Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(49): 31259-31266, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33229553

RESUMO

Triclosan (TCS), employed as an antiseptic and disinfectant, comes into direct contact with humans through a plethora of consumer products and its rising environmental release. We have demonstrated that TCS promotes liver tumorigenesis in mice, yet the biological and molecular mechanisms by which TCS exerts its toxicity, especially in early stages of liver disease, are largely unexplored. When mice were fed a high-fat diet (HFD), we found that fatty liver and dyslipidemia are prominent early signs of liver abnormality induced by TCS. The presumably protective HFD-induced hepatic expression of the metabolic regulator fibroblast growth factor 21 (FGF21) was blunted by TCS. TCS-altered Fgf21 expression aligned with aberrant expression of genes encoding metabolic enzymes manifested as profound systemic metabolic changes that disturb homeostasis of amino acids, fatty acids, and glucose. Using a type 1 diabetic animal model, TCS potentiates and accelerates the development of steatohepatitis and fibrosis, accompanied by increased levels of hepatic lipid droplets and oxidative stress. Analysis of fecal samples revealed that HFD-fed mice exhibited a reduction in fecal species richness, and that TCS further diminished microbial diversity and shifted the bacterial community toward lower Bacteriodetes and higher Firmicutes, resembling changes in microbiota composition in nonalcoholic steatohepatitis (NASH) patients. Using reverse-genetic approaches, we demonstrate that, along with HFD, TCS induces hepatic steatosis and steatohepatitis jointly regulated by the transcription factor ATF4 and the nuclear receptor PPARα, which participate in the transcriptional regulation of the Fgf21 gene. This study provides evidence linking nutritional imbalance and exposure to TCS with the progression of NASH.


Assuntos
Fatores de Crescimento de Fibroblastos/genética , Hepatopatia Gordurosa não Alcoólica/tratamento farmacológico , PPAR alfa/genética , Triclosan/farmacologia , Animais , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Ácidos Graxos/biossíntese , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/etiologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Camundongos , Hepatopatia Gordurosa não Alcoólica/etiologia , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/patologia , Obesidade/tratamento farmacológico , Obesidade/etiologia , Obesidade/genética , Obesidade/patologia
2.
Proc Natl Acad Sci U S A ; 114(8): E1432-E1440, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28167773

RESUMO

Severe neonatal hyperbilirubinemia (SNH) and the onset of bilirubin encephalopathy and kernicterus result in part from delayed expression of UDP-glucuronosyltransferase 1A1 (UGT1A1) and the inability to metabolize bilirubin. Although there is a good understanding of the early events after birth that lead to the rapid increase in serum bilirubin, the events that control delayed expression of UGT1A1 during development remain a mystery. Humanized UGT1 (hUGT1) mice develop SNH spontaneously, which is linked to repression of both liver and intestinal UGT1A1. In this study, we report that deletion of intestinal nuclear receptor corepressor 1 (NCoR1) completely diminishes hyperbilirubinemia in hUGT1 neonates because of intestinal UGT1A1 gene derepression. Transcriptomic studies and immunohistochemistry analysis demonstrate that NCoR1 plays a major role in repressing developmental maturation of the intestines. Derepression is marked by accelerated metabolic and oxidative phosphorylation, drug metabolism, fatty acid metabolism, and intestinal maturation, events that are controlled predominantly by H3K27 acetylation. The control of NCoR1 function and derepression is linked to IKKß function, as validated in hUGT1 mice with targeted deletion of intestinal IKKß. Physiological events during neonatal development that target activation of an IKKß/NCoR1 loop in intestinal epithelial cells lead to derepression of genes involved in intestinal maturation and bilirubin detoxification. These findings provide a mechanism of NCoR1 in intestinal homeostasis during development and provide a key link to those events that control developmental repression of UGT1A1 and hyperbilirubinemia.


Assuntos
Células Epiteliais/metabolismo , Hiperbilirrubinemia Neonatal/metabolismo , Mucosa Intestinal/metabolismo , Correpressor 1 de Receptor Nuclear/metabolismo , Animais , Animais Recém-Nascidos/metabolismo , Bilirrubina/metabolismo , Glucuronosiltransferase/metabolismo , Humanos , Quinase I-kappa B/metabolismo , Fígado/metabolismo , Camundongos
3.
Annu Rev Pharmacol Toxicol ; 56: 251-72, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26738475

RESUMO

Triclosan (TCS) is a broad-spectrum antimicrobial agent that has been added to personal care products, including hand soaps and cosmetics, and impregnated in numerous different materials ranging from athletic clothing to food packaging. The constant disposal of TCS into the sewage system is creating a major environmental and public health hazard. Owing to its chemical properties of bioaccumulation and resistance to degradation, TCS is widely detected in various environmental compartments in concentrations ranging from nanograms to micrograms per liter. Epidemiology studies indicate that significant levels of TCS are detected in body fluids in all human age groups. We document here the emerging evidence--from in vitro and in vivo animal studies and environmental toxicology studies--demonstrating that TCS exerts adverse effects on different biological systems through various modes of action. Considering the fact that humans are simultaneously exposed to TCS and many TCS-like chemicals, we speculate that TCS-induced adverse effects may be relevant to human health.


Assuntos
Anti-Infecciosos/administração & dosagem , Substâncias Perigosas/efeitos adversos , Triclosan/efeitos adversos , Animais , Meio Ambiente , Humanos
4.
Mol Pharmacol ; 91(5): 545-553, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28283555

RESUMO

Hyperbilirubinemia, caused by the accumulation of unconjugated bilirubin, is one of the most common clinical diagnoses in both premature and term newborns. Owing to the fact that bilirubin is metabolized solely through glucuronidation by UDP-glucuronosyltransferase (UGT) 1A1, it is now known that immaturity of UGT1A1, in combination with the overproduction of bilirubin during the developmental stage, acts as a bottleneck to bilirubin elimination and predisposes the infant to high total serum bilirubin levels. Although neonatal jaundice is mostly benign, excessively high levels of serum bilirubin in a small percentage of newborns can cause bilirubin-induced neurologic dysfunction, potentially leading to permanent brain damage, a condition known as kernicterus Although a large portion of hyperbilirubinemia cases in newborns are associated with hemolytic diseases, we emphasize here the impaired ability of UGT1A1 to eliminate bilirubin that contributes to hyperbilirubinemia-induced neurotoxicity in the developmental stage. As a series of hereditary UGT1A1 mutations have been identified that are associated with UGT1A1 deficiency, new evidence has verified that delayed expression of UGT1A1 during the early stages of neonatal development is a tightly controlled event involving coordinated intrahepatic and extrahepatic regulation. This review recapitulates the progress that has been made in recent years in understanding the causes and physiopathology of severe hyperbilirubinemia, investigating molecular mechanisms underlying bilirubin-induced encephalopathy, and searching for potential therapies for treating pathologic hyperbilirubinemia. Several animal models have been developed to make it possible to examine bilirubin-induced neurotoxicity from multiple directions. Moreover, environmental factors that may alleviate or worsen the condition of hyperbilirubinemia are discussed.


Assuntos
Hiperbilirrubinemia Neonatal/etiologia , Animais , Bilirrubina/biossíntese , Bilirrubina/sangue , Dieta , Glucuronosiltransferase/deficiência , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Hiperbilirrubinemia Neonatal/sangue , Hiperbilirrubinemia Neonatal/induzido quimicamente , Hiperbilirrubinemia Neonatal/genética , Recém-Nascido
5.
Drug Metab Dispos ; 45(7): 748-754, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28468837

RESUMO

The gastrointestinal tract is enriched with xenobiotic processing proteins that play important roles in xenobiotic bioactivation, metabolism, and detoxification. The application of genetically modified mouse models has been instrumental in characterizing the function of xenobiotic processing genes (XPG) and their proteins in drug metabolism. Here, we report the utilization of three-dimensional crypt organoid cultures from these animal models to study intestinal drug metabolism and toxicity. With the successful culturing of crypt organoids, we profiled the abundance of Phase I and Phase II XPG expression, drug transporter gene expression, and xenobiotic nuclear receptor (XNR) gene expression. Functions of XNRs were examined by treating crypt cells with XNR prototypical agonists. Real-time quantitative polymerase chain reaction demonstrated that the representative downstream target genes were induced. These findings were validated from cultures developed from XNR-null mice. In crypt cultures isolated from Pxr-/- mice, pregnenolone 16α-carbonitrile failed to induce Cyp3a11 gene expression; similarly, WY14643 failed to induce Cyp4a10 in the Pparα-/- crypts. Crypt cultures from control (Ugt1F/F ) and intestinal epithelial cell (IEC) specific Ugt1 null mice (Ugt1ΔIEC ) were treated with camptothecin-11, an anticancer prodrug with severe intestinal toxicity that originates from insufficient UGT1A1-dependent glucuronidation of its active metabolite SN-38. In the absence of Ugt1 gene expression, Ugt1ΔIEC crypt cultures exhibit very limited production of SN-38 glucuronide, concordant with increased apoptosis in comparison with Ugt1F/F crypt cultures. This study suggests crypt organoid cultures as an effective in vitro model for studying intestinal drug metabolism and toxicity.


Assuntos
Camptotecina/análogos & derivados , Inativação Metabólica/fisiologia , Organoides/metabolismo , Animais , Apoptose/fisiologia , Camptotecina/metabolismo , Técnicas de Cultura de Células/métodos , Citocromo P-450 CYP3A/metabolismo , Expressão Gênica/fisiologia , Mucosa Intestinal/metabolismo , Irinotecano , Taxa de Depuração Metabólica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Xenobióticos/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(48): 17200-5, 2014 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-25404284

RESUMO

Triclosan [5-chloro-2-(2,4-dichlorophenoxy)phenol; TCS] is a synthetic, broad-spectrum antibacterial chemical used in a wide range of consumer products including soaps, cosmetics, therapeutics, and plastics. The general population is exposed to TCS because of its prevalence in a variety of daily care products as well as through waterborne contamination. TCS is linked to a multitude of health and environmental effects, ranging from endocrine disruption and impaired muscle contraction to effects on aquatic ecosystems. We discovered that TCS was capable of stimulating liver cell proliferation and fibrotic responses, accompanied by signs of oxidative stress. Through a reporter screening assay with an array of nuclear xenobiotic receptors (XenoRs), we found that TCS activates the nuclear receptor constitutive androstane receptor (CAR) and, contrary to previous reports, has no significant effect on mouse peroxisome proliferation activating receptor α (PPARα). Using the procarcinogen diethylnitrosamine (DEN) to initiate tumorigenesis in mice, we discovered that TCS substantially accelerates hepatocellular carcinoma (HCC) development, acting as a liver tumor promoter. TCS-treated mice exhibited a large increase in tumor multiplicity, size, and incidence compared with control mice. TCS-mediated liver regeneration and fibrosis preceded HCC development and may constitute the primary tumor-promoting mechanism through which TCS acts. These findings strongly suggest there are adverse health effects in mice with long-term TCS exposure, especially on enhancing liver fibrogenesis and tumorigenesis, and the relevance of TCS liver toxicity to humans should be evaluated.


Assuntos
Carcinogênese/efeitos dos fármacos , Carcinoma Hepatocelular/induzido quimicamente , Neoplasias Hepáticas/induzido quimicamente , Fígado/efeitos dos fármacos , Triclosan/toxicidade , Alanina Transaminase/sangue , Alanina Transaminase/metabolismo , Animais , Anti-Infecciosos Locais/toxicidade , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Fibrose/induzido quimicamente , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
7.
Mol Pharmacol ; 89(1): 84-93, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26480925

RESUMO

Bilirubin-induced neurologic dysfunction (BIND) and kernicterus has been used to describe moderate to severe neurologic dysfunction observed in children exposed to excessive levels of total serum bilirubin (TSB) during the neonatal period. Here we use a new mouse model that targets deletion of the Ugt1 locus and the Ugt1a1 gene in liver to promote hyperbilirubinemia-induced seizures and central nervous system toxicity. The accumulation of TSB in these mice leads to diffuse yellow coloration of brain tissue and a marked cerebellar hypoplasia that we characterize as kernicterus. Histologic studies of brain tissue demonstrate that the onset of severe neonatal hyperbilirubinemia, characterized by seizures, leads to alterations in myelination and glia reactivity. Kernicterus presents as axonopathy with myelination deficits at different brain regions, including pons, medulla oblongata, and cerebellum. The excessive accumulation of TSB in the early neonatal period (5 days after birth) promotes activation of the myelin basic protein (Mbp) gene with an accelerated loss of MBP that correlates with a lack of myelin sheath formation. These changes were accompanied by increased astroglial and microglial reactivity, possibly as a response to myelination injury. Interestingly, cerebellum was the area most affected, with greater myelination impairment and glia burden, and showing a marked loss of Purkinje cells and reduced arborization of the remaining ones. Thus, kernicterus in this model displays not only axonal damage but also myelination deficits and glial activation in different brain regions that are usually related to the neurologic sequelae observed after severe hyperbilirubinemia.


Assuntos
Hiperbilirrubinemia Neonatal/metabolismo , Bainha de Mielina/metabolismo , Neuroglia/metabolismo , Índice de Gravidade de Doença , Animais , Humanos , Hiperbilirrubinemia Neonatal/genética , Hiperbilirrubinemia Neonatal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas de Transporte de Monossacarídeos/deficiência , Proteínas de Transporte de Monossacarídeos/genética , Bainha de Mielina/patologia , Neuroglia/patologia
8.
Proc Natl Acad Sci U S A ; 110(47): 19143-8, 2013 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-24191041

RESUMO

Camptothecin (CPT)-11 (irinotecan) has been used widely for cancer treatment, particularly metastatic colorectal cancer. However, up to 40% of treated patients suffer from severe late diarrhea, which prevents CPT-11 dose intensification and efficacy. CPT-11 is a prodrug that is hydrolyzed by hepatic and intestinal carboxylesterase to form SN-38, which in turn is detoxified primarily through UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed glucuronidation. To better understand the mechanism associated with toxicity, we generated tissue-specific Ugt1 locus conditional knockout mouse models and examined the role of glucuronidation in protecting against irinotecan-induced toxicity. We targeted the deletion of the Ugt1 locus and the Ugt1a1 gene specifically in the liver (Ugt1(ΔHep)) and the intestine (Ugt1(ΔGI)). Control (Ugt1(F/F)), Ugt1(ΔHep), and Ugt1(ΔGI) adult male mice were treated with different concentrations of CPT-11 daily for four consecutive days. Toxicities were evaluated with regard to tissue glucuronidation potential. CPT-11-treated Ugt1(ΔHep) mice showed a similar lethality rate to the CPT-11-treated Ugt1(F/F) mice. However, Ugt1(ΔGI) mice were highly susceptible to CPT-11-induced diarrhea, developing severe and lethal mucositis at much lower CPT-11 doses, a result of the proliferative cell loss and inflammation in the intestinal tract. Comparative expression levels of UGT1A1 in intestinal tumors and normal surrounding tissue are dramatically different, providing for the opportunity to improve therapy by differential gene regulation. Intestinal expression of the UGT1A proteins is critical toward the detoxification of SN-38, whereas induction of the UGT1A1 gene may serve to limit toxicity and improve the efficacy associated with CPT-11 treatment.


Assuntos
Camptotecina/análogos & derivados , Neoplasias Colorretais/tratamento farmacológico , Ácido Glucurônico/metabolismo , Glucuronosiltransferase/metabolismo , Mucosa Intestinal/metabolismo , Animais , Camptotecina/efeitos adversos , Camptotecina/metabolismo , Camptotecina/toxicidade , Regulação Neoplásica da Expressão Gênica/genética , Glucuronosiltransferase/genética , Técnicas Histológicas , Immunoblotting , Irinotecano , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Microscopia Confocal , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
J Biol Chem ; 289(8): 4699-709, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24403077

RESUMO

Biological and signaling events that connect developmentally induced hyperbilirubinemia to bilirubin-induced neurological dysfunction (BIND) and CNS toxicity in humans are poorly understood. In mammals, UDP-glucuronosyltransferase 1A1 (UGT1A1) is the sole enzyme responsible for bilirubin glucuronidation, a rate-limiting step necessary for bilirubin metabolism and clearance. Humanized mice that express the entire UGT1 locus (hUGT1) and the UGT1A1 gene, develop neonatal hyperbilirubinemia, with 8-10% of hUGT1 mice succumbing to CNS damage, a phenotype that is presented by uncontrollable seizures. We demonstrate that neuroinflammation and reactive gliosis are prominent features of bilirubin brain toxicity, and a disturbed redox status resulting from activation of NADPH oxidase is an important contributing mechanism found in BIND. Using knock-out mice and primary brain cells, we connect a key pattern recognition receptor, Toll-like receptor 2 (TLR2), to hyperbilirubinemia-induced signaling. We illustrate a requirement for TLR2 signaling in regulating gliosis, proinflammatory mediators, and oxidative stress when neonatal mice encounter severe hyperbilirubinemia. TLR2-mediated gliosis strongly correlates with pronounced neuroinflammation in the CNS with up-regulation of TNFα, IL-1ß, and IL-6, creating a pro-inflammatory CNS environment. Gene expression and immunohistochemistry staining show that hUGT1/Tlr2(-/-) mice fail to activate glial cells, proinflammatory cytokines, and stress response genes. In addition, bilirubin-induced apoptosis was significantly enhanced by blocking TLR2 signaling indicating its anti-apoptotic property. Consequently, a higher neonatal death rate (57.1%) in hUGT1/Tlr2(-/-) mice was observed when compared with hUGT1 mice (8.7%). These results suggest that TLR2 signaling and microglia neuroinflammation are linked to a repair and/or protection mode against BIND.


Assuntos
Bilirrubina/efeitos adversos , Glucuronosiltransferase/metabolismo , Síndromes Neurotóxicas/metabolismo , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Bilirrubina/sangue , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Imunofluorescência , Deleção de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Hiperbilirrubinemia/sangue , Hiperbilirrubinemia/complicações , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/patologia , Mediadores da Inflamação/metabolismo , Kernicterus/sangue , Kernicterus/complicações , Kernicterus/genética , Kernicterus/patologia , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , NADPH Oxidases/metabolismo , NF-kappa B/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neuroglia/patologia , Síndromes Neurotóxicas/sangue , Síndromes Neurotóxicas/complicações , Síndromes Neurotóxicas/genética , Oxirredução/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Receptor 2 Toll-Like/deficiência , Receptor 2 Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
10.
Hepatology ; 56(2): 658-67, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22371261

RESUMO

UNLABELLED: In humanized UDP glucuronosyltransferase-1 (hUGT1) mice that express the entire UGT1 locus, the maternal hepatic UGT1A genes are dramatically induced 12-14 days after conception. Steroid induction of the UGT1A1 gene indicates that xenobiotic sensors, such as the pregnane X receptor (PXR) and constitutive androstane receptor (CAR), may underlie the induction process. In contrast, neonatal hUGT1 mice display severe hyperbilirubinemia, with limited expression of the UGT1A genes. This study identifies PXR as both a positive and negative regulator of the UGT1A1 gene. Pregnancy hormones, in particular the glucocorticoids, target PXR as a positive regulator of human glucuronidation. Employing reverse genetics, where PXR has been genetically deleted, hUGT1/Pxr(-/-) mice show limited induction of the liver UGT1A genes during pregnancy, whereas the exact opposite occurs in newborn mice. Neonatal hUGT1 mice show delayed expression of hepatic UGT1A1 and are severely hyperbilirubinemic. However, in hUGT1/Pxr(-/-) mice, hyperbilirubinemia is greatly reduced due to induction of hepatic UGT1A1. Thus, PXR serves to repress UGT1A1 gene expression during development. Transcriptional silencing of the UGT1A1 gene was relieved in neonatal hUGT1 hepatocytes through interruption of PXR by small interfering RNA. CONCLUSION: PXR is a key regulator of pregnancy induced glucuronidation capacity in addition to modulating the severity of neonatal jaundice.


Assuntos
Ácido Glucurônico/metabolismo , Glucuronosiltransferase/metabolismo , Icterícia Neonatal/metabolismo , Fígado , Complicações na Gravidez/metabolismo , Receptores de Esteroides/metabolismo , Animais , Animais Recém-Nascidos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glucuronosiltransferase/genética , Hepatócitos/citologia , Hepatócitos/fisiologia , Humanos , Recém-Nascido , Fígado/embriologia , Fígado/crescimento & desenvolvimento , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Gravidez , Receptor de Pregnano X , Cultura Primária de Células , RNA Interferente Pequeno/genética , Esteroides/fisiologia
11.
J Steroid Biochem Mol Biol ; 109(3-5): 247-53, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18467088

RESUMO

Conjugation by UDP-Glucuronosyltransferase (UGT) is the major pathway of androgen metabolism and elimination in the human. High concentrations of glucuronide conjugates of androsterone (ADT) and androstane-3alpha,17beta-diol (3alpha-diol) are present in circulation and several studies over the last 30 years have concluded that the serum levels of these metabolites might reflect the androgen metabolism in several tissues, including the liver and androgen target tissues. Three UGT2B enzymes are responsible for the conjugation of DHT and its metabolites ADT and 3alpha-diol: UGT2B7, B15 and B17. UGT2B7 is expressed in the liver and skin whereas UGT2B15 and B17 were found in the liver, prostate and skin. Very specific antibodies against each UGT2B enzyme have been obtained and used for immunohistochemical studies in the human prostate. It was shown that UGT2B17 is expressed in basal cells whereas UGT2B15 is only localized in luminal cells, where it inactivates DHT. By using LNCaP cells, we have also demonstrated that the expression and activity of UGT2B15 and B17 are modulated by several endogenous prostate factors including androgen. Finally, to study the physiological role of UGT2B enzymes, transgenic mice bearing the human UGT2B15 gene were recently obtained. A decrease in reproductive tissue weight from transgenic animals compared to those from control animals was observed. In conclusion, the conjugation by UGT2B7, B15 and B17, which represents a non-reversible step in androgen metabolism, is an important means by which androgens are regulated locally. It is also postulated that UGT enzymes protect the tissue from deleteriously high concentrations of active androgen.


Assuntos
Androgênios/metabolismo , Glucuronosiltransferase/metabolismo , Transdução de Sinais , Animais , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/classificação , Glucuronosiltransferase/genética , Humanos , Polimorfismo Genético/genética , Esteroides/sangue
12.
Biochem Pharmacol ; 110-111: 37-46, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27060662

RESUMO

Humanized UDP-glucuronosyltransferase (UGT)-1 (hUGT1) mice encode the UGT1 locus including the UGT1A1 gene. During neonatal development, delayed expression of the UGT1A1 gene leads to hyperbilirubinemia as determined by elevated levels of total serum bilirubin (TSB). We show in this report that the redox-sensitive NF-κB pathway is crucial for intestinal expression of the UGT1A1 gene and control of TSB levels. Targeted deletion of IKKß in intestinal epithelial cells (hUGT1/Ikkß(ΔIEC) mice) leads to greater neonatal accumulation of TSB than observed in control hUGT1/Ikkß(F/F) mice. The elevation in TSB levels in hUGT1/Ikkß(ΔIEC) mice correlates with a reduction in intestinal UGT1A1 expression. As TSB levels accumulate in hUGT1/Ikkß(ΔIEC) mice during the neonatal period, the increase over that observed in hUGT1/Ikkß(F/F) mice leads to weight loss, seizures and eventually death. Bilirubin accumulates in brain tissue from hUGT1/Ikkß(ΔIEC) mice inducing an inflammatory state as shown by elevated TNFα, IL-1ß and IL-6, all of which can be prevented by neonatal induction of hepatic or intestinal UGT1A1 and lowering of TSB levels. Altering the redox state of the intestines by oral administration of cadmium or arsenic to neonatal hUGT1/Ikkß(F/F) and hUGT1/Ikkß(ΔIEC) mice leads to induction of UGT1A1 and a dramatic reduction in TSB levels. Microarray analysis following arsenic treatment confirms upregulation of oxidation-reduction processes and lipid metabolism, indicative of membrane repair or synthesis. Our findings indicate that the redox state in intestinal epithelial cells during development is important in maintaining UGT1A1 gene expression and control of TSB levels.


Assuntos
Arsênio/farmacologia , Cádmio/farmacologia , Glucuronosiltransferase/genética , Hiperbilirrubinemia/prevenção & controle , NF-kappa B/genética , Convulsões/prevenção & controle , Animais , Animais Recém-Nascidos , Bilirrubina/antagonistas & inibidores , Bilirrubina/biossíntese , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucuronosiltransferase/metabolismo , Humanos , Hiperbilirrubinemia/genética , Hiperbilirrubinemia/metabolismo , Hiperbilirrubinemia/patologia , Quinase I-kappa B/deficiência , Quinase I-kappa B/genética , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/patologia , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Convulsões/genética , Convulsões/metabolismo , Convulsões/patologia , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Redução de Peso/efeitos dos fármacos
13.
Cell Mol Gastroenterol Hepatol ; 2(1): 63-76.e5, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26807433

RESUMO

BACKGROUND & AIMS: The UDP-glucuronosyltransferases (UGTs) are part of the cells machinery that protects the tissues from a toxicant insult by environmental and host cell metabolites. We have investigated the mechanism behind tumor growth and UGT repression. METHODS: We initially silenced the Ugt1 locus in human colon cell lines and investigated markers and responses linked to p53 activation. To examine the role of the Ugt1 locus in p53-directed apoptosis and tumorigenesis, experiments were conducted to induce acute colon inflammation and chemical induced colon cancer in mice where we have selectively deleted the Ugt1 locus in the intestinal epithelial cells (Ugt1ΔIEC mice). RESULTS: Knockdown of the UGT1A proteins by RNAi in human colon cancer cells and knockout of the Ugt1 locus in intestinal crypt stem cells reduces phosphorylated p53 activation and compromises the ability of p53 to control apoptosis. Targeted deletion of intestinal Ugt1 expression in Ugt1ΔIEC mice represses colon inflammation-induced p53 production and pro-apoptotic protein activation. When we induced colon cancer, the size and number of the tumors were significantly greater in the Ugt1ΔIEC mice when compared to wild type mice. Furthermore, analysis of endoplasmic reticulum (ER) stress-related markers indicated that lack of UGT1A expression causes higher ER stress in intestinal epithelial cells and tissue, which may account for the lower expression of p53. CONCLUSIONS: Our results demonstrate that UGT1A expression is required to maintain and sustain p53 activation in stress-induced colon epithelial cells and has a significant impact on p53-mediated apoptosis and tumor suppression, thus protecting the colon tissue from neoplastic transformation.

14.
Methods Enzymol ; 400: 75-91, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16399344

RESUMO

UDP-glucuronosyltransferases (UGTs) catalyze a major metabolic pathway initiating the transfer of glucuronic acid from uridine 5'-diphosphoglucuronic acid to endogenous and exogenous substances. Endogenous substances include bile acids, steroids, phenolic neurotransmitters, and bilirubin. Xenobiotic substances include dietary substances, therapeutics, and environmental compounds. The versatility in the selection of substrates for glucuronidation results from the multiplicity of the UGTs in addition to the ability of these genes to be regulated. UDP-glucuronosyltransferase 1A1 (UGT1A1), responsible for the glucuronidation of bilirubin, is controlled in a tissue-specific manner and can be regulated following environmental exposure. This chapter describes materials and methods for the examination of molecular interactions that control UGT1A1 expression and induction in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Using an in vitro cell culture system, we mapped a regulatory sequence that contains a xenobiotic response element core sequence in the enhancer region of the UGT1A1 gene. Similar to regulation of CYP1A1, the transcriptional activation of UGT1A1 by TCDD is mediated through the aryl hydrocarbon receptor.


Assuntos
Regulação Enzimológica da Expressão Gênica , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Western Blotting , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Indução Enzimática/fisiologia , Genótipo , Glucuronosiltransferase/efeitos dos fármacos , Humanos , Dibenzodioxinas Policloradas/toxicidade , Regiões Promotoras Genéticas , Teratogênicos/toxicidade , Ativação Transcricional
15.
Toxicol In Vitro ; 19(2): 275-87, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15649641

RESUMO

CYP1A is a subfamily of cytochrome P450 enzymes involved in the metabolism of numerous therapeutic drugs and in the bioactivation of procarcinogens to mutagens. Because of their diverse metabolic capacities, differences in expression of CYP1A enzymes may profoundly influence drug-drug interactions and drug or carcinogen activation and detoxification. Here, we demonstrate that cell-based bioassays are capable of identifying xenobiotics that either alter aryl hydrocarbon receptor (AhR)-mediated CYP1A levels or produce inhibition of enzyme activity. To assess induction, a stable cell line harboring a luciferase reporter driven by multiple dioxin response elements (DREs) was developed. Using this cell line, AhR agonists and antagonists were identified among drugs, dietary agents, and environmental compounds. Of the chemicals examined, the therapeutic agent omeprazole induced reporter gene activity 12.5+/-0.41 fold above control, whereas the phytochemical, chrysin and environmental pollutant, benzanthracene enhanced luciferase activity 3.3+/-0.03 and 28.7+/-1.7 fold above control, respectively. Several natural products, polychlorinated biphenyls (PCBs) and polycyclic aromatic hydrocarbons (PAHs) prevented TCDD-mediated increases in luciferase expression. For example, the botanical kava inhibited TCDD-mediated induction by 88%. Northern blot analyses of CYP1A1 in HepG2 cells treated with similar agents validated results generated in the stable cell line. The stable cells were further used to identify inhibitors of CYP1A-mediated metabolism. Resveratrol and furafylline exhibited dose-dependent decreases in CYP1A1 and CYP1A2 enzyme activities with IC50 values of 1.89 and 0.79 microM, respectively. In summary, chemicals that possess the ability to alter CYP1A expression or inhibit CYP1A enzyme activities can be rapidly identified with the cell-based bioassays described here.


Assuntos
Bioensaio/métodos , Linhagem Celular Tumoral , Citocromo P-450 CYP1A1/biossíntese , Citocromo P-450 CYP1A2/biossíntese , Hepatócitos/enzimologia , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A2/genética , Relação Dose-Resposta a Droga , Indução Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/classificação , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica , Genes Reporter/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Humanos , Luciferases/genética , Receptores de Hidrocarboneto Arílico/agonistas , Receptores de Hidrocarboneto Arílico/antagonistas & inibidores , Xenobióticos/classificação , Xenobióticos/farmacologia
16.
Biochem Pharmacol ; 63(7): 1353-9, 2002 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11960612

RESUMO

Flavin-containing monooxygenases (FMOs) comprise a multi-gene family and catalyze the oxygenation of soft nucleophilic sulfur, nitrogen, phosphorus, and selenium in xenobiotics. Previous studies have demonstrated that FMO is regulated developmentally and by the administration of certain steroid hormones. This study examined the expression of FMO form 1 in the livers and kidneys of fetal and neonatal rabbits, from day 25 of gestation through 3 weeks of age, by assaying FMO1 mRNA and protein levels, as well as catalytic activity. FMO1 mRNA and protein expression and FMO catalytic activity were present in fetal livers at the earliest time point measured (day 25 of gestation), although at levels approximately 10% of that found in adult livers. Hepatic FMO1 mRNA levels increased during and after gestation; levels were not significantly different from those measured in adult male livers. FMO1 protein content and activity rose rapidly after birth to reach 70-80% of adult levels by 3 weeks of age. The expression of FMO1 in fetal and neonatal kidneys was markedly lower than in liver. FMO1 mRNA levels never averaged more than 3.4% of adult male liver levels, but did not differ from adult kidney levels at any of the points measured. Protein levels and enzyme activity rose significantly after birth to approximately 30% of the level in adult kidneys by 3 weeks of age. The early developmental appearance of FMO1 suggests a possible role in the metabolism of xenobiotics through transplacental or lactational exposures.


Assuntos
Rim/enzimologia , Fígado/enzimologia , Oxigenases/metabolismo , Animais , Desenvolvimento Embrionário e Fetal , Feminino , Feto/enzimologia , Rim/embriologia , Fígado/embriologia , Troca Materno-Fetal , Gravidez , Coelhos
17.
PLoS One ; 7(6): e37705, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22761658

RESUMO

Triclocarban (3,4,4'-trichlorocarbanilide, TCC) is used as a broad-based antimicrobial agent that is commonly added to personal hygiene products. Because of its extensive use in the health care industry and resistance to degradation in sewage treatment processes, TCC has become a significant waste product that is found in numerous environmental compartments where humans and wildlife can be exposed. While TCC has been linked to a range of health and environmental effects, few studies have been conducted linking exposure to TCC and induction of xenobiotic metabolism through regulation by environmental sensors such as the nuclear xenobiotic receptors (XenoRs). To identify the ability of TCC to activate xenobiotic sensors, we monitored XenoR activities in response to TCC treatment using luciferase-based reporter assays. Among the XenoRs in the reporter screening assay, TCC promotes both constitutive androstane receptor (CAR) and estrogen receptor alpha (ERα) activities. TCC treatment to hUGT1 mice resulted in induction of the UGT1A genes in liver. This induction was dependent upon the constitutive active/androstane receptor (CAR) because no induction occurred in hUGT1Car(-/-) mice. Induction of the UGT1A genes by TCC corresponded with induction of Cyp2b10, another CAR target gene. TCC was demonstrated to be a phenobarbital-like activator of CAR in receptor-based assays. While it has been suggested that TCC be classified as an endocrine disruptor, it activates ERα leading to induction of Cyp1b1 in female ovaries as well as in promoter activity. Activation of ERα by TCC in receptor-based assays also promotes induction of human CYP2B6. These observations demonstrate that TCC activates nuclear xenobiotic receptors CAR and ERα both in vivo and in vitro and might have the potential to alter normal physiological homeostasis. Activation of these xenobiotic-sensing receptors amplifies gene expression profiles that might represent a mechanistic base for potential human health effects from exposure to TCC.


Assuntos
Carbanilidas/farmacologia , Receptor alfa de Estrogênio/metabolismo , Glucuronosiltransferase/fisiologia , Microssomos Hepáticos/efeitos dos fármacos , Ovário/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/metabolismo , Xenobióticos/metabolismo , Animais , Anti-Infecciosos Locais/farmacologia , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Western Blotting , Células Cultivadas , Receptor Constitutivo de Androstano , Citocromo P-450 CYP1B1 , Citocromo P-450 CYP2B6 , Receptor alfa de Estrogênio/genética , Feminino , Regulação da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Inativação Metabólica , Camundongos , Microssomos Hepáticos/metabolismo , Ovário/metabolismo , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Citoplasmáticos e Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Biol Chem ; 282(12): 8749-58, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17259171

RESUMO

The formation of beta-D-glucopyranosides (glucuronides) by the UDP-glucuronosyltransferases (UGTs) is a significant metabolic pathway that facilitates the elimination of small hydrophobic molecules such as drugs, dietary constituents, steroids, and bile acids. We elucidate here that an anti-oxidative response leads to induction of UGT1A1 through the Nrf2-Keap1 pathway. When human HepG2 cells were treated with the prooxidants tert-butylhydroquinone and beta-naphthoflavone, cellular UGT1A1 glucuronidation activities were increased. The induction of UGT1A1 proceeded following the overexpression of Nrf2 and was blocked following overexpression of Keap1, demonstrating that Keap1 suppresses Nrf2 activation of the UGT1A1 gene. Loss of function analysis for Nrf2 conducted by small interfering RNA revealed that induction of UGT1A1 was not seen in Nrf2 knock-out cells. To examine the contribution of oxidants toward the regulation of human UGT1A1 in vivo, transgenic mice bearing the human UGT1 locus (Tg-UGT1) were treated with tert-butylhydroquinone. Human UGT1A1 was markedly increased in small and large intestines as well as in liver. Gene mapping experiments including transfections of UGT1A1 reporter gene constructs into HepG2 cells coupled with functional analysis of Nrf2 expression and binding to anti-oxidant-response elements (ARE) resulted in identification of an ARE in the phenobarbital-response enhancer module region of the UGT1A1 gene. The ARE flanks the recently identified Ah receptor xenobiotic-responsive element. The results suggest that Nrf2-Keap1-dependent UGT1A1 induction by prooxidants might represent a key adaptive response to cellular oxidative stress that defends against a variety of environmental insults, including electrophile attacks and chemical carcinogenesis.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas do Citoesqueleto/fisiologia , Regulação da Expressão Gênica , Glucuronosiltransferase/biossíntese , Glucuronosiltransferase/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Fator 2 Relacionado a NF-E2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas do Citoesqueleto/genética , Glucuronidase/metabolismo , Hepatócitos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Oxidantes/química , Oxidantes/metabolismo , Fenobarbital/farmacologia
19.
Drug Metab Dispos ; 35(3): 419-27, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17151188

RESUMO

The UDP-glucuronosyltransferase (UGT) 1A genes in humans have been shown to be differentially regulated in a tissue-specific fashion. Transgenic mice carrying the human UGT1 locus (Tg-UGT1) were recently created, demonstrating that expression of the nine UGT1A genes closely resembles the patterns of expression observed in human tissues. In the present study, UGT1A1, UGT1A3, UGT1A4, and UGT1A6 have been identified as targets of the peroxisome proliferator-activated receptor (PPAR) alpha in human hepatocytes and Tg-UGT1 mice. Oral administration of the PPARalpha agonist 4-chloro-6-(2,3-xylidino)-2-pyrimidinylthioacetic acid (pirinixic acid, WY-14643) to Tg-UGT1 mice led to induction of these proteins in either the liver, gastrointestinal tract, or kidney. The levels of induced UGT1A3 gene transcripts in liver and UGT1A4 protein in small intestine correlated with induced lamotrigine glucuronidation activity in these tissues. With UGT1A3 previously identified as the major human enzyme involved in human C24-glucuronidation of lithocholic acid (LCA), the dramatic induction of liver UGT1A3 RNA in Tg-UGT1 mice was consistent with the formation of LCA-24G in plasma. Furthermore, PPAR-responsive elements (PPREs) were identified flanking the UGT1A1, UGT1A3, and UGT1A6 genes by a combination of site-directed mutagenesis, specific binding to PPARalpha and retinoic acid X receptor alpha, and functional response of the concatenated PPREs in HepG2 cells overexpressing PPARalpha. In conclusion, these results suggest that oral fibrate treatment in humans will induce the UGT1A family of proteins in the gastrointestinal tract and liver, influencing bile acid glucuronidation and first-pass metabolism of other drugs that are taken concurrently with hypolipidemic therapy.


Assuntos
Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , PPAR alfa/agonistas , Proliferadores de Peroxissomos/farmacologia , Pirimidinas/farmacologia , Animais , Anticonvulsivantes/metabolismo , Linhagem Celular , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/enzimologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/enzimologia , Humanos , Lamotrigina , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Camundongos , Camundongos Transgênicos , Microssomos/efeitos dos fármacos , Microssomos/enzimologia , Triazinas/metabolismo
20.
Drug Metab Dispos ; 33(1): 38-48, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15466163

RESUMO

Exposure to certain xenochemicals can alter the catalytic activity of the major drug-metabolizing enzyme, CYP3A4, either by enhancing expression of this cytochrome P450 or inhibiting its activity. Such alterations can result in adverse consequences stemming from drug-drug interactions. A simplified and reliable tool for detecting the ability of candidate drugs to alter CYP3A4 levels or inhibit catalytic activity was developed by stable integration of human pregnane X receptor and a luciferase vector harboring the CYP3A4 enhancers. Treatment of stable transformants, namely DPX-2, with various concentrations of inducers including rifampicin, mifepristone, troglitazone, methoxychlor, and kava produced dose-dependent increases in luciferase expression (between 2- and 40-fold above dimethyl sulfoxide-treated cells). Northern blot analyses of CYP3A4 mRNA in DPX-2 cells exhibited a good correlation to results generated with the reporter gene assay (r(2) = 0.5, p < 0.01). Induction of CYP3A4 protein was examined by measuring catalytic activity with the CYP3A4 substrate, luciferin 6' benzyl ether (luciferin BE). Metabolism of luciferin BE by DPX-2 cells was enhanced 5.2-fold above dimethyl sulfoxide-treated cells by treatment with rifampicin. Constitutive androstane receptor-mediated regulation of CYP3A4 protein was addressed by measuring catalytic activity in a separate cell line over-expressing this receptor. Phenobarbital and dexamethasone produced 1.5- and 2.0-fold increases, respectively, above control in luciferin BE metabolism. To determine the utility of DPX-2 cells for identifying inhibitors of CYP3A4 catabolism, luciferin BE activity was measured in the presence of various concentrations of ketoconazole, erythromycin, or kava. These agents exhibited dose-dependent decreases in CYP3A4 activity with IC(50) values of 0.3 microM for ketoconazole, 108 microM for erythromycin, and 15.5 microg/ml for kava. Collectively, DPX-2 cells were used to identify xenobiotics that induce or inhibit CYP3A4 in a high throughput manner, demonstrating their applicability to early-stage drug development.


Assuntos
Bioensaio/métodos , Inibidores das Enzimas do Citocromo P-450 , Sistema Enzimático do Citocromo P-450/biossíntese , Linhagem Celular , Linhagem Celular Tumoral , Células Cultivadas , Citocromo P-450 CYP3A , Sistema Enzimático do Citocromo P-450/fisiologia , Relação Dose-Resposta a Droga , Interações Medicamentosas/fisiologia , Indução Enzimática/efeitos dos fármacos , Indução Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Xenobióticos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA