Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Mol Psychiatry ; 26(4): 1376-1398, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-31444474

RESUMO

Aberrant fear is a cornerstone of several psychiatric disorders. Consequently, there is large interest in elucidation of signaling mechanisms that link extracellular cues to changes in neuronal function and structure in brain pathways that are important in the generation and maintenance of fear memory and its behavioral expression. Members of the Plexin-B family of receptors for class 4 semaphorins play important roles in developmental plasticity of neurons, and their expression persists in some areas of the adult nervous system. Here, we aimed to elucidate the role of Semaphorin 4C (Sema4C) and its cognate receptor, Plexin-B2, in the expression of contextual and cued fear memory, setting a mechanistic focus on structural plasticity and exploration of contributing signaling pathways. We observed that Plexin-B2 and Sema4C are expressed in forebrain areas related to fear memory, such as the anterior cingulate cortex, amygdala and the hippocampus, and their expression is regulated by aversive stimuli that induce fear memory. By generating forebrain-specific Plexin-B2 knockout mice and analyzing fear-related behaviors, we demonstrate that Sema4C-PlexinB2 signaling plays a crucial functional role in the recent and remote recall of fear memory. Detailed neuronal morphological analyses revealed that Sema4C-PlexinB2 signaling largely mediates fear-induced structural plasticity by enhancing dendritic ramifications and modulating synaptic density in the adult hippocampus. Analyses on signaling-related mutant mice showed that these functions are mediated by PlexinB2-dependent RhoA activation. These results deliver important insights into the mechanistic understanding of maladaptive plasticity in fear circuits and have implications for novel therapeutic strategies against fear-related disorders.


Assuntos
Medo , Memória , Proteínas do Tecido Nervoso , Semaforinas , Animais , Moléculas de Adesão Celular , Camundongos , Proteínas do Tecido Nervoso/genética , Neurônios , Semaforinas/genética
2.
J Neurosci ; 40(35): 6664-6677, 2020 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-32616667

RESUMO

Wnt signaling represents a highly versatile signaling system, which plays critical roles in developmental morphogenesis as well as synaptic physiology in adult life and is implicated in a variety of neural disorders. Recently, we demonstrated that Wnt3a is able to recruit multiple noncanonical signaling pathways to alter peripheral sensory neuron function in a nociceptive modality-specific manner. Furthermore, several studies recently reported an important role for Wnt5a acting via canonical and noncanonical signaling in spinal processing of nociception in a number of pathologic pain disorders. Here, using diverse molecular, genetic, and behavioral approaches in mouse models of pain in vivo, we report a novel role for Wnt5a signaling in nociceptive modulation at the structural level. In models of chronic pain, using male and female mice, we found that Wnt5a is released spinally from peripheral sensory neurons, where it recruits the tyrosine kinase receptors Ror2 and Ryk to modulate dendritic spine rearrangement. Blocking the Wnt5a-Ryk/Ror2 axis in spinal dorsal horn neurons prevented activity-dependent dendritic spine remodeling and significantly reduced mechanical hypersensitivity induced by peripheral injury as well as inflammation. Moreover, we observed that peripheral Wnt3a signaling triggers the release of Wnt5a in the spinal cord, and inhibition of spinal Wnt5a signaling attenuates the functional impact of peripheral Wnt3a on nociceptive sensitivity. In conclusion, this study reports a novel role for the Wnt signaling axis in coordinating peripheral and spinal sensitization and shows that targeting Wnt5a-Ryk/ROR2 signaling alleviates both structural and functional mechanisms of nociceptive hypersensitivity in models of chronic pain in vivoSIGNIFICANCE STATEMENT There is a major need to elucidate molecular mechanisms underlying chronic pain disorders to develop novel therapeutic approaches. Wnt signaling represents a highly versatile signaling system, which plays critical roles during development and adult physiology, and it was implicated in several diseases, including chronic pain conditions. Using mouse models, our study identifies a novel role for Wnt5a signaling in nociceptive modulation at the spinal cord level. We observed that Wnt5a recruits Ror2 and Ryk receptors to enhance dendritic spine density, leading to nociceptive sensitization. Blocking the Wnt5a-Ryk/Ror2 interaction in the spinal dorsal horn prevented spine remodeling and significantly reduced inflammatory and neuropathic hypersensitivity. These findings provide proof-of-concept for targeting spinal Wnt signaling for alleviating nociceptive hypersensitivity in vivo.


Assuntos
Dor do Câncer/metabolismo , Espinhas Dendríticas/metabolismo , Neuralgia/metabolismo , Nociceptividade , Proteína Wnt-5a/metabolismo , Proteína Wnt3A/metabolismo , Animais , Dor do Câncer/fisiopatologia , Linhagem Celular Tumoral , Células Cultivadas , Espinhas Dendríticas/patologia , Espinhas Dendríticas/fisiologia , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Neuralgia/fisiopatologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores Órfãos Semelhantes a Receptor Tirosina Quinase/metabolismo , Transdução de Sinais , Proteína Wnt-5a/genética , Proteína Wnt3A/genética
3.
Gastroenterology ; 159(2): 665-681.e13, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32330476

RESUMO

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) is frequently accompanied by excruciating pain, which has been associated with attraction of cancer cells and their invasion of intrapancreatic sensory nerves. Neutralization of the chemokine CCL2 reduced cancer-associated pain in a clinical trial, but there have been no systematic analyses of the highly diverse chemokine families and their receptors in PDAC. METHODS: We performed an open, unbiased RNA-interference screen of mammalian chemokines in co-cultures of mouse PDAC cells (K8484) and mouse peripheral sensory neurons, and confirmed findings in studies of DT8082 PDAC cells. We studied the effects of chemokines on migration of PDAC cell lines. Orthotopic tumors were grown from K8484 cells in mice, and mice were given injections of neutralizing antibodies against chemokines, antagonists, or control antibodies. We analyzed abdominal mechanical hypersensitivity and collected tumors and analyzed them by histology and immunohistochemistry to assess neural remodeling. We collected PDAC samples and information on pain levels from 74 patients undergoing resection and measured levels of CXCR3 and CCR7 by immunohistochemistry and immunoblotting. RESULTS: Knockdown of 9 chemokines in DRG neurons significantly reduced migration of PDAC cells towards sensory neurons. Sensory neuron-derived CCL21 and CXCL10 promoted migration of PDAC cells via their receptors CCR7 and CXCR3, respectively, which were expressed by cells in orthotopic tumors and PDAC specimens from patients. Neutralization of CCL21 or CXCL10, or their receptors, in mice with orthotopic tumors significantly reduced nociceptive hypersensitivity and nerve fiber hypertrophy and improved behavioral parameters without affecting tumor infiltration by T cells or neutrophils. Increased levels of CXCR3 and CCR7 in human PDAC specimens were associated with increased frequency of cancer-associated pain, determined from patient questionnaires. CONCLUSIONS: In an unbiased screen of chemokines, we identified CCL21 and CXCL10 as proteins that promote migration of pancreatic cancer cells toward sensory neurons. Inhibition of these chemokines or their receptors reduce hypersensitivity in mice with orthotopic tumors, and patients with PDACs with high levels of the chemokine receptors of CXCR3 and CCR7 had increased frequency of cancer-associated pain.


Assuntos
Dor do Câncer/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Movimento Celular , Quimiocina CCL21/metabolismo , Quimiocina CXCL10/metabolismo , Gânglios Espinais/metabolismo , Neoplasias Pancreáticas/metabolismo , Células Receptoras Sensoriais/metabolismo , Analgésicos/farmacologia , Animais , Anticorpos Neutralizantes/farmacologia , Dor do Câncer/genética , Dor do Câncer/patologia , Dor do Câncer/prevenção & controle , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Quimiocina CCL21/antagonistas & inibidores , Quimiocina CCL21/genética , Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL10/genética , Técnicas de Cocultura , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/patologia , Humanos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Receptores CCR7/metabolismo , Receptores CXCR3/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/patologia , Transdução de Sinais
4.
Int J Mol Sci ; 22(16)2021 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-34445160

RESUMO

Throughout life, macrophages are located in every tissue of the body, where their main roles are to phagocytose cellular debris and recycle aging red blood cells. In the tissue niche, they promote homeostasis through trophic, regulatory, and repair functions by responding to internal and external stimuli. This in turn polarizes macrophages into a broad spectrum of functional activation states, also reflected in their iron-regulated gene profile. The fast adaptation to the environment in which they are located helps to maintain tissue homeostasis under physiological conditions.


Assuntos
Ferro/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Animais , Homeostase , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Ferro/metabolismo , Macrófagos/metabolismo , Fagocitose
5.
Cell Tissue Res ; 357(2): 407-26, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25012522

RESUMO

Acute physiological pain, the unpleasant sensory response to a noxious stimulus, is essential for animals and humans to avoid potential injury. Pathological pain that persists after the original insult or injury has subsided, however, not only results in individual suffering but also imposes a significant cost on society. Improving treatments for long-lasting pathological pain requires a comprehensive understanding of the biological mechanisms underlying pain perception and the development of pain chronicity. In this review, we aim to highlight some of the major findings related to the involvement of neuronal calcium signaling in the processes that mediate chronic pain.


Assuntos
Sinalização do Cálcio , Dor Crônica/metabolismo , Neurônios/patologia , Animais , Doença Crônica , Dor Crônica/fisiopatologia , Neuropatias Diabéticas/metabolismo , Neuropatias Diabéticas/fisiopatologia , Humanos , Neuralgia/metabolismo , Neuralgia/fisiopatologia , Plasticidade Neuronal , Neurônios/metabolismo
6.
Nat Metab ; 6(3): 494-513, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38443593

RESUMO

Long-lasting pain stimuli can trigger maladaptive changes in the spinal cord, reminiscent of plasticity associated with memory formation. Metabolic coupling between astrocytes and neurons has been implicated in neuronal plasticity and memory formation in the central nervous system, but neither its involvement in pathological pain nor in spinal plasticity has been tested. Here we report a form of neuroglia signalling involving spinal astrocytic glycogen dynamics triggered by persistent noxious stimulation via upregulation of the Protein Targeting to Glycogen (PTG) in spinal astrocytes. PTG drove glycogen build-up in astrocytes, and blunting glycogen accumulation and turnover by Ptg gene deletion reduced pain-related behaviours and promoted faster recovery by shortening pain maintenance in mice. Furthermore, mechanistic analyses revealed that glycogen dynamics is a critically required process for maintenance of pain by facilitating neuronal plasticity in spinal lamina 1 neurons. In summary, our study describes a previously unappreciated mechanism of astrocyte-neuron metabolic communication through glycogen breakdown in the spinal cord that fuels spinal neuron hyperexcitability.


Assuntos
Astrócitos , Dor , Camundongos , Animais , Astrócitos/metabolismo , Dor/metabolismo , Dor/patologia , Neurônios/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Glicogênio/metabolismo
7.
Mol Pain ; 9: 62, 2013 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-24294842

RESUMO

BACKGROUND: ATP-gated P2X3 receptors of sensory ganglion neurons are important transducers of pain as they adapt their expression and function in response to acute and chronic nociceptive signals. The present study investigated the role of calcium/calmodulin-dependent serine protein kinase (CASK) in controlling P2X3 receptor expression and function in trigeminal ganglia from Cacna1a R192Q-mutated knock-in (KI) mice, a genetic model for familial hemiplegic migraine type-1. RESULTS: KI ganglion neurons showed more abundant CASK/P2X3 receptor complex at membrane level, a result that likely originated from gain-of-function effects of R192Q-mutated CaV2.1 channels and downstream enhanced CaMKII activity. The selective CaV2.1 channel blocker ω-Agatoxin IVA and the CaMKII inhibitor KN-93 were sufficient to return CASK/P2X3 co-expression to WT levels. After CASK silencing, P2X3 receptor expression was decreased in both WT and KI ganglia, supporting the role of CASK in P2X3 receptor stabilization. This process was functionally observed as reduced P2X3 receptor currents. CONCLUSIONS: We propose that, in trigeminal sensory neurons, the CASK/P2X3 complex has a dynamic nature depending on intracellular calcium and related signaling, that are enhanced in a transgenic mouse model of genetic hemiplegic migraine.


Assuntos
Canais de Cálcio Tipo N/metabolismo , Guanilato Quinases/metabolismo , Receptores Purinérgicos P2X3/metabolismo , Células Receptoras Sensoriais/metabolismo , Transdução de Sinais/genética , Gânglio Trigeminal/citologia , Animais , Canais de Cálcio Tipo N/genética , Guanilato Quinases/genética , Camundongos , Camundongos Transgênicos , Mutação , Receptores Purinérgicos P2X3/genética
8.
Cells ; 12(8)2023 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-37190035

RESUMO

Chronic pain affects a significant amount of the population and is responsible for vast worldwide socio-economic costs [...].


Assuntos
Dor Crônica , Nociceptores , Humanos
9.
Cells ; 12(2)2023 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-36672219

RESUMO

Drugs enhancing the availability of noradrenaline are gaining prominence in the therapy of chronic neuropathic pain. However, underlying mechanisms are not well understood, and research has thus far focused on α2-adrenergic receptors and neuronal excitability. Adrenergic receptors are also expressed on glial cells, but their roles toward antinociception are not well deciphered. This study addresses the contribution of ß2-adrenergic receptors (ß2-ARs) to the therapeutic modulation of neuropathic pain in mice. We report that selective activation of ß2-ARs with Formoterol inhibits pro-inflammatory signaling in microglia ex vivo and nerve injury-induced structural remodeling and functional activation of microglia in vivo. Systemic delivery of Formoterol inhibits behaviors related to neuropathic pain, such as mechanical hypersensitivity, cold allodynia as well as the aversive component of pain, and reverses chronically established neuropathic pain. Using conditional gene targeting for microglia-specific deletion of ß2-ARs, we demonstrate that the anti-allodynic effects of Formoterol are primarily mediated by microglia. Although Formoterol also reduces astrogliosis at late stages of neuropathic pain, these functions are unrelated to ß2-AR signaling in microglia. Our results underline the value of developing microglial ß2-AR agonists for relief from neuropathic pain and clarify mechanistic underpinnings.


Assuntos
Microglia , Neuralgia , Camundongos , Animais , Neuralgia/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Receptores Adrenérgicos , Fumarato de Formoterol/farmacologia , Fumarato de Formoterol/uso terapêutico
10.
Cells ; 11(19)2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36231105

RESUMO

Chronic pain is a debilitating condition that influences the social, economic, and psychological aspects of patients' lives. Hence, the need for better treatment is drawing extensive interest from the research community. Developmental molecules such as Wnt, ephrins, and semaphorins are acknowledged as central players in the proper growth of a biological system. Their receptors and ligands are expressed in a wide variety in both neurons and glial cells, which are implicated in pain development, maintenance, and resolution. Thereby, it is not surprising that the impairment of those pathways affects the activities and functions of the entire cell. Evidence indicates aberrant activation of their pathways in the nervous system in rodent models of chronic pain. In those conditions, Wnt, ephrin, and semaphorin signaling participate in enhancing neuronal excitability, peripheral sensitization, synaptic plasticity, and the production and release of inflammatory cytokines. This review summarizes the current knowledge on three main developmental pathways and their mechanisms linked with the pathogenesis and progression of pain, considering their impacts on neuronal and glial cells in experimental animal models. Elucidations of the downstream pathways may provide a new mechanism for the involvement of Wnt, ephrin, and semaphorin pathways in pain chronicity.


Assuntos
Dor Crônica , Semaforinas , Animais , Orientação de Axônios , Citocinas , Efrinas/metabolismo , Semaforinas/metabolismo
11.
Neuron ; 109(9): 1413-1416, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33957067

RESUMO

How genetic loss of the sodium channel NaV1.7 results in painlessness is puzzling. MacDonald et al. (2021) demonstrate that instead of impairing peripheral excitability, NaV1.7 channels at central terminals of pain-sensing afferents play a pivotal role in the balance between pain and analgesia.


Assuntos
Analgesia , Gânglios Espinais , Animais , Humanos , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.8 , Dor/genética , Células Receptoras Sensoriais
12.
Mol Pain ; 6: 48, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20735819

RESUMO

BACKGROUND: The R192Q mutation of the CACNA1A gene, encoding for the α1 subunit of voltage-gated P/Q Ca2+ channels (Ca(v)2.1), is associated with familial hemiplegic migraine-1. We investigated whether this gain-of-function mutation changed the structure and function of trigeminal neuron P2X3 receptors that are thought to be important contributors to migraine pain. RESULTS: Using in vitro trigeminal sensory neurons of a mouse genetic model knockin for the CACNA1A R192Q mutation, we performed patch clamp recording and intracellular Ca2+ imaging that showed how these knockin ganglion neurons generated P2X3 receptor-mediated responses significantly larger than wt neurons. These enhanced effects were reversed by the Ca(v)2.1 blocker ω-agatoxin. We, thus, explored intracellular signalling dependent on kinases and phosphatases to understand the molecular regulation of P2X3 receptors of knockin neurons. In such cells we observed strong activation of CaMKII reversed by ω-agatoxin treatment. The CaMKII inhibitor KN-93 blocked CaMKII phosphorylation and the hyperesponsive P2X3 phenotype. Although no significant difference in membrane expression of knockin receptors was found, serine phosphorylation of knockin P2X3 receptors was constitutively decreased and restored by KN-93. No change in threonine or tyrosine phosphorylation was detected. Finally, pharmacological inhibitors of the phosphatase calcineurin normalized the enhanced P2X3 receptor responses of knockin neurons and increased their serine phosphorylation. CONCLUSIONS: The present results suggest that the CACNA1A mutation conferred a novel molecular phenotype to P2X3 receptors of trigeminal ganglion neurons via CaMKII-dependent activation of calcineurin that selectively impaired the serine phosphorylation state of such receptors, thus potentiating their effects in transducing trigeminal nociception.


Assuntos
Canais de Cálcio Tipo P/genética , Canais de Cálcio Tipo Q/genética , Gânglios Sensitivos/metabolismo , Ativação do Canal Iônico/fisiologia , Enxaqueca com Aura/genética , Mutação/genética , Dor/fisiopatologia , Receptores Purinérgicos P2X3/metabolismo , Trifosfato de Adenosina/metabolismo , Agatoxinas , Substituição de Aminoácidos/genética , Animais , Calcineurina/metabolismo , Canais de Cálcio Tipo N , Canais de Cálcio Tipo P/metabolismo , Canais de Cálcio Tipo Q/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Ativação Enzimática/efeitos dos fármacos , Gânglios Sensitivos/efeitos dos fármacos , Técnicas de Introdução de Genes , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Enxaqueca com Aura/complicações , Enxaqueca com Aura/fisiopatologia , Proteínas Mutantes/metabolismo , Dor/complicações , Fosforilação/efeitos dos fármacos , Fosfosserina/metabolismo , Potássio/metabolismo , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/metabolismo , Venenos de Aranha/farmacologia , Nervo Trigêmeo/efeitos dos fármacos , Nervo Trigêmeo/enzimologia , Nervo Trigêmeo/fisiopatologia
13.
Cell Mol Neurobiol ; 30(4): 505-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19960242

RESUMO

Cdk5 is an endogenous kinase activated by the neuronal-specific protein p35 and implicated in multiple neuronal functions, including modulation of certain pain responses. We investigated whether Cdk5 could regulate ATP-gated P2X(3) receptors that are members of the family of membrane proteins expressed by sensory neurons to transduce nociception in baseline and chronic pain. To study the potential P2X(3) receptor modulation by Cdk5, we co-transfected rat P2X(3) receptors and Cdk5 into HEK cells and observed increased P2X(3) receptor serine phosphorylation together with downregulation of receptor currents only when these genes were transfected together with the gene of the Cdk5 activator p35. The changes in receptor responses were limited to depressed current amplitude as desensitization and recovery were not altered. Transfection of p35 with P2X(3) similarly downregulated receptor responses, suggesting that this phenomenon could be observed even with constitutive Cdk5. The present data indicate a novel target to express the action of Cdk5 on membrane proteins involved in pain perception.


Assuntos
Quinase 5 Dependente de Ciclina/metabolismo , Dor/fisiopatologia , Receptores Purinérgicos P2/metabolismo , Serina/metabolismo , Animais , Linhagem Celular , Quinase 5 Dependente de Ciclina/genética , Regulação para Baixo , Humanos , Técnicas de Patch-Clamp , Fosforilação , Ratos , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2X3
14.
J Neurosci ; 27(31): 8190-201, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17670966

RESUMO

The molecular mechanisms of migraine pain are incompletely understood, although migraine mediators such as NGF and calcitonin gene-related peptide (CGRP) are believed to play an algogenic role. Although NGF block is proposed as a novel analgesic approach, its consequences on nociceptive purinergic P2X receptors of trigeminal ganglion neurons remain unknown. We investigated whether neutralizing NGF might change the function of P2X3 receptors natively coexpressed with NGF receptors on cultured mouse trigeminal neurons. Treatment with an NGF antibody (24 h) decreased P2X3 receptor-mediated currents and Ca2+ transients, an effect opposite to exogenously applied NGF. Recovery from receptor desensitization was delayed by anti-NGF treatment without changing desensitization onset. NGF neutralization was associated with decreased threonine phosphorylation of P2X3 subunits, presumably accounting for their reduced responses and slower recovery. Anti-NGF treatment could also increase the residual current typical of heteromeric P2X2/3 receptors, consistent with enhanced membrane location of P2X2 subunits. This possibility was confirmed with cross-linking and immunoprecipitation studies. NGF neutralization also led to increased P2X2e splicing variant at mRNA and membrane protein levels. These data suggest that NGF controlled plasticity of P2X3 subunits and their membrane assembly with P2X2 subunits. Despite anti-NGF treatment, CGRP could still enhance P2X3 receptor activity, indicating separate NGF- or CGRP-mediated mechanisms to upregulate P2X3 receptors. In an in vivo model of mouse trigeminal pain, anti-NGF pretreatment suppressed responses evoked by P2X3 receptor activation. Our findings outline the important contribution by NGF signaling to nociception of trigeminal sensory neurons, which could be counteracted by anti-NGF pretreatment.


Assuntos
Trifosfato de Adenosina/fisiologia , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/imunologia , Neurônios/metabolismo , Medição da Dor/métodos , Receptores Purinérgicos P2/fisiologia , Gânglio Trigeminal/metabolismo , Animais , Anticorpos/farmacologia , Cálcio/antagonistas & inibidores , Cálcio/metabolismo , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Crescimento Neural/fisiologia , Plasticidade Neuronal/fisiologia , Antagonistas do Receptor Purinérgico P2 , Receptores Purinérgicos P2X3 , Gânglio Trigeminal/efeitos dos fármacos
15.
J Neurosci ; 26(23): 6163-71, 2006 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-16763024

RESUMO

Recent evidence indicates a key role for the neuropeptide calcitonin gene-related peptide (CGRP) in migraine pain, as demonstrated by the strong analgesic action of CGRP receptor antagonists, although the mechanisms of this effect remain unclear. Most trigeminal nociceptive neurons releasing CGRP also express ATP-activated purinergic P2X3 receptors to transduce pain. To understand whether the CGRP action involves P2X3 receptor modulation, the model of trigeminal nociceptive neurons in culture was used to examine the long-term action of this peptide. Although 79% of CGRP-binding neurons expressed P2X3 receptors, acute application of CGRP did not change P2X3 receptor function. Nevertheless, after 1 h of CGRP treatment, strong enhancement of the amplitude of P2X3 receptor currents was observed together with accelerated recovery from desensitization. Receptor upregulation persisted up to 10 h (despite CGRP washout), was accompanied by increased P2X3 gene transcription, and was fully prevented by the CGRP antagonist CGRP(8-37). Surface biotinylation showed CGRP augmented P2X3 receptor expression, consistent with confocal microscopy data indicating enhanced P2X3 immunoreactivity beneath the neuronal membrane. These results suggest that CGRP stimulated trafficking of P2X3 receptors to the cell-surface membrane. Using pharmacological tools, we demonstrated that this effect of CGRP was dependent on protein kinase A and PKC activation and was prevented by the trafficking inhibitor brefeldin A. Capsaicin-sensitive TRPV1 vanilloid receptors were not upregulated. The present data demonstrate a new form of selective, slow upregulation of nociceptive P2X3 receptors on trigeminal neurons by CGRP. This mechanism might contribute to pain sensitization and represents a model of neuronal plasticity in response to a migraine mediator.


Assuntos
Trifosfato de Adenosina/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/fisiologia , Nociceptores/metabolismo , Receptores Purinérgicos P2/metabolismo , Nervo Trigêmeo/metabolismo , Regulação para Cima , Animais , Peptídeo Relacionado com Gene de Calcitonina/farmacologia , Membrana Celular/metabolismo , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Sinergismo Farmacológico , Eletrofisiologia , Camundongos , Camundongos Endogâmicos , Plasticidade Neuronal , Nociceptores/efeitos dos fármacos , Proteína Quinase C/fisiologia , Transporte Proteico , Receptores Purinérgicos P2/efeitos dos fármacos , Receptores Purinérgicos P2/fisiologia , Receptores Purinérgicos P2X3 , Canais de Cátion TRPV/efeitos dos fármacos , Canais de Cátion TRPV/metabolismo , Fatores de Tempo , Nervo Trigêmeo/citologia , Nervo Trigêmeo/efeitos dos fármacos
16.
Nat Commun ; 8(1): 176, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28765520

RESUMO

Semaphorins and their transmembrane receptors, Plexins, are key regulators of axon guidance and development of neuronal connectivity. B-type Plexins respond to Class IV semaphorins and mediate a variety of developmental functions. Here we report that the expression of Plexin-B2 and its high-affinity ligand, Sema4C, persists in peripheral sensory neurons in adult life and is markedly increased in states of persistent pain in mice. Genetic deletion of Sema4C as well as adult-onset loss of Plexin-B2 leads to impairment of the development and duration of inflammatory hypersensitivity. Remarkably, unlike the neurodevelopmental functions of Plexin-B2 that solely rely on Ras signaling, we obtained genetic and pharmacological evidence for a requirement of RhoA-ROCK-dependent mechanisms as well as TRPA1 sensitization in pronociceptive functions of Sema4C-Plexin-B2 signaling in adult life. These results suggest important roles for Plexin-B2 signaling in sensory function that may be of therapeutic relevance in pathological pain.Semaphorins and their receptors are involved in neurodevelopment, but their functions in the adult nervous system are not fully understood. This study finds that semaphorin 4C and its receptor Plexin B are expressed in sensory neurons and are pronociceptive in a mouse model of inflammatory pain.


Assuntos
Inflamação , Proteínas do Tecido Nervoso/genética , Nociceptividade , Dor/genética , Semaforinas/genética , Células Receptoras Sensoriais/metabolismo , Animais , Modelos Animais de Doenças , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Dor/metabolismo , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Semaforinas/metabolismo , Transdução de Sinais , Canal de Cátion TRPA1/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP
17.
Mol Pain ; 2: 11, 2006 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-16566843

RESUMO

BACKGROUND: Cultured sensory neurons are a common experimental model to elucidate the molecular mechanisms of pain transduction typically involving activation of ATP-sensitive P2X or capsaicin-sensitive TRPV1 receptors. This applies also to trigeminal ganglion neurons that convey pain inputs from head tissues. Little is, however, known about the plasticity of these receptors on trigeminal neurons in culture, grown without adding the neurotrophin NGF which per se is a powerful algogen. The characteristics of such receptors after short-term culture were compared with those of ganglia. Furthermore, their modulation by chronically-applied serotonin or NGF was investigated. RESULTS: Rat or mouse neurons in culture mainly belonged to small and medium diameter neurons as observed in sections of trigeminal ganglia. Real time RT-PCR, Western blot analysis and immunocytochemistry showed upregulation of P2X(3) and TRPV1 receptors after 1-4 days in culture (together with their more frequent co-localization), while P2X(2) ones were unchanged. TRPV1 immunoreactivity was, however, lower in mouse ganglia and cultures. Intracellular Ca(2+) imaging and whole-cell patch clamping showed functional P2X and TRPV1 receptors. Neurons exhibited a range of responses to the P2X agonist alpha, beta-methylene-adenosine-5'-triphosphate indicating the presence of homomeric P2X(3) receptors (selectively antagonized by A-317491) and heteromeric P2X(2/3) receptors. The latter were observed in 16 % mouse neurons only. Despite upregulation of receptors in culture, neurons retained the potential for further enhancement of P2X(3) receptors by 24 h NGF treatment. At this time point TRPV1 receptors had lost the facilitation observed after acute NGF application. Conversely, chronically-applied serotonin selectively upregulated TRPV1 receptors rather than P2X(3) receptors. CONCLUSION: Comparing ganglia and cultures offered the advantage of understanding early adaptive changes of nociception-transducing receptors of trigeminal neurons. Culturing did not prevent differential receptor upregulation by algogenic substances like NGF or serotonin, indicating that chronic application led to distinct plastic changes in the molecular mechanisms mediating pain on trigeminal nociceptors.


Assuntos
Gânglios/metabolismo , Regulação da Expressão Gênica , Fator de Crescimento Neural/metabolismo , Neurônios/metabolismo , Receptores Purinérgicos P2/fisiologia , Serotonina/metabolismo , Canais de Cátion TRPV/metabolismo , Nervo Trigêmeo/metabolismo , Animais , Cálcio/metabolismo , Capsaicina/farmacologia , Camundongos , Ratos , Receptores Purinérgicos P2X , Fármacos do Sistema Sensorial/farmacologia
18.
Nat Med ; 21(5): 518-23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25915831

RESUMO

Neuropathic pain is a major, intractable clinical problem and its pathophysiology is not well understood. Although recent gene expression profiling studies have enabled the identification of novel targets for pain therapy, classical study designs provide unclear results owing to the differential expression of hundreds of genes across sham and nerve-injured groups, which can be difficult to validate, particularly with respect to the specificity of pain modulation. To circumvent this, we used two outbred lines of rats, which are genetically similar except for being genetically segregated as a result of selective breeding for differences in neuropathic pain hypersensitivity. SerpinA3N, a serine protease inhibitor, was upregulated in the dorsal root ganglia (DRG) after nerve injury, which was further validated for its mouse homolog. Mice lacking SerpinA3N developed more neuropathic mechanical allodynia than wild-type (WT) mice, and exogenous delivery of SerpinA3N attenuated mechanical allodynia in WT mice. T lymphocytes infiltrate the DRG after nerve injury and release leukocyte elastase (LE), which was inhibited by SerpinA3N derived from DRG neurons. Genetic loss of LE or exogenous application of a LE inhibitor (Sivelastat) in WT mice attenuated neuropathic mechanical allodynia. Overall, we reveal a novel and clinically relevant role for a member of the serpin superfamily and a leukocyte elastase and crosstalk between neurons and T cells in the modulation of neuropathic pain.


Assuntos
Proteínas de Fase Aguda/metabolismo , Inibidores Enzimáticos/farmacologia , Elastase de Leucócito/antagonistas & inibidores , Serpinas/metabolismo , Linfócitos T/citologia , Animais , Separação Celular , Dependovirus/genética , Feminino , Gânglios Espinais/metabolismo , Hiperalgesia/fisiopatologia , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Neuralgia , Neurônios/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Dor/fisiopatologia , Fenótipo , Reação em Cadeia da Polimerase , Ratos , Regulação para Cima
19.
Neuron ; 83(1): 104-21, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24991956

RESUMO

Wnt signaling represents a highly versatile signaling system, which plays diverse and critical roles in various aspects of neural development. Sensory neurons of the dorsal root ganglia require Wnt signaling for initial cell-fate determination as well as patterning and synapse formation. Here we report that Wnt signaling pathways persist in adult sensory neurons and play a functional role in their sensitization in a pathophysiological context. We observed that Wnt3a recruits the Wnt-calcium signaling pathway and the Wnt planar cell polarity pathway in peripheral nerves to alter pain sensitivity in a modality-specific manner and we elucidated underlying mechanisms. In contrast, biochemical, pharmacological, and genetic studies revealed lack of functional relevance for the classical canonical ß-catenin pathway in peripheral sensory neurons in acute modulation of nociception. Finally, this study provides proof-of-concept for a translational potential for Wnt3a-Frizzled3 signaling in alleviating disease-related pain hypersensitivity in cancer-associated pain in vivo.


Assuntos
Receptores Frizzled/fisiologia , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Células Receptoras Sensoriais/metabolismo , Via de Sinalização Wnt/fisiologia , Proteína Wnt3A/fisiologia , Animais , Células Cultivadas , Gânglios Espinais/patologia , Células HEK293 , Humanos , Hiperalgesia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Células Receptoras Sensoriais/patologia
20.
Neuron ; 77(1): 43-57, 2013 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-23312515

RESUMO

Persistent pain induced by noxious stimuli is characterized by the transition from normosensitivity to hypersensitivity. Underlying mechanisms are not well understood, although gene expression is considered important. Here, we show that persistent nociceptive-like activity triggers calcium transients in neuronal nuclei within the superficial spinal dorsal horn, and that nuclear calcium is necessary for the development of long-term inflammatory hypersensitivity. Using a nucleus-specific calcium signal perturbation strategy in vivo complemented by gene profiling, bioinformatics, and functional analyses, we discovered a pain-associated, nuclear calcium-regulated gene program in spinal excitatory neurons. This includes C1q, a modulator of synaptic spine morphogenesis, which we found to contribute to activity-dependent spine remodelling on spinal neurons in a manner functionally associated with inflammatory hypersensitivity. Thus, nuclear calcium integrates synapse-to-nucleus communication following noxious stimulation and controls a spinal genomic response that mediates the transition between acute and long-term nociceptive sensitization by modulating functional and structural plasticity.


Assuntos
Sinalização do Cálcio/fisiologia , Núcleo Celular/fisiologia , Dor Crônica/genética , Genômica , Células do Corno Posterior/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Dor Crônica/patologia , Genômica/métodos , Inflamação/genética , Inflamação/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Neurônios/fisiologia , Medição da Dor/métodos , Células do Corno Posterior/patologia , Medula Espinal/citologia , Medula Espinal/patologia , Medula Espinal/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA