Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 224
Filter
1.
J Mol Neurosci ; 72(8): 1598-1608, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35507012

ABSTRACT

Dry eye disease (DED) is a multifactorial disorder with recognized pathology, but not entirely known pathomechanism. It is suggested to represent a continuum with neuropathic corneal pain with the paradox that DED is a pain-free disease in most cases, although it is regarded as a pain condition. The current paper puts into perspective that one gateway from physiology to pathophysiology could be a Piezo2 channelopathy, opening the pathway to a potentially quad-phasic non-contact injury mechanism on a multifactorial basis and with a heterogeneous clinical picture. The primary non-contact injury phase could be the pain-free microinjury of the Piezo2 ion channel at the corneal somatosensory nerve terminal. The secondary non-contact injury phase involves harsher corneal tissue damage with C-fiber contribution due to the lost or inadequate intimate cross-talk between somatosensory Piezo2 and peripheral Piezo1. The third injury phase of this non-contact injury is the neuronal sensitization process with underlying repeated re-injury of the Piezo2, leading to the proposed chronic channelopathy. Notably, sensitization may evolve in certain cases in the absence of the second injury phase. Finally, the quadric injury phase is the lingering low-grade neuroinflammation associated with aging, called inflammaging. This quadric phase could clinically initiate or augment DED, explaining why increasing age is a risk factor. We highlight the potential role of the NGF-TrkA axis as a signaling mechanism that could further promote the microinjury of the corneal Piezo2 in a stress-derived hyperexcited state. The NGF-TrkA-Piezo2 axis might explain why female sex represents a risk factor for DED.


Subject(s)
Channelopathies , Dry Eye Syndromes , Ion Channels , Neuralgia , Sex Characteristics , Channelopathies/genetics , Channelopathies/physiopathology , Dry Eye Syndromes/genetics , Dry Eye Syndromes/physiopathology , Female , Humans , Ion Channels/genetics , Male , Nerve Growth Factor/genetics , Receptor, trkA/genetics
2.
Biochem Pharmacol ; 192: 114746, 2021 10.
Article in English | MEDLINE | ID: mdl-34461117

ABSTRACT

Long QT syndrome (LQTS), Brugada syndrome (BrS), and catecholaminergic polymorphic ventricular tachycardia (CPVT) are genetic diseases of the heart caused by mutations in specific cardiac ion channels and are characterized by paroxysmal arrhythmias, which can deteriorate into ventricular fibrillation. In LQTS3 and BrS different mutations in the SCN5A gene lead to a gain-or a loss-of-function of the voltage-gated sodium channel Nav1.5, respectively. Although sharing the same gene mutation, these syndromes are characterized by different clinical manifestations and functional perturbations and in some cases even present an overlapping clinical phenotype. Several studies have shown that Na+ current abnormalities in LQTS3 and BrS can also cause Ca2+-signaling aberrancies in cardiomyocytes (CMs). Abnormal Ca2+ homeostasis is also the main feature of CPVT which is mostly caused by heterozygous mutations in the RyR2 gene. Large numbers of disease-causing mutations were identified in RyR2 and SCN5A but it is not clear how different variants in the SCN5A gene produce different clinical syndromes and if in CPVT Ca2+ abnormalities and drug sensitivities vary depending on the mutation site in the RyR2. These questions can now be addressed by using patient-specific in vitro models of these diseases based on induced pluripotent stem cells (iPSCs). In this review, we summarize different insights gained from these models with a focus on electrophysiological perturbations caused by different ion channel mutations and discuss how will this knowledge help develop better stratification and more efficient personalized therapies for these patients.


Subject(s)
Channelopathies/genetics , Electrophysiological Phenomena/physiology , Heart Diseases/genetics , Induced Pluripotent Stem Cells/physiology , Mutation/genetics , Myocytes, Cardiac/physiology , Animals , Channelopathies/pathology , Channelopathies/physiopathology , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/pathology
3.
Nat Rev Neurosci ; 22(5): 275-289, 2021 05.
Article in English | MEDLINE | ID: mdl-33828309

ABSTRACT

Fragile X syndrome (FXS) is the most common inherited form of intellectual disability and the leading monogenic cause of autism. The condition stems from loss of fragile X mental retardation protein (FMRP), which regulates a wide range of ion channels via translational control, protein-protein interactions and second messenger pathways. Rapidly increasing evidence demonstrates that loss of FMRP leads to numerous ion channel dysfunctions (that is, channelopathies), which in turn contribute significantly to FXS pathophysiology. Consistent with this, pharmacological or genetic interventions that target dysregulated ion channels effectively restore neuronal excitability, synaptic function and behavioural phenotypes in FXS animal models. Recent studies further support a role for direct and rapid FMRP-channel interactions in regulating ion channel function. This Review lays out the current state of knowledge in the field regarding channelopathies and the pathogenesis of FXS, including promising therapeutic implications.


Subject(s)
Channelopathies/etiology , Channelopathies/physiopathology , Fragile X Syndrome/complications , Fragile X Syndrome/physiopathology , Animals , Channelopathies/genetics , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Humans
4.
Cells ; 10(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925740

ABSTRACT

The rough endoplasmic reticulum (ER) of nucleated human cells has crucial functions in protein biogenesis, calcium (Ca2+) homeostasis, and signal transduction. Among the roughly one hundred components, which are involved in protein import and protein folding or assembly, two components stand out: The Sec61 complex and BiP. The Sec61 complex in the ER membrane represents the major entry point for precursor polypeptides into the membrane or lumen of the ER and provides a conduit for Ca2+ ions from the ER lumen to the cytosol. The second component, the Hsp70-type molecular chaperone immunoglobulin heavy chain binding protein, short BiP, plays central roles in protein folding and assembly (hence its name), protein import, cellular Ca2+ homeostasis, and various intracellular signal transduction pathways. For the purpose of this review, we focus on these two components, their relevant allosteric effectors and on the question of how their respective functional cycles are linked in order to reconcile the apparently contradictory features of the ER membrane, selective permeability for precursor polypeptides, and impermeability for Ca2+. The key issues are that the Sec61 complex exists in two conformations: An open and a closed state that are in a dynamic equilibrium with each other, and that BiP contributes to its gating in both directions in cooperation with different co-chaperones. While the open Sec61 complex forms an aqueous polypeptide-conducting- and transiently Ca2+-permeable channel, the closed complex is impermeable even to Ca2+. Therefore, we discuss the human hereditary and tumor diseases that are linked to Sec61 channel gating, termed Sec61-channelopathies, as disturbances of selective polypeptide-impermeability and/or aberrant Ca2+-permeability.


Subject(s)
Channelopathies/metabolism , Channelopathies/physiopathology , Ion Channel Gating , SEC Translocation Channels/metabolism , Allosteric Regulation , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology
6.
Physiol Rev ; 101(4): 1633-1689, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33769100

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.


Subject(s)
Brain/physiopathology , Channelopathies/physiopathology , Muscle, Skeletal/physiopathology , Sodium Channels , Animals , Channelopathies/genetics , Humans , Mice , Nervous System Diseases/genetics , Nervous System Diseases/physiopathology , Sodium Channels/genetics
7.
Int J Mol Sci ; 22(3)2021 Jan 31.
Article in English | MEDLINE | ID: mdl-33572566

ABSTRACT

Ion channels are macromolecular complexes present in the plasma membrane and intracellular organelles of cells. Dysfunction of ion channels results in a group of disorders named channelopathies, which represent an extraordinary challenge for study and treatment. In this review, we will focus on voltage-gated potassium channels (KV), specifically on the KV4-family. The activation of these channels generates outward currents operating at subthreshold membrane potentials as recorded from myocardial cells (ITO, transient outward current) and from the somata of hippocampal neurons (ISA). In the heart, KV4 dysfunctions are related to Brugada syndrome, atrial fibrillation, hypertrophy, and heart failure. In hippocampus, KV4.x channelopathies are linked to schizophrenia, epilepsy, and Alzheimer's disease. KV4.x channels need to assemble with other accessory subunits (ß) to fully reproduce the ITO and ISA currents. ß Subunits affect channel gating and/or the traffic to the plasma membrane, and their dysfunctions may influence channel pharmacology. Among KV4 regulatory subunits, this review aims to analyze the KV4/KChIPs interaction and the effect of small molecule KChIP ligands in the A-type currents generated by the modulation of the KV4/KChIP channel complex. Knowledge gained from structural and functional studies using activators or inhibitors of the potassium current mediated by KV4/KChIPs will better help understand the underlying mechanism involving KV4-mediated-channelopathies, establishing the foundations for drug discovery, and hence their treatments.


Subject(s)
Alzheimer Disease/physiopathology , Channelopathies/physiopathology , Epilepsy/physiopathology , Kv Channel-Interacting Proteins/pharmacology , Potassium Channels, Voltage-Gated/pharmacology , Schizophrenia/physiopathology , Shal Potassium Channels/pharmacology , Alzheimer Disease/etiology , Amino Acid Sequence , Channelopathies/complications , Epilepsy/etiology , Heart/physiopathology , Hippocampus/metabolism , Hippocampus/physiopathology , Humans , Kv Channel-Interacting Proteins/genetics , Kv Channel-Interacting Proteins/metabolism , Membrane Potentials , Models, Molecular , Neurons/metabolism , Potassium Channels, Voltage-Gated/genetics , Potassium Channels, Voltage-Gated/metabolism , Schizophrenia/etiology , Sequence Alignment , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism
8.
Cardiovasc Ther ; 2020: 6615038, 2020.
Article in English | MEDLINE | ID: mdl-33381229

ABSTRACT

The Transient Receptor Potential Melastatin 4 (TRPM4) is a transmembrane N-glycosylated ion channel that belongs to the large family of TRP proteins. It has an equal permeability to Na+ and K+ and is activated via an increase of the intracellular calcium concentration and membrane depolarization. Due to its wide distribution, TRPM4 dysfunction has been linked with several pathophysiological processes, including inherited cardiac arrhythmias. Many pathogenic variants of the TRPM4 gene have been identified in patients with different forms of cardiac disorders such as conduction defects, Brugada syndrome, and congenital long QT syndrome. At the cellular level, these variants induce either gain- or loss-of-function of TRPM4 channels for similar clinical phenotypes. However, the molecular mechanisms associating these functional alterations to the clinical phenotypes remain poorly understood. The main objective of this article is to review the major cardiac TRPM4 channelopathies and recent advances regarding their genetic background and the underlying molecular mechanisms.


Subject(s)
Arrhythmias, Cardiac/genetics , Channelopathies/genetics , Heart Rate/genetics , TRPM Cation Channels/genetics , Animals , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/physiopathology , Brugada Syndrome/genetics , Brugada Syndrome/physiopathology , Channelopathies/complications , Channelopathies/physiopathology , Genetic Predisposition to Disease , Heredity , Humans , Long QT Syndrome/genetics , Long QT Syndrome/physiopathology , Pedigree , Phenotype
9.
J Comput Neurosci ; 48(4): 377-386, 2020 11.
Article in English | MEDLINE | ID: mdl-33063225

ABSTRACT

Channelopathies involving acquired or genetic modifications of the delayed rectifier K+ channel Kv1.1 include phenotypes characterized by enhanced neuronal excitability. Affected Kv1.1 channels exhibit combinations of altered expression, voltage sensitivity, and rates of activation and deactivation. Computational modeling and analysis can reveal the potential of particular channelopathies to alter neuronal excitability. A dynamical systems approach was taken to study the excitability and underlying dynamical structure of the Hodgkin-Huxley (HH) model of neural excitation as properties of the delayed rectifier K+ channel were altered. Bifurcation patterns of the HH model were determined as the amplitude of steady injection current was varied simultaneously with single parameters describing the delayed rectifier rates of activation and deactivation, maximal conductance, and voltage sensitivity. Relatively modest changes in the properties of the delayed rectifier K+ channel analogous to what is described for its channelopathies alter the bifurcation structure of the HH model and profoundly modify excitability of the HH model. Channelopathies associated with Kv1.1 can reduce the threshold for onset of neural activity. These studies also demonstrate how pathological delayed rectifier K+ channels could lead to the observation of the generalized Hopf bifurcation and, perhaps, other variants of the Hopf bifurcation. The observed bifurcation patterns collectively demonstrate that properties of the nominal delayed rectifier in the HH model appear optimized to permit activation of the HH model over the broadest possible range of input currents.


Subject(s)
Channelopathies/physiopathology , Delayed Rectifier Potassium Channels/genetics , Membrane Potentials/physiology , Models, Neurological , Neurons/physiology , Animals , Channelopathies/genetics , Computer Simulation
10.
Neurol Clin ; 38(3): 481-491, 2020 08.
Article in English | MEDLINE | ID: mdl-32703462

ABSTRACT

Skeletal muscle channelopathies are rare genetic neuromuscular conditions that include the nondystrophic myotonias and periodic paralyses. They cause disabling muscle symptoms and can limit educational potential, work opportunities, socialization, and quality of life. Effective therapy is available, making it essential to recognize and treat this group of disorders. Here, the authors highlight important aspects regarding diagnosis and management using illustrative case reports.


Subject(s)
Andersen Syndrome/diagnosis , Andersen Syndrome/genetics , Channelopathies/diagnosis , Channelopathies/genetics , Hypokalemic Periodic Paralysis/diagnosis , Hypokalemic Periodic Paralysis/genetics , Adolescent , Andersen Syndrome/physiopathology , Channelopathies/physiopathology , Humans , Hypokalemic Periodic Paralysis/physiopathology , Male , Muscle, Skeletal/physiopathology , Mutation/genetics , Myotonic Disorders/diagnosis , Myotonic Disorders/genetics , Myotonic Disorders/physiopathology , Neuromuscular Junction Diseases/diagnosis , Neuromuscular Junction Diseases/genetics , Neuromuscular Junction Diseases/physiopathology
11.
Neuromuscul Disord ; 30(7): 539-545, 2020 07.
Article in English | MEDLINE | ID: mdl-32622512

ABSTRACT

The skeletal muscle channelopathies are a group of rare diseases and include non-dystrophic myotonia and periodic paralysis. Given their rarity, little has been published on the management of anaesthesia and pregnancy in this cohort despite being important aspects of care. We have conducted a large study of over 70 patients who underwent anaesthesia and 87 pregnancies to investigate the problems encountered following anaesthesia or during pregnancy. This was performed via patient surveys sent out to genetically confirmed channelopathy patients seen at the National Hospital for Neurology and Neurosurgery. Most significantly in our cohort, patients frequently experienced a worsening or precipitation of symptoms during pregnancy (75%) or following anaesthetic (31%). None of our patients developed malignant hyperthermia, although there are confirmed reports of this in patients with periodic paralysis and mutations in RYR1. There was a significantly higher number of miscarriages compared to the normal population. There was no significant difference in antenatal or delivery complications compared to the general population. However, three neonates did have complications, all of whom were found to carry mutations in SCN4A. This study highlights the importance of counselling patients and clinicians for the possibility of worsening symptoms during pregnancy or anaesthesia and the careful management of neonates following delivery.


Subject(s)
Anesthetics/adverse effects , Channelopathies/physiopathology , Muscle, Skeletal/physiopathology , Pregnancy Complications/physiopathology , Female , Humans , Infant, Newborn , Male , Middle Aged , Myotonia/physiopathology , Pregnancy , Surveys and Questionnaires
12.
Adv Genet ; 105: 137-174, 2020.
Article in English | MEDLINE | ID: mdl-32560786

ABSTRACT

Potassium channels are a heterogeneous group of membrane-bound proteins, whose functions support a diverse range of biological processes. Genetic disorders arising from mutations in potassium channels are classically recognized by symptoms arising from acute channel dysfunction, such as periodic paralysis, ataxia, seizures, or cardiac conduction abnormalities, often in a patient with otherwise normal examination findings. In this chapter, we review a distinct subgroup of rare potassium channelopathies whose presentations are instead suggestive of a developmental disorder, with features including intellectual disability, craniofacial dysmorphism or other physical anomalies. Known conditions within this subgroup are: Andersen-Tawil syndrome, Birk-Barel syndrome, Cantú syndrome, Keppen-Lubinsky syndrome, Temple-Baraitser syndrome, Zimmerman-Laband syndrome and a very similar disorder called Bauer-Tartaglia or FHEIG syndrome. Ion channelopathies are unlikely to be routinely considered in the differential diagnosis of children presenting with developmental concerns, and so detailed description and photographs of the clinical phenotype are provided to aid recognition. For several of these disorders, functional characterization of the genetic mutations responsible has led to identification of candidate therapies, including drugs already commonly used for other indications, which adds further impetus to their prompt recognition. Together, these cases illustrate the potential for mechanistic insights gained from genetic diagnosis to drive translational work toward targeted, disease-modifying therapies for rare disorders.


Subject(s)
Abnormalities, Multiple/genetics , Andersen Syndrome/genetics , Cardiomegaly/genetics , Channelopathies/genetics , Craniofacial Abnormalities/genetics , Fibromatosis, Gingival/genetics , Hallux/abnormalities , Hand Deformities, Congenital/genetics , Hypertrichosis/genetics , Intellectual Disability/genetics , Muscle Hypotonia/genetics , Nails, Malformed/genetics , Osteochondrodysplasias/genetics , Potassium Channels/genetics , Thumb/abnormalities , Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/pathology , Abnormalities, Multiple/physiopathology , Andersen Syndrome/drug therapy , Andersen Syndrome/pathology , Andersen Syndrome/physiopathology , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Channelopathies/drug therapy , Channelopathies/metabolism , Channelopathies/physiopathology , Child , Craniofacial Abnormalities/drug therapy , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/physiopathology , Fibromatosis, Gingival/drug therapy , Fibromatosis, Gingival/pathology , Fibromatosis, Gingival/physiopathology , Hallux/pathology , Hallux/physiopathology , Hand Deformities, Congenital/drug therapy , Hand Deformities, Congenital/pathology , Hand Deformities, Congenital/physiopathology , Humans , Hypertrichosis/drug therapy , Hypertrichosis/pathology , Hypertrichosis/physiopathology , Intellectual Disability/drug therapy , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Muscle Hypotonia/drug therapy , Muscle Hypotonia/pathology , Muscle Hypotonia/physiopathology , Nails, Malformed/drug therapy , Nails, Malformed/pathology , Nails, Malformed/physiopathology , Osteochondrodysplasias/drug therapy , Osteochondrodysplasias/pathology , Osteochondrodysplasias/physiopathology , Potassium Channels/metabolism , Thumb/pathology , Thumb/physiopathology
13.
Int J Mol Sci ; 21(7)2020 Apr 08.
Article in English | MEDLINE | ID: mdl-32276507

ABSTRACT

Myotonia congenita (MC) is a rare disorder characterized by stiffness and weakness of the limb and trunk muscles. Mutations in the SCN4A gene encoding the alpha-subunit of the voltage-gated sodium channel Nav1.4 have been reported to be responsible for sodium channel myotonia (SCM). The Nav1.4 channel is expressed in skeletal muscles, and its related channelopathies affect skeletal muscle excitability, which can manifest as SCM, paramyotonia and periodic paralysis. In this study, the missense mutation p.V445M was identified in two individual families with MC. To determine the functional consequences of having a mutated Nav1.4 channel, whole-cell patch-clamp recording of transfected Chinese hamster ovary cells was performed. Evaluation of the transient Na+ current found that a hyperpolarizing shift occurs at both the activation and inactivation curves with an increase of the window currents in the mutant channels. The Nav1.4 channel's co-expression with the Navß4 peptide can generate resurgent Na+ currents at repolarization following a depolarization. The magnitude of the resurgent currents is higher in the mutant than in the wild-type (WT) channel. Although the decay kinetics are comparable between the mutant and WT channels, the time to the peak of resurgent Na+ currents in the mutant channel is significantly protracted compared with that in the WT channel. These findings suggest that the p.V445M mutation in the Nav1.4 channel results in an increase of both sustained and resurgent Na+ currents, which may contribute to hyperexcitability with repetitive firing and is likely to facilitate recurrent myotonia in SCM patients.


Subject(s)
Mutation, Missense , Myotonia Congenita/genetics , Myotonia Congenita/physiopathology , NAV1.4 Voltage-Gated Sodium Channel/physiology , Amino Acid Sequence , Animals , Asian People , CHO Cells , Channelopathies/genetics , Channelopathies/metabolism , Channelopathies/physiopathology , Cricetulus , Female , Humans , Male , Myotonia Congenita/metabolism , NAV1.4 Voltage-Gated Sodium Channel/chemistry , NAV1.4 Voltage-Gated Sodium Channel/genetics , NAV1.4 Voltage-Gated Sodium Channel/metabolism , Patch-Clamp Techniques , Pedigree
14.
Mol Autism ; 11(1): 23, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32264956

ABSTRACT

Efforts to identify the causes of autism spectrum disorders have highlighted the importance of both genetics and environment, but the lack of human models for many of these disorders limits researchers' attempts to understand the mechanisms of disease and to develop new treatments. Induced pluripotent stem cells offer the opportunity to study specific genetic and environmental risk factors, but the heterogeneity of donor genetics may obscure important findings. Diseases associated with unusually high rates of autism, such as SCN2A syndromes, provide an opportunity to study specific mutations with high effect sizes in a human genetic context and may reveal biological insights applicable to more common forms of autism. Loss-of-function mutations in the SCN2A gene, which encodes the voltage-gated sodium channel NaV1.2, are associated with autism rates up to 50%. Here, we review the findings from experimental models of SCN2A syndromes, including mouse and human cell studies, highlighting the potential role for patient-derived induced pluripotent stem cell technology to identify the molecular and cellular substrates of autism.


Subject(s)
Autism Spectrum Disorder/physiopathology , Channelopathies/physiopathology , NAV1.2 Voltage-Gated Sodium Channel/physiology , Pluripotent Stem Cells , Animals , Humans
15.
Cardiovasc Res ; 116(9): 1557-1570, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32251506

ABSTRACT

The cardiac sodium channel NaV1.5, encoded by the SCN5A gene, is responsible for the fast upstroke of the action potential. Mutations in SCN5A may cause sodium channel dysfunction by decreasing peak sodium current, which slows conduction and facilitates reentry-based arrhythmias, and by enhancing late sodium current, which prolongs the action potential and sets the stage for early afterdepolarization and arrhythmias. Yet, some NaV1.5-related disorders, in particular structural abnormalities, cannot be directly or solely explained on the basis of defective NaV1.5 expression or biophysics. An emerging concept that may explain the large disease spectrum associated with SCN5A mutations centres around the multifunctionality of the NaV1.5 complex. In this alternative view, alterations in NaV1.5 affect processes that are independent of its canonical ion-conducting role. We here propose a novel classification of NaV1.5 (dys)function, categorized into (i) direct ionic effects of sodium influx through NaV1.5 on membrane potential and consequent action potential generation, (ii) indirect ionic effects of sodium influx on intracellular homeostasis and signalling, and (iii) non-ionic effects of NaV1.5, independent of sodium influx, through interactions with macromolecular complexes within the different microdomains of the cardiomyocyte. These indirect ionic and non-ionic processes may, acting alone or in concert, contribute significantly to arrhythmogenesis. Hence, further exploration of these multifunctional effects of NaV1.5 is essential for the development of novel preventive and therapeutic strategies.


Subject(s)
Action Potentials , Arrhythmias, Cardiac/metabolism , Channelopathies/metabolism , Heart Rate , Myocytes, Cardiac/metabolism , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/therapeutic use , Arrhythmias, Cardiac/drug therapy , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/physiopathology , Channelopathies/drug therapy , Channelopathies/genetics , Channelopathies/physiopathology , Genetic Predisposition to Disease , Heart Rate/drug effects , Heredity , Humans , Mutation , Myocytes, Cardiac/drug effects , NAV1.5 Voltage-Gated Sodium Channel/drug effects , NAV1.5 Voltage-Gated Sodium Channel/genetics , Phenotype , Risk Factors , Voltage-Gated Sodium Channel Blockers/therapeutic use
16.
Annu Rev Pharmacol Toxicol ; 60: 133-154, 2020 01 06.
Article in English | MEDLINE | ID: mdl-31537174

ABSTRACT

Voltage-gated sodium and calcium channels are evolutionarily related transmembrane signaling proteins that initiate action potentials, neurotransmission, excitation-contraction coupling, and other physiological processes. Genetic or acquired dysfunction of these proteins causes numerous diseases, termed channelopathies, and sodium and calcium channels are the molecular targets for several major classes of drugs. Recent advances in the structural biology of these proteins using X-ray crystallography and cryo-electron microscopy have given new insights into the molecular basis for their function and pharmacology. Here we review this recent literature and integrate findings on sodium and calcium channels to reveal the structural basis for their voltage-dependent activation, fast and slow inactivation, ion conductance and selectivity, and complex pharmacology at the atomic level. We conclude with the theme that new understanding of the diseases and therapeutics of these channels will be derived from application of the emerging structural principles from these recent structural analyses.


Subject(s)
Calcium Channels/drug effects , Channelopathies/drug therapy , Voltage-Gated Sodium Channels/drug effects , Calcium Channels/chemistry , Calcium Channels/metabolism , Channelopathies/physiopathology , Cryoelectron Microscopy , Crystallography, X-Ray , Humans , Molecular Targeted Therapy , Voltage-Gated Sodium Channels/chemistry , Voltage-Gated Sodium Channels/metabolism
17.
Physiol Rev ; 100(2): 725-803, 2020 04 01.
Article in English | MEDLINE | ID: mdl-31670612

ABSTRACT

The transient receptor potential ankyrin (TRPA) channels are Ca2+-permeable nonselective cation channels remarkably conserved through the animal kingdom. Mammals have only one member, TRPA1, which is widely expressed in sensory neurons and in non-neuronal cells (such as epithelial cells and hair cells). TRPA1 owes its name to the presence of 14 ankyrin repeats located in the NH2 terminus of the channel, an unusual structural feature that may be relevant to its interactions with intracellular components. TRPA1 is primarily involved in the detection of an extremely wide variety of exogenous stimuli that may produce cellular damage. This includes a plethora of electrophilic compounds that interact with nucleophilic amino acid residues in the channel and many other chemically unrelated compounds whose only common feature seems to be their ability to partition in the plasma membrane. TRPA1 has been reported to be activated by cold, heat, and mechanical stimuli, and its function is modulated by multiple factors, including Ca2+, trace metals, pH, and reactive oxygen, nitrogen, and carbonyl species. TRPA1 is involved in acute and chronic pain as well as inflammation, plays key roles in the pathophysiology of nearly all organ systems, and is an attractive target for the treatment of related diseases. Here we review the current knowledge about the mammalian TRPA1 channel, linking its unique structure, widely tuned sensory properties, and complex regulation to its roles in multiple pathophysiological conditions.


Subject(s)
Calcium Signaling , Mechanotransduction, Cellular , Nociception , Sensory Receptor Cells/metabolism , TRPA1 Cation Channel/metabolism , Thermosensing , Animals , Channelopathies/metabolism , Channelopathies/physiopathology , Chemoreceptor Cells/metabolism , Humans , Inflammation/metabolism , Inflammation/physiopathology , Mechanoreceptors/metabolism , Nociceptors/metabolism , Pain/metabolism , Pain/physiopathology , Thermoreceptors/metabolism
18.
Neurol Sci ; 41(4): 749-761, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31838630

ABSTRACT

Genetic brain channelopathies result from inherited or de novo mutations of genes encoding ion channel subunits within the central nervous system. Most neurological channelopathies arise in childhood with paroxysmal or episodic symptoms, likely because of a transient impairment of homeostatic mechanisms regulating membrane excitability, and the prototypical expression of this impairment is epilepsy. Migraine, episodic ataxia and alternating hemiplegia can also occur, as well as chronic phenotypes, such as spinocerebellar ataxias, intellectual disability and autism spectrum disorder. Voltage-gated and ligand-gated channels may be involved. In most cases, a single gene may be associated with a phenotypical spectrum that shows variable expressivity. Different clinical features may arise at different ages and the adult phenotype may be remarkably modified from the syndrome onset in childhood or adolescence. Recognizing the prominent phenotypical traits of brain channelopathies is essential to perform appropriate diagnostic investigations and to provide the better care not only in the paediatric setting but also for adult patients and their caregivers. Herein, we provide an overview of genetic brain channelopathies associated with epilepsy, highlight the different molecular mechanisms and describe the different clinical characteristics which may prompt the clinician to suspect specific syndromes and to possibly establish tailored treatments.


Subject(s)
Channelopathies , Epilepsy , Adult , Channelopathies/complications , Channelopathies/genetics , Channelopathies/physiopathology , Child , Epilepsy/etiology , Epilepsy/genetics , Epilepsy/physiopathology , Humans , Infant
19.
Clin Neurophysiol ; 131(1): 145-154, 2020 01.
Article in English | MEDLINE | ID: mdl-31765978

ABSTRACT

OBJECTIVE: Corneal confocal microscopy (CCM) has been identified as a non-invasive technique to assess corneal nerve fiber morphology. It is not known how corneal nerve changes relate to measures of peripheral nerve function in diabetic peripheral neuropathy (DPN). The present study investigates the relationship between nerve structure and function in DPN. METHODS: Fifty participants with type 1 diabetes (T1DM) and 29 healthy controls underwent CCM to assess corneal nerve fiber density (CNFD), branch density (CNBD), fiber length (CNFL), total branch density (CTBD), nerve fractal dimension (CNFrD) and inferior whorl length (IWL). The severity of DPN was assessed as Total Neuropathy Score (TNS). Motor nerve axonal excitability tests were conducted to assess axonal function. RESULTS: Significant correlations were noted between CNFD (rho = -0.783; P < 0.01) or superexcitability (rho = 0.435; P < 0.01) and TNS. CNFrD was significantly correlated with peak response to stimulus (r = 0.414; P < 0.01) and superexcitability (r = -0.467; P < 0.01) measurements. CONCLUSION: Corneal nerve loss demonstrates a significant association with axonal ion channel dysfunction in T1DM. SIGNIFICANCE: Detection of altered corneal nerve morphology may lead to the earlier diagnosis of DPN.


Subject(s)
Axons/physiology , Channelopathies/physiopathology , Cornea/innervation , Diabetes Mellitus, Type 1/physiopathology , Diabetic Neuropathies/physiopathology , Action Potentials/physiology , Adult , Analysis of Variance , Axons/pathology , Case-Control Studies , Channelopathies/diagnostic imaging , Cornea/diagnostic imaging , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/diagnosis , Diabetic Neuropathies/diagnostic imaging , Electric Stimulation/methods , Female , Humans , Male , Median Nerve/physiopathology , Microscopy, Confocal/methods , Middle Aged , Models, Neurological , Nerve Fibers/pathology , Nerve Fibers/physiology , Neural Conduction/physiology , Statistics, Nonparametric , Sural Nerve/physiopathology , Tibial Nerve/physiopathology
20.
J Clin Neurophysiol ; 37(3): 231-238, 2020 May.
Article in English | MEDLINE | ID: mdl-31567646

ABSTRACT

PURPOSE: Hereditary skeletal muscle channelopathies are characterized by muscle stiffness and/or periodic muscle weakness because of different gene mutations. The objective of this study was to investigate the clinical and electromyographic phenotypes in Chinese patients with different skeletal ion channel mutations. METHODS: The electromyographic results of 61 Chinese patients with skeletal muscle channelopathies were retrospectively reviewed and the differential features were characterized. RESULTS: Myotonic discharges were in patients with chloride voltage-gated channel 1 and sodium voltage-gated channel alpha subunit 4 mutations. Subclinical myotonia was identified in four patients with hypokalemic periodic paralysis because of sodium voltage-gated channel alpha subunit 4 mutations. Patients with potassium voltage-gated channel subfamily J member 2 mutations had an early decline after exercise (5.7 ± 4.9 minutes) and patients with calcium voltage-gated channel subunit alpha 1S mutations have a relatively lower baseline amplitude (4.6 ± 2 mV). Specific patterns were characterized in patients with Becker disease and paramyotonia congenital after short exercise. CONCLUSIONS: Myotonic discharges help to discriminate chloride and sodium from other channelopathies. Early decline and low baseline compound motor action potential amplitude in long exercise test are significant in patients with potassium voltage-gated channel subfamily J member 2 and calcium voltage-gated channel subunit alpha 1S mutations, respectively. Electromyographic patterns in the electromyography study and exercise test may help in better providing the comprehensive picture for patients with primary skeletal muscle channelopathies.


Subject(s)
Channelopathies/diagnosis , Channelopathies/physiopathology , Electromyography/methods , Exercise Test/methods , Action Potentials/physiology , Adolescent , Adult , Aged , Channelopathies/genetics , Child , Cohort Studies , Female , Humans , Male , Middle Aged , Mutation , Retrospective Studies , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...