Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncotarget ; 10(23): 2292-2305, 2019 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-31040920

RESUMO

Vitamin K serves as an essential co-factor in the γ-carboxylation of glutamate to γ-carboxyglutamate (GLA), a post-translational modification mediated by gamma-glutamyl carboxylase (GGCX) and vitamin K oxidoreductases (VKORC1 or VKORC1L1). While both phylloquinone (K1) and menaquinone (K2) support the synthesis of GLA-modified proteins, studies assessing K1 and/or K2 effects in cancer cells have reported minimal effects of K1 and anti-proliferative or pro-apoptotic effects of K2. qPCR results indicated highest expression of GGCX, VKORC1, and VKORC1L1 in triple negative breast cancer (TNBC) cell lines, Hs578T, MDA-MB-231 and SUM159PT, and in advanced stage disease. To assess differential effects of vitamin K, TNBC cells were cultured in media supplemented with K1 or K2. K1 treatment increased cell growth, and enhanced stemness and GLA-modified protein expression in TNBC lysates. Alternatively, lysates from cells exposed to vehicle, K2, or the VKOR antagonist, warfarin, did not express GLA-modified proteins. Further, K2 exposure reduced stemness and elicited anti-proliferative effects. These studies show that TNBC cells express a functional vitamin K pathway and that K1 and K2 exert distinct phenotypic effects. Clarification of the mechanisms by which K1 and K2 induce these effects may lead to relevant therapeutic strategies for manipulating this pathway in TNBC patients.

2.
Endocrinology ; 158(12): 4174-4188, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29029014

RESUMO

Genomic profiling has identified a subset of metabolic genes that are altered by 1,25-dihydroxyvitamin D (1,25D) in breast cells, including GLUL, the gene that encodes glutamine synthetase (GS). In this study, we explored the relevance of vitamin D modulation of GLUL and other metabolic genes in the context of glutamine utilization and dependence. We show that exposure of breast epithelial cells to glutamine deprivation or a GS inhibitor reduced growth and these effects were exacerbated by cotreatment with 1,25D. 1,25D downregulation of GLUL was sufficient to reduce abundance and activity of GS. Flow cytometry demonstrated that glutamine deprivation induced S phase arrest, likely due to reduced availability of glutamine for DNA synthesis. In contrast, 1,25D induced G0/G1 arrest, indicating that its effects are not solely due to reduced glutamine synthesis. Indeed, 1,25D also reduced expression of GLS1 and GLS2 genes, which code for glutaminases that shunt glutamine into the tricarboxylic acid (TCA) cycle. Consistent with reduced entry of glutamine into the TCA cycle, 1,25D inhibited glutamine oxidation and the metabolic response to exogenous glutamine as analyzed by Seahorse Bioscience extracellular flux assays. Effects of 1,25D on GLUL/GS expression and glutamine oxidation were retained in human mammary epithelial (HME) cells that express SV-40 (HME-LT cells) but not in those that express SV-40 and oncogenic H-Ras (HME-PR cells). Furthermore, HME-PR cells exhibited glutamine independence and expressed constitutively high levels of GLUL/GS, which were unaffected by 1,25D. Collectively, these data suggest that 1,25D alters glutamine availability, dependence, and metabolism in nontransformed and preneoplastic mammary epithelial cells in association with cell cycle arrest.


Assuntos
Células Epiteliais/efeitos dos fármacos , Glutamato-Amônia Ligase/metabolismo , Glutamina/metabolismo , Vitamina D/análogos & derivados , Western Blotting , Mama/citologia , Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Epiteliais/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glutamato-Amônia Ligase/genética , Glutamina/farmacologia , Humanos , Oxirredução/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vitamina D/farmacologia , Vitaminas/farmacologia
3.
Mol Cell Endocrinol ; 424: 34-41, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26774511

RESUMO

Genomic profiling of immortalized human mammary epithelial (hTERT-HME1) cells identified several metabolic genes, including the membrane glutamate transporter, SLC1A1, as 1,25-dihydroxyvitamin D3 (1,25D) regulated. In these studies we have surveyed the effects of 1,25D on known glutamate transporters and evaluated its impact on cellular glutamate handling. We confirm that expression of SLC1A1 and all of its known transcript variants are significantly upregulated in hTERT-HME1 cells following 1,25D treatment. Expression of the full-length cognate protein, EAAT3, is correspondingly increased in 1,25D treated hTERT-HME1 cells. Under the same conditions, the expression of two other glutamate transporters--SLC1A6 (EAAT4) and SLC1A2 (EAAT2 or GLT-1)--is enhanced by 1,25D while that of SLC1A3 (EAAT1 or GLAST) and SLC7A11 (xCT) is decreased. Glutamate is not essential for growth of hTERT-HME1 cells, and supplemental glutamate (up to 0.5 mM) does not abrogate the growth inhibitory effects of 1,25D. These data suggest that extracellular glutamate is not a major contributor to cellular energy metabolism in hTERT-HME1 cells under basal conditions and that the growth inhibitory effects of 1,25D are not secondary to its effects on glutamate handling. Instead, the effects of 1,25D on glutamate transporters translated to a decrease in cellular glutamate concentration and an increase in media glutamate concentration, suggesting that one or more of these transporters functions to export glutamate in response to 1,25D exposure. The reduced cellular glutamate concentration may also reflect its incorporation into the cellular glutathione (GSH) pool, which is increased upon 1,25D treatment. In support of this concept, the expression of GCLC (which codes for the rate-limiting enzyme in GSH synthesis) and genes which generate reducing equivalents in the form of NADPH (ie, G6PD, PGD, IDH2) are elevated in 1,25D-treated cells. Taken together, these data identify 1,25D as a physiological regulator of multiple membrane glutamate transporters that impacts on overall cellular glutamate handling.


Assuntos
Células Epiteliais/efeitos dos fármacos , Transportador 3 de Aminoácido Excitatório/genética , Transportador 3 de Aminoácido Excitatório/metabolismo , Ácido Glutâmico/farmacologia , Glândulas Mamárias Humanas/citologia , Vitamina D/análogos & derivados , Sistema y+ de Transporte de Aminoácidos/genética , Linhagem Celular , Meios de Cultura/farmacologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 2 de Aminoácido Excitatório , Transportador 4 de Aminoácido Excitatório/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Transporte de Glutamato da Membrana Plasmática/genética , Humanos , Glândulas Mamárias Humanas/efeitos dos fármacos , Glândulas Mamárias Humanas/metabolismo , Isoformas de Proteínas/metabolismo , Vitamina D/farmacologia
4.
J Cell Biochem ; 116(8): 1693-711, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25736056

RESUMO

To elucidate potential mediators of vitamin D receptor (VDR) action in breast cancer, we profiled the genomic effects of its ligand 1,25-dihydroxyvitamin D3 (1,25D) in cells derived from normal mammary tissue and breast cancer. In non-transformed hTERT-HME cells, 483 1,25D responsive entities in 42 pathways were identified, whereas in MCF7 breast cancer cells, 249 1,25D responsive entities in 31 pathways were identified. Only 21 annotated genes were commonly altered by 1,25D in both MCF7 and hTERT-HME cells. Gene set enrichment analysis highlighted eight pathways (including senescence/autophagy, TGFß signaling, endochondral ossification, and adipogenesis) commonly altered by 1,25D in hTERT-HME and MCF7 cells. Regulation of a subset of immune (CD14, IL1RL1, MALL, CAMP, SEMA6D, TREM1, CSF1, IL33, TLR4) and metabolic (ITGB3, SLC1A1, G6PD, GLUL, HIF1A, KDR, BIRC3) genes by 1,25D was confirmed in hTERT-HME cells and similar changes were observed in another comparable non-transformed mammary cell line (HME cells). The effects of 1,25D on these genes were retained in HME cells expressing SV40 large T antigen but were selectively abrogated in HME cells expressing SV40 + RAS and in MCF7 cells. Integration of the datasets from hTERT-HME and MCF7 cells with publically available RNA-SEQ data from 1,25D treated SKBR3 breast cancer cells enabled identification of an 11-gene signature representative of 1,25D exposure in all three breast-derived cell lines. Four of these 11 genes (CYP24A1, CLMN, EFTUD1, and SERPINB1) were also identified as 1,25D responsive in human breast tumor explants, suggesting that this gene signature may prove useful as a biomarker of vitamin D exposure in breast tissue.


Assuntos
Neoplasias da Mama/genética , Calcitriol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glândulas Mamárias Humanas/efeitos dos fármacos , Vitaminas/farmacologia , Linhagem Celular , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Células MCF-7 , Glândulas Mamárias Humanas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos
5.
J Steroid Biochem Mol Biol ; 148: 96-102, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25239595

RESUMO

Previous genomic profiling of immortalized, non-tumorigenic human breast epithelial cells identified a set of 1,25-dihydroxyvitamin D3 (1,25D) regulated genes with potential relevance to breast cancer prevention. In this report, we characterized the effect of 1,25D on a subset of these genes in six cell lines derived from mammary tissue and breast cancers. Non-tumorigenic cell lines included hTERT-HME1, HME and MCF10A cells which are often used to model normal breast epithelial cells. Breast cancer cell lines included MCF7 cells (a model of early stage, estrogen-dependent disease), DCIS.com cells (a derivative of MCF10A cells that models in situ breast cancer) and Hs578T cells (a model of metastatic disease). All of these cell lines express the vitamin D receptor (VDR) and exhibit anti-cancer responses to 1,25D such as changes in proliferation, apoptosis, metabolism, or invasion. Our comparative data demonstrate highly variable responses to 1,25D (100nM, 24h) between the cell lines. In both hTERT-HME1 and HME cell lines, CYP24A1, SLC1A1 and ITGB3 were up-regulated whereas KDR, GLUL and BIRC3 were down-regulated in response to 1,25D. In contrast, no changes in SLC1A1, ITGB3 or GLUL expression were detected in 1,25D treated MCF10A cells although KDR and BIRC3 were down-regulated by 1,25D. The effects of 1,25D on these genes in the breast cancer cell lines were blunted, with the DCIS.com cells exhibiting the most similar responses to the immortalized hTERT-HME1 and HME cells. The differences in cellular responses were not due to general impairment in VDR function as robust CYP24A1 induction was observed in all cell lines. Thus, our data indicate that the genomic changes induced by 1,25D are highly cell-type specific even in model cell lines derived from the same tissue. The implication of these findings is that genomic responses to changes in vitamin D status in vivo are likely to be distinct from individual to individual, particularly in neoplastic tissue. This article is part of a Special Issue entitled '17th Vitamin D Workshop'.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Mama/metabolismo , Calcitriol/farmacologia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Biomarcadores Tumorais/metabolismo , Mama/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Feminino , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Vitaminas/farmacologia
6.
Front Physiol ; 5: 213, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24982636

RESUMO

Nuclear receptors exert profound effects on mammary gland physiology and have complex roles in the etiology of breast cancer. In addition to receptors for classic steroid hormones such as estrogen and progesterone, the nuclear vitamin D receptor (VDR) interacts with its ligand 1α,25(OH)2D3 to modulate the normal mammary epithelial cell genome and subsequent phenotype. Observational studies suggest that vitamin D deficiency is common in breast cancer patients and that low vitamin D status enhances the risk for disease development or progression. Genomic profiling has characterized many 1α,25(OH)2D3 responsive targets in normal mammary cells and in breast cancers, providing insight into the molecular actions of 1α,25(OH)2D3 and the VDR in regulation of cell cycle, apoptosis, and differentiation. New areas of emphasis include regulation of tumor metabolism and innate immune responses. However, the role of VDR in individual cell types (i.e., epithelial, adipose, fibroblast, endothelial, immune) of normal and tumor tissues remains to be clarified. Furthermore, the mechanisms by which VDR integrates signaling between diverse cell types and controls soluble signals and paracrine pathways in the tissue/tumor microenvironment remain to be defined. Model systems of carcinogenesis have provided evidence that both VDR expression and 1α,25(OH)2D3 actions change with transformation but clinical data regarding vitamin D responsiveness of established tumors is limited and inconclusive. Because breast cancer is heterogeneous, analysis of VDR actions in specific molecular subtypes of the disease may help to clarify the conflicting data. The expanded use of genomic, proteomic and metabolomic approaches on a diverse array of in vitro and in vivo model systems is clearly warranted to comprehensively understand the network of vitamin D regulated pathways in the context of breast cancer.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...