Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 606(7912): 137-145, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614217

RESUMO

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


Assuntos
Hiperalgesia , Neuralgia , Nociceptores , Pele , Animais , Dor Crônica/fisiopatologia , Hiperalgesia/fisiopatologia , Mecanorreceptores/patologia , Camundongos , Neuralgia/fisiopatologia , Nociceptores/patologia , Pele/inervação , Pele/fisiopatologia
2.
Mol Pain ; 14: 1744806918817975, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30453826

RESUMO

Diabetic peripheral neuropathy is a major debilitating late complication of diabetes, which significantly reduces the quality of life in patients. Diabetic peripheral neuropathy is associated with a wide spectrum of sensory abnormalities, where in loss of sensation or hypoalgesia to applied external stimuli is paradoxically accompanied by debilitating tonic spontaneous pain. In numerous studies on animal models of diabetic peripheral neuropathy, behavioural measurements have been largely confined to analysis of evoked withdrawal to mechanical and thermal stimuli applied to dermatomes, whereas spontaneous, on-going pain has not been widely studied. In the Streptozotocin model of type 1 diabetes, we employed the Conditioned Place Preference test to assess tonic pain. Our results indicate that both phases, that is, early evoked hypersensitivity (i.e. 5-7 weeks post-Streptozotocin) as well as late stage hypoalgesia (i.e. 17-20 weeks post-Streptozotocin) are accompanied by significant tonic pain in mice with diabetic peripheral neuropathy. We also report on the temporal relation between on-going pain and neuropathological changes in the dorsal root ganglia of mice with diabetic peripheral neuropathy up to 6 months post-Streptozotocin. Neither early hypersensitivity nor late hypoalgesia were associated with markers of cellular stress in the dorsal root ganglia. Whereas significant neutrophil infiltration was observed in the dorsal root ganglia over both early and late stages post-Streptozotocin, T-cell infiltration in the dorsal root ganglia was prominent at late stages post-Streptozotocin. Thus, longitudinal analyses reveal that similar to patients with chronic diabetic peripheral neuropathy, mice show tonic pain despite sensory loss after several months in the Streptozotocin model, which is accompanied by neuroimmune interactions in the dorsal root ganglia.


Assuntos
Neuropatias Diabéticas/complicações , Neuropatias Diabéticas/imunologia , Gânglios Espinais/imunologia , Gânglios Espinais/patologia , Hiperalgesia/complicações , Neuralgia/complicações , Fator 3 Ativador da Transcrição/metabolismo , Animais , Complexo CD3/metabolismo , Neuropatias Diabéticas/patologia , Hiperalgesia/patologia , Masculino , Camundongos Endogâmicos C57BL , Neuralgia/patologia
3.
Mol Pain ; 13: 1744806917701743, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28326941

RESUMO

Abstract: Chronic pain represents a frequent and poorly understood public health issue. Numerous studies have documented the key significance of plastic changes along the somatosensory pain pathways in chronic pain states. Our recent study demonstrated that the cGMP-dependent protein kinase I (PKG-I) specifically localized in nociceptors constitutes a key mediator of hyperexcitability of primary sensory neurons and spinal synaptic plasticity after inflammation. However, whether PKG-I in nociceptors further affects the cortical plasticity in the ascending pain pathways under pathological states has remained elusive. The immediate-early gene c-fos and phosphorylated ERK1/2 (pERK1/2) are considered reliable indicators for the neuronal activation status and it permits a comprehensive and large-scale observation of nociceptive neuronal activity along the ascending pain pathways subjected to tissue injury. In the present study, we systemically demonstrated that peripheral injury in PKG-Ifl/fl mice produced a significant upregulation of c-Fos or pERK1/2 over from the periphery to the cortex along the pain pathways, including dorsal root ganglion, spinal dorsal horn, ventral posterolateral thalamus, primary somatosensory hindlimb cortex, anterior cingulate cortex, basolateral amygdala, periaqueductal gray, and parabrachial nucleus. In contrast, very few cells in the above regions showed c-Fos or pERK1/2 induction in nociceptor-specific knockout mice lacking PKG-I (SNS-PKG-I/ mice). Our results indicate that PKG-I expressed in nociceptors is not only a key determinant of dorsal root ganglion hyperexcitability and spinal synaptic plasticity but also an important modulator of cortical neuronal activity in pathological pain states and represent what we believe to be novel targets in the periphery for pain therapeutics.


Assuntos
Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Nociceptores/metabolismo , Animais , Camundongos Transgênicos , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Neurônios Aferentes/metabolismo , Transdução de Sinais/fisiologia
4.
PLoS Biol ; 10(3): e1001283, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22427743

RESUMO

Synaptic long-term potentiation (LTP) at spinal neurons directly communicating pain-specific inputs from the periphery to the brain has been proposed to serve as a trigger for pain hypersensitivity in pathological states. Previous studies have functionally implicated the NMDA receptor-NO pathway and the downstream second messenger, cGMP, in these processes. Because cGMP can broadly influence diverse ion-channels, kinases, and phosphodiesterases, pre- as well as post-synaptically, the precise identity of cGMP targets mediating spinal LTP, their mechanisms of action, and their locus in the spinal circuitry are still unclear. Here, we found that Protein Kinase G1 (PKG-I) localized presynaptically in nociceptor terminals plays an essential role in the expression of spinal LTP. Using the Cre-lox P system, we generated nociceptor-specific knockout mice lacking PKG-I specifically in presynaptic terminals of nociceptors in the spinal cord, but not in post-synaptic neurons or elsewhere (SNS-PKG-I(-/-) mice). Patch clamp recordings showed that activity-induced LTP at identified synapses between nociceptors and spinal neurons projecting to the periaqueductal grey (PAG) was completely abolished in SNS-PKG-I(-/-) mice, although basal synaptic transmission was not affected. Analyses of synaptic failure rates and paired-pulse ratios indicated a role for presynaptic PKG-I in regulating the probability of neurotransmitter release. Inositol 1,4,5-triphosphate receptor 1 and myosin light chain kinase were recruited as key phosphorylation targets of presynaptic PKG-I in nociceptive neurons. Finally, behavioural analyses in vivo showed marked defects in SNS-PKG-I(-/-) mice in several models of activity-induced nociceptive hypersensitivity, and pharmacological studies identified a clear contribution of PKG-I expressed in spinal terminals of nociceptors. Our results thus indicate that presynaptic mechanisms involving an increase in release probability from nociceptors are operational in the expression of synaptic LTP on spinal-PAG projection neurons and that PKG-I localized in presynaptic nociceptor terminals plays an essential role in this process to regulate pain sensitivity.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Potenciação de Longa Duração , Nociceptores/metabolismo , Dor/patologia , Aminoquinolinas/farmacologia , Animais , Comportamento Animal , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Proteína Quinase Dependente de GMP Cíclico Tipo I , Proteínas Quinases Dependentes de GMP Cíclico/genética , Ativação Enzimática , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Deleção de Genes , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Fibras Nervosas/metabolismo , Fibras Nervosas/patologia , Nociceptores/efeitos dos fármacos , Nociceptores/patologia , Dor/metabolismo , Técnicas de Patch-Clamp , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Transdução de Sinais , Especificidade por Substrato , Transmissão Sináptica
5.
Science ; 377(6602): eabo0924, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35737810

RESUMO

The human cerebral cortex houses 1000 times more neurons than that of the cerebral cortex of a mouse, but the possible differences in synaptic circuits between these species are still poorly understood. We used three-dimensional electron microscopy of mouse, macaque, and human cortical samples to study their cell type composition and synaptic circuit architecture. The 2.5-fold increase in interneurons in humans compared with mice was compensated by a change in axonal connection probabilities and therefore did not yield a commensurate increase in inhibitory-versus-excitatory synaptic input balance on human pyramidal cells. Rather, increased inhibition created an expanded interneuron-to-interneuron network, driven by an expansion of interneuron-targeting interneuron types and an increase in their synaptic selectivity for interneuron innervation. These constitute key neuronal network alterations in the human cortex.


Assuntos
Córtex Cerebral , Conectoma , Animais , Córtex Cerebral/ultraestrutura , Humanos , Interneurônios/ultraestrutura , Macaca , Camundongos , Células Piramidais/ultraestrutura
6.
Science ; 378(6626): 1336-1343, 2022 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-36548429

RESUMO

The primary motor cortex (M1) is involved in the control of voluntary movements and is extensively mapped in this capacity. Although the M1 is implicated in modulation of pain, the underlying circuitry and causal underpinnings remain elusive. We unexpectedly unraveled a connection from the M1 to the nucleus accumbens reward circuitry through a M1 layer 6-mediodorsal thalamus pathway, which specifically suppresses negative emotional valence and associated coping behaviors in neuropathic pain. By contrast, layer 5 M1 neurons connect with specific cell populations in zona incerta and periaqueductal gray to suppress sensory hypersensitivity without altering pain affect. Thus, the M1 employs distinct, layer-specific pathways to attune sensory and aversive-emotional components of neuropathic pain, which can be exploited for purposes of pain relief.


Assuntos
Córtex Motor , Vias Neurais , Neuralgia , Córtex Motor/citologia , Córtex Motor/fisiologia , Vias Neurais/citologia , Vias Neurais/fisiologia , Neuralgia/fisiopatologia , Neurônios/fisiologia , Substância Cinzenta Periaquedutal/citologia , Substância Cinzenta Periaquedutal/fisiologia , Tálamo/citologia , Tálamo/fisiologia , Animais , Camundongos
7.
Mol Pain ; 5: 68, 2009 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19925671

RESUMO

BACKGROUND: gamma-aminobutyric acid (GABA) is an important inhibitory neurotransmitter which mainly mediates its effects on neurons via ionotropic (GABA(A)) and metabotropic (GABA(B)) receptors. GABA(B) receptors are widely expressed in the central and the peripheral nervous system. Although there is evidence for a key function of GABA(B) receptors in the modulation of pain, the relative contribution of peripherally- versus centrally-expressed GABA(B) receptors is unclear. RESULTS: In order to elucidate the functional relevance of GABA(B) receptors expressed in peripheral nociceptive neurons in pain modulation we generated and analyzed conditional mouse mutants lacking functional GABA(B1) subunit specifically in nociceptors, preserving expression in the spinal cord and brain (SNS-GABA(B1)-/- mice). Lack of the GABA(B1) subunit precludes the assembly of functional GABA(B) receptor. We analyzed SNS-GABA(B1)-/- mice and their control littermates in several models of acute and neuropathic pain. Electrophysiological studies on peripheral afferents revealed higher firing frequencies in SNS-GABA(B1)-/- mice compared to corresponding control littermates. However no differences were seen in basal nociceptive sensitivity between these groups. The development of neuropathic and chronic inflammatory pain was similar across the two genotypes. The duration of nocifensive responses evoked by intraplantar formalin injection was prolonged in the SNS-GABAB(1)-/- animals as compared to their control littermates. Pharmacological experiments revealed that systemic baclofen-induced inhibition of formalin-induced nociceptive behaviors was not dependent upon GABA(B1) expression in nociceptors. CONCLUSION: This study addressed contribution of GABA(B) receptors expressed on primary afferent nociceptive fibers to the modulation of pain. We observed that neither the development of acute and chronic pain nor the analgesic effects of a systematically-delivered GABA(B) agonist was significantly changed upon a specific deletion of GABA(B) receptors from peripheral nociceptive neurons in vivo. This lets us conclude that GABA(B) receptors in the peripheral nervous system play a less important role than those in the central nervous system in the regulation of pain.


Assuntos
Nociceptores/metabolismo , Receptores de GABA-B/fisiologia , Animais , Western Blotting , Eletrofisiologia , Deleção de Genes , Hibridização In Situ , Inflamação/metabolismo , Camundongos , Camundongos Transgênicos , Neuralgia/metabolismo , Nervos Periféricos/metabolismo , Receptores de GABA-B/genética
8.
Nat Neurosci ; 20(11): 1591-1601, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28920932

RESUMO

The identity of cortical circuits mediating nociception and pain is largely unclear. The cingulate cortex is consistently activated during pain, but the functional specificity of cingulate divisions, the roles at distinct temporal phases of central plasticity and the underlying circuitry are unknown. Here we show in mice that the midcingulate division of the cingulate cortex (MCC) does not mediate acute pain sensation and pain affect, but gates sensory hypersensitivity by acting in a wide cortical and subcortical network. Within this complex network, we identified an afferent MCC-posterior insula pathway that can induce and maintain nociceptive hypersensitivity in the absence of conditioned peripheral noxious drive. This facilitation of nociception is brought about by recruitment of descending serotonergic facilitatory projections to the spinal cord. These results have implications for our understanding of neuronal mechanisms facilitating the transition from acute to long-lasting pain.


Assuntos
Córtex Cerebral/patologia , Córtex Cerebral/fisiologia , Giro do Cíngulo/patologia , Giro do Cíngulo/fisiologia , Dor/patologia , Dor/fisiopatologia , Vias Aferentes/química , Vias Aferentes/patologia , Vias Aferentes/fisiologia , Animais , Córtex Cerebral/química , Giro do Cíngulo/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética/métodos , Técnicas de Cultura de Órgãos , Medição da Dor/métodos
9.
Neuron ; 93(1): 179-193, 2017 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-27989460

RESUMO

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


Assuntos
Potenciais de Ação , Dor Aguda/genética , Mecanorreceptores/metabolismo , Fibras Nervosas Mielinizadas/metabolismo , Neurônios/metabolismo , Nociceptividade/fisiologia , Nociceptores/metabolismo , Somação de Potenciais Pós-Sinápticos , Animais , Comportamento Animal , Gânglios Espinais/citologia , Imuno-Histoquímica , Camundongos , Fibras Nervosas Mielinizadas/fisiologia , Dor Nociceptiva , Optogenética , Dor , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Neuropeptídeo Y/genética , Receptores de Neuropeptídeo Y/metabolismo , Reflexo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tato/fisiologia
10.
Neuron ; 87(4): 673-5, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26291151

RESUMO

How do spinal circuits mediating tactile sensation and pain get entangled to evoke allodynia, i.e., pain sensation, in response to a normally innocuous stimulus? Recent breakthroughs are now closing this long-standing, critical gap. VGLUT3-expressing neurons and their polysynaptic connectivity to calretinin-expressing neurons are now identified as key determinants of the spinal circuitry underlying mechanical allodynia.


Assuntos
Sistemas de Transporte de Aminoácidos Acídicos/biossíntese , Hiperalgesia/metabolismo , Rede Nervosa/metabolismo , Dor/metabolismo , Corno Dorsal da Medula Espinal/metabolismo , Tato , Animais
11.
Cancer Cell ; 27(6): 780-96, 2015 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-26058077

RESUMO

Cancer pain is a debilitating disorder and a primary determinant of the poor quality of life. Here, we report a non-vascular role for ligands of the Vascular Endothelial Growth Factor (VEGF) family in cancer pain. Tumor-derived VEGF-A, PLGF-2, and VEGF-B augment pain sensitivity through selective activation of VEGF receptor 1 (VEGFR1) expressed in sensory neurons in human cancer and mouse models. Sensory-neuron-specific genetic deletion/silencing or local or systemic blockade of VEGFR1 prevented tumor-induced nerve remodeling and attenuated cancer pain in diverse mouse models in vivo. These findings identify a therapeutic potential for VEGFR1-modifying drugs in cancer pain and suggest a palliative effect for VEGF/VEGFR1-targeting anti-angiogenic tumor therapies.


Assuntos
Neoplasias/patologia , Dor/metabolismo , Células Receptoras Sensoriais/metabolismo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Inibidores da Angiogênese/farmacologia , Animais , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias/metabolismo , Dor/tratamento farmacológico , Dor/patologia , Células Receptoras Sensoriais/efeitos dos fármacos , Células Receptoras Sensoriais/patologia , Regulação para Cima , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
12.
Dis Model Mech ; 6(4): 889-95, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23828645

RESUMO

There are two basic categories of pain: physiological pain, which serves an important protective function, and pathological pain, which can have a major negative impact on quality of life in the context of human disease. Major progress has been made in understanding the molecular mechanisms that drive sensory transduction, amplification and conduction in peripheral pain-sensing neurons, communication of sensory inputs to spinal second-order neurons, and the eventual modulation of sensory signals by spinal and descending circuits. This poster article endeavors to provide an overview of how molecular and cellular mechanisms underlying nociception in a physiological context undergo plasticity in pathophysiological states, leading to pain hypersensitivity and chronic pain.


Assuntos
Hipersensibilidade/metabolismo , Dor/metabolismo , Animais , Comunicação Celular , Dor Crônica/metabolismo , Dor Crônica/terapia , Humanos , Nociceptores/metabolismo , Transdução de Sinais
13.
Pain ; 154(12): 2801-2812, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23973358

RESUMO

The rich diversity of lipids and the specific signalling pathways they recruit provides tremendous scope for modulation of biological functions. Lysophosphatidylinositol (LPI) is emerging as a key modulator of cell proliferation, migration, and function, and holds important pathophysiological implications due to its high levels in diseased tissues, such as in cancer. Here we report a novel role for LPI in sensitization of peripheral sensory neurons, which was evident as exaggerated sensitivity to painful and innocuous pressure. Histopathological analyses indicated lack of involvement of myelin pathology and immune cell recruitment by LPI. Using pharmacological and conditional genetic tools in mice, we delineated receptor-mediated from non-receptor-mediated effects of LPI and we observed that GPR55, which functions as an LPI receptor when heterologously expressed in mammalian cells, only partially mediates LPI-induced actions in the context of pain sensitization in vivo; we demonstrate that, in vivo, LPI functions by activating Gα(13) as well as Gα(q/11) arms of G-protein signalling in sensory neurons. This study thus reports a novel pathophysiological function for LPI and elucidates underlying molecular mechanisms.


Assuntos
Subunidades alfa G12-G13 de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Lisofosfolipídeos/fisiologia , Nociceptividade/fisiologia , Células Receptoras Sensoriais/fisiologia , Transdução de Sinais/fisiologia , Animais , Relação Dose-Resposta a Droga , Proteínas de Ligação ao GTP/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nociceptividade/efeitos dos fármacos , Fosfolipídeos/farmacologia , Fosfolipídeos/fisiologia , Células Receptoras Sensoriais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
14.
J Clin Invest ; 121(4): 1608-23, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21383497

RESUMO

α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type (AMPA-type) glutamate receptors (AMPARs) play an important role in plasticity at central synapses. Although there is anatomical evidence for AMPAR expression in the peripheral nervous system, the functional role of such receptors in vivo is not clear. To address this issue, we generated mice specifically lacking either of the key AMPAR subunits, GluA1 or GluA2, in peripheral, pain-sensing neurons (nociceptors), while preserving expression of these subunits in the central nervous system. Nociceptor-specific deletion of GluA1 led to disruption of calcium permeability and reduced capsaicin-evoked activation of nociceptors. Deletion of GluA1, but not GluA2, led to reduced mechanical hypersensitivity and sensitization in models of chronic inflammatory pain and arthritis. Further analysis revealed that GluA1-containing AMPARs regulated the responses of nociceptors to painful stimuli in inflamed tissues and controlled the excitatory drive from the periphery into the spinal cord. Consequently, peripherally applied AMPAR antagonists alleviated inflammatory pain by specifically blocking calcium-permeable AMPARs, without affecting physiological pain or eliciting central side effects. These findings indicate an important pathophysiological role for calcium-permeable AMPARs in nociceptors and may have therapeutic implications for the treatment chronic inflammatory pain states.


Assuntos
Inflamação/fisiopatologia , Nociceptores/fisiologia , Dor/fisiopatologia , Receptores de AMPA/fisiologia , Vias Aferentes/crescimento & desenvolvimento , Vias Aferentes/fisiopatologia , Animais , Artrite Experimental/fisiopatologia , Benzodiazepinas/farmacologia , Cálcio/metabolismo , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/fisiopatologia , Inflamação/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dor/tratamento farmacológico , Permeabilidade , Receptores de AMPA/antagonistas & inibidores , Receptores de AMPA/deficiência , Receptores de AMPA/genética , Transmissão Sináptica
15.
Nat Med ; 15(7): 802-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19525966

RESUMO

Pain is one of the most severe and debilitating symptoms associated with several forms of cancer. Various types of carcinomas and sarcomas metastasize to skeletal bones and cause spontaneous bone pain and hyperalgesia, which is accompanied by bone degradation and remodeling of peripheral nerves. Despite recent advances, the molecular mechanisms underlying the development and maintenance of cancer-evoked pain are not well understood. Several types of non-hematopoietic tumors secrete hematopoietic colony-stimulating factors that act on myeloid cells and tumor cells. Here we report that receptors and signaling mediators of granulocyte- and granulocyte-macrophage colony-stimulating factors (G-CSF and GM-CSF) are also functionally expressed on sensory nerves. GM-CSF sensitized nerves to mechanical stimuli in vitro and in vivo, potentiated CGRP release and caused sprouting of sensory nerve endings in the skin. Interruption of G-CSF and GM-CSF signaling in vivo led to reduced tumor growth and nerve remodeling, and abrogated bone cancer pain. The key significance of GM-CSF signaling in sensory neurons was revealed by an attenuation of tumor-evoked pain following a sensory nerve-specific knockdown of GM-CSF receptors. These results show that G-CSF and GM-CSF are important in tumor-nerve interactions and suggest that their receptors on primary afferent nerve fibers constitute potential therapeutic targets in cancer pain.


Assuntos
Neoplasias Ósseas/fisiopatologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Dor Intratável/etiologia , Células Receptoras Sensoriais/fisiologia , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/fisiologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/fisiologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA