Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Med Chem ; 26(25): 4786-4798, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30836908

RESUMO

BACKGROUND: Preeclapmsia (PE) is characterized by early onset symptoms such as elevated blood pressure, proteinuria and edema in the pregnant woman, and may result in seizures in the affected female. Currently, there are no therapeutic drugs available to treat this condition, but there are interventions to regulate the symptoms based on the gestational period of the fetus, although the largely favored option is delivery of the fetus and placenta. OBJECTIVE: A search for biomolecules associated with PE was conducted so as to identify diagnostic markers and therapeutic leads. RESULTS: The literature search resulted in the identification of biomolecules such as Corin and Placental Protein 13 (PP13), among others that are associated with PE. Thereby, giving an insight into the various mechanistic pathways involved in the causation of PE. However, it is also evident that PE cannot be solely attributed to any single mechanism but is due to an interplay of different factors that have led to the development of this disease condition. CONCLUSION: The identified biomarkers would ultimately help in understanding this complex disease and perhaps lead to the discovery of potential effective molecular targets for clinical trials, thereby providing a valuable therapeutic option for affected pregnant women.


Assuntos
Adenosina/uso terapêutico , Pré-Eclâmpsia/tratamento farmacológico , Vasodilatadores/uso terapêutico , Animais , Feminino , Humanos , Pré-Eclâmpsia/diagnóstico , Pré-Eclâmpsia/metabolismo , Gravidez
2.
Neuroradiology ; 58(1): 45-50, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26423907

RESUMO

INTRODUCTION: To describe the unique MRI findings of superior cervical ganglia (SCG) that may help differentiate them from retropharyngeal lymph nodes (RPLNs). METHODS: A retrospective review of post-treatment NPC patients from 1999 to 2012 identified three patients previously irradiated for NPC that were suspected of having recurrent nodal disease in retropharyngeal lymph nodes during surveillance MRI. Subsequent surgical exploration revealed enlarged SCG only; no retropharyngeal nodal disease was found. A cadaveric head specimen was also imaged with a 3T MRI before and after dissection. In addition, SCG were also harvested from three cadaveric specimens and subjected to histologic analysis. RESULTS: The SCG were found at the level of the C2 vertebral body, medial to the ICA. They were ovoid on axial images and fusiform and elongated with tapered margins in the coronal plane. T2-weighted (T2W) signal was hyperintense. No central elevated T1-weighted (T1W) signal was seen within the ganglia in non-fat-saturated sequences to suggest the presence of a fatty hilum. Enhancement after gadolinium was present. A central "black dot" was seen on axial T2W and post-contrast images in two of the three SCG demonstrated. Histology showed the central black line was comprised of venules and interlacing neurites within the central portion of the ganglion. CONCLUSIONS: The SCG can be mistaken for enlarged RPLNs in post-treatment NPC patients. However, there are features which can help differentiate them from RPLNs, preventing unnecessary therapy. These imaging findings have not been previously described.


Assuntos
Neoplasias de Cabeça e Pescoço/diagnóstico , Linfadenopatia/diagnóstico , Imageamento por Ressonância Magnética , Gânglio Cervical Superior/diagnóstico por imagem , Cadáver , Diagnóstico Diferencial , Neoplasias de Cabeça e Pescoço/cirurgia , Humanos , Faringe , Estudos Retrospectivos
3.
Neuroscience ; 311: 398-414, 2015 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-26523980

RESUMO

Microglia are the prime cellular sources of reactive oxygen species (ROS) in the central nervous system (CNS). Chronic activation of microglia has been linked to aging-associated neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) since they produce excessive amounts of ROS for a prolonged duration leading to oxidative stress. The present study was aimed at investigating the expression and role of Sirtuin 3 (Sirt3), a protein deacetylase which is implicated in regulating cellular ROS levels. It has been shown that Sirt3 reduces cellular ROS levels by deacetylating forkhead box O 3a (Foxo3a), a transcription factor which transactivates antioxidant genes, catalase (Cat) and manganese superoxide dismutase (mnSod). In the present study, Sirt3 immunoreactivity was localized in the ameboid microglial cells distributed in the corpus callosum (CC) of the early postnatal rat brain and diminished in the ramified microglial cells in the CC of the adult rat brain. A marked induction of Sirt3 expression was seen in lipopolysaccharide (LPS)-activated microglia in vivo and in vitro as well as in adult rat brains subjected to traumatic brain injury (TBI). Knockdown of Sirt3 in microglia led to an increase in the cellular and mitochondrial ROS and decrease in the expression of antioxidant, mnSod which is indicative of the function of Sirt3 in ROS regulation in microglia. Conversely, Sirt3 overexpression led to increase in the expression of antioxidants Cat and mnSod. Further, increase in the expression and nuclear translocation of Foxo3a was observed following Sirt3 overexpression, suggesting that Sirt3 regulates ROS by inducing the expression of antioxidants via activation of Foxo3a. The above results point to an antioxidant defense mechanism presented by Sirt3 through the activation of Foxo3a, in microglia.


Assuntos
Antioxidantes/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Microglia/metabolismo , Sirtuína 3/metabolismo , Sirtuínas/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Animais Recém-Nascidos , Protocolos de Quimioterapia Combinada Antineoplásica , Lesões Encefálicas/metabolismo , Catalase/metabolismo , Linhagem Celular , Cisplatino , Corpo Caloso/metabolismo , Proteína Forkhead Box O3 , Técnicas de Silenciamento de Genes , Ifosfamida , Lipopolissacarídeos , Camundongos Knockout , Mitomicina , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Sirtuína 3/genética , Sirtuínas/genética , Superóxido Dismutase/metabolismo
4.
Neuroscience ; 253: 40-54, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-23988434

RESUMO

Microglia, the resident immune cells of the CNS, are known to respond to injuries, infection and inflammation in the CNS by producing proinflammatory cytokines and phagocytosing cell debris and pathogens. In this study, we investigated the expression pattern and role of dihydropyrimidinase-like 3 (Dpysl3), a member of collapsin response mediator protein family, on the inflammatory reaction of microglia. Microarray analysis comparing the global gene expression profile of ameboid and ramified microglia has shown that Dpysl3 is mainly expressed in ameboid microglia in the 5-day postnatal rat brain. Immunohistochemical analysis revealed that Dpysl3 was intensely expressed in ameboid microglial cells in the rat brain till postnatal 7th day and then gradually diminished in ramified microglia of 2 weeks postnatal rat brain. Further, in vitro analysis confirmed that Dpysl3 expression was induced in activated BV-2 microglia treated with lipopolysaccharide (LPS). It is well documented that microglial activation by LPS increased the expression of inducible nitric oxide synthase (iNOS) and proinflammatory cytokines through the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity in BV-2 microglia. However, siRNA-mediated knockdown of Dpysl3 prevented the LPS-induced expression of iNOS and cytokines including interleukin-1 beta, and tumor necrosis factor-alpha as well as nuclear translocation of NF-κB in microglia. Remarkably, knockdown of Dpysl3 inhibited the migration of activated microglia coupled with deranged actin filament configuration (as revealed by F-actin cytoskeleton expression) in lamellipodia projecting from the cells. Knockdown of Dpysl3 also inhibited the phagocytic ability of activated microglia. These findings suggest that knockdown of Dpysl3 can inhibit activation, migration and phagocytic capability of microglia and consequently reduce neuroinflammation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Inflamação/metabolismo , Inflamação/patologia , Microglia/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Linhagem Celular Transformada , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Sobrevivência Celular/efeitos dos fármacos , Comportamento Excretor Animal , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/terapia , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Microglia/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Fagócitos/efeitos dos fármacos , Ratos , Ratos Wistar , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
5.
Curr Med Chem ; 20(24): 3022-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23746279

RESUMO

BACKGROUND AND PURPOSE: Transplantation/infusion of mesenchymal stem cells (MSCs) is a promising new approach for treatment of spinal cord injury (SCI). Considering some defined chemokines of MSCs that may have adverse side effects in SCI repair, it is therefore desirable to search for a new chemokine, which should not only be harmless to the host, but also would attract more MSCs to the injury area of spinal cord. This study sought to demonstrate if neurotrophin- 3 (NT-3) would attract migration of MSCs with overexpressing tyrosine kinase C (TrkC) a NT-3 receptor. EXPERIMENTAL APPROACH: A micropipette containing NT-3 was placed in cell culture dish. After this, movement of TrkC gene modified MSCs was monitored for 240 min under an inverted microscope equipped with an imaging system. In vivo, a gelatin sponge scaffold containing TrkC gene modified MSCs was transplanted into the injury area of transected rat spinal cord. Following this, replication-deficient recombinant adenoviral vectors carrying human NT-3 gene (Ad-NT-3) was injected 1 mm caudal to the transplantation site to create an NT-3 enriched area. KEY RESULTS: The results showed that TrkC overexpressing MSCs migrated actively towards the source of NT-3 in the NT-3+TrkC-GFP-MSCs group in vitro. A similar phenomenon was not observed in the control groups. In vivo, transplanted MSCs overexpressing TrkC migrated into the NT-3 enriched area. The incidence of migrating MSCs as well as migration distance was significantly higher than the control groups. CONCLUSION AND IMPLICATIONS: The present results suggest that NT-3 may play a role in attracting MSCs with its high affinity for TrkC as a chemokine receptor.


Assuntos
Movimento Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/fisiologia , Neurotrofina 3/farmacologia , Receptor trkC/biossíntese , Animais , Humanos , Transplante de Células-Tronco Mesenquimais , Neurotrofina 3/genética , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/terapia
6.
Exp Eye Res ; 109: 83-97, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23375774

RESUMO

Hypoxia-induced glutamate accumulation in neural tissues results in damage to neurons through excitotoxic mechanisms via activation of glutamate receptors (GluRs). Here we examine whether hypoxia in the developing retina would cause activation of the ionotropic α-amino-3-hydroxy-5-methylisoxazole-4-propioate (AMPA) GluRs and increase in Ca(2+) influx into retinal ganglion cells (RGCs) that might ultimately lead to their death. Neonatal Wistar rats were subjected to hypoxia for 2h and then sacrificed at various time points after the exposure together with normal age matched control rats. Primary cultures of RGCs were also prepared and subjected to hypoxia. Expression of AMPA glutamate receptor (GluR) 1-4 was examined in the retina. Additionally, expression of GluRs, intracellular Ca(2+) influx, reactive oxygen species (ROS) generation and cell death were investigated in cultured RGCs. GluR1-4 mRNA and protein expression showed a significant increase (P < 0.01) over control values after the hypoxic exposure both in vivo and in vitro. Cells expressing GluR1-4 in the retina were identified as RGCs by double immunofluorescence labeling with Thy1.1. Increased intracellular Ca(2+) in cultured RGCs following hypoxic exposure was reduced (P < 0.01) by 10 µM AMPA antagonist 6, 7-dinitroquinoxaline-2,3-dione (DNQX). Our results suggest that following a hypoxic insult, an increased amount of glutamate accumulates in the neonatal retina. This would then activate AMPA receptors which may damage RGCs through increased Ca(2+) accumulation and ROS generation. The involvement of AMPA receptors in damaging the RGCs is evidenced by suppression of intracellular Ca(2+) influx by DNQX which also decreased ROS generation and cell death by 50%.


Assuntos
Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Quinoxalinas/farmacologia , Receptores de AMPA/metabolismo , Células Ganglionares da Retina/metabolismo , Animais , Animais Recém-Nascidos , Cálcio/metabolismo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipóxia/patologia , Fármacos Neuroprotetores/farmacologia , Cultura Primária de Células , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores de AMPA/antagonistas & inibidores , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia
7.
Neuroscience ; 237: 106-17, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23376738

RESUMO

Growing evidence has suggested that hydrogen sulfide (H2S) acts as a novel neuro-modulator and neuroprotective agent; however, it remains to be investigated whether H2S has a direct effect on neural stem cells (NSCs). We report here that NSCs expressed cystathionine ß synthase (CBS) and addition of exogenous H2S donor, L-cysteine, stimulated proliferation and increased the differentiation potential of NSCs to neurons and astroglia. Moreover, pre-treatment with aminooxyacetic acid, the inhibitor of CBS or knockdown of CBS in using siRNA, significantly attenuated the effects of L-cysteine on elevated H2S levels and the cell proliferation; it also effectively suppressed L-cysteine-induced neurogenesis and astrocytogenesis. Further analysis revealed that L-cysteine-induced proliferation was associated with phosphorylation of extracellular signal-regulated kinases 1/2 and differentiation with altered expression of differentiation-related genes. Taken together, the present data suggest that L-cysteine can enhance proliferation and differentiation of NSCs via the CBS/H2S pathway, which may serve as a useful inference for elucidating its role in regulating the fate of NSCs in physiological and pathological settings.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cistationina beta-Sintase/metabolismo , Cisteína/farmacologia , Sulfeto de Hidrogênio/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Análise de Variância , Animais , Bromodesoxiuridina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia , Telencéfalo/citologia , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Neuropathol Appl Neurobiol ; 39(5): 531-52, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23106634

RESUMO

AIMS: The effect of combustion smoke inhalation on the respiratory system is widely reported but its effects on the central nervous system remain unclear. Here, we aimed to determine the effects of smoke inhalation on the cerebellum and hippocampus which are areas vulnerable to hypoxia injury. METHODS: Adult male Sprague-Dawley rats were subjected to combustion smoke inhalation and sacrificed at 0.5, 3, 24 and 72 h after exposure. The cerebellum and hippocampus were subjected to Western analysis for VEGF, iNOS, eNOS, nNOS and AQP4 expression; ELISA analysis for cytokine and chemokine levels; and immunohistochemistry for GFAP/AQP4, RECA-1/RITC and TUNEL. Aminoguanidine (AG) was administered to determine the effects of iNOS after smoke inhalation. RESULTS: Both the cerebellum and hippocampus showed a significant increase in VEGF, iNOS, eNOS, nNOS and AQP4 expression with corresponding increases in inflammatory cytokines and chemokines and increased AQP4 expression and RITC permeability after smoke exposure. AG was able to decrease the expression of iNOS, followed by VEGF, eNOS, nNOS, RITC and AQP4 after smoke exposure. There was also a significant increase in TUNEL+ cells in the cerebellum and hippocampus which were not significantly reduced by AG. Beam walk test revealed immediate deficits after smoke inhalation which was attenuated with AG. CONCLUSION: The findings suggest that iNOS plays a major role in the central nervous system inflammatory pathophysiology after smoke inhalation exposure with concomitant increase in proinflammatory molecules, vascular permeability and oedema, for which the cerebellum appears to be more vulnerable to smoke exposure than the hippocampus.


Assuntos
Cerebelo/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Inflamação/induzido quimicamente , Fumaça/efeitos adversos , Animais , Aquaporina 4/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Óxido Nítrico Sintase/metabolismo , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Curr Med Chem ; 19(5): 770-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22214447

RESUMO

BACKGROUND AND PURPOSE: Microglial activation plays an important role in neurodegenerative diseases by producing an array of proinflammatory enzymes and cytokines. Ginsenoside Rg1 (Rg1), a well-known Chinese herbal medicine, has been well recognized for its anti-inflammatory effect. This study sought to determine the anti-inflammatory effects of Rg1 and its underlying mechanisms in lipopolysaccharide (LPS)-stimulated murine BV-2 microglial cells. EXPERIMENTAL APPROACH: Murine BV-2 microglial cells were treated with Rg1 (10, 20, and 40 µM) and/or LPS (1 µg·ml(-1)). The mRNA and protein levels of proinflammatory proteins and cytokines were analysed by RT-PCR assay and double immunofluorescence labeling, respectively. Phosphorylation levels of mitogen-activated protein kinases (MAPKs) cascades, inhibitor κB-α (IκB-α) and cyclic AMP- responsive element (CRE)-binding protein (CREB) were measured by western blot. U73122 (5 µM), a specific phospholipase C (PLC) inhibitor, was used to determine if PLC signaling pathway might be involved in Rg1's action on activated BV-2 cells. KEY RESULTS: Pretreatment with Rg1 significantly attenuated the LPS-induced expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and nuclear factor-κB (NF-κB) in BV-2 cells. U73122 blocked the effects of Rg1 on LPS-induced microglial activation. In addition, PLC-γ1 inhibition partially abolished the inhibitory effect of Rg1 on the phosphorylation of IκB-α, CREB, extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal protein kinase (JNK), and p38 mitogen-activated protein kinase (p38 MAPK). CONCLUSION AND IMPLICATIONS: This investigation demonstrates that Rg1 significantly attenuates overactivation of microglial cells by repressing expression levels of neurotoxic proinflammatory mediators and cytokines via activation of PLC-γ1 signaling pathway.


Assuntos
Ginsenosídeos/farmacologia , Inflamação/tratamento farmacológico , Lipopolissacarídeos/farmacologia , Microglia/imunologia , Fosfolipase C gama/metabolismo , Transdução de Sinais , Animais , Anti-Inflamatórios , Linhagem Celular , Fármacos do Sistema Nervoso Central , Citocinas , Relação Dose-Resposta a Droga , Ginsenosídeos/uso terapêutico , Lipopolissacarídeos/antagonistas & inibidores , Camundongos , Microglia/metabolismo , Fosforilação
10.
Neuroscience ; 202: 58-68, 2012 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-22178606

RESUMO

Astrocyte activation plays important roles both in physiological and pathological process in the CNS. In the latter, the process is further aggravated by hyperglycemia, leading to diabetes complications of CNS. We report here that high glucose (HG) treatment stimulated astrocytic morphological alteration coupled with changes in glial fibrillary acidic protein (GFAP) and vimentin expression. Additionally, HG upregulated the expression of tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), interleukin-1ß (IL-1ß), interleukin-4 (IL-4), and vascular endothelial growth factor (VEGF); however, its effects on transforming growth factor-ß (TGF-ß) expression were not evident. HG treatment induced increased production of reactive oxygen species (ROS) as well as activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and signal transducer and activator transcription 3 (STAT 3). HG-induced expression of TNF-α, IL-6, IL-1ß, IL-4, and VEGF was blocked by ROS scavenger and inhibitors specific for NF-κB and STAT 3, respectively. The results suggest that the aforementioned multiple inflammatory cytokines and mediators that may be linked to the pathogenesis of the diabetes complications of CNS are induced by HG via the key signaling pathways.


Assuntos
Astrócitos/metabolismo , Citocinas/biossíntese , Glucose/toxicidade , Inflamação/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Sequestradores de Radicais Livres/farmacologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação/induzido quimicamente , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Melatonina/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Prolina/análogos & derivados , Prolina/farmacologia , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/metabolismo , Tiocarbamatos/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Triterpenos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Neuroscience ; 192: 140-54, 2011 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-21729740

RESUMO

Microglial cells exhibit Notch-1 signaling expression which is enhanced upon activation. We reported previously that enhanced Notch-1 expression in activated microglia modulates production of proinflammatory cytokines and nitric oxide (NO). Furthermore, Notch-1 modulates transcription factor nuclear factor-kappa B (NF-κB). This study was aimed to investigate if NF-κB reciprocally modulates Notch signaling in BV-2 cells. In this connection, the cells were pretreated with caffeic acid phenethyl ester (Cape) followed by stimulating the cells with lipopolysaccharide (LPS). Cape+LPS treatment resulted in reduced translocation of NF-κB into the nucleus. Concomitantly, NF-κB DNA binding activity and the mRNA and protein expression levels of NF-κB/p65, Notch-1, intracellular domain of Notch-1 receptor (NICD), Hes-1, tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1ß) and inducible nitric oxide synthase (iNOS) along with nitrite level were significantly reduced. Additionally, flow cytometry analysis showed a decrease in expression levels of NF-κB/p65, Notch-1, NICD but an increase in that of signal transducers and activators of transcription 3 (Stat3). Furthermore, nuclear Hes-1, phosphorylated Stat3 (p-Stat3) and recombination signal-binding protein 1 for J-Kappa (RBP-JK) expression levels were significantly suppressed. The present results suggest that Cape inhibits NF-κB activation through suppressing its interaction with DNA. Cape-induced reduction of Hes-1 may be attributed to decreased interaction between NICD and RBP-JK whose levels were reduced concurrently. Hes-1 reduction may lead to decreased production of inflammatory cytokines and NO. It is concluded that NF-κB can modulate Notch-1 signaling. Both pathways operate synergistically for production of proinflammatory cytokines and NO in activated microglia.


Assuntos
Citocinas/biossíntese , Microglia/metabolismo , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Ácidos Cafeicos/farmacologia , Linhagem Celular , Separação Celular , Citotoxinas/farmacologia , Citometria de Fluxo , Imunofluorescência , Inflamação/metabolismo , Camundongos , Álcool Feniletílico/análogos & derivados , Álcool Feniletílico/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Curr Med Chem ; 18(6): 886-99, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21182477

RESUMO

Among potential radiological, nuclear, biological and chemical weapons, cholinergic nerve agents from chemical weapons remain a realistic terrorist threat due to its combination of high lethality, demonstrated use and relative abundance of un-destroyed stockpiles in various militaries around the world. While current fielded antidotes are able to mitigate acute poisoning, effective neuroprotection in the field remains a challenge amongst subjects with established status epilepticus following nerve agent intoxication. Due to ethical, safety and surety issues, extensive preclinical and clinical research on cholinergic nerve agents is not possible. This may have been a contributory factor for the slow progress in uncovering new neuroprotectants for nerve agent casualties with established status epilepticus. To overcome this challenge, comparative research with surrogate chemicals that produce similar hypercholinergic toxicity but with less security concerns would be a useful approach forward. In this paper, we will systemically compare the mechanism of seizure generation, propagation and the subsequent clinical, hematologic, and metabolic, biochemical, neuroinflammatory changes and current therapeutic approaches reported in pilocarpine, soman, and sarin models of seizures. This review will be an important first step in closing this knowledge gap among different closely related models of seizures and neurotoxicity. Hopefully, it will spur further efforts in using surrogate cholinergic models by the wider scientific community to expedite the development of a new generation of antidotes that are better able to protect against delayed neurological effects inflicted by nerve agents.


Assuntos
Substâncias para a Guerra Química/farmacologia , Pilocarpina/farmacologia , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/terapia , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Substâncias para a Guerra Química/toxicidade , Humanos , Pilocarpina/toxicidade , Sarina/farmacologia , Sarina/toxicidade , Soman/farmacologia , Soman/toxicidade , Estado Epiléptico/etiologia , Estado Epiléptico/metabolismo
13.
Curr Med Chem ; 17(36): 4492-510, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21062255

RESUMO

Spinal cord injury (SCI) damages axons and disrupts myelination interrupting sensory and motor neuronal transmission to and from the brain. Patients suffering from SCI although continue to survive, are often left chronically disabled and with no promise of a cure. Advances in stem cell biology has opened up doors for the use of human embryonic, adult neural and induced pluripotent stem cell strategies for SCI. Despite great promise from animal research, clinical trials have been limited and the jury is still out on its safety and efficacy. This review discusses the advantages and disadvantages of the various stem cell types, barriers hindering translation from animal to humans, and the need for established guidelines for standardization of clinical trials ensuring subsequent implementation. Ultimately, unrealistic expectations of stem cell therapy (SCT) as the elixir for SCI should be managed. The success of SCT for SCI lies in the network of research scientists, medical professionals and patients working cooperatively to build up a knowledge-intensive platform for a comprehensive risk-benefit assessment of SCT for SCI.


Assuntos
Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Animais , Humanos , Modelos Biológicos , Traumatismos da Medula Espinal/patologia
14.
J Neurosci Res ; 88(12): 2701-14, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20648656

RESUMO

Microglial cells constitutively express Notch-1 and nuclear factor-kappaB/p65 (NF-kappaB/p65), and both pathways modulate production of inflammatory mediators. This study sought to determine whether a functional relationship exists between them and, if so, to investigate whether they synergistically regulate common microglial cell functions. By immunofluorescence labeling, real-time polymerase chain reaction (RT-PCR), flow cytometry, and Western blot, BV-2 cells exhibited Notch-1 and NF-kappaB/p65 expression, which was significantly up-regulated in cells challenged with lipopolysaccharide (LPS). This was coupled with an increase in expression of Hes-1, tumor necrosis factor-alpha (TNF-alpha), and interleukin-1beta (IL-1beta). In BV-2 cells pretreated with N-[N-(3,5-difluorophenacetyl)-1-alany1]-S-phenyglycine t-butyl ester (DAPT), a gamma-secretase inhibitor, followed by LPS stimulation, Notch-1 expression level was enhanced but that of all other markers was suppressed. Additionally, Hes-1 expression and NF-kappaB nuclear translocation decreased as shown by flow cytometry. Notch-1's modulation of NF-kappaB/p65 was also evidenced in amoeboid microglial cells (AMC) in vivo. In 5-day-old rats given intraperitoneal injections of LPS, Notch-1, NF-kappaB/p65, TNF-alpha, and IL-1beta immunofluorescence in AMC was markedly enhanced. However, in rats given an intraperitoneal injection of DAPT prior to LPS, Notch-1 labeling was augmented, but that of TNF-alpha and IL-1beta was reduced. The results suggest that blocking of Notch-1 activation with DAPT would reduce the level of its downstream end product Hes-1 along with suppression of NF-kappaB/p65 translocation, resulting in suppressed production of proinflammatory cytokines. It is concluded that Notch-1 signaling can trans-activate NF-kappaB/p65 by amplifying NF-kappaB/p65-dependent proinflammatory functions in activated microglia.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Dipeptídeos/farmacologia , Microglia/metabolismo , Inibidores de Proteases/farmacologia , Receptor Notch1/metabolismo , Transdução de Sinais/fisiologia , Fator de Transcrição RelA/metabolismo , Secretases da Proteína Precursora do Amiloide/fisiologia , Animais , Animais Recém-Nascidos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Gliose/enzimologia , Gliose/metabolismo , Gliose/patologia , Mediadores da Inflamação/metabolismo , Mediadores da Inflamação/fisiologia , Camundongos , Microglia/efeitos dos fármacos , Microglia/enzimologia , Ratos , Ratos Wistar , Receptor Notch1/genética , Receptor Notch1/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética
15.
Neuroscience ; 167(3): 665-77, 2010 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-20206236

RESUMO

We reported previously that amoeboid microglial cells (AMC) in the developing brain exhibited endothelins (ETs) expression which diminished with advancing age and was undetected in microglia in the more mature brain. This study sought to explore if microglia in the adult would be induced to express ETs in altered conditions. By immunofluorescence microscopy, ETs and endothelin (ET)-B receptor were undetected in microglial cells in sham-operated and normal control rats. However, in adult rats subjected to middle cerebral artery occlusion (MCAO), lectin labeled activated microglia which occurred in large numbers in the marginal zones in the ischemic cortex at 3 days and 1 week intensely expressed ETs specifically endothelin (ET)-1 and ET-B receptor; ET-3 and ET-A receptor were absent in these cells. By RT-PCR and ELISA, ET-1 and -3 mRNA and protein expression level was progressively increased in the ischemic cerebral cortex after MCAO compared with the controls. ET-A and ET-B receptor mRNA and protein levels were concomitantly up-regulated. It is suggested that increased release of ET-1 following MCAO by massive activated microglia can exert an immediate constriction of local blood vessels bearing ET-A receptor. ET-1 may also interact with activated microglia endowed with ET-B receptor via an autocrine manner that may be linked to chemokines/cytokines production. ET-1, ET-3 and ET-B receptor were also localized in reactive astrocytes along with some oligodendrocytes. We conclude that activated microglia together with other glial cells in the marginal zone after MCAO are the main cellular source of ETs that may be involved in regulation of vascular constriction and glial chemokines/cytokines production. However, dissecting the role of individual component of the endothelin system in the various glial cells, notably activated microglia, would be vital in designing of an effective therapeutic strategy for clinical treatment of stroke in which microglial cells have been implicated.


Assuntos
Isquemia Encefálica/metabolismo , Encefalite/metabolismo , Endotelinas/metabolismo , Gliose/metabolismo , Microglia/metabolismo , Receptores de Endotelina/metabolismo , Animais , Isquemia Encefálica/fisiopatologia , Proliferação de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/fisiopatologia , Endotelina-1/genética , Endotelina-1/metabolismo , Endotelina-3/genética , Endotelina-3/metabolismo , Endotelinas/genética , Gliose/fisiopatologia , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Lectinas , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Endotelina A/genética , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Receptores de Endotelina/genética , Coloração e Rotulagem , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Vasoconstrição/fisiologia
16.
J Pineal Res ; 48(3): 185-193, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20136703

RESUMO

This study investigated the potential of melatonin in ameliorating hypoxic damage to the periventricular white matter (PWM) in the neonatal brain. Vascular endothelial growth factor (VEGF), nitric oxide (NO), glutathione (GSH) and malondialdehyde (MDA) content in the PWM of 1-day-old rats subjected to hypoxia for a period of 2 hr was examined. Vascular endothelial growth factor, NO and MDA concentration was increased whereas that of GSH was reduced after the hypoxic exposure. Additionally, degenerating axons, apoptotic and necrotic cells and vacuolation of capillary endothelial cells were observed in the PWM. The neighboring ependymal and choroid plexus cells also appeared to undergo structural alterations. Increased vascular permeability in the PWM of hypoxic rats was evidenced by the leakage of rhodamine isothiocyanate (RhIC) which was taken up by the amoeboid microglial cells. In vitro experiments showed increased apoptosis in OLN-93 cells, an oligodendrocytic cell line, following hypoxic exposure. Hypoxic rats treated with melatonin showed reduced VEGF, NO and MDA concentrations, increased GSH content and reduced RhIC leakage in the PWM. The ultrastructure of axons, endothelial, ependymal and choroid plexus epithelial cells appeared relatively normal in the hypoxic animals treated with melatonin. The incidence of apoptotic OLN-93 cells was also reduced with melatonin treatment. We suggest that the protective effects of melatonin on various parameters in the PWM of hypoxic neonatal brains were due to its antioxidant properties.


Assuntos
Ventrículos Cerebrais/efeitos dos fármacos , Hipóxia Encefálica/prevenção & controle , Melatonina/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Astrócitos/citologia , Astrócitos/metabolismo , Linhagem Celular , Ventrículos Cerebrais/metabolismo , Glutationa/metabolismo , Hipóxia Encefálica/metabolismo , Imuno-Histoquímica , Peroxidação de Lipídeos/efeitos dos fármacos , Malondialdeído/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Óxido Nítrico/biossíntese , Óxido Nítrico/metabolismo , Oligodendroglia/metabolismo , Ratos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo
17.
Neuroscience ; 165(2): 386-94, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19878709

RESUMO

Activation of microglial cells, the resident immune cells of the CNS causes neurotoxicity through the release of a wide array of inflammatory mediators including proinflammatory cytokines, chemokines and reactive oxygen species. In this study, we have investigated the expression of NG2 (also known as CSPG4), one of the members of transmembrane chondroitin sulfate proteoglycans family, in microglial cells and its role on inflammatory reaction of microglia by analyzing the expression of the proinflammation cytokines (interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF-alpha)), chemokines (stromal cell-derived factor-1alpha and monocyte chemotactic protein-1) and inducible nitric oxide synthase (iNOS). NG2 expression was not detectable in microglial cells expressing OX-42 in the brains of 1-day old postnatal rat pups and adult rats; it was, however, induced in activated microglial cells in pups and adult rats injected with lipopolysaccharide (LPS). In vitro analysis further confirmed that LPS induced the expression of NG2 in primary microglial cells and this was inhibited by dexamethasone. It has been well demonstrated that LPS induces the expression of iNOS and proinflammatory cytokines in microglia. However in this study, LPS did not induce the mRNA expression of iNOS and cytokines including IL-1beta, and TNF-alpha in microglial cells transfected with CSPG4 siRNA. On the contrary, mRNA expression of chemokines such as monocyte chemoattractant protein-1 (MCP-1) and stromal cell-derived factor-1alpha (SDF-1alpha) was significantly increased in LPS-activated microglial cells after CSPG4 siRNA transfection in comparison with the control. The above results indicate that NG2 mediates the induction of iNOS and inflammatory cytokine expression, but not the chemokine expression in activated microglia.


Assuntos
Antígenos/metabolismo , Encéfalo/imunologia , Encéfalo/fisiologia , Microglia/imunologia , Microglia/fisiologia , Proteoglicanas/metabolismo , Envelhecimento , Animais , Animais Recém-Nascidos , Anti-Inflamatórios/farmacologia , Encéfalo/efeitos dos fármacos , Quimiocina CCL2/metabolismo , Quimiocina CXCL12/metabolismo , Dexametasona/farmacologia , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
18.
Neuroscience ; 166(1): 132-44, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20036321

RESUMO

Microglial activation has been implicated as one of the causative factors for neuroinflammation in various neurodegenerative diseases. The sphingolipid metabolic pathway plays an important role in inflammation, cell proliferation, survival, chemotaxis, and immunity in peripheral macrophages. In this study, we demonstrate that sphingosine kinase1 (SphK1), a key enzyme of the sphingolipid metabolic pathway, and its receptors are expressed in the mouse BV2 microglial cells and SphK1 alters the expression and production of proinflammatory cytokines and nitric oxide in microglia treated with lipopolysaccharide (LPS). LPS treatment increased the SphK1 mRNA and protein expression in microglia as revealed by the RT-PCR, Western blot and immunofluorescence. Suppression of SphK1 by its inhibitor, N, N Dimethylsphingosine (DMS), or siRNA resulted in decreased mRNA expression of TNF-alpha, IL-1beta, and iNOS and release of TNF-alpha and nitric oxide (NO) in LPS-activated microglia. Moreover, addition of sphingosine 1 phosphate (S1P), a breakdown product of sphingolipid metabolism, increased the expression levels of TNF-alpha, IL-1beta and iNOS and production of TNF-alpha and NO in activated microglia. Hence to summarize, suppression of SphK1 in activated microglia inhibits the production of proinflammatory cytokines and NO and the addition of exogenous S1P to activated microglia enhances their inflammatory responses. Since the chronic proinflammatory cytokine production by microglia has been implicated in neuroinflammation, modulation of SphK1 and S1P in microglia could be looked upon as a future potential therapeutic method in the control of neuroinflammation in neurodegenerative diseases.


Assuntos
Citocinas/metabolismo , Encefalite/enzimologia , Gliose/enzimologia , Microglia/enzimologia , Óxido Nítrico/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Linhagem Celular , Encefalite/fisiopatologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/fisiologia , Gliose/fisiopatologia , Interleucina-1beta/metabolismo , Lipopolissacarídeos , Lisofosfolipídeos/metabolismo , Lisofosfolipídeos/farmacologia , Camundongos , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Interferência de RNA/fisiologia , RNA Mensageiro/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Esfingosina/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia
19.
Neuroscience ; 164(4): 1521-30, 2009 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-19782730

RESUMO

Granulocyte-colony stimulating factor (G-CSF) is a growth factor that regulates proliferation, differentiation and survival of hematopoietic progenitor cells. There is growing evidence to suggest that G-CSF exerts a powerful neuroprotective effect in different neurological disorders. However, it has remained to be elucidated if G-CSF has a direct effect on neural stem cells (NSCs). Here, we show that G-CSF could stimulate the proliferation of NSCs and promote their differentiation in vitro. Additionally, we have shown that G-CSF-induced proliferation of NSCs is associated with phosphorylation of STAT3, and the differentiation is linked to altered expression of differentiation-related genes. Remarkably, G-CSF could not initiate the differentiation of NSCs. The added roles of G-CSF in regulating proliferation and differentiation of NSCs as shown in this study would serve as a useful reference in designing new stem cell therapy strategies for promoting brain recovery and repair.


Assuntos
Astrócitos/citologia , Fator Estimulador de Colônias de Granulócitos/fisiologia , Neurônios/citologia , Células-Tronco/citologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Encéfalo/citologia , Diferenciação Celular , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Embrião de Mamíferos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação , Receptores de Fator Estimulador de Colônias de Granulócitos/biossíntese , Fator de Transcrição STAT3/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
20.
Neuroscience ; 164(3): 948-62, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19712723

RESUMO

This study aimed to investigate the effect of hypoxia on the expression of cyclooxygenase-1 (COX-1), cyclooxygenase-2 (COX-2), microsomal prostaglandin-E synthase (mPGES-1), E-prostanoid receptor 2 (EP2) in microglia; and the roles of EP2-cyclic adenosine monophosphate (cAMP) signaling pathway in the prostaglandin E(2) (PGE(2)) regulation of inflammatory mediators released by hypoxic BV-2 cells. Immunoexpression of COX-1, COX-2, mPGES-1 and EP2 was localized in the amoeboid microglial cells (AMC), a nascent brain macrophage in the developing brain, as confirmed by double labeling with OX-42 and lectin, specific markers of microglia. AMC emitted a more intense immunofluorescence in hypoxic rats when compared with the matching controls. In postnatal rats subjected to hypoxia, mRNA and protein expression levels of COX-1, COX-2 and mPGES-1 along with tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), inducible nitric-oxide synthase (iNOS) and PGE(2) product in the callosal tissue were significantly increased. The results were shared in the BV-2 cells except for COX-1 mRNA and protein whose levels remained unaltered. Interestingly, treatment with EP2 antagonist AH-6809 resulted in suppression of hypoxia induced EP2, IL-1beta and iNOS mRNA and protein expression, TNF-alpha protein expression and intracellular cAMP level in BV-2 cells. It is suggested that PGE(2) may regulate above inflammatory mediators in the activated microglia via EP2-cAMP signaling pathway in hypoxic conditions.


Assuntos
Dinoprostona/metabolismo , Encefalite/metabolismo , Hipóxia Encefálica/metabolismo , Oxirredutases Intramoleculares/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Receptores de Prostaglandina E/metabolismo , Animais , Animais Recém-Nascidos , Antígeno CD11b/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , AMP Cíclico/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/fisiopatologia , Hipóxia Encefálica/fisiopatologia , Oxirredutases Intramoleculares/genética , Camundongos , Microglia/metabolismo , Microglia/ultraestrutura , Microssomos/metabolismo , Microssomos/ultraestrutura , Óxido Nítrico Sintase Tipo II/metabolismo , Antagonistas de Prostaglandina/farmacologia , Prostaglandina-E Sintases , Prostaglandina-Endoperóxido Sintases/genética , RNA Mensageiro/metabolismo , Ratos , Receptores de Prostaglandina E Subtipo EP2 , Transdução de Sinais/fisiologia , Xantonas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...