Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Mol Pain ; 13: 1744806917727625, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28814148

RESUMO

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.


Assuntos
Barreira Hematoneural/metabolismo , Células Endoteliais/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Doença Crônica , Constrição Patológica , Proteínas Hedgehog/metabolismo , Masculino , Neuralgia/metabolismo , Traumatismos dos Nervos Periféricos/metabolismo , Ratos Sprague-Dawley , Receptor 4 Toll-Like/metabolismo , beta Catenina/metabolismo
2.
J Neurochem ; 136(1): 133-47, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26440453

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Janus Quinases/metabolismo , Microglia/metabolismo , Neurônios/metabolismo , Fator de Transcrição STAT3/metabolismo , Medula Espinal/metabolismo , Animais , Células Cultivadas , Feminino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Medula Espinal/citologia
3.
Brain Behav Immun ; 38: 38-52, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24263070

RESUMO

Functional interactions between the chemokine receptor CXCR4 and opioid receptors have been reported in the brain, leading to a decreased morphine analgesic activity. However the cellular mechanisms responsible for this loss of opioid analgesia are largely unknown. Here we examined whether Src family-kinases (SFK)-linked mechanisms induced by CXCR4 contributed to the loss of acute morphine analgesia and could represent a new physiological anti-opioid signaling pathway. In this way, we showed by immunohistochemistry and western blot that CXCL12 rapidly activated SFK phosphorylation in vitro in primary cultured lumbar rat dorsal root ganglia (DRG) but also in vivo in the DRG and the spinal cord. We showed that SFK activation occurred in a sub population of sensory neurons, in spinal microglia but also in spinal nerve terminals expressing mu-(MOR) and delta-opioid (DOR) receptor. In addition we described that CXCR4 is detected in MOR- and DOR-immunoreactive neurons in the DRG and spinal cord. In vivo, we demonstrated that an intrathecal administration of CXCL12 (1µg) significantly attenuated the subcutaneous morphine (4mg/kg) analgesia. Conversely, pretreatment with a potent CXCR4 antagonist (5µg) significantly enhanced morphine analgesia. Similar effects were obtained after an intrathecal injection of a specific SFK inhibitor, PP2 (10µg). Furthermore, PP2 abrogated CXCL12-induced decrease in morphine analgesia by suppressing SFK activation in the spinal cord. In conclusion, our data highlight that CXCL12-induced loss of acute morphine analgesia is linked to Src family kinases activation.


Assuntos
Analgésicos Opioides/farmacologia , Quimiocina CXCL12/farmacologia , Gânglios Espinais/enzimologia , Morfina/farmacologia , Receptores CXCR4/metabolismo , Quinases da Família src/metabolismo , Animais , Tolerância a Medicamentos , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Masculino , Microglia/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley
4.
J Neurosci ; 31(15): 5865-75, 2011 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-21490228

RESUMO

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.


Assuntos
Quimiocina CCL2/metabolismo , Inflamação/patologia , Neuralgia/patologia , Neurônios/metabolismo , Traumatismos dos Nervos Periféricos , Medula Espinal/metabolismo , Vesículas Sinápticas/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/farmacologia , Doença Crônica , Constrição Patológica , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Imunofluorescência , Hiperalgesia/patologia , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Inibidores de Proteínas Quinases/farmacologia , Pirrolidinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores CCR2/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nervo Isquiático/lesões , Nervo Isquiático/patologia , Medula Espinal/citologia
5.
Eur J Neurosci ; 36(5): 2619-31, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22694179

RESUMO

Initial studies implicated the chemokine CXC motif ligand 12 (CXCL12) and its cognate CXC motif receptor 4 (CXCR4) in pain modulation. However, there has been no description of the distribution, transport and axonal sorting of CXCL12 and CXCR4 in rat nociceptive structures, and their direct participation in nociception modulation has not been demonstrated. Here, we report that acute intrathecal administration of CXCL12 induced mechanical hypersensitivity in naive rats. This effect was prevented by a CXCR4-neutralizing antibody. To determine the morphological basis of this behavioural response, we used light and electron microscopic immunohistochemistry to map CXCL12- and CXCR4-immunoreactive elements in dorsal root ganglia, lumbar spinal cord, sciatic nerve and skin. Light microscopy analysis revealed CXCL12 and CXCR4 immunoreactivity in calcitonin gene related peptide-containing peptidergic primary sensory neurons, which were both conveyed to central and peripheral sensory nerve terminals. Electron microscopy clearly demonstrated CXCL12 and CXCR4 immunoreactivity in primary sensory nerve terminals in the dorsal horn; both were sorted into small clear vesicles and large dense-core vesicles. This suggests that CXCL12 and CXCR4 are trafficked from nerve cell bodies to the dorsal horn. Double immunogold labelling for CXCL12 and calcitonin gene related peptide revealed partial vesicular colocalization in axonal terminals. We report, for the first time, that CXCR4 receptors are mainly located on the neuronal plasma membrane, where they are present at pre-synaptic and post-synaptic sites of central terminals. Receptor inactivation experiments, behavioural studies and morphological analyses provide strong evidence that the CXCL12/CXCR4 system is involved in modulation of nociceptive signalling.


Assuntos
Quimiocina CXCL12/análise , Nociceptores/química , Receptores CXCR4/análise , Animais , Masculino , Nociceptores/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/química , Medula Espinal/química
6.
J Neuroinflammation ; 9: 36, 2012 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-22353423

RESUMO

BACKGROUND: Sustained neuroinflammation strongly contributes to the pathogenesis of pain. The clinical challenge of chronic pain relief led to the identification of molecules such as cytokines, chemokines and more recently matrix metalloproteinases (MMPs) as putative therapeutic targets. Evidence points to a founder member of the matricial CCN family, NOV/CCN3, as a modulator of these inflammatory mediators. We thus investigated the possible involvement of NOV in a preclinical model of persistent inflammatory pain. METHODS: We used the complete Freund's adjuvant (CFA)-induced model of persistent inflammatory pain and cultured primary sensory neurons for in vitro experiments. The mRNA expression of NOV and pro-inflammatory factors were measured with real-time quantitative PCR, CCL2 protein expression was assessed using ELISA, MMP-2 and -9 activities using zymography. The effect of drugs on tactile allodynia was evaluated by the von Frey test. RESULTS: NOV was expressed in neurons of both dorsal root ganglia (DRG) and dorsal horn of the spinal cord (DHSC). After intraplantar CFA injection, NOV levels were transiently and persistently down-regulated in the DRG and DHSC, respectively, occurring at the maintenance phase of pain (15 days). NOV-reduced expression was restored after treatment of CFA rats with dexamethasone. In vitro, results based on cultured DRG neurons showed that siRNA-mediated inhibition of NOV enhanced IL-1ß- and TNF-α-induced MMP-2, MMP-9 and CCL2 expression whereas NOV addition inhibited TNF-α-induced MMP-9 expression through ß1 integrin engagement. In vivo, the intrathecal delivery of MMP-9 inhibitor attenuated mechanical allodynia of CFA rats. Importantly, intrathecal administration of NOV siRNA specifically led to an up-regulation of MMP-9 in the DRG and MMP-2 in the DHSC concomitant with increased mechanical allodynia. Finally, NOV intrathecal treatment specifically abolished the induction of MMP-9 in the DRG and, MMP-9 and MMP-2 in the DHSC of CFA rats. This inhibitory effect on MMP is associated with reduced mechanical allodynia. CONCLUSIONS: This study identifies NOV as a new actor against inflammatory pain through regulation of MMPs thus uncovering NOV as an attractive candidate for therapeutic improvement in pain relief.


Assuntos
Proteínas Imediatamente Precoces/metabolismo , Inflamação/complicações , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Dor/etiologia , Dor/metabolismo , Análise de Variância , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células Cultivadas , Quimiocina CCL2/metabolismo , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Adjuvante de Freund , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Proteínas Imediatamente Precoces/genética , Inflamação/induzido quimicamente , Peptídeos e Proteínas de Sinalização Intercelular/genética , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Dor/tratamento farmacológico , Medição da Dor , Limiar da Dor/efeitos dos fármacos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Sprague-Dawley , Células Receptoras Sensoriais/efeitos dos fármacos , Medula Espinal/patologia , Fatores de Tempo , Transfecção , Regulação para Cima/efeitos dos fármacos
7.
Comput Methods Programs Biomed ; 222: 106908, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35716534

RESUMO

BACKGROUND AND OBJECTIVE: During lung cancer radiotherapy, the position of infrared reflective objects on the chest can be recorded to estimate the tumor location. However, radiotherapy systems have a latency inherent to robot control limitations that impedes the radiation delivery precision. Prediction with online learning of recurrent neural networks (RNN) allows for adaptation to non-stationary respiratory signals, but classical methods such as real-time recurrent learning (RTRL) and truncated backpropagation through time are respectively slow and biased. This study investigates the capabilities of unbiased online recurrent optimization (UORO) to forecast respiratory motion and enhance safety in lung radiotherapy. METHODS: We used nine observation records of the three-dimensional (3D) position of three external markers on the chest and abdomen of healthy individuals breathing during intervals from 73s to 222s. The sampling frequency was 10Hz, and the amplitudes of the recorded trajectories range from 6mm to 40mm in the superior-inferior direction. We forecast the 3D location of each marker simultaneously with a horizon value (the time interval in advance for which the prediction is made) between 0.1s and 2.0s, using an RNN trained with UORO. We compare its performance with an RNN trained with RTRL, least mean squares (LMS), and offline linear regression. We provide closed-form expressions for quantities involved in the gradient loss calculation in UORO, thereby making its implementation efficient. Training and cross-validation were performed during the first minute of each sequence. RESULTS: On average over the horizon values considered and the nine sequences, UORO achieves the lowest root-mean-square (RMS) error and maximum error among the compared algorithms. These errors are respectively equal to 1.3mm and 8.8mm, and the prediction time per time step was lower than 2.8ms (Dell Intel core i9-9900K 3.60 GHz). Linear regression has the lowest RMS error for the horizon values 0.1s and 0.2s, followed by LMS for horizon values between 0.3s and 0.5s, and UORO for horizon values greater than 0.6s. CONCLUSIONS: UORO can accurately predict the 3D position of external markers for intermediate to high response times with an acceptable time performance. This will help limit unwanted damage to healthy tissues caused by radiotherapy.


Assuntos
Neoplasias Pulmonares , Redes Neurais de Computação , Algoritmos , Humanos , Pulmão , Neoplasias Pulmonares/radioterapia , Movimento (Física) , Respiração
8.
Neuropsychopharmacology ; 47(2): 599-608, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34621016

RESUMO

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.


Assuntos
Fentanila , Hiperalgesia , Analgésicos/farmacologia , Analgésicos Opioides/farmacologia , Animais , Dieta , Fentanila/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/prevenção & controle , Masculino , Poliaminas/efeitos adversos , Ratos , Ratos Sprague-Dawley
9.
J Neurosci ; 30(16): 5754-66, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20410127

RESUMO

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.


Assuntos
Mediadores da Inflamação/antagonistas & inibidores , Janus Quinases/antagonistas & inibidores , Dor/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Neuropatia Ciática/metabolismo , Medula Espinal/metabolismo , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Humanos , Mediadores da Inflamação/fisiologia , Janus Quinases/fisiologia , Marmota , Dor/etiologia , Medição da Dor/métodos , Estimulação Física/métodos , Ratos , Fator de Transcrição STAT3/fisiologia , Neuropatia Ciática/complicações , Transdução de Sinais/fisiologia , Medula Espinal/patologia , Proteína 3 Supressora da Sinalização de Citocinas
10.
Comput Med Imaging Graph ; 91: 101941, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34265553

RESUMO

During the radiotherapy treatment of patients with lung cancer, the radiation delivered to healthy tissue around the tumor needs to be minimized, which is difficult because of respiratory motion and the latency of linear accelerator (LINAC) systems. In the proposed study, we first use the Lucas-Kanade pyramidal optical flow algorithm to perform deformable image registration (DIR) of chest computed tomography (CT) scan images of four patients with lung cancer. We then track three internal points close to the lung tumor based on the previously computed deformation field and predict their position with a recurrent neural network (RNN) trained using real-time recurrent learning (RTRL) and gradient clipping. The breathing data is quite regular, sampled at approximately 2.5 Hz, and includes artificially added drift in the spine direction. The amplitude of the motion of the tracked points ranged from 12.0 mm to 22.7 mm. Finally, we propose a simple method for recovering and predicting three-dimensional (3D) tumor images from the tracked points and the initial tumor image, based on a linear correspondence model and the Nadaraya-Watson non-linear regression. The root-mean-square (RMS) error, maximum error and jitter corresponding to the RNN prediction on the test set were smaller than the same performance measures obtained with linear prediction and least mean squares (LMS). In particular, the maximum prediction error associated with the RNN, equal to 1.51 mm, is respectively 16.1% and 5.0% lower than the error given by a linear predictor and LMS. The average prediction time per time step with RTRL is equal to 119 ms, which is less than the 400 ms marker position sampling time. The tumor position in the predicted images appears visually correct, which is confirmed by the high mean cross-correlation between the original and predicted images, equal to 0.955. The standard deviation of the Gaussian kernel and the number of layers in the optical flow algorithm were the parameters having the most significant impact on registration performance. Their optimization led respectively to a 31.3% and 36.2% decrease in the registration error. Using only a single layer proved to be detrimental to the registration quality because tissue motion in the lower part of the lung has a high amplitude relative to the resolution of the CT scan images. The random initialization of the hidden units and the number of these hidden units were found to be the most important factors affecting the performance of the RNN. Increasing the number of hidden units from 15 to 250 led to a 56.3% decrease in the prediction error on the cross-validation data. Similarly, optimizing the standard deviation of the initial Gaussian distribution of the synaptic weights σinitRNN led to a 28.4% decrease in the prediction error on the cross-validation data, with the error minimized for σinitRNN=0.02 with the four patients.


Assuntos
Neoplasias Pulmonares , Redes Neurais de Computação , Algoritmos , Humanos , Pulmão , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/radioterapia , Movimento (Física)
11.
J Neurosci ; 28(34): 8489-501, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18716207

RESUMO

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.


Assuntos
Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Neuralgia/etiologia , Neuralgia/metabolismo , Órbita/inervação , Nervo Isquiático , Fator 3 Ativador da Transcrição/genética , Animais , Antígenos de Diferenciação/genética , Comportamento Animal/efeitos dos fármacos , Biomarcadores/metabolismo , Constrição Patológica , Citocinas/genética , Ensaio de Imunoadsorção Enzimática , Gânglios Sensitivos/metabolismo , Proteína Glial Fibrilar Ácida/genética , Hiperestesia/etiologia , Hiperestesia/psicologia , Imuno-Histoquímica , Interleucina-6/genética , Masculino , Minociclina/farmacologia , Neuroglia/metabolismo , Neurônios/metabolismo , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nervo Isquiático/lesões , Fatores de Tempo , Traumatismos do Sistema Nervoso/complicações
12.
J Neurochem ; 107(1): 50-60, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18636982

RESUMO

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.


Assuntos
Interleucina-6/metabolismo , Janus Quinase 1/metabolismo , Microglia/metabolismo , Doenças do Sistema Nervoso Periférico/fisiopatologia , Fator de Transcrição STAT3/metabolismo , Medula Espinal/fisiopatologia , Animais , Anticorpos/farmacologia , Modelos Animais de Doenças , Ativação Enzimática/fisiologia , Hiperalgesia/enzimologia , Hiperalgesia/fisiopatologia , Interleucina-6/genética , Interleucina-6/imunologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Ligadura , Masculino , Microglia/enzimologia , Nervos Periféricos/enzimologia , Nervos Periféricos/fisiopatologia , Doenças do Sistema Nervoso Periférico/enzimologia , Doenças do Sistema Nervoso Periférico/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Medula Espinal/enzimologia , Medula Espinal/metabolismo , Nervos Espinhais/lesões , Nervos Espinhais/fisiopatologia , Nervos Espinhais/cirurgia , Regulação para Cima/fisiologia
13.
J Neurochem ; 106(2): 757-69, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18419759

RESUMO

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.


Assuntos
Quimiocina CCL2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/fisiopatologia , Neurônios Aferentes/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Pirrolidinas/farmacologia , Receptores CCR2/antagonistas & inibidores , Medula Espinal/citologia , Análise de Variância , Animais , Comportamento Animal , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Cálcio/metabolismo , Quimiocina CCL2/genética , Ensaio de Imunoadsorção Enzimática/métodos , Gânglios Espinais/citologia , Masculino , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Substância P/genética , Substância P/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
14.
J Neurosci Methods ; 167(2): 148-59, 2008 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-17949823

RESUMO

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.


Assuntos
Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Hiperalgesia/terapia , Lentivirus/fisiologia , Neuroglia/metabolismo , Medula Espinal/citologia , Animais , Embrião de Mamíferos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Injeções Espinhais/métodos , Atividade Motora/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Limiar da Dor/fisiologia , Ratos
15.
Mol Ther ; 15(4): 687-697, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28192702

RESUMO

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.

16.
PLoS Negl Trop Dis ; 11(11): e0006058, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29149212

RESUMO

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.


Assuntos
Anti-Inflamatórios/metabolismo , Macrolídeos/metabolismo , Mycobacterium ulcerans/metabolismo , Sistema Nervoso/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Neuralgia/fisiopatologia , Ratos
17.
J Neurosci ; 25(35): 7896-904, 2005 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-16135746

RESUMO

Keeping in mind the increased pain complaints reported in anxious or depressive patients, our goal was to investigate in rats the consequences of an experimentally provoked state of anxiety/depression on pain behavior and on its underlying mechanisms. We therefore used a model of social defeat consisting of a 30 min protected confrontation followed by a 15 min physical confrontation, repeated during 4 d, that elicited symptoms close to those observed in humans with anxiety or depression. Indeed, 5 d later, animals subjected to social-defeat confrontation were characterized by a decrease of sweet-water consumption and of body weight, and a hyperactivity of the hypothalamic-pituitary-adrenal axis, suggesting that the social-defeat procedure induced a prolonged state of anxiety. Rats subjected to the social-defeat procedure showed an enhanced nociceptive behavior to the subcutaneous administration of formalin, 5 d after the last confrontation session. Because chronic treatment with the established anxiolytic chlordiazepoxide (10 mg.kg(-1).d(-1)) prevented hyperalgesia, this strongly suggested that this experimental procedure might be a suitable animal model of "anxiety-induced hyperalgesia." Hyperalgesia associated with anxiety not only was related to a significant increase of CCKLM [cholecystokinin (CCK)-like material] in frontal cortex microdialysates but also was prevented by a CCK-B receptor antagonist [4-[[2-[[3-(1H-indol-3-yl)-2-methyl-1-oxo-2[[(tricyclo[3.3[12,17]dec-2-yloxy)-carbonyl]amino]-propyl]amino]-1-phenyethyl]amino]-4-oxo-[R-(R*, R*)]-butanoate N-methyl-D-glucamine (CI-988)] (2 mg/kg), strongly supporting the involvement of central CCKergic systems in these phenomena. Finally, combined treatments with CI-988 and morphine completely suppressed pain-related behavior, supporting the idea that the association of both compounds might represent a new therapeutic approach to reduce the increase of pain complaints highly prevalent among anxious or depressive patients.


Assuntos
Ansiedade/metabolismo , Ansiedade/psicologia , Córtex Cerebral/metabolismo , Colecistocinina/metabolismo , Hiperalgesia/metabolismo , Hiperalgesia/psicologia , Receptores da Colecistocinina/fisiologia , Animais , Peso Corporal/fisiologia , Masculino , Medição da Dor/métodos , Ratos , Ratos Sprague-Dawley
18.
Pain ; 157(4): 827-839, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26655733

RESUMO

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.


Assuntos
Barreira Hematoneural/metabolismo , Células Endoteliais/metabolismo , Neuralgia/fisiopatologia , Traumatismos dos Nervos Periféricos/metabolismo , Nervo Isquiático/fisiopatologia , Transdução de Sinais , Animais , Proteínas Hedgehog/metabolismo , Inflamação/metabolismo , Masculino , Neuralgia/metabolismo , Ratos Sprague-Dawley , Neuropatia Ciática/fisiopatologia , Receptor 4 Toll-Like/metabolismo
19.
J Neurosci ; 22(13): 5442-51, 2002 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12097496

RESUMO

Two proteins previously known as Na(+)-dependent phosphate transporters have been identified recently as vesicular glutamate transporters (VGLUT1 and VGLUT2). Together, VGLUT1 and VGLUT2 are operating at most central glutamatergic synapses. In this study, we characterized a third vesicular glutamate transporter (VGLUT3), highly homologous to VGLUT1 and VGLUT2. Vesicles isolated from endocrine cells expressing recombinant VGLUT3 accumulated l-glutamate with bioenergetic and pharmacological characteristics similar, but not identical, to those displayed by the type-1 and type-2 isoforms. Interestingly, the distribution of VGLUT3 mRNA was restricted to a small number of neurons scattered in the striatum, hippocampus, cerebral cortex, and raphe nuclei, in contrast to VGLUT1 and VGLUT2 transcripts, which are massively expressed in cortical and deep structures of the brain, respectively. At the ultrastructural level, VGLUT3 immunoreactivity was concentrated over synaptic vesicle clusters present in nerve endings forming asymmetrical as well as symmetrical synapses. Finally, VGLUT3-positive neurons of the striatum and raphe nuclei were shown to coexpress acetylcholine and serotonin transporters, respectively. Our study reveals a novel class of glutamatergic nerve terminals and suggests that cholinergic striatal interneurons and serotoninergic neurons from the brainstem may store and release glutamate.


Assuntos
Acetilcolina/análise , Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Proteínas de Membrana Transportadoras , Neurônios/metabolismo , Serotonina/análise , Proteínas de Transporte Vesicular , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos Acídicos/fisiologia , Animais , Encéfalo/metabolismo , Proteínas de Transporte/genética , Linhagem Celular , Clonagem Molecular , Ácido Glutâmico/farmacologia , Cinética , Dados de Sequência Molecular , Neurônios/química , Neurônios/efeitos dos fármacos , RNA Mensageiro/biossíntese , Núcleos da Rafe , Ratos , Alinhamento de Sequência , Vesículas Sinápticas/química , Distribuição Tecidual , Transcrição Gênica , Proteína Vesicular 1 de Transporte de Glutamato , Proteína Vesicular 2 de Transporte de Glutamato , Proteínas Vesiculares de Transporte de Glutamato
20.
Curr Gene Ther ; 3(3): 223-38, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12762481

RESUMO

Chronic pain is frequently associated with profound alterations of neuronal systems involved in pain processing and should be considered as an actual disease state of the nervous system. It should not only be relieved, but must really be treated in suffering patients. However, some forms of chronic pain, in particular those of neuropathic origin, are most often not satisfactorily managed with currently available pharmacological agents, some of which, in addition, may be poorly tolerated by some patients. In this context, gene-based approaches may contribute to the search for a better management of chronic pain. The question then arises regarding the most appropriate level for such an intervention using gene-transfer techniques. The first experimental protocols attempted the transfer of opioid precursor genes and their overexpression mainly at the spinal level. They demonstrated the feasibility and the real interest of these approaches by showing that local overproduction of opioid peptides induced antinociceptive effects in animal models of persistent pain, of inflammatory-, neuropathic- and even cancerous origin. Although really tempting data were obtained using gene-based techniques in experimental inflammatory diseases, the possible clinical interest of these approaches in chronic pain has still to be established. Nevertheless, targeting some proinflammatory cytokines, involved not only in inflammation but also in the induction and probably the perpetuation of pain, raises the possibility to block the "development" of chronic pain rather that to "simply" relieve established ongoing pain. Future gene-based protocols will certainly target some of the recently identified molecules involved in pain transduction mechanisms, sensory nerve sensitization or pain perpetuation, and evaluate their potential interest to ideally abolish or, at least, reduce chronic pain.


Assuntos
Terapia Genética , Manejo da Dor , Animais , Doença Crônica , Humanos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa