Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Mol Carcinog ; 61(6): 549-557, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35319799

RESUMEN

Pancreatic ductal adenocarcinoma (PDA) tumors have a highly immunosuppressive desmoplastic tumor microenvironment (TME) where immune checkpoint inhibition (ICI) therapy has been exceptionally ineffective. Transforming growth factor-ß (TGF-ß) receptor activation leads to cancer and immune cell proliferation and phenotype, and cytokine production leading to tumor progression and worse overall survival in PDA patients. We hypothesized that TGF-ß receptor inhibition may alter PDA progression and antitumor immunity in the TME. Here, we used a syngeneic preclinical murine model of PDA to explore the impact of TGF-ß pathway inhibitor galunisertib (GAL), dual checkpoint immunotherapy (anti-PD-L1 and CTLA-4), the chemotherapy gemcitabine (GEM), and their combinations on antitumor immune responses. Blockade of TGF-ß and ICI in immune-competent mice bearing orthotopically injected murine PDA cells significantly inhibited tumor growth and was accompanied by antitumor M1 macrophage infiltration. In contrast, GEM treatment resulted in increased PDA tumor growth, decreased antitumor M1 macrophages, and decreased cytotoxic CD8+ T cell subpopulation compared to control mice. Together, these findings demonstrate the ability of TGF-ß inhibition with GAL to prime antitumor immunity in the TME and the curative potential of combining GAL with dual ICI. These preclinical results indicate that targeted inhibition of TGF-ß may enhance the efficacy of dual immunotherapy in PDA. Optimal manipulation of the immune TME with non-ICI therapy may enhance therapeutic efficacy.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Carcinoma Ductal Pancreático/genética , Desoxicitidina/análogos & derivados , Humanos , Inmunoterapia/métodos , Ratones , Neoplasias Pancreáticas/patología , Receptores de Factores de Crecimiento Transformadores beta , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral , Gemcitabina , Neoplasias Pancreáticas
2.
Chromosome Res ; 26(4): 255-276, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30009337

RESUMEN

In recent years, some transcription factors have been observed to remain associated with mitotic chromatin. Based on these observations, it is suggested that these chromatin-bound transcription factors may serve as 'epigenetic marks' for transmission of pattern of gene expression from progenitor to progeny cells. In this context, our laboratory has reported that nuclear receptor PXR, a master regulator of xenobiotic metabolism, remains constitutively associated with mitotic chromatin. However, the region responsible for this interaction with chromatin remained unknown. In this study, we have shown, for the first time, that mitotic chromatin association of this factor is mediated by the combined action of two zinc fingers present in the DNA-binding domain of PXR. Overall, the nuclear localization signal (NLS) region appears to play a major role in this interaction with mitotic chromatin. Also, we have identified a sub-region of 11 amino acid residues within NLS region of PXR (R66-76R) essential for receptor interaction with the mitotic chromatin. Interestingly, this minimal region is sequence-specific and independent of its basic charge. We have termed this minimal sub-region as 'mitotic chromatin binding-determining region' (MCBR). It is suggested that this receptor region is essential for activation of its target genes. Additionally, we have shown that PXR remains associated with the everted repeat (ER6) region of its major target gene, CYP3A4 promoter during mitosis implying its suggested role in 'gene bookmarking'.


Asunto(s)
Cromatina/metabolismo , Señales de Localización Nuclear , Receptor X de Pregnano/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Citocromo P-450 CYP3A/genética , Humanos , Mitosis , Receptor X de Pregnano/química , Regiones Promotoras Genéticas , Receptores Citoplasmáticos y Nucleares , Dedos de Zinc
3.
Biochim Biophys Acta ; 1859(9): 1183-1197, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26962022

RESUMEN

Pregnane & Xenobiotic Receptor (PXR) acts as a xenosensing transcriptional regulator of many drug metabolizing enzymes and transporters of the 'detoxification machinery' that coordinate in elimination of xenobiotics and endobiotics from the cellular milieu. It is an accepted view that some individuals or specific populations display considerable differences in their ability to metabolize different drugs, dietary constituents, herbals etc. In this context we speculated that polymorphisms in PXR gene might contribute to variability in cytochrome P450 (CYP450) metabolizing enzymes of phase I, drug metabolizing components of phase II and efflux components of the detoxification machinery. Therefore, in this study, we have undertaken a comprehensive functional analysis of seventeen naturally occurring non-synonymous variants of human PXR. When compared, we observed that some of the PXR SNP variants exhibit distinct functional and dynamic responses on parameters which included transcriptional function, sub-cellular localization, mitotic chromatin binding, DNA-binding properties and other molecular interactions. One of the unique SNP located within the DNA-binding domain of PXR was found to be functionally null and distinct on other parameters. Similarly, some of the non-synonymous SNPs in PXR imparted reduced transactivation function as compared to wild type PXR. Interestingly, PXR is reported to be a mitotic chromatin binding protein and such an association has been correlated to an emerging concept of 'transcription memory' and altered transcription output. In view of the observations made herein our data suggest that some of the natural PXR variants may have adverse physiological consequences owing to its influence on the expression levels and functional output of drug-metabolizing enzymes and transporters. The present study is expected to explain not only the observed inter-individual responses to different drugs but may also highlight the mechanistic details and importance of PXR in drug clearance, drug-drug interactions and diverse metabolic disorders. This article is part of a Special Issue entitled: Xenobiotic nuclear receptors: New Tricks for An Old Dog, edited by Dr. Wen Xie.


Asunto(s)
Cromatina/química , Sistema Enzimático del Citocromo P-450/genética , ADN/química , Polimorfismo de Nucleótido Simple , Receptores de Esteroides/química , Alelos , Sustitución de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Cromatina/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , ADN/metabolismo , Exones , Regulación de la Expresión Génica , Frecuencia de los Genes , Células Hep G2 , Humanos , Inactivación Metabólica/genética , Mitosis , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Receptor X de Pregnano , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Homología Estructural de Proteína , Relación Estructura-Actividad
4.
Cell Biol Int ; 41(5): 570-576, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28198586

RESUMEN

The Pregnane and Xenobiotic Receptor (PXR; NR1I2) is a ligand-modulated transcription factor that belongs to the nuclear receptor superfamily. It is expressed at higher levels primarily in liver and intestine as compared to the levels in several other organs. It is activated by a broad spectrum of xenobiotics and endobiotics. The primary function of PXR is to regulate the expression of drug metabolizing enzymes and transporters and prevent the accumulation of toxic chemicals in the body, thereby maintaining body's homeostasis. In this study, we identified a C/T single nucleotide polymorphism at position -831 from the transcriptional start site of the PXR gene promoter and examined the functional significance of this variant using both the luciferase reporter gene assays and electrophoretic mobility shift assays (EMSA). Transient transfection experiments showed that the T-allele was associated with significantly greater transcriptional activity than the C-allele of SNP rs3814055. These results indicate that the -831C/T polymorphism has a direct effect on transcriptional regulation of PXR gene. This allelic variation may be a potential genetic marker that can help identify individuals at higher risk for Inflammatory Bowel Disease (IBD).


Asunto(s)
Polimorfismo de Nucleótido Simple/genética , Regiones Promotoras Genéticas , Receptores de Esteroides/genética , Alelos , Extractos Celulares , Hepatocitos/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Receptor X de Pregnano , Unión Proteica
5.
Exp Cell Res ; 339(2): 187-96, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26586566

RESUMEN

Pregnane and Xenobiotic Receptor (PXR), a member of nuclear receptor superfamily, acts as a 'xenosensor' in our body and modulates a network of genes involved in xenobiotic metabolism and elimination. Expression levels of PXR in certain metabolic disorders including cancer are reported to be altered and its induced expression is associated with the development of resistance towards chemotherapy and adverse drug-drug interactions. Though the transcriptional regulation of PXR target genes have been elucidated in significant details, the structure and functional control of PXR promoter itself remains inadequately explored. In this work, we identify a Composite Element (CE) located within the proximal PXR promoter region that consists of multiple overlapping cis-elements and demonstrated that CE interacts specifically with some critical nuclear proteins. Subsequent DNA-protein interaction studies revealed mutually exclusive interactions on CE occurring between Sp1 and two unidentified DNA binding proteins with molecular masses of 50 and 54kDa. Here, we report the identification of 54kDa CE binding protein as a heterogeneous nuclear ribonucleoprotein K (hnRNPK) and demonstrate the effect of hnRNP K and Sp1 on PXR promoter transcriptional activity. Overall, the study indicates that PXR gene is tightly regulated to maintain a low receptor level.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Receptores de Esteroides/genética , Animales , Sitios de Unión , Células Cultivadas , Proteínas de Unión al ADN/química , Regulación de la Expresión Génica , Células Hep G2 , Ribonucleoproteína Heterogénea-Nuclear Grupo K , Humanos , Receptor X de Pregnano , Regiones Promotoras Genéticas/genética , Receptores de Esteroides/metabolismo , Elementos de Respuesta/genética , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo
6.
Exp Cell Res ; 330(2): 398-411, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25265064

RESUMEN

Nuclear receptor PXR is predominantly expressed in liver and intestine. Expression of PXR is observed to be dysregulated in various metabolic disorders indicating its involvement in disease development. However, information available on mechanisms of PXR self-regulation is fragmentary. The present investigation identifies some of the regulatory elements responsible for its tight regulation and low cellular expression. Here, we report that the PXR-promoter is a target for some key transcription factors like PU.1/Ets-1, Pax5, LEF-1 and c-Jun. Interestingly, we observed that PXR-promoter responsiveness to Pax5, LEF-1 and c-Jun, is considerably enhanced by Ets transcription factors (PU.1 and Ets-1). Co-transfection of cells with Ets-1, LEF-1 and c-Jun increased PXR-promoter activity by 5-fold and also induced expression of endogenous human PXR. Site-directed mutagenesis and transfection studies revealed that two Ets binding sites and two of the three LEF binding sites in the PXR-promoter are functional and have a positive effect on PXR transcription. Results suggest that expression of Ets family members, in conjunction with Pax5, LEF-1 and c-Jun, lead to coordinated up-regulation of PXR gene transcription. Insights obtained on the regulation of PXR gene have relevance in offering important cues towards normal functioning as well as development of several metabolic disorders via PXR signaling.


Asunto(s)
Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Hígado/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factor de Transcripción PAX5/metabolismo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Receptores de Esteroides/biosíntesis , Sitios de Unión/genética , Ensayo de Cambio de Movilidad Electroforética , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Receptor X de Pregnano , Regiones Promotoras Genéticas , Unión Proteica/genética , Proteína Proto-Oncogénica c-ets-1/genética , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño , Receptores de Esteroides/genética , Transactivadores/metabolismo , Transcripción Genética , Activación Transcripcional/genética
7.
Mol Cancer Ther ; 22(8): 936-946, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37294884

RESUMEN

Identifying novel, unique, and personalized molecular targets for patients with pancreatic ductal adenocarcinoma (PDAC) remains the greatest challenge in altering the biology of fatal tumors. Bromo- and extra-terminal domain (BET) proteins are activated in a noncanonical fashion by TGFß, a ubiquitous cytokine in the PDAC tumor microenvironment (TME). We hypothesized that BET inhibitors (BETi) represent a new class of drugs that attack PDAC tumors via a novel mechanism. Using a combination of patient and syngeneic murine models, we investigated the effects of the BETi drug BMS-986158 on cellular proliferation, organoid growth, cell-cycle progression, and mitochondrial metabolic disruption. These were investigated independently and in combination with standard cytotoxic chemotherapy (gemcitabine + paclitaxel [GemPTX]). BMS-986158 reduced cell viability and proliferation across multiple PDAC cell lines in a dose-dependent manner, even more so in combination with cytotoxic chemotherapy (P < 0.0001). We found that BMS-986158 reduced both human and murine PDAC organoid growth (P < 0.001), with associated perturbations in the cell cycle leading to cell-cycle arrest. BMS-986158 disrupts normal cancer-dependent mitochondrial function, leading to aberrant mitochondrial metabolism and stress via dysfunctional cellular respiration, proton leakage, and ATP production. We demonstrated mechanistic and functional data that BETi induces metabolic mitochondrial dysfunction, abrogating PDAC progression and proliferation, alone and in combination with systemic cytotoxic chemotherapies. This novel approach improves the therapeutic window in patients with PDAC and offers another treatment approach distinct from cytotoxic chemotherapy that targets cancer cell bioenergetics.


Asunto(s)
Antineoplásicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Ratones , Animales , Gemcitabina , Línea Celular Tumoral , Carcinoma Ductal Pancreático/patología , Neoplasias Pancreáticas/patología , Antineoplásicos/farmacología , Desoxicitidina/uso terapéutico , Proliferación Celular , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Mitocondrias , Microambiente Tumoral
8.
Sci Rep ; 11(1): 1393, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-33446905

RESUMEN

Prostate cancer (PCa) is dependent on the androgen receptor (AR). Advanced PCa is treated with an androgen deprivation therapy-based regimen; tumors develop resistance, although they typically remain AR-dependent. Expression of constitutively active AR variants lacking the ligand-binding domain including the variant AR-V7 contributes to this resistance. AR and AR-V7, as transcription factors, regulate many of the same genes, but also have unique activities. In this study, the capacity of the two AR isoforms to regulate splicing was examined. RNA-seq data from models that endogenously express AR and express AR-V7 in response to doxycycline were used. Both AR isoforms induced multiple changes in splicing and many changes were isoform-specific. Analyses of two endogenous genes, PGAP2 and TPD52, were performed to examine differential splicing. A novel exon that appears to be a novel transcription start site was preferentially induced by AR-V7 in PGAP2 although it is induced to a lesser extent by AR. The previously described AR induced promoter 2 usage that results in a novel protein derived from TPD52 (PrLZ) was not induced by AR-V7. AR, but not AR-V7, bound to a site proximal to promoter 2, and induction was found to depend on FOXA1.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/biosíntesis , Neoplasias de la Próstata/metabolismo , Empalme del ARN , Receptores Androgénicos/biosíntesis , Línea Celular Tumoral , Humanos , Masculino , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , RNA-Seq , Receptores Androgénicos/genética
9.
Mol Cell Endocrinol ; 420: 194-207, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26549688

RESUMEN

Pregnane & Xenobiotic Receptor (PXR) is one of the 48 members of the nuclear receptor superfamily of ligand-modulated transcription factors. PXR plays an important role in metabolism and elimination of diverse noxious endobiotics and xenobiotics. Like in case of some nuclear receptors its function may also be differentially altered, positively or negatively, by various post-translational modifications. In this context, regulation of PXR function by SUMOylation is the subject of present investigation. Here, we report that human PXR is modified by SUMO-1 resulting in its enhanced transcriptional activity. RT-PCR analysis showed that PXR SUMOylation in presence of rifampicin also enhances the endogenous expression levels of key PXR-regulated genes like CYP3A4, CYP2C9, MDR1 and UGT1A1. In addition, mammalian two-hybrid assay exhibited enhanced interaction between PXR and co-activator SRC-1. EMSA results revealed that SUMOylation has no influence on the DNA binding ability of PXR. In silico analysis suggested that PXR protein contains four putative SUMOylation sites, centered at K108, K129, K160 and K170. In addition to this, we identified the presence of NDSM (Negative charge amino acid Dependent SUMOylation Motif) in PXR. Substitution of all its four putative lysine residues along with NDSM abolished the effect of SUMO-1-mediated transactivation function of PXR. Furthermore, we show that interaction between PXR and E2-conjugation enzyme UBCh9, an important step for implementation of SUMOylation event, was reduced in case of NDSM mutant PXRD115A. Overall, our results suggest that SUMOylation at specific sites on PXR protein are involved in enhancement of transcription function of this receptor.


Asunto(s)
Regulación de la Expresión Génica , Procesamiento Proteico-Postraduccional , Receptores de Esteroides/química , Receptores de Esteroides/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Línea Celular , ADN/metabolismo , Perfilación de la Expresión Génica , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Receptor X de Pregnano , Unión Proteica , Estabilidad Proteica , Relación Estructura-Actividad , Sumoilación , Transcripción Genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA