Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Pain ; 165(6): 1336-1347, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38739766

ABSTRACT

ABSTRACT: Evidence from previous studies supports the concept that spinal cord injury (SCI)-induced neuropathic pain (NP) has its neural roots in the peripheral nervous system. There is uncertainty about how and to which degree mechanoreceptors contribute. Sensorimotor activation-based interventions (eg, treadmill training) have been shown to reduce NP after experimental SCI, suggesting transmission of pain-alleviating signals through mechanoreceptors. The aim of the present study was to understand the contribution of mechanoreceptors with respect to mechanical allodynia in a moderate mouse contusion SCI model. After genetic ablation of tropomyosin receptor kinase B expressing mechanoreceptors before SCI, mechanical allodynia was reduced. The identical genetic ablation after SCI did not yield any change in pain behavior. Peptidergic nociceptor sprouting into lamina III/IV below injury level as a consequence of SCI was not altered by either mechanoreceptor ablation. However, skin-nerve preparations of contusion SCI mice 7 days after injury yielded hyperexcitability in nociceptors, not in mechanoreceptors, which makes a substantial direct contribution of mechanoreceptors to NP maintenance unlikely. Complementing animal data, quantitative sensory testing in human SCI subjects indicated reduced mechanical pain thresholds, whereas the mechanical detection threshold was not altered. Taken together, early mechanoreceptor ablation modulates pain behavior, most likely through indirect mechanisms. Hyperexcitable nociceptors seem to be the main drivers of SCI-induced NP. Future studies need to focus on injury-derived factors triggering early-onset nociceptor hyperexcitability, which could serve as targets for more effective therapeutic interventions.


Subject(s)
Disease Models, Animal , Hyperalgesia , Mechanoreceptors , Mice, Inbred C57BL , Spinal Cord Injuries , Animals , Spinal Cord Injuries/complications , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology , Mice , Hyperalgesia/physiopathology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Male , Humans , Pain Threshold/physiology , Female , Pain Measurement , Mice, Transgenic , Neuralgia/etiology , Neuralgia/metabolism , Neuralgia/physiopathology
2.
Br J Pharmacol ; 2024 May 13.
Article in English | MEDLINE | ID: mdl-38741464

ABSTRACT

BACKGROUND AND PURPOSE: The mechanistic target of rapamycin (mTOR) signalling pathway is a key regulator of cell growth and metabolism. Its deregulation is implicated in several diseases. The macrolide rapamycin, a specific inhibitor of mTOR, has immunosuppressive, anti-inflammatory and antiproliferative properties. Recently, we identified tacrolimus, another macrolide immunosuppressant, as a novel activator of TRPM8 ion channels, involved in cold temperature sensing, thermoregulation, tearing and cold pain. We hypothesized that rapamycin may also have agonist activity on TRPM8 channels. EXPERIMENTAL APPROACH: Using calcium imaging and electrophysiology in transfected HEK293 cells and wildtype or Trpm8 KO mouse DRG neurons, we characterized rapamycin's effects on TRPM8 channels. We also examined the effects of rapamycin on tearing in mice. KEY RESULTS: Micromolar concentrations of rapamycin activated rat and mouse TRPM8 channels directly and potentiated cold-evoked responses, effects also observed in human TRPM8 channels. In cultured mouse DRG neurons, rapamycin increased intracellular calcium levels almost exclusively in cold-sensitive neurons. Responses were markedly decreased in Trpm8 KO mice or by TRPM8 channel antagonists. Cutaneous cold thermoreceptor endings were also activated by rapamycin. Topical application of rapamycin to the eye surface evokes tearing in mice by a TRPM8-dependent mechanism. CONCLUSION AND IMPLICATIONS: These results identify TRPM8 cationic channels in sensory neurons as novel molecular targets of the immunosuppressant rapamycin. These findings may help explain some of its therapeutic effects after topical application to the skin and the eye surface. Moreover, rapamycin could be used as an experimental tool in the clinic to explore cold thermoreceptors.

3.
J Clin Invest ; 134(7)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38557489

ABSTRACT

Regulated exocytosis is initiated by increased Ca2+ concentrations in close spatial proximity to secretory granules, which is effectively prevented when the cell is at rest. Here we showed that exocytosis of zymogen granules in acinar cells was driven by Ca2+ directly released from acidic Ca2+ stores including secretory granules through NAADP-activated two-pore channels (TPCs). We identified OCaR1 (encoded by Tmem63a) as an organellar Ca2+ regulator protein integral to the membrane of secretory granules that controlled Ca2+ release via inhibition of TPC1 and TPC2 currents. Deletion of OCaR1 led to extensive Ca2+ release from NAADP-responsive granules under basal conditions as well as upon stimulation of GPCR receptors. Moreover, OCaR1 deletion exacerbated the disease phenotype in murine models of severe and chronic pancreatitis. Our findings showed OCaR1 as a gatekeeper of Ca2+ release that endows NAADP-sensitive secretory granules with an autoregulatory mechanism preventing uncontrolled exocytosis and pancreatic tissue damage.


Subject(s)
Calcium Channels , Calcium , Mice , Animals , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium/metabolism , Pancreas/metabolism , Exocytosis/physiology , Secretory Vesicles/genetics
4.
Nat Commun ; 14(1): 1899, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37019973

ABSTRACT

Mechanically silent nociceptors are sensory afferents that are insensitive to noxious mechanical stimuli under normal conditions but become sensitized to such stimuli during inflammation. Using RNA-sequencing and quantitative RT-PCR we demonstrate that inflammation upregulates the expression of the transmembrane protein TMEM100 in silent nociceptors and electrophysiology revealed that over-expression of TMEM100 is required and sufficient to un-silence silent nociceptors in mice. Moreover, we show that mice lacking TMEM100 do not develop secondary mechanical hypersensitivity-i.e., pain hypersensitivity that spreads beyond the site of inflammation-during knee joint inflammation and that AAV-mediated overexpression of TMEM100 in articular afferents in the absence of inflammation is sufficient to induce mechanical hypersensitivity in remote skin regions without causing knee joint pain. Thus, our work identifies TMEM100 as a key regulator of silent nociceptor un-silencing and reveals a physiological role for this hitherto enigmatic afferent subclass in triggering spatially remote secondary mechanical hypersensitivity during inflammation.


Subject(s)
Nociceptors , Pain , Animals , Mice , Inflammation/metabolism , Knee Joint , Nociceptors/metabolism , Pain/metabolism , Skin/metabolism
5.
Nature ; 606(7912): 137-145, 2022 06.
Article in English | MEDLINE | ID: mdl-35614217

ABSTRACT

Nerve injury leads to chronic pain and exaggerated sensitivity to gentle touch (allodynia) as well as a loss of sensation in the areas in which injured and non-injured nerves come together1-3. The mechanisms that disambiguate these mixed and paradoxical symptoms are unknown. Here we longitudinally and non-invasively imaged genetically labelled populations of fibres that sense noxious stimuli (nociceptors) and gentle touch (low-threshold afferents) peripherally in the skin for longer than 10 months after nerve injury, while simultaneously tracking pain-related behaviour in the same mice. Fully denervated areas of skin initially lost sensation, gradually recovered normal sensitivity and developed marked allodynia and aversion to gentle touch several months after injury. This reinnervation-induced neuropathic pain involved nociceptors that sprouted into denervated territories precisely reproducing the initial pattern of innervation, were guided by blood vessels and showed irregular terminal connectivity in the skin and lowered activation thresholds mimicking low-threshold afferents. By contrast, low-threshold afferents-which normally mediate touch sensation as well as allodynia in intact nerve territories after injury4-7-did not reinnervate, leading to an aberrant innervation of tactile end organs such as Meissner corpuscles with nociceptors alone. Genetic ablation of nociceptors fully abrogated reinnervation allodynia. Our results thus reveal the emergence of a form of chronic neuropathic pain that is driven by structural plasticity, abnormal terminal connectivity and malfunction of nociceptors during reinnervation, and provide a mechanistic framework for the paradoxical sensory manifestations that are observed clinically and can impose a heavy burden on patients.


Subject(s)
Hyperalgesia , Neuralgia , Nociceptors , Skin , Animals , Chronic Pain/physiopathology , Hyperalgesia/physiopathology , Mechanoreceptors/pathology , Mice , Neuralgia/physiopathology , Nociceptors/pathology , Skin/innervation , Skin/physiopathology
6.
Nat Commun ; 13(1): 1365, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35292651

ABSTRACT

A central question in mechanobiology is how mechanical forces acting in or on cells are transmitted to mechanically-gated PIEZO channels that convert these forces into biochemical signals. Here we examined the role of the intracellular domains of PIEZO2, which account for 25% of the channel, and demonstrate that these domains fine-tune properties such as poking and stretch-sensitivity, velocity coding and single channel conductance. Moreover, we show that the intrinsically disordered linker between the transmembrane helices twelve and thirteen (IDR5) is required for the activation of PIEZO2 by cytoskeleton-transmitted forces. The deletion of IDR5 abolishes PIEZO2-mediated inhibition of neurite outgrowth, while it only partially affected its sensitivity to cell indentation and does not alter its stretch sensitivity. Thus, we propose that PIEZO2 is a polymodal mechanosensor that detects different types of mechanical stimuli via different force transmission pathways, which highlights the importance of utilizing multiple complementary assays when investigating PIEZO function.


Subject(s)
Ion Channels , Mechanotransduction, Cellular , Cytoskeleton/metabolism , Ion Channels/metabolism , Mechanotransduction, Cellular/physiology
7.
Cell Rep ; 35(9): 109191, 2021 06 01.
Article in English | MEDLINE | ID: mdl-34077727

ABSTRACT

The vasculature is innervated by a network of peripheral afferents that sense and regulate blood flow. Here, we describe a system of non-peptidergic sensory neurons with cell bodies in the spinal ganglia that regulate vascular tone in the distal arteries. We identify a population of mechanosensitive neurons, marked by tropomyosin receptor kinase C (TrkC) and tyrosine hydroxylase in the dorsal root ganglia, which projects to blood vessels. Local stimulation of TrkC neurons decreases vessel diameter and blood flow, whereas systemic activation increases systolic blood pressure and heart rate variability via the sympathetic nervous system. Ablation of the neurons provokes variability in local blood flow, leading to a reduction in systolic blood pressure, increased heart rate variability, and ultimately lethality within 48 h. Thus, a population of TrkC+ sensory neurons forms part of a sensory-feedback mechanism that maintains cardiovascular homeostasis through the autonomic nervous system.


Subject(s)
Blood Pressure/physiology , Sensory Receptor Cells/physiology , Animals , Behavior, Animal , Fluorescein/metabolism , Ganglia, Spinal/physiology , Heart Rate/physiology , Mice, Transgenic , Receptor, trkC/metabolism
8.
Nat Neurosci ; 24(1): 74-81, 2021 01.
Article in English | MEDLINE | ID: mdl-33288907

ABSTRACT

Fingertip mechanoreceptors comprise sensory neuron endings together with specialized skin cells that form the end-organ. Exquisitely sensitive, vibration-sensing neurons are associated with Meissner's corpuscles in the skin. In the present study, we found that USH2A, a transmembrane protein with a very large extracellular domain, was found in terminal Schwann cells within Meissner's corpuscles. Pathogenic USH2A mutations cause Usher's syndrome, associated with hearing loss and visual impairment. We show that patients with biallelic pathogenic USH2A mutations also have clear and specific impairments in vibrotactile touch perception, as do mutant mice lacking USH2A. Forepaw rapidly adapting mechanoreceptors innervating Meissner's corpuscles, recorded from Ush2a-/- mice, showed large reductions in vibration sensitivity. However, the USH2A protein was not found in sensory neurons. Thus, loss of USH2A in corpuscular end-organs reduced mechanoreceptor sensitivity as well as vibration perception. Thus, a tether-like protein is required to facilitate detection of small-amplitude vibrations essential for the perception of fine-grained tactile surfaces.


Subject(s)
Extracellular Matrix Proteins/genetics , Mechanoreceptors/metabolism , Sensation/physiology , Vibration , Adult , Animals , Female , Humans , Male , Mice , Mice, Inbred CBA , Mice, Knockout , Mutation/genetics , Schwann Cells/physiology , Skin/innervation , Touch/physiology , Usher Syndromes/genetics
9.
Neuron ; 107(6): 1141-1159.e7, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32735781

ABSTRACT

Diabetic peripheral neuropathy (DPN) is a highly frequent and debilitating clinical complication of diabetes that lacks therapies. Cellular oxidative stress regulates post-translational modifications, including SUMOylation. Here, using unbiased screens, we identified key enzymes in metabolic pathways and ion channels as novel molecular targets of SUMOylation that critically regulated their activity. Sensory neurons of diabetic patients and diabetic mice demonstrated changes in the SUMOylation status of metabolic enzymes and ion channels. In support of this, profound metabolic dysfunction, accelerated neuropathology, and sensory loss were observed in diabetic gene-targeted mice selectively lacking the ability to SUMOylate proteins in peripheral sensory neurons. TRPV1 function was impaired by diabetes-induced de-SUMOylation as well as by metabolic imbalance elicited by de-SUMOylation of metabolic enzymes, facilitating diabetic sensory loss. Our results unexpectedly uncover an endogenous post-translational mechanism regulating diabetic neuropathy in patients and mouse models that protects against metabolic dysfunction, nerve damage, and altered sensory perception.


Subject(s)
Diabetic Neuropathies/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Nociception , Sensory Receptor Cells/metabolism , Sumoylation , TRPV Cation Channels/metabolism , Animals , Cells, Cultured , Citric Acid Cycle , Diabetic Neuropathies/physiopathology , Female , Ganglia, Spinal/cytology , Glycolysis , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL
10.
Proc Natl Acad Sci U S A ; 116(28): 14260-14269, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31235572

ABSTRACT

Piezo channels are mechanically activated ion channels that confer mechanosensitivity to a variety of different cell types. Piezos oligomerize as propeller-shaped homotrimers that are thought to locally curve the membrane into spherical domes that project into the cell. While several studies have identified domains and amino acids that control important properties such as ion permeability and selectivity as well as inactivation kinetics and voltage sensitivity, only little is known about intraprotein interactions that govern mechanosensitivity-the most unique feature of PIEZOs. Here we used site-directed mutagenesis and patch-clamp recordings to investigate the mechanogating mechanism of PIEZO2. We demonstrate that charged amino acids at the interface between the beam domain-i.e., a long α-helix that protrudes from the intracellular side of the "propeller" blade toward the inner vestibule of the channel-and the C-terminal domain (CTD) as well as hydrophobic interactions between the highly conserved Y2807 of the CTD and pore-lining helices are required to ensure normal mechanosensitivity of PIEZO2. Moreover, single-channel recordings indicate that a previously unrecognized intrinsically disordered domain located adjacent to the beam acts as a cytosolic plug that limits ion permeation possibly by clogging the inner vestibule of both PIEZO1 and PIEZO2. Thus, we have identified several intraprotein domain interfaces that control the mechanical activation of PIEZO1 and PIEZO2 and which might thus serve as promising targets for drugs that modulate the mechanosensitivity of Piezo channels.

11.
Cell Cycle ; 18(5): 580-595, 2019 03.
Article in English | MEDLINE | ID: mdl-30739521

ABSTRACT

The yeast ß-karyopherin Msn5 controls the SBF cell-cycle transcription factor, responsible for the periodic expression of CLN2 cyclin gene at G1/S, and the nuclear export of Cln2 protein. Here we show that Msn5 regulates Cln2 by an additional mechanism. Inactivation of Msn5 causes a severe reduction in the cellular content of Cln2. This occurs by a post-transcriptional mechanism, since CLN2 mRNA level is not importantly affected in asynchronous cultures. Cln2 stability is not significantly altered in msn5 cells and inactivation of Msn5 causes a reduction in protein level even when Cln2 is stabilized. Therefore, the reduced amount of Cln2 in msn5 cells is mainly due not to a higher rate of protein degradation but to a defect in Cln2 synthesis. In fact, analysis of polysome profiles indicated that Msn5 inactivation causes a shift of CLN2 and SWI5 mRNAs from heavy-polysomal to light-polysomal and non-polysomal fractions, supporting a defect in Cln2 and Swi5 protein synthesis in the msn5 mutant. The analysis of truncated versions of Cln2 and of chimeric cyclins combining distinct domains from Cln2 and the related Cln1 cyclin identified an internal region in Cln2 from 181 to 225 residues that when fused to GFP is able to confer Msn5-dependent regulation of protein cellular content. Finally, we showed that a high level of Cln2 is toxic in the absence of Msn5. In summary, we described that Msn5 is required for the proper protein synthesis of specific proteins, introducing a new level of control of cell cycle regulators.


Subject(s)
Cell Cycle Proteins/metabolism , Cyclins/metabolism , Karyopherins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/metabolism , Actins/genetics , Actins/metabolism , Cell Cycle Proteins/genetics , Cyclins/genetics , Gene Expression Regulation, Fungal , Karyopherins/genetics , Mutagenesis , Polyribosomes/metabolism , Protein Biosynthesis , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics
12.
Mol Pain ; 14: 1744806918814640, 2018.
Article in English | MEDLINE | ID: mdl-30387376

ABSTRACT

Nerve growth factor is an inflammatory mediator that induces long-lasting hyperalgesia, which can partially be attributed to nerve growth factor-induced sensitization of primary afferent nociceptors. It was shown that nerve growth factor increases the excitability of polymodal C-fibre nociceptors by modulating tetrodotoxin-sensitive and tetrodotoxin-resistant voltage-gated sodium channels, but hitherto only little is known about the effects of nerve growth factor on sodium currents in other nociceptor subtypes that express the nerve growth factor receptor TrkA. We previously characterized two reporter mouse lines that allow the unequivocal identification of two important subclasses of TrkA-expressing nociceptors - i.e. neuropeptide Y receptor type 2 (NPY2R+ ) Aδ-fibre nociceptors that mediate pinprick pain and nicotinic acetylcholine receptor alpha-3 subunit (CHRNA3+ ) silent nociceptors, which are the most abundant TrkA+ nociceptors in visceral organs and deep somatic tissues. Here, we utilized these mouse lines to investigate the expression patterns and the possible nerve growth factor-dependent modulation of sodium channels in these neurons using whole-cell patch-clamp recordings and quantitative real-time polymerase chain reaction. We demonstrate that NPY2R+ nociceptors, CHRNA3+ 'silent' nociceptors and polymodal C-fibre nociceptors express different combinations of sodium channel α- and ß-subunits and accordingly exhibit functionally different sodium currents. Moreover, we demonstrate that nerve growth factor produces robust hyperpolarizing shifts in the half-activation voltage of tetrodotoxin-resistant currents in NPY2R+ nociceptors and polymodal C-fibre nociceptors and also shifts the half-activation of tetrodotoxin-sensitive currents in polymodal C-fibre nociceptors. In silent nociceptors, however, nerve growth factor solely increases the current density of the tetrodotoxin-resistant current but does not alter other sodium channel properties. Considering the different peripheral target tissues and the previously reported roles in different forms of pain of the nociceptor subpopulations that were examined here, our results suggest that nerve growth factor differentially contributes to the development visceral and cutaneous pain hypersensitivity and highlights the importance of developing different therapeutic strategies for different forms of pain.


Subject(s)
Action Potentials/drug effects , Nerve Growth Factor/pharmacology , Nociceptors/metabolism , Tetrodotoxin/pharmacology , Voltage-Gated Sodium Channels/drug effects , Animals , Axons/drug effects , Axons/metabolism , Ganglia, Spinal/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Nociceptors/drug effects , Voltage-Gated Sodium Channels/metabolism
13.
Nat Commun ; 9(1): 1640, 2018 04 24.
Article in English | MEDLINE | ID: mdl-29691410

ABSTRACT

Mechanical allodynia is a major symptom of neuropathic pain whereby innocuous touch evokes severe pain. Here we identify a population of peripheral sensory neurons expressing TrkB that are both necessary and sufficient for producing pain from light touch after nerve injury in mice. Mice in which TrkB-Cre-expressing neurons are ablated are less sensitive to the lightest touch under basal conditions, and fail to develop mechanical allodynia in a model of neuropathic pain. Moreover, selective optogenetic activation of these neurons after nerve injury evokes marked nociceptive behavior. Using a phototherapeutic approach based upon BDNF, the ligand for TrkB, we perform molecule-guided laser ablation of these neurons and achieve long-term retraction of TrkB-positive neurons from the skin and pronounced reversal of mechanical allodynia across multiple types of neuropathic pain. Thus we identify the peripheral neurons which transmit pain from light touch and uncover a novel pharmacological strategy for its treatment.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hyperalgesia/therapy , Laser Therapy , Membrane Glycoproteins/metabolism , Neuralgia/metabolism , Neuralgia/therapy , Protein-Tyrosine Kinases/metabolism , Sensory Receptor Cells/radiation effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Female , Humans , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Ligands , Male , Membrane Glycoproteins/genetics , Mice , Neuralgia/genetics , Neuralgia/physiopathology , Protein-Tyrosine Kinases/genetics , Sensory Receptor Cells/metabolism , Touch/radiation effects
14.
Elife ; 72018 03 09.
Article in English | MEDLINE | ID: mdl-29521261

ABSTRACT

Piezo2 ion channels are critical determinants of the sense of light touch in vertebrates. Yet, their regulation is only incompletely understood. We recently identified myotubularin related protein-2 (Mtmr2), a phosphoinositide (PI) phosphatase, in the native Piezo2 interactome of murine dorsal root ganglia (DRG). Here, we demonstrate that Mtmr2 attenuates Piezo2-mediated rapidly adapting mechanically activated (RA-MA) currents. Interestingly, heterologous Piezo1 and other known MA current subtypes in DRG appeared largely unaffected by Mtmr2. Experiments with catalytically inactive Mtmr2, pharmacological blockers of PI(3,5)P2 synthesis, and osmotic stress suggest that Mtmr2-dependent Piezo2 inhibition involves depletion of PI(3,5)P2. Further, we identified a PI(3,5)P2 binding region in Piezo2, but not Piezo1, that confers sensitivity to Mtmr2 as indicated by functional analysis of a domain-swapped Piezo2 mutant. Altogether, our results propose local PI(3,5)P2 modulation via Mtmr2 in the vicinity of Piezo2 as a novel mechanism to dynamically control Piezo2-dependent mechanotransduction in peripheral sensory neurons.


Subject(s)
Ion Channels/genetics , Mechanotransduction, Cellular/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Sensory Receptor Cells/metabolism , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Ganglia, Spinal/growth & development , Ganglia, Spinal/physiology , Humans , Ion Channels/chemistry , Mice , Osmotic Pressure/physiology , Peripheral Nerves/metabolism , Peripheral Nerves/physiology , Phosphoinositide Phospholipase C/genetics , Phospholipids/chemistry , Phospholipids/genetics , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Sensory Receptor Cells/physiology
15.
Cell Rep ; 21(11): 3102-3115, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241539

ABSTRACT

Mechanical and thermal hyperalgesia (pain hypersensitivity) are cardinal signs of inflammation. Although the mechanism underlying thermal hyperalgesia is well understood, the cellular and molecular basis of mechanical hyperalgesia is poorly described. Here, we have identified a subset of peptidergic C-fiber nociceptors that are insensitive to noxious mechanical stimuli under normal conditions but become sensitized to such stimuli when exposed to the inflammatory mediator nerve growth factor (NGF). Strikingly, NGF did not affect mechanosensitivity of other nociceptors. We show that these mechanoinsensitive "silent" nociceptors are characterized by the expression of the nicotinic acetylcholine receptor subunit alpha-3 (CHRNA3) and that the mechanically gated ion channel PIEZO2 mediates NGF-induced mechanosensitivity in these neurons. Retrograde tracing revealed that CHRNA3+ nociceptors account for ∼50% of all peptidergic nociceptive afferents innervating visceral organs and deep somatic tissues. Hence, our data suggest that NGF-induced "un-silencing" of CHRNA3+ nociceptors significantly contributes to the development of mechanical hyperalgesia during inflammation.


Subject(s)
Hyperalgesia/genetics , Ion Channels/genetics , Mechanotransduction, Cellular , Nerve Growth Factor/pharmacology , Nociceptors/drug effects , Receptors, Nicotinic/genetics , Animals , Biomechanical Phenomena , Evoked Potentials, Somatosensory/drug effects , Evoked Potentials, Somatosensory/physiology , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Ion Channels/metabolism , Mice , Mice, Transgenic , Nociceptors/cytology , Nociceptors/metabolism , Pain/genetics , Pain/metabolism , Pain/physiopathology , Patch-Clamp Techniques , Primary Cell Culture , Receptors, Nicotinic/metabolism
16.
Neuron ; 93(1): 179-193, 2017 Jan 04.
Article in English | MEDLINE | ID: mdl-27989460

ABSTRACT

Painful mechanical stimuli activate multiple peripheral sensory afferent subtypes simultaneously, including nociceptors and low-threshold mechanoreceptors (LTMRs). Using an optogenetic approach, we demonstrate that LTMRs do not solely serve as touch receptors but also play an important role in acute pain signaling. We show that selective activation of neuropeptide Y receptor-2-expressing (Npy2r) myelinated A-fiber nociceptors evokes abnormally exacerbated pain, which is alleviated by concurrent activation of LTMRs in a frequency-dependent manner. We further show that spatial summation of single action potentials from multiple NPY2R-positive afferents is sufficient to trigger nocifensive paw withdrawal, but additional simultaneous sensory input from LTMRs is required for normal well-coordinated execution of this reflex. Thus, our results show that combinatorial coding of noxious and tactile sensory input is required for normal acute mechanical pain signaling. Additionally, we established a causal link between precisely defined neural activity in functionally identified sensory neuron subpopulations and nocifensive behavior and pain.


Subject(s)
Action Potentials , Acute Pain/genetics , Mechanoreceptors/metabolism , Nerve Fibers, Myelinated/metabolism , Neurons/metabolism , Nociception/physiology , Nociceptors/metabolism , Postsynaptic Potential Summation , Animals , Behavior, Animal , Ganglia, Spinal/cytology , Immunohistochemistry , Mice , Nerve Fibers, Myelinated/physiology , Nociceptive Pain , Optogenetics , Pain , Patch-Clamp Techniques , Real-Time Polymerase Chain Reaction , Receptors, Neuropeptide Y/genetics , Receptors, Neuropeptide Y/metabolism , Reflex , Reverse Transcriptase Polymerase Chain Reaction , Touch/physiology
17.
Adv Exp Med Biol ; 898: 265-304, 2016.
Article in English | MEDLINE | ID: mdl-27161233

ABSTRACT

ThermoTRPs are unique channels that mediate Na(+) and Ca(2+) currents in response to changes in ambient temperature. In combination with their activation by other physical and chemical stimuli, they are considered key integrators of environmental cues into neuronal excitability. Furthermore, roles of thermoTRPs in non-neuronal tissues are currently emerging such as insulin secretion in pancreatic ß-cells, and links to cancer. Calcium permeability through thermoTRPs appears a central hallmark for their physiological and pathological activities. Moreover, it is currently being proposed that beyond working as a second messenger, Ca(2+) can function locally by acting on protein complexes near the membrane. Interestingly, thermoTRPs can enhance and expand the inherent plasticity of signalplexes by conferring them temperature, pH and lipid regulation through Ca(2+) signalling. Thus, unveiling the local role of Ca(2+) fluxes induced by thermoTRPs on the dynamics of membrane-attached signalling complexes as well as their significance in cellular processes, are central issues that will expand the opportunities for therapeutic intervention in disorders involving dysfunction of thermoTRP channels.


Subject(s)
Calcium/metabolism , TRPC Cation Channels/metabolism , Animals , Humans , Ion Transport , Permeability , Protein Conformation , TRPC Cation Channels/chemistry
18.
Nat Commun ; 6: 8095, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26311398

ABSTRACT

Hyaluronan (HA) is present in the extracellular matrix of all body tissues, including synovial fluid in joints, in which it behaves as a filter that buffers transmission of mechanical forces to nociceptor nerve endings thereby reducing pain. Using recombinant systems, mouse-cultured dorsal root ganglia (DRG) neurons and in vivo experiments, we found that HA also modulates polymodal transient receptor potential vanilloid subtype 1 (TRPV1) channels. HA diminishes heat, pH and capsaicin (CAP) responses, thus reducing the opening probability of the channel by stabilizing its closed state. Accordingly, in DRG neurons, HA decreases TRPV1-mediated impulse firing and channel sensitization by bradykinin. Moreover, subcutaneous HA injection in mice reduces heat and capsaicin nocifensive responses, whereas the intra-articular injection of HA in rats decreases capsaicin joint nociceptor fibres discharge. Collectively, these results indicate that extracellular HA reduces the excitability of the ubiquitous TRPV1 channel, thereby lowering impulse activity in the peripheral nociceptor endings underlying pain.


Subject(s)
Adjuvants, Immunologic/pharmacology , Hyaluronic Acid/pharmacology , Neurons/drug effects , Nociceptive Pain , Nociceptors/drug effects , Stifle/drug effects , TRPV Cation Channels/drug effects , Animals , Behavior, Animal/drug effects , Bradykinin/pharmacology , CHO Cells , Calcium/metabolism , Capsaicin/pharmacology , Cell Line, Tumor , Cricetulus , Ganglia, Spinal/cytology , HEK293 Cells , Hot Temperature , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Models, Molecular , Mutagenesis, Site-Directed , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Sensory System Agents/pharmacology , Stifle/innervation , TRPA1 Cation Channel , TRPM Cation Channels/drug effects , TRPM Cation Channels/metabolism , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/drug effects , Transient Receptor Potential Channels/metabolism , Vasodilator Agents/pharmacology
19.
Biochim Biophys Acta ; 1848(9): 1818-27, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25838124

ABSTRACT

Transient receptor potential (TRP) proteins are a family of ion channels central for sensory signaling. These receptors and, in particular, those involved in thermal sensing are also involved in pain signaling. Noteworthy, thermosensory receptors are polymodal ion channels that respond to both physical and chemical stimuli, thus integrating different environmental clues. In addition, their activity is modulated by algesic agents and lipidergic substances that are primarily released in pathological states. Lipids and lipid-like molecules have been found that can directly activate some thermosensory channels or modulate their activity by either potentiating or inhibiting it. To date, more than 50 endogenous lipids that can regulate TRP channel activity in sensory neurons have been described, thus representing the majority of known endogenous TRP channel modulators. Lipid modulators of TRP channels comprise lipids from a variety of metabolic pathways, including metabolites of the cyclooxygenase, lipoxygenase and cytochrome-P450 pathways, phospholipids and lysophospholipids. Therefore, TRP-channels are able to integrate and interpret incoming signals from the different metabolic lipid pathways. Taken together, the large number of lipids that can activate, sensitize or inhibit neuronal TRP-channels highlights the pivotal role of these molecules in sensory biology as well as in pain transduction and perception. This article is part of a Special Issue entitled: Lipid-protein interactions. Guest Editors: Amitabha Chattopadhyay and Jean-Marie Ruysschaert.


Subject(s)
Membrane Lipids/metabolism , Pain/physiopathology , Signal Transduction/physiology , Transient Receptor Potential Channels/metabolism , Animals , Humans , Membrane Lipids/chemistry , Models, Molecular , Protein Binding , Protein Structure, Tertiary , Sensory Receptor Cells/metabolism , Transient Receptor Potential Channels/chemistry
20.
J Biol Chem ; 289(41): 28579-94, 2014 Oct 10.
Article in English | MEDLINE | ID: mdl-25157108

ABSTRACT

The ability of transient receptor potential (TRP) channels to sense and respond to environmental and endogenous cues is crucial in animal sensory physiology. The molecular mechanism of channel gating is yet elusive. The TRP box, a conserved region in the N-end of the C terminus domain, has been signaled as pivotal for allosteric activation in TRP channels. Here, we have examined the role of the linker region between the TRPM8 inner gate and the TRP box (referred to as the S6-TRP box linker) to identify structural determinants of channel gating. Stepwise substitutions of segments in the S6-TRP box linker of TRPM8 channel with the cognate TRPV1 channel sequences produced functional chimeric channels, and identified Tyr(981) as a central molecular determinant of channel function. Additionally, mutations in the 986-990 region had a profound impact on channel gating by voltage and menthol, as evidenced by the modulation of the conductance-to-voltage (G-V) relationships. Simulation of G-V curves using an allosteric model for channel activation revealed that these mutations altered the allosteric constants that couple stimuli sensing to pore opening. A molecular model of TRPM8, based on the recently reported TRPV1 structural model, showed that Tyr(981) may lie in a hydrophobic pocket at the end of the S6 transmembrane segment and is involved in inter-subunit interactions with residues from neighbor subunits. The 986-990 region holds intrasubunit interactions between the TRP domain and the S4-S5 linker. These findings substantiate a gating mechanism whereby the TRP domain acts as a coupling domain for efficient channel opening. Furthermore, they imply that protein-protein interactions of the TRP domain may be targets for channel modulation and drug intervention.


Subject(s)
Mutant Chimeric Proteins/chemistry , TRPM Cation Channels/chemistry , TRPV Cation Channels/chemistry , Allosteric Regulation , Amino Acid Sequence , Animals , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Ion Channel Gating , Membrane Potentials , Menthol/metabolism , Models, Molecular , Molecular Sequence Data , Mutant Chimeric Proteins/genetics , Mutant Chimeric Proteins/metabolism , Mutation , Patch-Clamp Techniques , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Rats , Sequence Alignment , Signal Transduction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...