Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Can J Physiol Pharmacol ; 100(7): 594-611, 2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35413210

RESUMO

1,2,3,4-tetrahydroisoquinoline (TIQ) is endogenously present in the human brain, and some of its derivatives are thought to contribute to the induction of Parkinson's disease (PD)-like signs in rodents and primates. In contrast, the endogenous TIQ derivative 1-methyl-TIQ (1-MeTIQ) is reported to be neuroprotective. In the present study, we compared the effects of artificially modified 1-MeTIQ derivatives (loading an N-propyl, N-propenyl, N-propargyl, or N-butynyl group) on 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD-like signs in mice. In a behavioral study, MPTP-induced bradykinesia was significantly decreased by all compounds. However, only 1-Me-N-propargyl-TIQ showed an inhibitory effect by blocking the MPTP-induced reduction in striatal dopamine content and the number of nigral tyrosine hydroxylase-positive cells. Western blot analysis showed that 1-Me-N-propargyl-TIQ and 1-Me-N-butynyl-TIQ potently prevented the MPTP-induced decrease in dopamine transporter expression, whereas 1-MeTIQ and 1-Me-N-propyl-TIQ did not. These results suggest that although loading an N-propargyl group on 1-MeTIQ clearly enhanced neuroprotective effects, other N-functional groups showed distinct pharmacological properties characteristic of their functional groups. Thus, the number of bonds and length of the N-functional group may contribute to the observed differences in effect.


Assuntos
Intoxicação por MPTP , Fármacos Neuroprotetores , Transtornos Parkinsonianos , Tetra-Hidroisoquinolinas , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , Animais , Humanos , Intoxicação por MPTP/tratamento farmacológico , Intoxicação por MPTP/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Transtornos Parkinsonianos/induzido quimicamente , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/prevenção & controle
2.
Drug Chem Toxicol ; 45(1): 393-397, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31899978

RESUMO

Chemotherapy-induced peripheral neuropathy has an important impact on the quality of life of cancer patients. Vincristine-induced neuropathy is a major dose-limiting side effect. Symptoms of peripheral neuropathy are spontaneous pain, allodynia, and hyperalgesia. To analyze the contribution of substance P to the development of vincristine-induced mechanical allodynia/hyperalgesia, substance P levels in the rat spinal dorsal horn were analyzed after vincristine treatment. Mechanical allodynia/hyperalgesia was tested with the von Frey filaments 14 days after intraperitoneal (i.p.) administration of vincristine 0.1 mg/kg/day in rats. Vincristine-induced mechanical allodynia/hyperalgesia after day 14 was significantly inhibited by the neurokinin 1 receptor antagonist, aprepitant (20 mg/kg, s.c.). Immunohistochemistry showed that vincristine treatment significantly increased substance P expression (30.3% ± 2.4%) compared to saline treatment in the superficial layers of the spinal dorsal horn. Moreover, vincristine treatment significantly increased the substance P level in the spinal cord. These results suggest that vincristine treatment increases substance P in the spinal dorsal horn, and that aprepitant attenuates mechanical allodynia/hyperalgesia in vincristine-induced neuropathic rats.


Assuntos
Neuralgia , Substância P , Animais , Modelos Animais de Doenças , Humanos , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Qualidade de Vida , Ratos , Ratos Sprague-Dawley , Medula Espinal , Vincristina/toxicidade
3.
Int J Mol Sci ; 21(12)2020 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-32570786

RESUMO

Painful peripheral neuropathy is a common adverse effect of paclitaxel (PTX) treatment. To analyze the contribution of transient receptor potential vanilloid 1 (TRPV1) in the development of PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia, TRPV1 expression in the rat spinal cord was analyzed after intraperitoneal administration of 2 and 4 mg/kg PTX. PTX treatment increased the expression of TRPV1 protein in the spinal cord. Immunohistochemistry showed that PTX (4 mg/kg) treatment increased TRPV1 protein expression in the superficial layers of the spinal dorsal horn 14 days after treatment. Behavioral assessment using the paw withdrawal response showed that PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia after 14 days was significantly inhibited by oral or intrathecal administration of the TRPV1 antagonist AMG9810. We found that intrathecal administration of small interfering RNA (siRNA) to knock down TRPV1 protein expression in the spinal cord significantly decreased PTX-induced mechanical allodynia/hyperalgesia and thermal hyperalgesia. Together, these results demonstrate that TRPV1 receptor expression in spinal cord contributes, at least in part, to the development of PTX-induced painful peripheral neuropathy. TRPV1 receptor antagonists may be useful in the prevention and treatment of PTX-induced peripheral neuropathic pain.


Assuntos
Hiperalgesia/metabolismo , Paclitaxel/efeitos adversos , Medula Espinal/metabolismo , Canais de Cátion TRPV/metabolismo , Acrilamidas/administração & dosagem , Acrilamidas/farmacologia , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Modelos Animais de Doenças , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Injeções Intraperitoneais , Injeções Espinhais , Masculino , Paclitaxel/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/farmacologia , Ratos , Canais de Cátion TRPV/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos
4.
J Pharmacol Sci ; 133(4): 254-260, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28410966

RESUMO

The clinical anti-cancer efficacy of vincristine is limited by the development of dose-dependent peripheral neuropathy. Up-regulation of transient receptor potential vanilloid 1 (TRPV1) is correlated with peripheral neuropathy following anti-cancer drug treatment. To analyze the contribution of TRPV1 to the development of vincristine-induced mechanical allodynia/hyperalgesia, TRPV1 expression in the rat dorsal root ganglion (DRG) was analyzed after vincristine treatment. Mechanical allodynia/hyperalgesia was tested with von Frey filaments 14 days after intraperitoneal administration of 0.1 mg/kg vincristine in rats. TRPV1 expression in DRGs following vincristine treatment was assessed with western blot analysis and in situ hybridization histochemistry. Vincristine-induced mechanical allodynia/hyperalgesia after day 14 was significantly inhibited by the TRP antagonist ruthenium red (3 mg/kg, s.c.) and the TRPV1 antagonist capsazepine (30 mg/kg, s.c.). Vincristine treatment increased the expression of TRPV1 protein in DRG neurons. In situ hybridization histochemistry revealed that most of the TRPV1 mRNA-labeled neurons in the DRG were small in size. Immunohistochemistry showed that isolectin B4-positive small DRG neurons co-expressed TRPV1 protein 14 days after treatment. These results suggest that vincristine treatment increases TRPV1 expression in small DRG neurons. TRPV1 expression may contribute to the development of vincristine-induced painful peripheral neuropathy.


Assuntos
Antineoplásicos Fitogênicos/toxicidade , Expressão Gênica/efeitos dos fármacos , Neuralgia/induzido quimicamente , Neuralgia/genética , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Vincristina/toxicidade , Animais , Capsaicina/análogos & derivados , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Humanos , Masculino , Neuralgia/tratamento farmacológico , Neurônios/metabolismo , Neurônios/patologia , Ratos Wistar , Rutênio Vermelho/farmacologia , Rutênio Vermelho/uso terapêutico , Canais de Cátion TRPV/antagonistas & inibidores , Regulação para Cima/efeitos dos fármacos
5.
J Pharmacol Sci ; 130(2): 117-22, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26883453

RESUMO

Oxaliplatin is a chemotherapeutic agent that is effective against various types of cancer including colorectal cancer. Acute cold hyperalgesia is a serious side effect of oxaliplatin treatment. Although the therapeutic drug pregabalin is beneficial for preventing peripheral neuropathic pain by targeting the voltage-dependent calcium channel α2δ-1 (Cavα2δ-1) subunit, the effect of oxaliplatin-induced acute cold hypersensitivity is uncertain. To analyze the contribution of the Cavα2δ-1 subunit to the development of oxaliplatin-induced acute cold hypersensitivity, Cavα2δ-1 subunit expression in the rat spinal cord was analyzed after oxaliplatin treatment. Behavioral assessment using the acetone spray test showed that 6 mg/kg oxaliplatin-induced cold hypersensitivity 2 and 4 days later. Oxaliplatin-induced acute cold hypersensitivity 4 days after treatment was significantly inhibited by pregabalin (50 mg/kg, p.o.). Oxaliplatin (6 mg/kg, i.p.) treatment increased the expression level of Cavα2δ-1 subunit mRNA and protein in the spinal cord 2 and 4 days after treatment. Immunohistochemistry showed that oxaliplatin increased Cavα2δ-1 subunit protein expression in superficial layers of the spinal dorsal horn 2 and 4 days after treatment. These results suggest that oxaliplatin treatment increases Cavα2δ-1 subunit expression in the superficial layers of the spinal cord and may contribute to functional peripheral acute cold hypersensitivity.


Assuntos
Antineoplásicos/toxicidade , Canais de Cálcio/metabolismo , Expressão Gênica/efeitos dos fármacos , Compostos Organoplatínicos/toxicidade , Medula Espinal/metabolismo , Doença Aguda , Animais , Canais de Cálcio/genética , Síndromes Periódicas Associadas à Criopirina/induzido quimicamente , Síndromes Periódicas Associadas à Criopirina/genética , Síndromes Periódicas Associadas à Criopirina/prevenção & controle , Masculino , Oxaliplatina , Pregabalina/administração & dosagem , Pregabalina/uso terapêutico , Ratos Wistar
6.
Mol Pain ; 11: 69, 2015 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-26567040

RESUMO

BACKGROUND: Peripheral cold neuropathic pain is a serious side effect of oxaliplatin treatment. However, the mechanism of oxaliplatin-induced cold hyperalgesia is unknown. In the present study, we investigated the effects of oxaliplatin on transient receptor potential ankyrin 1 (TRPA1) in dorsal root ganglion (DRG) neurons of rats. RESULTS: Behavioral assessment using the acetone spray test showed that 3 and 6 mg/kg oxaliplatin (i.p.) induced acute cold hypersensitivity after 1, 2, 4, and 7 days. Real-time PCR showed that oxaliplatin (6 mg/kg) significantly increased TRPA1 mRNA expression in DRGs at days 1, 2, and 4. Western blotting revealed that oxaliplatin significantly increased TRPA1 protein expression in DRGs at days 2, 4, and 7. Moreover, in situ hybridization histochemistry revealed that most TRPA1 mRNA-labeled neurons in the DRGs were small in size. Oxaliplatin significantly increased co-localization of TRPA1 expression and isolectin B4 binding in DRG neurons. Oxaliplatin induced a significant increase in the percent of TRPA1 mRNA-positive small neurons in DRGs at days 1, 2, and 4. In addition, we found that intrathecal administration of TRPA1 antisense, but not TRPA1 mismatched oligodeoxynucleotides, knocked down TRPA1 expression and decreased oxaliplatin-induced cold hyperalgesia. Double labeling showed that p-p38 mitogen-activated protein kinase (MAPK) was co-expressed in TRPA1 mRNA-labeled neurons at day 2 after oxaliplatin administration. Intrathecal administration of the p38 MAPK inhibitor, SB203580, significantly decreased oxaliplatin-induced acute cold hypersensitivity. CONCLUSIONS: Together, these results demonstrate that TRPA1 expression via activation of p38 MAPK in DRG neurons, at least in part, contributes to the development of oxaliplatin-induced acute cold hyperalgesia.


Assuntos
Síndromes Periódicas Associadas à Criopirina/induzido quimicamente , Gânglios Espinais/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Doença Aguda , Animais , Síndromes Periódicas Associadas à Criopirina/enzimologia , Síndromes Periódicas Associadas à Criopirina/fisiopatologia , Gânglios Espinais/enzimologia , Gânglios Espinais/fisiopatologia , Imidazóis/farmacologia , Masculino , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Ratos , Canal de Cátion TRPA1 , Proteínas Quinases p38 Ativadas por Mitógeno/genética
7.
Biol Pharm Bull ; 38(1): 134-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25342005

RESUMO

Psilocin (3-[2-(dimethylamino)ethyl]-1H-indol-4-ol) is a hallucinogenic component of the Mexican mushroom Psilocybe mexicana and a skeletal serotonin (5-HT) analogue. Psilocin is the active metabolite of psilocybin (3-[2-(dimethylamino)ethyl]-1H-indol-4-yl dihydrogen phosphate). In the present study, we examined the effects of systemically administered psilocin on extracellular dopamine and 5-HT concentrations in the ventral tegmental area (VTA), nucleus accumbens, and medial prefrontal cortex of the dopaminergic pathway in awake rats using in vivo microdialysis. Intraperitoneal administration of psilocin (5, 10 mg/kg) significantly increased extracellular dopamine levels in the nucleus accumbens. Psilocin did not affect the extracellular 5-HT level in the nucleus accumbens. Conversely, systemic administration of psilocin (10 mg/kg) significantly increased extracellular 5-HT levels in the medial prefrontal cortex of rats, but dopamine was decreased in this region. However, neither extracellular dopamine nor 5-HT levels in the VTA were altered by administration of psilocin. Behaviorally, psilocin significantly increased the number of head twitches. Thus, psilocin affects the dopaminergic system in the nucleus accumbens. In the serotonergic system, psilocin contribute to a crucial effect in the medial prefrontal cortex. The present data suggest that psilocin increased both the extracellular dopamine and 5-HT concentrations in the mesoaccumbens and/or mesocortical pathway.


Assuntos
Dopamina/metabolismo , Alucinógenos/farmacologia , Núcleo Accumbens/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Psilocibina/análogos & derivados , Serotonina/metabolismo , Animais , Masculino , Núcleo Accumbens/metabolismo , Córtex Pré-Frontal/metabolismo , Psilocibina/farmacologia , Ratos Wistar , Área Tegmentar Ventral/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
8.
Pharmacology ; 95(1-2): 87-94, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25633935

RESUMO

1,2,3,4-Tetrahydroisoquinoline (TIQ) and its derivatives, 1-methyl-TIQ (1-MeTIQ) and 1-benzyl-TIQ (1-BnTIQ), are endogenously present in the human brain. In this study, we compared the effects of TIQ derivatives on spontaneous nigral dopaminergic discharge in rats treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). In the low-to-middle dose range (0.01-1 mg/kg), intravenous administration of MPTP induced a transient and potent increase in the firing rate. TIQ (0.01-30 mg/kg) had no effects, and 1-MeTIQ and 1-BnTIQ (0.01-10 mg/kg) produced a weaker increase in the firing frequency immediately after intravenous administration. Pretreatment with 1-MeTIQ (80 mg/kg, i.p.) significantly inhibited the decrease in dopaminergic spontaneous firing induced by a high dose of MPTP. The nigral induction of thiobarbituric acid-reactive substances (TBARS) by MPTP was also significantly suppressed by pretreatment with 1-MeTIQ. These results suggest that the neurotoxicity induced by TIQ derivatives is relatively weak compared to that induced by MPTP. The neuroprotective effect of 1-MeTIQ from MPTP-induced toxicity may be partially due to a decrease in free radicals, as suggested by a decrease in TBARS. This action presumably prevents cell membrane degeneration.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Dopaminérgicos/farmacologia , Neurônios/efeitos dos fármacos , Neurotoxinas/farmacologia , Substância Negra/efeitos dos fármacos , Tetra-Hidroisoquinolinas/farmacologia , Animais , Dopamina/fisiologia , Masculino , Neurônios/metabolismo , Neurônios/fisiologia , Ratos Sprague-Dawley , Substância Negra/metabolismo , Substância Negra/fisiologia , Substâncias Reativas com Ácido Tiobarbitúrico/metabolismo
9.
Drug Chem Toxicol ; 38(2): 167-73, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24896366

RESUMO

An itch is experientially well known that the scratching response of conditions such as atopic dermatitis is enhanced under psychological stress. Morphine is typical narcotic drug that induces a scratching response upon local application as an adverse drug reaction. Although long-term treatment with morphine will cause tolerance and dependence, morphine withdrawal can cause psychologically and physiologically stressful changes in humans. In this study, we evaluated the effects of morphine withdrawal on histamine-induced scratching behavior in mice. Administration of morphine with progressively increasing doses (10-50 mg/kg, i.p.) was performed for 5 consecutive days. At 3, 24, 48, and 72 hr after spontaneous withdrawal from the final morphine dose, histamine was intradermally injected into the rostral part of the back and then the number of bouts of scratching in 60 min was recorded and summed. We found that at 24 hr after morphine withdrawal there was a significant increase in histamine-induced scratching behavior. The spinal c-Fos positive cells were also significantly increased. The relative adrenal weight increased and the relative thymus weight decreased, both significantly. Moreover, the plasma corticosterone levels changed in parallel with the number of scratching bouts. These results suggest that morphine withdrawal induces a stressed state and enhances in histamine-induced scratching behavior. Increased reaction against histamine in the cervical vertebrae will participate in this stress-induced itch enhancement.


Assuntos
Histamina/metabolismo , Morfina/administração & dosagem , Prurido/etiologia , Síndrome de Abstinência a Substâncias/fisiopatologia , Analgésicos Opioides/administração & dosagem , Animais , Comportamento Animal/efeitos dos fármacos , Corticosterona/sangue , Relação Dose-Resposta a Droga , Histamina/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fatores de Tempo
10.
Neurol Sci ; 35(2): 205-13, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23820959

RESUMO

Guillain-Barré syndrome, which is a variant of acute inflammatory neuropathy, is associated with anti-GM1 antibodies and causes ataxia. We investigated the effects of IgG anti-GM1 monoclonal antibody (IgG anti-GM1 mAb) on spontaneous muscle action potentials in a rat spinal cord-muscle co-culture system and the localization of IgG anti-GM1 mAb binding in the rat hemi-diaphragm. The frequency of spontaneous muscle action potentials in innervated muscle cells was acutely inhibited by IgG anti-GM1 mAb. When cultures were pretreated with GM2 synthase antisense oligodeoxynucleotide, IgG anti-GM1 mAb failed to inhibit spontaneous muscle action potentials, demonstrating the importance of the GM1 epitope in the action of IgG anti-GM1 mAb. Immunohistochemistry of rat hemi-diaphragm showed that IgG anti-GM1 mAb binding overlapped with neurofilament 200 (NF200) antibodies staining, but not α-bungarotoxin (α-BuTx) staining, demonstrating that IgG anti-GM1 mAb was localized at the presynaptic nerve terminal. IgG anti-GM1 mAb binding overlapped with syntaxin antibody and S-100 antibody in the nerve terminal. After collagenase treatment, IgG anti-GM1 mAb and NF200 antibodies did not show staining, but α-BuTx selectively stained the hemi-diaphragm. IgG anti-GM1 mAb binds to the presynaptic nerve terminal of neuromuscular junctions. Therefore, we suggest that the inhibitory effect of IgG anti-GM1 mAb on spontaneous muscle action potentials is related to the GM1 epitope in presynaptic motor nerve terminals at the NMJs.


Assuntos
Potenciais de Ação , Anticorpos Monoclonais/imunologia , Autoanticorpos/imunologia , Gangliosídeo G(M1)/imunologia , Imunoglobulina G/imunologia , Junção Neuromuscular/fisiologia , Animais , Células Cultivadas , Colagenases/metabolismo , Diafragma/efeitos dos fármacos , Diafragma/fisiologia , Técnicas In Vitro , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Proteínas de Neurofilamentos/metabolismo , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Terminações Pré-Sinápticas/fisiologia , Proteínas Qa-SNARE/metabolismo , Ratos , Ratos Wistar , Proteínas S100/metabolismo , Medula Espinal/fisiologia
11.
Neurol Res ; 46(6): 505-515, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38569563

RESUMO

OBJECTIVES: Neuropathic pain is characterized by long-lasting, intractable pain. Sciatic nerve ligation is often used as an animal model of neuropathic pain, and the spared nerve injury (SNI) model, in which the common peroneal nerve (CPN) and tibial nerve (TN) are ligated, is widely used. In the present study, we evaluated the analgesic effect of a cholinergic agonist, carbachol, on a neuropathic pain model prepared by sural nerve (SN) ligation in mice. METHODS: The SN was tightly ligated as a branch of the sciatic nerve. Mechanical and thermal allodynia, and hyperalgesia were assessed using von Frey filaments and heat from a hot plate. The analgesic effects of intracerebroventricularly-administered morphine and carbachol were compared. RESULTS: SN ligation resulted in a significant decrease in pain threshold for mechanical stimulation 1 day after ligation. In response to thermal stimulation, allodynia was observed at 50°C and hyperalgesia at 53 and 56°C 3 days after ligation. Content of thiobarbituric acid reactive substances (TBARS) in the spinal cord increased significantly at 6 and 12 h after ligation. Acetylcholine content of the spinal cord also increased at 5 and 7 days after ligation. Intracerebroventricular administration of carbachol at 7 days after ligation produced a marked analgesic effect against mechanical and thermal stimuli, which was stronger and longer-lasting than morphine at all experimental time points. CONCLUSION: These findings suggest that cholinergic nerves are involved in allodynia and hyperalgesia of the SN ligation neuropathic pain model.


Assuntos
Carbacol , Modelos Animais de Doenças , Hiperalgesia , Neuralgia , Nervo Sural , Animais , Hiperalgesia/tratamento farmacológico , Masculino , Neuralgia/tratamento farmacológico , Neuralgia/etiologia , Carbacol/farmacologia , Ligadura , Camundongos , Nervo Sural/efeitos dos fármacos , Agonistas Colinérgicos/farmacologia , Limiar da Dor/efeitos dos fármacos , Morfina/farmacologia , Analgésicos/farmacologia , Medição da Dor , Medula Espinal/efeitos dos fármacos , Acetilcolina/metabolismo
12.
Neurol Sci ; 34(10): 1735-44, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23389808

RESUMO

About 85 % of patients with generalized myasthenia gravis (MG) have anti-nicotinic acetylcholine receptor (nAChR) antibodies in their sera (seropositive MG; SPMG). The other 15 % (seronegative MG; SNMG) are also considered to have antibody-mediated disease, but the nature of the antibodies in SNMG is not fully understood. We investigated the effect of sera from patients with MG on spontaneous muscle action potentials and acetylcholine (ACh)-induced potentials, and we examined the localization of epitopes recognized by SPMG sera or SNMG sera. SPMG sera and SNMG sera inhibited spontaneous muscle action potentials and ACh-induced potentials in the spinal-muscle co-culture system. However, spontaneous muscle action potentials and ACh-induced potentials in the neuromuscular junctions were strongly blocked by SPMG serum, whereas they were weakly blocked by SNMG serum. Both types of sera reacted strongly with the neuromuscular junctions in normal rat muscles, as shown by double immunostaining with serum and α-bungarotoxin. The SPMG epitope remained in the neuromuscular junctions, whereas that of SNMG disappeared after denervation of the sciatic nerve. Therefore, we suggest that the skeletal muscle weakness in SNMG may be due to an interaction with presynaptic neuromuscular transmission and nAChR.


Assuntos
Miastenia Gravis/sangue , Junção Neuromuscular/efeitos dos fármacos , Acetilcolina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Adulto , Idoso , Animais , Anticorpos/farmacologia , Bungarotoxinas/imunologia , Agonistas Colinérgicos/farmacologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/fisiologia , Miastenia Gravis/imunologia , Junção Neuromuscular/imunologia , Ratos , Receptores Proteína Tirosina Quinases/imunologia , Receptores Colinérgicos/imunologia , Medula Espinal/citologia , Adulto Jovem
13.
Muscle Nerve ; 46(5): 705-10, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23055311

RESUMO

INTRODUCTION: We investigated the localization of a ganglioside, N-acetylgalactosaminyl GD1a (GalNAc-GD1a), in peripheral nerves with an IgG anti-GalNAc-GD1a antibody, which was produced in rabbits immunized with GalNAc-GD1a. METHODS: Teased fibers from ventral and dorsal roots and hemidiaphragm sections of rats were assessed using fluorescent double- and triple-labeling methods. RESULTS: The nodal and paranodal regions of teased fibers from ventral roots were immunostained with IgG anti-GalNAc-GD1a antibodies. After collagenase treatment, no staining was seen with IgG anti-GalNAc-GD1a or anti-NF200 antibodies, whereas α-bungarotoxin selectively stained nerve terminals. In cross-sectional and longitudinal sections of rat hemidiaphragm, IgG anti-GalNAc-GD1a antibodies overlapped with α-BuTx and anti-NF200 antibodies, indicating that GalNAc-GD1a is localized to the nerve terminal. IgG anti-GalNAc-GD1a antibody staining also overlapped with that of AChR clusters and syntaxin-positive presynaptic nerve terminals. CONCLUSION: GalNAc-GD1 is localized in both pre- and postsynaptic nerve terminals of neuromuscular junctions.


Assuntos
Sítios de Ligação de Anticorpos , Diafragma/metabolismo , Gangliosídeos/imunologia , Gangliosídeos/metabolismo , Imunoglobulina G/metabolismo , Junção Neuromuscular/metabolismo , Animais , Diafragma/química , Diafragma/imunologia , Feminino , Junção Neuromuscular/química , Junção Neuromuscular/imunologia , Ligação Proteica/imunologia , Coelhos , Ratos , Ratos Wistar
14.
Oncol Res ; 20(4): 179-85, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23461065

RESUMO

No established supportive therapy to prevent and treat chemotherapy-induced peripheral neuropathy (PN) is available. Minimizing the severity of PN is therefore critical in clinical use. We aimed to determine when and how often PN occurs in association with paclitaxel plus carboplatin (PC therapy), a regimen used to treat non-small cell lung cancer, and factors that exacerbate this condition. Patients who received PC therapy for non-small cell lung cancer at the Japanese Foundation for Cancer Research, Cancer Institute Hospital, between May 20, 2009, and November 30, 2010, were included. PN was evaluated by the study pharmacist using specific questions based on the Common Terminology Criteria for Adverse Events Version 3.0. Univariate analysis was used to compare a group with no, Grade 1, or Grade 2 PN (non-serious) and a group with Grade 3 PN (serious). Analyses were conducted using the Cox proportional hazard model with patient characteristics having p < or = 0.20 when assessed as independent variables. Of 50 patients, 38 (76.0%) developed PN by day 6 of the first course of anticancer treatment. Grade 3 PN had an incidence of 25.0% in the fourth course. In multivariate analysis with the Cox proportional hazard model, pack-year [hazard ratio = 1.029; 95% confidence interval (CI): 1.009-1.050, p = 0.005] and creatinine clearance (hazard ratio = 0.957; 95% CI: 0.920-0.996, p = 0.031) were significant factors. A high pack-year and a low creatinine clearance exacerbated PN in patients treated with PC. PN must be carefully evaluated in patients with exacerbating factors.


Assuntos
Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carboplatina/efeitos adversos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Paclitaxel/efeitos adversos , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais
15.
J Pharmacol Sci ; 120(3): 187-95, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23090716

RESUMO

Peripheral neuropathic pain is a serious side effect of paclitaxel treatment. However, the mechanism of this paclitaxel-induced neuropathic pain is unknown. In this study, we investigated the effects of paclitaxel on the voltage-dependent calcium channel (VDCC) current in rat dorsal root ganglion (DRG) neurons using the whole-cell patch clamp technique. Behavioral assessment using von Frey filament stimuli showed that 2 and 4 mg/kg paclitaxel treatment induced mechanical allodynia/hyperalgesia. Paclitaxel-induced mechanical hyperalgesia was significantly inhibited by gabapentin (100 mg/kg). Using the patch clamp method, we observed that paclitaxel (4 mg/kg) treatment significantly increased the VDCC current in small- and medium-diameter DRG neurons. Moreover, paclitaxel-induced increase in the VDCC current in medium-diameter DRG neurons was completely inhibited by 10 and 100 µM gabapentin. Similar effects in small-diameter DRG neurons were only seen with 100 µM gabapentin. Western blotting revealed that paclitaxel increased protein levels of the VDCC subunit α2δ-1 (Ca(v)α2δ-1) in DRG neurons. Immunohistochemistry showed that paclitaxel treatment increased Ca(v)α2δ-1 protein expression in DRG neurons. Thus, paclitaxel treatment increases the VDCC current in small- and medium-diameter DRG neurons and upregulates Ca(v)α2δ-1. The antihyperalgesic action of gabapentin may be due to inhibition of paclitaxel-induced increases in the VDCC current in DRG neurons.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologia , Agonistas dos Canais de Cálcio/farmacologia , Gânglios Espinais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Paclitaxel/farmacologia , Regulação para Cima/efeitos dos fármacos , Animais , Antineoplásicos Fitogênicos/efeitos adversos , Comportamento Animal/efeitos dos fármacos , Agonistas dos Canais de Cálcio/efeitos adversos , Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/química , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L , Tamanho Celular , Células Cultivadas , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Hiperalgesia/induzido quimicamente , Hiperalgesia/tratamento farmacológico , Hiperalgesia/metabolismo , Masculino , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neuralgia/induzido quimicamente , Neuralgia/tratamento farmacológico , Neuralgia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Síndromes Neurotóxicas/tratamento farmacológico , Síndromes Neurotóxicas/metabolismo , Paclitaxel/efeitos adversos , Ratos , Ratos Wistar
16.
Biol Pharm Bull ; 35(1): 127-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22223349

RESUMO

Cardanol (ginkgol) extracted from Ginkgo biloba leaves and cashew nutshell liquid enhances the growth of NSC-34 immortalized motor neuron-like cells and, when chronically administered to young rats, improves working memory-related learning ability as assessed by eight-arm radial maze tasks. These findings suggest that cardanol is one of the components in Ginkgo biloba leaves that improves cognitive learning ability.


Assuntos
Anacardium/química , Ginkgo biloba/química , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Memória de Curto Prazo/efeitos dos fármacos , Fenóis/farmacologia , Extratos Vegetais/farmacologia , Animais , Masculino , Nozes , Fenóis/administração & dosagem , Fenóis/uso terapêutico , Fitoterapia , Extratos Vegetais/uso terapêutico , Folhas de Planta , Ratos , Ratos Wistar
17.
J Neurochem ; 112(4): 913-23, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19968754

RESUMO

The Kv1.4 potassium channel is reported to exhibit higher cell surface expression than the Kv1.1 potassium channel when expressed as a homomer in cell lines. Kv1.4 also shows highly efficient trans-Golgi glycosylation whereas Kv1.1 is not glycosylated. The surface expression and glycosylation of Kv1.2 is intermediate between those of Kv1.1 and Kv1.4. Amino acid determinants controlling the surface expression of Kv1 channels were localized to the highly conserved pore region and both positive and negative determinants of Kv1.1 and Kv1.4 trafficking have been reported. In this study, we analyzed the effect of substituting amino acids in the pore region of Kv1.2 with the corresponding amino acid present in Kv1.1 or Kv1.4 on glycosylation and trafficking of Kv1.2. Mutations in the outer pore region of Kv1.2 of Arg(354) to Pro (corresponding to Kv1.4) and to Ala (corresponding to Kv1.1) enhanced and reduced, respectively, cell surface expression of Kv1.2. Mutations in a different outer pore region of Val(381) to Lys (Kv1.4) and Tyr (Kv1.1) both reduced the cell surface expression. In contrast, mutation in the deep pore region of Ser(371) to Thr (Kv1.4) markedly enhanced cell surface expression. These results suggest that the cell surface expression of Kv1.2 is regulated by specific amino acids in the pore region in a similar manner to Kv1.1 and Kv1.4, and that the cell surface expression of Kv1.2, a channel intermediate between Kv1.1 and Kv1.4, can be attributed to these specific residues.


Assuntos
Aminoácidos/metabolismo , Membrana Celular/metabolismo , Regulação da Expressão Gênica/genética , Canal de Potássio Kv1.2/química , Canal de Potássio Kv1.2/fisiologia , Aminoácidos/genética , Animais , Células CHO , Membrana Celular/genética , Cricetinae , Cricetulus , Regulação da Expressão Gênica/efeitos dos fármacos , Glicosilação , Ativação do Canal Iônico/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/genética , Modelos Moleculares , Dados de Sequência Molecular , Técnicas de Patch-Clamp/métodos , Transporte Proteico/genética , Transfecção/métodos , Rede trans-Golgi/genética , Rede trans-Golgi/metabolismo
18.
J Neurochem ; 113(2): 351-62, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20132479

RESUMO

Recent etiological studies have revealed that molecular mimicry between the lipo-oligosaccharide (LOS) component of Campylobacter jejuni and gangliosides of peripheral nervous system plays an important role in the pathogenesis of Guillain-Barré syndrome (GBS). Previously, we demonstrated GD3 ganglioside molecular mimicry in a model of GBS in Lewis rats by sensitization with GD3-like LOS (LOS(GD3)) from C. jejuni. Since the neuropathophysiological consequences were due largely to the anti-GD3-like antibodies, we subsequently focused our effort upon eliminating the pathogenic antibodies using several strategies to mimic GD3 in this model. Here, we have validated this strategy by the use of peptide glycomimics based on epitopic mimicry between carbohydrates and peptides. We treated rats by i.p. administration of phage-displayed GD3-like peptides. One GD3-like peptide (P(GD3)-4; RHAYRSMAEWGFLYS) induced in treated rats a remarkable restoration of motor nerve functions, as evidenced by improved histopathology, rotarod performance, and motor nerve conduction velocity. P(GD3)-4 effectively decreased the titer of anti-GD3/anti-LOS(GD3) antibodies and ameliorated peripheral nerve dysfunction in the sera of treated rats. The data suggest that peptide glycomimics of ganglioside may be potential powerful reagents for therapeutic intervention in GBS by neutralizing specific pathogenic anti-ganglioside antibodies.


Assuntos
Desenho de Fármacos , Gangliosídeos/química , Gangliosídeos/uso terapêutico , Neurite (Inflamação)/tratamento farmacológico , Peptídeos/uso terapêutico , Animais , Autoanticorpos/metabolismo , Peso Corporal/efeitos dos fármacos , Técnicas de Cocultura/métodos , Reações Cruzadas/fisiologia , Modelos Animais de Doenças , Feminino , Gangliosídeos/imunologia , Lipopolissacarídeos , Microscopia Eletrônica de Transmissão/métodos , Mimetismo Molecular , Atividade Motora/efeitos dos fármacos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Condução Nervosa/efeitos dos fármacos , Condução Nervosa/fisiologia , Neurite (Inflamação)/induzido quimicamente , Neurite (Inflamação)/patologia , Neurite (Inflamação)/fisiopatologia , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/fisiopatologia , Oligossacarídeos/uso terapêutico , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/fisiopatologia , Ratos , Ratos Endogâmicos Lew , Fatores de Tempo
19.
Neurochem Res ; 34(1): 149-57, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18478327

RESUMO

To investigate the pathophysiological mechanisms of immune-mediated peripheral neuropathies, we studied the effects of sera from patients with Guillain-Barré syndrome (GBS) on the Cav2.1 voltage-dependent calcium channel (VDCC) current in Purkinje cells. Using the whole-cell recording technique, Cav2.1 VDCC current was measured in cerebellar Purkinje cells in the presence of serum from GBS patients with acute motor axonal neuropathy (AMAN) or acute inflammatory demyelinating polyneuropathy (AIDP). The AMAN patient sera significantly inhibited the Cav2.1 VDCC current compared with healthy volunteer sera, and this inhibition was fully reversible by washing out the AMAN serum. Similarly, IgG purified from AMAN sera also inhibited the Cav2.1 VDCC current. However, the activation and inactivation kinetics of the Cav2.1 VDCC currents were not affected by serum from an AMAN patient. Moreover, the VDCC current of Purkinje cells was also inhibited by IgG anti-GM1 monoclonal antibody (anti-GM1 mAb). In an immunocytochemical study using double fluorescence staining, Purkinje cells were stained by monoclonal IgG anti-GM1 mAb. In contrast, AIDP patient and healthy volunteer sera did not affect the Cav2.1 VDCC current. These results suggest that in some case of GBS, particularly of AMAN patients with IgG anti-GM1 mAb, muscle weakness may be induced by dysfunction of Cav2.1 VDCC functioning at the motor nerve terminals.


Assuntos
Canais de Cálcio Tipo N/fisiologia , Canais de Cálcio/fisiologia , Síndrome de Guillain-Barré/sangue , Adulto , Canais de Cálcio/efeitos dos fármacos , Canais de Cálcio Tipo N/efeitos dos fármacos , Feminino , Síndrome de Guillain-Barré/fisiopatologia , Humanos , Imunoglobulina G/farmacologia , Masculino , Pessoa de Meia-Idade , Doenças do Sistema Nervoso Periférico/fisiopatologia , Polirradiculoneuropatia/imunologia , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/fisiopatologia , Células de Purkinje/efeitos dos fármacos , Células de Purkinje/fisiologia
20.
J Neurosci Res ; 86(15): 3359-74, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18627035

RESUMO

An infecting strain VLA2/18 of Campylobacter jejuni was obtained from an individual with campylobacteriosis and used to prepare chicken sera by experimental infection to investigate the role of serum anti-ganglioside antibodies in Guillain-Barré syndrome. Both sera of the patient and chicken contained anti-ganglioside antibodies and anti-Lipid A (anti-Kdo2-Lipid A) antibodies directed against the lipid A portion of the bacterial lipooligosaccharide. The anti-Kdo2-Lipid A activities inhibited voltage-gated Na (Nav) channel of NSC-34 cells in culture. We hypothesized that anti-Kdo2-Lipid A antibody acts on the functional inhibition of Nav1.4. To test this possibility, a rabbit peptide antibody (anti-Nav1.4 pAb) against a 19-mer peptide (KELKDNHILNHVGLTDGPR) on the alpha subunit of Nav1.4 was produced. Anti-Nav1.4 pAb was cross-reactive to Kdo2-Lipid A. Anti-Kdo2-lipid A antibody activity in the chicken serum was tested for the Na(+) current inhibition in NSC-34 cells in combination with mu-Conotoxin and tetrodotoxin. Contrary to our expectations, the anti-Kdo2-Lipid A antibody activity was extended to Nav channels other than Nav1.4. By overlapping structural analysis, it was found that there might be multiple peptide epitopes containing certain dipeptides showing a structural similarity with v-Lipid A. Thus, our study suggests the possibility that there are multiple epitopic peptides on the extracellular domains of Nav1.1 to 1.9, and some of them may represent target sites for anti-Kdo2-Lipid A antibody, to induce neurophysiological changes in GBS by disrupting the normal function of the Nav channels.


Assuntos
Autoanticorpos/imunologia , Autoantígenos/imunologia , Infecções por Campylobacter/imunologia , Campylobacter jejuni/imunologia , Lipídeo A/imunologia , Canais de Sódio/imunologia , Sequência de Aminoácidos , Animais , Especificidade de Anticorpos , Western Blotting , Galinhas , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Epitopos de Linfócito B/imunologia , Gangliosídeos/imunologia , Humanos , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Peptídeos/imunologia , Isoformas de Proteínas/imunologia , Canais de Sódio/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA