Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Sci Signal ; 17(840): eadc9142, 2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38861615

RESUMO

Neuroendocrine prostate cancer (PCa) (NEPC), an aggressive subtype that is associated with poor prognosis, may arise after androgen deprivation therapy (ADT). We investigated the molecular mechanisms by which ADT induces neuroendocrine differentiation in advanced PCa. We found that transmembrane protein 1 (MCTP1), which has putative Ca2+ sensing function and multiple Ca2+-binding C2 domains, was abundant in samples from patients with advanced PCa. MCTP1 was associated with the expression of the EMT-associated transcription factors ZBTB46, FOXA2, and HIF1A. The increased abundance of MCTP1 promoted PC3 prostate cancer cell migration and neuroendocrine differentiation and was associated with SNAI1-dependent EMT in C4-2 PCa cells after ADT. ZBTB46 interacted with FOXA2 and HIF1A and increased the abundance of MCTP1 in a hypoxia-dependent manner. MCTP1 stimulated Ca2+ signaling and AKT activation to promote EMT and neuroendocrine differentiation by increasing the SNAI1-dependent expression of EMT and neuroendocrine markers, effects that were blocked by knockdown of MCTP1. These data suggest an oncogenic role for MCTP1 in the maintenance of a rare and aggressive prostate cancer subtype through its response to Ca2+ and suggest its potential as a therapeutic target.


Assuntos
Diferenciação Celular , Transição Epitelial-Mesenquimal , Neoplasias da Próstata , Animais , Humanos , Masculino , Camundongos , Androgênios/metabolismo , Androgênios/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Fator 3-beta Nuclear de Hepatócito/metabolismo , Fator 3-beta Nuclear de Hepatócito/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Células Neuroendócrinas/metabolismo , Células Neuroendócrinas/patologia , Células PC-3 , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
2.
Mol Oncol ; 18(6): 1665-1686, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38381121

RESUMO

Prostate stromal cells play a crucial role in the promotion of tumor growth and immune evasion in the tumor microenvironment (TME) through intricate molecular alterations in their interaction with prostate cancer (PCa) cells. While the impact of these cells on establishing an immunosuppressive response and influencing PCa aggressiveness remains incompletely understood. Our study shows that the activation of the leukemia inhibitory factor (LIF)/LIF receptor (LIFR) pathway in both prostate tumor and stromal cells, following androgen deprivation therapy (ADT), leads to the development of an immunosuppressive TME. Activation of LIF/LIFR signaling in PCa cells induces neuroendocrine differentiation (NED) and upregulates immune checkpoint expression. Inhibition of LIF/LIFR attenuates these effects, underscoring the crucial role of LIF/LIFR in linking NED to immunosuppression. Prostate stromal cells expressing LIFR contribute to NED and immunosuppressive marker abundance in PCa cells, while LIFR knockdown in prostate stromal cells reverses these effects. ADT-driven LIF/LIFR signaling induces brain-derived neurotrophic factor (BDNF) expression, which, in turn, promotes NED, aggressiveness, and immune evasion in PCa cells. Clinical analyses demonstrate elevated BDNF levels in metastatic castration-resistant PCa (CRPC) and a positive correlation with programmed death-ligand 1 (PDL1) and immunosuppressive signatures. This study shows that the crosstalk between PCa cells and prostate stromal cells enhances LIF/LIFR signaling, contributing to an immunosuppressive TME and NED in PCa cells through the upregulation of BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Neoplasias da Próstata , Microambiente Tumoral , Masculino , Humanos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neoplasias da Próstata/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/imunologia , Linhagem Celular Tumoral , Microambiente Tumoral/imunologia , Transdução de Sinais/efeitos dos fármacos , Fator Inibidor de Leucemia/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/metabolismo , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/genética , Animais , Antagonistas de Androgênios/farmacologia , Antagonistas de Androgênios/uso terapêutico , Tumores Neuroendócrinos/patologia , Tumores Neuroendócrinos/metabolismo , Tumores Neuroendócrinos/imunologia , Diferenciação Celular
3.
Eur Arch Otorhinolaryngol ; 280(9): 4285-4290, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37466661

RESUMO

OBJECTIVE: Observational studies suggested that peripheral blood eosinophils were associated with the risk of nasal polyps. However, these studies did not confirm the causality. This study aims to apply Mendelian randomization (MR) method to comprehensively assess the potential causal association between peripheral blood eosinophils and nasal polyps. METHODS: Genetic instrumental variables were extracted from the largest available genome-wide association study (GWAS) of European participants, which were used to investigate the relationship between peripheral blood eosinophils and nasal polyps. The inverse variance weighted method, the MR Egger method, and the weighted median method were applied for this analysis. MR-Egger intercept tests, leave-one-out analyses, and funnel plots were performed for the sensitivity analysis. RESULTS: With the inverse variance weighted method, the MR analysis suggested that there was a significant difference between peripheral blood eosinophils and the risk of nasal polyps (ukb-a-97, OR 1.004, 95% CI 1.003-1.005, p < 0.001; ukb-a-541, OR 1.005, 95% CI 1.004-1.006, p < 0.001; ukb-b-7211, OR 1.004, 95% CI 1.003-1.005, p < 0.001; ukb-b-8425, OR 1.004, 95% CI 1.003-1.005, p < 0.001; finn-b-J10_NASALPOLYP, OR 3.089, 95% CI 2.537-3.761, p < 0.001). Consistent results were also proved by using the weighted median method and the MR Egger method. CONCLUSIONS: Our findings reveal the causal effect of peripheral blood eosinophils on the increased risk of nasal polyps.


Assuntos
Eosinófilos , Pólipos Nasais , Humanos , Pólipos Nasais/genética , Estudo de Associação Genômica Ampla , Análise da Randomização Mendeliana , Causalidade , Polimorfismo de Nucleotídeo Único
4.
Sci China Life Sci ; 66(8): 1869-1887, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37059927

RESUMO

Protein-biomolecule interactions play pivotal roles in almost all biological processes. For a biomolecule of interest, the identification of the interacting protein(s) is essential. For this need, although many assays are available, highly robust and reliable methods are always desired. By combining a substrate-based proximity labeling activity from the pupylation pathway of Mycobacterium tuberculosis and the streptavidin (SA)-biotin system, we developed the Specific Pupylation as IDEntity Reporter (SPIDER) method for identifying protein-biomolecule interactions. Using SPIDER, we validated the interactions between the known binding proteins of protein, DNA, RNA, and small molecule. We successfully applied SPIDER to construct the global protein interactome for m6A and mRNA, identified a variety of uncharacterized m6A binding proteins, and validated SRSF7 as a potential m6A reader. We globally identified the binding proteins for lenalidomide and CobB. Moreover, we identified SARS-CoV-2-specific receptors on the cell membrane. Overall, SPIDER is powerful and highly accessible for the study of protein-biomolecule interactions.


Assuntos
COVID-19 , Humanos , SARS-CoV-2 , Proteínas , Ligação Proteica
5.
Redox Biol ; 62: 102686, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36963289

RESUMO

Conventional treatment of prostate cancer (PCa) uses androgen-deprivation therapy (ADT) to inhibit androgen receptor (AR) signaling-driven tumor progression. ADT-induced PCa recurrence may progress to an AR-negative phenotype with neuroendocrine (NE) histologic features, which are associated with metabolic disturbances and poor prognoses. However, the metabolic pathways that regulate NE differentiation (NED) in PCa remain unclear. Herein, we show a regulatory mechanism in NED-associated metabolism dysfunction induced by ADT, whereby overexpression of pyruvate kinase L/R (PKLR) mediates oxidative stress through upregulation of reactive oxygen species modulator 1 (ROMO1), thereby promoting NED and aggressiveness. ADT mediates the nuclear translocation of PKLR, which binds to the MYCN/MAX complex to upregulate ROMO1 and NE-related genes, leading to altered mitochondrial function and NED of PCa. Targeting nuclear PKLR/MYCN using bromodomain and extra-terminal motif (BET) inhibitors has the potential to reduce PKLR/MYCN-driven NED. Abundant ROMO1 in serum samples may provide prognostic information in patients with ADT. Our results suggest that ADT resistance leads to upregulation of PKLR/MYCN/ROMO1 signaling, which may drive metabolic reprogramming and NED in PCa. We further show that increased abundance of serum ROMO1 may be associated with the development of NE-like PCa.


Assuntos
Neoplasias de Próstata Resistentes à Castração , Neoplasias da Próstata , Humanos , Masculino , Antagonistas de Androgênios/farmacologia , Linhagem Celular Tumoral , Proteínas de Membrana , Proteínas Mitocondriais/metabolismo , Proteína Proto-Oncogênica N-Myc/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Piruvato Quinase/metabolismo , Transdução de Sinais
6.
J Biol Chem ; 298(6): 102017, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35526564

RESUMO

Jumonji domain-containing 3 (JMJD3/KDM6B) is a histone demethylase that plays an important role in regulating development, differentiation, immunity, and tumorigenesis. However, the mechanisms responsible for the epigenetic regulation of inflammation during mastitis remain incompletely understood. Here, we aimed to investigate the role of JMJD3 in the lipopolysaccharide (LPS)-induced mastitis model. GSK-J1, a small molecule inhibitor of JMJD3, was applied to treat LPS-induced mastitis in mice and in mouse mammary epithelial cells in vivo and in vitro. Breast tissues were then collected for histopathology and protein/gene expression examination, and mouse mammary epithelial cells were used to investigate the mechanism of regulation of the inflammatory response. We found that the JMJD3 gene and protein expression were upregulated in injured mammary glands during mastitis. Unexpectedly, we also found JMJD3 inhibition by GSK-J1 significantly alleviated the severity of inflammation in LPS-induced mastitis. These results are in agreement with the finding that GSK-J1 treatment led to the recruitment of histone 3 lysine 27 trimethylation (H3K27me3), an inhibitory chromatin mark, in vitro. Furthermore, mechanistic investigation suggested that GSK-J1 treatment directly interfered with the transcription of inflammatory-related genes by H3K27me3 modification of their promoters. Meanwhile, we also demonstrated that JMJD3 depletion or inhibition by GSK-J1 decreased the expression of toll-like receptor 4 and negated downstream NF-κB proinflammatory signaling and subsequently reduced LPS-stimulated upregulation of Tnfa, Il1b, and Il6. Together, we propose that targeting JMJD3 has therapeutic potential for the treatment of inflammatory diseases.


Assuntos
Inibidores Enzimáticos , Histona Desmetilases com o Domínio Jumonji , Mastite , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Células Epiteliais , Feminino , Histonas/metabolismo , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Lipopolissacarídeos , Glândulas Mamárias Animais/citologia , Mastite/induzido quimicamente , Mastite/tratamento farmacológico , Camundongos
7.
Mol Cell Proteomics ; 17(9): 1720-1736, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29871872

RESUMO

Proteins, as the major executer for cell progresses and functions, its abundance and the level of post-translational modifications, are tightly monitored by regulators. Genetic perturbation could help us to understand the relationships between genes and protein functions. Herein, to explore the impact of the genome-wide interruption on certain protein, we developed a cell lysate microarray on kilo-conditions (CLICK) with 4837 knockout (YKO) and 322 temperature-sensitive (ts) mutant strains of yeast (Saccharomyces cerevisiae). Taking histone marks as examples, a general workflow was established for the global identification of upstream regulators. Through a single CLICK array test, we obtained a series of regulators for H3K4me3, which covers most of the known regulators in S. cerevisiae We also noted that several group of proteins are involved in negatively regulation of H3K4me3. Further, we discovered that Cab4p and Cab5p, two key enzymes of CoA biosynthesis, play central roles in histone acylation. Because of its general applicability, CLICK array could be easily adopted to rapid and global identification of upstream protein/enzyme(s) that regulate/modify the level of a protein or the posttranslational modification of a non-histone protein.


Assuntos
Redes Reguladoras de Genes , Código das Histonas/genética , Saccharomyces cerevisiae/genética , Acil Coenzima A/metabolismo , Acilação , Química Click , Histonas/metabolismo , Lisina/metabolismo , Metilação , Modelos Biológicos , Mutação/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Estresse Fisiológico
8.
Growth Factors ; 26(4): 192-200, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19378419

RESUMO

OBJECTIVE AND DESIGN: This study investigates the regulatory role of connective tissue growth factor (CTGF) on production of fractalkine, monocyte-chemoattractant protein-1 (MCP-1) and regulated on activation, normal T cell expressed and secreted (RANTES) in human mesangial cells, and explore the mechanisms of CTGF action. METHODS: Cultured human mesangial cells were treated with CTGF. Expressions of mRNA and proteins of fractalkine, MCP-1 and RANTES were analyzed by real-time polymerase chain reaction (PCR) and by enzyme-linked immunosorbent assay, respectively. Expressions of phosphorylated extracellular signal-regulated kinase 1/2 (ERK1/2), phosphoinositide 3-kinase (PI3-K) and protein kinase B (PKB) were assessed by Western blotting. Activities of nuclear factor-KB (NF-KB) were determined by NF-kappaB luciferase reporter assay. RESULTS: CTGF enhanced the mRNA expressions and protein release of fractalkine, MCP-1 and RANTES, and the expressions of phosphorylated ERK1/2, PI3-K and PKB, and activities of NF-KB. Blockade of ERK1/2 inhibited the CTGF-induced expression ofphosphorylated ERK1/2 and NF-kappaB, and partially decreased the expressions of the above chemokines. PI3-K blockade downregulated the CTGF-stimulated expressions of phosphorylated PI3-K, PKB and NF-kappaB but not phosphorylated ERK1/2, partially decreased the expressions of the above chemokines. NF-kappaB blockade abrogated the CTGF-activated NF-kappaB and partially decreased the expressions of the above chemokines. Soluble heparin and K252a, an inhibitor of Trk, blocked CTGF-induced production of the above chemokines and the activation of the above signaling proteins. CONCLUSION: These results demonstrated that CTGF induces production of fractalkine, MCP-1 and RANTES via ERK1/2 and PI3-K/PKB/NF-kappaB-dependent signal pathway mediated by cell surface heparin sulfate proteoglycans and the tyrosine kinase receptor TrkA in human mesangial cells.


Assuntos
Quimiocinas/biossíntese , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Mesângio Glomerular/metabolismo , Células Mesangiais/metabolismo , Transdução de Sinais/fisiologia , Células Cultivadas , Quimiocina CCL2/biossíntese , Quimiocina CCL5/biossíntese , Quimiocina CX3CL1/biossíntese , Mesângio Glomerular/citologia , Humanos , Células Mesangiais/química , RNA Mensageiro/metabolismo
9.
Zhonghua Nan Ke Xue ; 13(4): 315-9, 2007 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-17491263

RESUMO

OBJECTIVE: To assay the expression of KiSS-1 and GnRH in the male rat hypothalamus at different developmental stages, and to explore the significance of KiSS-1 in sex development onset and normal reproduction regulation. METHODS: Expression analyses of KiSS-1 and GnRH genes were conducted in the rat hypothalamus at different developmental stages with RT-PCR and real time-PCR. The testosterone level was assayed by chemoluminescence technique. RESULTS: KiSS-1 mRNA rose gradually during sex development in the rat hypothalamus, highest at puberty and lowered a little at adulthood. KiSS-1 mRNA of the prepubertal, early pubertal, pubertal and adult rats was 1.7, 2.1, 3.5 and 2.0 times higher than that of the infantile rats respectively. The expression of GnRH and KiSS-1 correlated positively (r = 0.905, P < 0.05). But the activation of GnRH neuron was later than KiSS-1. The expression of GnRH was the highest in the puberty rats. GnRH mRNA of the prepubertal, early pubertal, pubertal and adult rats was 1.1, 1.94, 2.42 and 1.92 times higher than that of the infantile rats respectively. The level of testosterone in the adult rats was significantly higher than that at the earlier stage and was the highest at the adult stage. CONCLUSION: The expression of KiSS-1 correlates positively with that of GnRH. KiSS-1 may participate in the regulation of GnRH and is relevant to puberty onset and the regulation of reproduction function.


Assuntos
Hormônio Liberador de Gonadotropina/biossíntese , Hipotálamo/metabolismo , Proteínas/metabolismo , Animais , Hormônio Liberador de Gonadotropina/genética , Kisspeptinas , Masculino , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Am J Respir Cell Mol Biol ; 34(1): 65-72, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16141446

RESUMO

Connective tissue growth factor (CTGF) plays an important role in pathways leading to lung fibrosis via the mitogenic action of CTGF on fibroblasts. Studies have shown that lipoxin A4 (LXA4) inhibits proliferation of renal mesangial cells induced by leukotriene D4 or platelet-derived growth factor. This study investigates the regulatory role of LXA4 on proliferation of human lung fibroblasts (HLF) induced by CTGF and mechanisms of LXA4 action. CTGF induced HLF proliferation; enhanced the expression of cyclin D1; phosphorylated extracellular signal-regulated kinase (ERK)1/2, phosphoinositide 3-kinase (PI3-K), protein kinase B (PKB), and DNA-binding activity of signal transducers and activators of transcription-3 (STAT3); and inhibited expression of p27(kip1). LXA4 downregulated the CTGF-stimulated HLF proliferation and expression of cyclin D1; and phosphorylated ERK1/2, PI3-K, PKB, and DNA-binding activity of STAT3. CTGF-induced decrement in expression of p27(kip1) was ameliorated by LXA4. PI3-K or STAT blockade but not ERK1/2 blockade partially inhibited the CTGF-activated proliferation of HLF. Transfection of the human LXA4 receptor gene into HLF intensified the inhibition of LXA4 on CTGF-induced cell proliferation. These results demonstrate that CTGF induces proliferation of HLF via upregulation of PI3-K/PKB, STAT3, and cyclin D1, and downregulation of p27(kip1). LXA4 inhibits these effects of CTGF on HLF.


Assuntos
Proliferação de Células , Fibroblastos/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lipoxinas/metabolismo , Pulmão/citologia , Animais , Ciclo Celular/fisiologia , Linhagem Celular , Fator de Crescimento do Tecido Conjuntivo , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/citologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Receptores de Lipoxinas/genética , Receptores de Lipoxinas/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia
11.
Kidney Int ; 68(1): 35-46, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15954894

RESUMO

BACKGROUND: Studies have shown that lipoxin A(4) (LXA(4)) and its analogues inhibited proliferation of glomerular mesangial cells induced by leukotriene D(4) (LTD(4)) or platelet-derived growth factor (PDGF), reduced the production of proinflammatory cytokines such as interleukin (IL)-1beta and IL-6 in renal tissue of ischemic injury. In the present studies, we examine whether LXA(4) have inhibitory effects on tumor necrosis factor-alpha (TNF-alpha)-induced productions of IL-1beta and IL-6 and proliferation of glomerular mesangial cells of rat, and explore the molecular mechanisms of signal pathway of LXA(4). METHODS: Cultured glomerular mesangial cells were treated with TNF-alpha (10 ng/mL), with or without preincubation with LXA(4) at the different concentrations. Cell proliferation was assessed by [(3)H]-thymidine incorporation. Proteins of IL-1beta and IL-6 in supernatant were analyzed by enzyme-linked immunosorbent assay (ELISA). Expressions of mRNA of IL-1beta and IL-6 were determined by real-time polymerase chain reaction (PCR) and cyclin E by reverse transcription (RT)-PCR. Proteins of cyclin E, threonine phosphorylated Akt(1) at 308 site (Thr(308)) and p27(kip1) were analyzed by Western blotting studies. Activities of signal transducers and activators of transcription-3 (STAT(3)), nuclear factor-kappaB (NF-kappaB) were determined by electrophroretic mobility shift assay (EMSA). Expression of Src homology (SH) 2-containing protein-tyrosine phosphatase (SHP-2) was assessed by immunoprecipitation and immunoblotting. RESULTS: TNF-alpha-stimulated proliferation, release of proteins and expressions of mRNA of IL-1beta and IL-6 in mesangial cells were inhibited by LXA(4) in a dose-dependent manner. The marked increments in mRNA expression and protein synthesis of cyclin E induced by TNF-alpha in parallel with proliferation of mesangial cells were down-regulated by LXA(4). LXA(4) antagonized the phosphorylation of SHP-2 and activity of NF-kappaB induced by TNF-alpha. Pretreatment of the cells with NF-kappaB inhibitor pyrrolidine dithio-carbamate (PDTC) blocked the productions of IL-1beta, IL-6, and activation of NF-kappaB induced by TNF-alpha. Stimulation of mesangial cells with TNF-alpha resulted in enhanced DNA-binding activity of STAT(3). This increment was inhibited by LXA(4) in a dose-dependent manner. Threonine phosphorylated Akt(1) protein at 308 site stimulated by TNF-alpha was reduced by LXA(4.) TNF-alpha-induced decrement in expression of p27(kip1) protein was ameliorated by LXA(4) in a dose-dependent manner. CONCLUSION: TNF-alpha-induced proliferation and increment of cyclin E of rat mesangial cells can be inhibited by LXA(4), and these inhibitory effects might be through the mechanisms of STAT(3) and Akt(1)/p27(kip1) pathway-dependent signal transduction. LXA(4) also antagonized TNF-alpha-stimulated IL-1beta and IL-6 synthesis, and these antagonisms were related to SHP-2 and NF-kappaB pathway-dependent signal transduction.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Lipoxinas/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Ciclina E/genética , Inibidor de Quinase Dependente de Ciclina p27 , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/imunologia , Mesângio Glomerular/imunologia , Interleucina-1/genética , Interleucina-1/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , NF-kappa B/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Transativadores/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Supressoras de Tumor/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-15936930

RESUMO

Studies have implicated that lipoxinA4 (LXA4) inhibited nuclear factor-kappaB (NF-kappaB), Akt/PKB and PI 3-kinase activity and proliferation of glomerular mesangial cells. It is speculated that LXA4 might serve as pro-apoptotic factor. Rat renal interstitial fibroblasts (NRK-49F cells) were exposed to LXA4 in 5% FCS for 24 h. LXA4 at 0.1 and 1 microM induced 9.83% and 33.82% apoptosis of the cells, respectively, upregulated the expression of calpain 10 and Smac, the levels of [Ca2+]i and activity of caspase-3, and downregulated the activity of STAT3 and threonine phosphorylated Akt1. Transfection of calpain 10 or Smac antisense oligodeoxynucleotide into the cells inhibited the LXA4-induced apoptosis, activity of caspase-3. Pretreatment of the cells with calcium inhibitor SK&F96365 inhibited the LXA4-induced apoptosis, levels of [Ca2+]i, expression of calpain 10 and Smac. In conclusion, LXA4 at high concentrations induced apoptosis of renal interstitial fibroblasts via [Ca2+]i-dependent upregulation of calpain 10 and Smac expression.


Assuntos
Apoptose , Fibroblastos/efeitos dos fármacos , Rim/citologia , Lipoxinas/farmacologia , Animais , Proteínas Reguladoras de Apoptose , Cálcio/metabolismo , Calpaína/genética , Calpaína/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 3 , Caspases/genética , Caspases/metabolismo , Células Cultivadas , Regulação para Baixo , Lipoxinas/administração & dosagem , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Oligodesoxirribonucleotídeos Antissenso/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Fosforilação , Ratos , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...