Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
J Am Heart Assoc ; 10(4): e014311, 2021 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-33522247

RESUMO

Background Heart pathological hypertrophy has been recognized as a predisposing risk factor for heart failure and arrhythmia. DUSP (dual-specificity phosphatase) 26 is a member of the DUSP family of proteins, which has a significant effect on nonalcoholic fatty liver disease, neuroblastoma, glioma, and so on. However, the involvement of DUSP26 in cardiac hypertrophy remains unclear. Methods and Results Our study showed that DUSP26 expression was significantly increased in mouse hearts in response to pressure overload as well as in angiotensin II-treated cardiomyocytes. Cardiac-specific overexpression of DUSP26 mice showed attenuated cardiac hypertrophy and fibrosis, while deficiency of DUSP26 in mouse hearts resulted in increased cardiac hypertrophy and deteriorated cardiac function. Similar effects were also observed in cellular hypertrophy induced by angiotensin II. Importantly, we showed that DUSP26 bound to transforming growth factor-ß activated kinase 1 and inhibited transforming growth factor-ß activated kinase 1 phosphorylation, which led to suppression of the mitogen-activated protein kinase signaling pathway. In addition, transforming growth factor-ß activated kinase 1-specific inhibitor inhibited cardiomyocyte hypertrophy induced by angiotensin II and attenuated the exaggerated hypertrophic response in DUSP26 conditional knockout mice. Conclusions Taken together, DUSP26 was induced in cardiac hypertrophy and protected against pressure overload induced cardiac hypertrophy by modulating transforming growth factor-ß activated kinase 1-p38/ c-Jun N-terminal kinase-signaling axis. Therefore, DUSP26 may provide a therapeutic target for treatment of cardiac hypertrophy and heart failure.


Assuntos
Cardiomegalia/tratamento farmacológico , Fosfatases de Especificidade Dupla/farmacologia , Regulação da Expressão Gênica , MAP Quinase Quinase Quinases/genética , Fosfatases da Proteína Quinase Ativada por Mitógeno/farmacologia , Miócitos Cardíacos/metabolismo , Animais , Cardiomegalia/genética , Cardiomegalia/metabolismo , Modelos Animais de Doenças , Ecocardiografia , MAP Quinase Quinase Quinases/biossíntese , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , RNA/genética , Transdução de Sinais
2.
Cell Cycle ; 20(3): 320-331, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33461373

RESUMO

Triple-negative breast cancer (TNBC) is the most aggressive histological subtype of breast cancer and is characterized by poor outcomes and a lack of specific-targeted therapies. Transforming growth factor-ß (TGF-ß) acts as the key cytokine in the epithelial-mesenchymal transition (EMT) and the metastasis of TNBC. However, the regulatory mechanisms of the TGF-ß signaling pathway remain largely unknown. In this study, we identified that the USP1/WDR48 complex could effectively enhance TGF-ß-mediated EMT and migration of TNBC cells. Furthermore, lower phosphorylation of Smad2/3, Erk, Jnk, and p38 was noted on the suppression of the expression of endogenous USP1 or WDR48. Moreover, the USP1-WDR48 complex was found to downregulate the polyubiquitination of TAK1 and mediate its in vitro stability. Therefore, our findings have shed a light on the novel role of the USP1/WDR48 complex in promoting TGF-ß-induced EMT and migration in TNBC via in vitro stabilization of TAK1.


Assuntos
Transição Epitelial-Mesenquimal/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , MAP Quinase Quinase Quinases/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Neoplasias de Mama Triplo Negativas/metabolismo , Proteases Específicas de Ubiquitina/biossíntese , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , MAP Quinase Quinase Quinases/genética , Estabilidade Proteica , Fator de Crescimento Transformador beta/genética , Neoplasias de Mama Triplo Negativas/genética , Proteases Específicas de Ubiquitina/genética
3.
Inflammation ; 43(4): 1548-1560, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32388658

RESUMO

The lncRNA nuclear enriched abundant transcript 1 (NEAT1) promotes sepsis-inflammatory responses and acute kidney injury (AKI), but little known about the underlying mechanisms. This study aims to investigate the roles of NEAT1 in regulating macrophage polarization and its potential for alleviating inflammatory responses during sepsis pathogenesis. Mouse RAW264.7 macrophages were treated with lipopolysaccharide (LPS) as a cellular inflammatory model. NEAT1 shRNA, miR-125a-5p mimics, and TRAF6-overexpressing vector were used to transfect RAW264.7 cells. NEAT1, miR-125a-5p, and mRNA levels of functional genes were detected by quantitative RT-PCR. Protein abundances were analyzed by western blotting. Macrophage polarization was evaluated by flow cytometry. The bindings of miR-125a-5p with NEAT1 or TRAF6 gene were validated by dual luciferase reporter assay. LPS treatment promoted NEAT1 and suppressed miR-125a-5p expression in mouse macrophage cells. NEAT1 silencing by shRNAs promoted macrophage M2 polarization under LPS treatment, which upregulated miR-125a-5p expression, repressed TRAF6 expression and TAK1 protein phosphorylation in macrophages. These cellular and molecular changes induced by NEAT1 shRNAs were abrogated by miR-125a-5p inhibitors. Moreover, miR-125a-5p mimics suppressed TRAF6 expression and TAK1 protein phosphorylation in LPS-treated macrophages, thus causing macrophage M2 polarization under LPS treatment. TRAF6 overexpression abrogated the miR-125a-5p mimics-induced macrophage M2 polarization. miR-125a-5p could directly bind to NEAT1 or TRAF6 gene in macrophages. lncRNA NEAT1 knockdown ameliorates LPS-induced inflammation by promoting macrophage M2 polarization via miR-125a-5p/TRAF6/TAK1 axis.


Assuntos
Polaridade Celular/fisiologia , MAP Quinase Quinase Quinases/biossíntese , Macrófagos/metabolismo , MicroRNAs/biossíntese , RNA Longo não Codificante/biossíntese , Fator 6 Associado a Receptor de TNF/biossíntese , Animais , Polaridade Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Células HEK293 , Humanos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/prevenção & controle , Lipopolissacarídeos/toxicidade , Macrófagos/efeitos dos fármacos , Camundongos , Células RAW 264.7 , RNA Longo não Codificante/antagonistas & inibidores
4.
Mol Cancer Ther ; 19(1): 247-257, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31562256

RESUMO

YAP and TAZ are central determinants of malignancy; however, their functions remain still undruggable. We identified TGFß-activated kinase 1 (TAK1) as a central hub integrating the most relevant signals sustaining pancreatic cancer aggressiveness and chemoresistance. Glycogen synthase kinase (GSK)3 is known to stabilize TAK1, and its inhibition causes a reduction in TAK1 levels. Here, we hypothesized that TAK1 could sustain YAP/TAZ program, and thus, modulation of TAK1 expression through the inhibition of GSK3 could impair YAP/TAZ functions in pancreatic cancer.Differentially expressed transcripts between pancreatic cancer cells expressing scramble or TAK1-specific shRNA were annotated for functional interrelatedness by ingenuity pathway analysis. TAK1 expression was modulated by using different GSK3 inhibitors, including LY2090314. In vivo activity of LY2090314 alone or in combination with nab-paclitaxel was evaluated in an orthotopic nude mouse model.Differential gene expression profiling revealed significant association of TAK1 expression with HIPPO and ubiquitination pathways. We measured a significant downregulation of YAP/TAZ and their regulated genes in shTAK1 cells. TAK1 prevented YAP/TAZ proteasomal degradation in a kinase independent manner, through a complex with TRAF6, thereby fostering their K63-ubiquitination versus K48-ubiquitination. Pharmacologic modulation of TAK1 by using GSK3 inhibitors significantly decreased YAP/TAZ levels and suppressed their target genes and oncogenic functions. In vivo, LY2090314 plus nab-paclitaxel significantly prolonged mice survival duration.Our study demonstrates a unique role for TAK1 in controlling YAP/TAZ in pancreatic cancer. LY2090314 is a novel agent that warrants further clinical development in combination with nab-paclitaxel for the treatment of pancreatic cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , MAP Quinase Quinase Quinases/biossíntese , Neoplasias Pancreáticas/metabolismo , Transativadores/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Albuminas/administração & dosagem , Albuminas/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Feminino , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Compostos Heterocíclicos com 3 Anéis/farmacologia , Xenoenxertos , Humanos , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Maleimidas/administração & dosagem , Maleimidas/farmacologia , Camundongos , Camundongos Nus , Paclitaxel/administração & dosagem , Paclitaxel/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Distribuição Aleatória , Transativadores/genética , Transativadores/metabolismo , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP
5.
Eur Rev Med Pharmacol Sci ; 23(15): 6497-6504, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31378889

RESUMO

OBJECTIVE: Esophagus squamous cell carcinoma (ESCC) was a dominant histological type of esophagus cancer, which has a very high incidence due to distant metastasis and local invasion. MicroRNA-148a (miR-148a) functioned as a tumor suppressor in a variety of cancers. The purpose of our study was to explore the vital role of miR-148a in esophagus squamous cell carcinoma. PATIENTS AND METHODS: The Kaplan-Meier method was applied to calculate the 5-year overall survival of esophagus squamous cell carcinoma patients. Real Time-quantitative Polymerase Chain Reaction (RT-qPCR) and Western blot were conducted to calculate the mRNA levels of miR-148a and genes. The cell counting kit-8 (CCK-8) and transwell assays were performed to measure the proliferative and invasive ability. RESULTS: MiR-148a was observed to be significantly downregulated and the downregulation of miR-148 predicted poor prognosis of esophagus squamous cell carcinoma patients. MAP3K9 was a target gene of miR-148a and its expression was mediated by miR-148a through directly binding to the 3'-untranslated region (3'-UTR) of its mRNA in the esophagus squamous cell carcinoma. Moreover, miR-148a remarkably inhibited the proliferation and invasion through directly targeting to MAP3K9 via extracellular-signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway and epithelial-mesenchymal transition (EMT) in the ESCC cells. In addition, overexpression of miR-148a inhibited the growth of ESCC in vivo. CONCLUSIONS: MiR-148a inhibited the proliferation and invasion through directly targeting to MAP3K9 by ERK/MAPK pathway and EMT in ESCC cells. The newly identified miR-148a/MAP3K9 axis provides a novel insight into the pathogenesis of the esophagus squamous cell carcinoma.


Assuntos
Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , MAP Quinase Quinase Quinases/biossíntese , Sistema de Sinalização das MAP Quinases/fisiologia , MicroRNAs/biossíntese , Animais , Proliferação de Células/fisiologia , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Nus , MicroRNAs/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
6.
Cell Death Differ ; 26(12): 2520-2534, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30850732

RESUMO

Melanoma cells are highly resistant to conventional genotoxic agents, and BRAFV600/MEK-targeted therapies as well as immunotherapies frequently remain inefficient. Alternative means to treat melanoma, in particular through the induction of programmed cell death modalities such as apoptosis or necroptosis, therefore still need to be explored. Here, we report that melanoma cell lines expressing notable amounts of RIPK1, RIPK3 and MLKL, the key players of necroptosis signal transduction, fail to execute necroptotic cell death. Interestingly, the activity of transforming growth factor ß-activated kinase 1 (TAK1) appears to prevent RIPK1 from contributing to cell death induction, since TAK1 inhibition by (5Z)-7-Oxozeaenol, deletion of MAP3K7 or the expression of inactive TAK1 were sufficient to sensitize melanoma cells to RIPK1-dependent cell death in response to TNFα or TRAIL based combination treatments. However, cell death was executed exclusively by apoptosis, even when RIPK3 expression was high. In addition, TAK1 inhibitor (5Z)-7-Oxozeaenol suppressed intrinsic or treatment-induced pro-survival signaling as well as the secretion of cytokines and soluble factors associated with melanoma disease progression. Correspondingly, elevated expression of TAK1 correlates with reduced disease free survival in patients diagnosed with primary melanoma. Overall, our results therefore demonstrate that TAK1 suppresses the susceptibility to RIPK1-dependent cell death and that high expression of TAK1 indicates an increased risk for disease progression in melanoma.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Melanoma/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Decitabina/farmacologia , Progressão da Doença , Humanos , MAP Quinase Quinase Quinases/biossíntese , MAP Quinase Quinase Quinases/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Necroptose , Proteína Serina-Treonina Quinases de Interação com Receptores/biossíntese , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Transdução de Sinais , Transfecção , Zearalenona/análogos & derivados , Zearalenona/farmacologia
7.
Oncogene ; 37(27): 3672-3685, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29622797

RESUMO

BMP receptor inhibitors induce death of cancer cells through the downregulation of antiapoptotic proteins XIAP, pTAK1, and Id1-Id3. However, the current most potent BMP receptor inhibitor, DMH2, does not downregulate BMP signaling in vivo because of metabolic instability and poor pharmacokinetics. Here we identified the site of metabolic instability of DMH2 and designed a novel BMP receptor inhibitor, JL5. We show that JL5 has a greater volume of distribution and suppresses the expression of Id1 and pTak1 in tumor xenografts. Moreover, we demonstrate JL5-induced tumor cell death and tumor regression in xenograft mouse models without immune cells and humanized with adoptively transferred human immune cells. In humanized mice, JL5 additionally induces the infiltration of immune cells within the tumor microenvironment. Our studies show that the BMP signaling pathway is targetable in vivo and BMP receptor inhibitors can be developed as a therapeutic to treat cancer patients.


Assuntos
Antineoplásicos/farmacologia , Receptores de Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Quinolonas/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Células A549 , Transferência Adotiva , Animais , Antineoplásicos/química , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Proteína 1 Inibidora de Diferenciação/biossíntese , Neoplasias Pulmonares/patologia , MAP Quinase Quinase Quinases/biossíntese , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Inibidores de Proteínas Quinases/química , Pirazóis/química , Pirazóis/uso terapêutico , Pirimidinas/química , Pirimidinas/uso terapêutico , Quinolonas/química , Quinolonas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Pathol Res Pract ; 213(4): 389-393, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28214207

RESUMO

Osteosarcoma is the most common type of bone cancer prevalent in young adults. Recent studies suggested that aberrant microRNA expression plays an essential role in osteosarcoma pathogenesis. In this study, we found miR-155 up-regulation in different osteosarcoma cell lines U2OS, Saos-2 and MG-63. Through bioinformatic prediction and analysis, we identified its target MAP3K10 that involved in cell proliferation and apoptosis. This work demonstrates novel interaction between microRNA, intercellular MAPK signaling and apoptosis in osteosarcoma, which will provide targets for therapeutic development.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , MAP Quinase Quinase Quinases/biossíntese , MicroRNAs/genética , Osteossarcoma/patologia , Animais , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase em Tempo Real
9.
Microrna ; 5(1): 57-65, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280801

RESUMO

BACKGROUND: The initiation of ß-cell proliferation to recover reduced ß-cell mass is considered as one of the attractive therapeutic approaches for type 1 and 2 diabetes. In this study, we investigated the involvement of miRNAs in ß-cell proliferation. METHODS: Global miRNA array analysis of pancreas tissue was carried out using a 60% partial pancreatectomy (PPx) rodent model, which is a well-characterized model for pancreatic regeneration with accelerated proliferation of ß-cells. To explore miRNAs with mitogenic activity on ß-cells, precursors and antisense oligonucleotides (ASOs) for miRNAs were transfected into a primary islet monolayer cell cultures isolated from adult rats in order to modify their expression and proliferation of ß-cells. RESULTS: We found that miR-199b-5p, which was up-regulated 2.6 times in the pancreas of the PPx treated group, significantly enhanced the proliferation of ß-cells when its precursor was over-expressed. Target genes of miR-199b-5p were investigated and Mixed lineage kinase-3 (MLK3) was identified as one of the candidates since its expression was down-regulated through an interaction with miR-199b-5p and siRNA treatment for MLK3 enhanced the proliferation of ß-cells. CONCLUSION: Our data suggest that the up-regulation of miR-199b-5p enhances proliferation of ß-cells at least in part through down-regulation of MLK3.


Assuntos
Proliferação de Células/genética , Células Secretoras de Insulina/citologia , MAP Quinase Quinase Quinases/genética , MicroRNAs/genética , Animais , Células Cultivadas , Ciclina D1/metabolismo , Ciclina E/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/terapia , Regulação para Baixo/genética , Regulação da Expressão Gênica/genética , MAP Quinase Quinase Quinases/biossíntese , Masculino , MicroRNAs/biossíntese , Oligonucleotídeos Antissenso/genética , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
10.
IUBMB Life ; 68(2): 145-55, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26748475

RESUMO

Inflammation of infiltrated macrophages in adipose tissue is a key contributor to the initiation of adipose insulin resistance. These macrophages are exposed to high local concentrations of free fatty acids (FFAs) and can be proinflammatory activated by saturated fatty acids (SFAs). However, the regulatory mechanisms on SFA-induced macrophage inflammation are still elusive. Peroxisome proliferator-activated receptor γ coactivator-1ß (PGC-1ß) is a member of the PGC-1 family of transcriptional coactivators and has been reported to play a key role in SFAs metabolism and in the regulation of inflammatory signaling. However, it remains unclear whether PGC-1ß is involved in SFA-induced macrophage inflammation. In this study, we found that PGC-1ß expression was significantly decreased in response to palmitic acid (PA) in macrophages in a dose dependent manner. PGC-1ß inhibited PA induced TNFα, MCP-1, and IL-1ß mRNA and protein expressions. Furthermore, PGC-1ß significantly antagonized PA induced macrophage nuclear factor-κB (NF-κB) p65 and JUN N-terminal kinase activation. Mechanistically, we revealed that TGF-ß-activated kinase 1 (TAK1) and its adaptor protein TAK1 binding protein 1 (TAB1) played a dominant role in the regulatory effects of PGC-1ß. We confirmed that PGC-1ß inhibited downstream inflammatory signals via binding with TAB1 and thus preventing TAB1/TAK1 binding and TAK1 activation. Finally, we showed that PGC-1ß overexpression in PA treated macrophages improved adipocytes PI3K-Akt insulin signaling in a paracrine fashion. Collectively, our results uncovered a novel mechanism on how macrophage inflammation induced by SFAs was regulated and suggest a potential target in the treatment of obesity induced insulin resistance.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Tecido Adiposo/metabolismo , Inflamação/genética , MAP Quinase Quinase Quinases/genética , Fatores de Transcrição/genética , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Tecido Adiposo/efeitos dos fármacos , Animais , Quimiocina CCL2/biossíntese , Regulação da Expressão Gênica , Humanos , Inflamação/induzido quimicamente , Inflamação/patologia , Resistência à Insulina/genética , Interleucina-1beta/biossíntese , MAP Quinase Quinase Quinases/biossíntese , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Ácido Palmítico/toxicidade , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Fatores de Transcrição/biossíntese , Ativação Transcricional/genética , Fator de Necrose Tumoral alfa/biossíntese
11.
Blood ; 126(24): 2649-52, 2015 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-26468227

Assuntos
Antineoplásicos/uso terapêutico , Imidazóis/uso terapêutico , Sarcoma de Células de Langerhans/tratamento farmacológico , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Oximas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Azetidinas/administração & dosagem , Ciclofosfamida/administração & dosagem , Doxorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos , Substituição de Medicamentos , Humanos , Indóis/administração & dosagem , Sarcoma de Células de Langerhans/diagnóstico por imagem , Sarcoma de Células de Langerhans/genética , Sarcoma de Células de Langerhans/patologia , Linfonodos/diagnóstico por imagem , Linfonodos/patologia , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/biossíntese , MAP Quinase Quinase Quinases/genética , Masculino , Pessoa de Meia-Idade , Imagem Multimodal , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Piperidinas/administração & dosagem , Mutação Puntual , Tomografia por Emissão de Pósitrons , Prednisona/administração & dosagem , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/biossíntese , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Recidiva , Sulfonamidas/administração & dosagem , Tomografia Computadorizada por Raios X , Vemurafenib , Vincristina/administração & dosagem
12.
J Ethnopharmacol ; 174: 195-9, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26297845

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: As documented in the Chinese Materia Medica Grand Dictionary (), a herbal formula (RL) consisting of Rosae Multiflorae Fructus (multiflora rose hips) and Lonicerae Japonicae Flos (Japanese honeysuckle flowers) has traditionally been used in treating inflammatory disorders. RL was previously reported to inhibit the expression of various inflammatory mediators regulated by NF-κB and MAPKs that are components of the TLR4 signalling pathways. AIM OF THE STUDY: This study aims to provide further justification for clinical application of RL in treating inflammatory disorders by further delineating the involvement of the TLR4 signalling cascades in the effects of RL on inflammatory mediators. MATERIALS AND METHODS: RL consisting of Rosae Multiflorae Fructus and Lonicerae Japonicae Flos (in 5:3 ratio) was extracted using absolute ethanol. We investigated the effect of RL on the production of cytokines and chemokines that are regulated by three key transcription factors of the TLR4 signalling pathways AP-1, NF-κB and IRF3 in LPS-stimulated RAW264.7 cells using the multiplex biometric immunoassay. Phosphorylation of AP-1, NF-κB, IRF3, IκB-α, IKKα/ß, Akt, TAK1, TBK1, IRAK-1 and IRAK-4 were examined in LPS-stimulated RAW264.7 cells and THP-1 cells using Western blotting. Nuclear localizations of AP-1, NF-κB and IRF3 were also examined using Western blotting. RESULTS: RL reduced the secretion of various pro-inflammatory cytokines and chemokines regulated by transcription factors AP-1, NF-κB and IRF3. Phosphorylation and nuclear protein levels of these transcription factors were decreased by RL treatment. Moreover, RL inhibited the activation/phosphorylation of IκB-α, IKKα/ß, TAK1, TBK1 and IRAK-1. CONCLUSIONS: Suppression of the IRAK-1/TAK1 and TBK1/IRF3 signalling pathways was associated with the effect of RL on inflammatory mediators in LPS-stimulated RAW264.7 and THP-1 cells. This provides further pharmacological basis for the clinical application of RL in the treatment of inflammatory disorders.


Assuntos
Fator Regulador 3 de Interferon/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/antagonistas & inibidores , Lonicera , MAP Quinase Quinase Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Rosa , Animais , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/farmacologia , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Fator Regulador 3 de Interferon/biossíntese , Quinases Associadas a Receptores de Interleucina-1/biossíntese , MAP Quinase Quinase Quinases/biossíntese , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia , Preparações de Plantas/isolamento & purificação , Preparações de Plantas/farmacologia , Proteínas Serina-Treonina Quinases/biossíntese
13.
J Cell Biochem ; 116(12): 2778-85, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25959272

RESUMO

Although evidence suggests that the RAF/MEK/ERK pathway plays an important role in triple negative breast cancer (TNBC), resistance to MEK inhibitors has been observed in TNBC cells. Different mechanisms have been hypothesized to be involved in this phenomenon, including receptor tyrosine kinase-dependent activation of the PI3K/AKT pathway. In this study, we analyzed the effects of the MEK1/2 inhibitor selumetinib in combination with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib in a panel of TNBC cell lines that showed different levels of sensitivity to single-agent selumetinib: SUM-149 and MDA-MB-231 cells resulted to be sensitive, whereas SUM-159, MDA-MB-468 and HCC70 cells were relatively resistant to the drug. Treatment of TNBC cells with selumetinib produced an increase of the phosphorylation of the EGFR both in selumetinib-sensitive SUM-149, MDA-MB-231 and in selumetinib-resistant MDA-MB-468 TNBC cells. The combination of selumetinib and gefitinib resulted in a synergistic growth inhibitory effect in all the TNBC cell lines, although the IC50 was not reached in SUM-159 and MDA-MB-468 cells. This effect was associated with an almost complete suppression of ERK1/2 activation and a reduction of selumetinib-induced AKT phosphorylation. In addition, in selumetinib-sensitive TNBC cells the combination of selumetinib and gefitinib induced a significant G0/G1 cell cycle arrest and apoptosis. Taken together, our data demonstrated that blockade of the EGFR might efficiently increase the antitumor activity of selumetinib in a subgroup of TNBC and that this phenomenon might be related to the effects of such combination on both ERK1/2 and AKT activation.


Assuntos
Benzimidazóis/administração & dosagem , Receptores ErbB/antagonistas & inibidores , MAP Quinase Quinase Quinases/antagonistas & inibidores , Quinazolinas/administração & dosagem , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Receptores ErbB/biossíntese , Feminino , Gefitinibe , Humanos , MAP Quinase Quinase Quinases/biossíntese , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Oncogênica v-akt/genética , Inibidores de Proteínas Quinases/administração & dosagem , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
14.
Mol Med Rep ; 12(1): 1535-43, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25815776

RESUMO

microRNAs (miRNAs; miR) are a class of small non-coding RNA molecules, which are involved in the pathogenesis of human diseases through the negative regulation of gene expression. Previous studies have demonstrated that miR-509-3p is a novel miRNA associated with cell proliferation and migration in 786-O renal cell carcinoma (RCC) cells. However, the mechanism of action of miR-509-3p in RCC remains to be elucidated. The present study aimed to examine the functional role and mechanism of miR-509-3p in the development of RCC. The results demonstrated that the expression levels of miR-509-3p were downregulated in the 786-O and ACHN RCC cell lines compared with the normal tissues of 10 patients with RCC, as determined by reverse transcription-quantitative polymerase chain reaction. The mRNA expression levels of mitogen-activated protein kinase kinase kinase 8 (MAP3K8) were upregulated in the RCC cell lines. Functional investigations demonstrated that the overexpression of miR-509-3p inhibited the migration and proliferation of the RCC cells, as determined by wound scratch and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays. Luciferase reporter assays revealed that the overexpression of miR-509-3p reduced the transcriptional activity of MAP3K8. Furthermore, the present study demonstrated that the ectopic transfection of miR-509-3p led to a significant reduction in the mRNA and protein expression levels of MAP3K8 in the RCC cells. Finally, knockdown of MAP3K8 inhibited the migration and proliferation of the RCC cells. Therefore, the results of the present study demonstrated that the miR-509-3p RCC suppressor was a significant regulator of the MAP3K8 oncogene, suggesting that it may have a potential therapeutic role in the treatment of RCC.


Assuntos
Carcinoma de Células Renais/genética , Proliferação de Células/genética , MAP Quinase Quinase Quinases/biossíntese , MicroRNAs/genética , Proteínas Proto-Oncogênicas/biossíntese , Adulto , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , MAP Quinase Quinase Quinases/genética , Masculino , MicroRNAs/biossíntese , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas/genética
15.
Mol Cell Biol ; 34(16): 3132-43, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24912674

RESUMO

Mixed-lineage kinase 3 (MLK3) activates mitogen-activated protein kinase (MAPK) signaling pathways and has important functions in migration, invasion, proliferation, tumorigenesis, and apoptosis. We investigated the role of the E3 ligase carboxyl terminus of Hsc70-interacting protein (CHIP) in the regulation of MLK3 protein levels. We show that CHIP interacts with MLK3 and, together with the E2 ubiquitin-conjugating enzyme UbcH5 (UbcH5a, -b, -c, or -d), ubiquitinates MLK3 in vitro. CHIP or Hsp70 overexpression promoted endogenous MLK3 ubiquitination and induced a decline in MLK3 protein levels in cells with Hsp90 inhibition. Furthermore, CHIP overexpression caused a proteasome-dependent reduction in exogenous MLK3 protein. Geldanamycin (GA), heat shock, and osmotic shock treatments also reduced the level of MLK3 protein via a CHIP-dependent mechanism. In addition, CHIP depletion in ovarian cancer SKOV3 cells increased cell invasion, and the enhancement of invasiveness was abrogated by small interfering RNA (siRNA)-mediated knockdown of MLK3. Thus, CHIP modulates MLK3 protein levels in response to GA and stress stimuli, and CHIP-dependent regulation of MLK3 is required for suppression of SKOV3 ovarian cancer cell invasion.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , Ubiquitina-Proteína Ligases/genética , Benzoquinonas/farmacologia , Linhagem Celular Tumoral , Movimento Celular/genética , Inibidores Enzimáticos/farmacologia , Células HEK293 , Proteínas de Choque Térmico HSP70/antagonistas & inibidores , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP90/biossíntese , Proteínas de Choque Térmico HSP90/metabolismo , Resposta ao Choque Térmico , Humanos , Lactamas Macrocíclicas/farmacologia , MAP Quinase Quinase Quinases/biossíntese , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Pressão Osmótica , Ligação Proteica , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Sorbitol/farmacologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitinação , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
16.
BMC Cancer ; 14: 182, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24628919

RESUMO

BACKGROUND: MLK3 gene mutations were described to occur in about 20% of microsatellite unstable gastrointestinal cancers and to harbor oncogenic activity. In particular, mutation P252H, located in the kinase domain, was found to have a strong transforming potential, and to promote the growth of highly invasive tumors when subcutaneously injected in nude mice. Nevertheless, the molecular mechanism underlying the oncogenic activity of P252H mutant remained elusive. METHODS: In this work, we performed Illumina Whole Genome arrays on three biological replicas of human HEK293 cells stably transfected with the wild-type MLK3, the P252H mutation and with the empty vector (Mock) in order to identify the putative signaling pathways associated with P252H mutation. RESULTS: Our microarray results showed that mutant MLK3 deregulates several important colorectal cancer- associated signaling pathways such as WNT, MAPK, NOTCH, TGF-beta and p53, helping to narrow down the number of potential MLK3 targets responsible for its oncogenic effects. A more detailed analysis of the alterations affecting the WNT signaling pathway revealed a down-regulation of molecules involved in the canonical pathway, such as DVL2, LEF1, CCND1 and c-Myc, and an up-regulation of DKK, a well-known negative regulator of canonical WNT signaling, in MLK3 mutant cells. Additionally, FZD6 and FZD10 genes, known to act as negative regulators of the canonical WNT signaling cascade and as positive regulators of the planar cell polarity (PCP) pathway, a non-canonic WNT pathway, were found to be up-regulated in P252H cells. CONCLUSION: The results provide an overall view of the expression profile associated with mutant MLK3, and they support the functional role of mutant MLK3 by showing a deregulation of several signaling pathways known to play important roles in the development and progression of colorectal cancer. The results also suggest that mutant MLK3 may be a novel modulator of WNT signaling, and pinpoint the activation of PCP pathway as a possible mechanism underlying the invasive potential of MLK3 mutant cells.


Assuntos
Neoplasias Colorretais/genética , MAP Quinase Quinase Quinases/genética , Transdução de Sinais/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Estudo de Associação Genômica Ampla , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/biossíntese , Mutagênese Sítio-Dirigida , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
17.
J Biol Chem ; 289(1): 299-311, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24275658

RESUMO

The induction of the intrinsic antiviral defense in mammals relies on the accumulation of foreign genetic material. As such, complete engagement of this response is limited to replication-competent viruses. Interferon regulatory factors (IRFs) are mediators of this defense with shared enhancer elements but display a spectrum of transcriptional potential. Here we describe a mechanism designed to enhance this response should a pathogen not be successfully inhibited. We find that activation of IRF7 results in the induction of MAP3K8 and restructuring of the antiviral transcriptome. MAP3K8 mediates the phosphorylation and repression of IRF3 homodimers to promote greater transcriptional activity through utilization of IRF3:IRF7 heterodimers. Among the genes influenced by the MAP3K8/IRF7 signaling axis are members of the SP100 gene family that serve as general transcriptional enhancers of the antiviral defense. We propose that this feed forward loop serves to reinforce the cellular response and is reserved for imminent threats to the host.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , MAP Quinase Quinase Quinases/biossíntese , Sistema de Sinalização das MAP Quinases , Proteínas Proto-Oncogênicas/biossíntese , Vesiculovirus/metabolismo , Animais , Linhagem Celular , Indução Enzimática/genética , Humanos , Fator Regulador 3 de Interferon/genética , Fator Regulador 7 de Interferon/genética , MAP Quinase Quinase Quinases/genética , Camundongos , Multimerização Proteica/genética , Proteínas Proto-Oncogênicas/genética , Infecções por Rhabdoviridae/genética , Infecções por Rhabdoviridae/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Vesiculovirus/genética
18.
Biomed Res Int ; 2013: 580135, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24151609

RESUMO

IL-6 and TNF α were significantly increased in the bone marrow aspirate samples of patients with active multiple myeloma (MM) compared to those of normal controls. Furthermore, MM patients with advanced aggressive disease had significantly higher levels of IL-6 and TNF α than those with MM in plateau phase. TNF α increased interleukin-6 (IL-6) production from MM cells. However, the detailed mechanisms involved in signaling pathways by which TNF α promotes IL-6 secretion from MM cells are largely unknown. In our study, we found that TNF α treatments induce MEK and AKT phosphorylation. TNF α -stimulated IL-6 production was abolished by inhibition of JAK2 and IKK ß or by small interfering RNA (siRNA) targeting TNF receptors (TNFR) but not by MEK, p38, and PI3K inhibitors. Also, TNF α increased phosphorylation of STAT3 (ser727) including c-Myc and cyclin D1. Three different types of JAK inhibitors decreased the activation of the previously mentioned pathways. In conclusion, blockage of JAK/STAT-mediated NF- κ B activation was highly effective in controlling the growth of MM cells and, consequently, an inhibitor of TNF α -mediated IL-6 secretion would be a potential new therapeutic agent for patients with multiple myeloma.


Assuntos
Interleucina-6/biossíntese , Janus Quinase 2/biossíntese , Mieloma Múltiplo/genética , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Quinase I-kappa B/metabolismo , Interleucina-6/metabolismo , Janus Quinase 2/antagonistas & inibidores , MAP Quinase Quinase Quinases/biossíntese , Masculino , Pessoa de Meia-Idade , Mieloma Múltiplo/patologia , Proteína Oncogênica v-akt/biossíntese , Proteína Oncogênica v-akt/metabolismo , Fosforilação , RNA Interferente Pequeno , Fatores de Transcrição STAT , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
19.
Biochem J ; 452(2): 359-65, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23557442

RESUMO

Activation of PKR (double-stranded-RNA-dependent protein kinase) by DNA plasmids decreases translation, and limits the amount of recombinant protein produced by transiently transfected HEK (human embryonic kidney)-293 cells. Co-expression with Ebola virus VP35 (virus protein 35), which blocked plasmid activation of PKR, substantially increased production of recombinant TPL-2 (tumour progression locus 2)-ABIN-2 [A20-binding inhibitor of NF-κB (nuclear factor κB) 2]-NF-κB1 p105 complex. VP35 also increased expression of other co-transfected proteins, suggesting that VP35 could be employed generally to boost recombinant protein production by HEK-293 cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Ebolavirus/fisiologia , MAP Quinase Quinase Quinases/biossíntese , Subunidade p50 de NF-kappa B/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Regulação para Cima/genética , Proteínas Virais Reguladoras e Acessórias/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Ebolavirus/genética , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/genética , Complexos Multiproteicos/biossíntese , Complexos Multiproteicos/genética , Subunidade p50 de NF-kappa B/genética , Proteínas Proto-Oncogênicas/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Transfecção , Proteínas Virais Reguladoras e Acessórias/genética
20.
Ann Thorac Surg ; 95(1): 285-90, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23272845

RESUMO

BACKGROUND: Transforming growth factor-ß activated kinase-1 (TAK1) is a serine/threonine kinase in the mitogen-activated protein kinase kinase family. Previous studies have reported the role of TAK1 in cancer occurrence and progression; however, its role and clinical significance in esophageal squamous cell carcinoma (ESCC) has not been elucidated. We investigated the correlation of TAK1 expression and clinical outcome in T3N1-3M0 surgically resected ESCCs. METHODS: Tissue microarrays constructed of 234 surgically resected T3N1-3M0 ESCC primary tumors were used for TAK1 evaluation by immunohistochemistry. The clinical and prognostic significance of TAK1 expression was analyzed statistically. RESULTS: Positive expression of TAK1 was observed in 38.5% (90 of 234). The expression level of TAK1 in ESCCs at stage N2-3 was significantly higher than the expression level in those at stage N1 (p = 0.041). Patients with negative TAK1 expression demonstrated better overall survival than those with positive expression (median, 22.7 vs 17.4 months; p = 0.023). Multivariate analysis showed that TAK1 expression was an independent prognostic factor in ESCC (relative risk, 1.429; p = 0.021). CONCLUSIONS: TAK1 expression correlates with lymph node metastasis and is a negative, independent prognostic factor in resected T3N1-3M0 ESCCs.


Assuntos
Carcinoma de Células Escamosas/genética , DNA de Neoplasias/genética , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , MAP Quinase Quinase Quinases/genética , Estadiamento de Neoplasias , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Carcinoma de Células Escamosas/enzimologia , Carcinoma de Células Escamosas/mortalidade , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/mortalidade , Esofagectomia , Esôfago/patologia , Esôfago/cirurgia , Feminino , Humanos , Imuno-Histoquímica , MAP Quinase Quinase Quinases/biossíntese , Masculino , Pessoa de Meia-Idade , Prognóstico , Estudos Retrospectivos , Análise Serial de Tecidos , Fator de Crescimento Transformador beta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA