Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 308
Filtrar
1.
Respir Res ; 23(1): 6, 2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35016680

RESUMO

BACKGROUND: Hypoxic pulmonary hypertension (HPH) is a chronic progressive advanced disorder pathologically characterized by pulmonary vascular remodeling. Notch4 as a cell surface receptor is critical for vascular development. However, little is known about the role and mechanism of Notch4 in the development of hypoxic vascular remodeling. METHODS: Lung tissue samples were collected to detect the expression of Notch4 from patients with HPH and matched controls. Human pulmonary artery smooth muscle cells (HPASMCs) were cultured in hypoxic and normoxic conditions. Real-time quantitative PCR and western blotting were used to examine the mRNA and protein levels of Notch4. HPASMCs were transfected with small interference RNA (siRNA) against Notch4 or Notch4 overexpression plasmid, respectively. Cell viability, cell proliferation, apoptosis, and migration were assessed using Cell Counting Kit-8, Edu, Annexin-V/PI, and Transwell assay. The interaction between Notch4 and ERK, JNK, P38 MAPK were analyzed by co-immunoprecipitation. Adeno-associated virus 1-mediated siRNA against Notch4 (AAV1-si-Notch4) was injected into the airways of hypoxic rats. Right ventricular systolic pressure (RVSP), right ventricular hypertrophy and pulmonary vascular remodeling were evaluated. RESULTS: In this study, we demonstrate that Notch4 is highly expressed in the media of pulmonary vascular and is upregulated in lung tissues from patients with HPH and HPH rats compared with control groups. In vitro, hypoxia induces the high expression of Delta-4 and Notch4 in HPASMCs. The increased expression of Notch4 promotes HPASMCs proliferation and migration and inhibits cells apoptosis via ERK, JNK, P38 signaling pathways. Furthermore, co-immunoprecipitation result elucidates the interaction between Notch4 and ERK/JNK/P38. In vivo, silencing Notch4 partly abolished the increase in RVSP and pulmonary vascular remodeling caused by hypoxia in HPH rats. CONCLUSIONS: These findings reveal an important role of the Notch4-ERK/JNK/P38 MAPK axis in hypoxic pulmonary remodeling and provide a potential therapeutic target for patients with HPH.


Assuntos
Regulação da Expressão Gênica , Hipertensão Pulmonar/genética , Hipóxia/complicações , Miócitos de Músculo Liso/metabolismo , Receptor Notch4/genética , Remodelação Vascular/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/patologia , Hipóxia/genética , Hipóxia/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Ratos , Ratos Sprague-Dawley , Receptor Notch4/biossíntese , Transdução de Sinais , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
2.
Diabet Med ; 39(2): e14713, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34614244

RESUMO

AIMS: Diabetic population have a twofold to threefold increased risk of developing liver cancer, and hyperglycaemia is a prime causative factor that propends the tumour cells to undergo aggressive metabolic growth. In this study, we aimed to examine the molecular mechanism by which lutein inhibits hyperglycaemia-induced human hepatocarcinoma (HepG2) cell proliferation. METHODS: The effect of lutein on high glucose-induced proliferation was measured using the WST-1 reagent. Its effect on intracellular reactive oxygen species (ROS) levels was measured by DCF assay. The effect on the expression of antioxidant enzymes, cell cycle regulatory proteins and intracellular protein kinases was analysed by western blotting. The modulatory effect of lutein on different phases of the cell cycle was analysed by flow cytometry. RESULTS: The data showed that lutein at 5 µM concentration significantly blocked glucose-promoted HepG2 cell proliferation. Suppression of high glucose-induced cell proliferation by lutein was not associated with apoptosis induction, but it was linked with inhibition of hyperglycaemia-mediated elevated ROS and upregulated expression of high glucose-mediated repressed heme oxygenase 1 (HO1). Furthermore, G2/M phase cell cycle arrest and associated phosphorylation of Cdk1 and P53 were found to be linked with suppressed hyperglycaemia-mediated cell proliferation by lutein. In addition, lutein inhibited hyperglycaemia-induced activation of P38 which relates to high glucose-induced ROS-mediated growth suppression and modulated the phosphorylation of Erk, JNK and Akt in hyperglycaemic HepG2 cells. CONCLUSION: Our findings portray that sufficient intake of lutein may offer a negative impact on diabetes-associated tumour growth.


Assuntos
Apoptose , Carcinoma Hepatocelular/genética , Hiperglicemia/complicações , Luteína/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Xantofilas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Carcinoma Hepatocelular/etiologia , Carcinoma Hepatocelular/patologia , Proliferação de Células , Células Hep G2 , Humanos , Hiperglicemia/genética , Hiperglicemia/metabolismo , Neoplasias Hepáticas , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
3.
Sci Rep ; 11(1): 16555, 2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34400718

RESUMO

Oxylipins modulate the behavior of immune cells in inflammation. Soluble epoxide hydrolase (sEH) converts anti-inflammatory epoxyeicosatrienoic acid (EET) to dihydroxyeicosatrienoic acid (DHET). An sEH-inhibitor, TPPU, has been demonstrated to ameliorate lipopolysaccharide (LPS)- and sepsis-induced inflammation via EETs. The immunomodulatory role of DHET is not well characterized. We hypothesized that TPPU dampens inflammation and that sEH-derived DHET alters neutrophil functionality in burn induced inflammation. Outbred mice were treated with vehicle, TPPU or 14,15-DHET and immediately subjected to either sham or dorsal scald 28% total body surface area burn injury. After 6 and 24 h, interleukin 6 (IL-6) serum levels and neutrophil activation were analyzed. For in vitro analyses, bone marrow derived neutrophil functionality and mRNA expression were examined. In vivo, 14,15-DHET and IL-6 serum concentrations were decreased after burn injury with TPPU administration. In vitro, 14,15-DHET impaired neutrophil chemotaxis, acidification, CXCR1/CXCR2 expression and reactive oxygen species (ROS) production, the latter independent from p38MAPK and PI3K signaling. We conclude that TPPU administration decreases DHET post-burn. Furthermore, DHET downregulates key neutrophil immune functions and mRNA expression. Altogether, these data reveal that TPPU not only increases anti-inflammatory and inflammation resolving EET levels, but also prevents potential impairment of neutrophils by DHET in trauma.


Assuntos
Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Anti-Inflamatórios/uso terapêutico , Queimaduras/tratamento farmacológico , Neutrófilos/imunologia , Compostos de Fenilureia/uso terapêutico , Piperidinas/uso terapêutico , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Queimaduras/imunologia , Queimaduras/metabolismo , Queimaduras/patologia , Citocinas/sangue , Epóxido Hidrolases/antagonistas & inibidores , Feminino , Lipopolissacarídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NADPH Oxidases/metabolismo , Neutrófilos/classificação , Neutrófilos/metabolismo , Fagocitose/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Fosfatidilinositol 3-Quinases/biossíntese , Fosfatidilinositol 3-Quinases/genética , Piperidinas/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Quimiocinas/fisiologia , Explosão Respiratória/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
4.
J Neuroinflammation ; 18(1): 150, 2021 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-34225736

RESUMO

BACKGROUND: Our recent studies have identified that the red nucleus (RN) dual-directionally modulates the development and maintenance of mononeuropathic pain through secreting proinflammatory and anti-inflammatory cytokines. Here, we further explored the action of red nucleus IL-33 in the early development of mononeuropathic pain. METHODS: In this study, male rats with spared nerve injury (SNI) were used as mononeuropathic pain model. Immunohistochemistry, Western blotting, and behavioral testing were used to assess the expressions, cellular distributions, and actions of red nucleus IL-33 and its related downstream signaling molecules. RESULTS: IL-33 and its receptor ST2 were constitutively expressed in the RN in naive rats. After SNI, both IL-33 and ST2 were upregulated significantly at 3 days and peaked at 1 week post-injury, especially in RN neurons, oligodendrocytes, and microglia. Blockade of red nucleus IL-33 with anti-IL-33 neutralizing antibody attenuated SNI-induced mononeuropathic pain, while intrarubral administration of exogenous IL-33 evoked mechanical hypersensitivity in naive rats. Red nucleus IL-33 generated an algesic effect in the early development of SNI-induced mononeuropathic pain through activating NF-κB, ERK, p38 MAPK, and JAK2/STAT3, suppression of NF-κB, ERK, p38 MAPK, and JAK2/STAT3 with corresponding inhibitors markedly attenuated SNI-induced mononeuropathic pain or IL-33-evoked mechanical hypersensitivity in naive rats. Red nucleus IL-33 contributed to SNI-induced mononeuropathic pain by stimulating TNF-α expression, which could be abolished by administration of inhibitors against ERK, p38 MAPK, and JAK2/STAT3, but not NF-κB. CONCLUSIONS: These results suggest that red nucleus IL-33 facilitates the early development of mononeuropathic pain through activating NF-κB, ERK, p38 MAPK, and JAK2/STAT3. IL-33 mediates algesic effect partly by inducing TNF-α through activating ERK, p38 MAPK and JAK2/STAT3.


Assuntos
Interleucina-33/biossíntese , Janus Quinase 2/biossíntese , Mononeuropatias/metabolismo , Neuralgia/metabolismo , Núcleo Rubro/metabolismo , Fator de Transcrição STAT3/biossíntese , Animais , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Mononeuropatias/patologia , Neuralgia/patologia , Ratos , Ratos Sprague-Dawley , Núcleo Rubro/patologia , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
5.
Pak J Pharm Sci ; 34(1): 129-134, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34248012

RESUMO

The aim of this study was to examine the effects of glycosaminoglycan (GAG) from Urechis unicinctus on the P2Y1 receptor pathway and expression of related factors in rat platelets. The concentration of calcium ion (Ca2+) in rat platelets was determined by double wavelength Fura-2 fluorescence spectrophotometry, and the concentrations of inositol trisphosphate (IP3) and glycoprotein IIb/IIIa (GPIIb/IIIa) in rat platelets were measured using the enzymatic immunoassay method. The phosphorylation levels of phospholipase C (PLC), phospholipase A2 (PLA2), protein kinase C (PKC), and p38 mitogen-activated protein kinase (p38MAPK) were also detected by Western blot. It was found that the GAG from U. unicinctus significantly reduced the Ca2+ and IP3 levels in rat platelets (p<0.05, p<0.01). Moreover, medium and high concentrations of GAG significantly reduced the concentration of the platelet membrane GPIIb/IIIa in rats (p<0.05, p<0.01). The phosphorylation levels of PLC, PLA)2), PKC and p38MAPK in rat platelets were also inhibited by GAG and P)2)Y)1) receptor blocker MRS2179 (p<0.05, p<0.01). However, the degree of inhibition of GAG was lower than that of MRS2179. The results laid a foundation for further utilization of the glycosaminoglycan.


Assuntos
Produtos Biológicos/farmacologia , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Glicosaminoglicanos/farmacologia , Receptores Purinérgicos P2Y1/biossíntese , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Animais , Produtos Biológicos/isolamento & purificação , Relação Dose-Resposta a Droga , Glicosaminoglicanos/isolamento & purificação , Nematoides , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
6.
Mol Neurodegener ; 16(1): 26, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863362

RESUMO

BACKGROUND: Apolipoprotein E4 (APOE4) is associated with a greater response to neuroinflammation and the risk of developing late-onset Alzheimer's disease (AD), but the mechanisms for this association are not clear. The activation of calcium-dependent cytosolic phospholipase A2 (cPLA2) is involved in inflammatory signaling and is elevated within the plaques of AD brains. The relation between APOE4 genotype and cPLA2 activity is not known. METHODS: Mouse primary astrocytes, mouse and human brain samples differing by APOE genotypes were collected for measuring cPLA2 expression, phosphorylation, and activity in relation to measures of inflammation and oxidative stress. RESULTS: Greater cPLA2 phosphorylation, cPLA2 activity and leukotriene B4 (LTB4) levels were identified in ApoE4 compared to ApoE3 in primary astrocytes, brains of ApoE-targeted replacement (ApoE-TR) mice, and in human brain homogenates from the inferior frontal cortex of patients with AD carrying APOE3/E4 compared to APOE3/E3. Greater cPLA2 phosphorylation was also observed in human postmortem frontal cortical synaptosomes and primary astrocytes after treatment with recombinant ApoE4 ex vivo. In ApoE4 astrocytes, the greater levels of LTB4, reactive oxygen species (ROS), and inducible nitric oxide synthase (iNOS) were reduced after cPLA2 inhibition. CONCLUSIONS: Our findings implicate greater activation of cPLA2 signaling system with APOE4, which could represent a potential drug target for mitigating the increased neuroinflammation with APOE4 and AD.


Assuntos
Apolipoproteína E4/metabolismo , Cálcio/farmacologia , Córtex Cerebral/enzimologia , Sistema de Sinalização das MAP Quinases , Fosfolipases A2 Citosólicas/metabolismo , Peptídeos beta-Amiloides/farmacologia , Animais , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E3/farmacologia , Apolipoproteína E4/genética , Apolipoproteína E4/farmacologia , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Córtex Cerebral/patologia , Ativação Enzimática/efeitos dos fármacos , Heterozigoto , Humanos , Inflamassomos , Inflamação , Leucotrieno B4/biossíntese , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Estresse Oxidativo , Fragmentos de Peptídeos/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Espécies Reativas de Oxigênio , Sinaptossomos/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
7.
Mol Cell Biochem ; 476(7): 2729-2738, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33683556

RESUMO

Colon cancer is one of the most familiar malignancies worldwide, with high morbidity and high mortality. This study intended to explore the role and mechanism of tectoridin (TEC) in regulating the progression of colon cancer. First, colon cancer cell lines (HCT116 and SW480 cells) were treated with different doses of TEC (0-200 µM). Then, CCK8 and clone formation experiments were performed to detect cell proliferation. Flow cytometry and western blot were conducted to examine apoptosis. Subsequently, Transwell assay and wound-healing test was employed to determine the effect of TEC on colon cancer cell invasion and migration. Next, western blot was performed to monitor the PKC/p38 MAPK pathway activation. In addition, a tumor model was established in nude mice to explore the effect of TEC on tumor growth in vivo. TEC dose-dependently dampened the proliferation, migration and invasion of colon cancer cells and facilitated their apoptosis. In addition, TEC abated the tumor cell growth in vivo. Besides, TEC dose-dependently suppressed the expression of PKC and p38 MAPK. Moreover, inhibiting the PKC pathway almost cancel out the anti-tumor effects induced by TEC. TEC attenuates the colon cancer progression by inhibiting the PKC/p38 MAPK pathway.


Assuntos
Neoplasias do Colo/enzimologia , Regulação para Baixo/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Isoflavonas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Proteína Quinase C/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/patologia , Células HCT116 , Humanos
8.
Minerva Gastroenterol Dietol ; 66(3): 208-210, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32218423

RESUMO

BACKGROUND: The P38-protein is known to be expressed in colorectal adenomas (CRA). Expression in low- and high-grade tubular adenomas is decreased when compared to adenocarcinomas and increased with regard to normal mucosa. We aimed to study P38 expression in human CRAs and the relationships to cell proliferation Ki67-protein, stem-phenotype CD133-protein and, to mTOR-protein (AKT pathway). METHODS: The immunohistochemical expression of P38 was evaluated in CRAs on tissue microarrays. Data were analyzed with the Kendall-rank-correlation test. RESULTS: Nuclear P38 correlated to low-grade dysplasia (Kendall P<0.01/tau=-0.254) and to decreased adenoma size (P<0.01/tau=-0.267). Nuclear P38 also correlated to cytoplasmic or membrane mTOR (P<0.01/tau=-0.223 and P<0.01/tau=-0.340) and to cytoplasmic CD133 (P<0.01/0.293). An inverse relationship was observed to Ki67 (P<0.00/ tau=-0.110). CONCLUSIONS: Our results suggest an interference of P38 with initial steps of colorectal adenomagenesis. The correlation to mTOR suggests a biological crosstalk between the MAPK- and AKT-signaling-pathways in colorectal adenomagenesis at P38 level.


Assuntos
Antígeno AC133 , Adenoma , Proliferação de Células , Neoplasias Colorretais , Antígeno Ki-67 , Serina-Treonina Quinases TOR , Proteínas Quinases p38 Ativadas por Mitógeno , Antígeno AC133/fisiologia , Adenoma/química , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Neoplasias Colorretais/química , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Feminino , Humanos , Antígeno Ki-67/fisiologia , Masculino , Fenótipo , Transdução de Sinais , Serina-Treonina Quinases TOR/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/análise , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
9.
Spine (Phila Pa 1976) ; 45(7): E355-E363, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-31725126

RESUMO

STUDY DESIGN: To evaluate the effect of p38 pathway on spinal cord injury (SCI), a rat model of SCI was performed. OBJECTIVE: We determined the effect of p38 on SCI and SCI related inflammation, apoptosis, and autophagy. SUMMARY OF BACKGROUND DATA: SCI is a severe clinical problem worldwide. It is difficult to prevent cell necroptosis and promote the survival of residual neurons after SCI. p38, a class of mitogen-activated protein kinases, its effect on SCI and SCI related inflammation, apoptosis, and autophagy have not been studied very well. METHODS: The rats were randomly divided into the following four groups: the sham-operated (sham) group, the SCI group, the SCI + vehicle group, and the SCI + SB203580 (10 mg/kg) group. The p38 inhibitor SB203580 was administered by oral (10 mg/kg/d) gavage once per day for 14 days. Neurological recovery was assessed using the Basso, Beattie, and Bresnahan locomotion rating scale. Apoptosis, autophagy, and inflammation related proteins were measured by enzyme-linked immunosorbent assay kits or western blotting. RESULTS: Our results showed that p38 was upregulated after SCI from day 3, which was paralleled with the levels of its proteins ATF-2, suggesting an increase in p38 activity. Our results showed administration of SB203580 attenuated histopathology and promoted locomotion recovery in rats after SCI. SB203580 administration significantly inhibited inflammatory cytokines levels as well as the inflammation signaling pathway. SB203580 administration also modulated the apoptosis and autophagy signaling pathway. CONCLUSION: Our findings suggest that p38 inhibitor SB203580 treatment alleviates secondary SCI by inhibiting inflammation and apoptosis, thereby promoting neurological and locomoter functional recovery, thus suggest the important role of p38 in neuronal protection after SCI. LEVEL OF EVIDENCE: N/A.


Assuntos
Apoptose/fisiologia , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Traumatismos da Medula Espinal/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Mediadores da Inflamação/antagonistas & inibidores , Locomoção/efeitos dos fármacos , Locomoção/fisiologia , Masculino , Piridinas/farmacologia , Piridinas/uso terapêutico , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
10.
J Invest Dermatol ; 140(4): 878-890.e5, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31622599

RESUMO

Melanoma cells can switch between distinct gene expression profiles, resulting in proliferative or invasive phenotypes. Signaling pathways involved in this switch were analyzed by gene expression profiling of a cohort of 22 patient-derived melanoma cell lines. CDH1 negativity was identified as a surrogate marker for the invasive phenotype. CDH1 expression could be turned on and off by modulating activity of p38 or its downstream target MK2, suggesting that this pathway controls melanoma progression. Mechanistically, MK2 inhibition prevented melanoma-induced vascular barrier disruption, reduced the expression of PODXL and DEL-1, and prevented vascular dissemination in vivo. PODXL and DEL-1 expression in patients with melanoma were associated with poor survival and thus can be used as prognostic markers. Downstream targets of MK2 may thus serve as candidate therapeutics.


Assuntos
Regulação Neoplásica da Expressão Gênica , Melanoma/genética , Neoplasias Cutâneas/genética , Neoplasias Vasculares/prevenção & controle , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Humanos , Melanoma/metabolismo , Melanoma/patologia , Invasividade Neoplásica , Prognóstico , Transdução de Sinais , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas , Neoplasias Vasculares/metabolismo , Neoplasias Vasculares/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
11.
Prostate ; 79(16): 1823-1831, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31509289

RESUMO

BACKGROUND: The amplification of gene COPS3 is closely related to the development of osteosarcoma and hepatocellular carcinoma. However, the effects of COPS3 on prostate cancer (PCa) are poorly understood. METHODS: In this study, the protein expression of COPS3 in PCa tissues, adjacent normal tissues, and bone metastasis tissues of PCa was analyzed by immunohistochemistry. Furthermore, cell proliferation, colony formation, migration, and invasion assay were performed in 22rv1 and PC-3 cells after knocking down COPS3 by small interfering RNAs. Furthermore, we performed western blot analysis to explore the potential mechanisms underlying it. RESULTS: This study found that the overall survival of the COPS3 high-expression group was significantly shorter than the low-expression group. This study discovered that the protein expression of COPS3 in PCa tissues was higher than that in the matched nontumor prostate tissues. In addition, tissues from bone metastasis of PCa had a high percentage of overexpressing COPS3. After knockdown of the COPS3 gene in 22rv1 and PC3 cells, two classic human PCa cell lines which had a high level of COPS3, the abilities of migration, invasion, and proliferation were inhibited. Finally, protein levels of phosphorylated P38 mitogen-activated protein kinase (MAPK) and N-cadherin were significantly decreased after knocking down the expression of COPS3, and the protein levels of E-cadherin were significantly increased. CONCLUSIONS: In conclusion, COPS3 may be closely related to the progress of PCa. Knockdown of COPS3 inhibited the progress of PCa through reducing the levels of phosphorylated P38 MAPK and impaired the epithelial-mesenchymal transition process.


Assuntos
Complexo do Signalossomo COP9/deficiência , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas/deficiência , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Complexo do Signalossomo COP9/biossíntese , Complexo do Signalossomo COP9/genética , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Transição Epitelial-Mesenquimal , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Masculino , Fosforilação , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/administração & dosagem , Taxa de Sobrevida , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
12.
Eur Rev Med Pharmacol Sci ; 23(10): 4491-4497, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31173326

RESUMO

OBJECTIVE: To explore the effect of long non-coding ribonucleic acid (lncRNA) H19 on the apoptosis of vascular endothelial cells in arteriosclerosis obliterans (ASO) via the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway. PATIENTS AND METHODS: Human umbilical vein endothelial cells (HUVECs) were cultured, and lncRNA H19 was inhibited by Si-H9 and overexpressed by H19-OE. Then, the apoptosis rate was detected by flow cytometry, the target of lncRNA H19 was detected by dual luciferase reporter gene assay, and changes in the protein level were determined via Western blotting (WB). RESULTS: LncRNA H19 exhibited high expression in serum of patients with ASO, and compared with that in congeneric normal mice, the expression of lncRNA H19 in ASO mice rose. Besides, the proliferation ability of cells transfected with H19-OE was markedly strengthened, and H19-OE treatment could down-regulate the expression level of the apoptin, active cysteinyl aspartate-specific proteinase-3 (Caspase-3). In addition, lncRNA H19 bound to micro ribonucleic acid (miR)-19a in a targeted way. After lncRNA H19 was overexpressed, the expression of the NF-κB pathway key factors, p38 and p65, were notably increased, and the nuclear translocation of p65 was significantly enhanced after transfection with miR-19a. CONCLUSIONS: LncRNA H19 promotes the proliferation of vascular endothelial cells in ASO and inhibits the apoptosis of them via the NF-κB pathway.


Assuntos
Apoptose/genética , Arteriosclerose Obliterante/genética , Arteriosclerose Obliterante/patologia , Células Endoteliais/patologia , Endotélio Vascular/patologia , RNA Longo não Codificante/genética , Animais , Arteriosclerose Obliterante/metabolismo , Caspase 3/biossíntese , Caspase 3/genética , Proliferação de Células , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Camundongos , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/biossíntese , Fator de Transcrição RelA/biossíntese , Fator de Transcrição RelA/genética , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/genética
13.
Stem Cell Reports ; 12(6): 1298-1312, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31130358

RESUMO

Physical exercise stimulates adult hippocampal neurogenesis and is considered a relevant strategy for preventing age-related cognitive decline in humans. The underlying mechanisms remains controversial. Here, we show that exercise increases proliferation of neural precursor cells (NPCs) of the mouse dentate gyrus (DG) via downregulation of microRNA 135a-5p (miR-135a). MiR-135a inhibition stimulates NPC proliferation leading to increased neurogenesis, but not astrogliogenesis, in DG of resting mice, and intriguingly it re-activates NPC proliferation in aged mice. We identify 17 proteins (11 putative targets) modulated by miR-135 in NPCs. Of note, inositol 1,4,5-trisphosphate (IP3) receptor 1 and inositol polyphosphate-4-phosphatase type I are among the modulated proteins, suggesting that IP3 signaling may act downstream miR-135. miR-135 is the first noncoding RNA essential modulator of the brain's response to physical exercise. Prospectively, the miR-135-IP3 axis might represent a novel target of therapeutic intervention to prevent pathological brain aging.


Assuntos
Células-Tronco Adultas/metabolismo , Envelhecimento/metabolismo , MicroRNAs/biossíntese , Células-Tronco Neurais/metabolismo , Neurogênese , Condicionamento Físico Animal , Animais , Proliferação de Células , Regulação da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , Ventrículos Laterais/citologia , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Knockout , Nicho de Células-Tronco , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
14.
Neurosci Lett ; 699: 115-121, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30735722

RESUMO

p38 mitogen-activated protein (MAP) kinase (p38) is a member of the MAP kinase family. Previous reports using p38 chemical inhibitors have suggested that its activation contributes to hippocampal neuronal cell death rather than cell survival. In this study, we used both a cell-permeable p38 protein containing the HIV protein transduction domain (PTD) and cultured adult hippocampal neurons, which were differentiated from cultured adult hippocampal neural stem/progenitor cells (NPCs), to evaluate the direct function of p38 on adult hippocampal neurons. Our immunocytochemical experiments demonstrated that wild-type cell-permeable p38 protein prevents cell death of adult hippocampal neurons induced by a low glucose condition. Our findings indicate that cell-permeable p38 protein may be useful in preventing the degeneration of higher brain function occurring through hippocampal neuronal cell death, and furthermore, that the maintenance of intracellular p38 levels could be another therapeutic target for neurodegenerative diseases such as Alzheimer's disease (AD).


Assuntos
Morte Celular , Hipocampo/citologia , Hipoglicemia/patologia , Neurônios/citologia , Neuroproteção/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Células Cultivadas , Hipocampo/metabolismo , Hipocampo/patologia , Imidazóis/farmacologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
15.
J Cell Physiol ; 234(7): 11360-11368, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30565699

RESUMO

Bladder urothelial carcinoma (BUC) is one of the most common urological malignancies. Our previous study found that adenosine A2b receptor (A2bR) was upregulated in BUC tissues and cells. In the present study, we investigated the effect of MRS1754 (a selective A2bR antagonist) on cell proliferation and migration in two well-studied invasive urothelial cell carcinoma lines EJ and T24. Our results showed that MRS1754 reduced BUC cell proliferation and induced a G0/G1 phase cell-cycle arrest. Next, MRS1754 inhibited cell migration and Bay60-6583 (a selective A2bR agonist) treatment could reverse the inhibitory effect of MRS1754 on BUC cells migration. Furthermore, our results showed MRS1754 treatment downregulated the protein levels of p-P38, p-JNK, and phospho-extracellular signal-regulated kinase (p-ERK). These findings suggest that MRS1754 can inhibit progression of BUC via mitogen-activated protein kinase (MAPK) pathway and indicate the therapeutic potential of A2B antagonists in BUC.


Assuntos
Acetamidas/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Purinas/farmacologia , Receptor A2B de Adenosina/efeitos dos fármacos , Neoplasias da Bexiga Urinária/tratamento farmacológico , Urotélio/patologia , Agonistas do Receptor A2 de Adenosina/farmacologia , Aminopiridinas/farmacologia , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
16.
Photodiagnosis Photodyn Ther ; 24: 198-205, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30268863

RESUMO

BACKGROUND: Esophageal cancer is a common gastrointestinal cancer. About 300,000 people die from esophageal cancer every year in the world. Photodynamic therapy (PDT) has attracted attention as a feasible cancer therap for this diagnosis. Sinoporphyrin sodium (DVDMs) is a novel sensitizer isolated from photofrin. In this study, we aimed to investigate the effects of DVDMs mediated photodynamic therapy and the possible mechanism on human esophageal cancer Eca-109 cells. METHODS: Cell viability was measured by MTT assay and cell apoptosis was determined by Annexin V-PE/7-AAD and western blot. MDC staining and western blot were used to evaluate cell autophagy. The production of intracellular reactive oxygen species (ROS) was detected by flow cytometry. The expression of MAPK and HO-1 were detected by western blot. RESULTS: DVDMs-PDT decreased cell viability and induced cell apoptosis and autophagy. Autophagy inhibition reduced cell apoptosis triggered by DVDMs-PDT in Eca-109 cells. Generation of ROS was detected in DVDMs-PDT group. p38MAPK, JNK and HO-1 were activated after PDT treatment and the activation were reversed by adding ROS scavenger NAC. CONCLUSIONS: Our studies demonstrated that DVDMs-PDT induced apoptosis and autophagy in Eca-109 cells. DVDMs-PDT induced ROS generation in Eca-109 cells, and the generation of ROS activated p38MAPK and JNK. Activation of p38MAPK and JNK may be involved in PDT-induced apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Esofágicas/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/farmacologia , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Heme Oxigenase-1/biossíntese , Humanos , Lasers Semicondutores , Espécies Reativas de Oxigênio , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese
17.
BMC Cancer ; 18(1): 1027, 2018 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-30352570

RESUMO

BACKGROUND: P38 mitogen activated protein kinase is an intermediary signal transduction factor with context-specific roles in breast cancer. Recent mechanistic studies add to the growing consensus that P38 is a tumour suppressor, and it may represent a novel target for breast cancer treatment. The aim of this study is to add definitive data on the prognostic value of P38 and its link with biomarkers in primary breast cancer. METHODS: A large, well-characterised series of 1332 primary breast cancer patients with long-term clinical follow-up was assessed for P38 expression by immunohistochemistry. Association of clinicopathological factors and a panel of breast cancer biomarkers was determined by chi-squared test, and multivariate survival analysis was performed using Cox Proportional Hazards regression modelling. RESULTS: This study shows that nuclear P38 is co-expressed with nuclear hormone receptors (p < 0.001) and is an independent prognostic marker of good long-term clinical outcome in primary breast cancer (hazard ratio 0.796, 95% confidence interval 0.662-0.957, p = 0.015). Significant association was found between expression of P38 and markers of DNA repair including nuclear BRCA1 and RAD51, and cleaved PARP1 (all p < 0.001). CONCLUSIONS: The findings support the proposed role for P38 as a tumour suppressor in breast cancer via upregulation of DNA repair proteins and provide novel hypothesis-generating information on the potential role of P38 in adjuvant therapy decision making.


Assuntos
Biomarcadores Tumorais/biossíntese , Neoplasias da Mama/metabolismo , Reparo do DNA , Receptores Citoplasmáticos e Nucleares/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Adulto , Idoso , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Feminino , Humanos , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Proteínas Quinases p38 Ativadas por Mitógeno/genética
18.
Toxicol Appl Pharmacol ; 358: 76-85, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30222980

RESUMO

Montelukast (MK),a cysteinyl leukotriene (CysLT1) receptor antagonist, latterly exhibited a remarkable neuroprotective activity in various neurodegenerative disorders. This study aims to elucidate the neuroprotective effect of MK in rotenone-induced Parkinson's disease(PD) model in rats. Ninety six male rats were split into four groups: vehicle control (0.2 ml/kg/48 h, sc), MK (10 mg/kg/day, ip), rotenone (1.5 mg/kg/48 h, sc.) and rotenone pretreated with MK. Rotenone treatment led to significant reduction in motor functioning and elevation in oxidative stress markers. Additionally, upregulation of p38 mitogen-activated protein kinase (p38 MAPK) and CysLT1 receptor expressions were anchored with enhanced striatal microglial activation generating a severe neuro-inflammatory milieu. Furthermore, an augmentation in p53 expression and cleaved caspases-3 activity increased apoptotic neurodegeneration synchronized with reduction of striatal tyrosine hydroxylase (TH) content. Changes in neuronal morphology was also noted. MK administration significantly mitigated motor impairment and rise in oxidative stress mediators. As well, the anti-inflammatory activity of MK was manifested by hindering the principal controller of inflammatory pathway, nuclear factor-kappa B, followed by its downstream pro-inflammatory cytokines (tumor necrosis factor-alpha and interleukin-1 beta), by attenuating striatal microglial activation and hampering the expression of both p38 MAPK and CysLT1. Moreover, MK revealed a decline in p53 expression with its downstream cleaved caspases-3 which resulted in preservation of striatal TH terminals as verified by increased striatal TH content and improvement in the histopathological changes incited by rotenone. In conclusion, MK endowed neuroprotective effects in rotenone-induced PD animal model via attenuation of microglial cell activation and p38 MAPK expression.


Assuntos
Acetatos/farmacologia , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Microglia/efeitos dos fármacos , Quinolinas/farmacologia , Rotenona/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Ciclopropanos , Regulação Enzimológica da Expressão Gênica , Inseticidas/toxicidade , Antagonistas de Leucotrienos/farmacologia , Masculino , Microglia/metabolismo , Ratos , Ratos Wistar , Sulfetos , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
Neurobiol Dis ; 119: 53-64, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30048803

RESUMO

Leucine-rich repeat kinase 2 (LRRK2) is the genetic cause of both familial and idiopathic Parkinson's disease (PD), and it is associated with neuronal death, vesicle trafficking, mitochondrial dysfunction, and inflammation. However, its role in secondary brain injury (SBI) induced by intracerebral hemorrhage (ICH) has not been evaluated. In this study, an ICH model was induced by injecting autologous whole blood into the right basal ganglia of adult rats. Meanwhile, primary rat cortical neurons treated with Oxyhemoglobin (OxyHb) were used as an in vitro ICH model. Protein levels of LRRK2 increased significantly in brain tissues after ICH. Upregulation of LRRK2 by genetic overexpression augmented inflammatory responses, behavioral and cognitive dysfunction, brain edema, blood-brain barrier (BBB) injury, and cell death involved in SBI following ICH. Downregulation of LRRK2 by GNE7915 (a specific chemical inhibitor of LRRK2) and genetic knockdown yielded opposite effects. Additionally, inhibiting LRRK2 by GNE7915 obviously reduced OxyHb-induced neuronal apoptosis in vitro and attenuated phosphorylation of p38 MAPK and Drosha both in vivo and in vitro. Therefore, we concluded that LRRK2 participated in ICH-induced SBI by mediating inflammatory responses, behavioral and cognitive dysfunction, brain edema, and BBB injury and by modulating neuronal death and dysfunction and regulating the p38 MAPK/Drosha pathway.


Assuntos
Lesões Encefálicas/metabolismo , Hemorragia Cerebral/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/biossíntese , Ribonuclease III/biossíntese , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas/patologia , Hemorragia Cerebral/patologia , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/antagonistas & inibidores , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/genética , Masculino , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/farmacologia , Ratos , Ribonuclease III/antagonistas & inibidores , Ribonuclease III/genética , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/genética
20.
Pain Physician ; 21(4): E307-E322, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-30045596

RESUMO

BACKGROUND: Interferon regulatory factor 8 (IRF8), which is induced by peripheral nerve injury (PNI), plays a key role in activating spinal microglia to release inflammatory cytokines in a p38-dependent way, thereafter results in formation of central sensitization. Pulsed radiofrequency (PRF) on dorsal root ganglion (DRG) alleviates neuropathic pain and inhibits the microglial activation in chronic constriction injury (CCI) rats. However, the consequences of PRF on spinal IRF8 of CCI rats remains unknown. OBJECTIVE: We explore if PRF on DRG of rats with CCI could restrain IRF8, microglia, and p38 hyperactivity in the spinal cord to alleviate neuropathic pain. STUDY DESIGN: A randomized, controlled animal study. SETTING: Department of Pain Management, Fujian Provincial Hospital, Fujian Key Laboratory of Geriatrics, Provincial Clinic College of Fujian Medical University. METHODS: The changes in pain behaviors and the expressions of IRF8, Iba1 and p-p38 in the spinal cord of CCI rats which were administrated with antisense/ mismatch oligodeoxynucleotide of IRF8 were studied. Rats in CCI+AS ODN group, CCI+MM ODN group or CCI+NS group were intrathecally treated with antisense oligodeoxynucleotide of IRF8, mismatch oligodeoxynucleotide of IRF8 or same volume 0.9% NaCl once daily respectively, beginning from the day after nerve transection 12 hours and lasting for 7 days. The effects of PRF on L4-5 DRG of rats with CCI were investigated. PRF was applied adjacent to the L4-5 DRG at an intensity of 45 V for 6 minutes after CCI, whereas the control rats were treated without radiofrequency current. The withdrawal thresholds were studied and the spinal levels of IRF8, ionized calcium-binding adapter molecule 1 (Iba1, microglia characteristic marker) and p-p38 were calculated by ELISA, western blot, RT-PCR, and immunofluorescence. RESULTS: Intrathecal administration of antisense oligodeoxynucleotide of IRF8 led to the reversal of CCI-induced allodynia, lower activation of spinal microglia and p-p38. Withdrawal thresholds were partially recovered after a single PRF treatment for 14 days. CCI-induced IRF8 upregulation, microglia hyperactivity, and p38 phosphorylation in the spinal cord were reduced due to PRF treatment. However, PRF did not alter pain behaviors and pain signals in normal rats. LIMITATIONS: In our study, one time point was selected just to assess the levels of microglia, and p-p38. The changes of IRF8, microglia, p-p38 in the ipsilateral DRG were not investigated. A more detailed study on how PRF on the DRG could further relieve NP is needed. CONCLUSIONS: Restraining IRF8, microglia and p38 hyperactivity in the spinal cord of CCI rats involved in the contribution to the long-lasting analgesia of PRF. KEY WORDS: Neuropathic pain, pulsed radiofrequency, dorsal root ganglion, microglia, p38MAPK, Interferon regulatory factor 8, chronic constriction injury of sciatic nerve.


Assuntos
Gânglios Espinais/metabolismo , Fatores Reguladores de Interferon/biossíntese , Neuralgia/metabolismo , Tratamento por Radiofrequência Pulsada/métodos , Proteínas Quinases p38 Ativadas por Mitógeno/biossíntese , Animais , Regulação para Baixo , Gânglios Espinais/fisiopatologia , Gânglios Espinais/efeitos da radiação , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatologia , Masculino , Microglia/metabolismo , Microglia/efeitos da radiação , Neuralgia/fisiopatologia , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/fisiopatologia , Ratos , Ratos Sprague-Dawley , Nervo Isquiático/lesões
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA