Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cereb Cortex ; 32(8): 1574-1592, 2022 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-34607362

RESUMEN

Plasticity is the mechanistic basis of development, aging, learning, and memory, both in healthy and pathological brains. Structural plasticity is rarely accounted for in computational network models due to a lack of insight into the underlying neuronal mechanisms and processes. Little is known about how the rewiring of networks is dynamically regulated. To inform such models, we characterized the time course of neural activity, the expression of synaptic proteins, and neural morphology employing an in vivo optogenetic mouse model. We stimulated pyramidal neurons in the anterior cingulate cortex of mice and harvested their brains at 1.5 h, 24 h, and $48\,\mathrm{h}$ after stimulation. Stimulus-induced cortical hyperactivity persisted up to 1.5 h and decayed to baseline after $24\,\mathrm{h}$ indicated by c-Fos expression. The synaptic proteins VGLUT1 and PSD-95, in contrast, were upregulated at $24\,\mathrm{h}$ and downregulated at $48\,\mathrm{h}$, respectively. Spine density and spine head volume were also increased at $24\,\mathrm{h}$ and decreased at $48\,\mathrm{h}$. This specific sequence of events reflects a continuous joint evolution of activity and connectivity that is characteristic of the model of homeostatic structural plasticity. Our computer simulations thus corroborate the observed empirical evidence from our animal experiments.


Asunto(s)
Giro del Cíngulo , Optogenética , Animales , Espinas Dendríticas/fisiología , Giro del Cíngulo/fisiología , Plasticidad Neuronal/fisiología , Células Piramidales/metabolismo
2.
Neurobiol Dis ; 155: 105363, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33845128

RESUMEN

Endogenous acetylcholine (ACh) is an important modulator of nociceptive sensory processing in the spinal cord. An increased level of spinal ACh induces analgesia both in humans and rodents while interfering with cholinergic signaling is allodynic, demonstrating that a basal tone of spinal ACh modulates nociceptive responses in naïve animals. The plasticity undergone by this cholinergic system in chronic pain situation is unknown, and the mere presence of this tone in neuropathic animals is controversial. We have addressed these issues in mice through behavioral experiments, histology, electrophysiology and molecular biology, in the cuff model of peripheral neuropathy. Our behavior experiments demonstrate the persistence, and even increased impact of the analgesic cholinergic tone acting through nicotinic receptors in cuff animals. The neuropathy does not affect the number or membrane properties of dorsal horn cholinergic neurons, nor specifically the frequency of their synaptic inputs. The alterations thus appear to be in the neurons receiving the cholinergic signaling, which is confirmed by the fact that subthreshold doses of acetylcholinesterase (AChE) inhibitors in sham animals become anti-allodynic in cuff mice and by the altered expression of the ß2 nicotinic receptor subunit. Our results demonstrate that endogenous cholinergic signaling can be manipulated to relieve mechanical allodynia in animal models of peripheral neuropathy. Until now, AChE inhibitors have mainly been used in the clinics in situations of acute pain (parturition, post-operative). The fact that lower doses (thus with fewer side effects) could be efficient in chronic pain conditions opens new avenues for the treatment of neuropathic pain. SIGNIFICANCE STATEMENT: Chronic pain continues to be the most common cause of disability that impairs the quality of life, accruing enormous and escalating socio-economic costs. A better understanding of the plasticity of spinal neuronal networks, crucially involved in nociceptive processing, could help designing new therapeutic avenues. We here demonstrate that chronic pain modifies the spinal nociceptive network in such a way that it becomes more sensitive to cholinergic modulations. The spinal cholinergic system is responsible for an analgesic tone that can be exacerbated by acetylcholinesterase inhibitors, a property used in the clinic to relief acute pain (child birth, post-op). Our results suggest that lower doses of acetylcholinesterases, with even fewer side effects, could be efficient to relieve chronic pain.


Asunto(s)
Analgesia/métodos , Neuronas Colinérgicas/metabolismo , Modelos Animales de Enfermedad , Neuralgia/metabolismo , Umbral del Dolor/fisiología , Médula Espinal/metabolismo , Acetilcolina/metabolismo , Acetilcolinesterasa/metabolismo , Animales , Neuronas Colinérgicas/efectos de los fármacos , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neuralgia/tratamiento farmacológico , Umbral del Dolor/efectos de los fármacos , Médula Espinal/efectos de los fármacos
3.
Eur J Neurosci ; 53(1): 236-270, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-31985104

RESUMEN

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.


Asunto(s)
Dolor Crónico , Trastorno Depresivo Mayor , Animales , Ansiedad , Comorbilidad , Depresión , Humanos , Roedores
4.
J Neurosci ; 38(12): 3102-3115, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29463643

RESUMEN

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.


Asunto(s)
Dolor Crónico/fisiopatología , Dolor Crónico/psicología , Giro del Cíngulo/fisiopatología , Neuralgia/fisiopatología , Neuralgia/psicología , Animales , Ansiedad/etiología , Ansiedad/fisiopatología , Depresión/etiología , Depresión/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
J Neurosci ; 38(46): 9934-9954, 2018 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-30249798

RESUMEN

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.


Asunto(s)
Amitriptilina/administración & dosificación , Antidepresivos/administración & dosificación , Clorhidrato de Duloxetina/administración & dosificación , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Norepinefrina/metabolismo , Adulto , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Manejo del Dolor/métodos , Receptor de Adenosina A2A/metabolismo
6.
Cell Tissue Res ; 377(1): 21-43, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30778732

RESUMEN

The increasing number of individuals with comorbidities poses an urgent need to improve the management of patients with multiple co-existing diseases. Among these comorbidities, chronic pain and mood disorders, two long-lasting disabling conditions that significantly reduce the quality of life, could be cited first. The recent development of animal models accelerated the studies focusing on the underlying mechanisms of the chronic pain and depression/anxiety comorbidity. This review provides an overview of clinical and pre-clinical studies performed over the past two decades addressing the molecular aspects of the comorbid relationship of chronic pain and depression. We thus focused on the studies that investigated the molecular characteristics of the comorbid relationship between chronic pain and mood disorders, especially major depressive disorders, from the genetic and epigenetic point of view to key neuromodulators which have been shown to play an important role in this comorbidity.


Asunto(s)
Dolor Crónico/epidemiología , Dolor Crónico/genética , Trastorno Depresivo Mayor/epidemiología , Trastorno Depresivo Mayor/genética , Animales , Trastornos de Ansiedad/tratamiento farmacológico , Trastornos de Ansiedad/epidemiología , Trastornos de Ansiedad/genética , Monoaminas Biogénicas/farmacología , Monoaminas Biogénicas/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Comorbilidad , Trastorno Depresivo Mayor/tratamiento farmacológico , Modelos Animales de Enfermedad , Epigénesis Genética , Humanos , Ratones , Calidad de Vida , Ratas , Factores de Transcripción/metabolismo
7.
Mol Pain ; 14: 1744806917749683, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29212409

RESUMEN

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.


Asunto(s)
Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Animales , Anticonvulsivantes/uso terapéutico , Antidepresivos/uso terapéutico , Dexametasona/uso terapéutico , Ketamina/uso terapéutico , Ratones Endogámicos C57BL
8.
Eur J Neurosci ; 48(5): 2231-2246, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30059180

RESUMEN

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.


Asunto(s)
Clorhidrato de Duloxetina/farmacología , Neuralgia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides delta/efectos de los fármacos , Animales , Antidepresivos Tricíclicos/farmacología , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Masculino , Ratones Transgénicos , Neuralgia/metabolismo , Nortriptilina/farmacología , Dimensión del Dolor/métodos , Receptores Opioides delta/metabolismo , Nervio Ciático/efectos de los fármacos , Nervio Ciático/metabolismo
9.
Mol Pain ; 122016.
Artículo en Inglés | MEDLINE | ID: mdl-27030724

RESUMEN

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.


Asunto(s)
Hiperalgesia/complicaciones , Hiperalgesia/tratamiento farmacológico , Neuralgia/complicaciones , Neuralgia/tratamiento farmacológico , Pregabalina/uso terapéutico , Receptores Opioides/metabolismo , Administración Oral , Animales , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones Endogámicos C57BL , Naloxona/farmacología , Naloxona/uso terapéutico , Pregabalina/administración & dosificación , Factores de Tiempo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo
12.
Eur J Neurosci ; 40(8): 3189-201, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25104469

RESUMEN

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.


Asunto(s)
Neuronas/fisiología , Asta Dorsal de la Médula Espinal/fisiología , Transmisión Sináptica , Canales Catiónicos TRPV/fisiología , Vías Aferentes , Animales , Capsaicina/farmacología , Células Cultivadas , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/fisiología , Hiperalgesia/inducido químicamente , Masculino , Neuronas/efectos de los fármacos , Nocicepción/efectos de los fármacos , Nocicepción/fisiología , Probenecid/farmacología , Ratas , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Canales Catiónicos TRPV/agonistas
14.
Curr Opin Anaesthesiol ; 27(5): 520-7, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25188218

RESUMEN

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.


Asunto(s)
Trastornos de Ansiedad/epidemiología , Dolor Crónico/epidemiología , Trastorno Depresivo/epidemiología , Comorbilidad , Humanos , Prevalencia
15.
Neurobiol Dis ; 60: 39-50, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23978467

RESUMEN

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.


Asunto(s)
Antidepresivos Tricíclicos/farmacología , Ganglios Espinales/metabolismo , Neuralgia/tratamiento farmacológico , Receptores Adrenérgicos beta 2/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Agonistas de Receptores Adrenérgicos beta 2/farmacología , Animales , Antiinflamatorios no Esteroideos/farmacología , Anticuerpos Monoclonales/farmacología , Antidepresivos Tricíclicos/uso terapéutico , Etanercept , Ganglios Espinales/patología , Inmunoglobulina G/farmacología , Infliximab , Masculino , Ratones , Ratones Endogámicos C57BL , Neuralgia/metabolismo , Norepinefrina/metabolismo , Nortriptilina/farmacología , Dimensión del Dolor , Receptores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo
16.
Int J Neuropsychopharmacol ; 16(7): 1649-60, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23425507

RESUMEN

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.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Analgésicos Opioides/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Morfina/farmacología , Puente/metabolismo , Amígdala del Cerebelo/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Adaptación a la Oscuridad/efectos de los fármacos , Dependovirus/genética , Conducta Exploratoria/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/efectos de los fármacos , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Puente/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Prueba de Desempeño de Rotación con Aceleración Constante
17.
Pharmacol Ther ; 251: 108546, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37832728

RESUMEN

Neuropathic pain, caused by a lesion or disease affecting the somatosensory system, affects between 3 and 17% of the general population. The treatment of neuropathic pain is challenging due to its heterogeneous etiologies, lack of objective diagnostic tools and resistance to classical analgesic drugs. First-line treatments recommended by the Special Interest Group on Neuropathic Pain (NeuPSIG) and European Federation of Neurological Societies (EFNS) include gabapentinoids, tricyclic antidepressants (TCAs) and selective serotonin noradrenaline reuptake inhibitors (SNRIs). Nevertheless these treatments have modest efficacy or dose limiting side effects. There is therefore a growing number of preclinical and clinical studies aim at developing new treatment strategies to treat neuropathic pain with better efficacy, selectivity, and less side effects. In this review, after a brief description of the mechanisms of action, efficacy, and limitations of current therapeutic drugs, we reviewed new preclinical and clinical targets currently under investigation, as well as promising non-pharmacological alternatives and their potential co-use with pharmacological treatments.


Asunto(s)
Antidepresivos , Neuralgia , Humanos , Antidepresivos/farmacología , Neuralgia/tratamiento farmacológico , Analgésicos/efectos adversos , Inhibidores Selectivos de la Recaptación de Serotonina , Norepinefrina
18.
Neurosci Biobehav Rev ; 153: 105362, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37595650

RESUMEN

Over the past 20 years, clinical and preclinical studies point to the anterior cingulate cortex (ACC) as a site of interest for several neurological and psychiatric conditions. The ACC plays a critical role in emotion, autonomic regulation, pain processing, attention, memory and decision making. An increasing number of studies have demonstrated the involvement of the ACC in the emotional component of pain and its comorbidity with emotional disorders such as anxiety and depression. Thanks to the development of animal models combined with state-of-the-art technologies, we now have a better mechanistic understanding of the functions of the ACC. Hence, the primary aim of this review is to compile the most recent preclinical studies on the role of ACC in the emotional component and consequences of chronic pain. Herein, we thus thoroughly describe the pain-induced electrophysiological, molecular and anatomical alterations in the ACC and in its related circuits. Finally, we discuss the next steps that are needed to strengthen our understanding of the involvement of the ACC in emotional and pain processing.

19.
Genome Biol ; 24(1): 261, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37968726

RESUMEN

BACKGROUND: Using mouse genetic studies and systematic assessments of brain neuroanatomical phenotypes, we set out to identify which of the 30 genes causes brain defects at the autism-associated 16p11.2 locus. RESULTS: We show that multiple genes mapping to this region interact to regulate brain anatomy, with female mice exhibiting far fewer brain neuroanatomical phenotypes. In male mice, among the 13 genes associated with neuroanatomical defects (Mvp, Ppp4c, Zg16, Taok2, Slx1b, Maz, Fam57b, Bola2, Tbx6, Qprt, Spn, Hirip3, and Doc2a), Mvp is the top driver implicated in phenotypes pertaining to brain, cortex, hippocampus, ventricles, and corpus callosum sizes. The major vault protein (MVP), the main component of the vault organelle, is a conserved protein found in eukaryotic cells, yet its function is not understood. Here, we find MVP expression highly specific to the limbic system and show that Mvp regulates neuronal morphology, postnatally and specifically in males. We also recapitulate a previously reported genetic interaction and show that Mvp+/-;Mapk3+/- mice exhibit behavioral deficits, notably decreased anxiety-like traits detected in the elevated plus maze and open field paradigms. CONCLUSIONS: Our study highlights multiple gene drivers in neuroanatomical phenotypes, interacting with each other through complex relationships. It also provides the first evidence for the involvement of the major vault protein in the regulation of brain size and neuroanatomy, specifically in male mice.


Asunto(s)
Trastorno Autístico , Masculino , Animales , Ratones , Femenino , Trastorno Autístico/genética , Trastorno Autístico/metabolismo , Neuroanatomía , Encéfalo/metabolismo , Fenotipo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas de Unión al Calcio/genética , Proteínas del Tejido Nervioso/metabolismo
20.
Nat Commun ; 14(1): 2198, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069164

RESUMEN

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.


Asunto(s)
Complejo Nuclear Basolateral , Dolor Crónico , Semaforinas , Ratones , Masculino , Humanos , Animales , Depresión/genética , Giro del Cíngulo/metabolismo , Complejo Nuclear Basolateral/metabolismo , Comorbilidad , Semaforinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA