Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 184(2): 294-296, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33482094

ABSTRACT

Children and adults with atopic dermatitis suffer from intractable chronic itch and can also experience acute itch flare ups that significantly increase itch intensity. In this issue of Cell, Wang et al. demonstrate that a subset of basophils activates sensory neurons to drive allergen-evoked itch flare ups in atopic dermatitis.


Subject(s)
Dermatitis, Atopic , Eczema , Allergens , Basophils , Humans , Pruritus
2.
Cell ; 155(2): 285-95, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24094650

ABSTRACT

Atopic dermatitis (AD) is a chronic itch and inflammatory disorder of the skin that affects one in ten people. Patients suffering from severe AD eventually progress to develop asthma and allergic rhinitis, in a process known as the "atopic march." Signaling between epithelial cells and innate immune cells via the cytokine thymic stromal lymphopoietin (TSLP) is thought to drive AD and the atopic march. Here, we report that epithelial cells directly communicate to cutaneous sensory neurons via TSLP to promote itch. We identify the ORAI1/NFAT calcium signaling pathway as an essential regulator of TSLP release from keratinocytes, the primary epithelial cells of the skin. TSLP then acts directly on a subset of TRPA1-positive sensory neurons to trigger robust itch behaviors. Our results support a model whereby calcium-dependent TSLP release by keratinocytes activates both primary afferent neurons and immune cells to promote inflammatory responses in the skin and airways.


Subject(s)
Cytokines/metabolism , Dermatitis, Atopic/pathology , Signal Transduction , Animals , Calcium/metabolism , Cells, Cultured , Dermatitis, Atopic/metabolism , Humans , Immunoglobulins/metabolism , Keratinocytes/metabolism , Pruritus/immunology , Receptors, Cytokine/metabolism , Sensory Receptor Cells/metabolism , Skin/metabolism , Skin/pathology , TRPA1 Cation Channel , Transient Receptor Potential Channels/metabolism , Thymic Stromal Lymphopoietin
3.
Cell ; 139(2): 267-84, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19837031

ABSTRACT

The nervous system detects and interprets a wide range of thermal and mechanical stimuli, as well as environmental and endogenous chemical irritants. When intense, these stimuli generate acute pain, and in the setting of persistent injury, both peripheral and central nervous system components of the pain transmission pathway exhibit tremendous plasticity, enhancing pain signals and producing hypersensitivity. When plasticity facilitates protective reflexes, it can be beneficial, but when the changes persist, a chronic pain condition may result. Genetic, electrophysiological, and pharmacological studies are elucidating the molecular mechanisms that underlie detection, coding, and modulation of noxious stimuli that generate pain.


Subject(s)
Nociceptors/physiology , Pain/physiopathology , Animals , Chronic Disease , Humans , Neuronal Plasticity
4.
FASEB J ; 35(10): e21899, 2021 10.
Article in English | MEDLINE | ID: mdl-34569661

ABSTRACT

The cornea of the eye differs from other mucosal surfaces in that it lacks a viable bacterial microbiome and by its unusually high density of sensory nerve endings. Here, we explored the role of corneal nerves in preventing bacterial adhesion. Pharmacological and genetic methods were used to inhibit the function of corneal sensory nerves or their associated transient receptor potential cation channels TRPA1 and TRPV1. Impacts on bacterial adhesion, resident immune cells, and epithelial integrity were examined using fluorescent labeling and quantitative confocal imaging. TRPA1/TRPV1 double gene-knockout mice were more susceptible to adhesion of environmental bacteria and to that of deliberately-inoculated Pseudomonas aeruginosa. Supporting the involvement of TRPA1/TRPV1-expressing corneal nerves, P. aeruginosa adhesion was also promoted by treatment with bupivacaine, or ablation of TRPA1/TRPV1-expressing nerves using RTX. Moreover, TRPA1/TRPV1-dependent defense was abolished by enucleation which severs corneal nerves. High-resolution imaging showed normal corneal ultrastructure and surface-labeling by wheat-germ agglutinin for TRPA1/TRPV1 knockout murine corneas, and intact barrier function by absence of fluorescein staining. P. aeruginosa adhering to corneas after perturbation of nerve or TRPA1/TRPV1 function failed to penetrate the surface. Single gene-knockout mice showed roles for both TRPA1 and TRPV1, with TRPA1-/- more susceptible to P. aeruginosa adhesion while TRPV1-/- corneas instead accumulated environmental bacteria. Corneal CD45+/CD11c+ cell responses to P. aeruginosa challenge, previously shown to counter bacterial adhesion, also depended on TRPA1/TRPV1 and sensory nerves. Together, these results demonstrate roles for corneal nerves and TRPA1/TRPV1 in corneal resistance to bacterial adhesion in vivo and suggest that the mechanisms involve resident immune cell populations.


Subject(s)
Bacterial Adhesion , Cornea , Pseudomonas aeruginosa/metabolism , TRPA1 Cation Channel/metabolism , TRPV Cation Channels/metabolism , Animals , Cornea/innervation , Cornea/metabolism , Cornea/microbiology , Female , Male , Mice , Mice, Knockout , TRPA1 Cation Channel/genetics , TRPV Cation Channels/genetics
5.
Nat Chem Biol ; 15(6): 623-631, 2019 06.
Article in English | MEDLINE | ID: mdl-31036923

ABSTRACT

Sphingosine-1-phosphate (S1P) plays important roles as a signaling lipid in a variety of physiological and pathophysiological processes. S1P signals via a family of G-protein-coupled receptors (GPCRs) (S1P1-5) and intracellular targets. Here, we report on photoswitchable analogs of S1P and its precursor sphingosine, respectively termed PhotoS1P and PhotoSph. PhotoS1P enables optical control of S1P1-3, shown through electrophysiology and Ca2+ mobilization assays. We evaluated PhotoS1P in vivo, where it reversibly controlled S1P3-dependent pain hypersensitivity in mice. The hypersensitivity induced by PhotoS1P is comparable to that induced by S1P. PhotoS1P is uniquely suited for the study of S1P biology in cultured cells and in vivo because it exhibits prolonged metabolic stability compared to the rapidly metabolized S1P. Using lipid mass spectrometry analysis, we constructed a metabolic map of PhotoS1P and PhotoSph. The formation of these photoswitchable lipids was found to be light dependent, providing a novel approach to optically probe sphingolipid biology.


Subject(s)
Lysophospholipids/metabolism , Sphingosine/analogs & derivatives , Animals , Lysophospholipids/chemistry , Mice , Models, Molecular , Molecular Structure , Optical Imaging , Photochemical Processes , Sphingosine/chemistry , Sphingosine/metabolism
6.
J Neurosci ; 38(36): 7833-7843, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30082422

ABSTRACT

Sphingosine 1-phosphate (S1P) is a bioactive signaling lipid associated with a variety of chronic pain and itch disorders. S1P signaling has been linked to cutaneous pain, but its role in itch has not yet been studied. Here, we find that S1P triggers itch and pain in male mice in a concentration-dependent manner, with low levels triggering acute itch alone and high levels triggering both pain and itch. Ca2+ imaging and electrophysiological experiments revealed that S1P signals via S1P receptor 3 (S1PR3) and TRPA1 in a subset of pruriceptors and via S1PR3 and TRPV1 in a subset of heat nociceptors. Consistent with these findings, S1P-evoked itch behaviors are selectively lost in mice lacking TRPA1, whereas S1P-evoked acute pain and heat hypersensitivity are selectively lost in mice lacking TRPV1. We conclude that S1P acts via different cellular and molecular mechanisms to trigger itch and pain. Our discovery elucidates the diverse roles that S1P signaling plays in somatosensation and provides insight into how itch and pain are discriminated in the periphery.SIGNIFICANCE STATEMENT Itch and pain are major health problems with few effective treatments. Here, we show that the proinflammatory lipid sphingosine 1-phosphate (S1P) and its receptor, S1P receptor 3 (S1PR3), trigger itch and pain behaviors via distinct molecular and cellular mechanisms. Our results provide a detailed understanding of the roles that S1P and S1PR3 play in somatosensation, highlighting their potential as targets for analgesics and antipruritics, and provide new insight into the mechanistic underpinnings of itch versus pain discrimination in the periphery.


Subject(s)
Lysophospholipids/metabolism , Pain/metabolism , Pruritus/metabolism , Receptors, Lysosphingolipid/metabolism , Signal Transduction/physiology , Sphingosine/analogs & derivatives , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Mice , Mice, Knockout , Pain/genetics , Pruritus/genetics , Receptors, Lysosphingolipid/genetics , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors , TRPV Cation Channels/genetics
7.
Nature ; 555(7698): 591-592, 2018 Mar.
Article in English | MEDLINE | ID: mdl-32094514
8.
Nature ; 555(7698): 591-592, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29595801
9.
Annu Rev Physiol ; 75: 181-200, 2013.
Article in English | MEDLINE | ID: mdl-23020579

ABSTRACT

Tissue damage evokes an inflammatory response that promotes the removal of harmful stimuli, tissue repair, and protective behaviors to prevent further damage and encourage healing. However, inflammation may outlive its usefulness and become chronic. Chronic inflammation can lead to a host of diseases, including asthma, itch, rheumatoid arthritis, and colitis. Primary afferent sensory neurons that innervate target organs release inflammatory neuropeptides in the local area of tissue damage to promote vascular leakage, the recruitment of immune cells, and hypersensitivity to mechanical and thermal stimuli. TRPA1 channels are required for neuronal excitation, the release of inflammatory neuropeptides, and subsequent pain hypersensitivity. TRPA1 is also activated by the release of inflammatory agents from nonneuronal cells in the area of tissue injury or disease. This dual function of TRPA1 as a detector and instigator of inflammatory agents makes TRPA1 a gatekeeper of chronic inflammatory disorders of the skin, airways, and gastrointestinal tract.


Subject(s)
Calcium Channels/physiology , Inflammation/physiopathology , Nerve Tissue Proteins/physiology , Signal Transduction/physiology , Transient Receptor Potential Channels/physiology , Humans , Neuropeptides/physiology , Pain/physiopathology , TRPA1 Cation Channel , Viscera/physiopathology
10.
Handb Exp Pharmacol ; 226: 177-90, 2015.
Article in English | MEDLINE | ID: mdl-25861780

ABSTRACT

Keratinocytes are epithelial cells that make up the stratified epidermis of the skin. Recent studies suggest that keratinocytes promote chronic itch. Changes in skin morphology that accompany a variety of chronic itch disorders and the multitude of inflammatory mediators secreted by keratinocytes that target both sensory neurons and immune cells highlight the importance of investigating the connection between keratinocytes and chronic itch. This chapter addresses some of the most recent data and models for the role keratinocytes play in the development and maintenance of chronic itch.


Subject(s)
Cell Communication/physiology , Keratinocytes/physiology , Pruritus/physiopathology , Sensory Receptor Cells/physiology , Animals , Chronic Disease , Humans
11.
J Neurosci ; 33(22): 9283-94, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23719797

ABSTRACT

Chronic itch is a debilitating condition that affects one in 10 people. Little is known about the molecules that mediate chronic itch in primary sensory neurons and skin. We demonstrate that the ion channel TRPA1 is required for chronic itch. Using a mouse model of chronic itch, we show that scratching evoked by impaired skin barrier is abolished in TRPA1-deficient animals. This model recapitulates many of the pathophysiological hallmarks of chronic itch that are observed in prevalent human diseases such as atopic dermatitis and psoriasis, including robust scratching, extensive epidermal hyperplasia, and dramatic changes in gene expression in sensory neurons and skin. Remarkably, TRPA1 is required for both transduction of chronic itch signals to the CNS and for the dramatic skin changes triggered by dry-skin-evoked itch and scratching. These data suggest that TRPA1 regulates both itch transduction and pathophysiological changes in the skin that promote chronic itch.


Subject(s)
Pruritus/physiopathology , Transient Receptor Potential Channels/physiology , Animals , Chronic Disease , Data Interpretation, Statistical , Gene Expression , Homeostasis/physiology , Hyperplasia/pathology , Male , Mice , Mice, Inbred C57BL , Microarray Analysis , Pruritus/genetics , Pruritus/pathology , Real-Time Polymerase Chain Reaction , Sensory Receptor Cells , Skin/innervation , Skin/pathology , TRPA1 Cation Channel , Transient Receptor Potential Channels/genetics
12.
bioRxiv ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-37503246

ABSTRACT

A key goal of evolutionary genomics is to harness molecular data to draw inferences about selective forces that have acted on genomes. The field progresses in large part through the development of advanced molecular-evolution analysis methods. Here we explored the intersection between classical sequence-based tests for selection and an empirical expression-based approach, using stem cells from Mus musculus subspecies as a model. Using a test of directional, cis-regulatory evolution across genes in pathways, we discovered a unique program of induction of translation genes in stem cells of the Southeast Asian mouse M. m. castaneus relative to its sister taxa. As a complement, we used sequence analyses to find population-genomic signatures of selection in M. m. castaneus, at the upstream regions of the translation genes, including at transcription factor binding sites. We interpret our data under a model of changes in lineage-specific pressures across Mus musculus in stem cells with high translational capacity. Together, our findings underscore the rigor of integrating expression and sequence-based methods to generate hypotheses about evolutionary events from long ago.

13.
bioRxiv ; 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38260476

ABSTRACT

SARS-CoV-2, the virus responsible for COVID-19, triggers symptoms such as sneezing, aches and pain.1 These symptoms are mediated by a subset of sensory neurons, known as nociceptors, that detect noxious stimuli, densely innervate the airway epithelium, and interact with airway resident epithelial and immune cells.2-6 However, the mechanisms by which viral infection activates these neurons to trigger pain and airway reflexes are unknown. Here, we show that the coronavirus papain-like protease (PLpro) directly activates airway-innervating trigeminal and vagal nociceptors in mice and human iPSC-derived nociceptors. PLpro elicits sneezing and acute pain in mice and triggers the release of neuropeptide calcitonin gene-related peptide (CGRP) from airway afferents. We find that PLpro-induced sneeze and pain requires the host TRPA1 ion channel that has been previously demonstrated to mediate pain, cough, and airway inflammation.7-9 Our findings are the first demonstration of a viral product that directly activates sensory neurons to trigger pain and airway reflexes and highlight a new role for PLpro and nociceptors in COVID-19.

14.
J Physiol ; 591(13): 3325-40, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23652591

ABSTRACT

In traditional medicine, the 'toothache tree' and other plants of the Zanthoxylum genus have been used to treat inflammatory pain conditions, such as toothache and rheumatoid arthritis. Here we examined the cellular and molecular mechanisms underlying the analgesic properties of hydroxy-α-sanshool, the active alkylamide produced by Zanthoxylum plants. Consistent with its analgesic effects in humans, sanshool treatment in mice caused a selective attenuation of mechanical sensitivity under naïve and inflammatory conditions, with no effect on thermal sensitivity. To elucidate the molecular mechanisms by which sanshool attenuates mechanical pain, we performed single fibre recordings, calcium imaging and whole-cell electrophysiology of cultured sensory neurons. We found that: (1) sanshool potently inhibits Aδ mechanonociceptors that mediate both sharp acute pain and inflammatory pain; (2) sanshool inhibits action potential firing by blocking voltage-gated sodium currents in a subset of somatosensory neurons, which express a unique combination of voltage-gated sodium channels; and (3) heterologously expressed Nav1.7 is most strongly inhibited by sanshool as compared to other sodium channels expressed in sensory neurons. These results suggest that sanshool targets voltage-gated sodium channels on Aδ mechanosensory nociceptors to dampen excitability and thus induce 'fast pain' analgesia.


Subject(s)
Amides/pharmacology , Pain/physiopathology , Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channels/physiology , Amides/therapeutic use , Animals , CHO Cells , Cells, Cultured , Cricetulus , Ganglia, Spinal/cytology , Hot Temperature , Male , Mechanoreceptors/physiology , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Pain/drug therapy , Pain/etiology , Sodium Channel Blockers/therapeutic use , Zanthoxylum
15.
Nature ; 448(7150): 204-8, 2007 Jul 12.
Article in English | MEDLINE | ID: mdl-17538622

ABSTRACT

Sensory nerve fibres can detect changes in temperature over a remarkably wide range, a process that has been proposed to involve direct activation of thermosensitive excitatory transient receptor potential (TRP) ion channels. One such channel--TRP melastatin 8 (TRPM8) or cold and menthol receptor 1 (CMR1)--is activated by chemical cooling agents (such as menthol) or when ambient temperatures drop below approximately 26 degrees C, suggesting that it mediates the detection of cold thermal stimuli by primary afferent sensory neurons. However, some studies have questioned the contribution of TRPM8 to cold detection or proposed that other excitatory or inhibitory channels are more critical to this sensory modality in vivo. Here we show that cultured sensory neurons and intact sensory nerve fibres from TRPM8-deficient mice exhibit profoundly diminished responses to cold. These animals also show clear behavioural deficits in their ability to discriminate between cold and warm surfaces, or to respond to evaporative cooling. At the same time, TRPM8 mutant mice are not completely insensitive to cold as they avoid contact with surfaces below 10 degrees C, albeit with reduced efficiency. Thus, our findings demonstrate an essential and predominant role for TRPM8 in thermosensation over a wide range of cold temperatures, validating the hypothesis that TRP channels are the principal sensors of thermal stimuli in the peripheral nervous system.


Subject(s)
Cold Temperature , TRPM Cation Channels/physiology , Thermosensing , Animals , Female , Ganglia, Sensory/physiology , Gene Targeting , Male , Menthol/metabolism , Mice , Mice, Inbred C57BL , TRPM Cation Channels/genetics
16.
Nat Neurosci ; 11(7): 772-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18568022

ABSTRACT

In traditional folk medicine, Xanthoxylum plants are referred to as 'toothache trees' because their anesthetic or counter-irritant properties render them useful in the treatment of pain. Psychophysical studies have identified hydroxy-alpha-sanshool as the compound most responsible for the unique tingling and buzzing sensations produced by Szechuan peppercorns or other Xanthoxylum preparations. Although it is generally agreed that sanshool elicits its effects by activating somatosensory neurons, the underlying cellular and molecular mechanisms remain a matter of debate. Here we show that hydroxy-alpha-sanshool excites two types of sensory neurons, including small-diameter unmyelinated cells that respond to capsaicin (but not mustard oil) as well as large-diameter myelinated neurons that express the neurotrophin receptor TrkC. We found that hydroxy-alpha-sanshool excites neurons through a unique mechanism involving inhibition of pH- and anesthetic-sensitive two-pore potassium channels (KCNK3, KCNK9 and KCNK18), providing a framework for understanding the unique and complex psychophysical sensations associated with the Szechuan pepper experience.


Subject(s)
Amides/pharmacology , Neurons, Afferent/drug effects , Piper nigrum/chemistry , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/physiology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Capsaicin/pharmacology , Cells, Cultured , Electric Stimulation/methods , Ganglia, Sensory/cytology , Gene Expression Regulation/drug effects , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Neurofilament Proteins/metabolism , Patch-Clamp Techniques , Potassium Channels, Tandem Pore Domain/drug effects , Potassium Chloride/pharmacology , Receptor, trkC/metabolism , TRPA1 Cation Channel , TRPV Cation Channels/deficiency , Transient Receptor Potential Channels/deficiency
17.
Biophys J ; 100(8): 1902-9, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21504726

ABSTRACT

Histone deacetylation and acetylation are catalyzed by histone deacetylase (HDAC) and histone acetyltransferase, respectively, which play important roles in the regulation of chromatin remodeling, gene expression, and cell functions. However, whether and how biophysical cues modulate HDAC activity and histone acetylation is not well understood. Here, we tested the hypothesis that microtopographic patterning and mechanical strain on the substrate regulate nuclear shape, HDAC activity, and histone acetylation. Bone marrow mesenchymal stem cells (MSCs) were cultured on elastic membranes patterned with parallel microgrooves 10 µm wide that kept MSCs aligned along the axis of the grooves. Compared with MSCs on an unpatterned substrate, MSCs on microgrooves had elongated nuclear shape, a decrease in HDAC activity, and an increase of histone acetylation. To investigate anisotropic mechanical sensing by MSCs, cells on the elastic micropatterned membranes were subjected to static uniaxial mechanical compression or stretch in the direction parallel or perpendicular to the microgrooves. Among the four types of loads, compression or stretch perpendicular to the microgrooves caused a decrease in HDAC activity, accompanied by the increase in histone acetylation and slight changes of nuclear shape. Knocking down nuclear matrix protein lamin A/C abolished mechanical strain-induced changes in HDAC activity. These results demonstrate that micropattern and mechanical strain on the substrate can modulate nuclear shape, HDAC activity, and histone acetylation in an anisotropic manner and that nuclear matrix mediates mechanotransduction. These findings reveal a new mechanism, to our knowledge, by which extracellular biophysical signals are translated into biochemical signaling events in the nucleus, and they will have significant impact in the area of mechanobiology and mechanotransduction.


Subject(s)
Biophysical Phenomena , Histones/metabolism , Mesenchymal Stem Cells/metabolism , Acetylation , Anisotropy , Biomechanical Phenomena , Cell Culture Techniques , Cell Nucleus/metabolism , Histone Deacetylases/metabolism , Humans , Lamin Type A/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Stress, Mechanical
18.
J Neurosci ; 30(12): 4353-61, 2010 Mar 24.
Article in English | MEDLINE | ID: mdl-20335471

ABSTRACT

Hydroxy-alpha-sanshool, the active ingredient in plants of the prickly ash plant family, induces robust tingling paresthesia by activating a subset of somatosensory neurons. However, the subtypes and physiological function of sanshool-sensitive neurons remain unknown. Here we use the ex vivo skin-nerve preparation to examine the pattern and intensity with which the sensory terminals of cutaneous neurons respond to hydroxy-alpha-sanshool. We found that sanshool excites virtually all D-hair afferents, a distinct subset of ultrasensitive light-touch receptors in the skin and targets novel populations of Abeta and C fiber nerve afferents. Thus, sanshool provides a novel pharmacological tool for discriminating functional subtypes of cutaneous mechanoreceptors. The identification of sanshool-sensitive fibers represents an essential first step in identifying the cellular and molecular mechanisms underlying tingling paresthesia that accompanies peripheral neuropathy and injury.


Subject(s)
Neurons, Afferent/physiology , Paresthesia/chemically induced , Paresthesia/pathology , Skin/innervation , Action Potentials/drug effects , Amides , Animals , Animals, Newborn , Capsaicin/pharmacology , Cells, Cultured , In Vitro Techniques , Isoquinolines/pharmacology , Mechanoreceptors/drug effects , Mechanoreceptors/physiology , Mice , Mice, Inbred C57BL , Mustard Plant , Nerve Fibers/drug effects , Nerve Fibers/physiology , Nerve Growth Factors/pharmacology , Neural Conduction/drug effects , Neural Conduction/physiology , Neurons, Afferent/drug effects , Paresthesia/physiopathology , Plant Oils/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/physiology , Sensory System Agents/pharmacology , Statistics, Nonparametric , Touch/physiology , Trigeminal Ganglion/cytology , Urea/analogs & derivatives , Urea/pharmacology , gamma-Aminobutyric Acid/pharmacology
19.
Proc Natl Acad Sci U S A ; 105(50): 20015-20, 2008 Dec 16.
Article in English | MEDLINE | ID: mdl-19060212

ABSTRACT

Primary afferent somatosensory neurons mediate our sense of touch in response to changes in ambient pressure. Molecules that detect and transduce thermal stimuli have been recently identified, but mechanisms underlying mechanosensation, particularly in vertebrate organisms, remain enigmatic. Traditionally, mechanically evoked responses in somatosensory neurons have been assessed one cell at a time by recording membrane currents in response to application of focal pressure, suction, or osmotic challenge. Here, we used radial stretch in combination with live-cell calcium imaging to gain a broad overview of mechanosensitive neuronal subpopulations. We found that different stretch intensities activate distinct subsets of sensory neurons as defined by size, molecular markers, or pharmacological attributes. In all subsets, stretch-evoked responses required extracellular calcium, indicating that mechanical force triggers calcium influx. This approach extends the repertoire of stimulus paradigms that can be used to examine mechanotransduction in mammalian sensory neurons, facilitating future physiological and pharmacological studies.


Subject(s)
Calcium/metabolism , Mechanotransduction, Cellular , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Mice , Potassium Chloride/pharmacology , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Stress, Mechanical
20.
Neuron ; 109(19): 3075-3087.e2, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34411514

ABSTRACT

Itch is a discrete and irritating sensation tightly coupled to a drive to scratch. Acute scratching developed evolutionarily as an adaptive defense against skin irritants, pathogens, or parasites. In contrast, the itch-scratch cycle in chronic itch is harmful, inducing escalating itch and skin damage. Clinically and preclinically, scratching incidence is currently evaluated as a unidimensional motor parameter and believed to reflect itch severity. We propose that scratching, when appreciated as a complex, multidimensional motor behavior, will yield greater insight into the nature of itch and the organization of neural circuits driving repetitive motor patterns. We outline the limitations of standard measurements of scratching in rodent models and present new approaches to observe and quantify itch-evoked scratching. We argue that accurate quantitative measurements of scratching are critical for dissecting the molecular, cellular, and circuit mechanisms underlying itch and for preclinical development of therapeutic interventions for acute and chronic itch disorders.


Subject(s)
Pruritus/physiopathology , Animals , Disease Models, Animal , Dogs , Humans , Mice , Pruritus/therapy , Rats
SELECTION OF CITATIONS
SEARCH DETAIL