia patients had lower FA (P = 10-11 ) and faster age-related decline in FA (P = 0.02) compared with controls. Tract-specific heterochronicity measures, that is, abnormal rates of adolescent maturation and aging explained approximately 50% of the regional variance effects of diagnosis and age-by-diagnosis interaction in patients. Interactive, three-dimensional visualization of the results is available at www.enigma-viewer.org. CONCLUSION WM tracts that mature later in life appeared more sensitive to the pathophysiology of schizophrenia and were more susceptible to faster age-related decline in FA values. Hum Brain Mapp 37:4673-4688, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Habib Ganjgahi
- Department of Statistics, University of Warwick, Warwick, United Kingdom
| | | | - Sinead Kelly
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, California
| | - Dinesh K Shukla
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Xiaoming Du
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Neda Jahanshad
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, California
| | - Laura Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hemalatha Sampath
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Binish Patel
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| | - Patricio O'Donnell
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc, 610 Main Street, Cambridge, Massachusetts, 02139
| | - Zhiyong Xie
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc, 610 Main Street, Cambridge, Massachusetts, 02139
| | - Sara A Paciga
- Enterprise Scientific Technology Operations, Worldwide Research and Development, Pfizer Inc, Eastern Point Rd, Groton, Connecticut, 06340
| | - Christian R Schubert
- Enterprise Scientific Technology Operations, Worldwide Research and Development, Pfizer Inc, Eastern Point Rd, Groton, Connecticut, 06340.,Biogen, Cambridge, Massachusetts, 02142
| | - Jian Chen
- Department of Computer Science and Electrical Engineering, University of Maryland, Baltimore County, Maryland, 21250
| | - Guohao Zhang
- Department of Computer Science and Electrical Engineering, University of Maryland, Baltimore County, Maryland, 21250
| | - Paul M Thompson
- Imaging Genetics Center, Keck School of Medicine of USC, Marina del Rey, California
| | - Thomas E Nichols
- Department of Statistics, University of Warwick, Warwick, United Kingdom
| | - L Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
24
|
Han Z, Jiang Y, Xiao F, Cao K, Wang DW. The effects of A-803467 on cardiac Nav1.5 channels. Eur J Pharmacol 2015; 754:52-60. [DOI: 10.1016/j.ejphar.2015.02.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 02/08/2015] [Accepted: 02/11/2015] [Indexed: 12/19/2022]
|
25
|
Bagriantsev SN, Gracheva EO. Molecular mechanisms of temperature adaptation. J Physiol 2015; 593:3483-91. [PMID: 25433072 DOI: 10.1113/jphysiol.2014.280446] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/21/2014] [Indexed: 11/08/2022] Open
Abstract
Thermal perception is a fundamental physiological process pertaining to the vast majority of organisms. In vertebrates, environmental temperature is detected by the primary afferents of the somatosensory neurons in the skin, which express a 'choir' of ion channels tuned to detect particular temperatures. Nearly two decades of research have revealed a number of receptor ion channels that mediate the perception of several temperature ranges, but most still remain molecularly orphaned. Yet even within this well-researched realm, most of our knowledge largely pertains to two closely related species of rodents, mice and rats. While these are standard biomedical research models, mice and rats provide a limited perspective to elucidate the general principles that drive somatosensory evolution. In recent years, significant advances have been made in understanding the molecular mechanism of temperature adaptation in evolutionarily distant vertebrates and in organisms with acute thermal sensitivity. These studies have revealed the remarkable versatility of the somatosensory system and highlighted adaptations at the molecular level, which often include changes in biophysical properties of ion channels from the transient receptor potential family. Exploiting non-standard animal models has the potential to provide unexpected insights into general principles of thermosensation and thermoregulation, unachievable using the rodent model alone.
Collapse
Affiliation(s)
- Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT , 06520, USA
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT , 06520, USA.,Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT , 06520, USA
| |
Collapse
|
26
|
Kochunov P, Chiappelli J, Wright SN, Rowland LM, Patel B, Wijtenburg SA, Nugent K, McMahon RP, Carpenter WT, Muellerklein F, Sampath H, Hong LE. Multimodal white matter imaging to investigate reduced fractional anisotropy and its age-related decline in schizophrenia. Psychiatry Res 2014; 223:148-56. [PMID: 24909602 PMCID: PMC4100065 DOI: 10.1016/j.pscychresns.2014.05.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 04/18/2014] [Accepted: 05/08/2014] [Indexed: 01/14/2023]
Abstract
We hypothesized that reduced fractional anisotropy (FA) of water diffusion and its elevated aging-related decline in schizophrenia patients may be caused by elevated hyperintensive white matter (HWM) lesions, by reduced permeability-diffusivity index (PDI), or both. We tested this hypothesis in 40/30 control/patient participants. FA values for the corpus callosum were calculated from high angular resolution diffusion tensor imaging (DTI). Whole-brain volume of HWM lesions was quantified by 3D-T2w-fluid-attenuated inversion recovery (FLAIR) imaging. PDI for corpus callosum was ascertained using multi b-value diffusion imaging (15 b-shells with 30 directions per shell). Patients had significantly lower corpus callosum FA values, and there was a significant age-by-diagnosis interaction. Patients also had significantly reduced PDI but no difference in HWM volume. PDI and HWM volume were significant predictors of FA and captured the diagnosis-related variance. Separately, PDI robustly explained FA variance in schizophrenia patients, but not in controls. Conversely, HWM volume made equally significant contributions to variability in FA in both groups. The diagnosis-by-age effect of FA was explained by a PDI-by-diagnosis interaction. Post hoc testing showed a similar trend for PDI of gray mater. Our study demonstrated that reduced FA and its accelerated decline with age in schizophrenia were explained by pathophysiology indexed by PDI, rather than HWM volume.
Collapse
Affiliation(s)
- Peter Kochunov
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA; Department of Physics, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| | - Joshua Chiappelli
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Susan N. Wright
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Laura M. Rowland
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Benish Patel
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - S. Andrea Wijtenburg
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Katie Nugent
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Robert P. McMahon
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - William T. Carpenter
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Florian Muellerklein
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - Hemalatha Sampath
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| | - L. Elliot Hong
- Maryland Psychiatric Research Center, Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD 21228, USA
| |
Collapse
|
27
|
Park DS, Fishman GI. Nav-igating through a complex landscape: SCN10A and cardiac conduction. J Clin Invest 2014; 124:1460-2. [PMID: 24642462 DOI: 10.1172/jci75240] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Genome-wide association studies (GWAS) have implicated SCN10A, which encodes a nociceptor-associated voltage-gated sodium channel subunit, as a modulator of cardiac conduction; however, this role has traditionally been ascribed to SCN5A, which is highly expressed in cardiac muscle. SCN10A is believed to affect cardiac conduction either directly through cardiomyocytes or indirectly via intracardiac neurons. In this issue of the JCI, van den Boogaard and colleagues introduce a third possibility: that the SCN10A locus acts as an enhancer of SCN5A gene expression. The authors demonstrate that SCN10A expression is negligible within human and murine hearts, and that a T-box enhancer within the SCN10A locus drives SCN5A expression within cardiomyocytes. This work reasserts SCN5A as the key determinant of cardiac conduction and highlights the importance of deciphering the functionality of coding versus noncoding regions when interpreting GWAS data.
Collapse
|
28
|
T-type calcium channels in chronic pain: mouse models and specific blockers. Pflugers Arch 2014; 466:707-17. [PMID: 24590509 DOI: 10.1007/s00424-014-1484-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 02/13/2014] [Indexed: 02/01/2023]
Abstract
Pain is a quite frequent complaint accompanying numerous pathologies. Among these pathological cases, neuropathies are retrieved with identified etiologies (chemotherapies, diabetes, surgeries…) and also more diffuse syndromes such as fibromyalgia. More broadly, pain is one of the first consequences of the majority of inherited diseases. Despite its importance for the quality of life, current pain management is limited to drugs that are either old or with a limited efficacy or that possess a bad benefit/risk ratio. As no new pharmacological concept has led to new analgesics in the last decades, the discovery of medications is needed, and to this aim the identification of new druggable targets in pain transmission is a first step. Therefore, studies of ion channels in pain pathways are extremely active. This is particularly true with ion channels in peripheral sensory neurons in dorsal root ganglia (DRG) known now to express unique sets of these channels. Moreover, both spinal and supraspinal levels are clearly important in pain modulation. Among these ion channels, we and others revealed the important role of low voltage-gated calcium channels in cellular excitability in different steps of the pain pathways. These channels, by being activated nearby resting membrane potential have biophysical characteristics suited to facilitate action potential generation and rhythmicity. In this review, we will review the current knowledge on the role of these channels in the perception and modulation of pain.
Collapse
|
29
|
A gain-of-function voltage-gated sodium channel 1.8 mutation drives intense hyperexcitability of A- and C-fiber neurons. Pain 2014; 155:896-905. [PMID: 24447515 DOI: 10.1016/j.pain.2014.01.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 12/27/2013] [Accepted: 01/14/2014] [Indexed: 12/12/2022]
Abstract
Therapeutic use of general sodium channel blockers, such as lidocaine, can substantially reduce the enhanced activity in sensory neurons that accompanies chronic pain after nerve or tissue injury. However, because these general blockers have significant side effects, there is great interest in developing inhibitors that specifically target subtypes of sodium channels. Moreover, some idiopathic small-fiber neuropathies are driven by gain-of-function mutations in specific sodium channel subtypes. In the current study, we focus on one subtype, the voltage-gated sodium channel 1.8 (Nav1.8). Nav1.8 is preferentially expressed in nociceptors, and gain-of-function mutations in Nav1.8 result in painful mechanical hypersensitivity in humans. Here, we used the recently developed gain-of-function Nav1.8 transgenic mouse strain, Possum, to investigate Nav1.8-mediated peripheral afferent hyperexcitability. This gain-of-function mutation resulted in markedly increased mechanically evoked action potential firing in subclasses of Aβ, Aδ, and C fibers. Moreover, mechanical stimuli initiated bursts of action potential firing in specific subpopulations that continued for minutes after removal of the force and were not susceptible to conduction failure. Surprisingly, despite the intense afferent firing, the behavioral effects of the Nav1.8 mutation were quite modest, as only frankly noxious stimuli elicited enhanced pain behavior. These data demonstrate that a Nav1.8 gain-of-function point mutation contributes to intense hyperexcitability along the afferent axon within distinct sensory neuron subtypes.
Collapse
|
30
|
Qi B, Wei Y, Chen S, Zhou G, Li H, Xu J, Ding Y, Lu X, Zhao L, Zhang F, Chen G, Zhao J, Liu S. Nav1.8 channels in ganglionated plexi modulate atrial fibrillation inducibility. Cardiovasc Res 2014; 102:480-6. [PMID: 24419303 DOI: 10.1093/cvr/cvu005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Emerging evidences indicate that SCN10A/NaV1.8 is associated with cardiac conduction and atrial fibrillation, but the exact role of NaV1.8 in cardiac electrophysiology remains poorly understood. The present study was designed to investigate the effects of blocking NaV1.8 channels in cardiac ganglionated plexi (GP) on modulating cardiac conduction and atrial fibrillation inducibility in the canine model. METHODS AND RESULTS Thirteen mongrel dogs were randomly enrolled. Right cervical vagus nerve stimulation (VNS) was applied to determine its effects on the sinus rate, ventricular rate during atrial fibrillation, PR interval, atrial effective refractory period, and the cumulative window of vulnerability. The NaV1.8 blocker A-803467 (1 μmol/0.5 mL per GP, n = 7) or 5% DMSO/95% polyethylene glycol (0.5 mL per GP, n = 6, control) was injected into the anterior right GP and the inferior right GP. The effects of VNS on the sinus rate, ventricular rate, PR interval, atrial effective refractory period, and the cumulative window of vulnerability were significantly eliminated at 10, 35, and 90 min after A-803467 injection. In separate experiments (n = 8), A-803467 blunted the slowing of sinus rate with increasing stimulation voltage of the anterior right GP at 10 min after local injection. CONCLUSIONS Blockade of NaV1.8 channels suppresses the effects of VNS on cardiac conduction and atrial fibrillation inducibility, most likely by inhibiting the neural activity of the cardiac GP.
Collapse
Affiliation(s)
- Baozhen Qi
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Yong Wei
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Songwen Chen
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Hongli Li
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Juan Xu
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Yu Ding
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Xiaofeng Lu
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Liqun Zhao
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Feng Zhang
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Gang Chen
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| | - Jing Zhao
- Wolfson Institute for Biomedical Research, University College London, Wing3.1, Cruciform Building, Gower Street, London WC1E 6BT, UK
| | - Shaowen Liu
- Department of Cardiology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiaotong University, NO 100, Haining Road, Hongkou District, Shanghai 200080, China
| |
Collapse
|
31
|
Gautron L, Lee CE, Lee S, Elmquist JK. Melanocortin-4 receptor expression in different classes of spinal and vagal primary afferent neurons in the mouse. J Comp Neurol 2013; 520:3933-48. [PMID: 22592759 DOI: 10.1002/cne.23137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Melanocortin-4 receptor (MC4R) ligands are known to modulate nociception, but the site of action of MC4R signaling on nociception remains to be elucidated. The current study investigated MC4R expression in dorsal root ganglia (DRG) of the MC4R-GFP reporter mouse. Because MC4R is known to be expressed in vagal afferent neurons in the nodose ganglion (NG), we also systematically compared MC4R-expressing vagal and spinal afferent neurons. Abundant green fluorescent protein (GFP) immunoreactivity was found in about 45% of DRG neuronal profiles (at the mid-thoracic level), the majority being small-sized profiles. Immunohistochemistry combined with in situ hybridization confirmed that GFP was genuinely produced in MC4R-expressing neurons in the DRG. While a large number of GFP profiles in the DRG coexpressed Nav1.8 mRNA (84%) and bound isolectin B4 (72%), relatively few GFP profiles were positive for NF200 (16%) or CGRP (13%), suggesting preferential MC4R expression in C-fiber nonpeptidergic neurons. By contrast, GFP in the NG frequently colocalized with Nav1.8 mRNA (64%) and NF200 (29%), but only to a moderate extent with isolectin B4 (16%). Lastly, very few GFP profiles in the NG expressed CGRP (5%) or CART (4%). Together, our findings demonstrate variegated MC4R expression in different classes of vagal and spinal primary afferent neurons, and underscore the role of the melanocortin system in modulating nociceptive and nonnociceptive peripheral sensory modalities.
Collapse
Affiliation(s)
- Laurent Gautron
- Department of Internal Medicine, Division of Hypothalamic Research, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9077, USA.
| | | | | | | |
Collapse
|
32
|
Wong K, Bumpstead S, Van Der Weyden L, Reinholdt LG, Wilming LG, Adams DJ, Keane TM. Sequencing and characterization of the FVB/NJ mouse genome. Genome Biol 2012; 13:R72. [PMID: 22916792 PMCID: PMC3491372 DOI: 10.1186/gb-2012-13-8-r72] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 08/23/2012] [Indexed: 01/13/2023] Open
Abstract
Background The FVB/NJ mouse strain has its origins in a colony of outbred Swiss mice established in 1935 at the National Institutes of Health. Mice derived from this source were selectively bred for sensitivity to histamine diphosphate and the B strain of Friend leukemia virus. This led to the establishment of the FVB/N inbred strain, which was subsequently imported to the Jackson Laboratory and designated FVB/NJ. The FVB/NJ mouse has several distinct characteristics, such as large pronuclear morphology, vigorous reproductive performance, and consistently large litters that make it highly desirable for transgenic strain production and general purpose use. Results Using next-generation sequencing technology, we have sequenced the genome of FVB/NJ to approximately 50-fold coverage, and have generated a comprehensive catalog of single nucleotide polymorphisms, small insertion/deletion polymorphisms, and structural variants, relative to the reference C57BL/6J genome. We have examined a previously identified quantitative trait locus for atherosclerosis susceptibility on chromosome 10 and identify several previously unknown candidate causal variants. Conclusion The sequencing of the FVB/NJ genome and generation of this catalog has increased the number of known variant sites in FVB/NJ by a factor of four, and will help accelerate the identification of the precise molecular variants that are responsible for phenotypes observed in this widely used strain.
Collapse
|
33
|
Verkerk AO, Remme CA, Schumacher CA, Scicluna BP, Wolswinkel R, de Jonge B, Bezzina CR, Veldkamp MW. Functional Na
V
1.8 Channels in Intracardiac Neurons. Circ Res 2012; 111:333-43. [DOI: 10.1161/circresaha.112.274035] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
The
SCN10A
gene encodes the neuronal sodium channel isoform Na
V
1.8. Several recent genome-wide association studies have linked
SCN10A
to PR interval and QRS duration, strongly suggesting an as-yet unknown role for Na
V
1.8 in cardiac electrophysiology.
Objective:
To demonstrate the functional presence of
SCN10A
/Nav1.8 in intracardiac neurons of the mouse heart.
Methods and Results:
Immunohistochemistry on mouse tissue sections showed intense Na
V
1.8 labeling in dorsal root ganglia and intracardiac ganglia and only modest Na
V
1.8 expression within the myocardium. Immunocytochemistry further revealed substantial Na
V
1.8 staining in isolated neurons from murine intracardiac ganglia but no Na
V
1.8 expression in isolated ventricular myocytes. Patch-clamp studies demonstrated that the Na
V
1.8 blocker A-803467 (0.5–2 μmol/L) had no effect on either mean sodium current (I
Na
) density or I
Na
gating kinetics in isolated myocytes but significantly reduced I
Na
density in intracardiac neurons. Furthermore, A-803467 accelerated the slow component of current decay and shifted voltage dependence of inactivation toward more negative voltages, as expected for blockade of Na
V
1.8-based I
Na
. In line with these findings, A-803467 did not affect cardiomyocyte action potential upstroke velocity but markedly reduced action potential firing frequency in intracardiac neurons, confirming a functional role for Na
V
1.8 in cardiac neural activity.
Conclusions:
Our findings demonstrate the functional presence of
SCN10A
/Na
V
1.8 in intracardiac neurons, indicating a novel role for this neuronal sodium channel in regulation of cardiac electric activity.
Collapse
Affiliation(s)
- Arie O. Verkerk
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Carol Ann Remme
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Cees A. Schumacher
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Brendon P. Scicluna
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Rianne Wolswinkel
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Berend de Jonge
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Connie R. Bezzina
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke W. Veldkamp
- From the Department of Clinical and Experimental Cardiology (C.A.R., C.A.S., B.P.S., R.W., C.R.B., M.W.V.) and the Department of Anatomy, Embryology, and Physiology (A.O.V., B.d.J.), Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
van den Boogaard M, Wong LE, Tessadori F, Bakker ML, Dreizehnter LK, Wakker V, Bezzina CR, ‘t Hoen PA, Bakkers J, Barnett P, Christoffels VM. Genetic variation in T-box binding element functionally affects SCN5A/SCN10A enhancer. J Clin Invest 2012; 122:2519-30. [PMID: 22706305 PMCID: PMC3386824 DOI: 10.1172/jci62613] [Citation(s) in RCA: 146] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Accepted: 05/10/2012] [Indexed: 12/26/2022] Open
Abstract
The contraction pattern of the heart relies on the activation and conduction of the electrical impulse. Perturbations of cardiac conduction have been associated with congenital and acquired arrhythmias as well as cardiac arrest. The pattern of conduction depends on the regulation of heterogeneous gene expression by key transcription factors and transcriptional enhancers. Here, we assessed the genome-wide occupation of conduction system-regulating transcription factors TBX3, NKX2-5, and GATA4 and of enhancer-associated coactivator p300 in the mouse heart, uncovering cardiac enhancers throughout the genome. Many of the enhancers colocalized with ion channel genes repressed by TBX3, including the clustered sodium channel genes Scn5a, essential for cardiac function, and Scn10a. We identified 2 enhancers in the Scn5a/Scn10a locus, which were regulated by TBX3 and its family member and activator, TBX5, and are functionally conserved in humans. We also provided evidence that a SNP in the SCN10A enhancer associated with alterations in cardiac conduction patterns in humans disrupts TBX3/TBX5 binding and reduces the cardiac activity of the enhancer in vivo. Thus, the identification of key regulatory elements for cardiac conduction helps to explain how genetic variants in noncoding regulatory DNA sequences influence the regulation of cardiac conduction and the predisposition for cardiac arrhythmias.
Collapse
Affiliation(s)
- Malou van den Boogaard
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - L.Y. Elaine Wong
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Federico Tessadori
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Martijn L. Bakker
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Lisa K. Dreizehnter
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent Wakker
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Connie R. Bezzina
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter A.C. ‘t Hoen
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Jeroen Bakkers
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Phil Barnett
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent M. Christoffels
- Department of Anatomy, Embryology, and Physiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, The Netherlands.
Department of Experimental Cardiology, Heart Failure Research Center, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
Center for Human and Clinical Genetics and Leiden Genome Technology Center, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|