Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neurotoxicology ; 95: 12-22, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36623431

RESUMO

Alzheimer's disease (AD) is a neurodegenerative disorder with progressive memory loss in dementia. Gold nanoparticles (AuNPs) were reported beneficial for human neural stem cells (hNSCs) treated with Amyloid-beta (Aß), but the neuroprotective mechanisms still are unknown. First, the hNSCs induced by Aß to construct AD cell model in vitro and AuNPs was performed to assess the therapeutic effect of Aß-targeted AD treatment. Then, we investigated the effects of AuNPs on hNSCs viability and proinflammatory factors (interleukin 6 and tumor necrosis factor-alpha) by Cell Counting Kit-8 (CCK-8) and enzyme-linked immunosorbent (ELISA). FACS was carried out to determinate Tuj-1 and glial fibrillary acidic protein (GFAP). Reactive oxygen species (ROS) generation and mitochondrial membrane potential was evaluated by ROS and JC-1 assay kit. In addition, miRNA array was used to systematically detect the differential miRNAs. Dual-luciferase reporter assay was applied to verify the targeting relationship between miR-21-5p and the suppressor of cytokine signalling 6(SOCS6). Quantitative PCR (qPCR) and Western blot assessments were also used to detect related gene expression intracellularly or in the supernatant. The results demonstrate that AuNPs co-treatment repressed the high expression of total tau (T-tau), phosphorylated tau (P-tau), and Aß protein, and reduced apoptosis rate of hNSCs. Aß-induced decreased mitochondrial membrane potential and mitochondria in the hNSCs were damaged, while AuNPs co-treatment showed a protective effect on mitochondrial membrane potential. Co-treatment with AuNPs significantly increased dynamin-related protein 1 (DRP1), nuclear respiratory factor 1 (NRF1), and mitochondrial transcription factor A (TFAM) mRNA levels. AuNPs may improve mitochondrial function impairment due to Aß by elevating mitochondrial membrane potential, upregulating regulators of mitochondrial biogenesis, and inhibiting ROS production. hNSCs transfected with miR-21-5p inhibitor reversed AuNPs mediated cytoprotection induced by Aß. AuNPs upregulation of miR-21-5p expression and exert a mitochondrial protective function. Overexpression of miR-21-5p contributes to enhancing the effect of cytoprotection of AuNPs. MiR-21-5p direct targeting SOCS6 and overexpression SOCS6 exerted opposite effects on hNSCs compared with miR-21-5p mimic group. In conclusion, AuNPs can protect hNSCs from Aß injury and decrease mitochondrial damage by regulating the miR-21-5p/SOCS6 pathway.


Assuntos
Doença de Alzheimer , Nanopartículas Metálicas , MicroRNAs , Células-Tronco Neurais , Humanos , Peptídeos beta-Amiloides/toxicidade , Peptídeos beta-Amiloides/metabolismo , Ouro/metabolismo , Ouro/farmacologia , Ouro/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Nanopartículas Metálicas/toxicidade , Doença de Alzheimer/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/farmacologia , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico
2.
Arterioscler Thromb Vasc Biol ; 34(9): 1953-60, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25012131

RESUMO

OBJECTIVE: Activation of Janus kinase/signal transducers and activators of transcription (STAT) pathway by hyperglycemia and dislypidemia contributes to the progression of diabetic complications, including atherosclerosis. Suppressor of cytokine signaling (SOCS) proteins negatively regulate Janus kinase/STAT and have emerged as promising target for anti-inflammatory therapies. We investigated whether a cell-permeable lipopeptide corresponding to the kinase inhibitory region of SOCS1 could reduce atherosclerosis in diabetic mice and identified the mechanisms involved. APPROACH AND RESULTS: Streptozotocin-induced diabetic apolipoprotein E-deficient mice (aged 8 and 22 weeks) were given intraperitoneal injections of vehicle, SOCS1-derived peptide, or control mutant peptide for 6 to 10 weeks. SOCS1 therapy suppressed STAT1/STAT3 activation in atherosclerotic plaques of diabetic mice and significantly reduced lesion size at both early and advanced stages of lesion development compared with vehicle group. Plaque characterization demonstrated that SOCS1 peptide decreased the accumulation of lipids, macrophages, and T lymphocytes, whereas increasing collagen and smooth muscle cell content. This atheroprotective effect was accompanied by systemic (reduced proinflammatory Ly6C(high) monocytes and splenic cytokine expression) and local (reduced aortic expression of chemokines and cytokines) mechanisms, without impact on metabolic parameters. In vitro, SOCS1 peptide dose dependently inhibited STAT1/STAT3 activation and target gene expression in vascular smooth muscle cells and macrophages and also suppressed cytokine-induced cell migration and adhesion processes. CONCLUSIONS: SOCS1-based targeting Janus kinase/STAT restrains key mechanisms of atherogenesis in diabetic mice, thereby preventing plaque formation and increasing plaque stability. Approaches to mimic native SOCS1 functions may have a therapeutic potential to retard the progression of diabetic complications.


Assuntos
Diabetes Mellitus Experimental/complicações , Inflamação/tratamento farmacológico , Janus Quinases/antagonistas & inibidores , Placa Aterosclerótica/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Fator de Transcrição STAT1/antagonistas & inibidores , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico , Sequência de Aminoácidos , Animais , Linhagem Celular , Dicroísmo Circular , Diabetes Mellitus Experimental/metabolismo , Progressão da Doença , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inflamação/enzimologia , Inflamação/etiologia , Interferon gama/farmacologia , Interleucina-6/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Dados de Sequência Molecular , Terapia de Alvo Molecular , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Placa Aterosclerótica/enzimologia , Placa Aterosclerótica/etiologia , Conformação Proteica , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Fator de Transcrição STAT1/fisiologia , Fator de Transcrição STAT3/fisiologia , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/química , Proteínas Supressoras da Sinalização de Citocina/farmacocinética , Proteínas Supressoras da Sinalização de Citocina/farmacologia
3.
Hepatology ; 47(1): 105-12, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17935223

RESUMO

UNLABELLED: The constitutive activation of signal transducer and activator of transcription 3 (STAT3) participates in carcinogenesis through up-regulation of genes encoding apoptosis inhibitors and cell cycle regulators, such as Bcl-xL, cyclins D1 and D2, and c-myc. Suppressor of cytokine signaling 3 (SOCS3) is one of the negative regulators of cytokine signaling and is frequently silenced in diverse cancers. In this study, we explored whether restoration of SOCS3 by oncolytic adenoviral vectors could inhibit the constitutive activation of the Janus kinase/STAT pathway and suppress tumor growth. Our data showed that SOCS3 was down-expressed in all liver tumor cell lines. The incorporation of SOCS3 or SOCS3 fused with cell-penetrating peptides (cpp-SOCS3) did not alter adenoviral replication selectively in liver tumor cells. The infection of cells with adenovirus CN305 (AdCN305)-SOCS3 and AdCN305-cpp-SOCS3 resulted in dramatic cytotoxicity in liver tumor cells. However, no cytotoxic effect was observed in normal cells infected with these vectors. Infection of liver tumor cells with AdCN305-SOCS3 and AdCN305-cpp-SOCS3 resulted in nearly complete inhibition of STAT3 phosphorylation and down-regulation of cyclin D1 and Bcl-xL. Treatment of the established tumor by AdCN305-SOCS3 and AdCN305-cpp-SOCS3 caused significant suppression of tumor growth. The suppression of tumor growth was due to the inhibition of STAT3 phosphorylation and induction of tumor cell death. CONCLUSION: This study suggests that transfer of SOCS3 by an oncolytic adenovirus represents a potent approach for cancer therapy.


Assuntos
Adenoviridae/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Terapia Viral Oncolítica/métodos , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico , Animais , Proteínas de Transporte/genética , Morte Celular/efeitos dos fármacos , Linhagem Celular , Expressão Gênica , Vetores Genéticos , Humanos , Camundongos , Camundongos Nus , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/farmacologia , Replicação Viral
5.
Cancer Gene Ther ; 13(2): 194-202, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16082380

RESUMO

Multiple myeloma (MM) accounts for 10% of hematological malignant disorders. Its refractory nature indicates the necessity of developing novel therapeutic modalities. Since interleukin 6 (IL-6) is one of the major growth factors for MM cells, we expressed suppressor of cytokine signaling-1 (SOCS-1), one of the blockades of IL-6 receptor downstream signaling, to suppress the proliferation of MM cells. Because MM cells are resistant to conventional adenoviral vector infection, we utilized infectivity-enhanced adenoviral vectors with an RGD4C motif in the adenoviral fiber-knob region (RGD-modified vector). In infectivity analysis, RGD-modified vectors were superior to unmodified controls in the majority of the MM cell lines tested. The overexpression of SOCS-1 using infectivity-enhanced adenoviral vectors achieved growth suppression in IL-6-dependent MM cells, but not in the IL-6-independent cells. IL-6-induced STAT3 phosphorylation was suppressed in IL-6-dependent cells, indicating that the signal transduction cascade of the IL-6 receptor signaling was blocked. In aggregate, SOCS-1 overexpression with RGD-modified adenoviral vectors achieved the antiproliferative effect in IL-6-dependent MM cells. These results provide an initial proof-of-principle of the anticancer effect of SOCS-1 expression vector as well as a promise for the future development of therapeutic modality for MM based on this vector.


Assuntos
Adenoviridae/genética , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/imunologia , Terapia Genética/métodos , Vetores Genéticos/genética , Mieloma Múltiplo/terapia , Transdução de Sinais/imunologia , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Western Blotting , Linhagem Celular Tumoral , Eletroforese em Gel de Poliacrilamida , Humanos , Interleucina-6/metabolismo , Mieloma Múltiplo/imunologia , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico
6.
J Neuroimmunol ; 168(1-2): 118-27, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16154209

RESUMO

Suppressors of cytokine signaling (SOCS) are implicated in immunopathogenic mechanisms of autoimmune diseases. We show here that SOCS expression in retina is temporarily correlated with progression of experimental autoimmune uveitis (EAU), an organ-specific autoimmune disease that serves as model of human uveitis. Peak of EAU correlates with highest SOCS genes expression while disease resolution coincides with their down-regulation. Surprisingly, SOCS5 is constitutively expressed in retina. SOCS5 expression increases significantly during EAU and remains elevated even after disease resolution. Our data suggest that cytokine-inducible SOCS members may be involved in negative regulation of inflammatory cytokines activities during EAU, while constitutively expressed SOCS5 may have neuroprotective functions.


Assuntos
Citocinas/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Retina/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico , Uveíte/metabolismo , Animais , Western Blotting/métodos , Antígenos CD4/metabolismo , Proliferação de Células , Citocinas/genética , Modelos Animais de Doenças , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Ensaio de Imunoadsorção Enzimática/métodos , Expressão Gênica/efeitos dos fármacos , Camundongos , Fármacos Neuroprotetores/farmacologia , RNA Mensageiro/biossíntese , Retina/metabolismo , Retina/fisiologia , Proteínas de Ligação ao Retinol , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Proteínas Supressoras da Sinalização de Citocina/farmacologia , Linfócitos T/metabolismo , Fatores de Tempo , Uveíte/induzido quimicamente , Uveíte/prevenção & controle
7.
Nat Med ; 11(8): 892-8, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16007096

RESUMO

Suppressor of cytokine signaling (SOCS) 3 attenuates proinflammatory signaling mediated by the signal transducer and activator of transcription (STAT) family of proteins. But acute inflammation can occur after exposure to pathogen-derived inducers staphylococcal enterotoxin B (SEB) and lipopolysaccharide (LPS), or the lectin concanavalin A (ConA), suggesting that physiologic levels of SOCS3 are insufficient to stem proinflammatory signaling under pathogenic circumstances. To test this hypothesis, we developed recombinant cell-penetrating forms of SOCS3 (CP-SOCS3) for intracellular delivery to counteract SEB-, LPS- and ConA-induced inflammation. We found that CP-SOCS3 was distributed in multiple organs within 2 h and persisted for at least 8 h in leukocytes and lymphocytes. CP-SOCS3 protected animals from lethal effects of SEB and LPS by reducing production of inflammatory cytokines and attenuating liver apoptosis and hemorrhagic necrosis. It also reduced ConA-induced liver apoptosis. Thus, replenishing the intracellular stores of SOCS3 with CP-SOCS3 effectively suppresses the devastating effects of acute inflammation.


Assuntos
Apoptose/efeitos dos fármacos , Enterotoxinas/toxicidade , Inflamação/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Infecções Estafilocócicas/complicações , Proteínas Supressoras da Sinalização de Citocina/uso terapêutico , Animais , Concanavalina A/toxicidade , Citocinas/sangue , Inflamação/etiologia , Leucócitos/metabolismo , Lipopolissacarídeos/toxicidade , Fígado/patologia , Linfócitos/metabolismo , Camundongos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT1/metabolismo , Infecções Estafilocócicas/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Proteínas Supressoras da Sinalização de Citocina/farmacocinética , Proteínas Supressoras da Sinalização de Citocina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...