Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 43(15): 2803-2814, 2023 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-36898840

RESUMO

The detection of environmental temperatures is critical for survival, yet inappropriate responses to thermal stimuli can have a negative impact on overall health. The physiological effect of cold is distinct among somatosensory modalities in that it is soothing and analgesic, but also agonizing in the context of tissue damage. Inflammatory mediators produced during injury activate nociceptors to release neuropeptides, such as calcitonin gene-related peptide (CGRP) and substance P, inducing neurogenic inflammation, which further exasperates pain. Many inflammatory mediators induce sensitization to heat and mechanical stimuli but, conversely, inhibit cold responsiveness, and the identity of molecules inducing cold pain peripherally is enigmatic, as are the cellular and molecular mechanisms altering cold sensitivity. Here, we asked whether inflammatory mediators that induce neurogenic inflammation via the nociceptive ion channels TRPV1 (vanilloid subfamily of transient receptor potential channel) and TRPA1 (transient receptor potential ankyrin 1) lead to cold pain in mice. Specifically, we tested cold sensitivity in mice after intraplantar injection of lysophosphatidic acid or 4-hydroxy-2-nonenal, finding that each induces cold pain that is dependent on the cold-gated channel transient receptor potential melastatin 8 (TRPM8). Inhibition of CGRP, substance P, or toll-like receptor 4 (TLR4) signaling attenuates this phenotype, and each neuropeptide produces TRPM8-dependent cold pain directly. Further, the inhibition of CGRP or TLR4 signaling alleviates cold allodynia differentially by sex. Last, cold pain induced by both inflammatory mediators and neuropeptides requires TRPM8, as well as the neurotrophin artemin and its receptor GDNF receptor α3 (GFRα3). These results are consistent with artemin-induced cold allodynia requiring TRPM8, demonstrating that neurogenic inflammation alters cold sensitivity via localized artemin release that induces cold pain via GFRα3 and TRPM8.SIGNIFICANCE STATEMENT The cellular and molecular mechanisms that generate pain are complex with a diverse array of pain-producing molecules generated during injury that act to sensitize peripheral sensory neurons, thereby inducing pain. Here we identify a specific neuroinflammatory pathway involving the ion channel TRPM8 (transient receptor potential cation channel subfamily M member 8) and the neurotrophin receptor GFRα3 (GDNF receptor α3) that leads to cold pain, providing select targets for potential therapies for this pain modality.


Assuntos
Nociceptores , Canais de Cátion TRPM , Animais , Camundongos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Temperatura Baixa , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hiperalgesia/metabolismo , Inflamação Neurogênica/metabolismo , Dor/metabolismo , Células Receptoras Sensoriais/fisiologia , Substância P/metabolismo , Substância P/farmacologia , Receptor 4 Toll-Like/metabolismo , Canal de Cátion TRPA1 , Canais de Cátion TRPM/metabolismo , Canais de Cátion TRPV/metabolismo , Masculino , Feminino
2.
bioRxiv ; 2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36747719

RESUMO

The detection of environmental temperatures is critical for survival, yet inappropriate responses to thermal stimuli can have a negative impact on overall health. The physiological effect of cold is distinct among somatosensory modalities in that it is soothing and analgesic, but also agonizing in the context of tissue damage. Inflammatory mediators produced during injury activate nociceptors to release neuropeptides, such as CGRP and substance P, inducing neurogenic inflammation which further exasperates pain. Many inflammatory mediators induce sensitization to heat and mechanical stimuli but, conversely, inhibit cold responsiveness, and the identity of molecules inducing cold pain peripherally is enigmatic, as are the cellular and molecular mechanisms altering cold sensitivity. Here, we asked if inflammatory mediators that induce neurogenic inflammation via the nociceptive ion channels TRPV1 and TRPA1 lead to cold pain in mice. Specifically, we tested cold sensitivity in mice after intraplantar injection of lysophosphatidic acid (LPA) or 4-hydroxy-2-nonenal (4HNE), finding each induces cold pain that is dependent on the cold-gated channel TRPM8. Inhibition of either CGRP, substance P, or toll-like receptor 4 (TLR4) signaling attenuates this phenotype, and each neuropeptide produces TRPM8-dependent cold pain directly. Further, the inhibition of CGRP or TLR4 signaling alleviates cold allodynia differentially by sex. Lastly, we find that cold pain induced by inflammatory mediators and neuropeptides requires the neurotrophin artemin and its receptor GFRα3. These results demonstrate that tissue damage alters cold sensitivity via neurogenic inflammation, likely leading to localized artemin release that induces cold pain via GFRα3 and TRPM8. Significance Statement: The cellular and molecular mechanisms that generate pain are complex with a diverse array of pain-producing molecules generated during injury that act to sensitize peripheral sensory neurons, thereby inducing pain. Here we identify a specific neuroinflammatory pathway involving the ion channel TRPM8 and the neurotrophin receptor GFRα3 that leads to cold pain, providing select targets for potential therapies for this pain modality.

3.
Nucleic Acids Res ; 50(20): 11738-11754, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-36321646

RESUMO

We describe a purified biochemical system to produce monoclonal antibodies (Abs) in vitro using activation-induced deoxycytidine deaminase (AID) and DNA polymerase η (Polη) to diversify immunoglobulin variable gene (IgV) libraries within a phage display format. AID and Polη function during B-cell affinity maturation by catalyzing somatic hypermutation (SHM) of immunoglobulin variable genes (IgV) to generate high-affinity Abs. The IgV mutational motif specificities observed in vivo are conserved in vitro. IgV mutations occurred in antibody complementary determining regions (CDRs) and less frequently in framework (FW) regions. A unique feature of our system is the use of AID and Polη to perform repetitive affinity maturation on libraries reconstructed from a preceding selection step. We have obtained scFv Abs against human glucagon-like peptide-1 receptor (GLP-1R), a target in the treatment of type 2 diabetes, and VHH nanobodies targeting Fatty Acid Amide Hydrolase (FAAH), involved in chronic pain, and artemin, a neurotropic factor that regulates cold pain. A round of in vitro affinity maturation typically resulted in a 2- to 4-fold enhancement in Ab-Ag binding, demonstrating the utility of the system. We tested one of the affinity matured nanobodies and found that it reduced injury-induced cold pain in a mouse model.


Assuntos
Anticorpos de Cadeia Única , Anticorpos de Domínio Único , Hipermutação Somática de Imunoglobulina , Animais , Humanos , Camundongos , Afinidade de Anticorpos/genética , Citidina Desaminase/metabolismo , Diabetes Mellitus Tipo 2 , Região Variável de Imunoglobulina/genética , Dor , Anticorpos de Domínio Único/genética , Anticorpos de Cadeia Única/genética
4.
Pain ; 163(12): 2380-2389, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-35353773

RESUMO

ABSTRACT: Migraine is a complex neurovascular disorder that is one of the leading causes of disability and a reduced quality of life. Even with such a high societal impact, our understanding of the cellular and molecular mechanisms that contribute to migraine headaches is limited. To address this complex disorder, several groups have performed genome-wide association studies to elucidate migraine susceptibility genes, with many identifying transient receptor potential melastatin 8 (TRPM8), a cold-sensitive cation channel expressed in peripheral afferents innervating the trigeminovascular system, and the principal mediator of cold and cold pain associated with injury and disease. Interestingly, these migraine-associated single-nucleotide polymorphisms reside in noncoding regions of TRPM8, with those correlated with reduced migraine risk exhibiting lower TRPM8 expression and decreased cold sensitivity. Nonetheless, as a role for TRPM8 in migraine has yet to be defined, we sought to address this gap in our knowledge using mouse genetics and TRPM8 antagonism to determine whether TRPM8 channels or neurons are required for migraine-like pain (mechanical allodynia and facial grimace) in inducible migraine models. Our results show that both evoked and spontaneous pain behaviors are dependent on both TRPM8 channels and neurons, as well as required in both acute and chronic migraine models. Moreover, inhibition of TRPM8 channels prevented acute but not established chronic migraine-like pain. These results are consistent with its association with migraine in genetic analyses and establish that TRPM8 channels are a component of the underlying mechanisms of migraine.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina , Transtornos de Enxaqueca , Canais de Cátion TRPM , Animais , Camundongos , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Temperatura Baixa , Estudo de Associação Genômica Ampla , Transtornos de Enxaqueca/induzido quimicamente , Transtornos de Enxaqueca/genética , Transtornos de Enxaqueca/metabolismo , Nitroglicerina/toxicidade , Dor , Qualidade de Vida , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo
5.
Pain ; 162(2): 609-618, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32826761

RESUMO

ABSTRACT: The proper detection and behavioral response to painfully cold temperatures is critical for avoiding potentially harmful tissue damage. Cold allodynia and hyperalgesia, pain associated with innocuous cooling and exaggerated pain with noxious cold, respectively, are common in patients with chronic pain. In peripheral somatosensory afferents, the ion channels transient receptor potential melastatin 8 (TRPM8) and transient receptor potential ankyrin 1 (TRPA1) are candidate receptors for innocuous and noxious cold temperatures, respectively. However, the role of TRPA1 as a cold sensor has remained controversial, and recent evidence suggests that TRPM8 channels and afferents mediate the detection of both pleasant and painful cold. To determine the role of TRPA1 afferents in cold-induced mouse behaviors in vivo, we used functional phenotyping by targeted nerve conduction block with the cell-impermeant lidocaine derivative QX-314. Surprisingly, we find that injection of QX-314 with TRPA1 agonists reduces cold-induced behaviors in mice, but does so in a TRPM8-dependent manner. Moreover, this effect is sexually dimorphic and requires the glial cell line-derived neurotrophic factor receptor GFRα3, as does cold hypersensitivity produced by the activation of TRPA1 channels. Taken together, these results suggest that under conditions of neurogenic inflammation, TRPA1 works upstream of GFRα3 and TRPM8 to produce cold hypersensitivity, providing novel insights into the role of TRPA1 channels in cold pain.


Assuntos
Mentol , Canais de Cátion TRPM , Animais , Anquirinas/genética , Temperatura Baixa , Humanos , Mentol/farmacologia , Camundongos , Inflamação Neurogênica , Nociceptividade , Receptores de Fator de Crescimento Neural , Canal de Cátion TRPA1/genética , Canais de Cátion TRPM/genética
6.
Sci Rep ; 9(1): 20324, 2019 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-31889131

RESUMO

Chemotherapy induced peripheral neuropathy (CIPN), a toxic side effect of some cancer treatments, negatively impacts patient outcomes and drastically reduces survivor's quality of life (QOL). Uncovering the mechanisms driving chemotherapy-induced CIPN is urgently needed to facilitate the development of effective treatments, as currently there are none. Observing that C57BL/6 (B6) and 129SvEv (129) mice are respectively sensitive and resistant to Paclitaxel-induced pain, we investigated the involvement of the gut microbiota in this extreme phenotypic response. Reciprocal gut microbiota transfers between B6 and 129 mice as well as antibiotic depletion causally linked gut microbes to Paclitaxel-induced pain sensitivity and resistance. Microglia proliferated in the spinal cords of Paclitaxel treated mice harboring the pain-sensitive B6 microbiota but not the pain-resistant 129 microbiota, which exhibited a notable absence of infiltrating immune cells. Paclitaxel decreased the abundance of Akkermansia muciniphila, which could compromise barrier integrity resulting in systemic exposure to bacterial metabolites and products - that acting via the gut-immune-brain axis - could result in altered brain function. Other bacterial taxa that consistently associated with both bacteria and pain as well as microglia and pain were identified, lending support to our hypothesis that microglia are causally involved in CIPN, and that gut bacteria are drivers of this phenotype.


Assuntos
Antineoplásicos/efeitos adversos , Microbioma Gastrointestinal , Neuralgia/etiologia , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Biodiversidade , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Masculino , Camundongos , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuralgia/metabolismo , Paclitaxel/efeitos adversos , Paclitaxel/farmacologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
7.
Handb Clin Neurol ; 156: 57-67, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30454609

RESUMO

Of somatosensory modalities cold is one of the more ambiguous percepts, evoking the pleasant sensation of cooling, the stinging bite of cold pain, and welcome relief from chronic pain. Moreover, unlike the precipitous thermal thresholds for heat activation of thermosensitive afferent neurons, thresholds for cold fibers are across a range of cool to cold temperatures that spans over 30°C. Until recently, how cold produces this myriad of biologic effects was unknown. However, recent advances in our understanding of cold mechanisms at the behavioral, physiologic, and cellular level have begun to provide insights into this sensory modality. The identification of a number of ion channels that either serve as the principal detectors of a cold stimulus in the peripheral nervous system, or are part of a differential expression pattern of channels that maintain cell excitability in the cold, endows select neurons with properties that are amenable to electric signaling in the cold. This chapter highlights the current understanding of the molecules involved in cold transduction in the mammalian peripheral nervous system, as well as presenting a hypothetic model to account for the broad range of cold thermal thresholds and distinct functions of cold fibers in perception, pain, and analgesia.


Assuntos
Regulação da Temperatura Corporal/fisiologia , Temperatura Baixa , Canais Iônicos/fisiologia , Transdução de Sinais/fisiologia , Sensação Térmica/fisiologia , Animais , Humanos , Nervos Periféricos/fisiologia , Células Receptoras Sensoriais/fisiologia
8.
Commun Biol ; 1: 53, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30271936

RESUMO

Treatment of pain with local anesthetics leads to an unfavorable decrease in general sensory acuity due to their indiscriminate block of both pain sensing (nociceptors) and non-pain sensing nerves. However, the cell impermeant lidocaine derivative QX-314 can be selectively targeted to only nociceptors by permeation through ligand-gated cation channels. Here we show that localized injection of QX-314 with agonists for the menthol receptor TRPM8 specifically blocks cold-evoked behaviors in mice, including cold allodynia and hyperalgesia. Remarkably, cooling stimuli also promotes QX-314-mediated inhibition of cold behaviors, and can be used to block cold allodynia, while retaining relatively normal cold sensation. The effects of both agonist and thermally evoked uptake of QX-314 are TRPM8-dependent, results demonstrating an effective approach to treat localized cold pain without altering general somatosensation.

9.
J Invest Dermatol ; 138(6): 1391-1399, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29288650

RESUMO

Cooling or the application of mentholated liniments to the skin has been used to treat itch for centuries, yet remarkably little is known about how counter-stimuli such as these induce itch relief. Indeed, there is no clear consensus in the scientific literature as to whether or not cooling does in fact block the transduction of itch signals or if it is simply a placebo effect. This gap in our understanding led us to hypothesize that cooling is antipruritic and, like cooling analgesia, requires function of the cold-gated ion channel TRPM8, a receptor for menthol expressed on peripheral afferent nerve endings. Using a combination of pharmacologic, genetic, and mouse behavioral assays, we find that cooling inhibits both histaminergic and non-histaminergic itch pathways, and that inhibition of itch by cooling requires TRPM8 channels or intact and functional TRPM8-expressing afferent neurons. The cold mimetic menthol is also effective in ameliorating itch in a TRPM8-dependent manner. Moreover, we find that chronic itch can be ameliorated by cooling, demonstrating that this counter-stimulus activates a specific neural circuit that leads to broad itch relief and a potential cellular mechanism for treatment of chronic itch.


Assuntos
Antipruriginosos/farmacologia , Crioterapia , Prurido/terapia , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Antipruriginosos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Capsaicina/toxicidade , Cloroquina/toxicidade , Doença Crônica/terapia , Modelos Animais de Doenças , Histamina/toxicidade , Humanos , Mentol/farmacologia , Mentol/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prurido/induzido quimicamente , Prurido/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Pele/efeitos dos fármacos , Pele/inervação , Pele/metabolismo , Pele/patologia , Resultado do Tratamento
10.
Neurosci Lett ; 639: 59-67, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-28038937

RESUMO

While most membrane channels are only capable of passing small ions, certain non-selective cation channels have been recently shown to have the capacity to permeate large cations. The mechanisms underlying large molecule permeation are unclear, but this property has been exploited pharmacologically to target molecules, such as nerve conduction blockers, to specific subsets of pain-sensing neurons (nociceptors) expressing the heat-gated transient receptor potential (TRP) channel TRPV1. However, it is not clear if the principal mediator of cold stimuli TRPM8 is capable of mediating the permeation large molecules across cell membranes, suggesting that TRPM8-positive nerves cannot be similarly targeted. Here we show that both heterologous cells and native sensory neurons expressing TRPM8 channels allow the permeation of the large fluorescent cation Po-Pro3. Po-Pro3 influx is blocked by TRPM8-specific antagonism and when channel activity is desensitized. The effects of the potent agonist WS-12 are TRPM8-specific and dye uptake mediated by TRPM8 channels is similar to that observed with TRPV1. Lastly, we find that as with TRPV1, activation of TRPM8 channels can be used as a means to target intracellular uptake of cell-impermeable sodium channel blockers. In a neuronal cell line expressing TRPM8 channels, voltage-gated sodium currents are blocked in the presence of the cell-impermeable, charged lidocaine derivative QX-314 and WS-12. These results show that the ability of somatosensory TRP channels to promote the permeation of large cations also includes TRPM8, thereby suggesting that novel approaches to alter cold pain can also be employed via conduction block in TRPM8-positive sensory neurons.


Assuntos
Nociceptores/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Anestésicos Locais/farmacologia , Animais , Linhagem Celular , Gânglios Espinais/metabolismo , Humanos , Lidocaína/análogos & derivados , Lidocaína/farmacologia , Dor/tratamento farmacológico , Dor/metabolismo , Ratos , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Sensação Térmica/efeitos dos fármacos , Sensação Térmica/fisiologia
11.
Proc Natl Acad Sci U S A ; 113(16): 4506-11, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27051069

RESUMO

Tissue injury prompts the release of a number of proalgesic molecules that induce acute and chronic pain by sensitizing pain-sensing neurons (nociceptors) to heat and mechanical stimuli. In contrast, many proalgesics have no effect on cold sensitivity or can inhibit cold-sensitive neurons and diminish cooling-mediated pain relief (analgesia). Nonetheless, cold pain (allodynia) is prevalent in many inflammatory and neuropathic pain settings, with little known of the mechanisms promoting pain vs. those dampening analgesia. Here, we show that cold allodynia induced by inflammation, nerve injury, and chemotherapeutics is abolished in mice lacking the neurotrophic factor receptor glial cell line-derived neurotrophic factor family of receptors-α3 (GFRα3). Furthermore, established cold allodynia is blocked in animals treated with neutralizing antibodies against the GFRα3 ligand, artemin. In contrast, heat and mechanical pain are unchanged, and results show that, in striking contrast to the redundant mechanisms sensitizing other modalities after an insult, cold allodynia is mediated exclusively by a single molecular pathway, suggesting that artemin-GFRα3 signaling can be targeted to selectively treat cold pain.


Assuntos
Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Hiperalgesia/metabolismo , Dor/metabolismo , Transdução de Sinais , Animais , Anticorpos Neutralizantes/farmacologia , Modelos Animais de Doenças , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/antagonistas & inibidores , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Hiperalgesia/genética , Hiperalgesia/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Dor/genética , Dor/patologia
12.
J Cell Biol ; 210(6): 891-8, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26347141

RESUMO

Myelin oligodendrocyte glycoprotein (MOG) is a central nervous system myelin-specific molecule expressed on the outer lamellae of myelin. To date, the exact function of MOG has remained unknown, with MOG knockout mice displaying normal myelin ultrastructure and no apparent specific phenotype. In this paper, we identify nerve growth factor (NGF) as a binding partner for MOG and demonstrate that this interaction is capable of sequestering NGF from TrkA-expressing neurons to modulate axon growth and survival. Deletion of MOG results in aberrant sprouting of nociceptive neurons in the spinal cord. Binding of NGF to MOG may offer widespread implications into mechanisms that underlie pain pathways.


Assuntos
Axônios/metabolismo , Gânglios Espinais/metabolismo , Glicoproteína Mielina-Oligodendrócito/metabolismo , Fator de Crescimento Neural/metabolismo , Oligodendroglia/metabolismo , Medula Espinal/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Sobrevivência Celular , Técnicas de Cocultura , Cricetulus , Gânglios Espinais/patologia , Genótipo , Camundongos Knockout , Dados de Sequência Molecular , Glicoproteína Mielina-Oligodendrócito/deficiência , Glicoproteína Mielina-Oligodendrócito/genética , Fenótipo , Ligação Proteica , Ratos Sprague-Dawley , Receptor trkA/metabolismo , Transdução de Sinais , Medula Espinal/patologia , Transfecção
13.
Curr Opin Neurobiol ; 34: 14-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25622298

RESUMO

Over a decade and a half of intensive study has shown that the Transient Receptor Potential family ion channels TRPV1 and TRPM8 are the primary sensors of heat and cold temperatures in the peripheral nervous system. TRPV homologues and TRPA1 are also implicated, but recent genetic evidence has diminished their significance in thermosensation and suggests that a number of newly identified thermosensitive channels, including TRPM3, two-pore potassium channels, and the chloride channel Ano1, require further consideration. In addition to novel thermostransducers, recent genetic and pharmacological approaches have begun to elucidate the afferent neurocircuits underlying temperature sensation, continuing the rapid expansion in our understanding of the cellular and molecular basis of thermosensation that began with the discovery of TRPV1 and TRPM8.


Assuntos
Canais Iônicos , Sensação Térmica , Animais , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Mamíferos , Sensação Térmica/fisiologia
14.
PLoS One ; 9(4): e94080, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24710558

RESUMO

Transient receptor potential melastatin 8 (TRPM8) ion channels mediate the detection of noxious and innocuous cold and are expressed by primary sensory neurons, but little is known about the processing of the TRPM8-mediated cold information within the trigeminal sensory nuclei (TSN) and the spinal dorsal horn (DH). To address this issue, we characterized TRPM8-positive (+) neurons in the trigeminal ganglion and investigated the distribution of TRPM8+ axons and terminals, and their synaptic organization in the TSN and in the DH using light and electron microscopic immunohistochemistry in transgenic mice expressing a genetically encoded axonal tracer in TRPM8+ neurons. TRPM8 was expressed in a fraction of small myelinated primary afferent fibers (23.7%) and unmyelinated fibers (76.3%), suggesting that TRPM8-mediated cold is conveyed via C and Aδ afferents. TRPM8+ axons were observed in all TSN, but at different densities in the dorsal and ventral areas of the rostral TSN, which dominantly receive sensory afferents from intra- and peri-oral structures and from the face, respectively. While synaptic boutons arising from Aδ and non-peptidergic C afferents usually receive many axoaxonic contacts and form complex synaptic arrangements, TRPM8+ boutons arising from afferents of the same classes of fibers showed a unique synaptic connectivity; simple synapses with one or two dendrites and sparse axoaxonic contacts. These findings suggest that TRPM8-mediated cold is conveyed via a specific subset of C and Aδ afferent neurons and is processed in a unique manner and differently in the TSN and DH.


Assuntos
Axônios/metabolismo , Neurônios Aferentes/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Canais de Cátion TRPM/metabolismo , Núcleos do Trigêmeo/metabolismo , Vias Aferentes/metabolismo , Animais , Camundongos , Camundongos Transgênicos , Fibras Nervosas Amielínicas/metabolismo , Terminações Pré-Sinápticas/metabolismo
15.
J Neurosci ; 33(30): 12543-52, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-23884957

RESUMO

Chronic pain associated with injury or disease can result from dysfunction of sensory afferents whereby the threshold for activation of pain-sensing neurons (nociceptors) is lowered. Neurotrophic factors control nociceptor development and survival, but also induce sensitization through activation of their cognate receptors, attributable, in part, to the modulation of ion channel function. Thermal pain is mediated by channels of the transient receptor potential (TRP) family, including the cold and menthol receptor TRPM8. Although it has been shown that TRPM8 is involved in cold hypersensitivity, the molecular mechanisms underlying this pain modality are unknown. Using microarray analyses to identify mouse genes enriched in TRPM8 neurons, we found that the glial cell line-derived neurotrophic factor (GDNF) family receptor GFRα3 is expressed in a subpopulation of TRPM8 sensory neurons that have the neurochemical profile of cold nociceptors. Moreover, we found that artemin, the specific GFRα3 ligand that evokes heat hyperalgesia, robustly sensitized cold responses in a TRPM8-dependent manner in mice. In contrast, GFRα1 and GFRα2 are not coexpressed with TRPM8 and their respective ligands GDNF and neurturin did not induce cold pain, whereas they did evoke heat hyperalgesia. Nerve growth factor induced mild cold sensitization, consistent with TrkA expression in TRPM8 neurons. However, bradykinin failed to alter cold sensitivity even though its receptor expresses in a subset of TRPM8 neurons. These results show for the first time that only select neurotrophic factors induce cold sensitization through TRPM8 in vivo, unlike the broad range of proalgesic agents capable of promoting heat hyperalgesia.


Assuntos
Dor Crônica/fisiopatologia , Temperatura Baixa/efeitos adversos , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Canais de Cátion TRPM/genética , Animais , Dor Crônica/genética , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/fisiologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Temperatura Alta , Hiperalgesia/genética , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Nociceptividade/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPM/metabolismo , Ativação Transcricional/fisiologia , Gânglio Trigeminal/citologia , Gânglio Trigeminal/fisiologia
16.
Am J Physiol Endocrinol Metab ; 305(1): E78-88, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23651844

RESUMO

Blood glucose concentration is tightly regulated by the rate of insulin secretion and clearance, a process partially controlled by sensory neurons serving as metabolic sensors in relevant tissues. The activity of these neurons is regulated by the products of metabolism which regulate transmitter release, and recent evidence suggests that neuronally expressed ion channels of the transient receptor potential (TRP) family function in this critical process. Here, we report the novel finding that the cold and menthol-gated channel TRPM8 is necessary for proper insulin homeostasis. Mice lacking TRPM8 respond normally to a glucose challenge while exhibiting prolonged hypoglycemia in response to insulin. Additionally, Trpm8-/- mice have increased rates of insulin clearance compared with wild-type animals and increased expression of insulin-degrading enzyme in the liver. TRPM8 channels are not expressed in the liver, but TRPM8-expressing sensory afferents innervate the hepatic portal vein, suggesting a TRPM8-mediated neuronal control of liver insulin clearance. These results demonstrate that TRPM8 is a novel regulator of serum insulin and support the role of sensory innervation in metabolic homeostasis.


Assuntos
Glicemia/metabolismo , Hipoglicemia/genética , Insulina/metabolismo , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPM/genética , Animais , Plasmídeos de Bacteriocinas , Diabetes Mellitus Experimental/metabolismo , Homeostase/fisiologia , Hipoglicemia/metabolismo , Células Secretoras de Insulina/metabolismo , Fígado/irrigação sanguínea , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Veia Porta/inervação , Ratos , Canais de Cátion TRPM/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(18): 7476-81, 2013 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-23596210

RESUMO

Transient Receptor Potential Melastatin-8 (TRPM8), a recently identified member of the transient receptor potential (TRP) family of ion channels, is activated by mild cooling and by chemical compounds such as the supercooling agent, icilin. Since cooling, possibly involving TRPM8 stimulation, diminishes injury-induced peripheral inflammation, we hypothesized that TRPM8 activation may also attenuate systemic inflammation. We thus studied the involvement of TRPM8 in regulating colonic inflammation using two mouse models of chemically induced colitis. TRPM8 expression, localized immunohistochemically in transgenic TRPM8(GFP) mouse colon, was up-regulated in both human- and murine-inflamed colon samples, as measured by real-time PCR. Wild-type mice (but not TRPM8-nulls) treated systemically with the TRPM8 agonist, icilin showed an attenuation of chemically induced colitis, as reflected by a decrease in macroscopic and microscopic damage scores, bowel thickness, and myeloperoxidase activity compared with untreated animals. Furthermore, icilin treatment reduced the 2,4,6-trinitrobenzenesulfonic acid-induced increase in levels of inflammatory cytokines and chemokines in the colon. In comparison with wild-type mice, Dextran Sodium Sulfate (DSS)-treated TRPM8 knockout mice showed elevated colonic levels of the inflammatory neuropeptide calcitonin-gene-related peptide, although inflammatory indices were equivalent for both groups. Further, TRPM8 activation by icilin blocked capsaicin-triggered calcitonin-gene-related peptide release from colon tissue ex vivo and blocked capsaicin-triggered calcium signaling in Transient Receptor Potential Vaniloid-1 (TRPV1) and TRPM8 transfected HEK cells. Our data document an anti-inflammatory role for TRPM8 activation, in part due to an inhibiton of neuropeptide release, pointing to a novel therapeutic target for colitis and other inflammatory diseases.


Assuntos
Colite/patologia , Colite/fisiopatologia , Inflamação/patologia , Inflamação/fisiopatologia , Ativação do Canal Iônico , Canais de Cátion TRPM/metabolismo , Animais , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Sinalização do Cálcio , Quimiocinas/metabolismo , Colite/complicações , Colite/tratamento farmacológico , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Modelos Animais de Doenças , Humanos , Inflamação/complicações , Mediadores da Inflamação/metabolismo , Camundongos , Camundongos Knockout , Pirimidinonas/farmacologia , Pirimidinonas/uso terapêutico , Canais de Cátion TRPM/deficiência , Canais de Cátion TRPM/genética , Canais de Cátion TRPV/metabolismo , Ácido Trinitrobenzenossulfônico
18.
J Neurosci ; 33(7): 2837-48, 2013 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-23407943

RESUMO

Many primary sensory neurons are polymodal, responding to multiple stimulus modalities (chemical, thermal, or mechanical), yet each modality is recognized differently. Although polymodality implies that stimulus encoding occurs in higher centers, such as the spinal cord or brain, recent sensory neuron ablation studies find that behavioral responses to different modalities require distinct subpopulations, suggesting the existence of modality-specific labeled lines at the level of the sensory afferent. Here we provide evidence that neurons expressing TRPM8, a cold- and menthol-gated channel required for normal cold responses in mammals, represents a labeled line solely for cold sensation. We examined the behavioral significance of conditionally ablating TRPM8-expressing neurons in adult mice, finding that, like animals lacking TRPM8 channels (Trpm8(-/-)), animals depleted of TRPM8 neurons ("ablated") are insensitive to cool to painfully cold temperatures. Ablated animals showed little aversion to noxious cold and did not distinguish between cold and a preferred warm temperature, a phenotype more profound than that of Trpm8(-/-) mice which exhibit only partial cold-avoidance and -preference behaviors. In addition to acute responses, cold pain associated with inflammation and nerve injury was significantly attenuated in ablated and Trpm8(-/-) mice. Moreover, cooling-induced analgesia after nerve injury was abolished in both genotypes. Last, heat, mechanical, and proprioceptive behaviors were normal in ablated mice, demonstrating that TRPM8 neurons are dispensable for other somatosensory modalities. Together, these data show that, although some limited cold sensitivity remains in Trpm8(-/-) mice, TRPM8 neurons are required for the breadth of behavioral responses evoked by cold temperatures.


Assuntos
Analgesia , Temperatura Baixa , Dor/fisiopatologia , Células Receptoras Sensoriais/metabolismo , Canais de Cátion TRPM/biossíntese , Animais , Comportamento Animal/fisiologia , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Toxina Diftérica/farmacologia , Força da Mão/fisiologia , Temperatura Alta , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Análise em Microsséries , Fibras Nervosas/fisiologia , Dor/induzido quimicamente , Dor/psicologia , Insensibilidade Congênita à Dor/genética , Estimulação Física , Propriocepção/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Células Receptoras Sensoriais/fisiologia , Canais de Cátion TRPM/genética , Sensação Térmica/genética , Sensação Térmica/fisiologia
19.
ACS Chem Neurosci ; 4(2): 238-47, 2013 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-23421674

RESUMO

Of somatosensory modalities, cold is one of the more ambiguous percepts, evoking the pleasant sensation of cooling, the stinging bite of cold pain, and welcome relief from chronic pain. Moreover, unlike the precipitous thermal thresholds for heat activation of thermosensitive afferent neurons, thresholds for cold fibers are across a range of cool to cold temperatures that spans over 30 °C. Until recently, how cold produces this myriad of biological effects has been poorly studied, yet new advances in our understanding of cold mechanisms may portend a better understanding of sensory perception as well as provide novel therapeutic approaches. Chief among these was the identification of a number of ion channels that either serve as the initial detectors of cold as a stimulus in the peripheral nervous system, or are part of rather sophisticated differential expression patterns of channels that conduct electrical signals, thereby endowing select neurons with properties that are amenable to electrical signaling in the cold. This review highlights the current understanding of the channels involved in cold transduction as well as presents a hypothetical model to account for the broad range of cold thermal thresholds and distinct functions of cold fibers in perception, pain, and analgesia.


Assuntos
Temperatura Baixa , Nociceptores/fisiologia , Canais de Cátion TRPM/fisiologia , Termorreceptores/fisiologia , Sensação Térmica/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Canais de Cálcio/fisiologia , Humanos , Proteínas do Tecido Nervoso/fisiologia , Neurônios Aferentes/fisiologia , Transdução de Sinais/fisiologia , Canal de Cátion TRPA1
20.
Hum Mol Genet ; 21(20): 4431-47, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22802075

RESUMO

A number of mouse models for spinal muscular atrophy (SMA) have been genetically engineered to recapitulate the severity of human SMA by using a targeted null mutation at the mouse Smn1 locus coupled with the transgenic addition of varying copy numbers of human SMN2 genes. Although this approach has been useful in modeling severe SMA and very mild SMA, a mouse model of the intermediate form of the disease would provide an additional research tool amenable for drug discovery. In addition, many of the previously engineered SMA strains are multi-allelic by design, containing a combination of transgenes and targeted mutations in the homozygous state, making further genetic manipulation difficult. A new genetic engineering approach was developed whereby variable numbers of SMN2 sequences were incorporated directly into the murine Smn1 locus. Using combinations of these alleles, we generated an allelic series of SMA mouse strains harboring no, one, two, three, four, five, six or eight copies of SMN2. We report here the characterization of SMA mutants in this series that displayed a range in disease severity from embryonic lethal to viable with mild neuromuscular deficits.


Assuntos
Atrofia Muscular Espinal/genética , Junção Neuromuscular/genética , Alelos , Animais , Comportamento Animal , Modelos Animais de Doenças , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos , Junção Neuromuscular/metabolismo , Fenótipo , Proteína 1 de Sobrevivência do Neurônio Motor/genética , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Proteína 2 de Sobrevivência do Neurônio Motor/genética , Proteína 2 de Sobrevivência do Neurônio Motor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...