Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Lipid Res ; 61(5): 696-706, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32170024

RESUMO

The pregnane X receptor (PXR) is a nuclear receptor that can be activated by numerous drugs and xenobiotic chemicals. PXR thereby functions as a xenobiotic sensor to coordinately regulate host responses to xenobiotics by transcriptionally regulating many genes involved in xenobiotic metabolism. We have previously reported that PXR has pro-atherogenic effects in animal models, but how PXR contributes to atherosclerosis development in different tissues or cell types remains elusive. In this study, we generated an LDL receptor-deficient mouse model with myeloid-specific PXR deficiency (PXRΔMyeLDLR-/-) to elucidate the role of macrophage PXR signaling in atherogenesis. The myeloid PXR deficiency did not affect metabolic phenotypes and plasma lipid profiles, but PXRΔMyeLDLR-/- mice had significantly decreased atherosclerosis at both aortic root and brachiocephalic arteries compared with control littermates. Interestingly, the PXR deletion did not affect macrophage adhesion and migration properties, but reduced lipid accumulation and foam cell formation in the macrophages. PXR deficiency also led to decreased expression of the scavenger receptor CD36 and impaired lipid uptake in macrophages of the PXRΔMyeLDLR-/- mice. Further, RNA-Seq analysis indicated that treatment with a prototypical PXR ligand affects the expression of many atherosclerosis-related genes in macrophages in vitro. These findings reveal a pivotal role of myeloid PXR signaling in atherosclerosis development and suggest that PXR may be a potential therapeutic target in atherosclerosis management.


Assuntos
Aterosclerose/imunologia , Aterosclerose/metabolismo , Macrófagos/metabolismo , Receptor de Pregnano X/deficiência , Receptores de LDL/deficiência , Animais , Antígenos CD36/metabolismo , Células Espumosas/citologia , Células Espumosas/metabolismo , Regulação da Expressão Gênica , Lipídeos/sangue , Camundongos , Fenótipo
2.
J Hepatol ; 70(5): 930-940, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30677459

RESUMO

BACKGROUND & AIMS: The most prescribed non-nucleoside reverse transcriptase inhibitor, efavirenz, has been associated with elevated risk of dyslipidemia and hepatic steatosis in HIV-infected patients but the underlying mechanisms remain elusive. Herein, we investigated the role of pregnane X receptor (PXR) in mediating the adverse effects of efavirenz on lipid homeostasis. METHODS: Cell-based reporter assays, primary cell culture, and multiple mouse models including conditional knockout and humanized mice were combined to study the impact of efavirenz on PXR activities and lipid homeostasis in vitro and in vivo. A novel liver-specific Pxr knockout mouse model was also generated to determine the contribution of hepatic PXR signaling to efavirenz-elicited dyslipidemia and hepatic steatosis. RESULTS: We found that efavirenz is a potent PXR-selective agonist that can efficiently activate PXR and induce its target gene expression in vitro and in vivo. Treatment with efavirenz-induced hypercholesterolemia and hepatic steatosis in mice but deficiency of hepatic PXR abolished these adverse effects. Interestingly, efavirenz-mediated PXR activation regulated the expression of several key hepatic lipogenic genes including fatty acid transporter CD36 and cholesterol biosynthesis enzyme squalene epoxidase (SQLE), leading to increased lipid uptake and cholesterol biosynthesis in hepatic cells. While CD36 is a known PXR target gene, we identified a DR-2-type of PXR-response element in the SQLE promoter and established SQLE as a direct transcriptional target of PXR. Since PXR exhibits considerable differences in its pharmacology across species, we also confirmed these findings in PXR-humanized mice and human primary hepatocytes. CONCLUSIONS: The widely prescribed antiretroviral drug efavirenz induces hypercholesterolemia and hepatic steatosis by activating PXR signaling. Activation of PXR should be taken into consideration for patients undergoing long-term treatment with PXR agonistic antiretroviral drugs. LAY SUMMARY: Efavirenz is widely prescribed for HIV-infected patients but has some side effects. It can increase lipid levels in patients' blood and liver. Here we show that efavirenz can activate a unique liver protein called PXR which mediates the adverse effects of efavirenz on lipid levels in mouse models.


Assuntos
Benzoxazinas/efeitos adversos , Fígado Gorduroso/induzido quimicamente , Hipercolesterolemia/induzido quimicamente , Receptor de Pregnano X/agonistas , Inibidores da Transcriptase Reversa/efeitos adversos , Alcinos , Animais , Antígenos CD36/fisiologia , Colesterol/biossíntese , Ciclopropanos , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Pregnano X/fisiologia , Transdução de Sinais/fisiologia , Esqualeno Mono-Oxigenase/fisiologia
3.
Arterioscler Thromb Vasc Biol ; 38(7): 1468-1478, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29724817

RESUMO

OBJECTIVE: The Wnt/ß-catenin signaling is an ancient and evolutionarily conserved pathway that regulates essential aspects of cell differentiation, proliferation, migration and polarity. Canonical Wnt/ß-catenin signaling has also been implicated in the pathogenesis of atherosclerosis. Macrophage is one of the major cell types involved in the initiation and progression of atherosclerosis, but the role of macrophage ß-catenin in atherosclerosis remains elusive. This study aims to investigate the impact of ß-catenin expression on macrophage functions and atherosclerosis development. APPROACH AND RESULTS: To investigate the role of macrophage canonical Wnt/ß-catenin signaling in atherogenesis, we generated ß-cateninΔmyeLDLR-/- mice (low-density lipoprotein receptor-deficient mice with myeloid-specific ß-catenin deficiency). As expected, deletion of ß-catenin decreased macrophage adhesion and migration properties in vitro. However, deficiency of ß-catenin significantly increased atherosclerotic lesion areas in the aortic root of LDLR-/- (low-density lipoprotein receptor-deficient) mice without affecting the plasma lipid levels and atherosclerotic plaque composition. Mechanistic studies revealed that ß-catenin can regulate activation of STAT (signal transducer and activator of transcription) pathway in macrophages, and ablation of ß-catenin resulted in STAT3 downregulation and STAT1 activation, leading to elevated macrophage inflammatory responses and increased atherosclerosis. CONCLUSIONS: This study demonstrates a critical role of myeloid ß-catenin expression in atherosclerosis by modulating macrophage inflammatory responses.


Assuntos
Aorta/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , Macrófagos/metabolismo , Placa Aterosclerótica , Receptores de LDL/deficiência , beta Catenina/deficiência , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/patologia , Adesão Celular , Movimento Celular , Dieta Hiperlipídica , Modelos Animais de Doenças , Progressão da Doença , Lipídeos/sangue , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Células RAW 264.7 , Receptores de LDL/genética , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT3/metabolismo , Fatores de Tempo , Via de Sinalização Wnt , beta Catenina/genética
4.
Stem Cells ; 34(7): 1883-95, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26991836

RESUMO

IκB kinase ß (IKKß), a central coordinator of inflammation through activation of nuclear factor-κB, has been identified as a potential therapeutic target for the treatment of obesity-associated metabolic dysfunctions. In this study, we evaluated an antisense oligonucleotide (ASO) inhibitor of IKKß and found that IKKß ASO ameliorated diet-induced metabolic dysfunctions in mice. Interestingly, IKKß ASO also inhibited adipocyte differentiation and reduced adiposity in high-fat (HF)-fed mice, indicating an important role of IKKß signaling in the regulation of adipocyte differentiation. Indeed, CRISPR/Cas9-mediated genomic deletion of IKKß in 3T3-L1 preadipocytes blocked these cells differentiating into adipocytes. To further elucidate the role of adipose progenitor IKKß signaling in diet-induced obesity, we generated mice that selectively lack IKKß in the white adipose lineage and confirmed the essential role of IKKß in mediating adipocyte differentiation in vivo. Deficiency of IKKß decreased HF-elicited adipogenesis in addition to reducing inflammation and protected mice from diet-induced obesity and insulin resistance. Further, pharmacological inhibition of IKKß also blocked human adipose stem cell differentiation. Our findings establish IKKß as a pivotal regulator of adipogenesis and suggest that overnutrition-mediated IKKß activation serves as an initial signal that triggers adipose progenitor cell differentiation in response to HF feeding. Inhibition of IKKß with antisense therapy may represent as a novel therapeutic approach to combat obesity and metabolic dysfunctions. Stem Cells 2016;34:1883-1895.


Assuntos
Adipócitos/patologia , Linhagem da Célula , Quinase I-kappa B/metabolismo , Síndrome Metabólica/tratamento farmacológico , Terapia de Alvo Molecular , Obesidade/tratamento farmacológico , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipogenia/efeitos dos fármacos , Animais , Linhagem da Célula/efeitos dos fármacos , Dieta , Fígado Gorduroso/patologia , Deleção de Genes , Técnicas de Silenciamento de Genes , Humanos , Quinase I-kappa B/deficiência , Insulina/farmacologia , Masculino , Síndrome Metabólica/patologia , Camundongos Endogâmicos C57BL , Obesidade/patologia , Oligonucleotídeos Antissenso/farmacologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
6.
Front Endocrinol (Lausanne) ; 15: 1398462, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38957441

RESUMO

Background: Cannabidiol (CBD), a non-psychoactive phytocannabinoid of cannabis, is therapeutically used as an analgesic, anti-convulsant, anti-inflammatory, and anti-psychotic drug. There is a growing concern about the adverse side effects posed by CBD usage. Pregnane X receptor (PXR) is a nuclear receptor activated by a variety of dietary steroids, pharmaceutical agents, and environmental chemicals. In addition to the role in xenobiotic metabolism, the atherogenic and dyslipidemic effects of PXR have been revealed in animal models. CBD has a low affinity for cannabinoid receptors, thus it is important to elucidate the molecular mechanisms by which CBD activates cellular signaling and to assess the possible adverse impacts of CBD on pro-atherosclerotic events in cardiovascular system, such as dyslipidemia. Objective: Our study aims to explore the cellular and molecular mechanisms by which exposure to CBD activates human PXR and increases the risk of dyslipidemia. Methods: Both human hepatic and intestinal cells were used to test if CBD was a PXR agonist via cell-based transfection assay. The key residues within PXR's ligand-binding pocket that CBD interacted with were investigated using computational docking study together with site-directed mutagenesis assay. The C57BL/6 wildtype mice were orally fed CBD in the presence of PXR antagonist resveratrol (RES) to determine how CBD exposure could change the plasma lipid profiles in a PXR-dependent manner. Human intestinal cells were treated with CBD and/or RES to estimate the functions of CBD in cholesterol uptake. Results: CBD was a selective agonist of PXR with higher activities on human PXR than rodents PXRs and promoted the dissociation of human PXR from nuclear co-repressors. The key amino acid residues Met246, Ser247, Phe251, Phe288, Trp299, and Tyr306 within PXR's ligand binding pocket were identified to be necessary for the agonistic effects of CBD. Exposure to CBD increased the circulating total cholesterol levels in mice which was partially caused by the induced expression levels of the key intestinal PXR-regulated lipogenic genes. Mechanistically, CBD induced the gene expression of key intestinal cholesterol transporters, which led to the increased cholesterol uptake by intestinal cells. Conclusion: CBD was identified as a selective PXR agonist. Exposure to CBD activated PXR signaling and increased the atherogenic cholesterol levels in plasma, which partially resulted from the ascended cholesterol uptake by intestinal cells. Our study provides potential evidence for the future risk assessment of CBD on cardiovascular disease, such as dyslipidemia.


Assuntos
Canabidiol , Colesterol , Camundongos Endogâmicos C57BL , Receptor de Pregnano X , Receptor de Pregnano X/metabolismo , Animais , Humanos , Camundongos , Canabidiol/farmacologia , Colesterol/metabolismo , Masculino , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Simulação de Acoplamento Molecular
7.
Mol Pharmacol ; 83(6): 1190-9, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23519392

RESUMO

Human immunodeficiency virus (HIV) protease inhibitors (PIs) have been used successfully in extending the life span of people infected with HIV. The use of PIs has also been associated with dyslipidemia and an increased risk of cardiovascular disease, but the underlying mechanisms remain elusive. Several PIs have been implicated in activating the nuclear receptor pregnane X receptor (PXR), which acts as a xenobiotic sensor to regulate xenobiotic metabolism in the liver and intestine. Recent studies indicate that PXR may also play an important role in the regulation of lipid homeostasis. In the present study, we identified amprenavir, a widely used HIV PI, as a potent PXR-selective agonist. Computational docking studies combined with site-direct mutagenesis identified several key residues within the ligand-binding pocket of PXR that constitute points of interaction with amprenavir. Amprenavir efficiently activated PXR and induced PXR target gene expression in vitro and in vivo. Short-term exposure to amprenavir significantly increased plasma total cholesterol and atherogenic low-density lipoprotein cholesterol levels in wild-type mice, but not in PXR-deficient mice. Amprenavir-mediated PXR activation stimulated the expression of several key intestinal genes involved in lipid homeostasis. These findings provide critical mechanistic insight for understanding the impact of PIs on cardiovascular disease and demonstrate a potential role of PXR in mediating the adverse effects of HIV PIs in humans.


Assuntos
Carbamatos/farmacologia , Dislipidemias/metabolismo , Inibidores da Protease de HIV/farmacologia , Receptores de Esteroides/metabolismo , Sulfonamidas/farmacologia , Animais , Sítios de Ligação , Carbamatos/efeitos adversos , Linhagem Celular , Furanos , Expressão Gênica , Inibidores da Protease de HIV/efeitos adversos , Humanos , Lipídeos/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Simulação de Acoplamento Molecular , Receptor de Pregnano X , Receptores de Esteroides/agonistas , Receptores de Esteroides/genética , Sulfonamidas/efeitos adversos
8.
Arterioscler Thromb Vasc Biol ; 32(12): 2869-76, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23023371

RESUMO

OBJECTIVE: Inflammatory responses are the driving force of atherosclerosis development. IκB kinase ß (IKKß), a central coordinator in inflammation through regulation of nuclear factor-κB, has been implicated in the pathogenesis of atherosclerosis. Macrophages play an essential role in the initiation and progression of atherosclerosis, yet the role of macrophage IKKß in atherosclerosis remains elusive and controversial. This study aims to investigate the impact of IKKß expression on macrophage functions and to assess the effect of myeloid-specific IKKß deletion on atherosclerosis development. METHODS AND RESULTS: To explore the issue of macrophage IKKß involvement of atherogenesis, we generated myeloid-specific IKKß-deficient low-density lipoprotein receptor-deficient mice (IKKß(ΔMye)LDLR(-/-)). Deficiency of IKKß in myeloid cells did not affect plasma lipid levels but significantly decreased diet-induced atherosclerotic lesion areas in the aortic root, brachiocephalic artery, and aortic arch of low-density lipoprotein receptor-deficient mice. Ablation of myeloid IKKß attenuated macrophage inflammatory responses and decreased atherosclerotic lesional inflammation. Furthermore, deficiency of IKKß decreased adhesion, migration, and lipid uptake in macrophages. CONCLUSIONS: The present study demonstrates a pivotal role for myeloid IKKß expression in atherosclerosis by modulating macrophage functions involved in atherogenesis. These results suggest that inhibiting nuclear factor-κB activation in macrophages may represent a feasible approach to combat atherosclerosis.


Assuntos
Aterosclerose/prevenção & controle , Quinase I-kappa B/deficiência , Células Mieloides/metabolismo , Receptores de LDL/deficiência , Animais , Aterosclerose/metabolismo , Aterosclerose/patologia , Adesão Celular , Movimento Celular , Modelos Animais de Doenças , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Metabolismo dos Lipídeos , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/genética , Receptores de LDL/metabolismo
9.
J Biol Chem ; 286(10): 8437-8447, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21190938

RESUMO

Metamorphosis in insects is regulated by juvenile hormone (JH) and ecdysteroids. The mechanism of 20-hydroxyecdysone (20E), but not of JH action, is well understood. A basic helix-loop-helix (bHLH)-Per-Arnt-Sim (PAS) family member, methoprene tolerant (Met), plays an important role in JH action. Microarray analysis and RNA interference (RNAi) were used to identify 69 genes that require Met for their hydroprene-regulated expression in the red flour beetle, Tribolium castaneum. Quantitative real time PCR analysis confirmed microarray data for 13 of the 16 hydroprene-response genes tested. The members of the bHLH-PAS family often function as heterodimers to regulate gene expression and Met is a member of this family. To determine whether other members of the bHLH-PAS family are required for the expression of JH-response genes, we employed RNAi to knockdown the expression of all 11 members of the bHLH-PAS family and studied the expression of JH-response genes in RNAi insects. These studies showed that besides Met, another member of this family, steroid receptor co-activator (SRC) is required for the expression of 15 JH-response genes tested. Moreover, studies in JH responsive Aag-2 cells revealed that Aedes aegypti homologues of both Met and SRC are required for the expression of the JH-response gene, kr-h1, and SRC is required for expression of ecdysone-response genes. These data suggest the steroid receptor co-activator plays key roles in both JH and 20E action suggesting that this may be an important molecule that mediates cross-talk between JH and 20E to prevent metamorphosis.


Assuntos
Proteínas de Insetos/metabolismo , Metoprene/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Tribolium/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica/fisiologia , Genes de Insetos/fisiologia , Sequências Hélice-Volta-Hélice , Proteínas de Insetos/genética , Coativadores de Receptor Nuclear/genética , Fatores de Transcrição/genética , Tribolium/genética
10.
J Lipid Res ; 52(9): 1652-9, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21685500

RESUMO

The pregnane X receptor (PXR, also known as SXR) is a nuclear hormone receptor activated by xenobiotics as well as diverse sterols and their metabolites. PXR functions as a xenobiotic sensor to coordinately regulate xenobiotic metabolism via transcriptional regulation of xenobiotic-detoxifying enzymes and transporters. Recent evidence indicates that PXR may also play an important role in lipid homeostasis and atherosclerosis. To define the role of PXR in atherosclerosis, we generated PXR and apoE double knockout (PXR(-/-)apoE(-/-)) mice. Here we show that deficiency of PXR did not alter plasma triglyceride and cholesterol levels in apoE(-/-) mice. However, PXR(-/-)apoE(-/-) mice had significantly decreased atherosclerotic cross-sectional lesion area in both the aortic root and brachiocephalic artery by 40% (P < 0.01) and 60% (P < 0.001), respectively. Interestingly, deficiency of PXR reduced the expression levels of CD36, lipid accumulation, and CD36-mediated oxidized LDL uptake in peritoneal macrophages of PXR(-/-)apoE(-/-) mice. Furthermore, immunofluorescence staining showed that PXR and CD36 were expressed in the atherosclerotic lesions of apoE(-/-) mice, and the expression levels of PXR and CD36 were diminished in the lesions of PXR(-/-)apoE(-/-) mice. Our findings indicate that deficiency of PXR attenuates atherosclerosis development, which may result from decreased CD36 expression and reduced lipid uptake in macrophages.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Receptores de Esteroides/deficiência , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/genética , Antígenos CD36/genética , Antígenos CD36/metabolismo , Colesterol/sangue , Humanos , Metabolismo dos Lipídeos , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Pregnano X , Receptores de Esteroides/genética , Triglicerídeos/sangue
11.
Environ Health Perspect ; 129(12): 127001, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34851150

RESUMO

BACKGROUND: Exposure to plastic-associated endocrine disrupting chemicals (EDCs) has been associated with an increased risk of cardiovascular disease (CVD) in humans. However, the underlying mechanisms for this association are unclear. Many EDCs have been shown to function as ligands of the nuclear receptor pregnane X receptor (PXR), which functions as xenobiotic sensor but also has pro-atherogenic effects in vivo. OBJECTIVE: We sought to investigate the contribution of PXR to the adverse effects dicyclohexyl phthalate (DCHP), a widely used phthalate plasticizer, on lipid homeostasis and CVD risk factors. METHODS: Cell-based assays, primary organoid cultures, and PXR conditional knockout and PXR-humanized mouse models were used to investigate the impact of DCHP exposure on PXR activation and lipid homeostasis in vitro and in vivo. Targeted lipidomics were performed to measure circulating ceramides, novel predictors for CVD. RESULTS: DCHP was identified as a potent PXR-selective agonist that led to higher plasma cholesterol levels in wild-type mice. DCHP was then demonstrated to activate intestinal PXR to elicit hyperlipidemia by using tissue-specific PXR-deficient mice. Interestingly, DCHP exposure also led to higher circulating ceramides in a PXR-dependent manner. DCHP-mediated PXR activation stimulated the expression of intestinal genes mediating lipogenesis and ceramide synthesis. Given that PXR exhibits considerable species-specific differences in receptor pharmacology, PXR-humanized mice were also used to replicate these findings. DISCUSSION: Although the adverse health effects of several well-known phthalates have attracted considerable attention, little is known about the potential impact of DCHP on human health. Our studies demonstrate that DCHP activated PXR to induce hypercholesterolemia and ceramide production in mice. These results indicate a potentially important role of PXR in contributing to the deleterious effects of plastic-associated EDCs on cardiovascular health in humans. Testing PXR activation should be considered for risk assessment of phthalates and other EDCs. https://doi.org/10.1289/EHP9262.


Assuntos
Receptores de Esteroides , Animais , Homeostase , Lipídeos , Camundongos , Camundongos Knockout , Ácidos Ftálicos , Receptor de Pregnano X , Receptores de Esteroides/agonistas , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo
12.
Environ Pollut ; 261: 114183, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32105967

RESUMO

Exposure to some environmental pollutants increases the risk of developing inflammatory disorders such as steatosis and cardiometabolic diseases. Diets high in fermentable fibers such as inulin can modulate the gut microbiota and lessen the severity of pro-inflammatory diseases, especially in individuals with elevated circulating cholesterol. Thus, we aimed to test the hypothesis that hyperlipidemic mice fed a diet enriched with 8% inulin would be protected from the pro-inflammatory toxic effects of PCB 126. Four groups of male Ldlr-/- mice were fed a high cholesterol diet containing 8% inulin or 8% cellulose (control) for 12 weeks. At weeks 2 and 4, mice were exposed to PCB 126 or vehicle (control). PCB 126 exposure induced wasting and impaired glucose tolerance, which were attenuated by inulin consumption. PCB 126 exposure induced hepatic lipid accumulation and increased inflammatory gene expression, which were both decreased by inulin consumption. In addition, inulin feeding decreased atherosclerotic lesion development in the aortic root and modulated the expression of enzymes related to glycolysis. Finally, 16S rRNA sequencing of gut microbial populations showed that PCB 126 modulated multiple microbiota genera (e.g., 3-fold decrease in Allobaculum and 3-fold increase in Coprococcus) which were normalized in inulin fed mice. Overall our data support the hypothesis that a dietary intervention that targets the gut microbiota may be an effective means of attenuating dioxin-like pollutant-mediated diseases.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Dioxinas , Microbioma Gastrointestinal , Animais , Disbiose , Inulina , Masculino , Camundongos , Bifenilos Policlorados , Prebióticos , RNA Ribossômico 16S
13.
Arch Insect Biochem Physiol ; 72(2): 74-87, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19557747

RESUMO

Trypsins belong to the serine endoproteases. They are the most important proteases in insects because of their key roles in food digestion and zymogens activation. But there has been little study of the trypsins in the integuments of insects. In this work, we cloned a trypsin-like protease gene from Helicoverpa armigera and named it trypsin-like protease 2 (Ha-TLP2). Semi-quantitative reverse transcription PCR analysis showed that Ha-TLP2 is constitutively expressed in the integument and can be down-regulated by 20-hydroxyecdysone (20E) and up-regulated by the juvenile hormone (JH) analog methoprene. Immunohistochemistry showed that Ha-TLP2 is located not only in the epidermis, but also in new and old cuticles. Immunoblotting and gelatin-SDS-PAGE revealed that Ha-TLP2 is constitutively expressed with activity in the integument during larval feeding, molting, and metamorphosis. This evidence suggests that Ha-TLP2 is involved in the remodeling of the integument.


Assuntos
Proteínas de Insetos/isolamento & purificação , Mariposas/enzimologia , Tripsina/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Eletroforese em Gel de Poliacrilamida , Regulação Enzimológica da Expressão Gênica/fisiologia , Immunoblotting , Proteínas de Insetos/genética , Larva/enzimologia , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Tripsina/genética
14.
JCI Insight ; 4(3)2019 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-30728326

RESUMO

Quetiapine, one of the most prescribed atypical antipsychotics, has been associated with hyperlipidemia and an increased risk for cardiovascular disease in patients, but the underlying mechanisms remain unknown. Here, we identified quetiapine as a potent and selective agonist for pregnane X receptor (PXR), a key nuclear receptor that regulates xenobiotic metabolism in the liver and intestine. Recent studies have indicated that PXR also plays an important role in lipid homeostasis. We generated potentially novel tissue-specific PXR-KO mice and demonstrated that quetiapine induced hyperlipidemia by activating intestinal PXR signaling. Quetiapine-mediated PXR activation stimulated the intestinal expression of cholesterol transporter Niemann-Pick C1-Like 1 (NPC1L1) and microsomal triglyceride transfer protein (MTP), leading to increased intestinal lipid absorption. While NPC1L1 is a known PXR target gene, we identified a DR-1-type PXR-response element in the MTP promoter and established MTP as a potentially novel transcriptional target of PXR. Quetiapine's effects on PXR-mediated gene expression and cholesterol uptake were also confirmed in cultured murine enteroids and human intestinal cells. Our findings suggest a potential role of PXR in mediating adverse effects of quetiapine in humans and provide mechanistic insights for certain atypical antipsychotic-associated dyslipidemia.

15.
Insect Mol Biol ; 17(5): 523-9, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18828838

RESUMO

A 967-bp cDNA, encoding a 31 kDa lipase with an isoelectric point of 8.8, was cloned from Helicoverpa armigera. The gene was named Ha-lipase. The Ha-lipase transcript was present in the head-thorax, midgut and fat body but not in integument and haemocytes from 5th instar larvae, with higher expression levels in feeding larvae than that in molting larvae. In the midgut, transcription of the gene during the 5th instar larval period was lower than during the 6th instar period. While in the fat body, lipase was not found at the 6th instar stage but only at the 5th instar stage. The expression of Ha-lipase could be up-regulated by methoprene and down-regulated by methoxyfenozide (RH-2485) and 20-hydroxyecdysone (20E).


Assuntos
Perfilação da Expressão Gênica , Gossypium/parasitologia , Hormônios de Inseto/farmacologia , Lepidópteros/enzimologia , Lepidópteros/genética , Lipase/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Clonagem Molecular , DNA Complementar/genética , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/enzimologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Íntrons/genética , Larva/efeitos dos fármacos , Larva/genética , Lepidópteros/efeitos dos fármacos , Lipase/química , Lipase/metabolismo , Dados de Sequência Molecular , Filogenia , Alinhamento de Sequência
16.
Endocrinology ; 159(4): 1595-1608, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29425287

RESUMO

Bisphenol A (BPA) is a base chemical used extensively in numerous consumer products, and human exposure to BPA is ubiquitous. Higher BPA exposure has been associated with an increased risk of atherosclerosis and cardiovascular disease (CVD) in multiple human population-based studies. However, the underlying mechanisms responsible for the associations remain elusive. We previously reported that BPA activates the xenobiotic receptor pregnane X receptor (PXR), which has proatherogenic effects in animal models. Because BPA is a potent agonist for human PXR but does not affect rodent PXR activity, a suitable PXR-humanized apolipoprotein E-deficient (huPXR•ApoE-/-) mouse model was developed to study BPA's atherogenic effects. Chronic BPA exposure increased atherosclerosis in the huPXR•ApoE-/- mice. We report that BPA exposure can also activate human PXR signaling in the heart tubes of huPXR•ApoE-/- embryos, and perinatal BPA exposure exacerbated atherosclerosis in adult male huPXR•ApoE-/- offspring. However, atherosclerosis development in female offspring was not affected by perinatal BPA exposure. Perinatal BPA exposure did not affect plasma lipid levels but increased aortic and atherosclerotic lesional fatty acid transporter CD36 expression in male huPXR•ApoE-/- offspring. Mechanistically, PXR epigenetically regulated CD36 expression by increasing H3K4me3 levels and decreasing H3K27me3 levels in the CD36 promoter in response to perinatal BPA exposure. The findings from the present study contribute to our understanding of the association between BPA exposure and increased atherosclerosis or CVD risk in humans, and activation of human PXR should be considered for future BPA risk assessment.


Assuntos
Aorta/efeitos dos fármacos , Aterosclerose/metabolismo , Compostos Benzidrílicos/administração & dosagem , Poluentes Ambientais/administração & dosagem , Fenóis/administração & dosagem , Receptores de Esteroides/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Aorta/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Antígenos CD36/genética , Antígenos CD36/metabolismo , Colesterol/sangue , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Receptor de Pregnano X
17.
JCI Insight ; 3(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29367460

RESUMO

Mesenchymal stem cells (MSCs) can give rise to both adipocytes and osteoblasts, but the molecular mechanisms underlying MSC fate determination remain poorly understood. IκB kinase ß (IKKß), a central coordinator of inflammation and immune responses through activation of NF-κB, has been implicated as a critical molecular link between obesity and metabolic disorders. Here, we show that IKKß can reciprocally regulate adipocyte and osteoblast differentiation of murine and human MSCs through an NF-κB-independent mechanism. IKKß is a ß-catenin kinase that phosphorylates the conserved degron motif of ß-catenin to prime it for ß-TrCP-mediated ubiquitination and degradation, thereby increasing adipogenesis and inhibiting osteogenesis in MSCs. Animal studies demonstrated that deficiency of IKKß in BM mesenchymal stromal cells increased bone mass and decreased BM adipocyte formation in adult mice. In humans, IKKß expression in adipose tissue was also positively associated with increased adiposity and elevated ß-catenin phosphorylation. These findings suggest IKKß as a key molecular switch that regulates MSC fate, and they provide potentially novel mechanistic insights into the understanding of the cross-regulation between the evolutionarily conserved IKKß and Wnt/ß-catenin signaling pathways. The IKKß-Wnt axis we uncovered may also have important implications for development, homeostasis, and disease pathogenesis.


Assuntos
Diferenciação Celular/fisiologia , Quinase I-kappa B/metabolismo , Células-Tronco Mesenquimais/metabolismo , Obesidade/patologia , beta Catenina/metabolismo , Gordura Abdominal/patologia , Adipócitos/fisiologia , Adipogenia/fisiologia , Adulto , Animais , Biópsia , Células Cultivadas , Feminino , Humanos , Quinase I-kappa B/análise , Quinase I-kappa B/genética , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Modelos Animais , Obesidade/sangue , Osteoblastos/fisiologia , Osteogênese/fisiologia , Fosforilação/fisiologia , Cultura Primária de Células , Proteólise , Ubiquitinação/fisiologia , Via de Sinalização Wnt/fisiologia , Proteínas Contendo Repetições de beta-Transducina/metabolismo
18.
Diabetes ; 65(6): 1616-29, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26993069

RESUMO

IκB kinase ß (IKKß), a central coordinator of inflammatory responses through activation of nuclear factor-κB (NF-κB), has been implicated as a critical molecular link between inflammation and metabolic disorders; however, the role of adipocyte IKKß in obesity and related metabolic disorders remains elusive. Here we report an essential role of IKKß in the regulation of adipose remodeling and adipocyte survival in diet-induced obesity. Targeted deletion of IKKß in adipocytes does not affect body weight, food intake, and energy expenditure but results in an exaggerated diabetic phenotype when challenged with a high-fat diet (HFD). IKKß-deficient mice have multiple histopathologies in visceral adipose tissue, including increased adipocyte death, amplified macrophage infiltration, and defective adaptive adipose remodeling. Deficiency of IKKß also leads to increased adipose lipolysis, elevated plasma free fatty acid (FFA) levels, and impaired insulin signaling. Mechanistic studies demonstrated that IKKß is a key adipocyte survival factor and that IKKß protects murine and human adipocytes from HFD- or FFA-elicited cell death through NF-κB-dependent upregulation of antiapoptotic proteins and NF-κB-independent inactivation of proapoptotic BAD protein. Our findings establish IKKß as critical for adipocyte survival and adaptive adipose remodeling in obesity.


Assuntos
Adipócitos/enzimologia , Adiposidade/fisiologia , Sobrevivência Celular/fisiologia , Quinase I-kappa B/fisiologia , Obesidade/enzimologia , Animais , Dieta Hiperlipídica/efeitos adversos , Quinase I-kappa B/deficiência , Insulina/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Obesidade/etiologia , Transdução de Sinais
19.
Mol Endocrinol ; 29(5): 765-76, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25811240

RESUMO

Recent studies have associated endocrine-disrupting chemical (EDC) exposure with the increased risk of cardiovascular disease in humans, but the underlying mechanisms responsible for these associations remain elusive. Many EDCs have been implicated in activation of the nuclear receptor pregnane X receptor (PXR), which acts as a xenobiotic sensor to regulate xenobiotic metabolism in the liver and intestine. Here we report an important role of intestinal PXR in linking xenobiotic exposure and hyperlipidemia. We identified tributyl citrate (TBC), one of a large group of Food and Drug Administration-approved plasticizers for pharmaceutical or food applications, as a potent and selective PXR agonist. TBC efficiently activated PXR and induced PXR target gene expression in vitro and in vivo. Interestingly, TBC activated intestinal PXR but did not affect hepatic PXR activity. Exposure to TBC increased plasma total cholesterol and atherogenic low-density lipoprotein cholesterol levels in wild-type mice, but not in PXR-deficient mice. TBC-mediated PXR activation stimulated the expression of an essential cholesterol transporter, Niemann-Pick C1-like 1 (NPC1L1), in the intestine. Promoter analysis revealed a DR-4 type of PXR response element in the human NPC1L1 promoter, and TBC promoted PXR recruitment onto the NPC1L1 promoter. Consistently, TBC treatment significantly increased lipid uptake by human and murine intestinal cells and deficiency of PXR inhibited TBC-elicited lipid uptake. These findings provide critical mechanistic insight for understanding the impact of EDC-mediated PXR activation on lipid homeostasis and demonstrate a potential role of PXR in mediating the adverse effects of EDCs on cardiovascular disease risk in humans.


Assuntos
Citratos/toxicidade , Disruptores Endócrinos/toxicidade , Ácidos Ftálicos/toxicidade , Plastificantes/toxicidade , Receptores de Esteroides/metabolismo , Animais , Colesterol/metabolismo , Células Hep G2 , Humanos , Hipercolesterolemia/sangue , Hipercolesterolemia/induzido quimicamente , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Masculino , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor de Pregnano X , Transcrição Gênica , Ativação Transcricional , Xenobióticos
20.
Insect Biochem Mol Biol ; 52: 23-32, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24931431

RESUMO

Juvenile hormone (JH) plays important roles in regulation of many physiological processes including development, reproduction and metabolism in insects. However, the molecular mechanisms of JH signaling pathway are not completely understood. To elucidate the molecular mechanisms of JH regulation of Krüppel homolog 1 gene (Kr-h1) in Aedes aegypti, we employed JH-sensitive Aag-2 cells developed from the embryos of this insect. In Aag-2 cells, AaKr-h1 gene is induced by nanomolar concentration of JH III, its expression peaked at 1.5 h after treatment with JH III. RNAi studies showed that JH induction of this gene requires the presence of Ae. aegypti methoprene-tolerant (AaMet). A conserved 13 nucleotide JH response element (JHRE, TGCCTCCACGTGC) containing canonical E box motif (underlined) identified in the promoter of AaKr-h1 is required for JH induction of this gene. Critical nucleotides in the JHRE required for JH action were identified by employing mutagenesis and reporter assays. Reporter assays also showed that basic helix loop helix (bHLH) domain of AaMet is required for JH induction of AaKr-h1. 5' rapid amplification of cDNA ends method identified two isoforms of AaKr-h1, AaKr-h1α and AaKr-h1ß, the expression of both isoforms is induced by JH III, but AaKr-h1α is the predominant isoform in both Aag-2 cells and Ae. aegypti larvae.


Assuntos
Aedes/genética , Elementos E-Box , Hormônios Juvenis/genética , Hormônios Juvenis/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Larva/genética , Larva/metabolismo , Aedes/crescimento & desenvolvimento , Animais , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição Kruppel-Like/genética , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Interferência de RNA , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA