Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Sci Rep ; 13(1): 7339, 2023 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-37147361

RESUMO

Renal cancer cells constitute a paradigm of tumor cells with a glycolytic reprogramming which drives metabolic alterations favouring cell survival and transformation. We studied the expression and activity of pyruvate dehydrogenase kinases (PDK1-4), key enzymes of the energy metabolism, in renal cancer cells. We analysed the expression, subcellular distribution and clinicopathological correlations of PDK1-4 by immunohistochemistry of tumor tissue microarray samples from a cohort of 96 clear cell renal cell carcinoma (ccRCC) patients. Gene expression analysis was performed on whole tumor tissue sections of a subset of ccRCC samples. PDK2 and PDK3 protein expression in tumor cells correlated with lower patient overall survival, whereas PDK1 protein expression correlated with higher patient survival. Gene expression analysis revealed molecular association of PDK2 and PDK3 expression with PI3K signalling pathway, as well as with T cell infiltration and exhausted CD8 T cells. Inhibition of PDK by dichloroacetate in human renal cancer cell lines resulted in lower cell viability, which was accompanied by an increase in pAKT. Together, our findings suggest a differential role for PDK enzymes in ccRCC progression, and highlight PDK as actionable metabolic proteins in relation with PI3K signalling and exhausted CD8 T cells in ccRCC.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Humanos , Piruvato Desidrogenase Quinase de Transferência de Acetil , Proteínas Serina-Treonina Quinases/genética , Carcinoma de Células Renais/genética , Neoplasias Renais/genética , Prognóstico , Oxirredutases , Piruvatos , Fosfatidilinositol 3-Quinases
3.
Sci Rep ; 9(1): 5839, 2019 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-30967582

RESUMO

Immunoregulatory protein B7-H3 is involved in the oncogenic and metastatic potential of cancer cells, as well as in drug resistance. Resistance to conventional chemotherapy is an important aspect of melanoma treatment, and a better understanding of how B7-H3 enhances drug resistance may lead to the development of more effective therapies. We investigated the in vitro and in vivo sensitivity of chemotherapeutic agents dacarbazine (DTIC) and cisplatin in sensitive and drug resistant melanoma cells with knockdown expression of B7-H3. We found that knockdown of B7-H3 increased in vitro and in vivo sensitivity of melanoma cells to the chemotherapeutic agents dacarbazine (DTIC) and cisplatin, in parallel with a decrease in p38 MAPK phosphorylation. Importantly, in B7-H3 knockdown cells we observed an increase in the expression of dual-specific MAP kinase phosphatase (MKP) DUSP10, a MKP known to dephosphorylate and inactivate p38 MAPK. DUSP10 knockdown by siRNA resulted in a reversion of the increased DTIC-sensitivity seen in B7-H3 knockdown cells. Our findings highlight the potential therapeutic benefit of combining chemotherapy with B7-H3 inhibition, and indicate that B7-H3 mediated chemoresistance in melanoma cells is driven through a mechanism involving DUSP10-mediated inactivation of p38 MAPK.


Assuntos
Antígenos B7/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Fosfatases de Especificidade Dupla/metabolismo , Fosfatases da Proteína Quinase Ativada por Mitógeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Antineoplásicos/farmacologia , Antígenos B7/genética , Linhagem Celular Tumoral , Cisplatino/farmacologia , Dacarbazina/farmacologia , Fosfatases de Especificidade Dupla/genética , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos , Fosfatases da Proteína Quinase Ativada por Mitógeno/genética , Fosforilação/efeitos dos fármacos
4.
Oncotarget ; 7(6): 6891-901, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26771843

RESUMO

B7 family proteins are important immune response regulators, and can mediate oncogenic signaling and cancer development. We have used human triple-negative breast cancer cell lines with different expression levels of B7-H3 to evaluate its effects on the sensitivity to 22 different anticancer compounds in a drug screen. API-2 (triciribidine) and everolimus (RAD-001), two inhibitors that target the PI3K/AKT/mTOR pathway, showed enhanced inhibition of cell viability and proliferation in B7-H3 knockdown tumor cells compared to their B7-H3 expressing counterparts. Similar inhibition was seen in control cells treated with an anti-B7-H3 monoclonal antibody. In B7-H3 overexpressing cells, the effects of the two drugs were reduced, supported also by in vivo experiments in which B7-H3 overexpressing xenografts were less sensitive to everolimus than control tumors. In API-2 and everolimus-treated B7-H3 overexpressing cells, phospho-mTOR levels were decreased. However, phosphorylation of p70S6K was differentially regulated in B7-H3 cells treated with API-2 or everolimus, suggesting a different B7-H3-mediated mechanism downstream of mTOR. Both API-2 and everolimus decreased the glycolysis of the cells, whereas knockdown of B7-H3 decreased and B7-H3 overexpression increased the glycolytic capacity. In conclusion, we have unveiled a previously unknown relationship between B7-H3 expression and glycolytic capacity in tumor cells, and found that B7-H3 confers resistance to API-2 and everolimus. The results provide novel insights into the function of B7-H3 in cancer, and suggest that targeting of B7-H3 may be a novel alternative to improve current anticancer therapies.


Assuntos
Antígenos B7/biossíntese , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Animais , Antígenos B7/antagonistas & inibidores , Antígenos B7/metabolismo , Linhagem Celular Tumoral , Feminino , Glicólise , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias de Mama Triplo Negativas/genética
5.
Clin Exp Metastasis ; 32(8): 755-67, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26349943

RESUMO

S100A4 promotes metastasis in several types of cancer, but the involved molecular mechanisms are still incompletely described. The protein is associated with a wide variety of biological functions and it locates to different subcellular compartments, including nuclei, cytoplasm and extracellular space. Nuclear expression of S100A4 has been associated with more advanced disease stage as well as poor outcome in colorectal cancer (CRC). The present study was initiated to investigate the nuclear function of S100A4 and thereby unravel potential biological mechanisms linking nuclear expression to a more aggressive phenotype. CRC cell lines show heterogeneity in nuclear S100A4 expression and preliminary experiments revealed cells in G2/M to have increased nuclear accumulation compared to G1 and S cells, respectively. Synchronization experiments validated nuclear S100A4 expression to be most prominent in the G2/M phase, but manipulating nuclear levels of S100A4 using lentiviral modified cells failed to induce changes in cell cycle distribution and proliferation. Proximity ligation assay did, however, demonstrate proximity between S100A4 and cyclin B1 in vitro, while confocal microscopy showed S100A4 to localize to areas corresponding to centrosomes in mitotic cells prior to chromosome segregation. This might indicate a novel and uncharacterized function of the metastasis-associated protein in CRC cells.


Assuntos
Núcleo Celular/química , Centrossomo/fisiologia , Neoplasias Colorretais/patologia , Ciclina B1/fisiologia , Proteínas S100/fisiologia , Animais , Divisão Celular , Linhagem Celular Tumoral , Fase G2 , Humanos , Camundongos , Proteína A4 de Ligação a Cálcio da Família S100 , Proteína Supressora de Tumor p53/fisiologia
6.
Cancer Lett ; 366(1): 71-83, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26095603

RESUMO

Invasiveness is a hallmark of aggressive cancer like malignant melanoma, and factors involved in acquisition or maintenance of an invasive phenotype are attractive targets for therapy. We investigated melanoma phenotype modulation induced by the metastasis-promoting microenvironmental protein S100A4, focusing on the relationship between enhanced cellular motility, dedifferentiation and metabolic changes. In poorly motile, well-differentiated Melmet 5 cells, S100A4 stimulated migration, invasion and simultaneously down-regulated differentiation genes and modulated expression of metabolism genes. Metabolic studies confirmed suppressed mitochondrial respiration and activated glycolytic flux in the S100A4 stimulated cells, indicating a metabolic switch toward aerobic glycolysis, known as the Warburg effect. Reversal of the glycolytic switch by dichloracetate induced apoptosis and reduced cell growth, particularly in the S100A4 stimulated cells. This implies that cells with stimulated invasiveness get survival benefit from the glycolytic switch and, therefore, become more vulnerable to glycolysis inhibition. In conclusion, our data indicate that transition to the invasive phenotype in melanoma involves dedifferentiation and metabolic reprogramming from mitochondrial oxidation to glycolysis, which facilitates survival of the invasive cancer cells. Therapeutic strategies targeting the metabolic reprogramming may therefore be effective against the invasive phenotype.


Assuntos
Melanoma/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Humanos , Melanoma/metabolismo , Mitocôndrias/efeitos dos fármacos , Invasividade Neoplásica , Fenótipo , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/farmacologia
7.
Dev Biol ; 352(2): 181-90, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21195708

RESUMO

High levels of the S100 calcium binding protein S100A4 also called fibroblast specific protein 1 (FSP1) have been established as an inducer of metastasis and indicator of poor prognosis in breast cancer. The mechanism by which S100A4 leads to increased cancer aggressiveness has yet to be established; moreover, the function of this protein in normal mammary gland biology has not been investigated. To address the role of S100A4 in normal mammary gland, its spatial and temporal expression patterns and possible function in branching morphogenesis were investigated. We show that the protein is expressed mainly in cells of the stromal compartment of adult humans, and during active ductal development, in pregnancy and in involution of mouse mammary gland. In 3D culture models, topical addition of S100A4 induced a significant increase in the TGFα mediated branching phenotype and a concomitant increase in expression of a previously identified branching morphogen, metalloproteinase-3 (MMP-3). These events were found to be dependent on MEK activation. Downregulation of S100A4 using shRNA significantly reduced TGFα induced branching and altered E-cadherin localization. These findings provide evidence that S100A4 is developmentally regulated and that it plays a functional role in mammary gland development, in concert with TGFα by activating MMP-3, and increasing invasion into the fat pad during branching. We suggest that S100A4-mediated effects during branching morphogenesis provide a plausible mechanism for how it may function in breast cancer progression.


Assuntos
Mama/crescimento & desenvolvimento , Glândulas Mamárias Animais/crescimento & desenvolvimento , Metástase Neoplásica/fisiopatologia , Proteínas S100/fisiologia , Animais , Sequência de Bases , Neoplasias da Mama/fisiopatologia , Adesão Celular/fisiologia , Linhagem Celular , Células Epiteliais/metabolismo , Feminino , Humanos , Glândulas Mamárias Animais/efeitos dos fármacos , Glândulas Mamárias Animais/metabolismo , Metaloproteinase 3 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Morfogênese , Gravidez , Prognóstico , RNA Interferente Pequeno/genética , Proteínas Recombinantes/farmacologia , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/antagonistas & inibidores , Proteínas S100/genética , Proteínas S100/farmacologia , Células Estromais/metabolismo
8.
Tumour Biol ; 28(1): 27-35, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17143014

RESUMO

We have previously shown that interferon-gamma (IFN-gamma) inhibits expression of the metastasis-promoting protein S100A4. In the present study, we further explore the mechanism behind the IFN-gamma-mediated effects on the human S100A4 promoter and demonstrate that IFN-gamma represses S100A4 promoter activity through induction of the class II transactivator (CIITA). The acidic domain in the N-terminal part of CIITA was crucial for the observed IFN-gamma-induced inhibition of S100A4 promoter activity, probably by binding the histone acetyltransferase CBP/p300. Importantly, overexpression of CIITA significantly reduced the expression of endogenous S100A4. Our data suggest a model where CIITA represses S100A4 transcription through sequestering of CBP/p300, thereby reducing the level of CBP/p300 at the S100A4 promoter, which in turn leads to inhibition of S100A4 transcription.


Assuntos
Interferon gama/farmacologia , Proteínas Nucleares/efeitos dos fármacos , Proteínas S100/efeitos dos fármacos , Transativadores/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Células COS , Linhagem Celular , Linhagem Celular Tumoral , Chlorocebus aethiops , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteossarcoma/genética , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Proteína A4 de Ligação a Cálcio da Família S100 , Proteínas S100/genética , Proteínas S100/metabolismo , Transativadores/genética , Transativadores/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...