Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J BUON ; 23(3): 604-610, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30003726

RESUMO

PURPOSE: To identify microRNAs (miRNAs) directly regulating the proto-oncogene Bmi-1 expression in the development of hepatocellular carcinoma (HCC) and to explore the underlying molecular mechanisms. METHODS: Four HCC cell lines, including HepG2, Bel7404, Huh7, and PLC5, the normal hepatocellular cell line MIHA, and 30 HCC biopsies were included in this study. Potential miRNAs, which interact with Bmi-1 and are involved in the development of HCC were identified through bioinformatic analyses. The expression of miRNA and Bmi-1 in HCC cell lines and HCC tissues was analyzed using fluorescence protein analysis, real-time quantitative PCR (RT-qPCR), and Western blotting. RESULTS: Bioinformatic analysis suggested that miR-218 is a potential miRNA regulating Bmi-1 expression. Fluorescence protein analysis, RT-qPCR, and Western blotting confirmed the direct interaction between miR-218 and Bmi-1. In addition, increased expression of Bmi-1 was detected in HCC cell lines and HCC tissues. In most HCC tissues, the expression of miR-218 was decreased and was associated with increased expression of Bmi-1. CONCLUSION: miR-p218 downregulates the expression of the proto-oncogene Bmi-1 in HCC, and it may be an effective target for the treatment of this disease.


Assuntos
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/genética , Proto-Oncogene Mas , Proto-Oncogenes , Transfecção
2.
Oncology ; 93 Suppl 1: 20-26, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29258116

RESUMO

BACKGROUND: Although the stem cell marker Bmi1 is overexpressed in many malignancies, its role in inflammation-associated cancer is unclear. Colitis-associated cancer (CAC) is caused by chronic intestinal inflammation and often results from refractory inflammatory bowel disease (IBD). METHODS: To assess the involvement of Bmi1 in the development of CAC, we analyzed the gene expression of colon tissues collected from 111 patients with IBD and CAC. RESULTS: In the colonic mucosa of patients with ulcerative colitis, the expression of Bmi1 correlated significantly with the expression of inflammatory cytokines such as IL-6, IL-17, IL-23, and tumor necrosis factor α (TNF-α). In the colonic mucosa of patients with Crohn's disease, the expression of Bmi1 correlated significantly with the expression of TNF-α and IL-23. The expression of Bmi1 was enhanced in the colonic mucosae of refractory IBD, suggesting that Bmi1 expression might be related to increased cancer risk. In addition, patients with high Bmi1 expression showed significantly lower response rates upon subsequent anti-TNF-α therapy as compared to patients with low Bmi1 expression. In human CAC specimens, the expression of Bmi1 was upregulated in nontumor tissues as well as tumors. CONCLUSIONS: Bmi1 expression is related to a refractory clinical course of IBD and upregulated in refractory IBD and CAC. Measurement of Bmi1 expression is a promising approach for the advanced treatment and personalized management of IBD patients.


Assuntos
Colite Ulcerativa/enzimologia , Doença de Crohn/enzimologia , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Adulto , Colite Ulcerativa/patologia , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Doença de Crohn/patologia , Citocinas/biossíntese , Feminino , Humanos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Masculino , Pessoa de Meia-Idade , Regulação para Cima
3.
Circulation ; 132(1): 47-58, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-25934838

RESUMO

BACKGROUND: Platelets have a pathophysiologic role in the ischemic microvascular environment of acute coronary syndromes. In comparison with platelet activation in normal healthy conditions, less attention is given to mechanisms of platelet activation in diseased states. Platelet function and mechanisms of activation in ischemic and reactive oxygen species-rich environments may not be the same as in normal healthy conditions. Extracellular regulated protein kinase 5 (ERK5) is a mitogen-activated protein kinase family member activated in hypoxic, reactive oxygen species-rich environments and in response to receptor-signaling mechanisms. Prior studies suggest a protective effect of ERK5 in endothelial and myocardial cells after ischemia. We present evidence that platelets express ERK5 and that platelet ERK5 has an adverse effect on platelet activation via selective receptor-dependent and receptor-independent reactive oxygen species-mediated mechanisms in ischemic myocardium. METHODS AND RESULTS: Using isolated human platelets and a mouse model of myocardial infarction (MI), we found that platelet ERK5 is activated post-MI and that platelet-specific ERK5(-/-) mice have less platelet activation, reduced MI size, and improved post-MI heart function. Furthermore, the expression of downstream ERK5-regulated proteins is reduced in ERK5(-/-) platelets post-MI. CONCLUSIONS: ERK5 functions as a platelet activator in ischemic conditions, and platelet ERK5 maintains the expression of some platelet proteins after MI, leading to infarct expansion. This demonstrates that platelet function in normal healthy conditions is different from platelet function in chronic ischemic and inflammatory conditions. Platelet ERK5 may be a target for acute therapeutic intervention in the thrombotic and inflammatory post-MI environment.


Assuntos
Plaquetas/enzimologia , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/patologia , Ativação Plaquetária/fisiologia , Animais , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 7 Ativada por Mitógeno/deficiência , Oxirredução
4.
Cancer Sci ; 107(4): 543-50, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26797961

RESUMO

Multistep hepatocarcinogenesis progresses from dysplastic nodules to early hepatocellular carcinoma (eHCC) and to advanced HCC. The aim of the present study was to investigate the detailed histopathological features of eHCC. We investigated 66 small vaguely nodular lesions resected from 40 patients. The degree of cellular and structural atypia and stromal invasion were assessed. The immunohistochemical expression of HCC-related markers adenylate cyclase-associated protein 2 (CAP2), heat shock protein 70 (HSP70), Bmi-1, CD34 and h-caldesmon were evaluated. Of the 66 nodules, 10 were diagnosed as low-grade dysplastic nodules (LGDN), 10 as high-grade dysplastic nodules (HGDN) and 46 as eHCC. Among the 46 eHCC, 18 nodules (39.1%) showed marked stromal invasion and/or the presence of the scirrhous component and were subclassified as high-grade eHCC (HGeHCC). The remaining 28 eHCC, which lacked these features, were subclassified as low-grade eHCC (LGeHCC) and were examined further. HGeHCC showed high levels of cellular and structural atypia and large tumor size. The immunohistochemical expression of CAP2 and the area of sinusoidal vascularization showed increases from LGDN to HGeHCC. The density of arterial tumor vessels was high in HGeHCC compared with other nodule types. Cluster analysis of these parameters subclassified 65 nodules into HGeHCC-dominant, LGeHCC and HGDN-dominant, and LGDN-dominant groups. These results indicate the increased malignant potential of HGeHCC and suggest that it is already a transitional stage to advanced HCC. We consider that our grading classification system may be valuable for considering treatment strategies for eHCC around 2 cm in diameter.


Assuntos
Carcinogênese/genética , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Proteínas de Neoplasias/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Idoso , Antígenos CD34/biossíntese , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Proteínas de Ligação a Calmodulina/biossíntese , Carcinoma Hepatocelular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico HSP70/biossíntese , Humanos , Neoplasias Hepáticas/genética , Masculino , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Estadiamento de Neoplasias
5.
J Recept Signal Transduct Res ; 36(5): 435-44, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27305962

RESUMO

Aldosterone acts on its target tissue through a classical mechanism or through the rapid pathway through a putative membrane-bound receptor. Our goal here was to better understand the molecular and biochemical rapid mechanisms responsible for aldosterone-induced cardiomyocyte hypertrophy. We have evaluated the hypertrophic process through the levels of ANP, which was confirmed by the analysis of the superficial area of cardiomyocytes. Aldosterone increased the levels of ANP and the cellular area of the cardiomyocytes; spironolactone reduced the aldosterone-increased ANP level and cellular area of cardiomyocytes. Aldosterone or spironolactone alone did not increase the level of cyclic 3',5'-adenosine monophosphate (cAMP), but aldosterone plus spironolactone led to increased cAMP level; the treatment with aldosterone + spironolactone + BAPTA-AM reduced the levels of cAMP. These data suggest that aldosterone-induced cAMP increase is independent of mineralocorticoid receptor (MR) and dependent on Ca(2+). Next, we have evaluated the role of A-kinase anchor proteins (AKAP) in the aldosterone-induced hypertrophic response. We have found that St-Ht31 (AKAP inhibitor) reduced the increased level of ANP which was induced by aldosterone; in addition, we have found an increase on protein kinase C (PKC) and extracellular signal-regulated kinase 5 (ERK5) activity when cells were treated with aldosterone alone, spironolactone alone and with a combination of both. Our data suggest that PKC could be responsible for ERK5 aldosterone-induced phosphorylation. Our study suggests that the aldosterone through its rapid effects promotes a hypertrophic response in cardiomyocytes that is controlled by an AKAP, being dependent on ERK5 and PKC, but not on cAMP/cAMP-dependent protein kinase signaling pathways. Lastly, we provide evidence that the targeting of AKAPs could be relevant in patients with aldosterone-induced cardiac hypertrophy and heart failure.


Assuntos
Proteínas de Ancoragem à Quinase A/metabolismo , Aldosterona/administração & dosagem , Insuficiência Cardíaca/tratamento farmacológico , Hipertrofia/tratamento farmacológico , Receptores de Mineralocorticoides/biossíntese , Proteínas de Ancoragem à Quinase A/genética , Animais , Fator Natriurético Atrial/biossíntese , Fator Natriurético Atrial/metabolismo , AMP Cíclico/metabolismo , Ácido Egtázico/administração & dosagem , Ácido Egtázico/análogos & derivados , Insuficiência Cardíaca/metabolismo , Humanos , Hipertrofia/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosforilação , Cultura Primária de Células , Proteína Quinase C/biossíntese , Ratos , Receptores de Mineralocorticoides/genética , Transdução de Sinais/efeitos dos fármacos , Espironolactona/administração & dosagem
6.
Tumour Biol ; 37(4): 5049-62, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26546432

RESUMO

Even though early detection methods and treatment options are greatly improved, chemoresistance is still a tremendous challenge for breast cancer therapy. Breast cancer stem cells (BCSCs) represent a subpopulation that is central to chemoresistance. We aim to investigate the relationship between SLC34A2 and chemoresistance in BCSCs and identify the underlying mechanisms by which SLC34A2 regulates chemoresistance in BCSCs. Fluorescence Activated Cell Sorting (FACS) analysis showed the presence of a variable fraction of CD44(+)CD24(-) cells in 25 out of 25 breast cancer samples. We cultured primary breast cancer sample cells and breast cancer cell line cells to induce sphere formation in serum-free medium. Following sorting of CD44(+)CD24(-) cells from spheres, we showed that CD44(+)CD24(-) cells displayed stem cell-like features and were resistant to chemotherapy drug doxorubicin. Significantly, enhanced SLC34A2 expression correlated with chemoresponse and survival of breast cancer patients. We subsequently indicated that increased SLC34A2 expression in BCSCs directly contributed to their chemoresistance by a series of in vitro and in vivo experiments. Furthermore, we demonstrated that SLC34A2 induced chemoresistance in BCSCs via SLC34A2-Bmi1-ABCC5 signaling. Finally, we showed that ABCC5 was a direct transcriptional target of Bmi1 by chromatin immunoprecipitation (ChIP). In conclusion, our work indicated that decreased SLC34A2 expression sensitized BCSCs to doxorubicin via SLC34A2-Bmi1-ABCC5 signaling and shed new light on understanding the mechanism of chemoresistance in BCSCs. This study not only bridges the missing link between stem cell-related transcription factor (Bmi1) and ABC transporter (ABCC5) but also contributes to development of potential therapeutics against breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteínas Associadas à Resistência a Múltiplos Medicamentos/biossíntese , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/biossíntese , Adulto , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Doxorrubicina/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Pessoa de Meia-Idade , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteínas Associadas à Resistência a Múltiplos Medicamentos/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mol Cancer ; 14: 159, 2015 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-26286140

RESUMO

BACKGROUND: The Sonic hedgehog (Shh) signaling pathway plays an important role in cerebellar development, and mutations leading to hyperactive Shh signaling have been associated with certain forms of medulloblastoma, a common form of pediatric brain cancer. While the fundamentals of this pathway are known, the molecular targets contributing to Shh-mediated proliferation and transformation are still poorly understood. Na,K-ATPase is a ubiquitous enzyme that maintains intracellular ion homeostasis and functions as a signaling scaffold and a cell adhesion molecule. Changes in Na,K-ATPase function and subunit expression have been reported in several cancers and loss of the ß1-subunit has been associated with a poorly differentiated phenotype in carcinoma but its role in medulloblastoma progression is not known. METHODS: Human medulloblastoma cell lines and primary cultures of cerebellar granule cell precursors (CGP) were used to determine whether Shh regulates Na,K-ATPase expression. Smo/Smo medulloblastoma were used to assess the Na,K-ATPase levels in vivo. Na,K-ATPase ß1-subunit was knocked down in DAOY cells to test its role in medulloblastoma cell proliferation and tumorigenicity. RESULTS: Na,K-ATPase ß1-subunit levels increased with differentiation in normal CGP cells. Activation of Shh signaling resulted in reduced ß1-subunit mRNA and protein levels and was mimicked by overexpression of Gli1and Bmi1, both members of the Shh signaling cascade; overexpression of Bmi1 reduced ß1-subunit promoter activity. In human medulloblastoma cells, low ß1-subunit levels were associated with increased cell proliferation and in vivo tumorigenesis. CONCLUSIONS: Na,K-ATPase ß1-subunit is a target of the Shh signaling pathway and loss of ß1-subunit expression may contribute to tumor development and progression not only in carcinoma but also in medulloblastoma, a tumor of neuronal origin.


Assuntos
Carcinogênese/genética , Proteínas Hedgehog/genética , Meduloblastoma/genética , ATPase Trocadora de Sódio-Potássio/biossíntese , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Proteínas Hedgehog/antagonistas & inibidores , Humanos , Meduloblastoma/patologia , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , ATPase Trocadora de Sódio-Potássio/genética , Fatores de Transcrição/biossíntese , Proteína GLI1 em Dedos de Zinco
8.
Am J Physiol Cell Physiol ; 306(11): C1089-100, 2014 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-24740537

RESUMO

Platelet-derived growth factor BB and its receptor (PDGFRß) play a pivotal role in the development of renal glomerular mesangial cells. Their roles in increased mesangial cell proliferation during mesangioproliferative glomerulonephritis have long been noted, but the operating logic of signaling mechanisms regulating these changes remains poorly understood. We examined the role of a recently identified MAPK, Erk5, in this process. PDGF increased the activating phosphorylation of Erk5 and tyrosine phosphorylation of proteins in a time-dependent manner. A pharmacologic inhibitor of Erk5, XMD8-92, abrogated PDGF-induced DNA synthesis and mesangial cell proliferation. Similarly, expression of dominant negative Erk5 or siRNAs against Erk5 blocked PDGF-stimulated DNA synthesis and proliferation. Inhibition of Erk5 attenuated expression of cyclin D1 mRNA and protein, resulting in suppression of CDK4-mediated phosphorylation of the tumor suppressor protein pRb. Expression of cyclin D1 or CDK4 prevented the dominant negative Erk5- or siErk5-mediated inhibition of DNA synthesis and mesangial cell proliferation induced by PDGF. We have previously shown that phosphatidylinositol 3-kinase (PI3-kinase) contributes to PDGF-induced proliferation of mesangial cells. Inhibition of PI3-kinase blocked PDGF-induced phosphorylation of Erk5. Since PI3-kinase acts through Akt, we determined the role of Erk5 on Akt phosphorylation. XMD8-92, dominant negative Erk5, and siErk5 inhibited phosphorylation of Akt by PDGF. Interestingly, we found inhibition of PDGF-induced Erk5 phosphorylation by a pharmacological inhibitor of Akt kinase and kinase dead Akt in mesangial cells. Thus our data unfold the presence of a positive feedback microcircuit between Erk5 and Akt downstream of PI3-kinase nodal point for PDGF-induced mesangial cell proliferation.


Assuntos
Proliferação de Células , Retroalimentação Fisiológica/fisiologia , Células Mesangiais/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Oncogênica v-akt/biossíntese , Fator de Crescimento Derivado de Plaquetas/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Retroalimentação Fisiológica/efeitos dos fármacos , Células Mesangiais/efeitos dos fármacos , Ratos
9.
J Cell Physiol ; 229(7): 856-67, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24264602

RESUMO

Mitogen-activated protein kinases (MAPKs) are important transducers of external signals for cell growth, survival, and other cellular responses including cell differentiation. Several MAPK cascades are known with the MEK1/2-ERK1/2, JNK, and p38MAPKs receiving most attention, but the role of MEK5-ERK5 in intracellular signaling deserves more scrutiny, as this pathway transmits signals that can complement ERK/2 signaling. We hypothesized that the ERK5 pathway plays a role in the control of monocytic differentiation, which is disturbed in myeloid leukemia. We therefore examined the cellular phenotype and key molecular events which occur when human myeloid leukemia cells, acute (AML) or chronic (CML), are forced to differentiate by vitamin D derivatives (VDDs). This study was performed using established cell lines HL60 and U937, and primary cultures of blasts from 10 patients with ML. We found that ERK5 and its direct downstream target transcription factor MEF2C are upregulated by 1,25D in parallel with monocytic differentiation. Further, inhibition of ERK5 activity by specific pharmacological agents BIX02189 and XMD8-92 alters the phenotype of these cells by reducing the abundance of the VDD-induced surface monocytic marker CD14, and concomitantly increasing surface expression of the general myeloid marker CD11b. Similar results were obtained when the expression of ERK5 was reduced by siRNA or short hairpin (sh) RNA. ERK5 inhibition resulted in an expected decrease in MEF2C activation. We also found that in AML cells the transcription factor C/EBPß is positively regulated, while C/EBPα is negatively regulated by ERK5. These findings provide new understanding of dysregulated differentiation in human myeloid leukemia.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Leucemia Mieloide/genética , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Monócitos/metabolismo , Compostos de Anilina/administração & dosagem , Benzodiazepinonas/administração & dosagem , Proteína beta Intensificadora de Ligação a CCAAT , Proteínas Estimuladoras de Ligação a CCAAT/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HL-60 , Humanos , Indóis/administração & dosagem , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/patologia , Fatores de Transcrição MEF2/biossíntese , Fatores de Transcrição MEF2/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/genética , Monócitos/citologia , Monócitos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células U937 , Vitamina D/administração & dosagem , Vitamina D/análogos & derivados
10.
Biochem Biophys Res Commun ; 449(4): 466-70, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24857985

RESUMO

Extracellular-signal-regulated kinase 5 (ERK5) is a member of the mitogen-activated protein kinase (MAPK) family and regulates a wide variety of cellular processes such as proliferation, differentiation, necrosis, apoptosis and degeneration. However, the expression of ERK5 and its role in degenerated human nucleus pulposus (NP) is hitherto unknown. In this study, we observed the differential expression of ERK5 in normal and degenerated human nucleus pulposus tissues by using immunohistochemical staining and Western blot. Treatment of NP cells with Pro-inflammatory cytokine, TNF-α decreased ERK5 gene expression as well as NP marker gene expression; including the type II collagen and aggrecan. Suppression of ERK5 gene expression in NP cells by ERK5 siRNA resulted in decreased gene expression of type II collagen and aggrecan. Furthermore, inhibition of ERK5 activation by BIX02188 (5µM) decreased the gene expression of type II collagen and aggrecan in NP cells. Our results document the expression of ERK5 in degenerated nucleus pulposus tissues, and suggest a potential involvement of ERK5 in human degenerated nucleus pulposus.


Assuntos
Degeneração do Disco Intervertebral/fisiopatologia , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Adolescente , Agrecanas/biossíntese , Compostos de Anilina/farmacologia , Células Cultivadas , Colágeno Tipo II/biossíntese , Humanos , Indóis/farmacologia , Disco Intervertebral/citologia , Disco Intervertebral/efeitos dos fármacos , Disco Intervertebral/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia
11.
Biochem Biophys Res Commun ; 421(3): 490-3, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22521884

RESUMO

We examined the effects of cadmium chloride (CdCl(2)) exposure on the phosphorylation and functionality of extracellular signal-regulated kinase 5 (ERK5), a recently identified member of the mitogen-activated protein kinase (MAPK) family, in HK-2 human renal proximal tubular cells. Following exposure to CdCl(2), ERK5 phosphorylation increased markedly, but the level of total ERK5 was unchanged. ERK5 phosphorylation following CdCl(2) exposure was rapid and transient, similar to the time course of ERK1/2 phosphorylation. Treatment of HK-2 cells with the MAPK/ERK kinase 5 inhibitor, BIX02189, suppressed CdCl(2)-induced ERK5 but not ERK1/2 phosphorylation. The CdCl(2)-induced increase of phosphorylated cAMP response element-binding protein (CREB) and activating transcription factor-1 (ATF-1), as well as the accumulation of mobility-shifted c-Fos protein, were suppressed by BIX02189 treatment. Furthermore, BIX02189 treatment enhanced cleavage of poly(ADP-ribose) polymerase and increased the level of cytoplasmic nucleosomes in HK-2 cells exposed to CdCl(2). These findings suggest that ERK5 pathway activation by CdCl(2) exposure might induce the phosphorylation of cell survival-transcription factors, such as CREB, ATF-1, and c-Fos, and may exert a partial anti-apoptotic role in HK-2 cells.


Assuntos
Poluentes Atmosféricos/toxicidade , Cádmio/toxicidade , Túbulos Renais Proximais/efeitos dos fármacos , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Compostos de Anilina/farmacologia , Cloreto de Cádmio/toxicidade , Linhagem Celular , Citoplasma/enzimologia , Humanos , Indóis/farmacologia , Túbulos Renais Proximais/enzimologia , Sistema de Sinalização das MAP Quinases , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Nucleossomos/enzimologia , Fosforilação , Poli(ADP-Ribose) Polimerases/metabolismo , Inibidores de Proteínas Quinases/farmacologia
12.
J Immunol ; 182(6): 3398-405, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265117

RESUMO

Tumor cell-based vaccines are currently used in clinical trails, but they are in general poorly immunogenic because they are composed of cell extracts or apoptotic cells. Live tumor cells should be much better Ags provided that they are properly processed by the host immune system. We show herein that stable expression of a small hairpin RNA for ERK5 (shERK5) decreases ERK5 levels in human and mouse leukemic cells and leads to their elimination by NK cells in vivo. The shERK5 cells show down-regulation of MHC class I expression at the plasma membrane. Accordingly, ectopic activation of the ERK5 pathway induces MHC class I gene expression. Coinjection of shERK5-expressing cells into the peritoneum diminishes survival of engrafted wild-type tumor cells. Moreover, s.c. injection of shERK5-expressing cells strongly diminishes tumor development by wild-type cells. Our results show that shERK5 expression in leukemia cells effectively attenuates their tumor activity and allows their use as a tumor cell-based vaccine.


Assuntos
Vacinas Anticâncer/imunologia , Técnicas de Silenciamento de Genes , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Leucemia L1210/prevenção & controle , Ativação Linfocitária/imunologia , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/genética , Animais , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Células Cultivadas , Citotoxicidade Imunológica/genética , Antígenos de Histocompatibilidade Classe I/biossíntese , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Jurkat , Células Matadoras Naturais/metabolismo , Leucemia L1210/enzimologia , Leucemia L1210/genética , Leucemia L1210/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , RNA Interferente Pequeno/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
13.
Oncogene ; 40(23): 3929-3941, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33981002

RESUMO

There is overwhelming clinical evidence that the extracellular-regulated protein kinase 5 (ERK5) is significantly dysregulated in human breast cancer. However, there is no definite understanding of the requirement of ERK5 in tumor growth and metastasis due to very limited characterization of the pathway in disease models. In this study, we report that a high level of ERK5 is a predictive marker of metastatic breast cancer. Mechanistically, our in vitro data revealed that ERK5 was critical for maintaining the invasive capability of triple-negative breast cancer (TNBC) cells through focal adhesion protein kinase (FAK) activation. Specifically, we found that phosphorylation of FAK at Tyr397 was controlled by a kinase-independent function of ERK5. Accordingly, silencing ERK5 in mammary tumor grafts impaired FAK phosphorylation at Tyr397 and suppressed TNBC cell metastasis to the lung without preventing tumor growth. Collectively, these results establish a functional relationship between ERK5 and FAK signaling in promoting malignancy. Thus, targeting the oncogenic ERK5-FAK axis represents a promising therapeutic strategy for breast cancer exhibiting aggressive clinical behavior.


Assuntos
Quinase 1 de Adesão Focal/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Neoplasias de Mama Triplo Negativas/enzimologia , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Antígenos CD/metabolismo , Caderinas/biossíntese , Caderinas/genética , Caderinas/metabolismo , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Progressão da Doença , Feminino , Xenoenxertos , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Nus , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/genética , Invasividade Neoplásica , Fosforilação , Prognóstico , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
14.
Oncogene ; 39(39): 6245-6262, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32843720

RESUMO

Triple negative breast cancer (TNBC) refers to tumors that do not express clinically significant levels of estrogen and progesterone receptors, and lack membrane overexpression or gene amplification of ErbB-2/HER2, a receptor tyrosine kinase. Transcriptome and proteome heterogeneity of TNBC poses a major challenge to precision medicine. Clinical biomarkers and targeted therapies for this disease remain elusive, so chemotherapy has been the standard of care for early and metastatic TNBC. Our present findings placed ErbB-2 in an unanticipated scenario: the nucleus of TNBC (NErbB-2). Our study on ErbB-2 alternative splicing events, using a PCR-sequencing approach combined with an RNA interference strategy, revealed that TNBC cells express either the canonical (wild-type) ErbB-2, encoded by transcript variant 1, or the non-canonical ErbB-2 isoform c, encoded by alternative variant 3 (RefSeq), or both. These ErbB-2 isoforms function in the nucleus as transcription factors. Evicting both from the nucleus or silencing isoform c only, blocks TN cell and tumor growth. This reveals not only NErbB-2 canonical and alternative isoforms role as targets of therapy in TNBC, but also isoform c dominant oncogenic potential. Furthermore, we validated our findings in the clinic and observed that NErbB-2 correlates with poor prognosis in primary TN tumors, disclosing NErbB-2 as a novel biomarker for TNBC. Our discoveries challenge the present scenario of drug development for personalized BC medicine that focuses on wild-type RefSeq proteins, which conserve the canonical domains and are located in their classical cellular compartments.


Assuntos
Receptor ErbB-2/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/enzimologia , Núcleo Celular/metabolismo , Proliferação de Células/fisiologia , Feminino , Humanos , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/genética , Inclusão em Parafina , Isoformas de Proteínas , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/biossíntese , Receptor ErbB-2/genética , Neoplasias de Mama Triplo Negativas/enzimologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/patologia
15.
Mol Biol Cell ; 16(2): 742-56, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15548597

RESUMO

BimC kinesins are required for mitotic spindle assembly in a variety of organisms. These proteins are localized to centrosomes, spindle microtubules, and the spindle midzone. We have previously shown that the Caenorhabditis elegans Aurora B kinase AIR-2 is required for the localization of the ZEN-4 kinesin protein to midzone microtubules. To determine whether the association of BimC kinesins with spindle microtubules is also dependent on AIR-2, we examined the expression pattern of BMK-1, a C. elegans BimC kinesin, in wild-type and AIR-2-deficient embryos. BMK-1 is highly expressed in the hermaphrodite gonad and is localized to meiotic spindle microtubules in the newly fertilized embryo. In mitotic embryos, BMK-1 is associated with spindle microtubules from prophase through anaphase and is concentrated at the spindle midzone during anaphase and telophase. In the absence of AIR-2, BMK-1 localization to meiotic and mitotic spindles is greatly reduced. This is not a consequence of loss of ZEN-4 localization because BMK-1 is appropriately localized in ZEN-4-deficient embryos. Furthermore, AIR-2 and BMK-1 directly interact with one another and the C-terminal tail domain of BMK-1 is specifically phosphorylated by AIR-2 in vitro. Together with our previous data, these results suggest that at least one function of the Aurora B kinases is to recruit spindle-associated motor proteins to their sites of action.


Assuntos
Proteínas de Caenorhabditis elegans/fisiologia , Caenorhabditis elegans/metabolismo , Proteínas de Helminto/metabolismo , Cinesinas/fisiologia , Mitose , Proteínas Serina-Treonina Quinases/fisiologia , Fuso Acromático , Sequência de Aminoácidos , Animais , Aurora Quinase B , Aurora Quinases , Western Blotting , Caenorhabditis elegans/enzimologia , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Sequência Conservada , Embrião não Mamífero , Glutationa Transferase/metabolismo , Imuno-Histoquímica , Cinesinas/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/química , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Fosforilação , Mutação Puntual , Testes de Precipitina , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína , Interferência de RNA , Proteínas Recombinantes/biossíntese , Homologia de Sequência de Aminoácidos , Temperatura , Treonina/química , Técnicas do Sistema de Duplo-Híbrido
16.
Cancer Res ; 65(13): 5683-9, 2005 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15994942

RESUMO

Expression of the neurotrophin-3 receptor, tyrosine kinase C (TrkC), is associated with favorable prognosis in medulloblastoma patients. This may be due to increased tumor apoptosis induced by TrkC activation. Neurotrophin-3/TrkC-induced apoptosis is inhibited by the mitogen-activated protein (MAP) kinase (MAPK) pharmacologic antagonists SB203580 and PD98059. In addition to extracellular signal-regulated kinase (ERK)-1/2, PD98059 also inhibits the more recently identified neurotrophin-responsive MAPK, ERK5 (big MAPK 1). In the present study, we investigate the contribution of ERK5 and its target myocyte enhancer factor 2 (MEF2) to neurotrophin-3/TrkC-induced medulloblastoma cell death. Neurotrophin-3 not only enhanced ERK5 phosphorylation but also significantly enhanced the transcriptional activity of MEF2, a specific target of ERK5. Overexpression of both ERK5 and MEF2 induced a statistically significant increase in cell death of neurotrophin-3-responsive and nonresponsive medulloblastoma cell lines (Daoy-trkC and Daoy) and primary cultures of patched heterozygous mouse medulloblastomas. Only those cells expressing MAP/ERK kinase 5 (MEK5) plus ERK5 or MEF2 constructs underwent apoptosis, indicating that overexpression of either is sufficient to induce medulloblastoma cell death. Expression of a dominant-negative MEF2 or small interfering RNA for the ERK5 activator, MEK5, significantly inhibited neurotrophin-3-induced cell death. The dominant-negative MEF2 construct also blocked MEK5/ERK5-induced cell death, supporting a role for MEF2 downstream of ERK5. Co-immunoprecipitation studies revealed direct interaction of phosphorylated ERK5 with MEF2 in response to neurotrophin-3. Our investigation of the mechanism of neurotrophin-3/TrkC-induced apoptosis has identified a novel role for both MEK5/ERK5 and MEF2 in cell death, suggesting that these molecules can be exploited to induce apoptosis in both TrkC-expressing and non-expressing medulloblastoma cells.


Assuntos
Neoplasias Cerebelares/patologia , Proteínas de Ligação a DNA/fisiologia , Meduloblastoma/patologia , Proteína Quinase 7 Ativada por Mitógeno/fisiologia , Fatores de Transcrição/fisiologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Tumoral , Neoplasias Cerebelares/enzimologia , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/metabolismo , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Humanos , Imidazóis/farmacologia , Fatores de Transcrição MEF2 , Meduloblastoma/enzimologia , Meduloblastoma/genética , Meduloblastoma/metabolismo , Camundongos , Proteína Quinase 7 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/genética , Fatores de Regulação Miogênica , Neurotrofina 3/farmacologia , Fosforilação , Piridinas/farmacologia , Receptor trkC/biossíntese , Receptor trkC/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
17.
Tumori ; 103(5): 483-488, 2017 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-26350187

RESUMO

PURPOSE: Osteosarcoma (OS) is the most common primary bone tumor and has low cure rates. Our study aimed to evaluate the roles of mitogen-activated protein kinase 7 (MAPK7) in cell proliferation, migration and invasion using the SOSP-M human OS cell line as an in vitro model. METHODS: SOSP-M cells were transfected with PCDNA3.1-MAPK7 and siRNA-MAPK7 plasmids using Lipofectamine 2000. Quantitative real-time polymerase chain reaction (RT-PCR) was performed to determine the relative expression level of MAPK7 and Western blot analysis was carried out to determine the expression level of ERK5 protein. Then MTT, scratch wound healing and Matrigel transwell assays were used to investigate the roles of MAPK7 expression in the proliferation, migration and invasion, respectively, of SOSP-M cells in vitro. RESULTS: RT-PCR analysis showed that the expression level of MAPK7 increased significantly after transfection with PCDNA3.1-MAPK7 plasmid compared with the blank group, while it decreased significantly after transfection with siRNA-MAPK7 plasmid. Similar results for ERK5 expression were obtained by Western blot analysis. In addition, the cell proliferation rate, cell migration rate and invasive cell number in the PCDNA3.1-MAPK7 transfection group increased significantly compared with the blank group, while they decreased significantly in the siRNA-MAPK7 transfection group. CONCLUSIONS: Our results indicate that overexpression of MAPK7 in human OS cells could promote cell proliferation, migration and invasion, whereas knockdown of MAPK7 expression had the opposite effect. All the results suggest that MAPK7 may serve as a potent target for drug development.


Assuntos
Movimento Celular/genética , Proliferação de Células/genética , Proteína Quinase 7 Ativada por Mitógeno/genética , Osteossarcoma/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Osteossarcoma/patologia , RNA Interferente Pequeno/genética
18.
Biotechnol Prog ; 33(4): 1046-1058, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28371547

RESUMO

In recent years, the number of complex but clinically effective biologicals such as multi-specific antibody formats and fusion proteins has increased dramatically. However, compared to classical monoclonal antibodies (mAbs), these rather artificially designed therapeutic proteins have never undergone millions of years of evolution and thus often turn out to be difficult-to-express using mammalian expression systems such as Chinese hamster ovary (CHO) cells. To provide access to these sophisticated but effective drugs, host cell engineering of CHO production cell lines represents a promising approach to overcome low production yields. MicroRNAs (miRNAs) have recently gained much attention as next-generation cell engineering tools. However, only very little is known about the capability of miRNAs to specifically increase production of difficult-to-express proteins. In a previous study we identified miR-143 amongst others to improve protein production in CHO cells. Thus, the aim of the present study was to examine if miR-143 might be suitable to improve production of low yield protein candidates. Both transient and stable overexpression of miR-143 significantly improved protein production without negatively affecting cell growth and viability of different recombinant CHO cells. In addition, mitogen-activated protein kinase 7 (MAPK7) was identified as a putative target gene of miR-143-3p in CHO cells. Finally, siRNA-mediated knock-down of MAPK7 could be demonstrated to phenocopy pro-productive effects of miR-143. In summary, our data suggest that miR-143 might represent a novel genetic element to enhance production of difficult-to-express proteins in CHO cells which may be partly mediated by down-regulation of MAPK7. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1046-1058, 2017.


Assuntos
MicroRNAs/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/genética , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Animais , Células CHO , Proliferação de Células , Sobrevivência Celular , Cricetulus , Humanos , MicroRNAs/genética , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Fenótipo
19.
Life Sci ; 79(19): 1839-46, 2006 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-16859717

RESUMO

Accumulating evidence implicates activation (phosphorylation) of mitogen-activated protein kinases (MAPK) during nonlethal ischemic preconditioning in the protection of hippocampal CA1 neuron against subsequent ischemic events. In this paper, we undertook to identify the role of extracellular signal regulated kinase (ERK) 5 in cerebral ischemic preconditioning (CIP). Three minutes of ischemia was induced as preconditioning stimulus. Three days later, 6 min of ischemia was induced. The levels of ERK5 protein expression and its activation were detected with or without the CIP in hippocampal CA1 and the dentate gyrus (DG) regions. Our results showed that ERK5 was activated selectively in hippocampal CA1 region with, but not without, the ischemic preconditioning. Notably, during the later phase of reperfusion, the rise in ERK5 activation was strong and persistent with a peak occurring at the third day. The activation peak was effectively prevented and ERK5 protein expression was significantly decreased by intracerebroventricular infusion of ERK5 antisense oligonucleotide (every 24 h for 3 days before the preconditioning), but not by sense oligonucleotide or vehicle. Subsequently, the CA1 neuronal loss was largely elevated. Moreover, both MK801 (10 microM), an antagonist of NMDA receptor, and EGTA (100 mM, but neither 50 nor 150 mM), an extracellular Ca2+ chelator, not only effectively inhibited the ERK5 activation but also markedly abolished CIP-induced survival of the CA1 neurons. These results suggested that activation of the ERK5 pathway by CIP was at least partly dependent on moderate Ca2+ influx via NMDA receptor, which might contribute to ischemic tolerance in hippocampal CA1 region of rats.


Assuntos
Isquemia Encefálica , Cálcio/metabolismo , Hipocampo , Precondicionamento Isquêmico , Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Traumatismo por Reperfusão , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Giro Denteado/irrigação sanguínea , Giro Denteado/enzimologia , Giro Denteado/metabolismo , Hipocampo/irrigação sanguínea , Hipocampo/enzimologia , Hipocampo/metabolismo , Hipocampo/patologia , Masculino , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Oligonucleotídeos Antissenso/farmacologia , Fosforilação , Células Piramidais/enzimologia , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/antagonistas & inibidores , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/prevenção & controle
20.
Oncol Rep ; 34(2): 771-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26043676

RESUMO

Epithelial-mesenchymal transition (EMT) is a crucial event required for the invasion and progression of carcinogenesis, inducing stem-like properties in epithelial cells. In the present study, the expression of BMI1, which controls self-renewal in stem cells, as well as that of ZEB1, a transcription factor that regulates EMT, was evaluated for its role in EMT and the carcinogenic processes of tongue squamous cell carcinoma (TSCC). Collagen invasion assays using two TSCC cells and 64 tongue specimens (32 carcinomas and 32 dysplasias) were employed and analyzed in the present study. We assessed the protein and mRNA expression levels of BMI1, ZEB1, vimentin and E-cadherin in the two cell lines and tumor tissues. The protein and mRNA expression of BMI1 and ZEB1 occurred at the invasion of TSCC. The elevated levels of BMI1 and ZEB1 were accompanied by the downregulation of E-cadherin and upregulation of vimentin at the invasive front, indicative of EMT in vitro and in vivo. The results showed that BMI1 and ZEB1 are important factors in association with the promotion of EMT and invasion of TSCC.


Assuntos
Carcinoma de Células Escamosas/genética , Proteínas de Homeodomínio/biossíntese , Proteína Quinase 7 Ativada por Mitógeno/biossíntese , Neoplasias da Língua/genética , Fatores de Transcrição/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Caderinas/biossíntese , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Humanos , Masculino , Pessoa de Meia-Idade , Proteína Quinase 7 Ativada por Mitógeno/genética , Neoplasias da Língua/patologia , Fatores de Transcrição/genética , Vimentina/biossíntese , Homeobox 1 de Ligação a E-box em Dedo de Zinco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA