Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Brain Behav Immun ; 115: 295-307, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37884161

RESUMO

GV1001 protects neural cells from amyloid-ß (Aß) toxicity and other stressors in in vitro studies and demonstrates clinically beneficial effects in patients with moderate to severe Alzheimer's disease (AD). Here, we investigated the protective effects and mechanism of action of GV1001 in triple transgenic AD (3xTg-AD) mice. We found that GV1001 improved memory and cognition in middle- and old-aged 3xTg-AD mice. Additionally, it reduced Aß oligomer and phospho-tau (Ser202 and Thr205) levels in the brain, and mitigated neuroinflammation by promoting a neuroprotective microglial and astrocyte phenotype while diminishing the neurotoxic ones. In vitro, GV1001 bound to gonadotropin releasing hormone receptors (GnRHRs) with high affinity. Levels of cyclic adenosine monophosphate, a direct downstream effector of activated GnRHRs, increased after GV1001 treatment. Furthermore, inhibition of GnRHRs blocked GV1001-induced effects. Thus, GV1001 might improve cognitive and memory functions of 3xTg-AD mice by suppressing neuroinflammation and reducing Aß oligomers levels and phospho-tau by activating GnRHRs and their downstream signaling pathways.


Assuntos
Doença de Alzheimer , Humanos , Camundongos , Animais , Pessoa de Meia-Idade , Idoso , Doença de Alzheimer/metabolismo , Camundongos Transgênicos , Receptores LHRH , Doenças Neuroinflamatórias , Proteínas tau/genética , Proteínas tau/metabolismo , Peptídeos beta-Amiloides/metabolismo , Hormônio Liberador de Gonadotropina , Modelos Animais de Doenças
2.
Medicine (Baltimore) ; 101(35): e30464, 2022 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-36107550

RESUMO

RATIONALE: Coronavirus disease 2019 (COVID-19) has become a global pandemic and COVID-19-associated anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis may occur through an immune-mediated pathomechanism. PATIENT CONCERNS: A 21-year-old woman with a history of COVID-19 presented to our hospital with memory decline and psychiatric symptoms. DIAGNOSIS: The patient was diagnosed with anti-NMDAR encephalitis. INTERVENTION: Intravenous methylprednisolone (1 g/day over 5 days) followed by immunoglobulin (0.4 g/kg/day over 5 days) were administered. The patient underwent laparoscopic salpingo-oophorectomy to remove an ovarian teratoma. OUTCOMES: The patient was discharged with sequelae of short-term memory impairment, without other neuropsychiatric symptoms. LESSONS: Cases of previously reported anti-NMDAR encephalitis with COVID-19 were reviewed and compared with the present case. Clinicians should be aware of the occurrence of anti-NMDAR encephalitis in patients who present with neuropsychiatric complaints during or after exposure to COVID-19. Further studies are required to determine the causal relationship between the 2 diseases and predict the prognosis of anti-NMDAR encephalitis after COVID-19 exposure.


Assuntos
Encefalite Antirreceptor de N-Metil-D-Aspartato , COVID-19 , Adulto , Encefalite Antirreceptor de N-Metil-D-Aspartato/complicações , Encefalite Antirreceptor de N-Metil-D-Aspartato/diagnóstico , COVID-19/complicações , Feminino , Humanos , Imunoglobulinas , Metilprednisolona/uso terapêutico , Receptores de N-Metil-D-Aspartato , Adulto Jovem
4.
Medicine (Baltimore) ; 99(17): e19882, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32332660

RESUMO

RATIONALE: Tolosa-Hunt syndrome (THS) is rare condition characterized by painful ophthalmoplegia that usually responds well to corticosteroid. About a half of THS patients experience recurrence within intervals of months to years from initial presentation. Recurrence is more common in younger patients, and can be ipsilateral, contralateral, or bilateral. Cyclosporine, azathioprine, methotrexate, mycophenolate mofetil, infliximab, and radiotherapy can be considered as second-line treatment. However, there is insufficient evidence for treatments preventing recurrence of THS. PATIENT CONCERNS: We experienced two patients with THS that recurred twice while tapering or after ceasing corticosteroid administration. DIAGNOSIS: Both patients were diagnosed as recurrent THS. INTERVENTIONS: Methotrexate was treated with a combination of corticosteroid after THS recurred twice with corticosteroid therapy alone. OUTCOMES: After adding methotrexate to the steroid regimen, their symptoms were successfully regulated and ceased to recur LESSONS:: These cases add to the evidence for the use of methotrexate as a second-line therapeutic agent for those patients with recurrent THS attacks. Further studies are in need to prove the risk and benefits of second-line treatments in THS.


Assuntos
Metotrexato/uso terapêutico , Síndrome de Tolosa-Hunt/tratamento farmacológico , Resultado do Tratamento , Adulto , Feminino , Humanos , Imageamento por Ressonância Magnética/métodos , Recidiva , Esteroides/uso terapêutico , Síndrome de Tolosa-Hunt/fisiopatologia
5.
Sci Rep ; 9(1): 15717, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31673096

RESUMO

Amlodipine, a L-type calcium channel blocker, has been reported to have a neuroprotective effect in brain ischemia. Mitochondrial calcium overload leads to apoptosis of cells in neurologic diseases. We evaluated the neuroprotective effects of amlodipine camsylate (AC) on neural stem cells (NSCs) injured by oxygen glucose deprivation (OGD) with a focus on mitochondrial structure and function. NSCs were isolated from rodent embryonic brains. Effects of AC on cell viability, proliferation, level of free radicals, and expression of intracellular signaling proteins were assessed in OGD-injured NSCs. We also investigated the effect of AC on mitochondrial structure in NSCs under OGD by transmission electron microscopy. AC increased the viability and proliferation of NSCs. This beneficial effect of AC was achieved by strong protection of mitochondria. AC markedly enhanced the expression of mitochondrial biogenesis-related proteins and mitochondrial anti-apoptosis proteins. Together, our results indicate that AC protects OGD-injured NSCs by protecting mitochondrial structure and function. The results of the present study provide insight into the mechanisms underlying the protective effects of AC on NSCs.


Assuntos
Anlodipino/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Cálcio/metabolismo , Glucose/metabolismo , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ativação Enzimática , Humanos , Transdução de Sinais
6.
Exp Neurobiol ; 28(5): 628-641, 2019 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-31698554

RESUMO

Leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) has been reported to play critical roles in the proliferation of various cancer cells. However, the roles of LGR5 in brain tumors and the specific intracellular signaling proteins directly associated with it remain unknown. Expression of LGR5 was first measured in normal brain tissue, meningioma, and pituitary adenoma of humans. To identify the downstream signaling pathways of LGR5, siRNA-mediated knockdown of LGR5 was performed in SH-SY5Y neuroblastoma cells followed by proteomics analysis with 2-dimensional polyacrylamide gel electrophoresis (2D-PAGE). In addition, the expression of LGR5-associated proteins was evaluated in LGR5-inhibited neuroblastoma cells and in human normal brain, meningioma, and pituitary adenoma tissue. Proteomics analysis showed 12 protein spots were significantly different in expression level (more than two-fold change) and subsequently identified by peptide mass fingerprinting. A protein association network was constructed from the 12 identified proteins altered by LGR5 knockdown. Direct and indirect interactions were identified among the 12 proteins. HSP 90-beta was one of the proteins whose expression was altered by LGR5 knockdown. Likewise, we observed decreased expression of proteins in the hnRNP subfamily following LGR5 knockdown. In addition, we have for the first time identified significantly higher hnRNP family expression in meningioma and pituitary adenoma compared to normal brain tissue. Taken together, LGR5 and its downstream signaling play critical roles in neuroblastoma and brain tumors such as meningioma and pituitary adenoma.

7.
Mol Neurobiol ; 56(4): 2964-2977, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30073508

RESUMO

Oxygen and glucose (OGD) deprivation is one of the most important pathogenic mechanisms in cerebral infarction and is widely used as an in vitro model for ischemic stroke. OGD also damages neural stem cells (NSCs), which are important in brain recovery after cerebral infarction. To enhance recovery, there have been many studies aimed at determining methods to protect NSCs after stroke. Because atorvastatin has diverse protective effects on neural cells, we studied whether it could rejuvenate NSCs injured by OGD. Primary cultured NSCs were exposed to OGD for 8 h, and the main characteristics of stem cells, such as survival, proliferation, migration, and differentiation, were evaluated to confirm the effect of OGD on NSCs. Next, cells were treated with various concentrations of atorvastatin with exposure to OGD for 8 h to confirm whether it could rejuvenate NSCs. OGD significantly affected the survival, proliferation, migration, and differentiation of NSCs. However, treatment with atorvastatin meaningfully restored survival, proliferation, migration, and differentiation of NSCs. These beneficial effects of atorvastatin were blocked by treatment with either a PI3K inhibitor or an ERK inhibitor. In conclusion, OGD damages NSCs and causes them to lose the main characteristics of stem cells so that they cannot contribute to brain recovery after cerebral infarction. However, treatment with atorvastatin after cerebral infarction can effectively rejuvenate NSCs through activating the PI3K and ERK pathways to aid in brain regeneration.


Assuntos
Atorvastatina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Glucose/deficiência , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Neurais/patologia , Neurônios/patologia , Oxigênio/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley
9.
Biomaterials ; 155: 80-91, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29169040

RESUMO

In vivo tracking of transplanted stem cells has been a central aim of stem cell therapy. Although many tracking systems have been introduced, no method has yet been validated for clinical applications. We developed a novel sophisticated peptide (GV1001) that mimics hTERT (human telomerase reverse transcriptase) and analysed its ability to track and protect stem cells after transplantation. Ferrocenecarboxylic acid-conjugated GV1001 (Fe-GV1001) efficiently penetrated stem cells with no adverse effects. Moreover, Fe-GV1001 improved the viability, proliferation, and migration of stem cells under hypoxia. After Fe-GV1001-labelled stem cells were transplanted into the brains of rats after stroke, the labelled cells were easily tracked by MRI. Our findings indicate that Fe-GV1001 can be used for the in vivo tracking of stem cells after transplantation into the brain and can improve the efficacy of stem cell therapy by sustaining and enhancing stem cell characteristics under disease conditions.


Assuntos
Compostos Ferrosos/química , Fragmentos de Peptídeos/química , Telomerase/química , Animais , Humanos , Células-Tronco Mesenquimais/efeitos dos fármacos , Metalocenos , Células-Tronco Neurais/efeitos dos fármacos , Fragmentos de Peptídeos/farmacologia , Ratos , Transplante de Células-Tronco
10.
Mol Neurobiol ; 53(10): 6644-6652, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-26643543

RESUMO

Deprivation of oxygen and glucose is the main cause of neuronal cell death during cerebral infarction and can result in severe morbidity and mortality. In general, the neuroprotective therapies that are applied after ischemic stroke have been unsuccessful, despite many investigations. Acetyl-L-carnitine (ALCAR) plays an important role in mitochondrial metabolism and in modulating the coenzyme A (CoA)/acyl-CoA ratio. We investigated the protective effects of ALCAR against oxygen-glucose deprivation (OGD) in neural stem cells (NSCs). We measured cell viability, proliferation, apoptosis, and intracellular signaling protein levels after treatment with varying concentrations of ALCAR under OGD for 8 h. ALCAR protected NSCs against OGD by reducing apoptosis and restoring proliferation. Its protective effects are associated with increases in the expression of survival-related proteins, such as phosphorylated Akt (pAkt), phosphorylated glycogen synthase kinase 3b (pGSK3b), B cell lymphoma 2 (Bcl-2), and Ki-67 in NSCs that were injured by OGD. ALCAR also reduced the expression of death-related proteins, such as Bax, cytosolic cytochrome C, cleaved caspase-9, and cleaved caspase-3. We concluded that ALCAR exhibits neuroprotective effects against OGD-induced damage to NSCs by enhancing the expression of survival signals and decreasing that of death signals.


Assuntos
Acetilcarnitina/farmacologia , Glucose/deficiência , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Fármacos Neuroprotetores/farmacologia , Oxigênio/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Ratos , Transdução de Sinais/efeitos dos fármacos
11.
J Stroke Cerebrovasc Dis ; 24(3): 704-10, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25601176

RESUMO

BACKGROUND: Pre-existing silent brain infarctions (SBIs) have been reported to be associated with better outcomes after first-ever symptomatic ischemic stroke, although the mechanism of this remains unclear. We investigated the association between SBIs, outcomes of acute lacunar infarction, and biomarkers including vascular endothelial growth factor (VEGF), stromal cell-derived factor-1α (SDF-1α), macrophage migration inhibitory factor (MIF), and high-mobility group box-1 (HMGB1). METHODS: A total of 68 consecutive patients diagnosed with first-ever lacunar infarction (<20 mm) within 24 hours of symptom onset were included in this study. Clinical, laboratory, and imaging data were obtained. Plasma levels of VEGF, SDF-1α, MIF, and HMGB1 were assessed using Enzyme-Linked Immunosorbent Assay kits. RESULTS: SBIs were noted in 31 of the 68 patients. Although the initial National Institutes of Health Stroke Scale scores were not related with the presence of SBIs (P = .313), patients with SBIs had better outcomes at 3 months (P = .029). Additionally, plasma VEGF levels were higher (P = .035) and SDF-1α levels were lower (P < .001) in patients with SBIs. Logistic regression analysis indicated that VEGF and SDF-1α were independently associated with the presence of SBIs. CONCLUSIONS: SBIs are associated with favorable outcomes in patients with first-ever acute lacunar infarction and higher levels of VEGF, and lower levels of SDF-1α in these patients may contribute to their more favorable prognosis.


Assuntos
Quimiocina CXCL12/sangue , Acidente Vascular Cerebral Lacunar/sangue , Fator A de Crescimento do Endotélio Vascular/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/sangue , Distribuição de Qui-Quadrado , Avaliação da Deficiência , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Modelos Logísticos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Razão de Chances , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco , Acidente Vascular Cerebral Lacunar/diagnóstico , Acidente Vascular Cerebral Lacunar/etiologia , Acidente Vascular Cerebral Lacunar/terapia , Fatores de Tempo , Regulação para Cima
12.
Mol Neurobiol ; 47(2): 757-69, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23254998

RESUMO

Phosphatidylinositol 3-kinase (PI3K) plays several important roles in neuronal survival. Activation of the pathway is essential for the neuroprotective mechanisms of materials that shield neuronal cells from many stressful conditions. However, there have been no reports to date about the effect of the direct activation of the pathway in hypoxic injury of neuronal cells. We investigated whether the direct activation of the PI3K pathway inhibits neuronal cell death induced by hypoxia. Primary cultured cortical neurons (PCCNs) were exposed to hypoxic conditions (less than 1 mol% O2) and/or treated with PI3K activator. Hypoxia reduced the viability of PCCNs in a time-dependent manner, but treatment with PI3K significantly restored viability in a concentration-dependent manner. Among the signaling proteins involved in the PI3K pathway, those associated with survival, including Akt and glycogen synthase kinase-3ß, were decreased shortly after exposure to hypoxia and those associated with cell death, including BAX, apoptosis-induced factor, cytochrome c, caspase-9, caspase-3, and poly(ADP-ribose) polymerase (PARP), were increased. However, treatment with PI3K activator normalized the expression levels of those signaling proteins. PARP activity and levels of ATP and NAD(+) altered by hypoxia were also normalized with direct PI3K activation. All these findings suggest that direct and early activation is important for protecting neuronal cells from hypoxic injury.


Assuntos
Trifosfato de Adenosina/metabolismo , Córtex Cerebral/metabolismo , NAD/metabolismo , Neurônios/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Regulação para Cima/fisiologia , Animais , Hipóxia Celular/fisiologia , Células Cultivadas , Córtex Cerebral/patologia , Regulação para Baixo/fisiologia , Ativação Enzimática/fisiologia , Neurônios/patologia , Fosfatidilinositol 3-Quinase/fisiologia , Poli(ADP-Ribose) Polimerases/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
13.
Brain Res ; 1478: 64-73, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-23046589

RESUMO

Recanalization and secondary prevention are the main therapeutic strategies for acute ischemic stroke. Neuroprotective therapies have also been investigated despite unsuccessful clinical results. Coenzyme Q10 (CoQ10), which is an essential cofactor for electron transport in mitochondria, is known to have an antioxidant effect. We investigated the protective effects of CoQ10 against hypoxia in neural stem cells (NSCs). We measured cell viability and levels of intracellular signaling proteins after treatment with several concentrations of CoQ10 under hypoxia-reperfusion. CoQ10 protected NSCs against hypoxia-reperfusion in a concentration-dependent manner by reducing growth inhibition and inhibiting free radical formation. It increased the expression of a number of survival-related proteins such as phosphorylated Akt (pAkt), phosphorylated glycogen synthase kinase 3-ß (pGSK3-ß), and B-cell lymphoma 2 (Bcl-2) in NSCs injured by hypoxia-reperfusion and reduced the expression of death-related proteins such as cleaved caspase-3. We conclude that CoQ10 has effects against hypoxia-reperfusion induced damage to NSCs by enhancing survival signals and decreasing death signals.


Assuntos
Antioxidantes/farmacologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Western Blotting , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Radicais Livres/metabolismo , Marcação In Situ das Extremidades Cortadas , Células-Tronco Neurais/citologia , Ratos , Traumatismo por Reperfusão/metabolismo , Ubiquinona/farmacologia
14.
J Clin Neurosci ; 18(11): 1567-9, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21868235

RESUMO

A cavernoma is a vascular malformation in the central nervous system. Brainstem cavernoma are relatively common and induce variable neurological symptoms. A 19-year-old woman visited our hospital with complaints of continuous dizziness. On a neurological examination, continuous conjugated upbeat nystagmus was observed in the primary position of gaze. A brain CT scan and MRI showed focal hemorrhagic signals in the central caudal medulla caused by a cavernoma. The spontaneous upbeat nystagmus disappeared gradually. To our knowledge, this is the first report of a probable cavernoma in the medulla oblongata presenting with upbeat nystagmus only.


Assuntos
Neoplasias Encefálicas/complicações , Hemangioma Cavernoso do Sistema Nervoso Central/complicações , Bulbo/patologia , Nistagmo Patológico/etiologia , Neoplasias Encefálicas/patologia , Feminino , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Humanos , Nistagmo Patológico/patologia , Adulto Jovem
15.
Neurotoxicology ; 32(6): 879-87, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21683736

RESUMO

The neurotoxicity of L-3,4-dihydroxyphenylalanine (L-DOPA), one of the most important drugs for the treatment of Parkinson's disease, still remains controversial, although much more data on L-DOPA neurotoxicity have been presented. Considering the well known neuroprotective effects of erythropoietin (EPO), the inhibitory effects of EPO on L-DOPA neurotoxicity need to be evaluated. Neuronally differentiated PC12 (nPC12) cells were treated with different concentrations of L-DOPA and/or EPO for 24h. Cell viability was evaluated using trypan blue, 4',6-diamidino-2-phenylindole (DAPI) and TUNEL staining, and cell counting. Free radicals and intracellular signaling protein levels were measured with 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and Western blotting, respectively. L-DOPA reduced nPC12 cell viability at higher concentrations, but combined treatment with EPO and L-DOPA significantly restored cell viability. Free radicals and hydroxyl radical levels increased by L-DOPA were decreased after combined treatment of L-DOPA and EPO. Levels of survival-related intracellular signaling proteins decreased in nPC12 cells treated with 200 µM L-DOPA but increased significantly in cells treated with 200µM L-DOPA and 5 µM EPO. However, cleaved caspase-3, a death-related protein, increased in nPC12 cells treated with 200 µM L-DOPA but decreased significantly in cells treated with 200 µM L-DOPA and 5 µM EPO. Pretreatment with LY294002, a phosphatidylinositol 3-kinase inhibitor, prior to combined treatment with EPO and L-DOPA almost completely blocked the protective effects of EPO. These results indicate that EPO can prevent L-DOPA neurotoxicity by activating the PI3K pathway as well as reducing oxidative stress.


Assuntos
Eritropoetina/farmacologia , Levodopa/toxicidade , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Caspase 3/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citoproteção , Relação Dose-Resposta a Droga , Humanos , Radical Hidroxila/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinase/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Ratos , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
16.
J Clin Neurosci ; 18(8): 1128-9, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21658952

RESUMO

Sparganosis is a rare parasitic infection caused by the migrating plerocercoid larva of the tapeworm Spirometra mansoni that rarely affects the central nervous system. When sparganosis involves the spinal cord, it often affects the thoracic area. Here we report a patient with sparganosis mimicking an intramedullary tumor of the cervical cord. A 51-year-old male presented with slowly progressive paresthesias in all extremities and left-sided motor weakness. MRI of the spinal cord revealed an enhancing lesion from C1 to C4 that was suggestive of an intramedullary tumor. The results of enzyme-linked immunosorbent assays of the cerebrospinal fluid were consistent with infection with Spirometra mansoni, and the larvae were surgically removed. Sparganosis may mimic an intramedullary tumor of the spinal cord and should be considered in the differential diagnosis of a cervical cord mass with an uncertain course.


Assuntos
Esparganose , Neoplasias da Medula Espinal/fisiopatologia , Medula Espinal/patologia , Spirometra/patogenicidade , Animais , Humanos , Masculino , Pessoa de Meia-Idade , Esparganose/etiologia , Esparganose/parasitologia , Esparganose/patologia , Medula Espinal/parasitologia
17.
Brain Res ; 1370: 53-63, 2011 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-21075093

RESUMO

Neurotoxic effects have been suggested for l-3,4-dihydroxyphenylalanine (L-DOPA), while neuroprotective effects have been proposed for statins. We investigated whether certain statins (simvastatin or pitavastatin) could inhibit L-DOPA neurotoxicity. Neuronally-differentiated PC12 (nPC12) cells were treated with L-DOPA and/or statins for 24h, and their viabilities were measured using a cell counting kit, trypan blue staining, and 4',6-diamidino-2-phenylindole (DAPI) staining. Free radical and specific intracellular signaling protein levels were measured with 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and Western blotting, respectively. High concentrations of l-DOPA reduced nPC12 cell viability, but combined treatment with statins restored viability. Treatment with 200 µM L-DOPA increased free radical and hydroxyl radical levels, but combined treatment with 5 µM statins decreased these levels. Survival-related signaling proteins were decreased in nPC12 cells treated with 200 µM L-DOPA, but combined treatment with 5µM statins significantly increased the levels of these proteins. Treatment with 200 µM L-DOPA significantly increased death-related signaling proteins, while combined treatment with 5 µM statins reduced the levels of these proteins. Pretreatment with LY294002, a phosphatidylinositol 3 kinase (PI3K) inhibitor, before combined treatment with statins and L-DOPA almost completely blocked the protective effects of statins. These results indicate that statins reduce L-DOPA neurotoxicity by lowering oxidative stress and by enhancing survival signals and inhibiting death signals via activation of the PI3K pathway.


Assuntos
Antiparkinsonianos/antagonistas & inibidores , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Levodopa/antagonistas & inibidores , Fármacos Neuroprotetores/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Antiparkinsonianos/toxicidade , Sequestradores de Radicais Livres/farmacologia , Sequestradores de Radicais Livres/uso terapêutico , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Levodopa/toxicidade , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Ratos
18.
J Clin Neurol ; 5(3): 146-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19826566

RESUMO

BACKGROUND: Multiple sclerosis (MS) is a demyelinating disease of the central nervous system. Secondary amyloidosis can occur as a complication of chronic systemic inflammatory and infectious diseases. Until now there has been no report of secondary amyloidosis associated with MS. We report herein a case of renal biopsy-proven secondary amyloidosis in a patient with MS. CASE REPORT: A 41-year-old woman with MS was hospitalized due to aggravated quadriparesis and edema in both lower extremities. Laboratory findings showed nephrotic-range proteinuria and hypoalbuminemia. A percutaneous renal biopsy procedure was performed, the results of which revealed secondary amyloid-A-type amyloidosis associated with MS. CONCLUSIONS: This is the first report of secondary amyloidosis associated with MS.

19.
Toxicology ; 265(3): 80-6, 2009 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-19786063

RESUMO

L-3,4-Dihydroxyphenylalanine (L-DOPA) is one of the most important drugs for the treatment of Parkinson's disease (PD). Although neurotoxicity of L-DOPA remains controversial, there are many reports suggesting that L-DOPA causes neuronal death. We investigated whether the neurotoxic effect of L-DOPA could be inhibited by the activation of the phosphatidylinositol 3-kinase (PI3K) pathway. Cell counting kit-8, trypan blue staining, and DAPI staining all showed that L-DOPA decreased nPC12 cell viability at high concentrations. However, combined treatment with the PI3K activator and L-DOPA significantly increased the viability of nPC12 cells when compared with treatment with L-DOPA only. Phosphorylated Akt (Ser473), phosphorylated glycogen synthase kinase-3beta (GSK-3beta) (Ser9), and heat shock transcription factor-1, which are survival-related signaling proteins, were decreased in nPC12 cells treated with 200 microM L-DOPA, but were significantly increased with combined treatment with the PI3K activator in a concentration-dependent manner. However, treatment of L-DOPA significantly increased expressions of cytosolic cytochrome c and cleaved caspase-3, which are death-related signaling proteins, in nPC12 cells, but combined treatment with the PI3K activator reduced those expressions. To confirm whether the effect of the PI3K activator is associated with direct activation of PI3K, LY294002, a PI3K inhibitor, was used to pretreat the nPC12 cells prior to combined treatment with the PI3K activator and L-DOPA. The protective effect of the PI3K activator was almost completely blocked. Together, these results suggest that L-DOPA neurotoxicity can be prevented by PI3K activation.


Assuntos
Inibidores Enzimáticos/farmacologia , Levodopa/toxicidade , Neurônios/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Sobrevivência Celular/efeitos dos fármacos , Corantes/metabolismo , Meios de Cultura Livres de Soro , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Corantes Fluorescentes/metabolismo , Indóis/metabolismo , Células PC12 , Ratos , Azul Tripano/metabolismo
20.
J Neurochem ; 111(1): 90-100, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19650875

RESUMO

We investigated the neuroprotective effect and mechanisms of action of cilnidipine, a long-acting, second-generation 1,4-dihydropyridine inhibitor of L- and N-type calcium channels, in PC12 cells that were neuronally differentiated by treatment with nerve growth factor (nPC12 cells). To evaluate the effect of cilnidipine on viability, nPC12 cells were treated with several concentrations of this drug before performing viability assays. Free radical levels and intracellular signaling proteins were measured with the fluorescent probe, 2',7'-dichlorodihydrofluorescein diacetate and western blotting, respectively. Cell viability was not affected by low concentrations of cilnidipine up to 150 microM, but it was slightly decreased at 200 microM cilnidipine. Following H(2)O(2) exposure, the viability of nPC12 cells decreased significantly; however, treatment with cilnidipine increased the viability of H(2)O(2)-injured nPC12 cells in a concentration-dependent manner. Treatment with H(2)O(2) resulted in a concentration-dependent increase in free radical levels in nPC12 cells, and cilnidipine treatment reduced free radical levels in H(2)O(2)-injured nPC12 cells in a dose-dependent manner. Cilnidipine treatment increased the expression of p85aPI3K (phosphatidylinositol 3-kinase) phosphorylated Akt, phosphorylated glycogen synthase kinase-3 (pGSK-3beta), and heat shock transcription factor (HSTF-1) which are proteins related to neuronal cell survival, and decreased levels of cytosolic cytochrome c, activated caspase 3, and cleaved poly (ADP-ribose) polymerase (PARP), which are associated with neuronal cell death, in H(2)O(2)-injured nPC12 cells. These results indicate that cilnidipine mediates its neuroprotective effects by reducing oxidative stress, enhancing survival signals (e.g., PI3K, phosphorylated Akt, pGSK-3beta, and HSTF-1), and inhibiting death signals from cytochrome c release, caspase 3 activation, and PARP cleavage.


Assuntos
Bloqueadores dos Canais de Cálcio/farmacologia , Di-Hidropiridinas/farmacologia , Sequestradores de Radicais Livres/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Di-Hidropiridinas/química , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Inibidores Enzimáticos/farmacologia , Peróxido de Hidrogênio/farmacologia , Indóis , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Células PC12 , Ratos , Transdução de Sinais/fisiologia , Sais de Tetrazólio , Tiazóis , ômega-Conotoxinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA