Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Mol Pain ; 13: 1744806917727625, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28814148

RESUMEN

Background: Blood­nerve barrier disruption is pivotal in the development of neuroinflammation, peripheral sensitization, and neuropathic pain after peripheral nerve injury. Activation of toll-like receptor 4 and inactivation of Sonic Hedgehog signaling pathways within the endoneurial endothelial cells are key events, resulting in the infiltration of harmful molecules and immunocytes within the nerve parenchyma. However, we showed in a previous study that preemptive inactivation of toll-like receptor 4 signaling or sustained activation of Sonic Hedgehog signaling did not prevent the local alterations observed following peripheral nerve injury, suggesting the implication of another signaling pathway. Methods: Using a classical neuropathic pain model, the infraorbital nerve chronic constriction injury (IoN-CCI), we investigated the role of the Wnt/ß-catenin pathway in chronic constriction injury-mediated blood­nerve barrier disruption and in its interactions with the toll-like receptor 4 and Sonic Hedgehog pathways. In the IoN-CCI model versus control, mRNA expression levels and/or immunochemical detection of major Wnt/Sonic Hedgehog pathway (Frizzled-7, vascular endothelial-cadherin, Patched-1 and Gli-1) and/or tight junction proteins (Claudin-1, Claudin-5, and Occludin) readouts were assessed. Vascular permeability was assessed by sodium fluorescein extravasation. Results: IoN-CCI induced early alterations in the vascular endothelial-cadherin/ß-catenin/Frizzled-7 complex, shown to participate in local blood­nerve barrier disruption via a ß-catenin-dependent tight junction protein downregulation. Wnt pathway also mediated a crosstalk between toll-like receptor 4 and Sonic Hedgehog signaling within endoneurial endothelial cells. Nevertheless, preemptive inhibition of Wnt/ß-catenin signaling before IoN-CCI could not prevent the downregulation of key Sonic Hedgehog pathway readouts or the disruption of the infraorbital blood­nerve barrier, suggesting that Sonic Hedgehog pathway inhibition observed following IoN-CCI is an independent event responsible for blood­nerve barrier disruption. Conclusion: A crosstalk between Wnt/ß-catenin- and Sonic Hedgehog-mediated signaling pathways within endoneurial endothelial cells could mediate the chronic disruption of the blood­nerve barrier following IoN-CCI, resulting in increased irreversible endoneurial vascular permeability and neuropathic pain development.


Asunto(s)
Barrera Hematonerviosa/metabolismo , Células Endoteliales/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Enfermedad Crónica , Constricción Patológica , Proteínas Hedgehog/metabolismo , Masculino , Neuralgia/metabolismo , Traumatismos de los Nervios Periféricos/metabolismo , Ratas Sprague-Dawley , Receptor Toll-Like 4/metabolismo , beta Catenina/metabolismo
2.
J Neurochem ; 136(1): 133-47, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26440453

RESUMEN

After peripheral nerve injury microglial reactivity change in the spinal cord is associated with an early activation of Janus kinase (JAK)/STAT3 transduction pathway whose blockade attenuates local inflammation and pain hypersensitivity. However, the consequences of microglial JAK/STAT3-mediated signaling on neighboring cells are unknown. Using an in vitro paradigm we assessed the impact of microglial JAK/STAT3 activity on functional characteristics of astrocytes and spinal cord neurons. Purified rat primary microglia was stimulated with JAK/STAT3 classical activator interleukin-6 in the presence or absence of a selective STAT3 inhibitor and rat primary astrocytes or spinal cord neurons were exposed to microglia conditioned media (CM). JAK/STAT3 activity-generated microglial CM modulated both astrocyte and neuron characteristics. Beyond inducing mRNA expression changes in various targets of interest in astrocytes and neurons, microglia CM activated c-Jun N-terminal kinase, STAT3 and NF-κB intracellular pathways in astrocytes and promoted their proliferation. Without modifying neuronal excitability or survival, CM affected the nerve processes morphology and distribution of the post-synaptic density protein 95, a marker of glutamatergic synaptic contacts. These findings show that JAK/STAT3 activity in microglia impacts the functional characteristics of astrocytes and neurons. This suggests its participation in spinal cord tissue plasticity and remodeling occurring after peripheral nerve injury. We show that the activity of JAK/STAT3 pathway in microglial cells confers them a specific signaling modality toward neighboring cells, promoting astrocyte proliferation and changes in neuronal morphology. These in vitro data suggest that the early JAK/STAT3 activation in spinal cord microglia, associated with peripheral nerve injury, participates in functional alteration of various cell populations and in spinal tissue remodeling.


Asunto(s)
Astrocitos/metabolismo , Quinasas Janus/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Factor de Transcripción STAT3/metabolismo , Médula Espinal/metabolismo , Animales , Células Cultivadas , Femenino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Médula Espinal/citología
3.
J Neurosci ; 31(15): 5865-75, 2011 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-21490228

RESUMEN

CCL2 chemokine and its receptor CCR2 may contribute to neuropathic pain development. We tested the hypothesis that injury to peripheral nerves triggers CCL2 release from afferents in the dorsal horn spinal cord (DHSC), leading to pronociceptive effects, involving the production of proinflammatory factors, in particular. Consistent with the release of CCL2 from primary afferents, electron microscopy showed the CCL2 immunoreactivity in glomerular boutons and secretory vesicles in the DHSC of naive rats. Through the ex vivo superfusion of DHSC slices, we demonstrated that the rate of CCL2 secretion was much lower in neonatal capsaicin-treated rats than in controls. Thus, much of the CCL2 released in the DHSC originates from nociceptive fibers bearing TRPV1 (transient receptor potential vanilloid 1). In contrast, high levels of CCL2 released from the DHSC were observed in neuropathic pain animal model induced by chronic constriction of the sciatic nerve (SN-CCI). The upregulated expression of proinflammatory markers and extracellular signal-regulated kinase (ERK) 1/2 pathway activation (ERK1/2 phosphorylation) in the DHSC of SN-CCI animals were reversed by intrathecal administration of the CCR2 antagonist INCB3344 (N-[2-[[(3S,4S)-1-E4-(1,3-benzodioxol-5-yl)-4-hydroxycyclohexyl]-4-ethoxy-3-pyrrolidinyl]amino]-2-oxoethyl]-3-(trifluoromethyl)benzamide). These pathological pain-associated changes in the DHSC were mimicked by the intrathecal injection of exogenous CCL2 in naive rats and were prevented by the administration of INCB3344 or ERK inhibitor (PD98059). Finally, mechanical allodynia, which was fully developed 2 weeks after SN-CCI in rats, was attenuated by the intrathecal injection of INCB3344. Our data demonstrate that CCL2 has the typical characteristics of a neuronal mediator involved in nociceptive signal processing and that antagonists of its receptor are promising agents from treating neuropathic pain.


Asunto(s)
Quimiocina CCL2/metabolismo , Inflamación/patología , Neuralgia/patología , Neuronas/metabolismo , Traumatismos de los Nervios Periféricos , Médula Espinal/metabolismo , Vesículas Sinápticas/metabolismo , Animales , Animales Recién Nacidos , Western Blotting , Quimiocina CCL2/antagonistas & inhibidores , Quimiocina CCL2/farmacología , Enfermedad Crónica , Constricción Patológica , Ensayo de Inmunoadsorción Enzimática , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Técnica del Anticuerpo Fluorescente , Hiperalgesia/patología , Inmunohistoquímica , Masculino , Microscopía Electrónica , Inhibidores de Proteínas Quinasas/farmacología , Pirrolidinas/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Receptores CCR2/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/lesiones , Nervio Ciático/patología , Médula Espinal/citología
4.
Neuropsychopharmacology ; 47(2): 599-608, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34621016

RESUMEN

Opioids are a mainstay of pain management but can induce unwanted effects, including analgesic tolerance and paradoxical hyperalgesia, either of which leads to increased pain. Clinically, however, the relationship between these two phenomena remains elusive. By evaluating changes in mechanical nociceptive threshold in male rats, we found that in contrast to a purely analgesic control response to a single subcutaneous administration of fentanyl (25 µg/kg), in rats subjected to inflammatory pain 2 weeks previously (Day0), the same test dose (D13) induced a bi-phasic response: initial decreased analgesia (tolerance) followed by hyperalgesia lasting several hours. Both the tolerance and hyperalgesia were further enhanced in rats that had additionally received fentanyl on D0. The dose-response profiles (5 fg to 50 µg/kg) of pain- and opioid-experienced rats were very different from pain/drug-naive rats. At ultra-low fentanyl doses (<5 ng/kg and <500 ng/kg for naïve control and pain/drug-experienced rats, respectively), solely hyperalgesia was observed in all cases. At higher doses, which now produced analgesia alone in naive rats, reduced analgesia (tolerance) coupled with hyperalgesia occurred in pain/fentanyl-experienced rats, with both phases increasing with dose. Transcriptomic and pharmacological data revealed that an overactivation of the spinal N-methyl-D-aspartate receptor-inducible NO synthase cascade plays a critical role in both acute tolerance and hyperalgesia, and together with the finding that the magnitudes of analgesia and associated hyperalgesia are negatively correlated, is indicative of closely related phenomena. Finally, a polyamine deficient diet prevented inducible NO synthase transcript upregulation, restored fentanyl's analgesic efficacy and suppressed the emergence of hyperalgesia.


Asunto(s)
Fentanilo , Hiperalgesia , Analgésicos/farmacología , Analgésicos Opioides/farmacología , Animales , Dieta , Fentanilo/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/prevención & control , Masculino , Poliaminas/efectos adversos , Ratas , Ratas Sprague-Dawley
5.
J Neurosci ; 30(16): 5754-66, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20410127

RESUMEN

Neuropathic pain after peripheral nerve injury, associated with local neuroinflammation in the spinal cord, is a severe incapacitating condition with which clinical treatment remains challenging. Inflammatory molecules signal through various intracellular transduction pathways, activation of which may amplify and cause spreading of the inflammatory response. We showed recently that spinal nerve lesion leads to rapid activation of Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signal transduction pathway in dorsal spinal cord microglia in relation with enhanced levels of spinal interleukin-6 (IL-6) protein. Here, we selectively inactivated JAK/STAT3 signaling in rat dorsal spinal cord glia through local, lentiviral-mediated production of the suppressor of cytokine signaling SOCS3, a physiologic inhibitory protein of JAK/STAT3, and analyzed its consequences in a preclinical model of neuropathic pain. The targeted blockade of JAK/STAT3 activity prevented the abnormal expression of IL-6, CC chemokine ligand CCL2, and activating transcription factor ATF3 induced in the spinal cord by chronic constriction injury of the sciatic nerve (CCI) and substantially attenuated mechanical hypersensitivity (allodynia) in rats. In naive rats, intrathecal administration of a proalgesic cytokine IL-6 rapidly activated microglial JAK/STAT3 and induced downstream changes closely resembling CCI-evoked alterations. We identified downstream mechanisms through which JAK/STAT3 pathway activation leads to the spreading of neuroinflammation. Our findings reveal that JAK/STAT3 signaling plays a major role in spinal cord plasticity and mechanical allodynia associated with peripheral nerve injury.


Asunto(s)
Mediadores de Inflamación/antagonistas & inhibidores , Quinasas Janus/antagonistas & inhibidores , Dolor/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Neuropatía Ciática/metabolismo , Médula Espinal/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Animales , Línea Celular , Células Cultivadas , Humanos , Mediadores de Inflamación/fisiología , Quinasas Janus/fisiología , Marmota , Dolor/etiología , Dimensión del Dolor/métodos , Estimulación Física/métodos , Ratas , Factor de Transcripción STAT3/fisiología , Neuropatía Ciática/complicaciones , Transducción de Señal/fisiología , Médula Espinal/patología , Proteína 3 Supresora de la Señalización de Citocinas
6.
J Neurosci ; 28(34): 8489-501, 2008 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-18716207

RESUMEN

Responses resulting from injury to the trigeminal nerve exhibit differences compared with those caused by lesion of other peripheral nerves. With the aim of elucidating the physiopathological mechanisms underlying cephalic versus extracephalic neuropathic pain, we determined the time course expression of proinflammatory cytokines interleukin-6 (IL-6) and IL-1beta, neuronal injury (ATF3), macrophage/microglial (OX-42), and satellite cells/astrocyte (GFAP) markers in central and ganglion tissues in rats that underwent unilateral chronic constriction injury (CCI) to either infraorbital nerve (IoN) (cephalic area) or sciatic nerve (SN) (extracephalic area). Whereas CCI induced microglial activation in both models, we observed a concomitant upregulation of IL-6 and ATF3 in the ipsilateral dorsal horn of the lumbar cord in SN-CCI rats but not in the ipsilateral spinal nucleus of the trigeminal nerve (Sp5c) in IoN-CCI rats. Preemptive treatment with minocycline (daily administration of 20 mg/kg, i.p., for 2 weeks) partially prevented pain behavior and microglial activation in SN-CCI rats but was ineffective in IoN-CCI rats. We show that IL-6 can upregulate OX-42 and ATF3 expression in cultured microglia and neurons from spinal cord, respectively, as well as in the dorsal horn after acute intrathecal administration of the cytokine. We propose that IL-6 could be one of the promoters of the signaling cascade leading to abnormal pain behavior in SN-CCI but not IoN-CCI rats. Our data further support the idea that different pathophysiological mechanisms contribute to the development of cephalic versus extracephalic neuropathic pain.


Asunto(s)
Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Neuralgia/etiología , Neuralgia/metabolismo , Órbita/inervación , Nervio Ciático , Factor de Transcripción Activador 3/genética , Animales , Antígenos de Diferenciación/genética , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Constricción Patológica , Citocinas/genética , Ensayo de Inmunoadsorción Enzimática , Ganglios Sensoriales/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Hiperestesia/etiología , Hiperestesia/psicología , Inmunohistoquímica , Interleucina-6/genética , Masculino , Minociclina/farmacología , Neuroglía/metabolismo , Neuronas/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Nervio Ciático/lesiones , Factores de Tiempo , Traumatismos del Sistema Nervioso/complicaciones
7.
J Neurochem ; 107(1): 50-60, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18636982

RESUMEN

Peripheral nerve lesion leads to the production of interleukin 6 (IL-6)-related neuropoietic cytokines involved in nerve protection and regeneration. This family of cytokines mainly signal through the signal transducer and activator of transcription (STAT) pathway that is locally activated in injured nerves. IL-6 is also involved in pain that frequently arises from peripheral nerve lesion. We investigated the possible activation of this major IL-6 signaling system in the spinal cord after peripheral nerve injury and its role in neuropathic pain. Ligation of L5-L6 spinal nerves (SNL) evoked an accumulation of active, phosphorylated form of STAT3 in microglial cells of dorsal spinal cord mostly in projection areas of injured nerves. SNL resulted also in a massive induction of IL-6 mRNA expression in dorsal root ganglia and increased concentration of IL-6 in dorsal spinal cord. Intrathecal injection of anti-rat IL-6 antibodies prevented the SNL-induced accumulation of phospho-STAT3 in the spinal cord. STAT3 pathway blockade with Janus kinase 2 inhibitor AG490 attenuated both mechanical allodynia and thermal hyperalgesia in SNL rats. These data show that in response to SNL injury Janus kinase/STAT3 system is activated mainly through IL-6 signaling in spinal microglia and that this transduction pathway participates in development of pain associated with nerve alteration.


Asunto(s)
Interleucina-6/metabolismo , Janus Quinasa 1/metabolismo , Microglía/metabolismo , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Factor de Transcripción STAT3/metabolismo , Médula Espinal/fisiopatología , Animales , Anticuerpos/farmacología , Modelos Animales de Enfermedad , Activación Enzimática/fisiología , Hiperalgesia/enzimología , Hiperalgesia/fisiopatología , Interleucina-6/genética , Interleucina-6/inmunología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Ligadura , Masculino , Microglía/enzimología , Nervios Periféricos/enzimología , Nervios Periféricos/fisiopatología , Enfermedades del Sistema Nervioso Periférico/enzimología , Enfermedades del Sistema Nervioso Periférico/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Médula Espinal/enzimología , Médula Espinal/metabolismo , Nervios Espinales/lesiones , Nervios Espinales/fisiopatología , Nervios Espinales/cirugía , Regulación hacia Arriba/fisiología
8.
J Neurochem ; 106(2): 757-69, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18419759

RESUMEN

A better understanding of the mechanisms linked to chemokine pronociceptive effects is essential for the development of new strategies to better prevent and treat chronic pain. Among chemokines, MCP-1/CCL2 involvement in neuropathic pain processing is now established. However, the mechanisms by which MCP-1/CCL2 exerts its pronociceptive effects are still poorly understood. In the present study, we demonstrate that MCP-1/CCL2 can alter pain neurotransmission in healthy rats. Using immunohistochemical studies, we first show that CCL2 is constitutively expressed by primary afferent neurons and their processes in the dorsal horn of the spinal cord. We also observe that CCL2 is co-localized with pain-related peptides (SP and CGRP) and capsaicin receptor (VR1). Accordingly, using in vitro superfusion system of lumbar dorsal root ganglion and spinal cord explants of healthy rats, we show that potassium or capsaicin evoke calcium-dependent release of CCL2. In vivo, we demonstrate that intrathecal administration of CCL2 to healthy rats produces both thermal hyperalgesia and sustained mechanical allodynia (up to four consecutive days). These pronociceptive effects of CCL2 are completely prevented by the selective CCR2 antagonist (INCB3344), indicating that CCL2-induced pain facilitation is elicited via direct spinal activation of CCR2 receptor. Therefore, preventing the activation of CCR2 might provide a fruitful strategy for treating pain.


Asunto(s)
Quimiocina CCL2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hiperalgesia/fisiopatología , Neuronas Aferentes/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Pirrolidinas/farmacología , Receptores CCR2/antagonistas & inhibidores , Médula Espinal/citología , Análisis de Varianza , Animales , Conducta Animal , Péptido Relacionado con Gen de Calcitonina/genética , Péptido Relacionado con Gen de Calcitonina/metabolismo , Calcio/metabolismo , Quimiocina CCL2/genética , Ensayo de Inmunoadsorción Enzimática/métodos , Ganglios Espinales/citología , Masculino , Cloruro de Potasio/farmacología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Sustancia P/genética , Sustancia P/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
9.
J Neurosci Methods ; 167(2): 148-59, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-17949823

RESUMEN

Activated glial cells in the dorsal spinal cord take an important part in the development of pain after peripheral nerve injury. Our understanding of mechanisms involved in functional changes of spinal glia remains incomplete. Excepting drugs that completely disrupt glial function, pharmacological studies fail to target glia and to modify locally its function in order to really discriminate the role of neuronal versus glial cells in chronic pain. We developed an intraspinal gene transfer approach using pseudotyped lentiviral-derived vector targeting highly preferentially glial cells. Single microinjection of vector expressing EGFP under a CMV promoter control (LV-EGFP) allowed vector diffusion along a rostro-caudal axis but strictly restricted to the grey matter of the ipsilateral dorsal spinal cord. EGFP transgene was mainly expressed in astrocytes and microglial cells whereas less than 9% of cells containing EGFP were neurons. Notably, LV-EGFP administration and EGFP overexpression in glial cells did neither modify glial activity, nor alter animal's nociceptive or locomotor behaviors. Targeted modulation of the expression of gene of interest in glial cells, closely restricted to a particular region of the spinal cord, may thus represent an interesting approach to refine the understanding of mechanisms by which spinal glial cells participate in pain processing.


Asunto(s)
Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Hiperalgesia/terapia , Lentivirus/fisiología , Neuroglía/metabolismo , Médula Espinal/citología , Animales , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inyecciones Espinales/métodos , Actividad Motora/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Umbral del Dolor/fisiología , Ratas
10.
Mol Ther ; 15(4): 687-697, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28192702

RESUMEN

Neuropathic pain developing after peripheral nerve injury is associated with altered neuronal and glial cell functions in the spinal cord. Activated glia produces algogenic mediators, exacerbating pain. Among the different intracellular pathways possibly involved in the modified glial function, the nuclear factor κB (NF-κB) system is of particular interest, as numerous genes encoding inflammation- and pain-related molecules are controlled by this transcription factor. NF-κB is a pleiotropic factor also involved in central nervous system homeostasy. To study its role in chronic pain, it is thus essential to inhibit the NF-κB pathway selectively in activated spinal glial cells. Here, we show that when restricted to spinal cord and targeted to glial cells, lentiviral vector-mediated delivery of NF-κB super- repressor IκBα resulted in an inhibition of the NF-κB pathway activated in the rat spinal cord after sciatic nerve injury (chronic constriction injury, CCI). Concomitantly, IκBα overproduction prevented the enhanced expression of interleukin-6 and of inducible nitric oxide synthase associated with chronic constriction injury and resulted in prolonged antihyperalgesic and antiallodynic effects. These data show that targeted blockade of NF-κB activity in spinal glia efficiently alleviates pain behavior in CCI rats, demonstrating the active participation of the glial NF-κB pathway in the development of neuropathic pain after peripheral nerve injury.

11.
Physiol Behav ; 194: 497-504, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29928887

RESUMEN

We explored the molecular and behavioral effects of a perineural Lipopolysaccharide (LPS)-mediated inflammatory priming on the development and maintenance of painful post-traumatic trigeminal neuropathy (PPTTN) following infra-orbital nerve chronic constriction injury (CCI-IoN) in rats. Rats were pretreated with repetitive perineural injections in the vicinity of the IoN of either LPS or vehicle (Vhcl) before being submitted to CCI-IoN. Orofacial pain-like behaviors (response to Von Frey Filament testing and spontaneous isolated face grooming) were measured during the period of LPS injections (three weeks) and following CCI-IoN surgery (two weeks). Local LPS administration induced an early pain-like behavior (i.e. an increase in spontaneous pain [SP] or mechanical static allodynia [MSA]) in both conditions, and following CCI-IoN, MSA and SP developed earlier and more severely in LPS-pretreated rats than in the control group. Ipsilateral increases of key neuropathic pain mRNA markers in the IoN parenchyma, trigeminal ganglia (TG) and spinal trigeminal nucleus caudalis (Sp5C) were observed in CCI-IoN injured animals as compared to controls. Although no significant molecular differences could be observed within the IoN parenchyma between LPS and Vhcl-pretreated animals, a significant increase of key inflammatory cytokine Interleukin 1 beta (IL - 1ß) could be found in the TG of LPS-pretreated CCI-injured animals versus controls. Finally, a higher increase of inducible nitric oxide synthase (iNOS) in ipsilateral Sp5C of LPS-pretreated animals was observed as compared to Sp5C of Vhcl-pretreated animals. These results suggest a key role of inflammatory priming in the development and maintenance of PPTTN implicating IL-1ß/iNOS-dependent central sensitization mechanisms.


Asunto(s)
Inflamación/fisiopatología , Lipopolisacáridos/farmacología , Nervio Maxilar/fisiopatología , Neuralgia/fisiopatología , Traumatismos del Nervio Trigémino/fisiopatología , Animales , Hiperalgesia/complicaciones , Hiperalgesia/fisiopatología , Inflamación/inducido químicamente , Inflamación/complicaciones , Interleucina-1beta/metabolismo , Masculino , Nervio Maxilar/metabolismo , Neuralgia/complicaciones , Neuralgia/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Dimensión del Dolor , Traumatismos de los Nervios Periféricos/complicaciones , Traumatismos de los Nervios Periféricos/fisiopatología , Ratas , Ganglio del Trigémino/metabolismo , Traumatismos del Nervio Trigémino/complicaciones , Traumatismos del Nervio Trigémino/metabolismo , Núcleo Espinal del Trigémino/metabolismo
12.
PLoS Negl Trop Dis ; 11(11): e0006058, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29149212

RESUMEN

BACKGROUND: Mycolactone is a macrolide produced by the skin pathogen Mycobacterium ulcerans, with cytotoxic, analgesic and immunomodulatory properties. The latter were recently shown to result from mycolactone blocking the Sec61-dependent production of pro-inflammatory mediators by immune cells. Here we investigated whether mycolactone similarly affects the inflammatory responses of the nervous cell subsets involved in pain perception, transmission and maintenance. We also investigated the effects of mycolactone on the neuroinflammation that is associated with chronic pain in vivo. METHODOLOGY/ PRINCIPLE FINDINGS: Sensory neurons, Schwann cells and microglia were isolated from mice for ex vivo assessment of mycolactone cytotoxicity and immunomodulatory activity by measuring the production of proalgesic cytokines and chemokines. In all cell types studied, prolonged (>48h) exposure to mycolactone induced significant cell death at concentrations >10 ng/ml. Within the first 24h treatment, nanomolar concentrations of mycolactone efficiently suppressed the cell production of pro-inflammatory mediators, without affecting their viability. Notably, mycolactone also prevented the pro-inflammatory polarization of cortical microglia. Since these cells critically contribute to neuroinflammation, we next tested if mycolactone impacts this pathogenic process in vivo. We used a rat model of neuropathic pain induced by chronic constriction of the sciatic nerve. Here, mycolactone was injected daily for 3 days in the spinal canal, to ensure its proper delivery to spinal cord. While this treatment failed to prevent injury-induced neuroinflammation, it decreased significantly the local production of inflammatory cytokines without inducing detectable cytotoxicity. CONCLUSION/ SIGNIFICANCE: The present study provides in vitro and in vivo evidence that mycolactone suppresses the inflammatory responses of sensory neurons, Schwann cells and microglia, without affecting the cell viability. Together with previous studies using peripheral blood leukocytes, our work implies that mycolactone-mediated analgesia may, at least partially, be explained by its anti-inflammatory properties.


Asunto(s)
Antiinflamatorios/metabolismo , Macrólidos/metabolismo , Mycobacterium ulcerans/metabolismo , Sistema Nervioso/efectos de los fármacos , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Neuralgia/fisiopatología , Ratas
13.
Pain ; 157(4): 827-839, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26655733

RESUMEN

Changes in the nerve's microenvironment and local inflammation resulting from peripheral nerve injury participate in nerve sensitization and neuropathic pain development. Taking part in these early changes, disruption of the blood-nerve barrier (BNB) allows for infiltration of immunocytes and promotes the neuroinflammation. However, molecular mechanisms engaged in vascular endothelial cells (VEC) dysfunction and BNB alterations remain unclear. In vivo, BNB permeability was assessed following chronic constriction injury (CCI) of the rat sciatic nerve (ScN) and differential expression of markers of VEC functional state, inflammation, and intracellular signaling was followed from 3 hours to 2 months postinjury. Several mechanisms potentially involved in functional alterations of VEC were evaluated in vitro using human VEC (hCMEC/D3), then confronted to in vivo physiopathological conditions. CCI of the ScN led to a rapid disruption of endoneurial vascular barrier that was correlated to a decreased production of endothelial tight-junction proteins and an early and sustained alteration of Hedgehog (Hh) signaling pathway. In vitro, activation of Toll-like receptor 4 in VEC downregulated the components of Hh pathway and altered the endothelial functional state. Inhibition of Hh signaling in the ScN of naive rats mimicked the biochemical and functional alterations observed after CCI and was, on its own, sufficient to evoke local neuroinflammation and sustained mechanical allodynia. Alteration of the Hh signaling pathway in VEC associated with peripheral nerve injury, is involved in BNB disruption and local inflammation, and could thus participate in the early changes leading to the peripheral nerve sensitization and, ultimately, neuropathic pain development.


Asunto(s)
Barrera Hematonerviosa/metabolismo , Células Endoteliales/metabolismo , Neuralgia/fisiopatología , Traumatismos de los Nervios Periféricos/metabolismo , Nervio Ciático/fisiopatología , Transducción de Señal , Animales , Proteínas Hedgehog/metabolismo , Inflamación/metabolismo , Masculino , Neuralgia/metabolismo , Ratas Sprague-Dawley , Neuropatía Ciática/fisiopatología , Receptor Toll-Like 4/metabolismo
14.
Pain ; 110(1-2): 22-32, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15275748

RESUMEN

Although clinically well controlled at the metabolic level, type I diabetes resulting from an insufficient insulin secretion remains the cause of severe complications. In particular, diabetes can be associated with neuropathic pain which fails to be treated by classical analgesics. In this study, we investigated the efficacy of a novel non opioid analgesic, cizolirtine, to reduce mechanical hyperalgesia associated with streptozotocin (STZ)-induced diabetes, in the rat. Cizolirtine was compared to paroxetine, an antidepressant drug with proven efficacy to relieve painful diabetic neuropathy. Under acute conditions, cizolirtine (30 and 80 mg/kgi.p.) significantly increased paw withdrawal and vocalization thresholds in the paw pressure test in diabetic rats displaying mechanical hyperalgesia. The antihyperalgesic effects of cizolirtine persisted under chronic treatment conditions, since pre-diabetes thresholds were recovered after a two week-treatment with the drug (3 mg/kg/day, s.c.). In this respect, cizolirtine was as efficient as paroxetine (5 mg/kg per day, s.c.) which, however, was inactive under acute treatment conditions. Measurements of the spinal release of calcitonin gene-related peptide (CGRP) through intrathecal perfusion under halothane-anesthesia showed that acute administration of cizolirtine (80 mg/kg, i.p.) significantly diminished (-36%) the peptide outflow in diabetic rats suffering from neuropathic pain. This effect as well as the antihyperalgesic effect of cizolirtine were prevented by the alpha(2)-adrenoreceptor antagonist idazoxan (2 mg/kg, i.p.). These data suggest that the antihyperalgesic effect of cizolirtine in diabetic rats suffering from neuropathic pain implies an alpha(2)-adrenoceptor-dependent presynaptic inhibition of CGRP-containing primary afferent fibers.


Asunto(s)
Analgésicos no Narcóticos/uso terapéutico , Péptido Relacionado con Gen de Calcitonina/metabolismo , Diabetes Mellitus Experimental/complicaciones , Hiperalgesia/tratamiento farmacológico , Pirazoles/uso terapéutico , Médula Espinal/efectos de los fármacos , Análisis de Varianza , Animales , Conducta Animal , Peso Corporal , Diabetes Mellitus Experimental/metabolismo , Relación Dosis-Respuesta a Droga , Ingestión de Líquidos/efectos de los fármacos , Interacciones Farmacológicas , Índice Glucémico/efectos de los fármacos , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Idazoxan/farmacología , Bombas de Infusión Implantables , Masculino , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Umbral del Dolor/efectos de los fármacos , Paroxetina/farmacología , Radioinmunoensayo/métodos , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Estreptozocina/farmacología , Factores de Tiempo
15.
Eur J Pharmacol ; 441(1-2): 47-55, 2002 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-12007919

RESUMEN

Although it is well established that adenosine exerts antinociceptive effects at the spinal level in various species including human, the mechanisms responsible for such effects are still a matter of debate. We presently investigated whether adenosine-induced antinociception might possibly be related to an inhibitory influence of this neuromodulator on the spinal release of neuropeptides implicated in the transfer and/or control of nociceptive signals. For this purpose, the K(+)-evoked overflow of substance P-, calcitonin gene-related peptide (CGRP)- and cholecystokinin-like materials was measured from slices of the dorsal half of the rat lumbar enlargement superfused with an artificial cerebrospinal fluid supplemented with increasing concentrations of various adenosine receptor ligands. The data showed that stimulation of adenosine A(1) and (possibly) A(3) receptors, but not A(2A) receptors, exerted an inhibitory influence on the spinal release of CGRP-like material. In contrast, none of the adenosine A(1), A(2A) and A(3) receptor agonists tested within relevant ranges of concentrations significantly affected the release of substance P- and cholecystokinin-like materials. These results support the idea that adenosine-induced antinociception at the spinal level might possibly be caused, at least partly, by the stimulation of inhibitory adenosine A(1) receptors located presynaptically on primary afferent fibres containing CGRP but not substance P.


Asunto(s)
Adenosina/análogos & derivados , Neuropéptidos/metabolismo , Receptores Purinérgicos P1/fisiología , Médula Espinal/metabolismo , Adenosina/farmacología , Adenosina-5'-(N-etilcarboxamida)/farmacología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colecistoquinina/metabolismo , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Masculino , Dolor/metabolismo , Fenetilaminas/farmacología , Potasio/farmacología , Agonistas del Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Ratas , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Sustancia P/metabolismo , Xantinas/farmacología
16.
Pain ; 150(2): 358-368, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20573451

RESUMEN

Chronic stressful events induce biochemical, physiological and psychological changes, resulting in stress-related neuropsychiatric disorders, such as anxiety or depression. Using repeated social defeat as a stressful event model, we show that this preclinical paradigm induces a transient increase in the expression of the genes encoding the pro-inflammatory molecules iNOS and COX-2. We provide the first demonstration that chronic stress affects spinal plasticity through a mechanism involving local neuroinflammation. The functional consequences of such neuroinflammation are associated with a transient decrease in the mechanical nociceptive threshold. Administration of the cholecystokinin(CCK)-2 receptor antagonist, CI-988, directly into the Rostral Ventromedial Medulla reverses the chronic stress-induced decrease in the nociceptive threshold. These data strongly suggest that chronic stress induces a spinal neuroinflammation associated with transient sensory hypersensitivity involving the activation of CCK-dependent nociceptive descending facilitatory pathways. Pharmacological data show that chronic social stress-induced long-lasting state of anxiety is not responsible for maintaining the spinal neuroinflammation and, therefore, for the associated sensory hypersensitivity. Conversely, an evaluation of pain-related behavior in the formalin model indicates that anxiety is directly related to prolonged hyperalgesia prevented by systemic benzodiazepine or CCK-2 receptor antagonist treatments. The present study highlights the adverse effects of chronic stress on spinal neuroinflammation triggering sensory hypersensitivity. Exploration of this phenomenon points out the divergence between pain sensitivity and anxiety-induced hyperalgesia, which is in agreement with clinical observations. Altogether, these data open up new perspectives for clinical research devoted to the evaluation and treatment of pain in anxio-depressive patients.


Asunto(s)
Ansiedad/metabolismo , Hiperalgesia/metabolismo , Médula Espinal/metabolismo , Estrés Psicológico/complicaciones , Estrés Psicológico/metabolismo , Análisis de Varianza , Animales , Ansiedad/complicaciones , Ansiedad/fisiopatología , Colecistoquinina/metabolismo , Dominación-Subordinación , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Indoles/farmacología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/fisiopatología , Masculino , Meglumina/análogos & derivados , Meglumina/farmacología , Dimensión del Dolor/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Receptores de Colecistoquinina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Médula Espinal/fisiopatología , Estrés Psicológico/fisiopatología
17.
PLoS One ; 4(9): e6874, 2009 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-19727441

RESUMEN

BACKGROUND: The functional significance of proenkephalin systems in processing pain remains an open question and indeed is puzzling. For example, a noxious mechanical stimulus does not alter the release of Met-enkephalin-like material (MELM) from segments of the spinal cord related to the stimulated area of the body, but does increase its release from other segments. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that, in the rat, a noxious mechanical stimulus applied to either the right or the left hind paw elicits a marked increase of MELM release during perifusion of either the whole spinal cord or the cervico-trigeminal area. However, these stimulatory effects were not additive and indeed, disappeared completely when the right and left paws were stimulated simultaneously. CONCLUSION/SIGNIFICANCE: We have concluded that in addition to the concept of a diffuse control of the transmission of nociceptive signals through the dorsal horn, there is a diffuse control of the modulation of this transmission. The "freezing" of Met-enkephalinergic functions represents a potential source of central sensitization in the spinal cord, notably in clinical situations involving multiple painful foci, e.g. cancer with metastases, poly-traumatism or rheumatoid arthritis.


Asunto(s)
Encefalina Metionina/metabolismo , Dolor/fisiopatología , Núcleo Caudal del Trigémino/metabolismo , Animales , Encefalinas/metabolismo , Extremidades , Masculino , Inhibición Neural , Precursores de Proteínas/metabolismo , Conejos , Ratas , Ratas Sprague-Dawley , Médula Espinal/fisiopatología , Núcleo Espinal del Trigémino/metabolismo
18.
Mol Ther ; 15(4): 687-97, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17299402

RESUMEN

Neuropathic pain developing after peripheral nerve injury is associated with altered neuronal and glial cell functions in the spinal cord. Activated glia produces algogenic mediators, exacerbating pain. Among the different intracellular pathways possibly involved in the modified glial function, the nuclear factor kappaB (NF-kappaB) system is of particular interest, as numerous genes encoding inflammation- and pain-related molecules are controlled by this transcription factor. NF-kappaB is a pleiotropic factor also involved in central nervous system homeostasy. To study its role in chronic pain, it is thus essential to inhibit the NF-kappaB pathway selectively in activated spinal glial cells. Here, we show that when restricted to spinal cord and targeted to glial cells, lentiviral vector-mediated delivery of NF-kappaB super- repressor IkappaBalpha resulted in an inhibition of the NF-kappaB pathway activated in the rat spinal cord after sciatic nerve injury (chronic constriction injury, CCI). Concomitantly, IkappaBalpha overproduction prevented the enhanced expression of interleukin-6 and of inducible nitric oxide synthase associated with chronic constriction injury and resulted in prolonged antihyperalgesic and antiallodynic effects. These data show that targeted blockade of NF-kappaB activity in spinal glia efficiently alleviates pain behavior in CCI rats, demonstrating the active participation of the glial NF-kappaB pathway in the development of neuropathic pain after peripheral nerve injury.


Asunto(s)
Lentivirus/genética , FN-kappa B/antagonistas & inhibidores , Neuralgia/terapia , Nervio Ciático/lesiones , Médula Espinal/fisiopatología , Animales , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos , Hiperalgesia/terapia , Proteínas I-kappa B/genética , Proteínas I-kappa B/fisiología , Interleucina-6/genética , Inhibidor NF-kappaB alfa , FN-kappa B/fisiología , Neuralgia/etiología , Neuralgia/fisiopatología , Neuroglía/fisiología , Óxido Nítrico Sintasa de Tipo II/genética , Ratas , Nervio Ciático/fisiopatología
19.
Mol Ther ; 11(4): 608-16, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15771963

RESUMEN

Trigeminal neuropathic pain represents a real challenge to therapy because commonly used drugs are devoid of real beneficial effect or patients frequently become intolerant or refractory to some of these compounds. In a rat model of trigeminal neuropathic pain, which shares numerous similarities with human trigeminal neuralgia and trigeminal neuropathic pain, we used a genomic herpes simplex virus-derived vector (HSVLatEnk) to examine the possible effect of a local overproduction of proenkephalin A (PA) targeted to the trigeminal primary sensory neurons. Unilateral peripheral inoculation of recombinant vectors on the vibrissal pad territory resulted in an about ninefold increase in proenkephalin A mRNA levels in trigeminal ganglion ipsilateral to the infected side. Transgene-derived met-enkephalin accumulated in numerous nerve cell bodies of trigeminal ganglion and was transported through the sensory nerve fibers located in the infraorbital nerve. Bilateral mechanical hyperresponsiveness, which developed 2 weeks after chronic constrictive injury of the left infraorbital nerve, was significantly attenuated in animals overproducing PA in the trigeminal ganglion ipsilateral to the lesioned infraorbital nerve. This antiallodynic effect was reversed by both the opioid receptor antagonist naloxone and the peripherally acting antagonist naloxone methiodide. Our data demonstrate that the local overproduction of PA-derived peptides in trigeminal ganglion sensory neurons evoked a potent antiallodynic effect through the stimulation of mainly peripherally located opioid receptors and suggest that targeted delivery of endogenous opioids may be of interest for the treatment of some severe forms of neuropathic pain.


Asunto(s)
Encefalinas/genética , Terapia Genética , Precursores de Proteínas/genética , Simplexvirus/genética , Ganglio del Trigémino/metabolismo , Neuralgia del Trigémino/terapia , Animales , Modelos Animales de Enfermedad , Encefalinas/análisis , Masculino , Naloxona/análogos & derivados , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/química , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Umbral del Dolor/efectos de los fármacos , Precursores de Proteínas/análisis , Compuestos de Amonio Cuaternario , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Activación Transcripcional/efectos de los fármacos , Ganglio del Trigémino/citología
20.
J Neurochem ; 81(3): 659-62, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-12065675

RESUMEN

Intrathecal infusion of the neuropeptide FF analogue, [D-Tyr1, (NMe)Phe3]neuropeptide FF (1DMe; 0.1 microm-0.1 mm) in anaesthetized rats produced a concentration-dependent decrease in the spinal outflow of dynorphin A (1-8)-like material, which persisted for at least 90 min after treatment with 10 microm-0.1 mm of the compound. Co-administration of d-Phe-Cys-Tyr-D-Trp-Orn-Thr-Pen-Thr-NH2 (CTOP; 1 microm) to block spinal micro-opioid receptors did not modify this effect, whereas naltrindole (10 microm) totally prevented it and nor-binaltorphimine (10 microm) reduced the post-effect. These data suggest that 1DMe triggers the release of endogenous opioids that stimulate mainly delta-opioid receptors, and secondarily kappa-opioid receptors, thereby exerting a negative influence on dynorphin A (1-8)-like material outflow. Because dynorphin has pronociceptive properties, such a decrease in spinal dynorphin A (1-8)-like material release might underlie the long-lasting antinociceptive effects of intrathecally administered neuropeptide FF and analogues.


Asunto(s)
Dinorfinas/líquido cefalorraquídeo , Naltrexona/análogos & derivados , Oligopéptidos/administración & dosificación , Fragmentos de Péptidos/líquido cefalorraquídeo , Somatostatina/análogos & derivados , Médula Espinal/efectos de los fármacos , Animales , Relación Dosis-Respuesta a Droga , Dinorfinas/metabolismo , Inyecciones Espinales , Masculino , Modelos Animales , Naltrexona/administración & dosificación , Antagonistas de Narcóticos/administración & dosificación , Fragmentos de Péptidos/metabolismo , Perfusión , Radioinmunoensayo , Ratas , Ratas Sprague-Dawley , Receptores Opioides delta/efectos de los fármacos , Receptores Opioides kappa/efectos de los fármacos , Receptores Opioides mu/antagonistas & inhibidores , Somatostatina/administración & dosificación , Médula Espinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA