Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.111
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 33: 291-353, 2015.
Article in English | MEDLINE | ID: mdl-25861976

ABSTRACT

Ion channels and transporters mediate the transport of charged ions across hydrophobic lipid membranes. In immune cells, divalent cations such as calcium, magnesium, and zinc have important roles as second messengers to regulate intracellular signaling pathways. By contrast, monovalent cations such as sodium and potassium mainly regulate the membrane potential, which indirectly controls the influx of calcium and immune cell signaling. Studies investigating human patients with mutations in ion channels and transporters, analysis of gene-targeted mice, or pharmacological experiments with ion channel inhibitors have revealed important roles of ionic signals in lymphocyte development and in innate and adaptive immune responses. We here review the mechanisms underlying the function of ion channels and transporters in lymphocytes and innate immune cells and discuss their roles in lymphocyte development, adaptive and innate immune responses, and autoimmunity, as well as recent efforts to develop pharmacological inhibitors of ion channels for immunomodulatory therapy.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Ion Channels/metabolism , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/metabolism , Immunotherapy/methods , Ion Channels/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Molecular Targeted Therapy , Mutation , Signal Transduction
2.
Annu Rev Biochem ; 90: 507-534, 2021 06 20.
Article in English | MEDLINE | ID: mdl-34153212

ABSTRACT

Mechanosensation is the ability to detect dynamic mechanical stimuli (e.g., pressure, stretch, and shear stress) and is essential for a wide variety of processes, including our sense of touch on the skin. How touch is detected and transduced at the molecular level has proved to be one of the great mysteries of sensory biology. A major breakthrough occurred in 2010 with the discovery of a family of mechanically gated ion channels that were coined PIEZOs. The last 10 years of investigation have provided a wealth of information about the functional roles and mechanisms of these molecules. Here we focus on PIEZO2, one of the two PIEZO proteins found in humans and other mammals. We review how work at the molecular, cellular, and systems levels over the past decade has transformed our understanding of touch and led to unexpected insights into other types of mechanosensation beyond the skin.


Subject(s)
Drug Discovery/methods , Ion Channels/chemistry , Ion Channels/physiology , Mechanotransduction, Cellular/physiology , Animals , Baroreflex/physiology , Humans , Ion Channels/genetics , Ion Channels/metabolism , Mice , Proprioception/physiology , Stem Cells/physiology , Touch
3.
Cell ; 184(4): 856-858, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33606984

ABSTRACT

In this issue of Cell, Ma et al. reveal a mechanistic role for PIEZO1 in iron homeostasis through molecular genetic mouse studies. They also demonstrate implications for human iron overload and deficiency syndromes, susceptibility to malarial infection, and red blood cell turnover in persons of African ancestries.


Subject(s)
Iron , Malaria , Animals , Erythrocytes , Homeostasis , Humans , Ion Channels/genetics , Mice
4.
Cell ; 181(4): 760-762, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32413297

ABSTRACT

Skin and other epithelial cell layers are frequently subjected to extensive deformations, yet sustain such mechanical stress without damage. In this issue of Cell, Nava and colleagues show that stretch induces rapid loss of heterochromatin that leads to transient softening of the nucleus, which, together with long-term cytoskeletal and supracellular rearrangements, protects nuclei from DNA damage.


Subject(s)
Heterochromatin , Ion Channels , Cell Nucleus , Ion Channels/genetics , Stress, Mechanical , Time
5.
Cell ; 182(3): 542-544, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32763186

ABSTRACT

Serotonin production by enterochromaffin cells (ECs) is microbiota-dependent, but the mechanism of this is unknown. In this issue of Cell, Sugisawa et al. demonstrate that Piezo1 in ECs senses single-strand RNA (ssRNA) from intestinal microbiota to promote serotonin production. Deletion of Piezo1 in intestinal epithelium promotes bone formation, decreases peristalsis, and protects from colitis because of decreased serotonin.


Subject(s)
Gastrointestinal Microbiome , Serotonin , Enterochromaffin Cells , Ion Channels/genetics , RNA
6.
Cell ; 180(5): 956-967.e17, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32084332

ABSTRACT

Mechanotransduction, the conversion of mechanical stimuli into electrical signals, is a fundamental process underlying essential physiological functions such as touch and pain sensing, hearing, and proprioception. Although the mechanisms for some of these functions have been identified, the molecules essential to the sense of pain have remained elusive. Here we report identification of TACAN (Tmem120A), an ion channel involved in sensing mechanical pain. TACAN is expressed in a subset of nociceptors, and its heterologous expression increases mechanically evoked currents in cell lines. Purification and reconstitution of TACAN in synthetic lipids generates a functional ion channel. Finally, a nociceptor-specific inducible knockout of TACAN decreases the mechanosensitivity of nociceptors and reduces behavioral responses to painful mechanical stimuli but not to thermal or touch stimuli. We propose that TACAN is an ion channel that contributes to sensing mechanical pain.


Subject(s)
Ion Channels/physiology , Mechanotransduction, Cellular/genetics , Nociceptors/metabolism , Pain/genetics , Touch/genetics , Animals , Gene Expression Regulation/genetics , Humans , Ion Channels/genetics , Lipids/genetics , Mice , Mice, Knockout , Pain/physiopathology , Patch-Clamp Techniques , Stress, Mechanical , Touch/physiology
7.
Cell ; 182(3): 609-624.e21, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32640190

ABSTRACT

Gastrointestinal enterochromaffin cells regulate bone and gut homeostasis via serotonin (5-hydroxytryptamine [5-HT]) production. A recent report suggested that gut microbes regulate 5-HT levels; however, the precise underlying molecular mechanisms are unexplored. Here, we reveal that the cation channel Piezo1 in the gut acts as a sensor of single-stranded RNA (ssRNA) governing 5-HT production. Intestinal epithelium-specific deletion of mouse Piezo1 profoundly disturbed gut peristalsis, impeded experimental colitis, and suppressed serum 5-HT levels. Because of systemic 5-HT deficiency, conditional knockout of Piezo1 increased bone formation. Notably, fecal ssRNA was identified as a natural Piezo1 ligand, and ssRNA-stimulated 5-HT synthesis from the gut was evoked in a MyD88/TRIF-independent manner. Colonic infusion of RNase A suppressed gut motility and increased bone mass. These findings suggest gut ssRNA as a master determinant of systemic 5-HT levels, indicating the ssRNA-Piezo1 axis as a potential prophylactic target for treatment of bone and gut disorders.


Subject(s)
Bone and Bones/metabolism , Colon/metabolism , Gastrointestinal Motility/genetics , Ion Channels/metabolism , RNA/metabolism , Serotonin/biosynthesis , Serotonin/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Bone and Bones/cytology , Calcium/metabolism , Colitis/genetics , Colitis/metabolism , Colitis/prevention & control , Colon/physiology , Feces/chemistry , Female , Gastrointestinal Motility/physiology , HEK293 Cells , Humans , Immunohistochemistry , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Ion Channels/genetics , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/drug effects , Myeloid Differentiation Factor 88/metabolism , Osteoclasts/metabolism , Pyrazines/pharmacology , RNA/pharmacology , Ribonuclease, Pancreatic/administration & dosage , Serotonin/blood , Serotonin/deficiency , Thiadiazoles/pharmacology
8.
Immunity ; 57(1): 52-67.e10, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38091995

ABSTRACT

The regulation of polymorphonuclear leukocyte (PMN) function by mechanical forces encountered during their migration across restrictive endothelial cell junctions is not well understood. Using genetic, imaging, microfluidic, and in vivo approaches, we demonstrated that the mechanosensor Piezo1 in PMN plasmalemma induced spike-like Ca2+ signals during trans-endothelial migration. Mechanosensing increased the bactericidal function of PMN entering tissue. Mice in which Piezo1 in PMNs was genetically deleted were defective in clearing bacteria, and their lungs were predisposed to severe infection. Adoptive transfer of Piezo1-activated PMNs into the lungs of Pseudomonas aeruginosa-infected mice or exposing PMNs to defined mechanical forces in microfluidic systems improved bacterial clearance phenotype of PMNs. Piezo1 transduced the mechanical signals activated during transmigration to upregulate nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 4, crucial for the increased PMN bactericidal activity. Thus, Piezo1 mechanosensing of increased PMN tension, while traversing the narrow endothelial adherens junctions, is a central mechanism activating the host-defense function of transmigrating PMNs.


Subject(s)
Cell Movement , Lung , Mechanotransduction, Cellular , Neutrophils , Animals , Mice , Cell Membrane , Ion Channels/genetics , Neutrophils/metabolism , Neutrophils/microbiology , Blood Bactericidal Activity/genetics , Mechanotransduction, Cellular/genetics
9.
Cell ; 173(2): 443-455.e12, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29576450

ABSTRACT

Hereditary xerocytosis is thought to be a rare genetic condition characterized by red blood cell (RBC) dehydration with mild hemolysis. RBC dehydration is linked to reduced Plasmodium infection in vitro; however, the role of RBC dehydration in protection against malaria in vivo is unknown. Most cases of hereditary xerocytosis are associated with gain-of-function mutations in PIEZO1, a mechanically activated ion channel. We engineered a mouse model of hereditary xerocytosis and show that Plasmodium infection fails to cause experimental cerebral malaria in these mice due to the action of Piezo1 in RBCs and in T cells. Remarkably, we identified a novel human gain-of-function PIEZO1 allele, E756del, present in a third of the African population. RBCs from individuals carrying this allele are dehydrated and display reduced Plasmodium infection in vitro. The existence of a gain-of-function PIEZO1 at such high frequencies is surprising and suggests an association with malaria resistance.


Subject(s)
Anemia, Hemolytic, Congenital/pathology , Black People/genetics , Hydrops Fetalis/pathology , Ion Channels/genetics , Malaria/pathology , Alleles , Anemia, Hemolytic, Congenital/genetics , Animals , Dehydration , Disease Models, Animal , Erythrocytes/cytology , Erythrocytes/metabolism , Gene Deletion , Genotype , Humans , Hydrops Fetalis/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/deficiency , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Ion Channels/chemistry , Malaria/genetics , Malaria/parasitology , Malaria/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Plasmodium berghei/growth & development , Plasmodium berghei/pathogenicity , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
10.
Cell ; 170(3): 594-594.e1, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28753432

ABSTRACT

Ion channel families are broadly classified into three types according to their major mechanisms of activation. This SnapShot highlights features of these three classes and conveys general modes of channel regulation. To view this SnapShot, open or download the PDF.


Subject(s)
Ion Channel Gating , Ion Channels/genetics , Ion Channels/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation , Humans , Ion Channels/chemistry
11.
Genes Dev ; 38(1-2): 31-45, 2024 02 13.
Article in English | MEDLINE | ID: mdl-38242633

ABSTRACT

Bacterial spores can remain dormant for decades yet rapidly germinate and resume growth in response to nutrients. GerA family receptors that sense and respond to these signals have recently been shown to oligomerize into nutrient-gated ion channels. Ion release initiates exit from dormancy. Here, we report that a distinct ion channel, composed of SpoVAF (5AF) and its newly discovered partner protein, YqhR (FigP), amplifies the response. At high germinant concentrations, 5AF/FigP accelerate germination; at low concentrations, this complex becomes critical for exit from dormancy. 5AF is homologous to the channel-forming subunit of GerA family receptors and is predicted to oligomerize around a central pore. 5AF mutations predicted to widen the channel cause constitutive germination during spore formation and membrane depolarization in vegetative cells. Narrow-channel mutants are impaired in germination. A screen for suppressors of a constitutively germinating 5AF mutant identified FigP as an essential cofactor of 5AF activity. We demonstrate that 5AF and FigP interact and colocalize with GerA family receptors in spores. Finally, we show that 5AF/FigP accelerate germination in B. subtilis spores that have nutrient receptors from another species. Our data support a model in which nutrient-triggered ion release by GerA family receptors activates 5AF/FigP ion release, amplifying the response to germinant signals.


Subject(s)
Bacillus subtilis , Membrane Proteins , Bacillus subtilis/genetics , Bacillus subtilis/metabolism , Membrane Proteins/genetics , Spores, Bacterial/genetics , Spores, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ion Channels/genetics , Ion Channels/metabolism
12.
Nat Immunol ; 20(11): 1506-1516, 2019 11.
Article in English | MEDLINE | ID: mdl-31611698

ABSTRACT

Fibroblastic reticular cells (FRCs) and their specialized collagen fibers termed 'conduits' form fundamental structural units supporting lymphoid tissues. In lymph nodes, conduits are known to transport interstitial fluid and small molecules from afferent lymphatics into the nodal parenchyma. However, the immunological contributions of conduit function have remained elusive. Here, we report that intestinal Peyer's patches (PPs) contain a specialized conduit system that directs the flow of water absorbed across the intestinal epithelium. Notably, PP FRCs responded to conduit fluid flow via the mechanosensitive ion channel Piezo1. Disruption of fluid flow or genetic deficiency of Piezo1 on CCL19-expressing stroma led to profound structural alterations in perivascular FRCs and associated high endothelial venules. This in turn impaired lymphocyte entry into PPs and initiation of mucosal antibody responses. These results identify a critical role for conduit-mediated fluid flow in the maintenance of PP homeostasis and mucosal immunity.


Subject(s)
Immunity, Mucosal , Intestinal Mucosa/immunology , Lymphocytes/immunology , Mechanotransduction, Cellular/immunology , Peyer's Patches/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Cell Movement/immunology , Chemokine CCL19/metabolism , Female , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Intestine, Small/metabolism , Ion Channels/genetics , Ion Channels/metabolism , Lymphocyte Activation , Lymphocytes/metabolism , Male , Mice , Mice, Knockout , Models, Animal , Peyer's Patches/metabolism , Water/metabolism
13.
Mol Cell ; 83(23): 4386-4397.e9, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37995686

ABSTRACT

The multi-pass transmembrane protein ACCELERATED CELL DEATH 6 (ACD6) is an immune regulator in Arabidopsis thaliana with an unclear biochemical mode of action. We have identified two loci, MODULATOR OF HYPERACTIVE ACD6 1 (MHA1) and its paralog MHA1-LIKE (MHA1L), that code for ∼7 kDa proteins, which differentially interact with specific ACD6 variants. MHA1L enhances the accumulation of an ACD6 complex, thereby increasing the activity of the ACD6 standard allele for regulating plant growth and defenses. The intracellular ankyrin repeats of ACD6 are structurally similar to those found in mammalian ion channels. Several lines of evidence link increased ACD6 activity to enhanced calcium influx, with MHA1L as a direct regulator of ACD6, indicating that peptide-regulated ion channels are not restricted to animals.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Ankyrins/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Death , Ion Channels/genetics , Ion Channels/metabolism , Plant Immunity/genetics
14.
Cell ; 162(4): 836-48, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26276633

ABSTRACT

Circadian clocks regulate membrane excitability in master pacemaker neurons to control daily rhythms of sleep and wake. Here, we find that two distinctly timed electrical drives collaborate to impose rhythmicity on Drosophila clock neurons. In the morning, a voltage-independent sodium conductance via the NA/NALCN ion channel depolarizes these neurons. This current is driven by the rhythmic expression of NCA localization factor-1, linking the molecular clock to ion channel function. In the evening, basal potassium currents peak to silence clock neurons. Remarkably, daily antiphase cycles of sodium and potassium currents also drive mouse clock neuron rhythms. Thus, we reveal an evolutionarily ancient strategy for the neural mechanisms that govern daily sleep and wake.


Subject(s)
Circadian Clocks , Circadian Rhythm , Drosophila/physiology , Animals , Biological Clocks , Cell Membrane/metabolism , Drosophila/cytology , Drosophila Proteins/metabolism , Gene Knockdown Techniques , Ion Channels/genetics , Ion Channels/metabolism , Membrane Proteins , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism
15.
Annu Rev Neurosci ; 44: 383-402, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34236889

ABSTRACT

Nearly all structures in our body experience mechanical forces. At a molecular scale, these forces are detected by ion channels that function as mechanotransducers converting physical forces into electrochemical responses. Here we focus on PIEZOs, a family of mechanically activated ion channels comprising PIEZO1 and PIEZO2. The significance of these channels is highlighted by their roles in touch and pain sensation as well as in cardiovascular and respiratory physiology, among others. Moreover, mutations in PIEZOs cause somatosensory, proprioceptive, and blood disorders. The goal here is to present the diverse physiology and pathophysiology of these unique channels, discuss ongoing research and critical gaps in the field, and explore the pharmaceutical interest in targeting PIEZOs for therapeutic development.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Ion Channels/genetics , Pain Perception
16.
Nat Rev Mol Cell Biol ; 18(12): 771-783, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28974772

ABSTRACT

Cellular mechanotransduction, the process of translating mechanical forces into biological signals, is crucial for a wide range of physiological processes. A role for ion channels in sensing mechanical forces has been proposed for decades, but their identity in mammals remained largely elusive until the discovery of Piezos. Recent research on Piezos has underscored their importance in somatosensation (touch perception, proprioception and pulmonary respiration), red blood cell volume regulation, vascular physiology and various human genetic disorders.


Subject(s)
Genetic Diseases, Inborn/metabolism , Ion Channel Gating , Ion Channels/metabolism , Proprioception , Respiratory Mechanics , Touch Perception , Animals , Genetic Diseases, Inborn/genetics , Humans , Ion Channels/genetics
17.
Cell ; 157(3): 664-75, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24746027

ABSTRACT

Sensory systems for detecting tactile stimuli have evolved from touch-sensing nerves in invertebrates to complicated tactile end organs in mammals. Merkel discs are tactile end organs consisting of Merkel cells and Aß-afferent nerve endings and are localized in fingertips, whisker hair follicles, and other touch-sensitive spots. Merkel discs transduce touch into slowly adapting impulses to enable tactile discrimination, but their transduction and encoding mechanisms remain unknown. Using rat whisker hair follicles, we show that Merkel cells rather than Aß-afferent nerve endings are primary sites of tactile transduction and identify the Piezo2 ion channel as the Merkel cell mechanical transducer. Piezo2 transduces tactile stimuli into Ca(2+)-action potentials in Merkel cells, which drive Aß-afferent nerve endings to fire slowly adapting impulses. We further demonstrate that Piezo2 and Ca(2+)-action potentials in Merkel cells are required for behavioral tactile responses. Our findings provide insights into how tactile end-organs function and have clinical implications for tactile dysfunctions.


Subject(s)
Ion Channels/metabolism , Merkel Cells/metabolism , Touch , Vibrissae/cytology , Vibrissae/physiology , Action Potentials , Animals , Calcium/metabolism , Gene Knockdown Techniques , Ion Channels/genetics , Mechanoreceptors/metabolism , Mechanotransduction, Cellular , Rats
18.
Cell ; 158(1): 69-83, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24995979

ABSTRACT

Brown fat can reduce obesity through the dissipation of calories as heat. Control of thermogenic gene expression occurs via the induction of various coactivators, most notably PGC-1α. In contrast, the transcription factor partner(s) of these cofactors are poorly described. Here, we identify interferon regulatory factor 4 (IRF4) as a dominant transcriptional effector of thermogenesis. IRF4 is induced by cold and cAMP in adipocytes and is sufficient to promote increased thermogenic gene expression, energy expenditure, and cold tolerance. Conversely, knockout of IRF4 in UCP1(+) cells causes reduced thermogenic gene expression and energy expenditure, obesity, and cold intolerance. IRF4 also induces the expression of PGC-1α and PRDM16 and interacts with PGC-1α, driving Ucp1 expression. Finally, cold, ß-agonists, or forced expression of PGC-1α are unable to cause thermogenic gene expression in the absence of IRF4. These studies establish IRF4 as a transcriptional driver of a program of thermogenic gene expression and energy expenditure.


Subject(s)
Adipose Tissue, Brown/metabolism , Interferon Regulatory Factors/metabolism , Thermogenesis , Transcription Factors/metabolism , Transcriptional Activation , Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cold Temperature , Cyclic AMP/metabolism , Energy Metabolism , Humans , Ion Channels/genetics , Mice , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Obesity/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Thinness/metabolism , Transcriptional Activation/drug effects , Uncoupling Protein 1
19.
Nature ; 607(7917): 104-110, 2022 07.
Article in English | MEDLINE | ID: mdl-35732741

ABSTRACT

Itch triggers scratching, a behavioural defence mechanism that aids in the removal of harmful irritants and parasites1. Chemical itch is triggered by many endogenous and exogenous cues, such as pro-inflammatory histamine, which is released during an allergic reaction1. Mechanical itch can be triggered by light sensations such as wool fibres or a crawling insect2. In contrast to chemical itch pathways, which have been extensively studied, the mechanisms that underlie the transduction of mechanical itch are largely unknown. Here we show that the mechanically activated ion channel PIEZO1 (ref. 3) is selectively expressed by itch-specific sensory neurons and is required for their mechanically activated currents. Loss of PIEZO1 function in peripheral neurons greatly reduces mechanically evoked scratching behaviours and both acute and chronic itch-evoked sensitization. Finally, mice expressing a gain-of-function Piezo1 allele4 exhibit enhanced mechanical itch behaviours. Our studies reveal the polymodal nature of itch sensory neurons and identify a role for PIEZO1 in the sensation of itch.


Subject(s)
Ion Channels , Pruritus , Alleles , Animals , Ion Channels/deficiency , Ion Channels/genetics , Ion Channels/metabolism , Mice , Pruritus/genetics , Pruritus/physiopathology , Sensation , Sensory Receptor Cells/metabolism
20.
Annu Rev Neurosci ; 42: 67-86, 2019 07 08.
Article in English | MEDLINE | ID: mdl-30699050

ABSTRACT

The genetic approach, based on the study of inherited forms of deafness, has proven to be particularly effective for deciphering the molecular mechanisms underlying the development of the peripheral auditory system, the cochlea and its afferent auditory neurons, and how this system extracts the physical parameters of sound. Although this genetic dissection has provided little information about the central auditory system, scattered data suggest that some genes may have a critical role in both the peripheral and central auditory systems. Here, we review the genes controlling the development and function of the peripheral and central auditory systems, focusing on those with demonstrated intrinsic roles in both systems and highlighting the current underappreciation of these genes. Their encoded products are diverse, from transcription factors to ion channels, as are their roles in the central auditory system, mostly evaluated in brainstem nuclei. We examine the ontogenetic and evolutionary mechanisms that may underlie their expression at different sites.


Subject(s)
Auditory Pathways/physiology , Gene Expression Regulation, Developmental , Genes , Neurogenesis/genetics , Animals , Auditory Pathways/growth & development , Biological Evolution , Cochlea/embryology , Cochlea/growth & development , Cochlea/physiology , Gene Ontology , Hair Cells, Auditory/cytology , Hair Cells, Auditory/physiology , Hearing Disorders/genetics , Humans , Ion Channels/genetics , Ion Channels/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Rhombencephalon/embryology , Rhombencephalon/growth & development , Rhombencephalon/physiology , Sensory Receptor Cells/physiology , Transcription Factors/genetics , Transcription Factors/physiology
SELECTION OF CITATIONS
SEARCH DETAIL