Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Physiol Rev ; 99(2): 1153-1222, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30724669

RESUMO

UDP-glycosyltransferases (UGTs) catalyze the covalent addition of sugars to a broad range of lipophilic molecules. This biotransformation plays a critical role in elimination of a broad range of exogenous chemicals and by-products of endogenous metabolism, and also controls the levels and distribution of many endogenous signaling molecules. In mammals, the superfamily comprises four families: UGT1, UGT2, UGT3, and UGT8. UGT1 and UGT2 enzymes have important roles in pharmacology and toxicology including contributing to interindividual differences in drug disposition as well as to cancer risk. These UGTs are highly expressed in organs of detoxification (e.g., liver, kidney, intestine) and can be induced by pathways that sense demand for detoxification and for modulation of endobiotic signaling molecules. The functions of the UGT3 and UGT8 family enzymes have only been characterized relatively recently; these enzymes show different UDP-sugar preferences to that of UGT1 and UGT2 enzymes, and to date, their contributions to drug metabolism appear to be relatively minor. This review summarizes and provides critical analysis of the current state of research into all four families of UGT enzymes. Key areas discussed include the roles of UGTs in drug metabolism, cancer risk, and regulation of signaling, as well as the transcriptional and posttranscriptional control of UGT expression and function. The latter part of this review provides an in-depth analysis of the known and predicted functions of UGT3 and UGT8 enzymes, focused on their likely roles in modulation of levels of endogenous signaling pathways.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Glicosiltransferases/classificação , Animais , Mamíferos/metabolismo , Família Multigênica , Transdução de Sinais/fisiologia
2.
Drug Metab Dispos ; 52(6): 526-538, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38565302

RESUMO

The human UDP-glucuronosyltransferases (UGTs) have crucial roles in metabolizing and clearing numerous small lipophilic compounds. The UGT1A locus generates nine UGT1A mRNAs, 65 spliced transcripts, and 34 circular RNAs. In this study, our analysis of published UGT-RNA capture sequencing (CaptureSeq) datasets identified novel splice junctions that predict 24 variant UGT1A transcripts derived from ligation of exon 2 to unique sequences within the UGT1A first-exon region using cryptic donor splice sites. Of these variants, seven (1A1_n1, 1A3_n3, 1A4_n4, 1A5_n1, 1A8_n2, 1A9_n2, 1A10_n7) are predicted to encode UGT1A proteins with truncated aglycone-binding domains. We assessed their expression profiles and deregulation in cancer using four RNA sequencing (RNA-Seq) datasets of paired normal and cancerous drug-metabolizing tissues from large patient cohorts. Variants were generally coexpressed with their canonical counterparts with a higher relative abundance in tumor than in normal tissues. Variants showed tissue-specific expression with high interindividual variability but overall low abundance. However, 1A8_n2 showed high abundance in normal and cancerous colorectal tissues, with levels that approached or surpassed canonical 1A8 mRNA levels in many samples. We cloned 1A8_n2 and showed expression of the predicted protein (1A8_i3) in human embryonic kidney (HEK)293T cells. Glucuronidation assays with 4-methylumbelliferone (4MU) showed that 1A8_i3 had no activity and was unable to inhibit the activity of 1A8_i1 protein. In summary, the activation of cryptic donor splice sites within the UGT1A first-exon region expands the UGT1A transcriptome and proteome. The 1A8_n2 cryptic donor splice site is highly active in colorectal tissues, representing an important cis-regulatory element that negatively regulates the function of the UGT1A8 gene through pre-mRNA splicing. SIGNIFICANT STATEMENT: The UGT1A locus generates nine canonical mRNAs, 65 alternately spliced transcripts, and 34 different circular RNAs. The present study reports a series of novel UDP-glucuronosyltransferase (UGT)1A variants resulting from use of cryptic donor splice sites in both normal and cancerous tissues, several of which are predicted to encode variant UGT1A proteins with truncated aglycone-binding domains. Of these, 1A8_n2 shows exceptionally high abundance in colorectal tissues, highlighting its potential role in the first-pass metabolism in gut through the glucuronidation pathway.


Assuntos
Éxons , Glucuronosiltransferase , Sítios de Splice de RNA , Humanos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Éxons/genética , Sítios de Splice de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Domínios Proteicos/genética , Processamento Alternativo/genética
3.
Drug Metab Rev ; 54(2): 120-140, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35275773

RESUMO

The human UGT gene superfamily is divided into four subfamilies (UGT1, UGT2, UGT3 and UGT8) that encodes 22 functional enzymes. UGTs are critical for the metabolism and clearance of numerous endogenous and exogenous compounds, including steroid hormones, bile acids, bilirubin, fatty acids, carcinogens, and therapeutic drugs. Therefore, the expression and activities of UGTs are tightly regulated by multiple processes at the transcriptional, post-transcriptional and post-translational levels. During recent years, nearly twenty studies have investigated the post-transcriptional regulation of UGT genes by miRNAs using human cancer cell lines (predominantly liver cancer). Overall, 14 of the 22 UGT mRNAs (1A1, 1A3, 1A4, 1A6, 1A8, 1A9, 1A10, 2A1, 2B4, 2B7, 2B10, 2B15, 2B17, UGT8) have been shown to be regulated by various miRNAs through binding to their respective 3' untranslated regions (3'UTRs). Three 3'UTRs (UGT1A, UGT2B7 and UGT2B15) contain the largest number of functional miRNA target sites; in particular, the UGT1A 3'UTR contains binding sites for 12 miRNAs (548d-5p, 183-5p, 214-5p, 486-3p, 200a-3p, 491-3p, 141-3p, 298, 103b, 376b-3p, 21-3p, 1286). Although all nine UGT1A family members have the same 3'UTR, these miRNA target sites appear to be functional in an isoform-specific and cellular context-dependent manner. Collectively, these observations demonstrate that miRNAs represent important post-transcriptional regulators of the UGT gene superfamily. In this article, we present a comprehensive review of reported UGT/miRNA regulation studies, describe polymorphisms within functional miRNA target sites that may affect their functionalities, and discuss potential cooperative and competitive regulation of UGT mRNAs by miRNAs through adjacently located miRNA target sites.


Assuntos
MicroRNAs , Regiões 3' não Traduzidas , Ácidos Graxos , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Glicosiltransferases/genética , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Difosfato de Uridina
4.
Development ; 146(6)2019 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-30683662

RESUMO

Canonical Wnts promote myoblast differentiation; however, the role of ß-catenin in adult myogenesis has been contentious, and its mechanism(s) unclear. Using CRISPR-generated ß-catenin-null primary adult mouse myoblasts, we found that ß-catenin was essential for morphological differentiation and timely deployment of the myogenic gene program. Alignment, elongation and fusion were grossly impaired in null cells, and myogenic gene expression was not coordinated with cytoskeletal and membrane remodeling events. Rescue studies and genome-wide analyses extended previous findings that a ß-catenin-TCF/LEF interaction is not required for differentiation, and that ß-catenin enhances MyoD binding to myogenic loci. We mapped cellular pathways controlled by ß-catenin and defined novel targets in myoblasts, including the fusogenic genes myomaker and myomixer. We also showed that interaction of ß-catenin with α-catenin was important for efficient differentiation. Overall the study suggests dual roles for ß-catenin: a TCF/LEF-independent nuclear function that coordinates an extensive network of myogenic genes in cooperation with MyoD; and an α-catenin-dependent membrane function that helps control cell-cell interactions. ß-Catenin-TCF/LEF complexes may function primarily in feedback regulation to control levels of ß-catenin and thus prevent precocious/excessive myoblast fusion.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteína MyoD/metabolismo , Mioblastos/metabolismo , alfa Catenina/metabolismo , beta Catenina/metabolismo , Animais , Diferenciação Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citoesqueleto/metabolismo , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Genômica , Células HEK293 , Humanos , Camundongos , Desenvolvimento Muscular , Fenótipo , Regiões Promotoras Genéticas , Transdução de Sinais , Transcriptoma , Transfecção , Proteínas Wnt/metabolismo
5.
Mol Pharmacol ; 99(6): 488-503, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33824186

RESUMO

The human UDP-glycosyltransferase (UGT) gene superfamily generates 22 canonical transcripts coding for functional enzymes and also produces nearly 150 variant UGT transcripts through alternative splicing and intergenic splicing. In the present study, our analysis of circRNA databases identified backsplicing events that predicted 85 circRNAs from UGT genes, with 33, 11, and 19 circRNAs from UGT1A, UGT2B4, UGT8, respectively. Most of these UGT circRNAs were reported by one database and had low abundance in cell- or tissue-specific contexts. Using reverse-transcriptase polymerase chain reaction with divergent primers and cDNA samples from human tissues and cell lines, we found 13 circRNAs from four UGT genes: UGT1A (three), UGT2B7 (one), UGT2B10 (one), and UGT8 (eight). Notably, all eight UGT8 circRNAs contain open reading frames that include the canonical start AUG codon and encode variant proteins that all have the common 274-amino acidN-terminal region of wild-type UGT8 protein. We further showed that one UGT8 circRNA (circ_UGT8-1) was broadly expressed in human tissues and cell lines, resistant to RNase R digestion, and predominately present in the cytoplasm. We cloned five UGT8 circRNAs into the Zinc finger with KRAB and SCAN domains 1 vector and transfected them into HEK293T cells. All these vectors produced both circRNAsand linear transcripts with varying circular/linear ratios (0.17-1.14).Western blotting and mass spectrometry assays revealed that only linear transcripts and not circRNAs were translated. In conclusion, our findings of nearly 100 circRNAs greatly expand the complexity and diversity of the UGT transcriptome; however, UGT circRNAs are expressed at a very low level in specific cellular contexts, and their biologic functions remain to be determined. SIGNIFICANCE STATEMENT: The human UGT gene transcriptome comprises 22 canonical transcripts coding for functional enzymes and approximately 150 alternatively spliced and chimeric variant transcripts. The present study identified nearly 100 circRNAs from UGT genes, thus greatly expanding the complexity and diversity of the UGT transcriptome. UGT circRNAs were expressed broadly in human tissues and cell lines; however, most showed very low abundance in tissue- and cell-specific contexts, and therefore their biological functions remain to be investigated.


Assuntos
Glucuronosiltransferase/genética , RNA Circular/metabolismo , Transcriptoma , Processamento Alternativo , Linhagem Celular Tumoral , Clonagem Molecular , Humanos , RNA Circular/genética
6.
Int J Mol Sci ; 22(9)2021 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-33919234

RESUMO

Breast cancer MCF-7 cell-line-derived mammospheres were shown to be enriched in cells with a CD44+/CD24- surface profile, consistent with breast cancer stem cells (BCSC). These BCSC were previously reported to express key sphingolipid signaling effectors, including pro-oncogenic sphingosine kinase 1 (SphK1) and sphingosine-1-phosphate receptor 3 (S1P3). In this study, we explored intracellular trafficking and localization of SphK1 and S1P3 in parental MCF-7 cells, and MCF-7 derived BCSC-enriched mammospheres treated with growth- or apoptosis-stimulating agents. Intracellular trafficking and localization were assessed using confocal microscopy and cell fractionation, while CD44+/CD24- marker status was confirmed by flow cytometry. Mammospheres expressed significantly higher levels of S1P3 compared to parental MCF-7 cells (p < 0.01). Growth-promoting agents (S1P and estrogen) induced SphK1 and S1P3 translocation from cytoplasm to nuclei, which may facilitate the involvement of SphK1 and S1P3 in gene regulation. In contrast, pro-apoptotic cytokine tumor necrosis factor α (TNFα)-treated MCF-7 cells demonstrated increased apoptosis and no nuclear localization of SphK1 and S1P3, suggesting that TNFα can inhibit nuclear translocation of SphK1 and S1P3. TNFα inhibited mammosphere formation and induced S1P3 internalization and degradation. No nuclear translocation of S1P3 was detected in TNFα-stimulated mammospheres. Notably, SphK1 and S1P3 expression and localization were highly heterogenous in mammospheres, suggesting the potential for a large variety of responses. The findings provide further insights into the understanding of sphingolipid signaling and intracellular trafficking in BCs. Our data indicates that the inhibition of SphK1 and S1P3 nuclear translocation represents a novel method to prevent BCSCs proliferation.


Assuntos
Neoplasias da Mama/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais , Receptores de Esfingosina-1-Fosfato/metabolismo , Neoplasias da Mama/genética , Estrogênios/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Lisofosfolipídeos/metabolismo , Células MCF-7 , Transporte Proteico , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
7.
J Cell Biochem ; 120(8): 12740-12751, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30945349

RESUMO

The downregulation of Pax7 expression is an essential requirement for adult muscle progenitor cell differentiation during muscle regeneration. We previously found that canonical Wnt signals drive myogenic differentiation by positively regulating the expression of myogenic target genes, and also by negatively regulating Pax7 expression. To better understand how Wnt signals repress Pax7 expression, we screened for Wnt-regulated microRNAs (miRNAs) that could target the Pax7 3'untranslated region (UTR). Using wild-type and ß-catenin null primary mouse myoblasts, we identified several Wnt/ß-catenin regulated miRNAs, the most abundant of which were miR-133b and miR-206. While miR-206 was previously identified as Pax7 regulator, miR-133b has never been shown to regulate the Pax7 transcript. We show here that miR-133b is a more potent inhibitor of Pax7 expression than miR-206, and that it acts via a site adjacent to the miR-206 binding site in the Pax7 3'UTR. The primary transcript encoding miR-133b/miR-206 is specifically induced by Wnt/ß-catenin, while the miR-1/miR-133a transcript is not; moreover, Wnt signals increase the secretion of mature miR-133b/miR-206 into exosomes. Overall, we conclude that miR-133b and to a lesser degree miR-206, but not miR-1 or miR-133a, are key components of the canonical Wnt-mediated pathway that allows differentiation to proceed by relieving Pax7-mediated repression of the myogenic program. Secretion of these miRNAs into exosomes may allow them to directly control the differentiation of neighboring cells.


Assuntos
MicroRNAs/genética , Mioblastos/citologia , Fator de Transcrição PAX7/genética , Regiões 3' não Traduzidas , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Camundongos , Mioblastos/metabolismo , Via de Sinalização Wnt , beta Catenina/genética
8.
J Pharmacol Exp Ther ; 368(3): 363-381, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30578287

RESUMO

Genes involved in drug absorption, distribution, metabolism, and excretion (ADME) are called ADME genes. Currently, 298 genes that encode phase I and II drug metabolizing enzymes, transporters, and modifiers are designated as ADME genes by the PharmaADME Consortium. ADME genes are highly expressed in the liver and their levels can be influenced by liver diseases such as hepatocellular carcinoma (HCC). In this study, we obtained RNA-sequencing and microRNA (miRNA)-sequencing data from 371 HCC patients via The Cancer Genome Atlas liver hepatocellular carcinoma project and performed ADME gene-targeted differential gene expression analysis and expression correlation analysis. Two hundred thirty-three of the 298 ADME genes (78%) were expressed in HCC. Of these genes, almost one-quarter (58 genes) were significantly downregulated, while only 6% (15) were upregulated in HCC relative to healthy liver. Moreover, one-half (14/28) of the core ADME genes (CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2E1, CYP3A4, NAT1, NAT2, UGT2B7, SLC22A1, SLCO1B1, and SLCO1B3) were downregulated. In addition, about one-half of the core ADME genes were positively correlated with each other and were also positively (AHR, ARNT, HNF4A, PXR, CAR, PPARA, and RXRA) or negatively (PPARD and PPARG) correlated with transcription factors known as ADME modifiers. Finally, we show that most miRNAs known to regulate core ADME genes are upregulated in HCC. Collectively, these data reveal 1) an extensive transcription factor-mediated ADME coexpression network in the liver that efficiently coordinates the metabolism and elimination of endogenous and exogenous compounds; and 2) a widespread deregulation of this network in HCC, most likely due to deregulation of both transcriptional and post-transcriptional (miRNA) pathways.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Absorção Gastrointestinal/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Estudos de Coortes , Feminino , Absorção Gastrointestinal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Distribuição Tecidual , Adulto Jovem
9.
Br J Clin Pharmacol ; 85(1): 216-226, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30340248

RESUMO

AIMS: Demonstrate the presence of cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) proteins and mRNAs in isolated human plasma exosomes and evaluate the capacity for exosome-derived biomarkers to characterize variability in CYP3A4 activity. METHODS: The presence of CYP and UGT protein and mRNA in exosomes isolated from human plasma and HepaRG cell culture medium was determined by mass spectrometry and reverse transcription-polymerase chain reaction, respectively. The concordance between exosome-derived CYP3A4 biomarkers and midazolam apparent oral clearance (CL/F) was evaluated in a small proof-of-concept study involving six genotyped (CYP3A4 *1/*1 and CYP3A5 *3/*3) Caucasian males. RESULTS: Exosomes isolated from human plasma contained peptides and mRNA originating from CYP 1A2, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 2 J2, 3A4 and 3A5, UGT 1A1, 1A3, 1A4, 1A6, 1A9, 2B4, 2B7, 2B10 and 2B15, and NADPH-cytochrome P450 reductase. Mean (95% confidence interval) exosome-derived CYP3A4 protein expression pre- and post-rifampicin dosing was 0.24 (0.2-0.28) and 0.42 (0.21-0.65) ng ml-1 exosome concentrate. Mean (95% confidence interval) exosome CYP3A4 mRNA expression pre- and post-rifampicin dosing was 6.0 (1.1-32.7) and 48.3 (11.3-104) × 10-11 2-ΔΔCt , respectively. R2 values for correlations of exosome-derived CYP3A4 protein expression, CYP3A4 mRNA expression, and ex vivo CYP3A4 activity with midazolam CL/F were 0.905, 0.787 and 0.832, respectively. CONCLUSIONS: Consistent strong concordance was observed between exosome-derived CYP3A4 biomarkers and midazolam CL/F. The significance of these results is that CYP3A4 is the drug-metabolizing enzyme of greatest clinical importance and variability in CYP3A4 activity is poorly described by existing precision dosing strategies.


Assuntos
Variação Biológica da População , Citocromo P-450 CYP3A/metabolismo , Monitoramento de Medicamentos/métodos , Exossomos/química , Administração Oral , Adulto , Biomarcadores/análise , Linhagem Celular , Estudos de Coortes , Citocromo P-450 CYP3A/análise , Citocromo P-450 CYP3A/genética , Técnicas de Genotipagem , Glucuronosiltransferase/análise , Glucuronosiltransferase/genética , Voluntários Saudáveis , Humanos , Masculino , Espectrometria de Massas , Taxa de Depuração Metabólica , Midazolam/administração & dosagem , Midazolam/farmacocinética , Estudo de Prova de Conceito , RNA Mensageiro/análise , Adulto Jovem
10.
Mol Pharmacol ; 93(5): 541-552, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29519853

RESUMO

The gastrointestinal tract expresses several UDP-glucuronosyltransferases (UGTs) that act as a first line of defense against dietary toxins and contribute to the metabolism of orally administered drugs. The expression of UGT1A8, UGT1A9, and UGT1A10 in gastrointestinal tissues is known to be at least partly directed by the caudal homeodomain transcription factor, CDX2. We sought to further define the factors involved in regulation of the UGT1A8-1A10 genes and identified a novel composite element located within the proximal promoters of these three genes that binds to both CDX2 and the hepatocyte nuclear factor (HNF) 4α, and mediates synergistic activation by these factors. We also show that HNF4α and CDX2 are required for the expression of these UGT genes in colon cancer cell lines, and show robust correlation of UGT expression with CDX2 and HNF4α levels in normal human colon. Finally, we show that these factors are involved in the differential expression pattern of UGT1A8 and UGT1A10, which are intestinal specific, and that of UGT1A9, which is expressed in both intestine and liver. These studies lead to a model for the developmental patterning of UGT1A8, UGT1A9, and UGT1A10 in hepatic and/or extrahepatic tissues involving discrete regulatory modules that may function (independently and cooperatively) in a context-dependent manner.


Assuntos
Fator de Transcrição CDX2/metabolismo , Glucuronosiltransferase/metabolismo , Fator 4 Nuclear de Hepatócito/metabolismo , Intestinos/enzimologia , Células CACO-2 , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Glucuronosiltransferase/genética , Humanos , Fígado/enzimologia , Regiões Promotoras Genéticas , UDP-Glucuronosiltransferase 1A
11.
Mol Pharmacol ; 94(3): 938-952, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29959221

RESUMO

Recent studies have investigated alternative splicing profiles of UDP-glucuronosyltransferase (UGT) genes and identified over 130 different alternatively spliced UGT transcripts. Although UGT genes are highly clustered, the formation of chimeric transcripts by intergenic splicing between two or more UGT genes has not yet been reported. This study identified 12 chimeric transcripts (chimeras A-L) containing exons from two or three genes of the four neighboring UGT genes (UGT2B15, UGT2B29P2, UGT2B17, and UGT2B29P1) in human liver and prostate cancer cells. These chimeras typically contain the first five exons of UGT2B15 or UGT2B17 (exons 1-5) spliced to a terminal exon (exon 6) from a downstream UGT gene. Hence they encode truncated UGTs with novel C-terminal peptides. Functional assays of representative chimeric UGT proteins (termed chimeric UGT2B15 and chimeric UGT2B17) showed that they are inactive and can repress the activity of wild-type UGTs. Coimmunoprecipitation assays demonstrated heterotypic interactions between chimeric UGT2B15 (or chimeric UGT2B17) and the UGT2B7 protein. Thus oligomerization of the chimeric UGTs with wild-type UGTs may explain their inhibitory activity. Studies in breast and prostate cancer cells showed that both wild-type and chimeric UGT2B15 and UGT2B17 transcripts are regulated in a similar way at the transcriptional level by sex hormones through their canonical promoters but are differentially regulated at the post-transcriptional level by micro-RNA 376c via their unique 3'-untranslated regions. In conclusion, the formation of chimeric transcripts by intergenic splicing among UGT genes represents a novel mechanism contributing to the diversity of the human UGT transcriptome and proteome. The differential post-transcriptional regulation of wild-type and variant transcripts by micro-RNAs may contribute to their deregulated expression in cancer.


Assuntos
DNA Intergênico/genética , Variação Genética/fisiologia , Glucuronídeos/genética , Glucuronosiltransferase/genética , Antígenos de Histocompatibilidade Menor/genética , Domínios e Motivos de Interação entre Proteínas/fisiologia , Células Cultivadas , DNA Intergênico/metabolismo , Glucuronídeos/metabolismo , Glucuronosiltransferase/metabolismo , Células HEK293 , Humanos , Fígado/metabolismo , Células MCF-7 , Antígenos de Histocompatibilidade Menor/metabolismo , Splicing de RNA/fisiologia
12.
J Pharmacol Exp Ther ; 365(1): 48-59, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29367276

RESUMO

UGT2B15 is an important androgen-metabolizing UDP-glucuronosyltransferase (UGT) and the mechanisms controlling its expression are of considerable interest. Recent studies showed that miR-376c regulates UGT2B15 in prostate cancer cells via a canonical target site in the 3' untranslated region (3'UTR). The UGT2B15 3'UTR also contains a canonical miR-331-5p target site; previous work indicated that deleting this site reduced, but did not abolish, the ability of miR-331-5p to repress a luciferase reporter carrying the UGT2B15 3'UTR We report here the discovery and characterization of a second, noncanonical miR-331-5p target site in the UGT2B15 3'UTR miR-331-5p-mediated repression of a UGT2B15 3'UTR-reporter was partly inhibited by mutating either of the two miR-331-5p target sites separately, but completely abolished by mutating the two sites simultaneously, indicating that the two sites act cooperatively. miR-331-5p mimics significantly reduced both UGT2B15 mRNA levels and glucuronidation activity in prostate cancer cells, confirming that the native transcript is a miR-331-5p target. Transfection of either miR-331-5p or miR-376c mimics repressed the activity of the UGT2B15 3'UTR-reporter; however, cotransfection of both microRNAs (miRNAs) further reduced activity, indicating cooperative regulation by these two miRNAs. A significant negative correlation between miR-331 and UGT2B15 mRNA levels was observed in a tissue RNA panel, and analysis of The Cancer Genome Atlas (TCGA) hepatocellular carcinoma data set provided further evidence that miR-331 may play an important role in regulation of UGT2B15 in vivo. There was no significant correlation between miR-331 and UGT2B15 mRNA levels in the TCGA prostate adenocarcinoma cohort, which may reflect the complexity of androgen-mediated regulation in determining UGT2B15 levels in prostate cancer. Finally, we show that miR-331-5p does not regulate UGT2B17, providing the first evidence for a post-transcriptional mechanism that differentially regulates these two important androgen-metabolizing UGTs.


Assuntos
Glucuronosiltransferase/genética , MicroRNAs/genética , Neoplasias da Próstata/patologia , Regiões 3' não Traduzidas/genética , Sequência de Bases , Linhagem Celular Tumoral , Humanos , Masculino , Antígenos de Histocompatibilidade Menor/genética
13.
J Pharmacol Exp Ther ; 361(3): 482-491, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28404691

RESUMO

Exemestane (EXE) is an aromatase inhibitor indicated for endocrine therapy of breast cancer in postmenopausal women. The primary active metabolite of EXE, 17-hydroexemestane (17-HE), is inactivated via glucuronidation, mainly by UDP-glucuronosyltransferase 2B17 (UGT2B17). UGT2B17 also has a primary role in inactivation of endogenous androgens testosterone and dihydrotestosterone and may play an important role in regulation of breast and prostate tumor intracrinology. We recently reported that UGT2B17 could be induced by both estrogenic and androgenic ligands in breast cancer cells via binding of the estrogen receptor α (ERα) or the androgen receptor (AR) to a complex regulatory unit in the proximal UGT2B17 promoter. In this study we show that both EXE and 17-HE increase UGT2B17 mRNA levels in breast cancer MCF-7 and MDA-MB-453 cells, and increase glucuronidation of UGT2B17 substrates, including 17-HE and androsterone. Using antagonists of ERα and AR as well as inhibition mediated by small interfering RNA (siRNA) we demonstrate that EXE and 17-HE induce UGT2B17 expression primarily via the AR. This result is consistent with previous reports that 17-HE can act as an AR ligand. In vitro studies suggest that multiple steroid-responsive DNA elements within the proximal promoter are involved in the response to 17-HE-liganded AR. The up-regulation of UGT2B17 by EXE and 17-HE in breast cancer cells might enhance the local metabolism of 17-HE as well as that of endogenous androgens, hence impacting potentially on treatment outcomes.


Assuntos
Androstadienos/metabolismo , Androstadienos/farmacologia , Inibidores da Aromatase/metabolismo , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/enzimologia , Glucuronosiltransferase/biossíntese , Antígenos de Histocompatibilidade Menor/biossíntese , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucuronosiltransferase/genética , Humanos , Células MCF-7 , Antígenos de Histocompatibilidade Menor/genética
14.
J Pharmacol Exp Ther ; 361(3): 386-397, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28389526

RESUMO

The transcriptional regulation of UDP-glucuronosyltransferases UGT2B4 and UGT2B7 has been well studied using liver cancer cell lines, and post-transcriptional regulation of these two UGTs by microRNA (miRNA/miR) miR-216b-5p was recently reported. This study describes novel miRNA-mediated regulation of UGT2B4 and UGT2B7 in liver cancer cells. Bioinformatic analyses identified a putative miR-3664-3p binding site in the UGT2B7 3'-untranslated region (UTR) and binding sites for both miR-135a-5p and miR-410-3p in the UGT2B4 3'-UTR. These sites were functionally characterized using miRNA mimics and reporter constructs. A miR-3664-3p mimic induced repression of a luciferase reporter carrying the UGT2B7 3'-UTR in liver cancer cell lines; mutation of the miR-3664-3p site abrogated the response of the reporter to the mimic. Similarly, mutation of the miR-135a-5p site or miR-410-3p site in a luciferase reporter bearing UGT2B4 3'-UTR abrogated the ability of miR-135a-5p or miR-410-3p mimics to reduce reporter activity. Transfection of miR-3664-3p mimics in HepG2 liver cancer cells significantly reduced mRNA and protein levels of UGT2B7, and this led to reduced enzymatic activity. Transfection of miR-135a-5p or miR-410-3p mimics significantly decreased UGT2B4 mRNA levels in Huh7 liver cancer cells. The expression levels of miR-410-3p were inversely correlated with UGT2B4 mRNA levels in The Cancer Genome Atlas cohort of liver hepatocellular carcinoma (371 specimens) and a panel of ten normal human tissues. Similarly, there was an inverse correlation between miR-135a and UGT2B4 mRNA levels in a panel of 18 normal human liver tissues. Together, these data suggest that miR-135a and miR-410 control UGT2B4 and that miR-3664 controls UGT2B7 expression in liver cancer and/or normal liver cells.


Assuntos
Carcinoma Hepatocelular/metabolismo , Glucuronosiltransferase/fisiologia , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Sítios de Ligação/fisiologia , Carcinoma Hepatocelular/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , MicroRNAs/genética
15.
Stem Cells ; 34(8): 2169-82, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27144473

RESUMO

Satellite cells are the resident stem cells of skeletal muscle; quiescent in adults until activated by injury to generate proliferating myoblasts. The canonical Wnt signalling pathway, mediated by T-cell factor/lymphoid enhancer factor (TCF/LEF) and ß-catenin effector proteins, controls myoblast differentiation in vitro, and recent work suggests that timely termination of the Wnt/ß-catenin signal is important for normal adult myogenesis. We recently identified the Barx2 and Pax7 homeobox proteins as novel components of the Wnt effector complex. Here, we examine molecular and epigenetic mechanisms by which Barx2 and Pax7 regulate the canonical Wnt target gene Axin2, which mediates critical feedback to terminate the transcriptional response to Wnt signals. Barx2 is recruited to the Axin2 gene via TCF/LEF binding sites, recruits ß-catenin and the coactivator GRIP-1, and induces local H3K-acetylation. Barx2 also promotes nuclear localization of ß-catenin. Conversely, Pax7 represses Axin2 promoter/intron activity and inhibits Barx2-mediated H3K-acetylation via the corepressor HDAC1. Wnt3a not only induces Barx2 mRNA, but also stabilises Barx2 protein in myoblasts; conversely, Wnt3a potently inhibits Pax7 protein expression. As Barx2 promotes myogenic differentiation and Pax7 suppresses it, this novel posttranscriptional regulation of Barx2 and Pax7 by Wnt3a may be involved in the specification of differentiation-competent and -incompetent myoblast populations. Finally, we propose a model for dual function of Barx2 downstream of Wnt signals: activation of myogenic target genes in association with canonical myogenic regulatory factors, and regulation of the negative feedback loop that limits the response of myoblasts to Wnt signals via direct interaction of Barx2 with the TCF/ß-catenin complex. Stem Cells 2016;34:2169-2182.


Assuntos
Proteína Axina/genética , Montagem e Desmontagem da Cromatina , Proteínas de Homeodomínio/metabolismo , Mioblastos/metabolismo , Fator de Transcrição PAX7/metabolismo , beta Catenina/metabolismo , Acetilação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteína Axina/metabolismo , Sequência de Bases , Núcleo Celular/metabolismo , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Regulação da Expressão Gênica , Células HEK293 , Histona Acetiltransferases/metabolismo , Histona Desacetilase 1/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Íntrons/genética , Camundongos , Modelos Biológicos , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas/genética , Ligação Proteica , Processamento de Proteína Pós-Traducional/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Via de Sinalização Wnt/genética
16.
Respirology ; 22(2): 315-321, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27868302

RESUMO

BACKGROUND AND OBJECTIVE: We previously showed that alveolar macrophages from COPD patients are defective in their ability to phagocytose apoptotic cells ('efferocytosis') and that this defect is potentially linked to the sphingosine-1 phosphate (S1P) system, in particular the sphingosine-1 phosphate receptor 5 (S1PR5). In alveolar macrophages from COPD patients, S1PR5 mRNA expression levels increased and were correlated with both lung function and efferocytosis. However, it us unknown whether these changes are under epigenetic control via DNA methylation or whether DNA methylation directly modulates macrophage function. METHODS: Bisulfite sequencing was used to assess DNA methylation levels at CpG islands associated with genes encoding selected S1P system components, including sphingosine kinase 1 (SPHK1), S1PR1 and S1PR5, in alveolar macrophages from 20 COPD patients, 7 healthy smokers and 10 healthy non/ex-smokers) by methyl quantitative real-time PCR (methyl qPCR). The effect of the DNA methyltransferase inhibitor, 5-azacytidine on the efferocytosis capacity of THP-1 macrophages was assessed using flow cytometry. RESULTS: Among the S1P system genes examined, S1PR5 was the single target that showed significant changes in DNA methylation between patient groups. Alveolar macrophages isolated from COPD patients showed lower methylation levels in the same region compared to macrophages from non/ex-smokers. in vitro studies using THP-1 macrophages showed that DNA demethylation with 5-azacytidine increased the efferocytosis capacity and dose-dependently rescued the cells from the cigarette smoke-induced defect in efferocytosis. CONCLUSION: Macrophage function can be modulated epigenetically. Reduced methylation may underlie the increased expression of the S1PR5 gene in alveolar macrophages and associated defective efferocytosis in COPD.


Assuntos
Azacitidina/farmacologia , Metilação de DNA , Lisofosfolipídeos/metabolismo , Macrófagos Alveolares/fisiologia , Fagocitose , Receptores de Lisoesfingolipídeo , Esfingosina/análogos & derivados , Adulto , Idoso , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/fisiologia , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Epigenômica , Feminino , Citometria de Fluxo , Humanos , Pessoa de Meia-Idade , Fagocitose/efeitos dos fármacos , Fagocitose/fisiologia , Doença Pulmonar Obstrutiva Crônica/genética , Receptores de Lisoesfingolipídeo/genética , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/metabolismo
17.
Drug Metab Rev ; 48(1): 47-69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26828111

RESUMO

Identification of genetic polymorphisms that contribute to the risk of developing cancers is important for cancer prevention. The most recent human genome GRCh38/hg38 assembly (2013) reveals thousands of genetic polymorphisms in human uridine diphosphoglucuronosyltransferase (UGT) genes. Among these, a large number of polymorphisms at the UGT1A and UGT2B genes have been shown to modulate UGT gene promoter activity or enzymatic activity. Glucuronidation plays an important role in the metabolism and clearance of endogenous and exogenous carcinogenic compounds, and this reaction is primarily catalyzed by the UGT1A and UGT2B enzymes. Therefore, it has long been hypothesized that UGT polymorphisms that reduce the capacity to glucuronidate carcinogens and other types of cancer-promoting molecules (e.g. sex hormones) are associated with an increased risk of developing cancers. A large number of case-control studies have investigated this hypothesis and these studies identified numerous UGT polymorphisms in UGT1A and UGT2B genes as genetic risk factors for a wide variety of cancers, including bladder, breast, colorectal, endometrial, esophageal, head and neck, liver, lung, prostate, and thyroid. These UGT polymorphisms may be cancer causative polymorphisms, or be linked to as yet undefined causative polymorphisms, either in UGT genes or neighboring genes. This article presents a comprehensive review of these case-control studies, discusses current areas of uncertainty, and highlights future research directions in this field.


Assuntos
Glucuronosiltransferase/genética , Neoplasias/enzimologia , Neoplasias/genética , Predisposição Genética para Doença , Glucuronosiltransferase/metabolismo , Humanos , Polimorfismo de Nucleotídeo Único
18.
Int J Mol Sci ; 18(1)2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-28035996

RESUMO

Nearly 20 different transcripts of the human androgen receptor (AR) are reported with two currently listed as Refseq isoforms in the NCBI database. Isoform 1 encodes wild-type AR (type 1 AR) and isoform 2 encodes the variant AR45 (type 2 AR). Both variants contain eight exons: they share common exons 2-8 but differ in exon 1 with the canonical exon 1 in isoform 1 and the variant exon 1b in isoform 2. Splicing of exon 1 or exon 1b is reported to be mutually exclusive. In this study, we identified a novel exon 1b (1b/TAG) that contains an additional TAG trinucleotide upstream of exon 1b. Moreover, we identified AR transcripts in both normal and cancerous breast and prostate cells that contained either exon 1b or 1b/TAG spliced between the canonical exon 1 and exon 2, generating nine-exon AR transcripts that we have named isoforms 3a and 3b. The proteins encoded by these new AR variants could regulate androgen-responsive reporters in breast and prostate cancer cells under androgen-depleted conditions. Analysis of type 3 AR-GFP fusion proteins showed partial nuclear localization in PC3 cells under androgen-depleted conditions, supporting androgen-independent activation of the AR. Type 3 AR proteins inhibited androgen-induced growth of LNCaP cells. Microarray analysis identified a small set of type 3a AR target genes in LNCaP cells, including genes known to modulate growth and proliferation of prostate cancer (PCGEM1, PEG3, EPHA3, and EFNB2) or other types of human cancers (TOX3, ST8SIA4, and SLITRK3), and genes that are diagnostic/prognostic biomarkers of prostate cancer (GRINA3, and BCHE).


Assuntos
Neoplasias da Mama/genética , Éxons , Neoplasias da Próstata/genética , Receptores Androgênicos/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Humanos , Masculino , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , Splicing de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Androgênicos/metabolismo
19.
Mol Pharmacol ; 87(3): 442-50, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25519837

RESUMO

The human UDP glycosyltransferase (UGT) superfamily comprises four families of enzymes that catalyze the addition of sugar residues to small lipophilic chemicals. The UGT1 and UGT2 enzymes use UDP-glucuronic acid, and UGT3 enzymes use UDP-N-acetylglucosamine, UDP-glucose, and UDP-xylose to conjugate xenobiotics, including drugs and endobiotics such as metabolic byproducts, hormones, and signaling molecules. This metabolism renders the substrate more polar and more readily excreted from the body and/or functionally inactive. The fourth UGT family, called UGT8, contains only one member that, unlike other UGTs, is considered biosynthetic. UGT8 uses UDP galactose to galactosidate ceramide, a key step in the synthesis of brain sphingolipids. To date other substrates for this UGT have not been identified and there has been no suggestion that UGT8 is involved in metabolism of endo- or xenobiotics. We re-examined the functions of UGT8 and discovered that it efficiently galactosidates bile acids and drug-like bile acid analogs. UGT8 conjugates bile acids ∼60-fold more efficiently than ceramide based on in vitro assays with substrate preference deoxycholic acid > chenodeoxycholic acid > cholic acid > hyodeoxycholic acid > ursodeoxycholic acid. Activities of human and mouse UGT8 are qualitatively similar. UGT8 is expressed at significant levels in kidney and gastrointestinal tract (intestine, colon) where conjugation of bile acids is likely to be metabolically significant. We also investigate the structural determinants of UDP-galactose selectivity. Our novel findings suggest a new role for UGT8 as a modulator of bile acid homeostasis and signaling.


Assuntos
Ácidos e Sais Biliares/metabolismo , Gangliosídeo Galactosiltransferase/química , Gangliosídeo Galactosiltransferase/fisiologia , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Linhagem Celular Tumoral , Células HEK293 , Humanos , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína
20.
Drug Metab Rev ; 47(3): 335-45, 2015 08.
Artigo em Inglês | MEDLINE | ID: mdl-26289097

RESUMO

Enzymes of the human uridine diphosphate (UDP)-glycosyltransferase (UGT) superfamily typically catalyze the covalent addition of a sugar from UDP-sugar cofactors to relatively small lipophilic compounds. The sugar conjugates are often biologically less active with improved water-solubility, facilitating more effective elimination from the body. Experimental data indicate that UGT proteins exhibit differing selectivities toward various UDP-sugars. Although, three-dimensional (3D) structures of UGT proteins are required to provide insights into the UDP-sugar selectivities observed for the various UGT proteins, there are currently, no experimental structures available for human UGTs bound to UDP-sugar(s). Thus, the absence of 3D structures poses a major challenge for analyzing UDP-sugar selectivity at an atomic level. In this commentary, we highlight the application of comparative homology modeling for understanding the UDP-sugar selectivities of UGT proteins. Homology models of the C-terminal (CT) domain indicate a highly conserved structural fold across the UGT family with backbone root mean-squared deviations (rmsds) between 0.066 and 0.079 Å with respect to the UGT2B7-CT X-ray crystal structure. The models show that four residues in the terminal portion of the CT signature sequence play an important role in UDP-sugar selectivity. The N-terminal domain is less likely to be associated with UDP-sugar selectivity, although, a conserved residue, Arg-259 (UGT2B7 numbering) in the UGT 1 and 2 families may influence UDP-sugar selectivity. Overall, the models demonstrate excellent agreement with experimental observations in predicting the key residues that influence the selectivity of UDP-sugar binding.


Assuntos
Glucuronosiltransferase/metabolismo , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Proteínas de Plantas/metabolismo , Açúcares de Uridina Difosfato/metabolismo , Configuração de Carboidratos , Glucuronosiltransferase/química , Humanos , Proteínas de Plantas/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade , Especificidade por Substrato , Açúcares de Uridina Difosfato/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA