Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Arch Virol ; 168(4): 128, 2023 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-37002434

RESUMO

Due to recurrence and resistance to chemotherapy, the current standard therapeutics are not fully effective against ovarian cancer. Therefore, we aimed to find an effective approach to improve the prognosis and therapy of ovarian cancer. NG34ScFvPD-1 is a modified oncolytic herpes simplex virus NG34 strain that expresses a single-chain antibody against programmed cell death protein 1 (PD-1) (ScFvPD-1). We assessed its efficacy and its regulatory mechanism in a mouse model of ovarian cancer. Enzyme-linked immunosorbent assay and western blot techniques were used to measure protein expression. Oncolysis caused by NG34ScFvPD-1 was examined using cytotoxicity and replication assays. The mechanism by which NG34ScFvPD-1 regulates apoptosis of ovarian cancer cells in vitro was also evaluated. We assessed the antitumor immunity and therapeutic potency of NG34ScFvPD-1 in combination with a phosphoinositide 3-kinase (PI3K) inhibitor. We found that NG34ScFvPD-1-infected ovarian cancer cells expressed and secreted ScFvPD-1, which bound mouse PD-1. The insertion of the ScFvPD-1 sequence did not inhibit the oncolytic activity of NG34ScFvPD-1, which induced apoptosis of ovarian cancer cells via the caspase-dependent pathway in vitro and activated the PI3K/AKT signaling pathway. Synergy was observed between NG34ScFvPD-1 and a PI3K inhibitor, and the combination was able to suppress tumor development, to prolong survival, and to elicit potent antitumor immunity. Thus, inhibition of PI3K enhanced the potent antitumor immunity induced by NG34ScFvPD-1 against ovarian cancer.


Assuntos
Herpesvirus Humano 1 , Terapia Viral Oncolítica , Vírus Oncolíticos , Neoplasias Ovarianas , Anticorpos de Cadeia Única , Humanos , Feminino , Camundongos , Animais , Fosfatidilinositol 3-Quinases , Inibidores de Fosfoinositídeo-3 Quinase , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Anticorpos de Cadeia Única/genética , Receptor de Morte Celular Programada 1 , Neoplasias Ovarianas/terapia , Neoplasias Ovarianas/patologia , Linhagem Celular Tumoral
2.
Am J Respir Cell Mol Biol ; 64(6): 747-759, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33705682

RESUMO

Smoking-mediated reprogramming of the phenotype and function of airway basal cells (BCs) disrupts airway homeostasis and is an early event in chronic obstructive pulmonary disease (COPD)-associated airway remodeling. Here, we examined the expression and regulation of the transmembrane glycoprotein TROP2 (trophoblast antigen 2), a putative stem cell marker in airway BCs, in lung tissue samples from healthy smokers and healthy nonsmokers and in models in culture to identify therapeutic targets. TROP2 expression was upregulated in the airway epithelia of smokers and positively correlated with the smoking index. In vitro, cigarette smoke extract (CSE) induced TROP2 expression in airway BCs in a time- and dose-dependent manner. The p38 MAPK and NF-κB pathways were also activated by CSE, and their specific antagonists inhibited CSE-induced TROP2 expression. A therapeutic component derived from traditional Chinese medicine, ginsenoside Rb3, inhibited CSE-induced TROP2 expression as well as activation of the p38 MAPK and NF-κB pathways in BCs in monolayer culture. Furthermore, ginsenoside Rb3 prevented the increase in TROP2 expression and antagonized CSE-induced BC hyperplasia and expression of inflammatory factors and epithelial-mesenchymal transition changes in an air-liquid culture model. Thus, CSE-induced TROP2 is a possible biomarker for early changes in the epithelium of smokers, and ginsenoside Rb3 may serve as a therapeutic molecule, preventing the disruption of epithelial homeostasis in COPD.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Ginsenosídeos/farmacologia , Pulmão/patologia , NF-kappa B/metabolismo , Transdução de Sinais , Fumar/efeitos adversos , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Epitélio/metabolismo , Feminino , Humanos , Hiperplasia , Mediadores da Inflamação/metabolismo , Masculino , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
3.
BMC Neurosci ; 20(1): 41, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31387531

RESUMO

BACKGROUND: Oxidative stress and neuroinflammation are central pathogenic mechanisms common to many neurological diseases. Isoliquiritigenin (ISL) is a flavonoid in licorice with multiple pharmacological properties, including anti-inflammatory activity, and has demonstrated protective efficacy against acute neural injury. However, potential actions against cognitive impairments have not been examined extensively. We established a rat model of cognitive impairment by intracerebroventricular injection of lipopolysaccharide (LPS), and examined the effects of ISL pretreatment on cognitive function, hippocampal injury, and hippocampal expression of various synaptic proteins, antioxidant enzymes, pro-inflammatory cytokines, and signaling factors controlling anti-oxidant and pro-inflammatory responses. RESULTS: Rats receiving LPS alone demonstrated spatial learning deficits in the Morris water maze test as evidenced by longer average escape latency, fewer platform crossings, and shorter average time in the target quadrant than untreated controls. ISL pretreatment reversed these deficits as well as LPS-induced decreases in the hippocampal expression levels of synaptophysin, postsynaptic density-95, brain-derived neurotrophic factor, superoxide dismutase, glutathione peroxidase, and BCL-2. ISL pretreatment also reversed LPS-induced increases in TUNEL-positive (apoptotic) cells, BAX/BCL-2 ratio, and expression levels of tumor necrosis factor-α, interleukin (IL)-1ß, IL-6, and C-C motif chemokine ligand 3. Pretreatment with ISL increased the expression levels of phosphorylated (p)-GSK-3ß, nuclear NRF2, HO-1 mRNA, and NQO1 mRNA, and reversed LPS-induced nuclear translocation of nuclear factor (NF)-κB. CONCLUSIONS: ISL protects against LPS-induced cognitive impairment and neuronal injury by promoting or maintaining antioxidant capacity and suppressing neuroinflammation, likely through phosphorylation-dependent inactivation of GSK-3ß, enhanced expression of NRF2-responsive antioxidant genes, and suppression of NF-κB-responsive pro-inflammatory genes.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Chalconas/farmacologia , Disfunção Cognitiva/prevenção & controle , Animais , Antioxidantes/metabolismo , Apoptose/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo , Disfunção Cognitiva/induzido quimicamente , Citocinas/biossíntese , Proteína 4 Homóloga a Disks-Large/biossíntese , Glutationa Peroxidase/biossíntese , Hipocampo/metabolismo , Lipopolissacarídeos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Superóxido Dismutase/biossíntese , Sinaptofisina/biossíntese , Proteína X Associada a bcl-2/biossíntese
4.
Inflammopharmacology ; 27(6): 1143-1153, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31037573

RESUMO

Epileptogenesis is a complex pathological process that occurs after an initial brain injury and involves a series of molecular events. Isoliquiritigenin (ISL), a flavonoid in licorice, is reported to have anti-inflammatory and antioxidant effects in various experimental models, but its specific roles and molecular mechanisms in the epileptogenic process following kainic acid (KA) treatment remain unclear. The purpose of this study was to explore the effects of ISL pretreatment in KA-induced epileptic rats and the underlying mechanisms. Our findings show that ISL pretreatment significantly attenuated the KA-induced expression of ionized calcium-binding adapter molecule 1 (IBα1)-labeled microglia (F(3, 20) = 97.29, p < 0.01, ηp2 = 0.94) and glial fibrillary acidic protein (GFAP)-positive astrocytes (F(3, 20) = 72.48, p < 0.01, ηp2 = 0.92), and the release of inflammatory mediators, such as TNF-α (F(3, 20) = 133.14, p < 0.01, ηp2 = 0.95), IL-1ß, and C-C motif chemokine ligand 3 (CCL3). ISL pretreatment given before KA also significantly prevented apoptotic neuronal injury by upregulating the activities of superoxide dismutase and glutathione peroxidase. It also significantly suppressed the protein levels of Toll-like receptor 4 (TLR4) (F(3, 20) = 63.23, p < 0.01, ηp2 = 0.91) and its downstream molecules, myeloid differentiation primary response 88 (MYD88), phosphorylated (p-)IκBα, and p-NF-κB. Blocking TLR4/MYD88 signaling also attenuated KA-induced neuroinflammation and neuronal damage in the hippocampus. Overall, our study demonstrates that ISL pretreatment plays neuroprotective and anti-inflammatory roles in KA-induced epileptogenesis, which may be mediated by the TLR4/MYD88 signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Chalconas/farmacologia , Epilepsia/tratamento farmacológico , Ácido Caínico/farmacologia , Fator 88 de Diferenciação Mieloide/fisiologia , Fármacos Neuroprotetores/farmacologia , Receptor 4 Toll-Like/fisiologia , Animais , Citocinas/análise , Epilepsia/fisiopatologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Microglia/efeitos dos fármacos , Microglia/fisiologia , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Ratos , Ratos Wistar , Transdução de Sinais/fisiologia , Receptor 4 Toll-Like/antagonistas & inibidores , Proteína X Associada a bcl-2/análise
5.
Biochem Biophys Res Commun ; 486(3): 693-699, 2017 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-28341121

RESUMO

The Eph family of receptor tyrosine kinases serves as key modulators of various cellular functions, including inflammation, hypertrophy and fibrosis. Recent analyses have revealed that a member of the Eph family, EphA1, plays a pivotal role in regulating insulin metabolism and kidney injury. However, the importance of EphA1 in diabetic nephropathy has not been recognized. We established a diabetic nephropathy mouse model using a high-fat diet and streptozotocin (STZ) injection. Then, the recombinant adeno-associated virus type 9 (AAV9) overexpressing EphA1 or a negative control was injected locally into the kidney. Metabolite testing and histopathological analyses of kidney fibrosis, pancreatic islet function and signaling pathways were evaluated. Our study showed that hyperglycemia, insulin resistance, and renal fibrosis accompanied the deterioration of kidney function in diabetic mice. The overexpression of EphA1 in the kidney attenuated renal fibrosis and improved kidney function but did not affect systemic glucose metabolism and pancreatic islet function. Furthermore, the overexpression of EphA1 decreased the phosphorylation of ERK1/2, JNK and MYPT1 (a substrate of Rho kinase). The overexpression of EphA1 can be therapeutically targeted to inhibit diabetic renal fibrosis, which suggests that the EphA1-Epha receptor axis may be a novel therapy target for diabetic nephropathy. Mechanistically, the overexpression of EphA1 could inhibit MAPK and the Rho pathway in diabetic kidneys.


Assuntos
Diabetes Mellitus Experimental/terapia , Nefropatias Diabéticas/terapia , Efrina-A1/genética , Vetores Genéticos/uso terapêutico , Hiperglicemia/terapia , Receptor EphA1/genética , Animais , Dependovirus/genética , Dependovirus/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Dieta Hiperlipídica , Efrina-A1/agonistas , Efrina-A1/metabolismo , Fibrose , Regulação da Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Hiperglicemia/induzido quimicamente , Hiperglicemia/genética , Hiperglicemia/patologia , Resistência à Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Glomérulos Renais/metabolismo , Glomérulos Renais/patologia , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/genética , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Receptor EphA1/agonistas , Receptor EphA1/metabolismo , Transdução de Sinais , Estreptozocina
6.
J Cell Mol Med ; 19(4): 836-49, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25661015

RESUMO

Macrophage migration inhibitory factor (MIF) involves the pathogenesis of atherosclerosis (AS) and increased plasma MIF levels in diabetes mellitus (DM) patients are associated with AS. Here, we have been suggested that MIF could be a critical contributor for the pathological process of diabetes-associated AS by using adenovirus-mediated RNA interference. First, streptozotocin (STZ)-induced diabetic animal model was constructed in 114 apolipoprotein E-deficient mice (apoE-/- mice) fed on a regular chow diet. Then, the animals were randomly divided into three groups: Adenovirus-mediated MIF interference (Ad-MIFi), Ad-enhanced green fluorescent protein (EGFP) and normal saline (NS) group (n ≈ 33/group). Non-diabetic apoE-/- mice (n = 35) were served as controls. Ad-MIFi, Ad-EGFP and NS were, respectively, injected into the tail vein of mice from Ad-MIFi, Ad-EGFP and NS group, which were injected repeatedly 4 weeks later. Physical, biochemical, morphological and molecular parameters were measured. The results showed that diabetic apoE-/- mice had significantly aggravated atherosclerotic lesions. MIF gene interference attenuated atherosclerotic lesions and stabilized atheromatous plaque, accompanied by the decreased macrophages and lipids deposition and inflammatory cytokines production, improved glucose intolerance and plasma cholesterol level, the decreased ratio of matrix matalloproteinase-2/tissue inhibitor of metalloproteinase-1 and plaque instability index. An increased expression of MIF and its ligand CD74 was also detected in the diabetic patients with coronary artery disease. The results suggest that MIF gene interference is able to inhibit atherosclerotic lesions and increase plaque stability in diabetic apoE-/-mice. MIF inhibition could be a novel and promising approach to the treatment of DM-associated AS.


Assuntos
Apolipoproteínas E/metabolismo , Aterosclerose/metabolismo , Diabetes Mellitus Experimental/metabolismo , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Animais , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Diferenciação de Linfócitos B/metabolismo , Apolipoproteínas E/genética , Apoptose/genética , Aterosclerose/complicações , Aterosclerose/genética , Western Blotting , Colesterol/sangue , Citocinas/genética , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Expressão Gênica , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Imuno-Histoquímica , Mediadores da Inflamação/metabolismo , Oxirredutases Intramoleculares/genética , Receptores X do Fígado , Fatores Inibidores da Migração de Macrófagos/genética , Camundongos Knockout , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
J Biol Chem ; 287(39): 32546-55, 2012 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-22859306

RESUMO

The inflammatory responses accompanying stroke are recognized to contribute to secondary ischemic injury. TIPE2 is a very recently identified negative regulator of inflammation that maintains immune homeostasis. However, it is unknown whether TIPE2 is expressed in the brain and contributes to the regulation of cerebral diseases. In this study, we explored the potential roles of TIPE2 in cerebral ischemia/reperfusion injury. TIPE2(-/-) mice were used to assess whether TIPE2 provides neuroprotection following cerebral ischemia/reperfusion induced by middle cerebral artery occlusion (MCAO), and in vitro primary cerebral cell cultures were used to investigate the expression and regulation of TIPE2. Our results show that genetic ablation of the Tipe2 gene significantly increased the cerebral volume of infarction and neurological dysfunction in mice subjected to MCAO. Flow cytometric analysis revealed more infiltrating macrophages, neutrophils, and lymphocytes in the ischemic hemisphere of TIPE2(-/-) mice. The responses to inflammatory cytokines and chemokines were significantly increased in TIPE2(-/-) mouse brain after MCAO. We further observed that TIPE2 was highly induced in WT mice after cerebral ischemia and was expressed mainly in microglia/macrophages, but not in neurons and astrocytes. Finally, we found that regulation of TIPE2 expression was associated with NADPH oxidase activity. These findings demonstrate, for the first time, that TIPE2 is involved in the pathogenesis of stroke and suggest that TIPE2 plays an essential role in a signal transduction pathway that links the inflammatory immune response to specific conditions after cerebral ischemia. Targeting TIPE2 may be a new therapeutic strategy for stroke treatment.


Assuntos
Cerebelo/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais , Acidente Vascular Cerebral/metabolismo , Animais , Cerebelo/imunologia , Cerebelo/patologia , Modelos Animais de Doenças , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout , Microglia/imunologia , Microglia/metabolismo , Microglia/patologia , NADPH Oxidases/genética , NADPH Oxidases/imunologia , NADPH Oxidases/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/imunologia , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/patologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/patologia
8.
Neurochem Res ; 38(4): 742-52, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23471594

RESUMO

Ethyl pyruvate (EP) has been reported to be neuroprotective in several models of brain injury, yet its influence on periventricular leukomalacia still remains elusive. Here we investigated whether repeated administration of EP could protect against white matter injury after hypoxia-ischemia (HI) (right common carotid artery ligation and 6 % O2 for 60 min) in post-natal 3 day rat pups. EP was injected (50 mg/kg, intraperitoneally) 10 min, 1 and 24 h after HI insult. Treatment with EP significantly reduced HI-induced ventricular enlargement, loss of developing oligodendrocytes, and hypomyelination. We further demonstrated a marked inhibitory effect of EP on inflammatory responses, as indicated by the decreased number of activated microglia and astrocytes and the reduced release of proinflammatory cytokines. Moreover, EP down-regulated the expression of cleaved caspase-3 and Bax, and up-regulated Bcl-2 expression after HI exposure. In conclusion, our results demonstrated that EP was able to provide potent protection on white matter injury through blocking the cerebral inflammatory responses and modulating the apoptotic death program of oligodendrocytes, indicating a potential neuroprotective agent in neonatal brain injury.


Assuntos
Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Piruvatos/uso terapêutico , Animais , Animais Recém-Nascidos , Astrócitos/metabolismo , Encéfalo/efeitos dos fármacos , Caspase 3/biossíntese , Ventrículos Cerebrais/patologia , Citocinas/antagonistas & inibidores , Encefalite/prevenção & controle , Hipóxia-Isquemia Encefálica/patologia , Oligodendroglia/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Ratos , Ratos Sprague-Dawley , Proteína X Associada a bcl-2/biossíntese
9.
Epilepsy Behav ; 27(1): 9-17, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23353000

RESUMO

Our aim was to examine whether neonatal lipopolysaccharide (LPS) exposure is associated with changes in microglia and whether these alternations could influence later seizure-induced neurobehavioral outcomes. Male pups were first injected intraperitoneally with either LPS or saline on postnatal day 3 (P3) and postnatal day 5 (P5). Immunohistochemical analysis showed that LPS-treated animals exhibited increased microglia activation that persisted into adolescence. At P45, seizures were induced in rats by intraperitoneal injection of kainic acid (KA). Rats treated with LPS neonatally showed significantly greater proinflammatory responses and performed significantly worse in the Y-maze, Morris water maze, and inhibitory avoidance tasks after KA insult. Treatment with minocycline at the time of neonatal LPS exposure to block LPS-induced microglia alternation attenuated the exaggerated neuroinflammatory responses and alleviated memory impairment associated with the KA insult. Our findings suggest that neonatal immune activation can predispose the brain to exacerbated behavioral deficits following seizures in adulthood, possibly by priming microglia.


Assuntos
Citocinas/metabolismo , Hipocampo/fisiologia , Transtornos da Memória/etiologia , Convulsões/complicações , Convulsões/imunologia , Análise de Variância , Animais , Animais Recém-Nascidos , Aprendizagem da Esquiva/efeitos dos fármacos , Citocinas/genética , Modelos Animais de Doenças , Feminino , Hipocampo/efeitos dos fármacos , Ácido Caínico/toxicidade , Lipopolissacarídeos/farmacologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Minociclina/farmacologia , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/induzido quimicamente , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
10.
Cells ; 11(17)2022 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-36078037

RESUMO

Atrial fibrillation (AF) is a form of sustained cardiac arrhythmia and microRNAs (miRs) play crucial roles in the pathophysiology of AF. To identify novel miR-mRNA pairs, we performed RNA-seq from atrial biopsies of persistent AF patients and non-AF patients with normal sinus rhythm (SR). Differentially expressed miRs (11 down and 9 up) and mRNAs (95 up and 82 down) were identified and hierarchically clustered in a heat map. Subsequently, GO, KEGG, and GSEA analyses were run to identify deregulated pathways. Then, miR targets were predicted in the miRDB database, and a regulatory network of negatively correlated miR-mRNA pairs was constructed using Cytoscape. To select potential candidate genes from GSEA analysis, the top-50 enriched genes in GSEA were overlaid with predicted targets of differentially deregulated miRs. Further, the protein-protein interaction (PPI) network of enriched genes in GSEA was constructed, and subsequently, GO and canonical pathway analyses were run for genes in the PPI network. Our analyses showed that TNF-α, p53, EMT, and SYDECAN1 signaling were among the highly affected pathways in AF samples. SDC-1 (SYNDECAN-1) was the top-enriched gene in p53, EMT, and SYDECAN1 signaling. Consistently, SDC-1 mRNA and protein levels were significantly higher in atrial samples of AF patients. Among negatively correlated miRs, miR-302b-3p was experimentally validated to suppress SDC-1 transcript levels. Overall, our results suggested that the miR-302b-3p/SDC-1 axis may be involved in the pathogenesis of AF.


Assuntos
Fibrilação Atrial , MicroRNAs , Fibrilação Atrial/patologia , Átrios do Coração/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/genética , Proteína Supressora de Tumor p53
11.
Acta Pharmacol Sin ; 31(10): 1395-400, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20871618

RESUMO

AIM: To examine whether danshensu could protect vascular endothelia in a rat model of hyperhomocysteinemia. METHODS: The model was established by feeding rats with a methionine-rich diet (1 g·kg⁻¹·d⁻¹) for 3 months. Immediately following the discontinuation of methionine-rich diet, rats were treated with danshensu (67.5 mg·kg⁻¹·d⁻¹, po) or saline for 3 additional months. One group of rats receiving vitamin mixture (folic acid, vitamin B12 and vitamin B6) was included as a positive control. One group of rats not exposed to methionine-rich diet was also included as a blank control. The expression of tumor necrosis factor-alpha (TNF-alpha) and intercellular adhesion molecule-1 (ICAM-1) protein in the descending aorta was examined using immunohistochemistry and Western blot. Homocysteine and blood concentration of endothelin and nitric oxide (NO) was also examined. RESULTS: Methionine-rich diet resulted in accumulation of "foam cells", up-regulated expression of TNF-alpha and ICAM-1 in the descending aorta, and significantly increased serum homocysteine. Plasma endothelin concentration was significantly increased; NO was decreased. Danshensu treatment, either simultaneous to methionine-rich diet or afterwards, attenuated the above mentioned changes. CONCLUSION: Chronic treatment with danshensu could prevent/attenuate the formation of atherosclerosis. Potential mechanisms include inhibited expression of representative proinflammatory cytokines and adhesion molecules in arterial endothelia. Changes in homocysteine and circulating molecules that control vascular contraction/relaxation via endothelial cells (eg, endothelin and NO) were also implicated.


Assuntos
Fármacos Cardiovasculares/farmacologia , Endotélio Vascular/efeitos dos fármacos , Hiper-Homocisteinemia/tratamento farmacológico , Lactatos/farmacologia , Animais , Aorta Torácica/metabolismo , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Fármacos Cardiovasculares/uso terapêutico , Dieta , Modelos Animais de Doenças , Endotelinas/sangue , Endotélio Vascular/metabolismo , Feminino , Hiper-Homocisteinemia/metabolismo , Hiper-Homocisteinemia/patologia , Molécula 1 de Adesão Intercelular/sangue , Lactatos/uso terapêutico , Masculino , Metionina , Óxido Nítrico/sangue , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-32371529

RESUMO

OBJECTIVE: Diabetic cardiomyopathy (DCM) is characterized by cardiac remodeling, dysfunction, and insulin resistance; however, the underlying mechanism has not been fully elucidated. Programmed cell death 4 (PDCD4) is a novel inflammation and apoptosis gene, but its role in type 2 DCM remains elusive. We aimed to determine if PDCD4 intervention improves DCM by affecting left ventricular remodeling, function, and insulin resistance. RESEARCH DESIGN AND METHODS: We designed a PDCD4-/- rat, established a type 2 diabetes animal model, and constructed a PDCD4 overexpressed adenovirus and PDCD4 small interfer RNA (siRNA) vectors to alter PDCD4 expression in H9c2 cardiomyocytes. Thereafter, glucose levels, lipid metabolism, echocardiography, and extent of myocardial fibrosis, inflammation, and apoptosis were compared in vivo and in vitro. RESULTS: PDCD4 deficiency improved insulin resistance, cardiac remodeling, and dysfunction in type 2 DCM rats and improved myocardial hypertrophy, fibrosis, inflammation, and apoptosis. Proliferation and transformation of cardiac fibroblasts was reduced via PDCD4 downregulation in vitro under high-glucose stimulation. Furthermore, PDCD4 regulated the myocardial phosphatidylinositol 3-kinase-protein kinase B (PI3K-AKT) pathway in vivo and in vitro. PDCD4 intervention affected cardiac remodeling, dysfunction, and insulin resistance by influencing fibrosis, inflammation, and apoptosis via the PI3K-AKT pathway in vivo. CONCLUSIONS: PDCD4 knockdown protected against left ventricular remodeling, dysfunction, and insulin resistance in type 2 DCM rats. The underlying mechanisms may involve reducing cardiomyocyte apoptosis, inflammation, fibrosis, and normalized PI3K-AKT phosphorylation. To the best of our knowledge, our study is the first to report the effects and underlying mechanisms of PDCD4 in type 2 DCM. These results provide a potential new treatment avenue for improving the prognosis of patients with type 2 DCM.


Assuntos
Diabetes Mellitus Tipo 2 , Cardiomiopatias Diabéticas , Resistência à Insulina , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/genética , Cardiomiopatias Diabéticas/genética , Humanos , Resistência à Insulina/genética , Fosfatidilinositol 3-Quinases , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Remodelação Ventricular
13.
Genet Test Mol Biomarkers ; 23(11): 807-814, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31638427

RESUMO

Background: The gene programmed cell death 5 (PDCD5) has recently been characterized as a tumor suppressor gene and is believed to be an important prognostic cancer marker; it is frequently involved in neoplastic transformation and apoptosis of tumor cells. Several studies have demonstrated a decrease or loss of expression of PDCD5 in certain tumors. However, the relevance of PDCD5 expression in human osteoclastoma and its clinicopathological significance have not been extensively studied. Methods: The aim of this study was to explore the relative transcriptional and translational expression levels of PDCD5 in 79 osteoclastoma samples using multi-modal methods of analysis. Results: Our findings showed that 52% (15/29) of osteoclastoma cases exhibited reduced PDCD5 expression at the transcriptional level, and 56% (44/79) exhibited lower PDCD5 expression at the protein level, when compared with nontumor tissue. In addition, the statistical significance of the altered PDCD5 protein expression was examined using the Campanacci grading system for osteoclastoma. More importantly, the decreased expression at the translational level was observed to have a negative association with the Ki-67 staining index. Conclusion: Based on these findings, abnormal PDCD5 expression might be an important biomarker in human osteoclastoma and may contribute to tumor progression and malignant cell proliferation.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Tumor de Células Gigantes do Osso/genética , Proteínas de Neoplasias/genética , Adulto , Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , China , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Tumor de Células Gigantes do Osso/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/metabolismo , Osteoblastoma/genética , Transcriptoma/genética
14.
Int Immunopharmacol ; 72: 358-366, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31030091

RESUMO

Epileptogenesis is a dynamic process initiated by insults to brain and commonly accompanied by cognitive impairment. Isoliquiritigenin (ISL), a flavonoid in licorice, has a broad spectrum of biological effects including anti-inflammatory and antioxidant activities. However, the protective effects of ISL against cognitive impairment in epileptic processes and the underlying molecular mechanism are not well understood. To address these questions, we established an reproducible seizure model by intracerebroventricular injection of kainic acid (KA) in 21-day-old rats; ISL was intraperitoneally administered three times prior to KA injection, and changes in cognitive function; synaptic plasticity; neuronal injury; number of glial cells; and expression of pro-inflammatory cytokines and nuclear factor-like (NRF)2 signaling and NACHT, LRR, and PYD domains-containing protein (NLRP)3 inflammasome components in the hippocampus were examined. Rats with KA-induced seizures showed longer average escape latency and decreases in the number of platform crossings and average time spent in the target quadrant in the Morris water maze; ISL pretreatment reversed this decline in cognitive impairment and increased the protein levels of synaptophysin, postsynaptic density-95 and brain-derived neurotrophic factor while reducing the number of Fluoro Jade B-positive cells, microglia, and astrocytes; cleaved-Caspase-3 and -9 protein levels; and tumor necrosis factor-α, interleukin (IL)-1ß, and IL-18 production. It also enhanced the nuclear localization of NRF2, hemeoxygenase-1, and NAD(P)H:quinone oxidoreductase (NQO) 1, and reversed the upregulation of NLRP3 inflammasome components NLRP3 and Caspase-1 induced by KA injection. Thus, ISL protects against cognitive impairment in KA-induced epileptic processes possibly through regulation of NRF2 signaling and the NLRP3 inflammasome pathway.


Assuntos
Anti-Inflamatórios/uso terapêutico , Chalconas/uso terapêutico , Disfunção Cognitiva/tratamento farmacológico , Encefalite/tratamento farmacológico , Fármacos Neuroprotetores/uso terapêutico , Convulsões/tratamento farmacológico , Animais , Anti-Inflamatórios/farmacologia , Chalconas/farmacologia , Disfunção Cognitiva/metabolismo , Encefalite/metabolismo , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Inflamassomos/metabolismo , Ácido Caínico , Masculino , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Ratos Wistar , Convulsões/induzido quimicamente , Convulsões/metabolismo , Sinapses/efeitos dos fármacos
15.
Oncol Lett ; 16(3): 3805-3811, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30127992

RESUMO

Programmed cell death 4 (PDCD4) has been recognized as a novel tumor suppressor gene, which inhibits the activation and translation of activator protein (AP)-1. Dysregulated expression of PDCD4 is also involved in various human tumors and is linked to tumor progression and development. However, the function and clinical implication of PDCD4 in giant cell tumors of the bone (GCTBs) has not been previously investigated. In the present study, PDCD4 expression was determined in 83 samples of GCTBs at mRNA and protein levels by quantitative reverse transcription-polymerase chain reaction, western blotting and immunohistochemistry. The results demonstrated that PDCD4 mRNA expression was reduced in 63% of GCTB samples (17/27) and protein expression was decreased in 65% of samples (54/83), compared with adjacent non-tumor tissues. Furthermore, decreased expression of PDCD4 was significantly associated with certain clinicopathological characteristics, including the Campanacci grade and recurrence. A strong negative correlation was determined between PDCD4 expression and the Ki-67 positive rate in GCTBs (r=-0.6392; P<0.001). The results of the present study suggest that PDCD4 may serve a role in the malignant progression of human GCTBs and may be an important prediction factor for prognosis.

16.
J Nutr Biochem ; 51: 91-98, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29107826

RESUMO

Atherosclerosis is an age-associated disease; however, diabetic atherosclerosis has higher severity beyond age range for accumulative premature senescent cells in diabetes. Recent findings suggest that rutin, a flavonoid, has potential benefits for diabetic individuals. This study was designed to evaluate the effects of rutin on premature senescence and atherosclerosis. Apolipoprotein E knockout mice exhibiting insulin resistance after 6 weeks of high-fat diet were administered with a low dose of streptozotocin (STZ) to induce diabetes. After 8 weeks of STZ administration, rutin (40 mg/kg/d) was supplemented by gavage for the last 6 weeks. We evaluated the prosperity of the plaque and diabetes using serial echocardiography, histopathologic and metabolite analysis. Premature senescence induced by hydrogen peroxide in primary vascular smooth muscle cells (VSMCs) was used to analyze the underlying mechanism. Mice with diabetes showed more severe plaque burden on aortic arteries and less smooth muscle cells but larger senescent cell ratio in plaque compared with mice with control diets. Rutin significantly improves glucose and lipid metabolic disturbance in diabetes. Moreover, rutin decreased the atherosclerotic burden and senescent cell number and increased the VSMC ratio in aortic root plaque. In vitro, we demonstrated that rutin ameliorated premature senescence induced by oxidative stress, and the protective function may be mediated by inhibiting oxidative stress and protecting telomere. Rutin administration attenuates atherosclerosis burden and stabilizes plaque by improving metabolic disturbance and alleviating premature senescence of VSMCs. Inhibition of VSMCs premature senescence with rutin may be an effective therapy for diabetic atherosclerosis.


Assuntos
Anti-Inflamatórios não Esteroides/uso terapêutico , Antioxidantes/uso terapêutico , Aterosclerose/dietoterapia , Angiopatias Diabéticas/dietoterapia , Suplementos Nutricionais , Músculo Liso Vascular/metabolismo , Rutina/uso terapêutico , Animais , Aorta , Aterosclerose/complicações , Aterosclerose/metabolismo , Aterosclerose/patologia , Biomarcadores/sangue , Biomarcadores/metabolismo , Células Cultivadas , Senescência Celular , Diabetes Mellitus Experimental/complicações , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/metabolismo , Angiopatias Diabéticas/patologia , Dieta Hiperlipídica/efeitos adversos , Resistência à Insulina , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/patologia , Estresse Oxidativo , Homeostase do Telômero
17.
Zhonghua Yu Fang Yi Xue Za Zhi ; 41(4): 290-4, 2007 Jul.
Artigo em Zh | MEDLINE | ID: mdl-17959051

RESUMO

OBJECTIVE: To investigate the anti-fibrotic effects of Qidan granule in rats. METHODS: The rats were randomly divided into six experimental groups: normal group, model group, Qidan group, Tetrandrine group. All rats except normal group were treated with silicon dioxide (50 mg/rat) by intratracheal instillation to induce silicosis. Qidan group and Tetrandrine group were treated with Qidan granule (3125 mg/kg) or treated with Tetrandrine (22 mg/kg) respectively. All the rats were sacrificed after 5 months. Calculate Lung/body coefficient by weighting the lung wet weight and the body weight of rats. Content of Hydroxyproline was measured by alkaline hydrolysis. The gene expression of transforming growth factor-beta1 was examined by using enzyme-linked immunosorbent assay (ELISA). Paraffin embedded lung sections with HE staining, VG staining and Gomori staining were observed under light microscope. RESULTS: In Qidan group and Tetrandrine group, Lung/body coefficient and content of Hydroxyproline and expression of transforming growth factor-beta1 were lower as compared with model group (P < 0.05). Model group mainly showed III approximately IV grade silicotic nodule, which contained thick collagen and sparse reticulum fibe; Qidan group and Tetrandrine group appeared with II grade silicotic nodule, which contained tiny collagen and intensive reticulum fibe. Tetrandrine group showed injury of kidney, and others were normal. CONCLUSION: Qidan granule extract should prevent and from inhibit the remarkably silicotic fibrosis in rats.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Fitoterapia , Fibrose Pulmonar/prevenção & controle , Silicose/tratamento farmacológico , Animais , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Fibrose Pulmonar/patologia , Ratos , Ratos Wistar , Silicose/metabolismo , Silicose/patologia , Fator de Crescimento Transformador beta/biossíntese
18.
Acta Neurobiol Exp (Wars) ; 76(4): 304-317, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28094821

RESUMO

Growing evidence suggests that macrophage inflammatory protein (MIP)-1alpha (synonym CCL3) is upregulated in the neuroinflammatory processes initiated by some brain disorders, but its precise role and regulatory mechanism remain unclear. The present work aims to evaluate the role of CCL3/MIP-1alpha in lipopolysaccharide (LPS)-induced brain injury, and investigate whether the MAPKs and NF-kappaB regulate CCL3/MIP-1alpha expression. We firstly examined the patterns of CCL3/MIP-1alpha expression and phosphorylation of MAPKs in the brains of rats 6, 24, and 72 h after LPS administration. Additionally, LPS-treated rats were administered an anti-MIP-1alpha neutralizing antibody, and the microglial reaction and the expression of both cyclooxygenase-2 and inducible nitric oxide synthase (iNOS) were analyzed. We finally evaluated the effect of an inhibitor of P38 MAPK, an inhibitor of ERK1/2, or an inhibitor of NF-kappaB, on the levels of CCL3/MIP-1alpha protein and numbers of microglia in the brain. In the observation period, LPS induced CCL3/MIP-1alpha expression, which localized to OX-42-labeled microglia, leading to time-dependent increases in the phosphorylation of P38 MAPK and ERK1/2. The expression pattern of induced CCL3/MIP-1alpha was partly consistent with the phosphorylation of MAPKs (P38 MAPK, ERK1/2). Anti-MIP-1alpha attenuated microglial accumulation and the upregulation of cyclooxygenase-2 and iNOS. The inhibition of P38 MAPK, ERK1/2, or NF-kappaB signaling reduced the induced upregulation of CCL3/MIP-1alpha and the microglial accumulation. Our data suggest that upregulated CCL3/MIP-1alpha mediates the accumulation of microglia and the neuroinflammatory reaction, and its expression may be regulated by MAPKs and NF-kappaB in LPS-induced brain injury.


Assuntos
Lesões Encefálicas/complicações , Quimiocina CCL3/metabolismo , Encefalite/patologia , Microglia/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Regulação para Cima/fisiologia , Animais , Anticorpos/farmacologia , Antioxidantes/farmacologia , Lesões Encefálicas/induzido quimicamente , Lesões Encefálicas/tratamento farmacológico , Antígeno CD11b/metabolismo , Quimiocina CCL3/genética , Quimiocina CCL3/imunologia , Modelos Animais de Doenças , Encefalite/etiologia , Ativadores de Enzimas/farmacologia , Inibidores Enzimáticos/farmacologia , Proteína Glial Fibrilar Ácida/metabolismo , Lipopolissacarídeos/toxicidade , Microglia/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Prolina/análogos & derivados , Prolina/farmacologia , Ratos , Ratos Wistar , Tiocarbamatos/farmacologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
19.
Oncotarget ; 7(13): 16194-204, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26908443

RESUMO

Vasa vasorum (VV) neovascularization contributes to atherogenesis and its expansion and distribution is correlated with intraplaque expression of angiogenic factors. The present study investigated the roles of Tongxinluo (TXL), a traditional Chinese medication, on VV proliferation and atherogenesis. In vitro, TXL pre-treatment reversed the tumor necrosis factor-a (TNF-a) induced expression of vascular endothelial growth factor A (VEGF-A) and angiopoietin-1 (ANGPT-1) but not ANGPT-2, leading to increased ratio of ANGPT-1 to ANGPT-2. Consistently, TXL treatment (at a dosage of 0.38, 0.75, 1.5 g/kg/d, respectively) decreased the expression of VEGF-A while increased that of ANGPT-1 in early atherosclerotic lesions of apolipoprotein E deficient (apoE-/-) mice. On aortic ring assay, microvessels sprouting from aortas were significantly inhibited in TXL-treated mice. Moreover, VV neovascularization in plaques was markedly reduced with TXL treatment. Histological and morphological analysis demonstrated that TXL treatment reduced plaque burden, plaque size and changed the plaque composition. These data suggest that TXL inhibits early atherogenesis through regulating angiogenic factor expression and inhibiting VV proliferation in atherosclerotic plaque. Our study shed new light on the anti-atherosclerotic effect of TXL.


Assuntos
Inibidores da Angiogênese/farmacologia , Aterosclerose/patologia , Medicamentos de Ervas Chinesas/farmacologia , Neovascularização Patológica/patologia , Angiopoietina-1/biossíntese , Animais , Aterosclerose/metabolismo , Masculino , Camundongos , Camundongos Knockout para ApoE , Neovascularização Patológica/metabolismo , Células RAW 264.7 , Vasa Vasorum/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos
20.
Zhonghua Jie He He Hu Xi Za Zhi ; 28(11): 760-2, 2005 Nov.
Artigo em Zh | MEDLINE | ID: mdl-16324271

RESUMO

OBJECTIVE: To observe the protective role of recombinant Chinese interferon-gamma (INF-gamma) in pulmonary injury (PI) induced by bleomycin (BLM) in C57 mice. METHODS: Seventy-five C57 mice were randomly divided into a control group and groups treated with BLM, BLM + INF-gamma minimum dose (0.25 microg/d), BLM + INF-gamma medium dose (0.5 microg/d), and BLM + INF-gamma maximum dose (1.0 microg/d, with 15 mice each). PI was induced by BLM, and intervention with different doses of INF-gamma was carried out in the experiment groups, but no treatment was administered in the control mice. Measurement of pulmonary hydroxyproline (Hyp), image analysis of collagen I and III, and measurement of the ratio between lung alveoli and interstitial areas were performed. RESULTS: Lung Hyp content and collagen I and III deposition were increased as compared with the control after administration of BLM (0.78 +/- 0.08 vs 0.65 +/- 0.06, P < 0.01; 0.048 +/- 0.006 vs 0.004 +/- 0.001, P < 0.01). Hyp content and collagen I and III deposition were reduced in group INF-gamma maximum dose (0.67 +/- 0.08 vs 0.78 +/- 0.08, P < 0.05; 0.008 +/- 0.001 vs 0.048 +/- 0.006, P < 0.01). The ratio of lung collagen to lung tissue areas was increased with the increase of INF-gamma doses (1.78, 0.12, 0.67, 0.73, 1.65 respectively). CONCLUSION: INF-gamma is effective in alleviating BLM induced pulmonary injury in mice, possibly by inhibition of transformation of fibroblasts to myofibroblasts and collagen synthesis.


Assuntos
Interferon gama/uso terapêutico , Lesão Pulmonar/prevenção & controle , Animais , Bleomicina/efeitos adversos , Colágeno Tipo I/metabolismo , Colágeno Tipo III/metabolismo , Modelos Animais de Doenças , Humanos , Hidroxiprolina/metabolismo , Pulmão/química , Lesão Pulmonar/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA