Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cephalalgia ; 38(5): 833-845, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-28554243

RESUMO

Background Recent genome-wide association studies have identified transient receptor potential M8 ( TRPM8) as a migraine susceptibility gene. TRPM8 is a nonselective cation channel that mediates cool perception. However, its precise role in migraine pathophysiology is elusive. Transient receptor potential V1 (TRPV1) is a nonselective cation channel activated by noxious heat. Both TRPM8 and TRPV1 are expressed in trigeminal ganglion (TG) neurons. Methods We investigated the functional roles of TRPM8 and TRPV1 in a meningeal inflammation-based migraine model by measuring the effects of facial TRPM8 activation on thermal allodynia and assessing receptor coexpression changes in TG neurons. We performed retrograde tracer labeling to identify TG neurons innervating the face and dura. Results We found that pharmacological TRPM8 activation reversed the meningeal inflammation-induced lowering of the facial heat pain threshold, an effect abolished by genetic ablation of TRPM8. No significant changes in the heat pain threshold were seen in sham-operated animals. Meningeal inflammation caused dynamic alterations in TRPM8/TRPV1 coexpression patterns in TG neurons, and colocalization was most pronounced when the ameliorating effect of TRPM8 activation on thermal allodynia was maximal. Our tracer assay disclosed the presence of dura-innervating TG neurons sending collaterals to the face. Approximately half of them were TRPV1-positive. We also demonstrated functional inhibition of TRPV1 by TRPM8 in a cell-based assay using c-Jun N-terminal kinase phosphorylation as a surrogate marker. Conclusions Our findings provide a plausible mechanism to explain how facial TRPM8 activation can relieve migraine by suppressing TRPV1 activity. Facial TRPM8 appears to be a promising therapeutic target for migraine.


Assuntos
Transtornos de Enxaqueca/metabolismo , Transtornos de Enxaqueca/fisiopatologia , Canais de Cátion TRPM/biossíntese , Canais de Cátion TRPV/biossíntese , Gânglio Trigeminal/metabolismo , Gânglio Trigeminal/fisiopatologia , Animais , Dor Facial/metabolismo , Dor Facial/fisiopatologia , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células PC12 , Medição da Dor/métodos , Ratos
2.
J Cereb Blood Flow Metab ; 37(3): 890-901, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27142867

RESUMO

Single episodes of cortical spreading depression (CSD) are believed to cause typical migraine aura, whereas clusters of spreading depolarizations have been observed in cerebral ischemia and subarachnoid hemorrhage. We recently demonstrated that the release of high-mobility group box 1 (HMGB1) from cortical neurons after CSD in a rodent model is dependent on the number of CSD episodes, such that only multiple CSD episodes can induce significant HMGB1 release. Here, we report that only multiple CSD inductions caused microglial hypertrophy (activation) accompanied by a greater impact on the transcription activity of the HMGB1 receptor genes, TLR2 and TLR4, while the total number of cortical microglia was not affected. Both an HMGB1-neurtalizing antibody and the HMGB1 inhibitor glycyrrhizin abrogated multiple CSD-induced microglial hypertrophy. Moreover, multiple CSD inductions failed to induce microglial hypertrophy in TLR2/4 double knockout mice. These results strongly implicate the HMGB1-TLR2/4 axis in the activation of microglia following multiple CSD inductions. Increased expression of the lysosomal acid hydrolase cathepsin D was detected in activated microglia by immunostaining, suggesting that lysosomal phagocytic activity may be enhanced in multiple CSD-activated microglia.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical , Proteína HMGB1/fisiologia , Microglia/metabolismo , Animais , Catepsina D/metabolismo , Hipertrofia , Camundongos , Camundongos Knockout , Microglia/citologia , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
3.
Brain ; 139(Pt 9): 2380-94, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27368346

RESUMO

Mutations in RNA-binding proteins, including fused in sarcoma (FUS) and TAR DNA-binding protein 43 (TDP-43, encoded by TARDBP), are associated with sporadic and familial amyotrophic lateral sclerosis. A major question is whether neuronal loss is caused by toxic gain-of-function cytoplasmic aggregates or loss of nuclear RNA-binding protein function. We generated a transgenic mouse overexpressing exogenous FUS without a nuclear localization signal (ΔNLS-FUS), which developed progressive spastic motor deficits and neuronal loss in the motor cortex. The ΔNLS-FUS protein was restricted to the cytoplasm and formed ubiquitin/p62-positive aggregates. Endogenous FUS expression, nuclear localization, and splicing activity were not altered, indicating that mislocated FUS is sufficient for proteinopathy. Crossing ΔNLS-FUS with wild-type human TDP-43 transgenic mice exacerbated pathological and behavioural phenotypes, suggesting that both proteins are involved in a common cascade. RNA-sequence analysis revealed specific transcriptome alterations, including genes regulating dynein-associated molecules and endoplasmic reticulum stress. ΔNLS-FUS mice are promising tools for understanding amyotrophic lateral sclerosis pathogenesis and testing new therapeutic approaches.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Sinais de Localização Nuclear/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Animais , Comportamento Animal , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora , Fenótipo
4.
Neurosci Res ; 112: 57-62, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27312532

RESUMO

Cortical spreading depression (CSD) has been implicated in a variety of neurological disorders. However, the relationship between serum sex hormones and susceptibility to the development of CSD in naturally estrous cycling female animals is largely unknown. The natural estrous cycle of mice consists of four stages, namely, proestrus, estrus, metestrus and diestrus. We measured the serum concentration of estradiol and progesterone in estrus and diestrus and compared the minimum potassium concentrations necessary to evoke CSD in each stage and in males. In diestrus, the minimum potassium concentration required to evoke CSD was significantly lower compared to the other three phases and male animals. The serum level of estradiol is significantly higher and serum level of progesterone is significantly lower in diestrus compared to estrus. Furthermore, when we administered an estrogen receptor antagonist, the susceptibility to the development of CSD was decreased. Conversely, the administration of a progesterone receptor antagonist increased the susceptibility to CSD. Our results demonstrated that neuronal excitability related to CSD induction differs among the natural estrous phases in mice.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical , Ciclo Estral/fisiologia , Potássio/metabolismo , Animais , Diestro/fisiologia , Estradiol/sangue , Estro/fisiologia , Feminino , Masculino , Camundongos Endogâmicos C57BL , Progesterona/sangue , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Progesterona/antagonistas & inibidores
5.
Neurosci Res ; 106: 74-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26739227

RESUMO

We examined the ability of trigeminal nerve activation to induce cortical spreading depression in rats. Capsaicin was injected into the bilateral plantar or whisker pad for either 4 or 6 days in rats. The number and duration of cortical spreading depressions induced by potassium were significantly increased in animals injected with capsaicin in the bilateral whisker pad compared with animals injected in the bilateral plantar or in controls, while administration of a GABAA receptor agonist decreased these effects. Repetitive nociceptive stimulation of the trigeminal nerve lowers the threshold for the induction of cortical spreading depression by altering GABAergic neuronal activity.


Assuntos
Capsaicina/farmacologia , Depressão Alastrante da Atividade Elétrica Cortical/efeitos dos fármacos , Nociceptividade , Nervo Trigêmeo/efeitos dos fármacos , Animais , Masculino , Transtornos de Enxaqueca/fisiopatologia , Ratos Sprague-Dawley , Nervo Trigêmeo/fisiologia
6.
Cephalalgia ; 36(1): 44-52, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25862357

RESUMO

INTRODUCTION: Cortical spreading depression (CSD) has recently been shown to induce the release of the nuclear protein termed high-mobility group box 1 from neurons, causing activation of the trigeminovascular system. Here, we explored the effects of single and multiple cortical spreading depression inductions on high-mobility group box 1 (HMGB1) transcriptional activity relative to high-mobility group box 1 protein expression levels and intracellular localization in cortical neurons and astrocytes. METHODS: Single or multiple cortical spreading depression inductions were achieved by KCl application to the mouse cerebral cortex. The animals were sacrificed at 30 minutes, 3 hours and 24 hours after cortical spreading depression induction. High-mobility group box 1 expression levels were explored with in situ hybridization, Western blotting and immunostaining. RESULTS: Cortical spreading depression up-regulated high-mobility group box 1 transcriptional activity in neurons at 3 hours in a manner that was dependent on the number of cortical spreading depression inductions. At 24 hours, the high-mobility group box 1 transcriptional activity had returned to basal levels. Cortical spreading depression induced a reduction in high-mobility group box 1 protein expression at 3 hours, which was also dependent on the number of cortical spreading depression inductions. Following cortical spreading depression, the release of high-mobility group box 1 from the nucleus was observed in a small proportion of neurons, but not in astrocytes. CONCLUSION: Cortical spreading depression induced translocation of high-mobility group box 1 from neuronal nuclei, driving transcriptional up-regulation of high-mobility group box 1 to maintain protein levels.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical/fisiologia , Proteína HMGB1/biossíntese , Lobo Parietal/metabolismo , Animais , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Proteína HMGB1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Tempo
7.
Neurosci Res ; 77(1-2): 110-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23962823

RESUMO

Extracellular signal-regulated kinase (ERK) is known to be phosphorylated after exposure to noxious stimuli. In this study, we investigated the response in the dura mater to nociceptive stimulation, which is thought to be responsible for the pathogenesis of headaches, including migraines. We also examined the level of ERK phosphorylation in the trigeminal ganglion following cortical spreading depression (CSD), which is thought to play an important role in migraine pathophysiology. Western blot and immunohistochemical analyses showed a significant increase in the ERK phosphorylation levels 3 min following an application of 10mM capsaicin to the dura mater. This increase was inhibited after an application of the TRPV1 antagonist capsazepine or a MEK inhibitor. An immunohistochemical analysis revealed that most of the small-sized trigeminal ganglion neurons with TRPV1-immunoreactivity that innervate the dura mater exhibited pERK-immunoreactivity, suggesting that these neurons had responded to nociceptive stimulation. CSD increased the level of ERK phosphorylation 30 min after its elicitation, and this response was inhibited by a prior intraventricular administration of TRPV1 antagonist. These results indicate that CSD can activate dural TRPV1 to send nociceptive signals to the trigeminal system, and they provide important clues regarding the relationship between CSD and the trigeminovascular system.


Assuntos
Depressão Alastrante da Atividade Elétrica Cortical , Dura-Máter/enzimologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gânglio Trigeminal/enzimologia , Animais , Capsaicina/farmacologia , Dura-Máter/efeitos dos fármacos , Masculino , Nociceptividade/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Fármacos do Sistema Sensorial/farmacologia , Gânglio Trigeminal/efeitos dos fármacos
8.
Rinsho Shinkeigaku ; 52(11): 1297-8, 2012.
Artigo em Japonês | MEDLINE | ID: mdl-23196597

RESUMO

Acupuncture is known as the effective tool for headache, but the mechanism of the effect is unknown. We already revealed the acupuncture effect in the clinical team of headache center in Keio University Hospital. Therefore, we tried to establish the animal model for elucidation of mechanism of acupuncture effect in the pathophysiology of headache. Resent study, we reveal the threshold-reduction of the genesis of the cortical spreading depression (CSD; thought as the trigger of migraine attack) during the trigeminal nerve stimulation. This result suggests that, the somatosensory stimulation may influence the occurrence and severity of the pathogenesis of migraine. Furthermore, we assume that our result may lead to the underlying mechanism of acupuncture effect.


Assuntos
Terapia por Acupuntura , Cefaleia/terapia , Animais , Humanos
9.
Rinsho Shinkeigaku ; 52(11): 1365-8, 2012.
Artigo em Japonês | MEDLINE | ID: mdl-23196620

RESUMO

Among many conditions causing small vessel diseases, lipohyalinosis is the leading pathology next to microatheroma. Lipohyalinosis affects penetrating arteries distally than microatheroma. Proliferation of smooth muscle cells may occlude the lumen, reducing the blood flow and inducing lacunar infarction. In contrast, fibrinoid necrosis of smooth muscle cells in the media may weaken the vascular constriction, increasing the perfusion pressure in the capillary and damaging the blood brain barrier which can induce white matter lesion. Neurovascular unit (NVU) is a concept that neurons, astrocytes, and vessels function as a unit to support neuronal activity. NVU is involved in the maintenance of synapse, transmitter, energy metabolism, blood-brain barrier, and blood flow. Change in neuronal activity is transmitted to capillaries through NVU, where the information is collected along vessels proximally and regulates blood flow (proximal integration model). CARASIL and CADASIL both affect vascular smooth muscle cells, resulting in vascular dilatation, damaging NVU, and inducing white matter lesion. Occlusion of the affected vessels, causing cerebral ischemia, under these diseases is relatively infrequent. Similarity in pathophysiology between hypertensive arteriolar disease and the familial angiopathy may indicate that injury to NVU may indicate the common pathophysiology of white matter lesions.


Assuntos
Alopecia/patologia , CADASIL/patologia , Infarto Cerebral/patologia , Transtornos Cerebrovasculares/patologia , Leucoencefalopatias/patologia , Doenças da Coluna Vertebral/patologia , Alopecia/fisiopatologia , CADASIL/fisiopatologia , Infarto Cerebral/fisiopatologia , Humanos , Leucoencefalopatias/fisiopatologia , Microcirculação/fisiologia , Músculo Liso Vascular/patologia , Doenças da Coluna Vertebral/fisiopatologia
10.
Neurobiol Dis ; 48(3): 367-78, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22820141

RESUMO

Botulinum neurotoxin type-A (BoNT-A) is clinically used for patients with pain disorders and dystonia. The precise mechanism whereby BoNT-A controls pain remains elusive. Here, we studied how BoNT-A affects the expression of the transient receptor potential vanilloid subfamily member 1 (TRPV1), a cation channel critically implicated in nociception, in the trigeminal system. Histological studies revealed that subcutaneous BoNT-A injection (0.25, 0.5, or 5 ng/kg) into the face targeted the ophthalmic division of trigeminal ganglion (TG) neurons and decreased TRPV1-immunoreactive neurons in the TG and TRPV1-immunoreactive fibers in rat trigeminal terminals. Of note, TG neurons that received projections from the dura mater, a principal site of headache generation, had reduced TRPV1 expression. BoNT-A-induced cleavage of SNAP25 (synaptosomal-associated protein of 25-kDa) in the TG became obvious 2 days after BoNT-A administration and persisted for at least 14 days. Quantitative real-time RT-PCR (reverse transcription-polymerase chain reaction) data indicated that the TRPV1-decreasing effects of BoNT-A were not mediated by transcriptional downregulation. By employing a surface protein biotin-labeling assay, we demonstrated that BoNT-A inhibited TRPV1 trafficking to the plasma membrane in primary TG neurons. Moreover, Y200F-mutated TRPV1, which is incapable of trafficking to the plasma membrane, was expressed in PC12 cells by transfection, and pharmacological studies revealed that TRPV1 in the cytoplasm was more predisposed to proteasome-mediated proteolysis than plasma membrane-located TRPV1. We conclude that the mechanism by which BoNT-A reduces TRPV1 expression involves the inhibition of TRPV1 plasma membrane trafficking and proteasome-mediated degradation in the cytoplasm. This paradigm seems to explain how BoNT-A alleviates TRPV1-mediated pain. Our data reveal a likely molecular mechanism whereby BoNT-A treatment reduces TRPV1 expression in the trigeminal system and provide important clues to novel therapeutic measures for ameliorating craniofacial pain.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Fármacos Neuromusculares/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Cátion TRPV/biossíntese , Animais , Western Blotting , Imuno-Histoquímica , Masculino , Células PC12 , Dor/tratamento farmacológico , Dor/metabolismo , Transporte Proteico/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Gânglio Trigeminal/efeitos dos fármacos , Gânglio Trigeminal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...