Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Eur J Neurosci ; 53(1): 236-270, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-31985104

RESUMO

Mood disorders such as depression and anxiety are frequently observed in patients suffering from chronic pain. Over time, different tests and models have been developed in rodents to study the anxiodepressive-like consequences of chronic pain. This review describes these preclinical tools (models and tests) used for studying behavioural aspects of the comorbid relationship between chronic pain and anxiety and/or major depressive disorder. Three major types of chronic pain strongly associated with anxiodepressive-like comorbidity as well as their animal models are presented: neuropathic pain, inflammatory pain and fibromyalgia. After a description of chronic pain animal models and of the tests that allow determining nociceptive responses, this review presents and discusses the various behavioural tests that have been used to assess anxiety and depressive-like behaviours in these models of chronic pain. Finally, this review highlights the progress that remains to be made to homogenize the results in the field of pain-induced mood disorders and summarizes the recent advances achieved through these tests and models.


Assuntos
Dor Crônica , Transtorno Depressivo Maior , Animais , Ansiedade , Comorbidade , Depressão , Humanos , Roedores
2.
Appetite ; 167: 105597, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34273421

RESUMO

Central dopamine signaling regulates reward-related aspects of feeding behavior, and during diet-induced obesity dopamine receptor signaling is altered. Yet, the influence of dopamine signaling on the consumption of specific dietary components remains to be elucidated. We have previously shown that 6-hydroxydopamine-mediated lesions of dopamine neuron terminals in the lateral shell of the nucleus accumbens promotes fat intake in rats fed a multi-component free-choice high-fat high-sugar (fcHFHS) diet. It is however not yet determined which dopamine receptors are responsible for this shift towards fat preference. In this study, we assess the effects of D1-or D2 receptor acute inhibition in the lateral shell of the nucleus accumbens on fcHFHS diet consumption. We report that infusion of the D1 receptor antagonist SCH2 3390, but not the D2 receptor antagonist raclopride, promotes dietary fat consumption in male Sprague Dawley rats on a fcHFHS diet during 2 h after infusion. Furthermore, anatomical analysis of infusion sites revealed that the rostral region, but not the caudal region, of the lateral shell of the nucleus accumbens is sensitive to the D1 receptor inhibition effects on fat consumption. Our data highlight a role for D1 receptors in the rostral region of the lateral shell of the nucleus accumbens to control dietary fat consumption.


Assuntos
Núcleo Accumbens , Receptores de Dopamina D1 , Animais , Gorduras na Dieta , Masculino , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D2
3.
Neurobiol Dis ; 139: 104818, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32087289

RESUMO

Parkinson's disease is a neurodegenerative disorder partly caused by the loss of the dopamine neurons of the nigrostriatal pathway. It is accompanied by motor as well as non-motor symptoms, including pain and depression. The tail of the ventral tegmental area (tVTA) or rostromedial tegmental nucleus (RMTg) is a GABAergic mesopontine structure that acts as a major inhibitory brake for the substantia nigra pars compacta (SNc) dopamine cells, thus controlling their neuronal activity and related motor functions. The present study tested the influence of suppressing this tVTA brake on motor and non-motor symptoms in a rat model of Parkinson's disease. Using behavioral approaches, we showed that male Sprague-Dawley rats with bilateral and partial 6-hydroxydopamine SNc lesion displayed motor impairments in the rotarod test, impairments that were no more present following a co-lesion of the tVTA. Using a larger set of behavioral tests, we then showed that such SNc lesion also led to non-motor symptoms, including lower body weight, lower mechanical nociceptive thresholds in the forceps test and lower thermal nociceptive thresholds in the incremented hot-plate test, and a decreased sucrose preference in a 2-bottle choice paradigm. The excitotoxic co-lesion of the tVTA led to compensation of body weight, mechanical nociceptive thresholds and anhedonia-like behavior. These findings illustrate the major influence that the tVTA exerts on the dopamine system, modulating the motor and non-motor symptoms related to a partial loss of dopamine cells.


Assuntos
Doença de Parkinson/metabolismo , Área Tegmentar Ventral/metabolismo , Anedonia , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Masculino , Modelos Teóricos , Vias Neurais/metabolismo , Oxidopamina/farmacologia , Parte Compacta da Substância Negra/metabolismo , Ratos , Ratos Sprague-Dawley , Teste de Desempenho do Rota-Rod , Substância Negra/metabolismo
4.
J Neurosci ; 38(12): 3102-3115, 2018 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-29463643

RESUMO

Pain associates both sensory and emotional aversive components, and often leads to anxiety and depression when it becomes chronic. Here, we characterized, in a mouse model, the long-term development of these sensory and aversive components as well as anxiodepressive-like consequences of neuropathic pain and determined their electrophysiological impact on the anterior cingulate cortex (ACC, cortical areas 24a/24b). We show that these symptoms of neuropathic pain evolve and recover in different time courses following nerve injury in male mice. In vivo electrophysiological recordings evidence an increased firing rate and bursting activity within the ACC when anxiodepressive-like consequences developed, and this hyperactivity persists beyond the period of mechanical hypersensitivity. Whole-cell patch-clamp recordings also support ACC hyperactivity, as shown by increased excitatory postsynaptic transmission and contribution of NMDA receptors. Optogenetic inhibition of the ACC hyperactivity was sufficient to alleviate the aversive and anxiodepressive-like consequences of neuropathic pain, indicating that these consequences are underpinned by ACC hyperactivity.SIGNIFICANCE STATEMENT Chronic pain is frequently comorbid with mood disorders, such as anxiety and depression. It has been shown that it is possible to model this comorbidity in animal models by taking into consideration the time factor. In this study, we aimed at determining the dynamic of different components and consequences of chronic pain, and correlated them with electrophysiological alterations. By combining electrophysiological, optogenetic, and behavioral analyses in a mouse model of neuropathic pain, we show that the mechanical hypersensitivity, ongoing pain, anxiodepressive consequences, and their recoveries do not necessarily exhibit temporal synchrony during chronic pain processing, and that the hyperactivity of the anterior cingulate cortex is essential for driving the emotional impact of neuropathic pain.


Assuntos
Dor Crônica/fisiopatologia , Dor Crônica/psicologia , Giro do Cíngulo/fisiopatologia , Neuralgia/fisiopatologia , Neuralgia/psicologia , Animais , Ansiedade/etiologia , Ansiedade/fisiopatologia , Depressão/etiologia , Depressão/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
5.
J Neurosci ; 38(46): 9934-9954, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30249798

RESUMO

In addition to treating depression, antidepressant drugs are also a first-line treatment for neuropathic pain, which is pain secondary to lesion or pathology of the nervous system. Despite the widespread use of these drugs, the mechanism underlying their therapeutic action in this pain context remains partly elusive. The present study combined data collected in male and female mice from a model of neuropathic pain and data from the clinical setting to understand how antidepressant drugs act. We show two distinct mechanisms by which the selective inhibitor of serotonin and noradrenaline reuptake duloxetine and the tricyclic antidepressant amitriptyline relieve neuropathic allodynia. One of these mechanisms is acute, central, and requires descending noradrenergic inhibitory controls and α2A adrenoceptors, as well as the mu and delta opioid receptors. The second mechanism is delayed, peripheral, and requires noradrenaline from peripheral sympathetic endings and ß2 adrenoceptors, as well as the delta opioid receptors. We then conducted a transcriptomic analysis in dorsal root ganglia, which suggested that the peripheral component of duloxetine action involves the inhibition of neuroimmune mechanisms accompanying nerve injury, including the downregulation of the TNF-α-NF-κB signaling pathway. Accordingly, immunotherapies against either TNF-α or Toll-like receptor 2 (TLR2) provided allodynia relief. We also compared duloxetine plasma levels in the animal model and in patients and we observed that patients' drug concentrations were compatible with those measured in animals under chronic treatment involving the peripheral mechanism. Our study highlights a peripheral neuroimmune component of antidepressant drugs that is relevant to their delayed therapeutic action against neuropathic pain.SIGNIFICANCE STATEMENT In addition to treating depression, antidepressant drugs are also a first-line treatment for neuropathic pain, which is pain secondary to lesion or pathology of the nervous system. However, the mechanism by which antidepressant drugs can relieve neuropathic pain remained in part elusive. Indeed, preclinical studies led to contradictions concerning the anatomical and molecular substrates of this action. In the present work, we overcame these apparent contradictions by highlighting the existence of two independent mechanisms. One is rapid and centrally mediated by descending controls from the brain to the spinal cord and the other is delayed, peripheral, and relies on the anti-neuroimmune action of chronic antidepressant treatment.


Assuntos
Amitriptilina/administração & dosagem , Antidepressivos/administração & dosagem , Cloridrato de Duloxetina/administração & dosagem , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Norepinefrina/metabolismo , Adulto , Idoso , Animais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Manejo da Dor/métodos , Receptor A2A de Adenosina/metabolismo
6.
Mol Pain ; 14: 1744806917749683, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29212409

RESUMO

Background Surgeries causing nerve injury can result in chronic neuropathic pain, which is clinically managed by using antidepressant or anticonvulsant drugs. Currently, there is a growing interest for investigating preemptive treatments that would prevent this long-term development of neuropathic pain. Our aim was to compare analgesic drugs using two distinct treatment modalities: either treatment onset at surgery time or following a couple of weeks of neuropathic pain. Methods In male C57BL/6J mice, neuropathic pain was induced by cuffing the sciatic nerve, and allodynia was assessed using von Frey filaments. We tested the effect of anticonvulsants (gabapentin 10 mg/kg and carbamazepine 40 mg/kg), antidepressants (desipramine 5 mg/kg, duloxetine 10 mg/kg, and fluoxetine 10 mg/kg), dexamethasone (2 mg/kg), and ketamine (15 mg/kg). Drugs were injected daily or twice a day, starting either at surgery time or on day 25 postsurgery (15 days of treatment for antidepressants and 10 days for other drugs). Results Ketamine was the only effective treatment during the early postsurgical period. Although early anticonvulsant treatment was not immediately effective, it prevented chronification of allodynia. When treatments started at day 25 postsurgery, desipramine, duloxetine, and anticonvulsants suppressed the mechanical allodynia. Conclusions Our data show that allodynia measured in experimental neuropathic pain model likely results from a combination of different processes (early vs. late allodynia) that display different sensitivity to treatments. We also propose that early anticonvulsant treatment with gabapentin or carbamazepine may have a prophylactic effect on the chronification of allodynia following nerve injury.


Assuntos
Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Neuralgia/complicações , Neuralgia/tratamento farmacológico , Animais , Anticonvulsivantes/uso terapêutico , Antidepressivos/uso terapêutico , Dexametasona/uso terapêutico , Ketamina/uso terapêutico , Camundongos Endogâmicos C57BL
7.
Eur J Neurosci ; 48(5): 2231-2246, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30059180

RESUMO

Peripheral delta opioid (DOP) receptors are essential for the antiallodynic effect of the tricyclic antidepressant nortriptyline. However, the population of DOP-expressing cells affected in neuropathic conditions or underlying the antiallodynic activity of antidepressants remains unknown. Using a mouse line in which DOP receptors were selectively ablated in cells expressing Nav1.8 sodium channels (DOP cKO), we established that these DOP peripheral receptors were mandatory for duloxetine to alleviate mechanical allodynia in a neuropathic pain model based on sciatic nerve cuffing. We then examined the impact of nerve cuffing and duloxetine treatment on DOP-positive populations using a knock-in mouse line expressing a fluorescent version of the DOP receptor fused with the enhanced green fluorescent protein (DOPeGFP). Eight weeks postsurgery, we observed a reduced proportion of DOPeGFP-positive small peptidergic sensory neurons (calcitonin gene-related peptide (CGRP) positive) in dorsal root ganglia and a lower density of DOPeGFP-positive free nerve endings in the skin. These changes were not present in nerve-injured mice chronically treated with oral duloxetine. In addition, increased DOPeGFP translocation to the plasma membrane was observed in neuropathic conditions but not in duloxetine-treated neuropathic mice, which may represent an additional level of control of the neuronal activity by DOP receptors. Our results therefore established a parallel between changes in the expression profile of peripheral DOP receptors and mechanical allodynia induced by sciatic nerve cuffing.


Assuntos
Cloridrato de Duloxetina/farmacologia , Neuralgia/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/efeitos dos fármacos , Animais , Antidepressivos Tricíclicos/farmacologia , Modelos Animais de Doenças , Feminino , Gânglios Espinais/metabolismo , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Camundongos Transgênicos , Neuralgia/metabolismo , Nortriptilina/farmacologia , Medição da Dor/métodos , Receptores Opioides delta/metabolismo , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo
8.
Mol Pain ; 122016.
Artigo em Inglês | MEDLINE | ID: mdl-27030724

RESUMO

BACKGROUND: Clinical management of neuropathic pain, which is pain arising as a consequence of a lesion or a disease affecting the somatosensory system, partly relies on the use of anticonvulsant drugs such as gabapentinoids. Therapeutic action of gabapentinoids such as gabapentin and pregabalin, which act by the inhibition of calcium currents through interaction with the α2δ-1 subunit of voltage-dependent calcium channels, is well documented. However, some aspects of the downstream mechanisms are still to be uncovered. Using behavioral, genetic, and pharmacological approaches, we tested whether opioid receptors are necessary for the antiallodynic action of acute and/or long-term pregabalin treatment in the specific context of neuropathic pain. RESULTS: Using the cuff model of neuropathic pain in mice, we show that acute pregabalin administration at high dose has a transitory antiallodynic action, while prolonged oral pregabalin treatment leads to sustained antiallodynic action, consistent with clinical observations. We show that pregabalin remains fully effective in µ-opioid receptor, in δ-opioid receptor and in κ-opioid receptor deficient mice, either female or male, and its antiallodynic action is not affected by acute naloxone. Our work also shows that long-term pregabalin treatment suppresses tumor necrosis factor-α overproduction induced by sciatic nerve constriction in the lumbar dorsal root ganglia. CONCLUSIONS: We demonstrate that neither acute nor long-term antiallodynic effect of pregabalin in a context of neuropathic pain is mediated by the endogenous opioid system, which differs from opioid treatment of pain and antidepressant treatment of neuropathic pain. Our data are also supportive of an impact of gabapentinoid treatment on the neuroimmune aspect of neuropathic pain.


Assuntos
Hiperalgesia/complicações , Hiperalgesia/tratamento farmacológico , Neuralgia/complicações , Neuralgia/tratamento farmacológico , Pregabalina/uso terapêutico , Receptores Opioides/metabolismo , Administração Oral , Animais , Relação Dose-Resposta a Droga , Feminino , Masculino , Camundongos Endogâmicos C57BL , Naloxona/farmacologia , Naloxona/uso terapêutico , Pregabalina/administração & dosagem , Fatores de Tempo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo
10.
Proc Natl Acad Sci U S A ; 110(40): 16223-8, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-24046366

RESUMO

Climbing fibers, the projections from the inferior olive to the cerebellar cortex, carry sensorimotor error and clock signals that trigger motor learning by controlling cerebellar Purkinje cell synaptic plasticity and discharge. Purkinje cells target the deep cerebellar nuclei, which are the output of the cerebellum and include an inhibitory GABAergic projection to the inferior olive. This pathway identifies a potential closed loop in the olivo-cortico-nuclear network. Therefore, sets of Purkinje cells may phasically control their own climbing fiber afferents. Here, using in vitro and in vivo recordings, we describe a genetically modified mouse model that allows the specific optogenetic control of Purkinje cell discharge. Tetrode recordings in the cerebellar nuclei demonstrate that focal stimulations of Purkinje cells strongly inhibit spatially restricted sets of cerebellar nuclear neurons. Strikingly, such stimulations trigger delayed climbing-fiber input signals in the stimulated Purkinje cells. Therefore, our results demonstrate that Purkinje cells phasically control the discharge of their own olivary afferents and thus might participate in the regulation of cerebellar motor learning.


Assuntos
Cerebelo/citologia , Vias Eferentes/citologia , Núcleo Olivar/citologia , Células de Purkinje/fisiologia , Animais , Channelrhodopsins , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Optogenética , Teste de Desempenho do Rota-Rod
14.
Eur J Neurosci ; 40(8): 3189-201, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25104469

RESUMO

Probenecid, an agonist of transient receptor vanilloid (TRPV) type 2, was used to evaluate the effects of TRPV2 activation on excitatory and inhibitory synaptic transmission in the dorsal horn (DH) of the rat spinal cord and on nociceptive reflexes induced by thermal heat and mechanical stimuli. The effects of probenecid were compared with those of capsaicin, a TRPV1 agonist. Calcium imaging experiments on rat dorsal root ganglion (DRG) and DH cultures indicated that functional TRPV2 and TRPV1 were expressed by essentially non-overlapping subpopulations of DRG neurons, but were absent from DH neurons and DH and DRG glial cells. Pretreatment of DRG cultures with small interfering RNAs against TRPV2 suppressed the responses to probenecid. Patch-clamp recordings from spinal cord slices showed that probenecid and capsaicin increased the frequencies of spontaneous excitatory postsynaptic currents (sEPSCs) and spontaneous inhibitory postsynaptic currents in a subset of laminae III-V neurons. In contrast to capsaicin, probenecid failed to stimulate synaptic transmission in lamina II. Intrathecal or intraplantar injections of probenecid induced mechanical hyperalgesia/allodynia without affecting nociceptive heat responses. Capsaicin induced both mechanical hyperalgesia/allodynia and heat hyperalgesia. Activation of TRPV1 or TRPV2 in distinct sets of primary afferents increased the sEPSC frequencies in a largely common population of DH neurons in laminae III-V, and might underlie the development of mechanical hypersensitivity following probenecid or capsaicin treatment. However, only TRPV1-expressing afferents facilitated excitatory and/or inhibitory transmission in a subpopulation of lamina II neurons, and this phenomenon might be correlated with the induction of thermal heat hyperalgesia.


Assuntos
Neurônios/fisiologia , Corno Dorsal da Medula Espinal/fisiologia , Transmissão Sináptica , Canais de Cátion TRPV/fisiologia , Vias Aferentes , Animais , Capsaicina/farmacologia , Células Cultivadas , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Hiperalgesia/induzido quimicamente , Masculino , Neurônios/efeitos dos fármacos , Nociceptividade/efeitos dos fármacos , Nociceptividade/fisiologia , Probenecid/farmacologia , Ratos , Ratos Sprague-Dawley , Corno Dorsal da Medula Espinal/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Canais de Cátion TRPV/agonistas
15.
Proc Natl Acad Sci U S A ; 108(39): 16446-50, 2011 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-21930931

RESUMO

Morphine is a highly potent analgesic with high addictive potential in specific contexts. Although dopamine neurons of the ventral tegmental area (VTA) are widely believed to play an essential role in the development of drug addiction, neuronal circuits underlying morphine action on dopamine neurons have not been fully elucidated. Here we combined in vivo electrophysiology, tract-tracing experiments, and targeted neuronal inactivation to dissect a neural circuit for acute morphine action on dopamine neurons in rats. We found that in vivo, morphine targets the GABAergic tail of the VTA, also called the rostromedial tegmental nucleus, to increase the firing of dopamine neurons through the activation of VTA µ opioid receptors expressed on tail of the VTA/rostromedial tegmental nucleus efferents. Our data also reveal that in the absence of VTA glutamatergic tone, there is no morphine-induced activation of dopamine neurons. These results define the anatomical organization and functional role of a neural circuit for acute morphine action on dopamine neurons.


Assuntos
Dopamina/metabolismo , Morfina/farmacologia , Neurônios/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Ácido Glutâmico/metabolismo , Neurônios/metabolismo , Neurônios/fisiologia , Ratos , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/metabolismo , Ácido gama-Aminobutírico/metabolismo
16.
Curr Opin Anaesthesiol ; 27(5): 520-7, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25188218

RESUMO

PURPOSE OF REVIEW: Chronic pain is often accompanied by mood, sleep and cognitive complications affecting the patient's quality of life. This reviews aims to provide a synthesis of the recent clinical and preclinical findings concerning the chronic pain and mood disorder comorbidity. RECENT FINDINGS: The possible mechanisms underlying the presence of anxiety and/or depression in neuropathic pain, chronic widespread pain (fibromyalgia) and inflammatory pain are reviewed based on recent evidences from neuroimaging, anatomical, behavioral, pharmacological, genetic and biochemical studies. Clinical data from patients and preclinical findings from pain models in rodents are considered. SUMMARY: The epidemiological studies report a high prevalence of mood disorders in patients with chronic pain, and these consequences of pain can be preclinically modeled. This comorbidity may be explained by shared morphological and functional alterations observed in both chronic pain and mood disorders. However, mechanistic studies also highlight differences in such alterations depending on the type of chronic pain. Better understanding of the genetic and environmental determinants of pain-induced mood disorders and of the various neurobiological bases of this comorbidity depending on the pain subtype could provide the clinician with important diagnosis and treatment tools. Such progress benefits from translational effort between clinical and preclinical research.


Assuntos
Transtornos de Ansiedade/epidemiologia , Dor Crônica/epidemiologia , Transtorno Depressivo/epidemiologia , Comorbidade , Humanos , Prevalência
17.
J Neurosci ; 32(41): 14094-101, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23055478

RESUMO

A new mesopontine structure exerting a strong influence on dopamine systems has recently been defined: the tail of the ventral tegmental area/rostromedial tegmental nucleus (tVTA/RMTg). This review presents a neuroanatomical, physiological, and behavioral overview of some of the recent and ongoing research on this brain region and its relationship with dopamine systems. The tVTA/RMTg sends dense GABA projections to VTA and substantia nigra neurons. The inhibitory influence of tVTA/RMTg on dopamine neurons is supported by both neuroanatomical and electrophysiology data. The latter studies also reveal the tVTA/RMTg as a substrate for morphine and cannabinoid action on dopamine cells. In primates, the tVTA/RMTg has been implicated in reward prediction error signals, through a basal ganglia-lateral habenula-tVTA/RMTg-dopamine-basal ganglia circuit. In rodents, the tVTA/RMTg has been shown to play a critical role in aversive behaviors, particularly those involving behavioral inhibition, such as freezing and avoidance. These findings highlight the functional importance of the tVTA/RMTg as a major GABA brake for dopamine systems.


Assuntos
Corpo Estriado/fisiologia , Dopamina/fisiologia , Rede Nervosa/fisiologia , Substância Negra/fisiologia , Área Tegmentar Ventral/fisiologia , Ácido gama-Aminobutírico/fisiologia , Animais , Humanos
18.
Neurobiol Dis ; 60: 39-50, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23978467

RESUMO

Neuropathic pain is pain arising as a direct consequence of a lesion or disease affecting the somatosensory system. It is usually chronic and challenging to treat. Some antidepressants are first-line pharmacological treatments for neuropathic pain. The noradrenaline that is recruited by the action of the antidepressants on reuptake transporters has been proposed to act through ß2-adrenoceptors (ß2-ARs) to lead to the observed therapeutic effect. However, the complex downstream mechanism mediating this action remained to be identified. In this study, we demonstrate in a mouse model of neuropathic pain that an antidepressant's effect on neuropathic allodynia involves the peripheral nervous system and the inhibition of cytokine tumor necrosis factor α (TNFα) production. The antiallodynic action of nortriptyline is indeed lost after peripheral sympathectomy, but not after lesion of central descending noradrenergic pathways. More particularly, we report that antidepressant-recruited noradrenaline acts, within dorsal root ganglia, on ß2-ARs expressed by non-neuronal satellite cells. This stimulation of ß2-ARs decreases the neuropathy-induced production of membrane-bound TNFα, resulting in relief of neuropathic allodynia. This indirect anti-TNFα action was observed with the tricyclic antidepressant nortriptyline, the selective serotonin and noradrenaline reuptake inhibitor venlafaxine and the ß2-AR agonist terbutaline. Our data revealed an original therapeutic mechanism that may open novel research avenues for the management of painful peripheral neuropathies.


Assuntos
Antidepressivos Tricíclicos/farmacologia , Gânglios Espinais/metabolismo , Neuralgia/tratamento farmacológico , Receptores Adrenérgicos beta 2/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anticorpos Monoclonais/farmacologia , Antidepressivos Tricíclicos/uso terapêutico , Etanercepte , Gânglios Espinais/patologia , Imunoglobulina G/farmacologia , Infliximab , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Norepinefrina/metabolismo , Nortriptilina/farmacologia , Medição da Dor , Receptores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
19.
Int J Neuropsychopharmacol ; 16(7): 1649-60, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23425507

RESUMO

In addition to its neurotrophic role, brain-derived neurotrophic factor (BDNF) is involved in a wide array of functions, including anxiety and pain. The central amygdaloid nucleus (CeA) contains a high concentration of BDNF in terminals, originating from the pontine parabrachial nucleus. Since the spino-parabrachio-amygdaloid neural pathway is known to convey nociceptive information, we hypothesized a possible involvement of BDNF in supraspinal pain-related processes. To test this hypothesis, we generated localized deletion of BDNF in the parabrachial nucleus using local bilateral injections of adeno-associated viruses in adult floxed-BDNF mice. Basal thresholds of thermal and mechanical nociceptive responses were not altered by BDNF loss and no behavioural deficit was noticed in anxiety and motor tests. However, BDNF-deleted animals displayed a major decrease in the analgesic effect of morphine. In addition, intra-CeA injections of the BDNF scavenger TrkB-Fc in control mice also decreased morphine-induced analgesia. Finally, the number of c-Fos immunoreactive nuclei after acute morphine injection was decreased by 45% in the extended amygdala of BDNF-deleted animals. The absence of BDNF in the parabrachial nucleus thus altered the parabrachio-amygdaloid pathway. Overall, our study provides evidence that BDNF produced in the parabrachial nucleus modulates the functions of the parabrachio-amygdaloid pathway in opiate analgesia.


Assuntos
Tonsila do Cerebelo/metabolismo , Analgésicos Opioides/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Morfina/farmacologia , Ponte/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Adaptação à Escuridão/efeitos dos fármacos , Dependovirus/genética , Comportamento Exploratório/efeitos dos fármacos , Hiperalgesia/tratamento farmacológico , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Vias Neurais/efeitos dos fármacos , Medição da Dor , Limiar da Dor/efeitos dos fármacos , Ponte/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Teste de Desempenho do Rota-Rod
20.
Nat Commun ; 14(1): 2198, 2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37069164

RESUMO

While depression and chronic pain are frequently comorbid, underlying neuronal circuits and their psychopathological relevance remain poorly defined. Here we show in mice that hyperactivity of the neuronal pathway linking the basolateral amygdala to the anterior cingulate cortex is essential for chronic pain-induced depression. Moreover, activation of this pathway in naive male mice, in the absence of on-going pain, is sufficient to trigger depressive-like behaviors, as well as transcriptomic alterations that recapitulate core molecular features of depression in the human brain. These alterations notably impact gene modules related to myelination and the oligodendrocyte lineage. Among these, we show that Sema4a, which was significantly upregulated in both male mice and humans in the context of altered mood, is necessary for the emergence of emotional dysfunction. Overall, these results place the amygdalo-cingulate pathway at the core of pain and depression comorbidity, and unravel the role of Sema4a and impaired myelination in mood control.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Dor Crônica , Semaforinas , Camundongos , Masculino , Humanos , Animais , Depressão/genética , Giro do Cíngulo/metabolismo , Complexo Nuclear Basolateral da Amígdala/metabolismo , Comorbidade , Semaforinas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA