Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 119: 750-766, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710336

ABSTRACT

Chronic pain is a heavily debilitating condition and a huge socio-economic burden, with no efficient treatment. Over the past decade, the gut microbiota has emerged as an important regulator of nervous system's health and disease states. Yet, its contribution to the pathogenesis of chronic somatic pain remains poorly documented. Here, we report that male but not female mice lacking Myosin1a (KO) raised under single genotype housing conditions (KO-SGH) are predisposed to develop chronic pain in response to a peripheral tissue injury. We further underscore the potential of MYO1A loss-of-function to alter the composition of the gut microbiota and uncover a functional connection between the vulnerability to chronic pain and the dysbiotic gut microbiota of KO-SGH males. As such, parental antibiotic treatment modifies gut microbiota composition and completely rescues the injury-induced pain chronicity in male KO-SGH offspring. Furthermore, in KO-SGH males, this dysbiosis is accompanied by a transcriptomic activation signature in the dorsal root ganglia (DRG) macrophage compartment, in response to tissue injury. We identify CD206+CD163- and CD206+CD163+ as the main subsets of DRG resident macrophages and show that both are long-lived and self-maintained and exhibit the capacity to monitor the vasculature. Consistently, in vivo depletion of DRG macrophages rescues KO-SGH males from injury-induced chronic pain underscoring a deleterious role for DRG macrophages in a Myo1a-loss-of function context. Together, our findings reveal gene-sex-microbiota interactions in determining the predisposition to injury-induced chronic pain and point-out DRG macrophages as potential effector cells.


Subject(s)
Chronic Pain , Dysbiosis , Ganglia, Spinal , Gastrointestinal Microbiome , Mice, Knockout , Myosin Type I , Animals , Female , Male , Mice , Chronic Pain/metabolism , Chronic Pain/microbiology , Dysbiosis/metabolism , Ganglia, Spinal/metabolism , Gastrointestinal Microbiome/physiology , Macrophages/metabolism , Mice, Inbred C57BL , Myosin Type I/metabolism
2.
iScience ; 27(4): 109396, 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38510134

ABSTRACT

The recent development of single-cell and single-nucleus RNA sequencing has highlighted the extraordinary diversity of dorsal root ganglia neurons. However, the few available genetic tools limit our understanding of the functional significance of this heterogeneity. We generated a new mouse line expressing the flippase recombinase from the scn10a locus. By crossing Nav1.8Ires-FLPo mice with the AdvillinCre and RC::FL-hM3Dq mouse lines in an intersectional genetics approach, we were able to obtain somatodendritic expression of hM3Dq-mCherry selectively in the Nav1.8 lineage. The bath application of clozapine N-oxide triggered strong calcium responses selectively in mCherry+ neurons. The intraplantar injection of CNO caused robust flinching, shaking, and biting responses accompanied by strong cFos activation in the ipsilateral lumbar spinal cord. The Nav1.8Ires-FLPo mouse model will be a valuable tool for extending our understanding of the in vivo functional specialization of neuronal subsets of the Nav1.8 lineage for which inducible Cre lines are available.

3.
Pain ; 165(1): 192-201, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37578506

ABSTRACT

ABSTRACT: Decreased GABA levels in injury-induced loss of spinal inhibition are still under intense interest and debate. Here, we show that GAD67 haplodeficient mice exhibited a prolonged injury-induced mechanical hypersensitivity in postoperative, inflammatory, and neuropathic pain models. In line with this, we found that loss of 1 copy of the GAD67-encoding gene Gad1 causes a significant decrease in GABA contents in spinal GABAergic neuronal profiles. Consequently, GAD67 haplodeficient males and females were unresponsive to the analgesic effect of diazepam. Remarkably, all these phenotypes were more pronounced in GAD67 haplodeficient females. These mice had significantly much lower amount of spinal GABA content, exhibited an exacerbated pain phenotype during the second phase of the formalin test, developed a longer lasting mechanical hypersensitivity in the chronic constriction injury of the sciatic nerve model, and were unresponsive to the pain relief effect of the GABA-transaminase inhibitor phenylethylidenehydrazine. Our study provides strong evidence for a role of GABA levels in the modulation of injury-induced mechanical pain and suggests a potential role of the GABAergic system in the prevalence of some painful diseases among females.


Subject(s)
Hypersensitivity , Neuralgia , Male , Female , Mice , Animals , Neuralgia/drug therapy , Neuralgia/etiology , Sciatic Nerve/injuries , Pain Management , gamma-Aminobutyric Acid/therapeutic use
4.
Sci Rep ; 13(1): 9515, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37308519

ABSTRACT

Mounting evidence shows sex-related differences in the experience of pain with women suffering more from chronic pain than men. Yet, our understanding of the biological basis underlying those differences remains incomplete. Using an adapted model of formalin-induced chemical/inflammatory pain, we report here that in contrast to male mice, females distinctly display two types of nocifensive responses to formalin, distinguishable by the duration of the interphase. Females in proestrus and in metestrus exhibited respectively a short-lasting and a long-lasting interphase, underscoring the influence of the estrus cycle on the duration of the interphase, rather than the transcriptional content of the dorsal horn of the spinal cord (DHSC). Additionally, deep RNA-sequencing of DHSC showed that formalin-evoked pain was accompanied by a male-preponderant enrichment in genes associated with the immune modulation of pain, revealing an unanticipated contribution of neutrophils. Taking advantage of the male-enriched transcript encoding the neutrophil associated protein Lipocalin 2 (Lcn2) and using flow cytometry, we confirmed that formalin triggered the recruitment of LCN2-expressing neutrophils in the pia mater of spinal meninges, preferentially in males. Our data consolidate the contribution of female estrus cycle to pain perception and provide evidence supporting a sex-specific immune regulation of formalin-evoked pain.


Subject(s)
Chronic Pain , Spinal Cord , Female , Male , Humans , Animals , Mice , Pain Perception , Oncogenes , Formaldehyde
5.
Cell Rep ; 38(13): 110588, 2022 03 29.
Article in English | MEDLINE | ID: mdl-35354022

ABSTRACT

This review highlights recent findings of different amplitude ranges, roles, and modulations of A-type K+ currents (IA) in excitatory (GAD67-GFP-) and inhibitory (GAD67-GFP+) interneurons in mouse spinal cord pain pathways. Endogenous neuropeptides, such as TAFA4, oxytocin, and dynorphin in particular, have been reported to modulate IA in these pain pathways, but only TAFA4 has been shown to fully reverse the opposing modulations that occur selectively in LIIo GAD67-GFP- and LIIi GAD67-GFP+ interneurons following both neuropathic and inflammatory pain. If, as hypothesized here, Kv4 subunits underlie IA in both GAD67-GFP- and GAD67-GFP+ interneurons, then IA diversity in spinal cord pain pathways may depend on the interneuron-subtype-selective expression of Kv4 auxiliary subunits with functionally different N-terminal variants. Thus, IA emerges as a good candidate for explaining the mechanisms underlying injury-induced mechanical hypersensitivity.


Subject(s)
Pain , Spinal Cord , Animals , Cytokines/metabolism , Green Fluorescent Proteins/metabolism , Interneurons/metabolism , Mice , Pain/metabolism , Spinal Cord/metabolism
6.
Brain ; 145(10): 3637-3653, 2022 10 21.
Article in English | MEDLINE | ID: mdl-34957475

ABSTRACT

Patients with bi-allelic loss of function mutations in the voltage-gated sodium channel Nav1.7 present with congenital insensitivity to pain (CIP), whilst low threshold mechanosensation is reportedly normal. Using psychophysics (n = 6 CIP participants and n = 86 healthy controls) and facial electromyography (n = 3 CIP participants and n = 8 healthy controls), we found that these patients also have abnormalities in the encoding of affective touch, which is mediated by the specialized afferents C-low threshold mechanoreceptors (C-LTMRs). In the mouse, we found that C-LTMRs express high levels of Nav1.7. Genetic loss or selective pharmacological inhibition of Nav1.7 in C-LTMRs resulted in a significant reduction in the total sodium current density, an increased mechanical threshold and reduced sensitivity to non-noxious cooling. The behavioural consequence of loss of Nav1.7 in C-LTMRs in mice was an elevation in the von Frey mechanical threshold and less sensitivity to cooling on a thermal gradient. Nav1.7 is therefore not only essential for normal pain perception but also for normal C-LTMR function, cool sensitivity and affective touch.


Subject(s)
NAV1.7 Voltage-Gated Sodium Channel , Pain Insensitivity, Congenital , Animals , Humans , Mice , Mechanoreceptors , NAV1.7 Voltage-Gated Sodium Channel/genetics , Pain Insensitivity, Congenital/genetics , Sodium
7.
Pain ; 163(7): e837-e849, 2022 07 01.
Article in English | MEDLINE | ID: mdl-34561389

ABSTRACT

ABSTRACT: Rheumatoid arthritis is frequently associated with chronic pain that still remains difficult to treat. Targeting nerve growth factor (NGF) seems very effective to reduce pain in at least osteoarthritis and chronic low back pain but leads to some potential adverse events. Our aim was to better understand the involvement of the intracellular signalling pathways activated by NGF through its specific tyrosine kinase type A (TrkA) receptor in the pathophysiology of rheumatoid arthritis using the complete Freund adjuvant model in our knock-in TrkA/C mice. Our multimodal study demonstrated that knock-in TrkA/C mice exhibited a specific decrease of mechanical allodynia, weight-bearing deficit, peptidergic (CGRP+) and sympathetic (TH+) peripheral nerve sprouting in the joints, a reduction in osteoclast activity and bone resorption markers, and a decrease of CD68-positive cells in the joint with no apparent changes in joint inflammation compared with wild-type mice after arthritis. Finally, transcriptomic analysis shows several differences in dorsal root ganglion mRNA expression of putative mechanotransducers, such as acid-sensing ionic channel 3 and TWIK-related arachidonic acid activated K+ channel, as well as intracellular pathways, such as c-Jun, in the joint or dorsal root ganglia. These results suggest that TrkA-specific intracellular signalling pathways are specifically involved in mechanical hypersensitivity and bone alterations after arthritis using TrkA/C mice.


Subject(s)
Arthritis, Rheumatoid , Hyperalgesia , Receptor, trkA , Signal Transduction , Animals , Arthritis, Rheumatoid/complications , Disease Models, Animal , Ganglia, Spinal/metabolism , Hyperalgesia/etiology , Hyperalgesia/metabolism , Mice , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, trkA/genetics
8.
Cell Rep ; 37(4): 109884, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34706225

ABSTRACT

Pain, whether acute or persistent, is a serious medical problem worldwide. However, its management remains unsatisfactory, and new analgesic molecules are required. We show here that TAFA4 reverses inflammatory, postoperative, and spared nerve injury (SNI)-induced mechanical hypersensitivity in male and female mice. TAFA4 requires functional low-density lipoprotein receptor-related proteins (LRPs) because their inhibition by RAP (receptor-associated protein) dose-dependently abolishes its antihypersensitive actions. SNI selectively decreases A-type K+ current (IA) in spinal lamina II outer excitatory interneurons (L-IIo ExINs) and induces a concomitant increase in IA and decrease in hyperpolarization-activated current (Ih) in lamina II inner inhibitory interneurons (L-IIi InhINs). Remarkably, SNI-induced ion current alterations in both IN subtypes were rescued by TAFA4 in an LRP-dependent manner. We provide insights into the mechanism by which TAFA4 reverses injury-induced mechanical hypersensitivity by restoring normal spinal neuron activity and highlight the considerable potential of TAFA4 as a treatment for injury-induced mechanical pain.


Subject(s)
Cytokines/metabolism , Hyperalgesia/metabolism , Pain/metabolism , Potassium/metabolism , Receptors, LDL/metabolism , Spinal Cord Dorsal Horn/metabolism , Animals , CHO Cells , Cricetulus , HEK293 Cells , Humans , Mice , RAW 264.7 Cells
9.
Sci Rep ; 11(1): 13691, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34211067

ABSTRACT

Integrating -omics data with biological networks such as protein-protein interaction networks is a popular and useful approach to interpret expression changes of genes in changing conditions, and to identify relevant cellular pathways, active subnetworks or network communities. Yet, most -omics data integration tools are restricted to static networks and therefore cannot easily be used for analyzing time-series data. Determining regulations or exploring the network structure over time requires time-dependent networks which incorporate time as one component in their structure. Here, we present a method to project time-series data on sequential layers of a multilayer network, thus creating a temporal multilayer network (tMLN). We implemented this method as a Cytoscape app we named TimeNexus. TimeNexus allows to easily create, manage and visualize temporal multilayer networks starting from a combination of node and edge tables carrying the information on the temporal network structure. To allow further analysis of the tMLN, TimeNexus creates and passes on regular Cytoscape networks in form of static versions of the tMLN in three different ways: (i) over the entire set of layers, (ii) over two consecutive layers at a time, (iii) or on one single layer at a time. We combined TimeNexus with the Cytoscape apps PathLinker and AnatApp/ANAT to extract active subnetworks from tMLNs. To test the usability of our app, we applied TimeNexus together with PathLinker or ANAT on temporal expression data of the yeast cell cycle and were able to identify active subnetworks relevant for different cell cycle phases. We furthermore used TimeNexus on our own temporal expression data from a mouse pain assay inducing hindpaw inflammation and detected active subnetworks relevant for an inflammatory response to injury, including immune response, cell stress response and regulation of apoptosis. TimeNexus is freely available from the Cytoscape app store at https://apps.cytoscape.org/apps/TimeNexus .

10.
Nature ; 594(7861): 94-99, 2021 06.
Article in English | MEDLINE | ID: mdl-34012116

ABSTRACT

Inflammation is a defence response to tissue damage that requires tight regulation in order to prevent impaired healing. Tissue-resident macrophages have a key role in tissue repair1, but the precise molecular mechanisms that regulate the balance between inflammatory and pro-repair macrophage responses during healing remain poorly understood. Here we demonstrate a major role for sensory neurons in promoting the tissue-repair function of macrophages. In a sunburn-like model of skin damage in mice, the conditional ablation of sensory neurons expressing the Gαi-interacting protein (GINIP) results in defective tissue regeneration and in dermal fibrosis. Elucidation of the underlying molecular mechanisms revealed a crucial role for the neuropeptide TAFA4, which is produced in the skin by C-low threshold mechanoreceptors-a subset of GINIP+ neurons. TAFA4 modulates the inflammatory profile of macrophages directly in vitro. In vivo studies in Tafa4-deficient mice revealed that TAFA4 promotes the production of IL-10 by dermal macrophages after UV-induced skin damage. This TAFA4-IL-10 axis also ensures the survival and maintenance of IL-10+TIM4+ dermal macrophages, reducing skin inflammation and promoting tissue regeneration. These results reveal a neuroimmune regulatory pathway driven by the neuropeptide TAFA4 that promotes the anti-inflammatory functions of macrophages and prevents fibrosis after tissue damage, and could lead to new therapeutic perspectives for inflammatory diseases.


Subject(s)
Cytokines/metabolism , Macrophages/metabolism , Regeneration , Sensory Receptor Cells/metabolism , Wound Healing , Animals , Cell Survival , Cytokines/deficiency , Disease Models, Animal , Female , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Fibrosis/prevention & control , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Macrophages/radiation effects , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/radiation effects , Skin/pathology , Skin/radiation effects , Sunburn/complications , Sunburn/etiology , Sunburn/metabolism , Sunburn/pathology , Ultraviolet Rays/adverse effects
11.
Nat Commun ; 12(1): 2936, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34006861

ABSTRACT

Host protection against cutaneous herpes simplex virus 1 (HSV-1) infection relies on the induction of a robust adaptive immune response. Here, we show that Nav1.8+ sensory neurons, which are involved in pain perception, control the magnitude of CD8 T cell priming and expansion in HSV-1-infected mice. The ablation of Nav1.8-expressing sensory neurons is associated with extensive skin lesions characterized by enhanced inflammatory cytokine and chemokine production. Mechanistically, Nav1.8+ sensory neurons are required for the downregulation of neutrophil infiltration in the skin after viral clearance to limit the severity of tissue damage and restore skin homeostasis, as well as for eliciting robust CD8 T cell priming in skin-draining lymph nodes by controlling dendritic cell responses. Collectively, our data reveal an important role for the sensory nervous system in regulating both innate and adaptive immune responses to viral infection, thereby opening up possibilities for new therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Nociceptive Pain/immunology , Sensory Receptor Cells/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cytokines/immunology , Cytokines/metabolism , Female , Herpes Simplex/genetics , Herpes Simplex/virology , Herpesvirus 1, Human/physiology , Humans , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , NAV1.8 Voltage-Gated Sodium Channel/immunology , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neutrophil Infiltration/immunology , Nociceptive Pain/genetics , Nociceptive Pain/metabolism , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/virology , Skin/immunology , Skin/metabolism , Skin/virology
12.
Cell Mol Neurobiol ; 41(2): 247-262, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32306148

ABSTRACT

C-nociceptors (C-Ncs) and non-nociceptive C-low threshold mechanoreceptors (C-LTMRs) are two subpopulations of small unmyelinated non-peptidergic C-type neurons of the dorsal root ganglia (DRGs) with central projections displaying a specific pattern of termination in the spinal cord dorsal horn. Although these two subpopulations exist in several animals, remarkable neurochemical differences occur between mammals, particularly rat/humans from one side and mouse from the other. Mouse is widely investigated by transcriptomics. Therefore, we here studied the immunocytochemistry of murine C-type DRG neurons and their central terminals in spinal lamina II at light and electron microscopic levels. We used a panel of markers for peptidergic (CGRP), non-peptidergic (IB4), nociceptive (TRPV1), non-nociceptive (VGLUT3) C-type neurons and two strains of transgenic mice: the TAFA4Venus knock-in mouse to localize the TAFA4+ C-LTMRs, and a genetically engineered ginip mouse that allows an inducible and tissue-specific ablation of the DRG neurons expressing GINIP, a key modulator of GABABR-mediated analgesia. We confirmed that IB4 and TAFA4 did not coexist in small non-peptidergic C-type DRG neurons and separately tagged the C-Ncs and the C-LTMRs. We then showed that TRPV1 was expressed in only about 7% of the IB4+ non-peptidergic C-Ncs and their type Ia glomerular terminals within lamina II. Notably, the selective ablation of GINIP did not affect these neurons, whereas it reduced IB4 labeling in the medial part of lamina II and the density of C-LTMRs glomerular terminals to about one half throughout the entire lamina. We discuss the significance of these findings for interspecies differences and functional relevance.


Subject(s)
Mechanoreceptors/ultrastructure , Myelin Sheath/ultrastructure , Nociceptors/ultrastructure , Peptides/metabolism , Spinal Cord/metabolism , Spinal Cord/ultrastructure , Animals , Calcitonin Gene-Related Peptide/metabolism , Cytokines/metabolism , Ganglia, Spinal/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice, Transgenic , Plant Lectins/metabolism , Sensory Receptor Cells/metabolism , Spinal Cord Dorsal Horn/metabolism , TRPV Cation Channels/metabolism
13.
Cell Rep ; 30(3): 602-610.e6, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31968239

ABSTRACT

C-LTMRs are known to convey affective aspects of touch and to modulate injury-induced pain in humans and mice. However, a role for these neurons in temperature sensation has been suggested, but not fully demonstrated. Here, we report that deletion of C-low-threshold mechanoreceptor (C-LTMR)-expressed bhlha9 causes impaired thermotaxis behavior and exacerbated formalin-evoked pain in male, but not female, mice. Positive modulators of GABAA receptors failed to relieve inflammatory formalin pain and failed to decrease the frequency of spontaneous excitatory post-synaptic currents (sEPSCs) selectively in bhlha9 knockout (KO) males. This could be explained by a drastic change in the GABA content of lamina II inner inhibitory interneurons contacting C-LTMR central terminals. Finally, C-LTMR-specific deep RNA sequencing revealed more genes differentially expressed in male than in female bhlha9 KO C-LTMRs. Our data consolidate the role of C-LTMRs in modulation of formalin pain and provide in vivo evidence of their role in the discriminative aspects of temperature sensation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/deficiency , Pain/pathology , Sex Characteristics , Taxis Response , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Formaldehyde , Ganglia, Spinal/pathology , Gene Expression Regulation , Interneurons/metabolism , Male , Mechanoreceptors/metabolism , Mice, Knockout , Spinal Cord/pathology , Synaptic Transmission , gamma-Aminobutyric Acid/metabolism
14.
Pain ; 161(5): 1109-1123, 2020 05.
Article in English | MEDLINE | ID: mdl-31977937

ABSTRACT

Mechanical allodynia is a cardinal sign of several inflammatory pain disorders where nerve growth factor, a prototypic neurotrophin, plays a crucial role by binding to TrkA receptors. Here, we took the advantage of our generated knock-in mouse model expressing a chimeric TrkA/TrkC receptor that seems to not specifically develop mechanical allodynia after inflammation, to identify the TrkA downstream pathways involved in this phenomenon. We confirmed and extended that disrupting TrkA-specific pathways leads to a specific deficit in mechanical hypersensitivity development after somatic (systemic nerve growth factor administration and paw incision) and, to a lesser extent, visceral injuries. Despite a deficit in thin, mainly peptidergic, fibre innervation in TrkAC mice, thermal hyperalgesia development was not different from WT mice. Inflammatory reaction (oedema, IL-6 content), pain behaviours after intraplantar capsaicin, as well as TRPV1 calcium imaging response of dorsal root ganglion neurons were similar between TrkAC and WT mice. This deficiency in mechanical allodynia development in TrkAC mice is likely due to the alteration of the expression of different TrkA transduction pathways (ie, Akt, p38 MAPK, and c-Jun) especially p38 MAPK, in the dorsal root ganglion cell bodies, ultimately leading to an alteration of at least, ASIC3 channel overexpression, known to participate in nociceptor mechanosensory function.


Subject(s)
Hyperalgesia , Animals , Ganglia, Spinal , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System , Mice , Nerve Growth Factor/genetics , Receptor, trkA/genetics , Receptor, trkC , p38 Mitogen-Activated Protein Kinases
15.
Nat Commun ; 10(1): 4137, 2019 09 12.
Article in English | MEDLINE | ID: mdl-31515492

ABSTRACT

Developmental cell death plays an important role in the construction of functional neural circuits. In vertebrates, the canonical view proposes a selection of the surviving neurons through stochastic competition for target-derived neurotrophic signals, implying an equal potential for neurons to compete. Here we show an alternative cell fitness selection of neurons that is defined by a specific neuronal heterogeneity code. Proprioceptive sensory neurons that will undergo cell death and those that will survive exhibit different molecular signatures that are regulated by retinoic acid and transcription factors, and are independent of the target and neurotrophins. These molecular features are genetically encoded, representing two distinct subgroups of neurons with contrasted functional maturation states and survival outcome. Thus, in this model, a heterogeneous code of intrinsic cell fitness in neighboring neurons provides differential competitive advantage resulting in the selection of cells with higher capacity to survive and functionally integrate into neural networks.


Subject(s)
Models, Biological , Sensory Receptor Cells/cytology , Animals , Cell Death/drug effects , Cell Survival/drug effects , Chick Embryo , Core Binding Factor Alpha 3 Subunit/metabolism , Mice, Inbred C57BL , Proprioception/drug effects , Receptor, trkC/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology
16.
Sci Rep ; 9(1): 3112, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30816223

ABSTRACT

The T-type calcium channel, Cav3.2, is necessary for acute pain perception, as well as mechanical and cold allodynia in mice. Being found throughout sensory pathways, from excitatory primary afferent neurons up to pain matrix structures, it is a promising target for analgesics. In our study, Cav3.2 was detected in ~60% of the lamina II (LII) neurons of the spinal cord, a site for integration of sensory processing. It was co-expressed with Tlx3 and Pax2, markers of excitatory and inhibitory interneurons, as well as nNOS, calretinin, calbindin, PKCγ and not parvalbumin. Non-selective T-type channel blockers slowed the inhibitory but not the excitatory transmission in LII neurons. Furthermore, T-type channel blockers modified the intrinsic properties of LII neurons, abolishing low-threshold activated currents, rebound depolarizations, and blunting excitability. The recording of Cav3.2-positive LII neurons, after intraspinal injection of AAV-DJ-Cav3.2-mcherry, showed that their intrinsic properties resembled those of the global population. However, Cav3.2 ablation in the dorsal horn of Cav3.2GFP-Flox KI mice after intraspinal injection of AAV-DJ-Cav3.2-Cre-IRES-mcherry, had drastic effects. Indeed, it (1) blunted the likelihood of transient firing patterns; (2) blunted the likelihood and the amplitude of rebound depolarizations, (3) eliminated action potential pairing, and (4) remodeled the kinetics of the action potentials. In contrast, the properties of Cav3.2-positive neurons were only marginally modified in Cav3.1 knockout mice. Overall, in addition to their previously established roles in the superficial spinal cord and in primary afferent neurons, Cav3.2 channel appear to be necessary for specific, significant and multiple controls of LII neuron excitability.


Subject(s)
Calcium Channels, T-Type/metabolism , Neurons/cytology , Spinal Nerves/cytology , Action Potentials , Animals , Hyperalgesia/metabolism , Mice , Neurons/metabolism , Patch-Clamp Techniques , Spinal Nerves/metabolism , Synaptic Transmission
17.
Sci Signal ; 11(561)2018 12 18.
Article in English | MEDLINE | ID: mdl-30563864

ABSTRACT

Irritable bowel syndrome (IBS) is a common gastrointestinal disorder that is characterized by chronic abdominal pain concurrent with altered bowel habit. Polyunsaturated fatty acid (PUFA) metabolites are increased in abundance in IBS and are implicated in the alteration of sensation to mechanical stimuli, which is defined as visceral hypersensitivity. We sought to quantify PUFA metabolites in patients with IBS and evaluate their role in pain. Quantification of PUFA metabolites by mass spectrometry in colonic biopsies showed an increased abundance of 5-oxoeicosatetraenoic acid (5-oxoETE) only in biopsies taken from patients with IBS with predominant constipation (IBS-C). Local administration of 5-oxoETE to mice induced somatic and visceral hypersensitivity to mechanical stimuli without causing tissue inflammation. We found that 5-oxoETE directly acted on both human and mouse sensory neurons as shown by lumbar splanchnic nerve recordings and Ca2+ imaging of dorsal root ganglion (DRG) neurons. We showed that 5-oxoETE selectively stimulated nonpeptidergic, isolectin B4 (IB4)-positive DRG neurons through a phospholipase C (PLC)- and pertussis toxin-dependent mechanism, suggesting that the effect was mediated by a G protein-coupled receptor (GPCR). The MAS-related GPCR D (Mrgprd) was found in mouse colonic DRG afferents and was identified as being implicated in the noxious effects of 5-oxoETE. Together, these data suggest that 5-oxoETE, a potential biomarker of IBS-C, induces somatic and visceral hyperalgesia without inflammation in an Mrgprd-dependent manner. Thus, 5-oxoETE may play a pivotal role in the abdominal pain associated with IBS-C.


Subject(s)
Arachidonic Acids/metabolism , Irritable Bowel Syndrome/pathology , Nociception , Receptors, G-Protein-Coupled/physiology , Sensory Receptor Cells/pathology , Animals , Calcium/metabolism , Case-Control Studies , Colon/drug effects , Colon/metabolism , Colon/pathology , Constipation/chemically induced , Constipation/physiopathology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Humans , Irritable Bowel Syndrome/etiology , Irritable Bowel Syndrome/metabolism , Male , Mice , Mice, Inbred C57BL , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction
18.
Cell Rep ; 22(11): 2886-2897, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29539418

ABSTRACT

C-low-threshold mechanoreceptors (C-LTMRs) are sensory neurons that, beyond conveying pleasant touch, modulate nociceptive transmission within the spinal cord. However, pain alleviation by C-LTMRs remains poorly understood. Here, we show that the C-LTMR-derived TAFA4 chemokine induces a reinforcement of inhibitory synaptic transmission within spinal networks, which consequently depresses local excitatory synapses and impairs synaptic transmission from high-threshold C-fibers. In animals with inflammation induced by Freund's complete adjuvant, TAFA4 decreases the noxious stimulus-induced neuronal responses recorded in vivo and alleviates mechanical pain. Both effects are blocked by antagonists of GABAergic transmission. Furthermore, TAFA4 promotes microglial retraction in inflammation and increases the number of inhibitory synapses on lamina IIi somata. Altogether, these results demonstrate GABAergic interneurons to be the first integration relay for C-LTMRs and highlight a tight interplay between sensory neurons, microglial cells, and spinal interneurons, which fine-tunes inhibitory activity and nociceptive transmission in pathological conditions.


Subject(s)
Cytokines/pharmacology , GABAergic Neurons/drug effects , Hyperalgesia/drug therapy , Animals , GABAergic Neurons/pathology , Hyperalgesia/pathology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , Nociception/drug effects , Nociception/physiology , Patch-Clamp Techniques , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/pathology , Synaptic Transmission
19.
J Invest Dermatol ; 138(3): 688-696, 2018 03.
Article in English | MEDLINE | ID: mdl-29054601

ABSTRACT

Sensing environmental temperature is a key factor allowing individuals to maintain thermal homeostasis via thermoregulatory mechanisms, including changes to skin blood flow. Among transient receptor potential channels, transient receptor potential vanilloid 3 (TRPV3) is a heat-activated cation channel highly expressed in keratinocytes. However, the role of TRPV3 in triggering heat-evoked cutaneous vasodilation is unknown. Using a murine in vivo model of local acute environmental heat exposure in the skin, we show that TRPV3 is involved in the local thermoregulatory control of skin blood flow by initiating the release of calcitonin gene-related peptide and nitric oxide in response to local heating of the skin. In addition to their contribution in local heat-evoked vasodilation, TRPV3, calcitonin gene-related peptide, and nitric oxide also contribute to internal body temperature stability during passive whole-body heating. This study provides in vivo demonstration of the role of TRPV3 as a strong modulator of cutaneous vascular thermoregulatory mechanisms.


Subject(s)
Body Temperature Regulation/physiology , Calcitonin Gene-Related Peptide/physiology , TRPV Cation Channels/physiology , Vasodilation/physiology , Animals , Hot Temperature , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/biosynthesis , Skin/blood supply
20.
Sci Rep ; 7: 43493, 2017 02 27.
Article in English | MEDLINE | ID: mdl-28240741

ABSTRACT

Primary sensory neurons are heterogeneous by myriad of molecular criteria. However, the functional significance of this remarkable heterogeneity is just emerging. We precedently described the GINIP+ neurons as a new subpopulation of non peptidergic C-fibers encompassing the free nerve ending cutaneous MRGPRD+ neurons and C-LTMRs. Using our recently generated ginip mouse model, we have been able to selectively ablate the GINIP+ neurons and assess their functional role in the somatosensation. We found that ablation of GINIP+ neurons affected neither the molecular contents nor the central projections of the spared neurons. GINIP-DTR mice exhibited impaired sensation to gentle mechanical stimuli applied to their hairy skin and had normal responses to noxious mechanical stimuli applied to their glabrous skin, under acute and injury-induced conditions. Importantly, loss of GINIP+ neurons significantly altered formalin-evoked first pain and drastically suppressed the second pain response. Given that MRGPRD+ neurons have been shown to be dispensable for formalin-evoked pain, our study suggest that C-LTMRs play a critical role in the modulation of formalin-evoked pain.


Subject(s)
Gene Expression , Intracellular Signaling Peptides and Proteins/genetics , Pain/etiology , Sensory Receptor Cells/metabolism , Animals , Biomarkers , Disease Models, Animal , Formaldehyde/adverse effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Ganglia, Spinal/physiopathology , Gene Knockdown Techniques , Genotype , Male , Mice , Mice, Knockout , Organ Specificity/genetics , Pain/metabolism , Pain/physiopathology , Physical Stimulation , Sensory Thresholds , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...