Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1084: 145-174, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30039439

RESUMO

Multiple sclerosis (MS) is a chronic inflammatory, autoimmune, and neurodegenerative disease of the central nervous system (CNS). It is characterized by demyelination and neuronal loss that is induced by attack of autoreactive T cells to the myelin sheath and endogenous remyelination failure, eventually leading to functional neurological disability. Although recent evidence suggests that MS relapses are induced by environmental and exogenous triggers such as viral infections in a genetic background, its very complex pathogenesis is not completely understood. Therefore, the efficiency of current immunosuppression-based therapies of MS is too low, and emerging disease-modifying immunomodulatory agents such as fingolimod and dimethyl fumarate cannot stop progressive neurodegenerative process. Thus, the cell replacement therapy approach that aims to overcome neuronal cell loss and remyelination failure and to increase endogenous myelin repair capacity is considered as an alternative treatment option. A wide variety of preclinical studies, using experimental autoimmune encephalomyelitis model of MS, have recently shown that grafted cells with different origins including mesenchymal stem cells (MSCs), neural precursor and stem cells, and induced-pluripotent stem cells have the ability to repair CNS lesions and to recover functional neurological deficits. The results of ongoing autologous hematopoietic stem cell therapy studies, with the advantage of peripheral administration to the patients, have suggested that cell replacement therapy is also a feasible option for immunomodulatory treatment of MS. In this chapter, we overview cell sources and applications of the stem cell therapy for treatment of MS. We also discuss challenges including those associated with administration route, immune responses to grafted cells, integration of these cells to existing neural circuits, and risk of tumor growth. Finally, future prospects of stem cell therapy for MS are addressed.


Assuntos
Esclerose Múltipla , Transplante de Células-Tronco , Animais , Humanos , Esclerose Múltipla/terapia , Bainha de Mielina , Transplante de Células-Tronco/tendências
2.
Antioxidants (Basel) ; 10(5)2021 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-34066647

RESUMO

Ethyl pyruvate is a molecule with anti-inflammatory and pro-metabolic effects. Ethyl pyruvate has been shown to ameliorate the clinical and pathological findings of neurodegenerative diseases such as Alzheimer's and Parkinson's Diseases in rodents. Its anti-inflammatory and neuroprotective effects are widely investigated in animal and cellular models. Our study aimed to investigate the mechanism of the impact of Ethyl pyruvate on NLRP3 inflammasome activation in the N9 microglial cell line. Our results indicated that ethyl pyruvate significantly suppressed LPS and ATP-induced NLRP3 inflammasome activation, decreased active caspase-1 level, secretion of IL-1ß and IL-18 cytokines, and reduced the level of pyroptotic cell death resulting from inflammasome activation. Furthermore, ethyl pyruvate reduced the formation of total and mitochondrial ROS and suppressed inflammasome-induced HMGB1 upregulation and nuclear NF-κB translocation and reversed the inflammasome activation-induced miRNA expression profile for miR-223 in N9 cells. Our study suggests that ethyl pyruvate effectively suppresses the NLRP3 inflammasome activation in microglial cells regulation by miR-223 and NF-κB/HMGB1 axis.

3.
Front Immunol ; 12: 737065, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858398

RESUMO

NLRP3 inflammasome activation contributes to several pathogenic conditions, including lipopolysaccharide (LPS)-induced sickness behavior characterized by reduced mobility and depressive behaviors. Dimethyl fumarate (DMF) is an immunomodulatory and anti-oxidative molecule commonly used for the symptomatic treatment of multiple sclerosis and psoriasis. In this study, we investigated the potential use of DMF against microglial NLRP3 inflammasome activation both in vitro and in vivo. For in vitro studies, LPS- and ATP-stimulated N9 microglial cells were used to induce NLRP3 inflammasome activation. DMF's effects on inflammasome markers, pyroptotic cell death, ROS formation, and Nrf2/NF-κB pathways were assessed. For in vivo studies, 12-14 weeks-old male BALB/c mice were treated with LPS, DMF + LPS and ML385 + DMF + LPS. Behavioral tests including open field, forced swim test, and tail suspension test were carried out to see changes in lipopolysaccharide-induced sickness behavior. Furthermore, NLRP3 and Caspase-1 expression in isolated microglia were determined by immunostaining. Here we demonstrated that DMF ameliorated LPS and ATP-induced NLRP3 inflammasome activation by reducing IL-1ß, IL-18, caspase-1, and NLRP3 levels, reactive oxygen species formation and damage, and inhibiting pyroptotic cell death in N9 murine microglia via Nrf2/NF-κB pathways. DMF also improved LPS-induced sickness behavior in male mice and decreased caspase-1/NLRP3 levels via Nrf2 activation. Additionally, we showed that DMF pretreatment decreased miR-146a and miR-155 both in vivo and in vitro. Our results proved the effectiveness of DMF on the amelioration of microglial NLRP3 inflammasome activation. We anticipate that this study will provide the foundation consideration for further studies aiming to suppress NLRP3 inflammasome activation associated with in many diseases and a better understanding of its underlying mechanisms.


Assuntos
Fumarato de Dimetilo/uso terapêutico , Comportamento de Doença/fisiologia , Fatores Imunológicos/uso terapêutico , Inflamassomos/metabolismo , Inflamação/tratamento farmacológico , Microglia/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Humanos , Inflamação/imunologia , Lipopolissacarídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Esclerose Múltipla/tratamento farmacológico , Psoríase/tratamento farmacológico , Transdução de Sinais
4.
Immunol Lett ; 233: 20-30, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33711331

RESUMO

The NLRP3 inflammasome is a multiprotein complex that activates caspase-1 and triggers the release of the proinflammatory cytokines IL-1ß and IL-18 in response to diverse signals. Although inflammasome activation plays critical roles against various pathogens in host defense, overactivation of inflammasome contributes to the pathogenesis of inflammatory diseases, including acute CNS injuries and chronic neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. In the current study, we demonstrated that Sulforaphane (SFN), a dietary natural product, inhibits NLRP3 inflammasome mediated IL-1ß and IL-18 secretion and pyroptosis in murine microglial cells. SFN decreased the secretion of IL-1ß and IL-18, and their mRNA levels in LPS primed microglia triggered by ATP. SFN suppressed the overexpression of cleaved caspase-1 and NLRP3 protein expressions as measured by caspase activity assay and western blot, respectively. SFN also prevented caspase-1 dependent pyroptotic cell death in microglia. Our data indicate that SFN suppresses NLRP3 inflammasome via the inhibition of NF-κB nuclear translocation and Nrf2 mediated miRNAs expression modulation in murine microglia.


Assuntos
Inflamassomos/metabolismo , Isotiocianatos/farmacologia , MicroRNAs/genética , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Sulfóxidos/farmacologia , Animais , Caspase 1/metabolismo , Citocinas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Camundongos , Piroptose/efeitos dos fármacos , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo
5.
Cell Biochem Funct ; 28(3): 197-201, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20229611

RESUMO

Erythropoietin (Epo) exerts neuroprotective, glioprotective, and vascular protective effects in the nervous system. However, the mechanisms of the cytoprotective effect of Epo have not been fully clarified. Here, we investigated whether Epo affects the transcription and activation of nuclear factor erythroid 2-related factor 2 (Nrf2), which is a key transcription factor of the cellular anti-oxidant defense system, and mRNA expression of its target genes including heme oxygenase-1 (HO-1). Epo was added to SH-SY5Y cells at 1 U mL(-1) and cultures were incubated for 24 h and then mRNA expression of Nrf2 target genes were analyzed with real-time PCR. SH-SY5Y cells were incubated with Epo at different time points and the nuclear and cytoplasmic levels of Nrf2 protein expression were examined by Western blotting and immunohistochemistry. Specific inhibitors of mitogen-activated protein kinases (MAPKs) and phosphatidylinositol-3 kinase (PI3K) were used to find out the possible signaling pathways that mediate the activating effect of Epo on Nrf2 activation. In cultured human SH-SY5Y neuroblastoma cells, Western blotting, immunohistochemistry, and real-time PCR analysis demonstrated that Epo-induced nuclear translocation of Nrf2 and upregulates HO-1 expression. Inhibitors of MAPKs and PI3K decreased Epo-induced nuclear translocation of Nrf2 and HO-1 mRNA expression. These results suggest that Epo induces neural HO-1 expression through the activation of PI3K, MAPK, and Nrf2 pathways, and this may unveil a novel mechanism which mediates the cytoprotective responses elicited by Epo.


Assuntos
Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Linhagem Celular/efeitos dos fármacos , Eritropoetina/farmacologia , Heme Oxigenase-1/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Linhagem Celular/metabolismo , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Humanos , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
6.
Adv Protein Chem Struct Biol ; 119: 247-308, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31997770

RESUMO

Multiple sclerosis (MS) is a chronic, autoimmune and neuroinflammatory disease of the central nervous system (CNS) mediated by autoreactive T cells directed against myelin antigens. Although the crucial role of adaptive immunity is well established in MS, the contribution of innate immunity has only recently been appreciated. Microglia are the main innate immune cells of the CNS. Similar to other myeloid cells, microglia recognize both exogenous and host-derived endogenous danger signals through pattern recognition receptors (PRRs) localized on their cell surface such as Toll Like receptor 4, or in the cytosol such as NLRP3. The second one is the sensor protein of the multi-molecular NLRP3 inflammasome complex in activated microglia that promotes the maturation and secretion of proinflammatory cytokines, interleukin-1ß and interleukin-18. Overactivation of microglia and aberrant activation of the NLRP3 inflammasome have been implicated in the pathogenesis of MS. Indeed, experimental data, together with post-mortem and clinical studies have revealed an increased expression of NLRP3 inflammasome complex elements in microglia and other immune cells. In this review, we focus on microglial NLRP3 inflammasome activation in MS. First, we overview the basic knowledge about MS, microglia and the NLRP3 inflammasome. Then, we summarize studies about microglial NLRP3 inflammasome activation in MS and its animal models. We also highlight experimental therapeutic approaches that target different steps of NLRP inflammasome activation. Finally, we discuss future research avenues and new methods in this rapidly evolving area.


Assuntos
Inflamassomos/imunologia , Microglia/imunologia , Esclerose Múltipla/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Animais , Humanos , Microglia/patologia , Esclerose Múltipla/patologia
7.
Cell Biochem Funct ; 27(6): 395-401, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19639578

RESUMO

The definite diagnosis of Alzheimer's disease (AD) is based on post mortem pathological examination. To date, there is no laboratory test that can discriminate AD patients from healthy individuals. In the perspective of recent knowledge, there are three cerebrospinal fluid (CSF) markers which have the highest sensitivity and specificity: A beta(1-40), A beta(1-42), and p-tau. In the present study, 15 'Probable Alzheimer's Disease' (PAD) patients and 15 control subjects were included. PAD patients were selected from the patients of Dokuz Eylül University Neurology Department Dementia outpatient clinic and control subjects were selected from the patients who were undergone epidural anesthesia because of any surgical operation. The concentrations of Ab1-40, Ab1-42, and p-tau in CSF were quantified by using ELISA. Also, the effects of 'PAD patients' CSF on the survival of PC12 cell line were assessed. There was a significant decrease of Ab1-40 and increase of p-tau in patients with AD when compared with controls. Ab1-42 concentration was not significantly different between groups. There was a positive correlation between duration of the disease and CSF of p-tau concentration in patients with AD. There was no significant difference in cell line viability values between groups.


Assuntos
Doença de Alzheimer/líquido cefalorraquidiano , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Biomarcadores/líquido cefalorraquidiano , Citotoxinas/líquido cefalorraquidiano , Fragmentos de Peptídeos/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano , Idoso , Doença de Alzheimer/diagnóstico , Peptídeos beta-Amiloides/toxicidade , Animais , Sobrevivência Celular , Distribuição de Qui-Quadrado , Citotoxinas/toxicidade , Demência , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Células PC12 , Fragmentos de Peptídeos/toxicidade , Curva ROC , Ratos , Estatísticas não Paramétricas , Proteínas tau/toxicidade
8.
Neurol Sci ; 30(3): 263-7, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19294332

RESUMO

In the present study, we determined the significance of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in Alzheimer's disease (AD). We characterized the expression of TRAIL protein in the cerebrospinal fluid (CSF) and serum with ELISA and TRAIL mRNA in the peripheral blood mononuclear cells (PBMCs) with real-time PCR in 22 patients with AD and 20 control cases. We could not find TRAIL protein in the CSF samples. The concentration of TRAIL protein in sera from patients with AD was not different from controls. However, there was an inverse correlation between serum TRAIL levels and Mini-Mental State Examination scores in AD patients. Also we did not find significant difference in TRAIL mRNA in the PBMCs of patients with AD when compared with control group. Our data indicate that TRAIL serum level decreases in the late stage of disease.


Assuntos
Doença de Alzheimer/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Idoso , Doença de Alzheimer/sangue , Doença de Alzheimer/líquido cefalorraquidiano , Biomarcadores/sangue , Biomarcadores/líquido cefalorraquidiano , Estudos de Casos e Controles , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Entrevista Psiquiátrica Padronizada , Pessoa de Meia-Idade , RNA Mensageiro/análise , Valores de Referência , Estatísticas não Paramétricas , Ligante Indutor de Apoptose Relacionado a TNF/sangue , Ligante Indutor de Apoptose Relacionado a TNF/líquido cefalorraquidiano , Ligante Indutor de Apoptose Relacionado a TNF/genética
9.
Front Immunol ; 10: 1511, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31327964

RESUMO

Inflammation is a crucial component of various stress-induced responses that contributes to the pathogenesis of major depressive disorder (MDD). Depressive-like behavior (DLB) is characterized by decreased mobility and depressive behavior that occurs in systemic infection induced by Lipopolysaccharide (LPS) in experimental animals and is considered as a model of exacerbation of MDD. We assessed the effects of melatonin on behavioral changes and inflammatory cytokine expression in hippocampus of mice in LPS-induced DLB, as well as its effects on NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome activation, oxidative stress and pyroptotic cell death in murine microglia in vitro. Intraperitoneal 5 mg/kg dose of LPS was used to mimic depressive-like behaviors and melatonin was given at a dose of 500 mg/kg for 4 times with 6 h intervals, starting at 2 h before LPS administration. Behavioral assessment was carried out at 24 h post-LPS injection by tail suspension and forced swimming tests. Additionally, hippocampal cytokine and NLRP3 protein levels were estimated. Melatonin increased mobility time of LPS-induced DLB mice and suppressed NLRP3 expression and interleukin-1ß (IL-1ß) cleavage in the hippocampus. Immunofluorescence staining of hippocampal tissue showed that NLRP3 is mainly expressed in ionized calcium-binding adapter molecule 1 (Iba1) -positive microglia. Our results show that melatonin prevents LPS and Adenosine triphosphate (ATP) induced NLRP3 inflammasome activation in murine microglia in vitro, evidenced by inhibition of NLRP3 expression, Apoptosis-associated speck-like protein containing a CARD (ASC) speck formation, caspase-1 cleavage and interleukin-1ß (IL-1ß) maturation and secretion. Additionally, melatonin inhibits pyroptosis, production of mitochondrial and cytosolic reactive oxygen species (ROS) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling. The beneficial effects of melatonin on NLRP3 inflammasome activation were associated with nuclear factor erythroid 2-related factor 2 (Nrf2) and Silent information regulator 2 homolog 1 (SIRT1) activation, which were reversed by Nrf2 siRNA and SIRT1 inhibitor treatment.


Assuntos
Depressão/tratamento farmacológico , Inflamassomos/metabolismo , Melatonina/farmacologia , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Sirtuína 1/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Linhagem Celular Transformada , Sobrevivência Celular/efeitos dos fármacos , Depressão/induzido quimicamente , Feminino , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Microglia/citologia , Microglia/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/genética , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/genética , Transfecção
10.
Adv Protein Chem Struct Biol ; 112: 309-357, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29680240

RESUMO

During the past 35 years, recombinant DNA technology has allowed the production of a wide range of hematopoietic and neurotrophic growth factors including erythropoietin (EPO). These have emerged as promising protein drugs in various human diseases. Accumulated evidences have recently demonstrated the neuroprotective effect of EPO in preclinical models of acute and chronic degenerative disorders. Nevertheless, tissue protective effect of EPO could not be translated to the clinical trials because of common lethal thromboembolic events, erythropoiesis and hypertension. Although chemically modified nonerythropoietic analogs of EPO bypass these side effects, high expense, development of antidrug antibodies, and promotion of tumorigenicity are still concern especially in long-term use. As an alternative, nonerythropoietic EPO mimetic peptides can be used as candidate drugs with their high potency and selectivity, easy production, low cost, and immunogenicity properties. Recent experimental studies suggest that these peptides prevent ischemic brain injury and neuroinflammation. The results of clinical trial in patients with neuropathic pain are also promising. Herein, we summarize these studies and review advanced experimental and in silico methods in peptide drug discovery.


Assuntos
Eritropoetina/química , Inflamação/tratamento farmacológico , Doenças Neurodegenerativas/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Peptídeos/uso terapêutico , Animais , Humanos , Fármacos Neuroprotetores/química , Peptídeos/química
11.
Front Immunol ; 9: 36, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29410668

RESUMO

Sulforaphane (SFN) is a natural product with cytoprotective, anti-inflammatory, and antioxidant effects. In this study, we evaluated the mechanisms of its effects on lipopolysaccharide (LPS)-induced cell death, inflammation, oxidative stress, and polarization in murine microglia. We found that SFN protects N9 microglial cells upon LPS-induced cell death and suppresses LPS-induced levels of secreted pro-inflammatory cytokines, tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6. SFN is also a potent inducer of redox sensitive transcription factor, nuclear factor erythroid 2-related factor 2 (Nrf2), which is responsible for the transcription of antioxidant, cytoprotective, and anti-inflammatory genes. SFN induced translocation of Nrf2 to the nucleus via extracellular signal-regulated kinase 1/2 (ERK1/2) pathway activation. siRNA-mediated knockdown study showed that the effects of SFN on LPS-induced reactive oxygen species, reactive nitrogen species, and pro-inflammatory cytokine production and cell death are partly Nrf2 dependent. Mox phenotype is a novel microglial phenotype that has roles in oxidative stress responses. Our results suggested that SFN induced the Mox phenotype in murine microglia through Nrf2 pathway. SFN also alleviated LPS-induced expression of inflammatory microRNA, miR-155. Finally, SFN inhibits microglia-mediated neurotoxicity as demonstrated by conditioned medium and co-culture experiments. In conclusion, SFN exerts protective effects on microglia and modulates the microglial activation state.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Isotiocianatos/farmacologia , MicroRNAs/biossíntese , Microglia/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Elementos de Resposta Antioxidante/fisiologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Citocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Inflamação/tratamento farmacológico , Lipopolissacarídeos , Fator de Transcrição MafK/metabolismo , Camundongos , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/genética , Sulfóxidos
12.
Brain Res ; 1164: 14-23, 2007 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-17644074

RESUMO

Periventricular leukomalacia (PVL), the dominant form of brain injury in premature infants, is characterized by white matter injury (WMI) and is associated with cerebral palsy. The pathogenesis of PVL is complex and likely involves ischemia/reperfusion, free radical formation, excitotoxicity, impaired regulation of cerebral blood flow, a procoagulant state, and inflammatory mechanisms associated with maternal and/or fetal infection. Using an established animal model of human PVL, we investigated whether activated protein C (APC), an anti-coagulant factor with anti-inflammatory, anti-apoptotic, anti-oxidant, and cytoprotective activities, could reduce endotoxin-induced WMI in the developing rat brain. Intraperitoneal injections of lipopolysaccharide (LPS) (0.5 mg/kg body weight) were given at embryonic days 18 (E18) and 19 (E19) to pregnant Sprague-Dawley rats; control rats were injected with sterile saline. A single intravenous injection of recombinant human (rh) APC (0.2 mg /kg body weight) was given to pregnant rats following the second LPS dose on embryonic day 19 (E19). Reduced cell death in white matter and hypomyelination were shown on TUNEL and myelin basic protein (MBP) staining, respectively, on late postnatal days (P7) in APC-treated groups. There were significantly fewer TUNEL+nuclei in the periventricular WM in the APC+LPS group than in the untreated LPS group. Compared to the APC+LPS and control group, MBP expression was weak in the LPS group on P7, indicating endotoxin-induced hypomyelination in the developing rat brain. APC attenuated the LPS-induced protein expression of inflammatory cytokines, tumor necrosis factor-alpha, and interleukin-6, as evaluated by ELISA in neonatal rat brains. A single intraperitoneal injection of rhAPC (0.2 mg/kg body weight) to neonatal rats on P1 also had similar protective and anti-inflammatory effects against maternally administered LPS. Collectively, these data support the hypothesis that APC may provide protection against an endotoxin-evoked inflammatory response and WMI in the developing rat brain. Moreover, our results suggest that the possible use of APC in treatment of preterm infants and pregnant women with maternal or placental infection may minimize the risk of PVL and cerebral palsy.


Assuntos
Encéfalo/efeitos dos fármacos , Encefalite/tratamento farmacológico , Endotoxinas/antagonistas & inibidores , Leucomalácia Periventricular/tratamento farmacológico , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Proteína C/farmacologia , Animais , Animais Recém-Nascidos , Anticoagulantes/metabolismo , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Infecções Bacterianas do Sistema Nervoso Central/complicações , Infecções Bacterianas do Sistema Nervoso Central/fisiopatologia , Paralisia Cerebral/etiologia , Paralisia Cerebral/fisiopatologia , Paralisia Cerebral/prevenção & controle , Modelos Animais de Doenças , Encefalite/complicações , Encefalite/microbiologia , Endotoxinas/toxicidade , Feminino , Humanos , Recém-Nascido , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Injeções Intravenosas , Leucomalácia Periventricular/fisiopatologia , Leucomalácia Periventricular/prevenção & controle , Lipopolissacarídeos/farmacologia , Proteína Básica da Mielina/efeitos dos fármacos , Proteína Básica da Mielina/metabolismo , Fibras Nervosas Mielinizadas/imunologia , Fibras Nervosas Mielinizadas/patologia , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Gravidez , Proteína C/metabolismo , Proteína C/uso terapêutico , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico
13.
Vitam Horm ; 105: 249-271, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28629521

RESUMO

Erythropoietin (EPO) is an erythropoiesis stimulating growth factor and hormone. EPO has been widely used in the treatment of chronic renal failure, cancer, and chemotherapy-related anemia for three decades. However, many clinical trials showed that EPO treatment may be associated with tumorigenesis and cancer progression. EPO is able to cross blood-brain barriers, and this may lead to an increased possibility of central nervous system tumors such as glioblastoma. Indeed, EPO promotes glioblastoma growth and invasion in animal studies. Additionally, EPO increases glioblastoma cell survival, proliferation, migration, invasion, and chemoresistancy in vitro. However, the exact mechanisms of cancer progression induced by EPO treatment are not fully understood. Posttranscriptional gene regulation through microRNAs may contribute to EPO's cellular and biological effects in tumor progression. Here, we aimed to study whether tumor suppressive microRNA, miR-451, counteracts the positive effects of EPO on U87 human glioblastoma cell line. Migration and invasion were evaluated by scratch assay and transwell invasion assay, respectively. We found that EPO decreased basal miR-451 expression and increased cell proliferation, migration, invasion, and cisplatin chemoresistancy in vitro. miR-451 overexpression by transfection of its mimic significantly reversed these effects. Furthermore, ectopic expression of miR-451 inhibited expression of its own target genes, such as metalloproteinases-2 and -9, which are stimulated by EPO treatment and involved in carcinogenesis processes, especially invasion. These findings suggest that miR-451 mimic delivery may be useful as adjuvant therapy in addition to chemotherapy and anemia treatment by EPO and should be tested in experimental glioblastoma models.


Assuntos
Eritropoetina/farmacologia , Glioblastoma/metabolismo , MicroRNAs/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Movimento Celular , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos , Eritropoetina/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , MicroRNAs/genética , Invasividade Neoplásica , Polienos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia
16.
Front Cell Neurosci ; 9: 209, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26074776

RESUMO

Lithium is a mood stabilizing agent commonly used for the treatment of bipolar disorder. Here, we investigated the potential neuroprotective effect of lithium against paraquat toxicity and its underlying mechanisms in vitro. SH-SY5Y human neuroblastoma cells were treated with paraquat (PQ) 0.5 mM concentration after lithium pretreatment to test lithium's capability in preventing cell toxicity. Cell death was evaluated by LDH, WST-8, and tryphan blue assays. Apoptosis was analyzed using DNA fragmentation, Annexin V immunostaining, Sub G1 cell cycle analysis, and caspase-3 activity assays. BCL2, BAX, and NRF2 protein expression were evaluated by Western-blotting and the BDNF protein level was determined with ELISA. mRNA levels of BCL2, BAX, BDNF, and NRF2 target genes (HO-1, GCS, NQO1), as well as miR-34a expression were analyzed by qPCR assay. Functional experiments were done via transfection with NRF2 siRNA and miR-34a mimic. Lithium treatment prevented paraquat induced cell death and apoptosis. Lithium treated cells showed increased anti-apoptotic protein BCL2 and decreased pro-apoptotic protein BAX expression. Lithium exerted a neurotrophic effect by increasing BDNF protein expression. It also diminished reactive oxygen species production and activated the redox sensitive transcription factor NRF2 and increased its target genes expression. Knockdown of NRF2 abolished neuroprotective, anti-apoptotic, and anti-oxidant effects of lithium. Furthermore, lithium significantly decreased both basal and PQ-induced expression of miR-34a. Transfection of miR-34a specific mimic reversed neuroprotective, anti-apoptotic, and anti-oxidant effects of lithium against PQ-toxicity. Our results revealed two novel mechanisms of lithium neuroprotection, namely NRF2 activation and miR-34a suppression.

17.
Immunol Lett ; 85(3): 271-4, 2003 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-12663142

RESUMO

In this study, it is reported that neonatal murine microglia and N9 murine microglial cell line express tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL). TRAIL protein and mRNA expression in murine microglia greatly upregulate upon stimulation with interferon gamma (IFNgamma) or lipopolysaccharide (LPS) as revealed by immunoprecipitation-immunoblotting, reverse transcriptase-polymerase chain reaction (RT-PCR) and flow cytometry techniques. IFNgamma and LPS act synergistically to induce TRAIL expression on both translational and transcriptional levels. The upregulated microglial TRAIL in inflammatory conditions may involve in the cytotoxic effect of these cells and play a role in neurodegenerative processes.


Assuntos
Interferon gama/metabolismo , Lipopolissacarídeos/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglia/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima/fisiologia , Animais , Proteínas Reguladoras de Apoptose , Citometria de Fluxo , Immunoblotting , Glicoproteínas de Membrana/genética , Camundongos , Testes de Precipitina , RNA Mensageiro , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/genética
18.
Brain Res Mol Brain Res ; 132(2): 260-70, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15582163

RESUMO

One of the most exciting findings in recent years has been the discovery of RNA interference (RNAi). RNAi is an important system that allows sequence-specific gene silencing through targeted degradation of mRNA by cognate double-stranded RNA (dsRNA). RNAi plays a role in endogenous cellular processes, such as developmental control and heterochromatin formation, and serves as an antiviral defense mechanism. Recent findings suggest that RNAi and related pathways are involved in protecting the genome against instabilities caused by transposons and repetitive sequences. Several rapidly developing RNAi methodologies provide a new approach for elucidation of gene functions. RNAi technology is also currently being evaluated as a potentially useful method to develop highly specific dsRNA-based gene-silencing therapeutics. In this paper, we review the use of RNAi in neuroscience research and as a possible therapeutic tool for treatment of neurological diseases.


Assuntos
Genômica , Doenças Neurodegenerativas/fisiopatologia , Neurociências/métodos , Interferência de RNA , Animais , Humanos , Doenças Neurodegenerativas/genética
19.
Restor Neurol Neurosci ; 22(2): 105-19, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15272145

RESUMO

PURPOSE: To provide an overview of the current knowledge on neuroprotective properties of Erythropoietin (Epo), mechanisms by which Epo produces neuroprotection, and signaling pathways regulated by Epo in the nervous system. METHODS: The Medline database was searched for articles on the neuroprotective properties of Epo. Experimental and clinical studies were systematically reviewed. RESULTS: In addition to promoting the survival, proliferation, and differentiation of immature erythroid cells, Epo and the Epo receptor (EpoR) have recently been shown to exist and function in the nervous system. The Epo/EpoR system plays a critical role in neurodevelopment and neuroprotection. Epo ameliorates or prevents neuronal injury by neuroprotective, anti-apoptotic, anti-inflammatory, anti-oxidant, angiogenic, neurogenic and neurotrophic effects in cell culture and animal models of neurological diseases. The clinical effectiveness of recombinant human Epo in ischemic stroke in human patients has also been reported recently. CONCLUSION: Recent studies suggest that Epo is a potential novel neurotherapeutic agent and further clinical studies are warranted.


Assuntos
Eritropoetina/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Animais , Humanos , Doenças do Sistema Nervoso/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
20.
Behav Brain Res ; 153(1): 77-86, 2004 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-15219709

RESUMO

It is well known that neonatal hypoxic-ischemic brain injury leads to mental retardation and deficits in cognitive abilities such as learning and memory in human beings. The ameliorative effect of erythropoietin (Epo) on experimental hypoxic-ischemic brain injury in neonatal rats has been recently reported. However, the effect of Epo on cognitive abilities in the hypoxic-ischemic brain injury model is unknown. The aim of this study is to investigate the effects of Epo on learning-memory, behavior and neurodegeneration induced by hypoxia-ischemia. Seven days old Wistar Albino rat pups have been used in the study (n = 28). Experimental groups in the study were: (1) saline-treated hypoxia-ischemia group, (2) Epo-treated (i.p., 1000 U/kg) hypoxia-ischemia group, (3) sham-operated group, (4) control group. In hypoxia-ischemia groups, left common carotid artery was ligated permanently on the seventh postnatal day. Two hours after the procedure, hypoxia (92% nitrogen and 8% oxygen) was induced for 2.5 h. Epo was administered as a single dose immediately after the hypoxia period. When pups were 22 days old, learning experiments were performed using Morris water maze. On the 20th week, when brain development is accepted to be complete, learning experiments were repeated. Rats were then perfused and brains removed for macroscopic and microscopic evaluation. Epo treatment immediately after hypoxic-ischemic insult significantly improved long-term neurobehavioral achievements when tested during the subsequent phase of brain maturation and even into adulthood. Histopathological evaluation demonstrated that Epo also significantly diminished brain injury and spared hippocampal CA1 neurons. In conclusion, Epo administrated as a single dose immediately after neonatal hypoxic-ischemic insult provides benefit over a prolonged period in the still developing rat brain. Since the wide use of Epo in premature newborns, this agent may be potentially beneficial in treating asphyxial brain damage in the perinatal period.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Eritropoetina/uso terapêutico , Hipóxia-Isquemia Encefálica/complicações , Transtornos da Memória/tratamento farmacológico , Análise de Variância , Animais , Animais Recém-Nascidos , Comportamento Animal , Lesões Encefálicas/etiologia , Contagem de Células/métodos , Reação de Fuga/efeitos dos fármacos , Lateralidade Funcional , Hipocampo/citologia , Hipóxia-Isquemia Encefálica/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Transtornos da Memória/etiologia , Neurônios/patologia , Ratos , Ratos Wistar , Tempo de Reação/efeitos dos fármacos , Comportamento Espacial/efeitos dos fármacos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA