1
|
Delgado‐Bermúdez A, Yeste M, Bonet S, Pinart E. Physiological role of potassium channels in mammalian germ cell differentiation, maturation, and capacitation. Andrology 2025; 13:184-201. [PMID: 38436215 PMCID: PMC11815548 DOI: 10.1111/andr.13606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 03/05/2024]
Abstract
BACKGROUND Ion channels are essential for differentiation and maturation of germ cells, and even for fertilization in mammals. Different types of potassium channels have been identified, which are grouped into voltage-gated channels (Kv), ligand-gated channels (Kligand), inwardly rectifying channels (Kir), and tandem pore domain channels (K2P). MATERIAL-METHODS The present review includes recent findings on the role of potassium channels in sperm physiology of mammals. RESULTS-DISCUSSION While most studies conducted thus far have been focused on the physiological role of voltage- (Kv1, Kv3, and Kv7) and calcium-gated channels (SLO1 and SLO3) during sperm capacitation, especially in humans and rodents, little data about the types of potassium channels present in the plasma membrane of differentiating germ cells exist. In spite of this, recent evidence suggests that the content and regulation mechanisms of these channels vary throughout spermatogenesis. Potassium channels are also essential for the regulation of sperm cell volume during epididymal maturation and for preventing premature membrane hyperpolarization. It is important to highlight that the nature, biochemical properties, localization, and regulation mechanisms of potassium channels are species-specific. In effect, while SLO3 is the main potassium channel involved in the K+ current during sperm capacitation in rodents, different potassium channels are implicated in the K+ outflow and, thus, plasma membrane hyperpolarization during sperm capacitation in other mammalian species, such as humans and pigs. CONCLUSIONS Potassium conductance is essential for male fertility, not only during sperm capacitation but throughout the spermiogenesis and epididymal maturation.
Collapse
Affiliation(s)
- Ariadna Delgado‐Bermúdez
- Biotechnology of Animal and Human Reproduction (TechnoSperm)Institute of Food and Agricultural TechnologyUniversity of GironaGironaSpain
- Department of BiologyFaculty of SciencesUnit of Cell BiologyUniversity of GironaGironaSpain
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm)Institute of Food and Agricultural TechnologyUniversity of GironaGironaSpain
- Department of BiologyFaculty of SciencesUnit of Cell BiologyUniversity of GironaGironaSpain
- Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| | - Sergi Bonet
- Biotechnology of Animal and Human Reproduction (TechnoSperm)Institute of Food and Agricultural TechnologyUniversity of GironaGironaSpain
- Department of BiologyFaculty of SciencesUnit of Cell BiologyUniversity of GironaGironaSpain
| | - Elisabeth Pinart
- Biotechnology of Animal and Human Reproduction (TechnoSperm)Institute of Food and Agricultural TechnologyUniversity of GironaGironaSpain
- Department of BiologyFaculty of SciencesUnit of Cell BiologyUniversity of GironaGironaSpain
| |
Collapse
|
2
|
Pei S, Wang N, Mei Z, Zhangsun D, Craik DJ, McIntosh JM, Zhu X, Luo S. Conotoxins Targeting Voltage-Gated Sodium Ion Channels. Pharmacol Rev 2024; 76:828-845. [PMID: 38914468 PMCID: PMC11331937 DOI: 10.1124/pharmrev.123.000923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
Voltage-gated sodium (NaV) channels are intimately involved in the generation and transmission of action potentials, and dysfunction of these channels may contribute to nervous system diseases, such as epilepsy, neuropathic pain, psychosis, autism, and cardiac arrhythmia. Many venom peptides selectively act on NaV channels. These include conotoxins, which are neurotoxins secreted by cone snails for prey capture or self-defense but which are also valuable pharmacological tools for the identification and/or treatment of human diseases. Typically, conotoxins contain two or three disulfide bonds, and these internal crossbraces contribute to conotoxins having compact, well defined structures and high stability. Of the conotoxins containing three disulfide bonds, some selectively target mammalian NaV channels and can block, stimulate, or modulate these channels. Such conotoxins have great potential to serve as pharmacological tools for studying the functions and characteristics of NaV channels or as drug leads for neurologic diseases related to NaV channels. Accordingly, discovering or designing conotoxins targeting NaV channels with high potency and selectivity is important. The amino acid sequences, disulfide bond connectivity, and three-dimensional structures are key factors that affect the biological activity of conotoxins, and targeted synthetic modifications of conotoxins can greatly improve their activity and selectivity. This review examines NaV channel-targeted conotoxins, focusing on their structures, activities, and designed modifications, with a view toward expanding their applications. SIGNIFICANCE STATEMENT: NaV channels are crucial in various neurologic diseases. Some conotoxins selectively target NaV channels, causing either blockade or activation, thus enabling their use as pharmacological tools for studying the channels' characteristics and functions. Conotoxins also have promising potential to be developed as drug leads. The disulfide bonds in these peptides are important for stabilizing their structures, thus leading to enhanced specificity and potency. Together, conotoxins targeting NaV channels have both immediate research value and promising future application prospects.
Collapse
Affiliation(s)
- Shengrong Pei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Nan Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Zaoli Mei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Dongting Zhangsun
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - David J Craik
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - J Michael McIntosh
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Xiaopeng Zhu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| | - Sulan Luo
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning, China (S.P., N.W., Z.M., D.Z., X.Z., S.L.); Key Laboratory of Tropical Biological Resources of Ministry of Education, Hainan University, Haikou, China (D.Z., S.L.); Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, Brisbane, Queensland, Australia (D.J.C.); Departments of Biology and Psychiatry, University of Utah, Salt Lake City, Utah (J.M.M.); and George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah (J.M.M.)
| |
Collapse
|
3
|
Arratia LM, Bermudes-Contreras JD, Juarez-Monroy JA, Romero-Macías EA, Luna-Rojas JC, López-Hidalgo M, Vega AV, Zamorano-Carrillo A. Experimental and computational evidence that Calpain-10 binds to the carboxy terminus of Na V1.2 and Na V1.6. Sci Rep 2024; 14:6761. [PMID: 38514708 PMCID: PMC10957924 DOI: 10.1038/s41598-024-57117-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
Voltage-gated sodium channels (NaV) are pivotal proteins responsible for initiating and transmitting action potentials. Emerging evidence suggests that proteolytic cleavage of sodium channels by calpains is pivotal in diverse physiological scenarios, including ischemia, brain injury, and neuropathic pain associated with diabetes. Despite this significance, the precise mechanism by which calpains recognize sodium channels, especially given the multiple calpain isoforms expressed in neurons, remains elusive. In this work, we show the interaction of Calpain-10 with NaV's C-terminus through a yeast 2-hybrid assay screening of a mouse brain cDNA library and in vitro by GST-pulldown. Later, we also obtained a structural and dynamic hypothesis of this interaction by modeling, docking, and molecular dynamics simulation. These results indicate that Calpain-10 interacts differentially with the C-terminus of NaV1.2 and NaV1.6. Calpain-10 interacts with NaV1.2 through domains III and T in a stable manner. In contrast, its interaction with NaV1.6 involves domains II and III, which could promote proteolysis through the Cys-catalytic site and C2 motifs.
Collapse
Affiliation(s)
- Luis Manuel Arratia
- Carrera de Médico Cirujano, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. Mex, Mexico
- Laboratorio de Biofísica Computacional, Doctorado en Biotecnología, SEPI-ENMH Instituto Politécnico Nacional, Av. Guillermo Massieu Helguera 239, Fracc. La Escalera, Ticomán, Gustavo A. Madero, 07320, Mexico City, Mexico
| | - Juan David Bermudes-Contreras
- Laboratorio de Biofísica Computacional, Doctorado en Biotecnología, SEPI-ENMH Instituto Politécnico Nacional, Av. Guillermo Massieu Helguera 239, Fracc. La Escalera, Ticomán, Gustavo A. Madero, 07320, Mexico City, Mexico
| | - Jorge Armando Juarez-Monroy
- Laboratorio de Biofísica Computacional, Doctorado en Biotecnología, SEPI-ENMH Instituto Politécnico Nacional, Av. Guillermo Massieu Helguera 239, Fracc. La Escalera, Ticomán, Gustavo A. Madero, 07320, Mexico City, Mexico
| | - Erik Alan Romero-Macías
- Carrera de Médico Cirujano, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. Mex, Mexico
- Doctorado en Ciencias Biomédicas, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla Edo, Mexico City, Mexico
| | - Julio Cesar Luna-Rojas
- Carrera de Médico Cirujano, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. Mex, Mexico
- Maestría en Neurobiología, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla Edo, Mexico City, Mexico
| | - Marisol López-Hidalgo
- Laboratorio de Biofísica Computacional, Doctorado en Biotecnología, SEPI-ENMH Instituto Politécnico Nacional, Av. Guillermo Massieu Helguera 239, Fracc. La Escalera, Ticomán, Gustavo A. Madero, 07320, Mexico City, Mexico
| | - Ana Victoria Vega
- Carrera de Médico Cirujano, FES Iztacala, UNAM, Av. de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Edo. Mex, Mexico.
| | - Absalom Zamorano-Carrillo
- Laboratorio de Biofísica Computacional, Doctorado en Biotecnología, SEPI-ENMH Instituto Politécnico Nacional, Av. Guillermo Massieu Helguera 239, Fracc. La Escalera, Ticomán, Gustavo A. Madero, 07320, Mexico City, Mexico.
| |
Collapse
|
4
|
Wright K, Jiang H, Xia W, Murphy MB, Boronina TN, Nwafor JN, Kim H, Iheanacho AM, Azurmendi PA, Cole RN, Cole PA, Gabelli SB. The C-Terminal of Na V1.7 Is Ubiquitinated by NEDD4L. ACS BIO & MED CHEM AU 2023; 3:516-527. [PMID: 38144259 PMCID: PMC10739247 DOI: 10.1021/acsbiomedchemau.3c00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/10/2023] [Accepted: 09/25/2023] [Indexed: 12/26/2023]
Abstract
NaV1.7, the neuronal voltage-gated sodium channel isoform, plays an important role in the human body's ability to feel pain. Mutations within NaV1.7 have been linked to pain-related syndromes, such as insensitivity to pain. To date, the regulation and internalization mechanisms of the NaV1.7 channel are not well known at a biochemical level. In this study, we perform biochemical and biophysical analyses that establish that the HECT-type E3 ligase, NEDD4L, ubiquitinates the cytoplasmic C-terminal (CT) region of NaV1.7. Through in vitro ubiquitination and mass spectrometry experiments, we identify, for the first time, the lysine residues of NaV1.7 within the CT region that get ubiquitinated. Furthermore, binding studies with an NEDD4L E3 ligase modulator (ubiquitin variant) highlight the dynamic partnership between NEDD4L and NaV1.7. These investigations provide a framework for understanding how NEDD4L-dependent regulation of the channel can influence the NaV1.7 function.
Collapse
Affiliation(s)
- Katharine
M. Wright
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Hanjie Jiang
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Wendy Xia
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | | | - Tatiana N. Boronina
- Mass
Spectrometry
and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Justin N. Nwafor
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - HyoJeon Kim
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Akunna M. Iheanacho
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Physiology, The Johns Hopkins School
of Medicine, Baltimore, Maryland 21205, United States
| | - P. Aitana Azurmendi
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
| | - Robert N. Cole
- Mass
Spectrometry
and Proteomics Facility, Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Philip A. Cole
- Division
of Genetics, Department of Medicine, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sandra B. Gabelli
- Department
of Biophysics and Biophysical Chemistry, The Johns Hopkins School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Medicine, The Johns Hopkins University
School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Oncology, The Johns Hopkins University
School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
5
|
Azizan EAB, Drake WM, Brown MJ. Primary aldosteronism: molecular medicine meets public health. Nat Rev Nephrol 2023; 19:788-806. [PMID: 37612380 PMCID: PMC7615304 DOI: 10.1038/s41581-023-00753-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
Primary aldosteronism is the most common single cause of hypertension and is potentially curable when only one adrenal gland is the culprit. The importance of primary aldosteronism to public health derives from its high prevalence but huge under-diagnosis (estimated to be <1% of all affected individuals), despite the consequences of poor blood pressure control by conventional therapy and enhanced cardiovascular risk. This state of affairs is attributable to the fact that the tools used for diagnosis or treatment are still those that originated in the 1970-1990s. Conversely, molecular discoveries have transformed our understanding of adrenal physiology and pathology. Many molecules and processes associated with constant adrenocortical renewal and interzonal metamorphosis also feature in aldosterone-producing adenomas and aldosterone-producing micronodules. The adrenal gland has one of the most significant rates of non-silent somatic mutations, with frequent selection of those driving autonomous aldosterone production, and distinct clinical presentations and outcomes for most genotypes. The disappearance of aldosterone synthesis and cells from most of the adult human zona glomerulosa is the likely driver of the mutational success that causes aldosterone-producing adenomas, but insights into the pathways that lead to constitutive aldosterone production and cell survival may open up opportunities for novel therapies.
Collapse
Affiliation(s)
- Elena A B Azizan
- Department of Medicine, Faculty of Medicine, The National University of Malaysia (UKM), Kuala Lumpur, Malaysia
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - William M Drake
- St Bartholomew's Hospital, Barts Health NHS Trust, London, United Kingdom
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Morris J Brown
- Endocrine Hypertension, Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom.
- NIHR Barts Biomedical Research Centre, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
6
|
Shemarova I. The Dysfunction of Ca 2+ Channels in Hereditary and Chronic Human Heart Diseases and Experimental Animal Models. Int J Mol Sci 2023; 24:15682. [PMID: 37958665 PMCID: PMC10650855 DOI: 10.3390/ijms242115682] [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: 09/11/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Chronic heart diseases, such as coronary heart disease, heart failure, secondary arterial hypertension, and dilated and hypertrophic cardiomyopathies, are widespread and have a fairly high incidence of mortality and disability. Most of these diseases are characterized by cardiac arrhythmias, conduction, and contractility disorders. Additionally, interruption of the electrical activity of the heart, the appearance of extensive ectopic foci, and heart failure are all symptoms of a number of severe hereditary diseases. The molecular mechanisms leading to the development of heart diseases are associated with impaired permeability and excitability of cell membranes and are mainly caused by the dysfunction of cardiac Ca2+ channels. Over the past 50 years, more than 100 varieties of ion channels have been found in the cardiovascular cells. The relationship between the activity of these channels and cardiac pathology, as well as the general cellular biological function, has been intensively studied on several cell types and experimental animal models in vivo and in situ. In this review, I discuss the origin of genetic Ca2+ channelopathies of L- and T-type voltage-gated calcium channels in humans and the role of the non-genetic dysfunctions of Ca2+ channels of various types: L-, R-, and T-type voltage-gated calcium channels, RyR2, including Ca2+ permeable nonselective cation hyperpolarization-activated cyclic nucleotide-gated (HCN), and transient receptor potential (TRP) channels, in the development of cardiac pathology in humans, as well as various aspects of promising experimental studies of the dysfunctions of these channels performed on animal models or in vitro.
Collapse
Affiliation(s)
- Irina Shemarova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| |
Collapse
|
7
|
Castro EV, Shepherd JW, Guggenheim RS, Sengvoravong M, Hall BC, Chappell MK, Hearn JA, Caraccio ON, Bissman C, Lantow S, Buehner D, Costlow HR, Prather DM, Zonza AM, Witt M, Zahratka JA. ChanFAD: A Functional Annotation Database for Ion Channels. FRONTIERS IN BIOINFORMATICS 2022; 2:835805. [PMID: 36304304 PMCID: PMC9580856 DOI: 10.3389/fbinf.2022.835805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
Ion channels are integral membrane protein complexes critical for regulation of membrane potential, cell volume, and other signaling events. As complex molecular assemblies with many interacting partners, ion channels have multiple structural and functional domains. While channel sequence and functional data are readily available across multiple online resources, there is an unmet need for functional annotation directly relating primary sequence information, 2D interactions, and three-dimensional protein structure. To this end, we present ChanFAD (Channel Functional Annotation Database), to provide the research community with a centralized resource for ion channel structure and functional data. ChanFAD provides functional annotation of PDB structures built on the National Center for Biotechnology Information’s iCn3D structure viewing tool while providing additional information such as primary sequence, organism, and relevant links to other databases. Here we provide a brief tour of ChanFAD functionality while showing example use cases involving drug-channel interactions and structural changes based on mutation. ChanFAD is freely available and can be accessed at https://www.chanfad.org/.
Collapse
Affiliation(s)
- Elizabeth V. Castro
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Psychology, Baldwin Wallace University, Berea, OH, United States
| | - John W. Shepherd
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - Ryan S. Guggenheim
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Psychology, Baldwin Wallace University, Berea, OH, United States
| | | | - Bailey C. Hall
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - McKenzie K. Chappell
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Biology and Geology, Baldwin Wallace University, Berea, OH, United States
| | - Jessica A. Hearn
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Biology and Geology, Baldwin Wallace University, Berea, OH, United States
| | - Olivia N. Caraccio
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - Cora Bissman
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Biology and Geology, Baldwin Wallace University, Berea, OH, United States
| | - Sydney Lantow
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - Damian Buehner
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - Harry R. Costlow
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - David M. Prather
- Department of Chemistry, Baldwin Wallace University, Berea, OH, United States
| | - Abigail M. Zonza
- Department of Biology and Geology, Baldwin Wallace University, Berea, OH, United States
| | - Mallory Witt
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
| | - Jeffrey A. Zahratka
- Department of Neuroscience, Baldwin Wallace University, Berea, OH, United States
- Department of Biology and Geology, Baldwin Wallace University, Berea, OH, United States
- *Correspondence: Jeffrey A. Zahratka,
| |
Collapse
|
8
|
Ren P, Wang J, Li N, Li G, Ma H, Zhao Y, Li Y. Sigma-1 Receptors in Depression: Mechanism and Therapeutic Development. Front Pharmacol 2022; 13:925879. [PMID: 35784746 PMCID: PMC9243434 DOI: 10.3389/fphar.2022.925879] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/26/2022] [Indexed: 12/26/2022] Open
Abstract
Depression is the most common type of neuropsychiatric illness and has increasingly become a major cause of disability. Unfortunately, the recent global pandemic of COVID-19 has dramatically increased the incidence of depression and has significantly increased the burden of mental health care worldwide. Since full remission of the clinical symptoms of depression has not been achieved with current treatments, there is a constant need to discover new compounds that meet the major clinical needs. Recently, the roles of sigma receptors, especially the sigma-1 receptor subtype, have attracted increasing attention as potential new targets and target-specific drugs due to their translocation property that produces a broad spectrum of biological functions. Even clinical first-line antidepressants with or without affinity for sigma-1 receptors have different pharmacological profiles. Thus, the regulatory role of sigma-1 receptors might be useful in treating these central nervous system (CNS) diseases. In addition, long-term mental stress disrupts the homeostasis in the CNS. In this review, we discuss the topical literature concerning sigma-1 receptor antidepressant mechanism of action in the regulation of intracellular proteostasis, calcium homeostasis and especially the dynamic Excitatory/Inhibitory (E/I) balance in the brain. Furthermore, based on these discoveries, we discuss sigma-1 receptor ligands with respect to their promise as targets for fast-onset action drugs in treating depression.
Collapse
Affiliation(s)
- Peng Ren
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jingya Wang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Nanxi Li
- Department of Pharmaceutical Sciences, Beijng Institute of Radiation Medicine, Beijing, China
| | - Guangxiang Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yongqi Zhao
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| | - Yunfeng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Hui Ma, ; Yongqi Zhao, ; Yunfeng Li,
| |
Collapse
|
9
|
Wang M, Tu X. The Genetics and Epigenetics of Ventricular Arrhythmias in Patients Without Structural Heart Disease. Front Cardiovasc Med 2022; 9:891399. [PMID: 35783865 PMCID: PMC9240357 DOI: 10.3389/fcvm.2022.891399] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/25/2022] [Indexed: 12/19/2022] Open
Abstract
Ventricular arrhythmia without structural heart disease is an arrhythmic disorder that occurs in structurally normal heart and no transient or reversible arrhythmia factors, such as electrolyte disorders and myocardial ischemia. Ventricular arrhythmias without structural heart disease can be induced by multiple factors, including genetics and environment, which involve different genetic and epigenetic regulation. Familial genetic analysis reveals that cardiac ion-channel disorder and dysfunctional calcium handling are two major causes of this type of heart disease. Genome-wide association studies have identified some genetic susceptibility loci associated with ventricular tachycardia and ventricular fibrillation, yet relatively few loci associated with no structural heart disease. The effects of epigenetics on the ventricular arrhythmias susceptibility genes, involving non-coding RNAs, DNA methylation and other regulatory mechanisms, are gradually being revealed. This article aims to review the knowledge of ventricular arrhythmia without structural heart disease in genetics, and summarizes the current state of epigenetic regulation.
Collapse
|
10
|
Rare CACNA1H and RELN variants interact through mTORC1 pathway in oligogenic autism spectrum disorder. Transl Psychiatry 2022; 12:234. [PMID: 35668055 PMCID: PMC9170683 DOI: 10.1038/s41398-022-01997-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/19/2022] [Accepted: 05/25/2022] [Indexed: 11/09/2022] Open
Abstract
Oligogenic inheritance of autism spectrum disorder (ASD) has been supported by several studies. However, little is known about how the risk variants interact and converge on causative neurobiological pathways. We identified in an ASD proband deleterious compound heterozygous missense variants in the Reelin (RELN) gene, and a de novo splicing variant in the Cav3.2 calcium channel (CACNA1H) gene. Here, by using iPSC-derived neural progenitor cells (NPCs) and a heterologous expression system, we show that the variant in Cav3.2 leads to increased calcium influx into cells, which overactivates mTORC1 pathway and, consequently, further exacerbates the impairment of Reelin signaling. Also, we show that Cav3.2/mTORC1 overactivation induces proliferation of NPCs and that both mutant Cav3.2 and Reelin cause abnormal migration of these cells. Finally, analysis of the sequencing data from two ASD cohorts-a Brazilian cohort of 861 samples, 291 with ASD; the MSSNG cohort of 11,181 samples, 5,102 with ASD-revealed that the co-occurrence of risk variants in both alleles of Reelin pathway genes and in one allele of calcium channel genes confer significant liability for ASD. Our results support the notion that genes with co-occurring deleterious variants tend to have interconnected pathways underlying oligogenic forms of ASD.
Collapse
|
11
|
Contreras GF, Saavedra J, Navarro-Quezada N, Mellado G, Gonzalez C, Neely A. Direct inhibition of Ca V2.3 by Gem is dynamin dependent and does not require a direct alfa/beta interaction. Biochem Biophys Res Commun 2022; 586:107-113. [PMID: 34837834 DOI: 10.1016/j.bbrc.2021.11.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/21/2021] [Accepted: 11/14/2021] [Indexed: 11/17/2022]
Abstract
The Rad, Rem, Rem2, and Gem/Kir (RGK) sub-family of small GTP-binding proteins are crucial in regulating high voltage-activated (HVA) calcium channels. RGK proteins inhibit calcium current by either promoting endocytosis or reducing channel activity. They all can associate directly with Ca2+ channel β subunit (CaVβ), and the binding between CaVα1/CaVβ appears essential for the endocytic promotion of CaV1.X, CaV2.1, and CaV2.2 channels. In this study, we investigated the inhibition of CaV2.3 channels by RGK proteins in the absence of CaVβ. To this end, Xenopus laevis oocytes expressing CaV2.3 channels devoid of auxiliary subunit were injected with purified Gem and Rem and found that only Gem had an effect. Ca currents and charge movements were reduced by injection of Gem, pointing to a reduction in the number of channels in the plasma membrane. Since this reduction was ablated by co-expression of the dominant-negative mutant of dynamin K44A, enhanced endocytosis appears to mediate this reduction in the number of channels. Thus, Gem inhibition of CaV2.3 channels would be the only example of a CaVβ independent promotion of dynamin-dependent endocytosis.
Collapse
Affiliation(s)
- Gustavo F Contreras
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile; Centro Interdisciplinario de Neurociencias Valparaíso, Chile
| | - Jonathan Saavedra
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile; Centro Interdisciplinario de Neurociencias Valparaíso, Chile; Doctorado en Ciencias Mención Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nieves Navarro-Quezada
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile; Centro Interdisciplinario de Neurociencias Valparaíso, Chile
| | - Guido Mellado
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile; Centro Interdisciplinario de Neurociencias Valparaíso, Chile; Doctorado en Ciencias Mención Biofisica y Biología Computacional, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlos Gonzalez
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile; Centro Interdisciplinario de Neurociencias Valparaíso, Chile; Cardiovascular Research, Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, USA
| | - Alan Neely
- Instituto de Neurociencia, Facultad de Ciencias, Universidad de Valparaiso, Chile
| |
Collapse
|
12
|
Holm A, Hansen SN, Klitgaard H, Kauppinen S. Clinical advances of RNA therapeutics for treatment of neurological and neuromuscular diseases. RNA Biol 2021; 19:594-608. [PMID: 35482908 PMCID: PMC9067473 DOI: 10.1080/15476286.2022.2066334] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/11/2022] [Indexed: 11/17/2022] Open
Abstract
RNA therapeutics comprise a diverse group of oligonucleotide-based drugs such as antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), and short hairpin RNAs (shRNAs) that can be designed to selectively interact with drug targets currently undruggable with small molecule-based drugs or monoclonal antibodies. Furthermore, RNA-based therapeutics have the potential to modulate entire disease pathways, and thereby represent a new modality with unprecedented potential for generating disease-modifying drugs for a wide variety of human diseases, including central nervous system (CNS) disorders. Here, we describe different strategies for delivering RNA drugs to the CNS and review recent advances in clinical development of ASO drugs and siRNA-based therapeutics for the treatment of neurological diseases and neuromuscular disorders.Abbreviations 2'-MOE: 2'-O-(2-methoxyethyl); 2'-O-Me: 2'-O-methyl; 2'-F: 2'-fluoro; AD: Alzheimer's disease; ALS: Amyotrophic lateral sclerosis; ALSFRS-R: Revised Amyotrophic Lateral Sclerosis Functional Rating Scale; ARC: Antibody siRNA Conjugate; AS: Angelman Syndrome; ASGRP: Asialoglycoprotein receptor; ASO: Antisense oligonucleotide; AxD: Alexander Disease; BBB: Blood brain barrier; Bp: Basepair; CNM: Centronuclear myopathies; CNS: Central Nervous System; CPP: Cell-penetrating Peptide; CSF: Cerebrospinal fluid; DMD: Duchenne muscular dystrophy; DNA: Deoxyribonucleic acid; FAP: Familial amyloid polyneuropathy; FALS: Familial amyotrophic lateral sclerosis; FDA: The United States Food and Drug Administration; GalNAc: N-acetylgalactosamine; GoF: Gain of function; hATTR: Hereditary transthyretin amyloidosis; HD: Huntington's disease; HRQOL: health-related quality of life; ICV: Intracerebroventricular; IT: Intrathecal; LNA: Locked nucleic acid; LoF: Loss of function; mRNA: Messenger RNA; MS: Multiple Sclerosis; MSA: Multiple System Atrophy; NBE: New Biological Entity; NCE: New Chemical Entity; NHP: Nonhuman primate; nt: Nucleotide; PD: Parkinson's disease; PNP: Polyneuropathy; PNS: Peripheral nervous system; PS: Phosphorothioate; RISC: RNA-Induced Silencing Complex; RNA: Ribonucleic acid; RNAi: RNA interference; s.c.: Subcutaneous; siRNA: Small interfering RNA; SMA: Spinal muscular atrophy; SMN: Survival motor neuron; TTR: Transthyretin.
Collapse
Affiliation(s)
- Anja Holm
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, A.C. Meyers Vænge 15, 2450Copenhagen, Denmark
| | - Stine N. Hansen
- Neumirna Therapeutics, A.C. Meyers Vænge 15, 2450Copenhagen, Denmark
| | - Henrik Klitgaard
- Neumirna Therapeutics, A.C. Meyers Vænge 15, 2450Copenhagen, Denmark
| | - Sakari Kauppinen
- Center for RNA Medicine, Department of Clinical Medicine, Aalborg University, A.C. Meyers Vænge 15, 2450Copenhagen, Denmark
| |
Collapse
|
13
|
Speigel IA, Hemmings HC. Selective inhibition of gamma aminobutyric acid release from mouse hippocampal interneurone subtypes by the volatile anaesthetic isoflurane. Br J Anaesth 2021; 127:587-599. [PMID: 34384592 DOI: 10.1016/j.bja.2021.06.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The cellular and molecular mechanisms by which general anaesthesia occurs is poorly understood. Hippocampal interneurone subpopulations, which are critical regulators of cognitive function, have diverse neurophysiological and synaptic properties, but their responses to anaesthetics are unclear. METHODS We used live-cell imaging of fluorescent biosensors expressed in mouse hippocampal neurones to delineate interneurone subtype-specific effects of isoflurane on synaptic vesicle exocytosis. The role of voltage-gated sodium channel (Nav) subtype expression in determining isoflurane sensitivity was probed by overexpression or knockdown of specific Nav subtypes in identified interneurones. RESULTS Clinically relevant concentrations of isoflurane differentially inhibited synaptic vesicle exocytosis: to 83.1% (11.7%) of control in parvalbumin-expressing interneurones, and to 58.6% (13.3%) and 64.5% (8.5%) of control in somatostatin-expressing interneurones and glutamatergic neurones, respectively. The relative expression of Nav1.1 (associated with lower sensitivity) and Nav1.6 (associated with higher sensitivity) determined the sensitivity of exocytosis to isoflurane. CONCLUSIONS Isoflurane inhibits synaptic vesicle exocytosis from hippocampal glutamatergic neurones and GABAergic interneurones in a cell-type-specific manner depending on their expression of voltage-gated sodium channel subtypes.
Collapse
Affiliation(s)
- Iris A Speigel
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA.
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
14
|
Younis W, Schini-Kerth VB, Junior AG, Nocchi SR, Silva DB, Roberts RE. Endothelium-independent vasorelaxant effect of Asphodelus tenuifolius Cav. via inhibition of myosin light chain kinase activity in the porcine coronary artery. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113693. [PMID: 33326818 DOI: 10.1016/j.jep.2020.113693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 04/11/2020] [Accepted: 12/09/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Asphodelus tenuifolius Cav. (Asphodelaceae), a wild, terrestrial, annual stemless herb, is widely used in traditional medicine for the treatment of hypertension, diabetes, atherosclerosis and circulatory problems. A previous research study from our laboratory revealed that A. tenuifolius has beneficial effects in reducing blood pressure and improves aortic endothelial dysfunction in chronically glucose fed rats. Despite the fact that A. tenuifolius reduces blood pressure and improves endothelial function in vivo, there are no detailed studies about its possible mechanism of action. AIM OF THE STUDY This study was designed to provide pharmacological basis and mechanism of action for the traditional use of A. tenuifolius in hypertension and circulatory problems. We explored the vasorelaxant effect of A. tenuifolius and its underlying vasorelaxation mechanism in porcine coronary artery rings. MATERIALS AND METHODS Aqueous methanolic crude extract of A. tenuifolius was prepared by maceration process and then activity guided fractionation was carried out by using different polarity based solvents. Phytochemical studies were carried out using LC-DAD-MS. Segments of porcine distal coronary artery were set up in a wire myograph for isometric force measurements. Extract/fractions of A. tenuifolius seeds were tested for vasodilator activity by measurement of changes in tone after pre-contraction with the thromboxane mimetic U46619 in the presence or absence of inhibitors of intracellular signaling cascades. RESULTS Crude extract/fractions of A. tenuifolius produced dose dependent endothelium independent vasorelaxant response in coronary rings, whereas, the butanol fraction of A. tenuifolius (BS-AT) produced the largest relaxation response with 100% relaxation at 1 mg/ml, therefore the mechanism of relaxation of this fraction was determined. The relaxation to BS-AT was unaffected by removal of the endothelium, pre-contraction with KCl, or the presence of the non-selective potassium channel blocker tetraethylammonium, indicating that the relaxation was endothelium-independent, and does not involve activation of potassium channels. BS-AT (1 mg/ml) inhibited the contractile response to calcium,the L-type calcium channel activator BAY K8664,and ionomycin, indicating that it inhibits calcium-induced contractions. The relaxation response to BS-AT was attenuated in the absence of extracellular calcium. However, relaxations to BS-AT were also reduced after deletion of calcium from intracellular stores with cyclopiazonic acid. Incubation with 1 mg/ml BS-AT also inhibited phosphorylation of myosin light chains in homogenates of coronary artery. CONCLUSION The butanol extract of Asphodelus tenuifolius produces a large endothelium-independent relaxation of the porcine coronary artery through inhibition of calcium-induced contractions. The effect appears to be downstream of calcium influx, possibly through inhibition of myosin light chain kinase. This study supports previous studies demonstrating that A. tenuifolius reduces blood pressure. Future studies will aim to determine the active compounds underlying this response.
Collapse
Affiliation(s)
- Waqas Younis
- Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, 40100, Pakistan; Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom
| | - V B Schini-Kerth
- UMR 1260 INSERM Nanomédecine Régénérative Faculté de Pharmacie, Université de Strasbourg, Illkirch, France
| | - Arquimedes Gasparotto Junior
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Federal University of Grande Dourados (UFGD), P.O. Box 533, 79.804-970, Dourados, MS, Brazil
| | - Samara Requena Nocchi
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Denise Brentan Silva
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Richard E Roberts
- Pharmacology Research Group, School of Life Sciences, University of Nottingham Medical School, Queen's Medical Centre, Nottingham, NG7 2UH, United Kingdom.
| |
Collapse
|
15
|
Mantegazza M, Cestèle S, Catterall WA. Sodium channelopathies of skeletal muscle and brain. Physiol Rev 2021; 101:1633-1689. [PMID: 33769100 DOI: 10.1152/physrev.00025.2020] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated sodium channels initiate action potentials in nerve, skeletal muscle, and other electrically excitable cells. Mutations in them cause a wide range of diseases. These channelopathy mutations affect every aspect of sodium channel function, including voltage sensing, voltage-dependent activation, ion conductance, fast and slow inactivation, and both biosynthesis and assembly. Mutations that cause different forms of periodic paralysis in skeletal muscle were discovered first and have provided a template for understanding structure, function, and pathophysiology at the molecular level. More recent work has revealed multiple sodium channelopathies in the brain. Here we review the well-characterized genetics and pathophysiology of the periodic paralyses of skeletal muscle and then use this information as a foundation for advancing our understanding of mutations in the structurally homologous α-subunits of brain sodium channels that cause epilepsy, migraine, autism, and related comorbidities. We include studies based on molecular and structural biology, cell biology and physiology, pharmacology, and mouse genetics. Our review reveals unexpected connections among these different types of sodium channelopathies.
Collapse
Affiliation(s)
- Massimo Mantegazza
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France.,INSERM, Valbonne-Sophia Antipolis, France
| | - Sandrine Cestèle
- Université Cote d'Azur, Valbonne-Sophia Antipolis, France.,CNRS UMR7275, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne-Sophia Antipolis, France
| | | |
Collapse
|
16
|
Zhang XD, Thai PN, Lieu DK, Chiamvimonvat N. Cardiac small-conductance calcium-activated potassium channels in health and disease. Pflugers Arch 2021; 473:477-489. [PMID: 33624131 PMCID: PMC7940285 DOI: 10.1007/s00424-021-02535-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
Small-conductance Ca2+-activated K+ (SK, KCa2) channels are encoded by KCNN genes, including KCNN1, 2, and 3. The channels play critical roles in the regulation of cardiac excitability and are gated solely by beat-to-beat changes in intracellular Ca2+. The family of SK channels consists of three members with differential sensitivity to apamin. All three isoforms are expressed in human hearts. Studies over the past two decades have provided evidence to substantiate the pivotal roles of SK channels, not only in healthy heart but also with diseases including atrial fibrillation (AF), ventricular arrhythmia, and heart failure (HF). SK channels are prominently expressed in atrial myocytes and pacemaking cells, compared to ventricular cells. However, the channels are significantly upregulated in ventricular myocytes in HF and pulmonary veins in AF models. Interests in cardiac SK channels are further fueled by recent studies suggesting the possible roles of SK channels in human AF. Therefore, SK channel may represent a novel therapeutic target for atrial arrhythmias. Furthermore, SK channel function is significantly altered by human calmodulin (CaM) mutations, linked to life-threatening arrhythmia syndromes. The current review will summarize recent progress in our understanding of cardiac SK channels and the roles of SK channels in the heart in health and disease.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA
| | - Deborah K Lieu
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, One Shields Avenue, GBSF 6315, Davis, CA, 95616, USA.
- Department of Veterans Affairs, Northern California Health Care System, 10535 Hospital Way, Mather, CA, 95655, USA.
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA.
| |
Collapse
|
17
|
Banerji R, Huynh C, Figueroa F, Dinday MT, Baraban SC, Patel M. Enhancing glucose metabolism via gluconeogenesis is therapeutic in a zebrafish model of Dravet syndrome. Brain Commun 2021; 3:fcab004. [PMID: 33842883 PMCID: PMC8023476 DOI: 10.1093/braincomms/fcab004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/14/2020] [Accepted: 12/16/2020] [Indexed: 01/18/2023] Open
Abstract
Energy-producing pathways are novel therapeutic targets for the treatment of neurodevelopmental disorders. Here, we focussed on correcting metabolic defects in a catastrophic paediatric epilepsy, Dravet syndrome which is caused by mutations in sodium channel NaV1.1 gene, SCN1A. We utilized a translatable zebrafish model of Dravet syndrome (scn1lab) which exhibits key characteristics of patients with Dravet syndrome and shows metabolic deficits accompanied by down-regulation of gluconeogenesis genes, pck1 and pck2. Using a metabolism-based small library screen, we identified compounds that increased gluconeogenesis via up-regulation of pck1 gene expression in scn1lab larvae. Treatment with PK11195, a pck1 activator and a translocator protein ligand, normalized dys-regulated glucose levels, metabolic deficits, translocator protein expression and significantly decreased electrographic seizures in mutant larvae. Inhibition of pck1 in wild-type larvae mimicked metabolic and behaviour defects observed in scn1lab mutants. Together, this suggests that correcting dys-regulated metabolic pathways can be therapeutic in neurodevelopmental disorders such as Dravet syndrome arising from ion channel dysfunction.
Collapse
Affiliation(s)
- Rajeswari Banerji
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| | - Christopher Huynh
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| | - Francisco Figueroa
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Matthew T Dinday
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Scott C Baraban
- Department of Neurological Surgery, Epilepsy Research Laboratory, University of California, San Francisco, CA 94143, USA
| | - Manisha Patel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, CA 80045, USA
| |
Collapse
|
18
|
Murakami M, Xu F, Ohba T, Kobayashi T, Inoue Y, Murakami AM, Miyoshi I, Ono K, Tohse N. Requirement of the Ca 2+ channel β 2 subunit for sympathetic PKA phosphorylation. J Pharmacol Sci 2021; 145:253-261. [PMID: 33602505 DOI: 10.1016/j.jphs.2020.12.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 12/10/2020] [Accepted: 12/28/2020] [Indexed: 11/29/2022] Open
Abstract
Facilitation of cardiac function in response to signals from the sympathetic nervous system is initiated by the phosphorylation of L-type voltage-dependent Ca2+ channels (VDCCs) by protein kinase A (PKA), which in turn is activated by β-adrenoceptors. Among the five subunits (α1, β, α2/δ, and γ) of VDCCs, the α1 subunit and the family of β subunits are substrates for PKA-catalyzed phosphorylation; however, the subunit responsible for β-adrenergic augmentation of Ca2+ channel function has yet to be specifically identified. Here we show that the VDCC β2 subunit is required for PKA phosphorylation upon sympathetic acceleration. In mice with β2 subunit-null mutations, cardiac muscle contraction in response to isoproterenol was reduced and there was no significant increase in Ca2+ channel currents upon PKA-dependent phosphorylation. These findings indicate that within the sympathetic nervous system the β2 subunit of VDCCs is required for physiological PKA-dependent channel phosphorylation.
Collapse
Affiliation(s)
- Manabu Murakami
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Aomori, 036-8562, Japan.
| | - Feng Xu
- Department of Pharmacology, Akita University, Graduate School of Medicine, Akita, 010-8543, Japan
| | - Takayoshi Ohba
- Department of Cell Physiology, Akita University, Graduate School of Medicine, Akita, 010-8543, Japan
| | - Takeshi Kobayashi
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| | - Yoshiro Inoue
- Division of Tumor Animals Institute for Experimental Animals, Institute of Development, Aging and Cancer, Tohoku University School of Medicine, Sendai, Japan
| | - Agnieszka M Murakami
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Aomori, 036-8562, Japan
| | - Ichiro Miyoshi
- Animal Care Center, Tohoku University School of Medicine, Sendai, Japan
| | - Kyoichi Ono
- Department of Cell Physiology, Akita University, Graduate School of Medicine, Akita, 010-8543, Japan
| | - Noritsugu Tohse
- Department of Cellular Physiology and Signal Transduction, Sapporo Medical University School of Medicine, Sapporo, 060-8556, Japan
| |
Collapse
|
19
|
Pauzi FA, Azizan EA. Functional Characteristic and Significance of Aldosterone-Producing Cell Clusters in Primary Aldosteronism and Age-Related Hypertension. Front Endocrinol (Lausanne) 2021; 12:631848. [PMID: 33763031 PMCID: PMC7982842 DOI: 10.3389/fendo.2021.631848] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/01/2021] [Indexed: 12/02/2022] Open
Abstract
Primary aldosteronism (PA) is one of the most frequent curable forms of secondary hypertension. It can be caused by the overproduction of aldosterone in one or both adrenal glands. The most common subtypes of PA are unilateral aldosterone over-production due to aldosterone-producing adenomas (APA) or bilateral aldosterone over-production due to bilateral hyperaldosteronism (BHA). Utilizing the immunohistochemical (IHC) detection of aldosterone synthase (CYP11B2) has allowed the identification of aldosterone-producing cell clusters (APCCs) with unique focal localization positive for CYP11B2 expression in the subcapsular portion of the human adult adrenal cortex. The presence of CYP11B2 supports that synthesis of aldosterone can occur in these cell clusters and therefore might contribute to hyperaldosteronism. However, the significance of the steroidogenic properties of APCCs especially in regards to PA remains unclear. Herein, we review the available evidence on the presence of APCCs in normal adrenals and adrenal tissues adjacent to APAs, their aldosterone-stimulating somatic gene mutations, and their accumulation during the ageing process; raising the possibility that APCCs may play a role in the development of PA and age-related hypertension.
Collapse
|
20
|
Wadsworth PA, Singh AK, Nguyen N, Dvorak NM, Tapia CM, Russell WK, Stephan C, Laezza F. JAK2 regulates Nav1.6 channel function via FGF14 Y158 phosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118786. [PMID: 32599005 PMCID: PMC7984254 DOI: 10.1016/j.bbamcr.2020.118786] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Protein interactions between voltage-gated sodium (Nav) channels and accessory proteins play an essential role in neuronal firing and plasticity. However, a surprisingly limited number of kinases have been identified as regulators of these molecular complexes. We hypothesized that numerous as-of-yet unidentified kinases indirectly regulate the Nav channel via modulation of the intracellular fibroblast growth factor 14 (FGF14), an accessory protein with numerous unexplored phosphomotifs and required for channel function in neurons. METHODS Here we present results from an in-cell high-throughput screening (HTS) against the FGF14: Nav1.6 complex using >3000 diverse compounds targeting an extensive range of signaling pathways. Regulation by top kinase targets was then explored using in vitro phosphorylation, biophysics, mass-spectrometry and patch-clamp electrophysiology. RESULTS Compounds targeting Janus kinase 2 (JAK2) were over-represented among HTS hits. Phosphomotif scans supported by mass spectrometry revealed FGF14Y158, a site previously shown to mediate both FGF14 homodimerization and interactions with Nav1.6, as a JAK2 phosphorylation site. Following inhibition of JAK2, FGF14 homodimerization increased in a manner directly inverse to FGF14:Nav1.6 complex formation, but not in the presence of the FGF14Y158A mutant. Patch-clamp electrophysiology revealed that through Y158, JAK2 controls FGF14-dependent modulation of Nav1.6 channels. In hippocampal CA1 pyramidal neurons, the JAK2 inhibitor Fedratinib reduced firing by a mechanism that is dependent upon expression of FGF14. CONCLUSIONS These studies point toward a novel mechanism by which levels of JAK2 in neurons could directly influence firing and plasticity by controlling the FGF14 dimerization equilibrium, and thereby the availability of monomeric species for interaction with Nav1.6.
Collapse
Affiliation(s)
- Paul A Wadsworth
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA; Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Aditya K Singh
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Nghi Nguyen
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, USA
| | - Nolan M Dvorak
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, The University of Texas Medical Branch, Galveston, TX, USA
| | - Clifford Stephan
- HTS Screening Core, Center for Translational Cancer Research, Texas A&M Health Science Center: Institute of Biosciences and Technology, Houston, TX, USA
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX, USA; Center for Addiction Research, The University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
21
|
The Sodium Channel B4-Subunits are Dysregulated in Temporal Lobe Epilepsy Drug-Resistant Patients. Int J Mol Sci 2020; 21:ijms21082955. [PMID: 32331418 PMCID: PMC7216270 DOI: 10.3390/ijms21082955] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/14/2020] [Accepted: 04/21/2020] [Indexed: 01/09/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is the most common type of partial epilepsy referred for surgery due to antiepileptic drug (AED) resistance. A common molecular target for many of these drugs is the voltage-gated sodium channel (VGSC). The VGSC consists of four domains of pore-forming α-subunits and two auxiliary β-subunits, several of which have been well studied in epileptic conditions. However, despite the β4-subunits' role having been reported in some neurological conditions, there is little research investigating its potential significance in epilepsy. Therefore, the purpose of this work was to assess the role of SCN4β in epilepsy by using a combination of molecular and bioinformatics approaches. We first demonstrated that there was a reduction in the relative expression of SCN4B in the drug-resistant TLE patients compared to non-epileptic control specimens, both at the mRNA and protein levels. By analyzing a co-expression network in the neighborhood of SCN4B we then discovered a linkage between the expression of this gene and K+ channels activated by Ca2+, or K+ two-pore domain channels. Our approach also inferred several potential effector functions linked to variation in the expression of SCN4B. These observations support the hypothesis that SCN4B is a key factor in AED-resistant TLE, which could help direct both the drug selection of TLE treatments and the development of future AEDs.
Collapse
|
22
|
Mackrill JJ, Shiels HA. Evolution of Excitation-Contraction Coupling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:281-320. [DOI: 10.1007/978-3-030-12457-1_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
23
|
Soriani O, Kourrich S. The Sigma-1 Receptor: When Adaptive Regulation of Cell Electrical Activity Contributes to Stimulant Addiction and Cancer. Front Neurosci 2019; 13:1186. [PMID: 31780884 PMCID: PMC6861184 DOI: 10.3389/fnins.2019.01186] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022] Open
Abstract
The sigma-1 receptor (σ1R) is an endoplasmic reticulum (ER)-resident chaperone protein that acts like an inter-organelle signaling modulator. Among its several functions such as ER lipid metabolisms/transports and indirect regulation of genes transcription, one of its most intriguing feature is the ability to regulate the function and trafficking of a variety of functional proteins. To date, and directly relevant to the present review, σ1R has been found to regulate both voltage-gated ion channels (VGICs) belonging to distinct superfamilies (i.e., sodium, Na+; potassium, K+; and calcium, Ca2+ channels) and non-voltage-gated ion channels. This regulatory function endows σ1R with a powerful capability to fine tune cells’ electrical activity and calcium homeostasis—a regulatory power that appears to favor cell survival in pathological contexts such as stroke or neurodegenerative diseases. In this review, we present the current state of knowledge on σ1R’s role in the regulation of cellular electrical activity, and how this seemingly adaptive function can shift cell homeostasis and contribute to the development of very distinct chronic pathologies such as psychostimulant abuse and tumor cell growth in cancers.
Collapse
Affiliation(s)
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, Montréal, QC, Canada.,Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois, Université du Québec à Montréal, Montréal, QC, Canada.,Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
24
|
Johansson JK, Karema-Jokinen VI, Hakanen S, Jylhä A, Uusitalo H, Vihinen-Ranta M, Skottman H, Ihalainen TO, Nymark S. Sodium channels enable fast electrical signaling and regulate phagocytosis in the retinal pigment epithelium. BMC Biol 2019; 17:63. [PMID: 31412898 PMCID: PMC6694495 DOI: 10.1186/s12915-019-0681-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/11/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Voltage-gated sodium (Nav) channels have traditionally been considered a trademark of excitable cells. However, recent studies have shown the presence of Nav channels in several non-excitable cells, such as astrocytes and macrophages, demonstrating that the roles of these channels are more diverse than was previously thought. Despite the earlier discoveries, the presence of Nav channel-mediated currents in the cells of retinal pigment epithelium (RPE) has been dismissed as a cell culture artifact. We challenge this notion by investigating the presence and possible role of Nav channels in RPE both ex vivo and in vitro. RESULTS Our work demonstrates that several subtypes of Nav channels are found in human embryonic stem cell (hESC)-derived and mouse RPE, most prominently subtypes Nav1.4, Nav1.6, and Nav1.8. Whole cell patch clamp recordings from the hESC-derived RPE monolayers showed that the current was inhibited by TTX and QX-314 and was sensitive to the selective blockers of the main Nav subtypes. Importantly, we show that the Nav channels are involved in photoreceptor outer segment phagocytosis since blocking their activity significantly reduces the efficiency of particle internalization. Consistent with this role, our electron microscopy results and immunocytochemical analysis show that Nav1.4 and Nav1.8 accumulate on phagosomes and that pharmacological inhibition of Nav channels as well as silencing the expression of Nav1.4 with shRNA impairs the phagocytosis process. CONCLUSIONS Taken together, our study shows that Nav channels are present in RPE, giving this tissue the capacity of fast electrical signaling. The channels are critical for the physiology of RPE with an important role in photoreceptor outer segment phagocytosis.
Collapse
Affiliation(s)
- Julia K Johansson
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Viivi I Karema-Jokinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Satu Hakanen
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Antti Jylhä
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Hannu Uusitalo
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Tays Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Maija Vihinen-Ranta
- Department of Biological and Environmental Science and Nanoscience Center, University of Jyväskylä, Jyväskylä, Finland
| | - Heli Skottman
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu O Ihalainen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Soile Nymark
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| |
Collapse
|
25
|
Qu YS, Lazzerini PE, Capecchi PL, Laghi-Pasini F, El Sherif N, Boutjdir M. Autoimmune Calcium Channelopathies and Cardiac Electrical Abnormalities. Front Cardiovasc Med 2019; 6:54. [PMID: 31119135 PMCID: PMC6507622 DOI: 10.3389/fcvm.2019.00054] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/16/2019] [Indexed: 12/24/2022] Open
Abstract
Patients with autoimmune diseases are at increased risk for developing cardiovascular diseases, and abnormal electrocardiographic findings are common. Voltage-gated calcium channels play a major role in the cardiovascular system and regulate cardiac excitability and contractility. Particularly, by virtue of their localization and expression in the heart, calcium channels modulate pace making at the sinus node, conduction at the atrioventricular node and cardiac repolarization in the working myocardium. Consequently, emerging evidence suggests that calcium channels are targets to autoantibodies in autoimmune diseases. Autoimmune-associated cardiac calcium channelopathies have been recognized in both sinus node dysfunction atrioventricular block in patients positive for anti-Ro/La antibodies, and ventricular arrhythmias in patients with dilated cardiomyopathy. In this review, we discuss mechanisms of autoimmune-associated calcium channelopathies and their relationship with the development of cardiac electrical abnormalities.
Collapse
Affiliation(s)
- Yongxia Sarah Qu
- Department of Cardiology, New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY, United States.,VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Pietro Enea Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pier Leopoldo Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Nabil El Sherif
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Mohamed Boutjdir
- VA New York Harbor Healthcare System and State University of New York Downstate Medical Center, Brooklyn, NY, United States.,NYU School of Medicine, New York, NY, United States
| |
Collapse
|
26
|
Identification of CACNA1D variants associated with sinoatrial node dysfunction and deafness in additional Pakistani families reveals a clinical significance. J Hum Genet 2018; 64:153-160. [PMID: 30498240 DOI: 10.1038/s10038-018-0542-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 01/08/2023]
Abstract
Sinoatrial node dysfunction and deafness (SANDD) syndrome is rare and characterized by a low heart beat and severe-to-profound deafness. Additional features include fatigue, dizziness, and episodic syncope. The sinoatrial node (SAN) drives heart automaticity and continuously regulates heart rate. The CACNA1D gene encoding the Cav1.3 protein expressed in inner hair cells, atria and SAN, induces loss-of-function in channel activity and underlies SANDD. To date, only one variant c.1208_1209insGGG:p.(G403_V404insG) has been reported for SANDD syndrome. We studied five Pakistani families with SANDD and characterized a new missense variant p.(A376V) in CACNA1D in one family, and further characterized the founder variant p.(G403_V404insG) in four additional pedigrees. We show that affected individuals in the four families which segregate p.(G403_V404insG) share a 1.03 MB haplotype on 3p21.1 suggesting they share a common distant ancestor. In conclusion, we identified new and known variants in CACNA1D in five Pakistani families with SANDD. This study is of clinical importance as the CACNA1D founder variant is only observed in families from the Khyber Pakhtunkhwa (KPK) province, in Pakistan. Therefore, screening patients with congenital deafness for SAN dysfunction in this province could ensure adequate follow-up and prevent cardiac failure associated with SAN.
Collapse
|
27
|
T-Type voltage gated calcium channels: a target in breast cancer? Breast Cancer Res Treat 2018; 173:11-21. [PMID: 30242580 DOI: 10.1007/s10549-018-4970-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 09/15/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE The purpose of this review article is to discuss the potential of T-type voltage gated calcium channels (VGCCs) as drug targets in breast cancer. Breast cancer, attributable to the different molecular subtypes, has a crucial need for therapeutic strategies to counter the mortality rate. VGCCs play an important role in regulating cytosolic free calcium levels which regulate cellular processes like tumorigenesis and cancer progression. In the last decade, T-type VGCCs have been investigated in breast cancer proliferation. Calcium channel blockers, in general, have shown an anti-proliferative and cytotoxic effects. T-type VGCC antagonists have shown growth inhibition owing to the inhibition of CaV3.2 isoform. CaV3.1 isoform has been indicated as a tumour-suppressor gene candidate and is reported to support anti-proliferative and apoptotic activity in breast cancer. The distribution of T-type VGCC isoforms in different breast cancer molecular subtypes is diverse and needs to be further investigated. The role of T-type VGCCs in breast cancer migration, metastasis and more importantly in epithelial to mesenchymal transition (EMT) is yet to be elucidated. In addition, interlaced therapy, using a combination of chemotherapy drugs and T-type VGCC blockers, presents a promising therapeutic approach for breast cancer but more validation and clinical trials are needed. Also, for investigating the potential of T-type VGCC blocker therapy, there is a need for isoform-specific agonists/antagonists to define and discover roles of T-type VGCC specific isoforms. CONCLUSION Our article provides a review of the role of T-type VGCCs in breast cancer and also discusses future of the research in this area so that it can be ascertained whether there is any potential of T-type VGCCs as drug targets in breast cancer.
Collapse
|
28
|
El Zein RM, Boulkroun S, Fernandes-Rosa FL, Zennaro MC. Molecular genetics of Conn adenomas in the era of exome analysis. Presse Med 2018; 47:e151-e158. [PMID: 30072045 DOI: 10.1016/j.lpm.2018.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aldosterone-producing adenomas (APA) are a major cause of primary aldosteronism (PA), the most common form of secondary hypertension. Exome analysis of APA has allowed the identification of recurrent somatic mutations in KCNJ5, CACNA1D, ATP1A1, and ATP2B3 in more than 50 % of sporadic cases. These gain of function mutations in ion channels and pumps lead to increased and autonomous aldosterone production. In addition, somatic CTNNB1 mutations have also been identified in APA. The CTNNB1 mutations were also identified in cortisol-producing adenomas and adrenal cancer, but their role in APA development and the mechanisms specifying the hormonal production or the malignant phenotype remain unknown. The role of the somatic mutations in the regulation of aldosterone production is well understood, while the impact of these mutations on cell proliferation remains to be established. Furthermore, the sequence of events leading to APA formation is currently the focus of many studies. There is evidence for a two-hit model where the somatic mutations are second hits occurring in a previously remodeled adrenal cortex. On the other hand, the APA-driver mutations were also identified in aldosterone-producing cell clusters (APCC) in normal adrenals, suggesting that these structures may represent precursors for APA development. As PA due to APA can be cured by surgical removal of the affected adrenal gland, the identification of the underlying genetic abnormalities by novel biomarkers could improve diagnostic and therapeutic approaches of the disease. In this context, recent data on steroid profiling in peripheral venous samples of APA patients and on new drugs capable of inhibiting mutated potassium channels provide promising preliminary data with potential for translation into clinical care.
Collapse
Affiliation(s)
- Rami M El Zein
- Paris Cardiovascular Research Center, INSERM, UMRS 970, 56, rue Leblanc, 75015 Paris, France; University Paris Descartes, Sorbonne Paris cité, 12, rue de l'École-de-médecine, 75006 Paris, France
| | - Sheerazed Boulkroun
- Paris Cardiovascular Research Center, INSERM, UMRS 970, 56, rue Leblanc, 75015 Paris, France; University Paris Descartes, Sorbonne Paris cité, 12, rue de l'École-de-médecine, 75006 Paris, France
| | - Fabio Luiz Fernandes-Rosa
- Paris Cardiovascular Research Center, INSERM, UMRS 970, 56, rue Leblanc, 75015 Paris, France; University Paris Descartes, Sorbonne Paris cité, 12, rue de l'École-de-médecine, 75006 Paris, France; Assistance publique-Hôpitaux de Paris, hôpital européen Georges-Pompidou, service de génétique, 20, rue Leblanc, 75015 Paris, France
| | - Maria-Christina Zennaro
- Paris Cardiovascular Research Center, INSERM, UMRS 970, 56, rue Leblanc, 75015 Paris, France; University Paris Descartes, Sorbonne Paris cité, 12, rue de l'École-de-médecine, 75006 Paris, France; Assistance publique-Hôpitaux de Paris, hôpital européen Georges-Pompidou, service de génétique, 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
29
|
Endo C, Sato T, Yajima T, Igarashi K, Ichikawa H. Innervation of the Human Incisive Papilla: Comparison with Other Oral Regions. Cells Tissues Organs 2018; 205:93-104. [DOI: 10.1159/000488278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 03/08/2018] [Indexed: 01/18/2023] Open
Abstract
Immunohistochemistry for several neurochemical substances was performed on the human incisive papilla and other oral structures. Sodium channel alpha subunit 7 (SCN7A) protein-immunoreactive (IR) Schwann cells and protein gene product 9.5 (PGP 9.5)-IR nerve fibers made nerve plexuses beneath the epithelium of the palate, including the incisive papilla, tongue, and lip. SCN7A immunoreactivity could also be detected in lamellated and nonlamellated capsules of corpuscle endings. Lamellated SCN7A-IR corpuscle endings were mostly restricted to the mucous and cutaneous lips. These endings had thick and spiral-shaped PGP 9.5-IR axons without ramification. Nonlamellated SCN7A-IR corpuscle endings were most numerous in the incisive papilla among the oral regions. On the basis of axonal morphology, the nonlamellated endings were divided into simple and complex types. PGP 9.5-IR terminal axons in the simple type ran straight or meandered with slight ramification, whereas those in the complex type were densely entangled with abundant ramification. Substance P (SP)-, calcitonin gene-related peptide (CGRP)-, and transient receptor potential cation channel subfamily V member 2 (TRPV2)-IR varicose fibers were rarely seen beneath the epithelium of oral structures. The present study indicates that the human incisive papilla has many low-threshold mechanoreceptors with nonlamellated capsules. SP-, CGRP-, and TRPV2-containing nociceptors may be infrequent in the incisive papilla and other oral regions.
Collapse
|
30
|
Regulation of aldosterone production by ion channels: From basal secretion to primary aldosteronism. Biochim Biophys Acta Mol Basis Dis 2018; 1864:871-881. [DOI: 10.1016/j.bbadis.2017.12.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/05/2017] [Accepted: 12/23/2017] [Indexed: 01/07/2023]
|
31
|
Kanellopoulos AH, Koenig J, Huang H, Pyrski M, Millet Q, Lolignier S, Morohashi T, Gossage SJ, Jay M, Linley JE, Baskozos G, Kessler BM, Cox JJ, Dolphin AC, Zufall F, Wood JN, Zhao J. Mapping protein interactions of sodium channel Na V1.7 using epitope-tagged gene-targeted mice. EMBO J 2018; 37:427-445. [PMID: 29335280 PMCID: PMC5793798 DOI: 10.15252/embj.201796692] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 11/30/2017] [Accepted: 12/05/2017] [Indexed: 11/24/2022] Open
Abstract
The voltage-gated sodium channel NaV1.7 plays a critical role in pain pathways. We generated an epitope-tagged NaV1.7 mouse that showed normal pain behaviours to identify channel-interacting proteins. Analysis of NaV1.7 complexes affinity-purified under native conditions by mass spectrometry revealed 267 proteins associated with Nav1.7 in vivo The sodium channel β3 (Scn3b), rather than the β1 subunit, complexes with Nav1.7, and we demonstrate an interaction between collapsing-response mediator protein (Crmp2) and Nav1.7, through which the analgesic drug lacosamide regulates Nav1.7 current density. Novel NaV1.7 protein interactors including membrane-trafficking protein synaptotagmin-2 (Syt2), L-type amino acid transporter 1 (Lat1) and transmembrane P24-trafficking protein 10 (Tmed10) together with Scn3b and Crmp2 were validated by co-immunoprecipitation (Co-IP) from sensory neuron extract. Nav1.7, known to regulate opioid receptor efficacy, interacts with the G protein-regulated inducer of neurite outgrowth (Gprin1), an opioid receptor-binding protein, demonstrating a physical and functional link between Nav1.7 and opioid signalling. Further information on physiological interactions provided with this normal epitope-tagged mouse should provide useful insights into the many functions now associated with the NaV1.7 channel.
Collapse
Affiliation(s)
| | - Jennifer Koenig
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Honglei Huang
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - Martina Pyrski
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - Queensta Millet
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Stéphane Lolignier
- Molecular Nociception Group, WIBR, University College London, London, UK
- Université Clermont Auvergne, Inserm U1107 Neuro-Dol, Pharmacologie Fondamentale et Clinique de la Douleur, Clermont-Ferrand, France
| | - Toru Morohashi
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Samuel J Gossage
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Maude Jay
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - John E Linley
- Molecular Nociception Group, WIBR, University College London, London, UK
- Neuroscience, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | | | - Benedikt M Kessler
- TDI Mass Spectrometry Laboratory, Target Discovery Institute, University of Oxford, Oxford, UK
| | - James J Cox
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Annette C Dolphin
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Frank Zufall
- Center for Integrative Physiology and Molecular Medicine, Saarland University, Homburg, Germany
| | - John N Wood
- Molecular Nociception Group, WIBR, University College London, London, UK
| | - Jing Zhao
- Molecular Nociception Group, WIBR, University College London, London, UK
| |
Collapse
|
32
|
Zhang Q, Chen J, Qin Y, Wang J, Zhou L. Mutations in voltage-gated L-type calcium channel: implications in cardiac arrhythmia. Channels (Austin) 2018; 12:201-218. [PMID: 30027834 PMCID: PMC6104696 DOI: 10.1080/19336950.2018.1499368] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/08/2018] [Accepted: 07/05/2018] [Indexed: 02/06/2023] Open
Abstract
The voltage-gated L-type calcium channel (LTCC) is essential for multiple cellular processes. In the heart, calcium influx through LTCC plays an important role in cardiac electrical excitation. Mutations in LTCC genes, including CACNA1C, CACNA1D, CACNB2 and CACNA2D, will induce the dysfunctions of calcium channels, which result in the abnormal excitations of cardiomyocytes, and finally lead to cardiac arrhythmias. Nevertheless, the newly found mutations in LTCC and their functions are continuously being elucidated. This review summarizes recent findings on the mutations of LTCC, which are associated with long QT syndromes, Timothy syndromes, Brugada syndromes, short QT syndromes, and some other cardiac arrhythmias. Indeed, we describe the gain/loss-of-functions of these mutations in LTCC, which can give an explanation for the phenotypes of cardiac arrhythmias. Moreover, we present several challenges in the field at present, and propose some diagnostic or therapeutic approaches to these mutation-associated cardiac diseases in the future.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, the Second Affiliated Hospital of Nantong University, Nantong First Hospital, Nantong, Jiangsu, China
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junjie Chen
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Qin
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Juejin Wang
- Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
33
|
Zennaro MC, Boulkroun S, Fernandes-Rosa F. Genetic Causes of Functional Adrenocortical Adenomas. Endocr Rev 2017; 38:516-537. [PMID: 28973103 DOI: 10.1210/er.2017-00189] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/28/2017] [Indexed: 12/14/2022]
Abstract
Aldosterone and cortisol, the main mineralocorticoid and glucocorticoid hormones in humans, are produced in the adrenal cortex, which is composed of three concentric zones with specific functional characteristics. Adrenocortical adenomas (ACAs) can lead to the autonomous secretion of aldosterone responsible for primary aldosteronism, the most frequent form of secondary arterial hypertension. In the case of cortisol production, ACAs lead to overt or subclinical Cushing syndrome. Genetic analysis driven by next-generation sequencing technology has enabled the discovery, during the past 7 years, of the genetic causes of a large subset of ACAs. In particular, somatic mutations in genes regulating intracellular ionic homeostasis and membrane potential have been identified in aldosterone-producing adenomas. These mutations all promote increased intracellular calcium concentrations, with activation of calcium signaling, the main trigger for aldosterone production. In cortisol-producing adenomas, recurrent somatic mutations in PRKACA (coding for the cyclic adenosine monophosphate-dependent protein kinase catalytic subunit α) affect cyclic adenosine monophosphate-dependent protein kinase A signaling, leading to activation of cortisol biosynthesis. In addition to these specific pathways, the Wnt/β-catenin pathway appears to play an important role in adrenal tumorigenesis, because β-catenin mutations have been identified in both aldosterone- and cortisol-producing adenomas. This, together with different intermediate states of aldosterone and cortisol cosecretion, raises the possibility that the two conditions share a certain degree of genetic susceptibility. Alternatively, different hits might be responsible for the diseases, with one hit leading to adrenocortical cell proliferation and nodule formation and the second specifying the hormonal secretory pattern.
Collapse
Affiliation(s)
- Maria-Christina Zennaro
- French National Institute of Health and Medical Research (INSERM), Unité Mixte de Recherche Scientifique (UMRS)_970, Paris Cardiovascular Research Center, France.,Université Paris Descartes, Sorbonne Paris Cité, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, France
| | - Sheerazed Boulkroun
- French National Institute of Health and Medical Research (INSERM), Unité Mixte de Recherche Scientifique (UMRS)_970, Paris Cardiovascular Research Center, France.,Université Paris Descartes, Sorbonne Paris Cité, France
| | - Fabio Fernandes-Rosa
- French National Institute of Health and Medical Research (INSERM), Unité Mixte de Recherche Scientifique (UMRS)_970, Paris Cardiovascular Research Center, France.,Université Paris Descartes, Sorbonne Paris Cité, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Service de Génétique, France
| |
Collapse
|
34
|
Hastoy B, Clark A, Rorsman P, Lang J. Fusion pore in exocytosis: More than an exit gate? A β-cell perspective. Cell Calcium 2017; 68:45-61. [PMID: 29129207 DOI: 10.1016/j.ceca.2017.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/17/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022]
Abstract
Secretory vesicle exocytosis is a fundamental biological event and the process by which hormones (like insulin) are released into the blood. Considerable progress has been made in understanding this precisely orchestrated sequence of events from secretory vesicle docked at the cell membrane, hemifusion, to the opening of a membrane fusion pore. The exact biophysical and physiological regulation of these events implies a close interaction between membrane proteins and lipids in a confined space and constrained geometry to ensure appropriate delivery of cargo. We consider some of the still open questions such as the nature of the initiation of the fusion pore, the structure and the role of the Soluble N-ethylmaleimide-sensitive-factor Attachment protein REceptor (SNARE) transmembrane domains and their influence on the dynamics and regulation of exocytosis. We discuss how the membrane composition and protein-lipid interactions influence the likelihood of the nascent fusion pore forming. We relate these factors to the hypothesis that fusion pore expansion could be affected in type-2 diabetes via changes in disease-related gene transcription and alterations in the circulating lipid profile. Detailed characterisation of the dynamics of the fusion pore in vitro will contribute to understanding the larger issue of insulin secretory defects in diabetes.
Collapse
Affiliation(s)
- Benoit Hastoy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK.
| | - Anne Clark
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK; Metabolic Research, Institute of Neuroscience and Physiology, University of Goteborg, Medicinaregatan 11, S-41309 Göteborg, Sweden
| | - Jochen Lang
- Laboratoire de Chimie et Biologie des Membranes et Nano-objets (CBMN), CNRS UMR 5248, Université de Bordeaux, Allée de Geoffrey St Hilaire, 33600 Pessac, France.
| |
Collapse
|
35
|
Fernandes-Rosa FL, Boulkroun S, Zennaro MC. Somatic and inherited mutations in primary aldosteronism. J Mol Endocrinol 2017; 59:R47-R63. [PMID: 28400483 DOI: 10.1530/jme-17-0035] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 04/11/2017] [Indexed: 01/22/2023]
Abstract
Primary aldosteronism (PA), the most common form of secondary hypertension, is caused in the majority of cases by unilateral aldosterone-producing adenoma (APA) or bilateral adrenal hyperplasia. Over the past few years, somatic mutations in KCNJ5, CACNA1D, ATP1A1 and ATP2B3 have been proven to be associated with APA development, representing more than 50% of sporadic APA. The identification of these mutations has allowed the development of a model for APA involving modification on the intracellular ionic equilibrium and regulation of cell membrane potential, leading to autonomous aldosterone overproduction. Furthermore, somatic CTNNB1 mutations have also been identified in APA, but the link between these mutations and APA development remains unknown. The sequence of events responsible for APA formation is not completely understood, in particular, whether a single hit or a double hit is responsible for both aldosterone overproduction and cell proliferation. Germline mutations identified in patients with early-onset PA have expanded the classification of familial forms (FH) of PA. The description of germline KCNJ5 and CACNA1H mutations has identified FH-III and FH-IV based on genetic findings; germline CACNA1D mutations have been identified in patients with very early-onset PA and severe neurological abnormalities. This review summarizes current knowledge on the genetic basis of PA, the association of driver gene mutations and clinical findings and in the contribution to patient care, plus the current understanding on the mechanisms of APA development.
Collapse
Affiliation(s)
- Fabio Luiz Fernandes-Rosa
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| | - Sheerazed Boulkroun
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
| | - Maria-Christina Zennaro
- INSERMUMRS_970, Paris Cardiovascular Research Center, Paris, France
- University Paris DescartesSorbonne Paris Cité, Paris, France
- Assistance Publique-Hôpitaux de ParisHôpital Européen Georges Pompidou, Service de Génétique, Paris, France
| |
Collapse
|
36
|
Wang Y, Fehlhaber KE, Sarria I, Cao Y, Ingram NT, Guerrero-Given D, Throesch B, Baldwin K, Kamasawa N, Ohtsuka T, Sampath AP, Martemyanov KA. The Auxiliary Calcium Channel Subunit α2δ4 Is Required for Axonal Elaboration, Synaptic Transmission, and Wiring of Rod Photoreceptors. Neuron 2017; 93:1359-1374.e6. [PMID: 28262416 PMCID: PMC5364038 DOI: 10.1016/j.neuron.2017.02.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 12/31/2016] [Accepted: 02/08/2017] [Indexed: 11/24/2022]
Abstract
Neural circuit wiring relies on selective synapse formation whereby a presynaptic release apparatus is matched with its cognate postsynaptic machinery. At metabotropic synapses, the molecular mechanisms underlying this process are poorly understood. In the mammalian retina, rod photoreceptors form selective contacts with rod ON-bipolar cells by aligning the presynaptic voltage-gated Ca2+ channel directing glutamate release (CaV1.4) with postsynaptic mGluR6 receptors. We show this coordination requires an extracellular protein, α2δ4, which complexes with CaV1.4 and the rod synaptogenic mediator, ELFN1, for trans-synaptic alignment with mGluR6. Eliminating α2δ4 in mice abolishes rod synaptogenesis and synaptic transmission to rod ON-bipolar cells, and disrupts postsynaptic mGluR6 clustering. We further find that in rods, α2δ4 is crucial for organizing synaptic ribbons and setting CaV1.4 voltage sensitivity. In cones, α2δ4 is essential for CaV1.4 function, but is not required for ribbon organization, synaptogenesis, or synaptic transmission. These findings offer insights into retinal pathologies associated with α2δ4 dysfunction.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Katherine E Fehlhaber
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ignacio Sarria
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yan Cao
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Norianne T Ingram
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Debbie Guerrero-Given
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Ben Throesch
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Kristin Baldwin
- Department of Molecular and Cellular Neuroscience, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92121, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute, 1 Max Planck Way, Jupiter, FL 33458, USA
| | - Toshihisa Ohtsuka
- Department of Biochemistry, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Alapakkam P Sampath
- Jules Stein Eye Institute, Department of Ophthalmology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kirill A Martemyanov
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
37
|
Hosseinzadeh Z, Singh Y, Shimshek DR, van der Putten H, Wagner CA, Lang F. Leucine-Rich Repeat Kinase 2 (Lrrk2)-Sensitive Na +/K + ATPase Activity in Dendritic Cells. Sci Rep 2017; 7:41117. [PMID: 28120865 PMCID: PMC5264149 DOI: 10.1038/srep41117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/14/2016] [Indexed: 12/18/2022] Open
Abstract
Leucine-rich repeat kinase 2 (Lrrk2) has been implicated in the pathophysiology of Parkinson's disease. Lrrk2 is expressed in diverse cells including neurons and dendritic cells (DCs). In DCs Lrrk2 was shown to up-regulate Na+/Ca2+-exchanger activity. The elimination of Ca2+ by Na+/Ca2+ -exchangers requires maintenance of the Na+ gradient by the Na+/K+ -ATPase. The present study thus explored whether Lrrk2 impacts on Na+/K+ -ATPase expression and function. To this end DCs were isolated from gene-targeted mice lacking Lrrk2 (Lrrk2-/-) and their wild-type littermates (Lrrk2+/+). Na+/K+ -ATPase activity was estimated from K+ induced, ouabain sensitive, current determined by whole cell patch clamp. Na+/K+ -ATPase α1 subunit transcript and protein levels were determined by RT-qPCR and flow cytometry. As a result, the K+ induced current was significantly smaller in Lrrk2-/- than in Lrrk2+/+ DCs and was completely abolished by ouabain (100 μM) in both genotypes. The K+ induced, ouabain sensitive, current in Lrrk2+/+ DCs was significantly blunted by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). The Na+/K+ -ATPase α1 subunit transcript and protein levels were significantly lower in Lrrk2-/- than in Lrrk2+/+ DCs and significantly decreased by Lrrk2 inhibitor GSK2578215A (1 μM, 24 hours). In conclusion, Lrrk2 is a powerful regulator of Na+/K+ -ATPase expression and activity in dendritic cells.
Collapse
Affiliation(s)
- Zohreh Hosseinzadeh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
- Experimental Retinal Prosthetics Group, Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| | - Derya R. Shimshek
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Herman van der Putten
- Department of Neuroscience, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
- National Contest for Life (NCL) Foundation, 203555 Hamburg, Germany
| | - Carsten A. Wagner
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen, Germany
| |
Collapse
|
38
|
Abstract
The sigma-1 receptor (Sig-1R), via interaction with various proteins, including voltage-gated and ligand-gated ion channels (VGICs and LGICs), is involved in a plethora of neuronal functions. This capability to regulate a variety of ion channel targets endows the Sig-1R with a powerful capability to fine tune neuronal excitability, and thereby the transmission of information within brain circuits. This versatility may also explain why the Sig-1R is associated to numerous diseases at both peripheral and central levels. To date, how the Sig-1R chooses its targets and how the combinations of target modulations alter overall neuronal excitability is one of the challenges in the field of Sig-1R-dependent regulation of neuronal activity. Here, we will describe and discuss the latest findings on Sig-1R-dependent modulation of VGICs and LGICs, and provide hypotheses that may explain the diverse excitability outcomes that have been reported so far.
Collapse
Affiliation(s)
- Saïd Kourrich
- Department of Psychiatry, University of Texas Southwestern Medical Center, 2201 Inwood Road, Dallas, TX, 75390-9070, USA.
| |
Collapse
|
39
|
Kambouris M, Thevenon J, Soldatos A, Cox A, Stephen J, Ben-Omran T, Al-Sarraj Y, Boulos H, Bone W, Mullikin JC, Masurel-Paulet A, St-Onge J, Dufford Y, Chantegret C, Thauvin-Robinet C, Al-Alami J, Faivre L, Riviere JB, Gahl WA, Bassuk AG, Malicdan MCV, El-Shanti H. Biallelic SCN10A mutations in neuromuscular disease and epileptic encephalopathy. Ann Clin Transl Neurol 2016; 4:26-35. [PMID: 28078312 PMCID: PMC5221474 DOI: 10.1002/acn3.372] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/02/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022] Open
Abstract
Objectives Two consanguineous families, one of Sudanese ethnicity presenting progressive neuromuscular disease, severe cognitive impairment, muscle weakness, upper motor neuron lesion, anhydrosis, facial dysmorphism, and recurrent seizures and the other of Egyptian ethnicity presenting with neonatal hypotonia, bradycardia, and recurrent seizures, were evaluated for the causative gene mutation. Methods and Results Homozygosity mapping and whole exome sequencing (WES) identified damaging homozygous variants in SCN10A, namely c.4514C>T; p.Thr1505Met in the first family and c.4735C>T; p.Arg1579* in the second family. A third family, of Western European descent, included a child with febrile infection‐related epilepsy syndrome (FIRES) who also had compound heterozygous missense mutations in SCN10A, namely, c.3482T>C; p.Met1161Thr and c.4709C>A; p.Thr1570Lys. A search for SCN10A variants in three consortia datasets (EuroEPINOMICS, Epi4K/EPGP, Autism/dbGaP) identified an additional five individuals with compound heterozygous variants. A Hispanic male with infantile spasms [c.2842G>C; p.Val948Leu and c.1453C>T; p.Arg485Cys], and a Caucasian female with Lennox–Gastaut syndrome [c.1529C>T; p.Pro510Leu and c.4984G>A; p.Gly1662Ser] in the epilepsy databases and three in the autism databases with [c.4009T>A; p.Ser1337Thr and c.1141A>G; p.Ile381Val], [c.2972C>T; p.Pro991Leu and c.2470C>T; p.His824Tyr], and [c.4009T>A; p.Ser1337Thr and c.2052G>A; p.Met684Ile]. Interpretation SCN10A is a member of the voltage‐gated sodium channel (VGSC) gene family. Sodium channels are responsible for the instigation and proliferation of action potentials in central and peripheral nervous systems. Heterozygous mutations in VGSC genes cause a wide range of epileptic and peripheral nervous system disorders. This report presents autosomal recessive mutations in SCN10A that may be linked to epilepsy‐related phenotypes, Lennox–Gastaut syndrome, infantile spasms, and Autism Spectrum Disorder.
Collapse
Affiliation(s)
- Marios Kambouris
- Pathology-Genetics Sidra Medical and Research Center Doha Qatar; Qatar Biomedical Research Institute Medical Genetics Center Hamad Bin Khalifa University Doha Qatar; Genetics Yale University School of Medicine New Haven Chicago
| | | | - Ariane Soldatos
- Medical Genetics Branch National Human Genome Research Institute National Institutes of Health Bethesda Maryland; Undiagnosed Diseases Program National Human Genome Research Institute National Institutes of Health Bethesda Maryland
| | - Allison Cox
- Pediatrics University of Iowa Iowa City Iowa
| | - Joshi Stephen
- Medical Genetics Branch National Human Genome Research Institute National Institutes of Health Bethesda Maryland
| | - Tawfeg Ben-Omran
- Clinical and Metabolic Genetics Pediatrics Hamad Medical Corporation Doha Qatar; Weill Cornell Medical College Doha Qatar
| | - Yasser Al-Sarraj
- Qatar Biomedical Research Institute Medical Genetics Center Hamad Bin Khalifa University Doha Qatar
| | - Hala Boulos
- Human Genetics University of Chicago Chicago Illinois
| | - William Bone
- Undiagnosed Diseases Program National Human Genome Research Institute National Institutes of Health Bethesda Maryland
| | - James C Mullikin
- Intramural Sequencing Center and Comparative Genomics Unit Genome Technology Branch National Genome Research Institute National Institutes of Health Bethesda Maryland
| | | | | | | | | | | | | | | | | | | | - William A Gahl
- Medical Genetics Branch National Human Genome Research Institute National Institutes of Health Bethesda Maryland; Undiagnosed Diseases Program National Human Genome Research Institute National Institutes of Health Bethesda Maryland
| | | | - May Christine V Malicdan
- Undiagnosed Diseases Program National Human Genome Research Institute National Institutes of Health Bethesda Maryland
| | - Hatem El-Shanti
- Qatar Biomedical Research Institute Medical Genetics Center Hamad Bin Khalifa University Doha Qatar; Pediatrics University of Iowa Iowa City Iowa; Pediatrics University of Jordan Amman Jordan
| |
Collapse
|
40
|
The effects of ropivacaine hydrochloride on the expression of CaMK II mRNA in the dorsal root ganglion neurons. Biomed Pharmacother 2016; 84:2014-2019. [PMID: 27863837 DOI: 10.1016/j.biopha.2016.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 01/27/2023] Open
Abstract
In this study, we identified the subtype of Calcium/calmodulin-dependent protein kinase II (CaMK II) mRNA in dorsal root ganglion neurons and observed the effects of ropivacaine hydrochloride in different concentration and different exposure time on the mRNA expression. Dorsal root ganglion neurons were isolated from the SD rats and cultured in vitro. The mRNA of the CaMK II subtype in dorsal root ganglion neurons were detected by real-time PCR. As well as, the dorsal root ganglion neurons were treated with ropivacaine hydrochloride in different concentration (1mM,2mM, 3mM and 4mM) for the same exposure time of 4h, or different exposure time (0h,2h,3h,4h and 6h) at the same concentration(3mM). The changes of the mRNA expression of the CaMK II subtype were observed with real-time PCR. All subtype mRNA of the CaMK II, CaMK IIα, CaMK IIβ, CaMK II δ, CaMK IIγ, can be detected in dorsal root ganglion neurons. With the increased of the concentration and exposure time of the ropivacaine hydrochloride, all the subtype mRNA expression increased. Ropivacaine hydrochloride up-regulate the CaMK IIβ, CaMK IIδ, CaMK IIg mRNA expression with the concentration and exposure time increasing. The nerve blocking or the neurotoxicity of the ropivacaine hydrochloride maybe involved with CaMK II.
Collapse
|
41
|
Motomura M, Nakata R, Shiraishi H. Lambert-Eaton myasthenic syndrome: Clinical review. ACTA ACUST UNITED AC 2016. [DOI: 10.1111/cen3.12326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Masakatsu Motomura
- Medical Engineering Course; Department of Engineering; Faculty of Engineering; Nagasaki Institute of Applied Science; Nagasaki Japan
- Department of Neurology and Strokology; Nagasaki University Hospital; Nagasaki Japan
| | - Ruka Nakata
- Department of Neurology and Strokology; Nagasaki University Hospital; Nagasaki Japan
- Department of Neurology; Nagasaki Kita Hospital; Nagasaki Japan
| | - Hirokazu Shiraishi
- Department of Neurology and Strokology; Nagasaki University Hospital; Nagasaki Japan
| |
Collapse
|
42
|
Catterall WA. Regulation of Cardiac Calcium Channels in the Fight-or-Flight Response. Curr Mol Pharmacol 2016; 8:12-21. [PMID: 25966697 DOI: 10.2174/1874467208666150507103417] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 02/17/2015] [Accepted: 04/20/2015] [Indexed: 11/22/2022]
Abstract
Intracellular calcium transients generated by activation of voltage-gated calcium (CaV) channels generate local signals, which initiate physiological processes such as secretion, synaptic transmission, and excitation-contraction coupling. Regulation of calcium entry through CaV channels is crucial for control of these physiological processes. In this article, I review experimental results that have emerged over several years showing that cardiac CaV1.2 channels form a local signaling complex, in which their proteolytically processed distal C-terminal domain, an A-Kinase Anchoring Protein, and cyclic AMP-dependent protein kinase (PKA) interact directly with the transmembrane core of the ion channel through the proximal C-terminal domain. This signaling complex is the substrate for β-adrenergic up-regulation of the CaV1.2 channel in the heart during the fight-or-flight response. Protein phosphorylation of two sites at the interface between the distal and proximal C-terminal domains contributes importantly to control of basal CaV1.2 channel activity, and phosphorylation of Ser1700 by PKA at that interface up-regulates CaV1.2 activity in response to β-adrenergic signaling. Thus, the intracellular C-terminal domain of CaV1.2 channels serves as a signaling platform, mediating beat-to-beat physiological regulation of channel activity and up-regulation by β-adrenergic signaling in the fight-or-flight response.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, Box 357280, University of Washington, Seattle, WA 98195-7280.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Aldosterone regulation in the adrenal plays an important role in blood pressure. The commonest curable cause of hypertension is primary aldosteronism. Recently, mutations in novel genes have been identified to cause primary aldosteronism. Elucidating the mechanism of action of these genetic abnormalities may help understand the cause of primary aldosteronism and the physiological regulation of aldosterone in the zona glomerulosa. RECENT FINDINGS KCNJ5, ATP1A1, ATP2B3, CACNA1D, CTNNB1, and CACNA1H mutations are causal of primary aldosteronism. ARMC5 may cause bilateral lesions resulting in primary aldosteronism.LGR5, DACH1, and neuron-specific proteins are highly expressed in the zona glomerulosa and regulate aldosterone production. SUMMARY Most mutations causing primary aldosteronism are in genes encoding cation channels or pumps, leading to increased calcium influx. Genotype-phenotype analyses identified two broad subtypes of aldosterone-producing adenomas (APAs), zona fasciculata-like and zona glomerulosa-like, and the likelihood of under-diagnosed zona glomerulosa-like APAs because of small size. Zona fasciculata-like APAs are only associated with KCNJ5 mutations, whereas zona glomerulosa-like APAs are associated with mutations in ATPase pumps, CACNA1D, and CTNNB1. The frequency of APAs, and the multiplicity of causal mutations, suggests a pre-existing drive for these mutations. We speculate that these mutations are selected for protecting against tonic inhibition of aldosterone in human zona glomerulosa, which express genes inhibiting aldosterone production.
Collapse
Affiliation(s)
- Elena A B Azizan
- aDepartment of Medicine, Faculty of Medicine, The National University of Malaysia (UKM) Medical Centre, Kuala Lumpur, Malaysia bThe Barts Heart Centre, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
44
|
Nanba K, Chen AX, Omata K, Vinco M, Giordano TJ, Else T, Hammer GD, Tomlins SA, Rainey WE. Molecular Heterogeneity in Aldosterone-Producing Adenomas. J Clin Endocrinol Metab 2016; 101:999-1007. [PMID: 26765578 PMCID: PMC4803171 DOI: 10.1210/jc.2015-3239] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT The use of next-generation sequencing has resulted in the identification of recurrent somatic mutations underlying primary aldosteronism (PA). However, significant gaps remain in our understanding of the relationship between tumor aldosterone synthase (CYP11B2) expression and somatic mutation status. OBJECTIVE The objective of the study was to investigate tumor CYP11B2 expression and somatic aldosterone-driver gene mutation heterogeneity. METHODS Fifty-one adrenals from 51 PA patients were studied. Immunohistochemistry for CYP11B2 was performed. Aldosterone-producing adenomas with intratumor CYP11B2 heterogeneity were analyzed for mutation status using targeted next-generation sequencing. DNA was isolated from CYP11B2-positive, CYP11B2-negative, and adjacent normal areas from formalin-fixed, paraffin-embedded sections. RESULTS Of 51 adrenals, seven (14 %) showed distinct heterogeneity in CYP11B2 by immunohistochemistry, including six adenomas with intratumor heterogeneity and one multinodular hyperplastic adrenal with both CYP11B2-positive and -negative nodules. Of the six adrenocortical adenomas with CYP11B2 heterogeneity, three had aldosterone-regulating mutations (CACNA1D p.F747C, KCNJ5 p.L168R, ATP1A1 p.L104R) only in CYP11B2-positive regions, and one had two different mutations localized to two histologically distinct CYP11B2-positive regions (ATP2B3 p.L424_V425del, KCNJ5 p.G151R). Lastly, one adrenal with multiple CYP11B2-expressing nodules showed different mutations in each (CACNA1D p.F747V and ATP1A1 p.L104R), and no mutations were identified in CYP11B2-negative nodule or adjacent normal adrenal. CONCLUSIONS Adrenal tumors in patients with PA can demonstrate clear heterogeneity in CYP11B2 expression and somatic mutations in driver genes for aldosterone production. These findings suggest that aldosterone-producing adenoma tumorigenesis can occur within preexisting nodules through the acquisition of somatic mutations that drive aldosterone production.
Collapse
Affiliation(s)
- Kazutaka Nanba
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Andrew X Chen
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Kei Omata
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Michelle Vinco
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Thomas J Giordano
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Tobias Else
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Gary D Hammer
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - Scott A Tomlins
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| | - William E Rainey
- Departments of Molecular and Integrative Physiology and Internal Medicine (K.N., A.X.C., G.D.H., W.E.R.), Pathology (K.O., M.V., T.J.G., S.A.T.), and Urology (S.A.T.), Comprehensive Cancer Center (T.J.G., T.E., S.A.T.), Division of Metabolism, Endocrinology, and Diabetes (T.J.G., T.E., G.D.H.), Endocrine Oncology Program (T.E., G.D.H.), Center for Organogenesis, and Michigan Center for Translational Pathology (S.A.T.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
45
|
Tewari D, Mukhopadhyay M, Nekkanti MS, Vallabhaneni S, Sahu G, Jetti SK, Preethidan D, Bera AK. Cytoprotective effect of Centella asiatica is mediated through the modulation of mitochondrial voltage-dependent anion channel (VDAC) and scavenging of free radicals. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
46
|
Heine M, Ciuraszkiewicz A, Voigt A, Heck J, Bikbaev A. Surface dynamics of voltage-gated ion channels. Channels (Austin) 2016; 10:267-81. [PMID: 26891382 DOI: 10.1080/19336950.2016.1153210] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Neurons encode information in fast changes of the membrane potential, and thus electrical membrane properties are critically important for the integration and processing of synaptic inputs by a neuron. These electrical properties are largely determined by ion channels embedded in the membrane. The distribution of most ion channels in the membrane is not spatially uniform: they undergo activity-driven changes in the range of minutes to days. Even in the range of milliseconds, the composition and topology of ion channels are not static but engage in highly dynamic processes including stochastic or activity-dependent transient association of the pore-forming and auxiliary subunits, lateral diffusion, as well as clustering of different channels. In this review we briefly discuss the potential impact of mobile sodium, calcium and potassium ion channels and the functional significance of this for individual neurons and neuronal networks.
Collapse
Affiliation(s)
- Martin Heine
- a RG Molecular Physiology, Leibniz Institute for Neurobiology, Center for Behavioral Brain Science, Otto-von-Guericke-University of Magdeburg , Magdeburg , Germany
| | - Anna Ciuraszkiewicz
- a RG Molecular Physiology, Leibniz Institute for Neurobiology, Center for Behavioral Brain Science, Otto-von-Guericke-University of Magdeburg , Magdeburg , Germany
| | - Andreas Voigt
- b Lehrstuhl Systemverfahrenstechnik, Otto-von-Guericke-University of Magdeburg , Magdeburg , Germany
| | - Jennifer Heck
- a RG Molecular Physiology, Leibniz Institute for Neurobiology, Center for Behavioral Brain Science, Otto-von-Guericke-University of Magdeburg , Magdeburg , Germany
| | - Arthur Bikbaev
- a RG Molecular Physiology, Leibniz Institute for Neurobiology, Center for Behavioral Brain Science, Otto-von-Guericke-University of Magdeburg , Magdeburg , Germany
| |
Collapse
|
47
|
Jeevaratnam K, Guzadhur L, Goh YM, Grace AA, Huang CLH. Sodium channel haploinsufficiency and structural change in ventricular arrhythmogenesis. Acta Physiol (Oxf) 2016; 216:186-202. [PMID: 26284956 DOI: 10.1111/apha.12577] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 06/11/2015] [Accepted: 07/24/2015] [Indexed: 12/19/2022]
Abstract
Normal cardiac excitation involves orderly conduction of electrical activation and recovery dependent upon surface membrane, voltage-gated, sodium (Na(+) ) channel α-subunits (Nav 1.5). We summarize experimental studies of physiological and clinical consequences of loss-of-function Na(+) channel mutations. Of these conditions, Brugada syndrome (BrS) and progressive cardiac conduction defect (PCCD) are associated with sudden, often fatal, ventricular tachycardia (VT) or fibrillation. Mouse Scn5a(+/-) hearts replicate important clinical phenotypes modelling these human conditions. The arrhythmic phenotype is associated not only with the primary biophysical change but also with additional, anatomical abnormalities, in turn dependent upon age and sex, each themselves exerting arrhythmic effects. Available evidence suggests a unified binary scheme for the development of arrhythmia in both BrS and PCCD. Previous biophysical studies suggested that Nav 1.5 deficiency produces a background electrophysiological defect compromising conduction, thereby producing an arrhythmic substrate unmasked by flecainide or ajmaline challenge. More recent reports further suggest a progressive decline in conduction velocity and increase in its dispersion particularly in ageing male Nav 1.5 haploinsufficient compared to WT hearts. This appears to involve a selective appearance of slow conduction at the expense of rapidly conducting pathways with changes in their frequency distributions. These changes were related to increased cardiac fibrosis. It is thus the combination of the structural and biophysical changes both accentuating arrhythmic substrate that may produce arrhythmic tendency. This binary scheme explains the combined requirement for separate, biophysical and structural changes, particularly occurring in ageing Nav 1.5 haploinsufficient males in producing clinical arrhythmia.
Collapse
Affiliation(s)
- K. Jeevaratnam
- Faculty of Health and Medical Science; University of Surrey; Guilford UK
- Perdana University - Royal College of Surgeons Ireland; Serdang Selangor Darul Ehsan Malaysia
| | - L. Guzadhur
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
- Niche Science & Technology; Richmond UK
| | - Y. M. Goh
- Department of Preclinical Sciences; Faculty of Veterinary Medicine; University Putra Malaysia; Serdang Selangor Darul Ehsan Malaysia
| | - A. A. Grace
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
| | - C. L.-H. Huang
- Division of Cardiovascular Biology; Department of Biochemistry; University of Cambridge; Cambridge UK
- Physiological Laboratory; University of Cambridge; Cambridge UK
| |
Collapse
|
48
|
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280.
| |
Collapse
|
49
|
Åkerström T, Willenberg HS, Cupisti K, Ip J, Backman S, Moser A, Maharjan R, Robinson B, Iwen KA, Dralle H, D Volpe C, Bäckdahl M, Botling J, Stålberg P, Westin G, Walz MK, Lehnert H, Sidhu S, Zedenius J, Björklund P, Hellman P. Novel somatic mutations and distinct molecular signature in aldosterone-producing adenomas. Endocr Relat Cancer 2015; 22:735-44. [PMID: 26285814 DOI: 10.1530/erc-15-0321] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Aldosterone-producing adenomas (APAs) are found in 1.5-3.0% of hypertensive patients in primary care and can be cured by surgery. Elucidation of genetic events may improve our understanding of these tumors and ultimately improve patient care. Approximately 40% of APAs harbor a missense mutation in the KCNJ5 gene. More recently, somatic mutations in CACNA1D, ATP1A1 and ATP2B3, also important for membrane potential/intracellular Ca(2) (+) regulation, were observed in APAs. In this study, we analyzed 165 APAs for mutations in selected regions of these genes. We then correlated mutational findings with clinical and molecular phenotype using transcriptome analysis, immunohistochemistry and semiquantitative PCR. Somatic mutations in CACNA1D in 3.0% (one novel mutation), ATP1A1 in 6.1% (six novel mutations) and ATP2B3 in 3.0% (two novel mutations) were detected. All observed mutations were located in previously described hotspot regions. Patients with tumors harboring mutations in CACNA1D, ATP1A1 and ATP2B3 were operated at an older age, were more often male and had tumors that were smaller than those in patients with KCNJ5 mutated tumors. Microarray transcriptome analysis segregated KCNJ5 mutated tumors from ATP1A1/ATP2B3 mutated tumors and those without mutation. We observed significant transcription upregulation of CYP11B2, as well as the previously described glomerulosa-specific gene NPNT, in ATP1A1/ATP2B3 mutated tumors compared to KCNJ5 mutated tumors. In summary, we describe novel somatic mutations in proteins regulating the membrane potential/intracellular Ca(2) (+) levels, and also a distinct mRNA and clinical signature, dependent on genetic alteration.
Collapse
Affiliation(s)
- Tobias Åkerström
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Holger Sven Willenberg
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Kenko Cupisti
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Julian Ip
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Samuel Backman
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Ana Moser
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Rajani Maharjan
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Bruce Robinson
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - K Alexander Iwen
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Henning Dralle
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Cristina D Volpe
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Martin Bäckdahl
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Johan Botling
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Peter Stålberg
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Gunnar Westin
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Martin K Walz
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Hendrik Lehnert
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Stan Sidhu
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Jan Zedenius
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Peyman Björklund
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| | - Per Hellman
- Department of Surgical SciencesUppsala University, Uppsala, SwedenDepartment of Endocrinology and MetabolismRostock University Medical Center, GermanyGeneralVisceral and Pediatric Surgery University Hospital Düsseldorf, Düsseldorf, GermanyUniversity of SydneyEndocrine Surgical Unit and Cancer Genetics, Hormones and Cancer Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, AustraliaDepartment of Medicine IUniversity of Lübeck, University Hospital, Lübeck, GermanyDepartment of GeneralVisceral and Vascular Surgery, University Hospital, University of Halle-Wittenberg, Halle/Saale, GermanyDepartment of Molecular Medicine and SurgeryEndocrine Surgery Unit, Karolinska Institutet, Karolinska University Hospital, Stockholm, SwedenDepartment of ImmunologyGenetics and Pathology, Uppsala University, Uppsala, SwedenKlinik für Chirurgie und Zentrum für Minimal Invasive ChirurgieKliniken Essen-Mitte, Essen, Germany
| |
Collapse
|
50
|
Campiglio M, Flucher BE. The role of auxiliary subunits for the functional diversity of voltage-gated calcium channels. J Cell Physiol 2015; 230:2019-31. [PMID: 25820299 PMCID: PMC4672716 DOI: 10.1002/jcp.24998] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/18/2022]
Abstract
Voltage-gated calcium channels (VGCCs) represent the sole mechanism to convert membrane depolarization into cellular functions like secretion, contraction, or gene regulation. VGCCs consist of a pore-forming α(1) subunit and several auxiliary channel subunits. These subunits come in multiple isoforms and splice-variants giving rise to a stunning molecular diversity of possible subunit combinations. It is generally believed that specific auxiliary subunits differentially regulate the channels and thereby contribute to the great functional diversity of VGCCs. If auxiliary subunits can associate and dissociate from pre-existing channel complexes, this would allow dynamic regulation of channel properties. However, most auxiliary subunits modulate current properties very similarly, and proof that any cellular calcium channel function is indeed modulated by the physiological exchange of auxiliary subunits is still lacking. In this review we summarize available information supporting a differential modulation of calcium channel functions by exchange of auxiliary subunits, as well as experimental evidence in support of alternative functions of the auxiliary subunits. At the heart of the discussion is the concept that, in their native environment, VGCCs function in the context of macromolecular signaling complexes and that the auxiliary subunits help to orchestrate the diverse protein-protein interactions found in these calcium channel signalosomes. Thus, in addition to a putative differential modulation of current properties, differential subcellular targeting properties and differential protein-protein interactions of the auxiliary subunits may explain the need for their vast molecular diversity.
Collapse
Affiliation(s)
- Marta Campiglio
- Division of Physiology, Department of Physiology and Medical Physics, Medical University InnsbruckInnsbruck, Austria
| | - Bernhard E Flucher
- Division of Physiology, Department of Physiology and Medical Physics, Medical University InnsbruckInnsbruck, Austria
| |
Collapse
|