Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 299(3): 103026, 2023 03.
Article in English | MEDLINE | ID: mdl-36796516

ABSTRACT

Autoimmune hepatitis (AIH) is a typical T cell-mediated chronic liver disease with a higher incidence in females. However, the molecular mechanism for the female predisposition is poorly understood. Estrogen sulfotransferase (Est) is a conjugating enzyme best known for its function in sulfonating and deactivating estrogens. The goal of this study is to investigate whether and how Est plays a role in the higher incidence of AIH in females. Concanavalin A (ConA) was used to induce T cell-mediated hepatitis in female mice. We first showed that Est was highly induced in the liver of ConA-treated mice. Systemic or hepatocyte-specific ablation of Est, or pharmacological inhibition of Est, protected female mice from ConA-induced hepatitis regardless of ovariectomy, suggesting the effect of Est inhibition was estrogen independent. In contrast, we found that hepatocyte-specific transgenic reconstitution of Est in the whole-body Est knockout (EstKO) mice abolished the protective phenotype. Upon the ConA challenge, EstKO mice exhibited a more robust inflammatory response with elevated production of proinflammatory cytokines and changed liver infiltration of immune cells. Mechanistically, we determined that ablation of Est led to the hepatic induction of lipocalin 2 (Lcn2), whereas ablation of Lcn2 abolished the protective phenotype of EstKO females. Our findings demonstrate that hepatocyte Est is required for the sensitivity of female mice to ConA-induced and T cell-mediated hepatitis in an estrogen-independent manner. Est ablation may have protected female mice from ConA-induced hepatitis by upregulating Lcn2. Pharmacological inhibition of Est might be a potential strategy for the treatment of AIH.


Subject(s)
Estrogens , Hepatitis, Autoimmune , Mice , Female , Animals , Concanavalin A/toxicity , Estrogens/pharmacology , T-Lymphocytes , Hepatocytes , Liver , Hepatitis, Autoimmune/genetics , Hepatitis, Autoimmune/prevention & control , Mice, Knockout , Mice, Inbred C57BL
2.
Gastroenterology ; 162(4): 1226-1241, 2022 04.
Article in English | MEDLINE | ID: mdl-34954226

ABSTRACT

BACKGROUND & AIMS: Sulfoconjugation of small molecules or protein peptides is a key mechanism to ensure biochemical and functional homeostasis in mammals. The PAPS synthase 2 (PAPSS2) is the primary enzyme to synthesize the universal sulfonate donor 3'-phosphoadenosine 5'-phosphosulfate (PAPS). Acetaminophen (APAP) overdose is the leading cause of acute liver failure (ALF), in which oxidative stress is a key pathogenic event, whereas sulfation of APAP contributes to its detoxification. The goal of this study was to determine whether and how PAPSS2 plays a role in APAP-induced ALF. METHODS: Gene expression was analyzed in APAP-induced ALF in patients and mice. Liver-specific Papss2-knockout mice using Alb-Cre (Papss2ΔHC) or AAV8-TBG-Cre (Papss2iΔHC) were created and subjected to APAP-induced ALF. Primary human and mouse hepatocytes were used for in vitro mechanistic analysis. RESULTS: The hepatic expression of PAPSS2 was decreased in APAP-induced ALF in patients and mice. Surprisingly, Papss2ΔHC mice were protected from APAP-induced hepatotoxicity despite having a decreased APAP sulfation, which was accompanied by increased hepatic antioxidative capacity through the activation of the p53-p2-Nrf2 axis. Treatment with a sulfation inhibitor also ameliorated APAP-induced hepatotoxicity. Gene knockdown experiments showed that the hepatoprotective effect of Papss2ΔHC was Nrf2, p53, and p21 dependent. Mechanistically, we identified p53 as a novel substrate of sulfation. Papss2 ablation led to p53 protein accumulation by preventing p53 sulfation, which disrupts p53-MDM2 interaction and p53 ubiquitination and increases p53 protein stability. CONCLUSIONS: We have uncovered a previously unrecognized and p53-mediated role of PAPSS2 in controlling oxidative response. Inhibition of p53 sulfation may be explored for the clinical management of APAP overdose.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Failure, Acute , Acetaminophen/toxicity , Animals , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Humans , Liver/metabolism , Liver Failure, Acute/chemically induced , Liver Failure, Acute/metabolism , Liver Failure, Acute/prevention & control , Mammals/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Tumor Suppressor Protein p53/metabolism
3.
Gastroenterology ; 161(1): 271-286.e11, 2021 07.
Article in English | MEDLINE | ID: mdl-33819483

ABSTRACT

BACKGROUND & AIMS: Sulfation is a conjugation reaction essential for numerous biochemical and cellular functions in mammals. The 3'-phosphoadenosine 5'-phosphosulfate (PAPS) synthase 2 (PAPSS2) is the key enzyme to generate PAPS, which is the universal sulfonate donor for all sulfation reactions. The goal of this study was to determine whether and how PAPSS2 plays a role in colitis and colonic carcinogenesis. METHODS: Tissue arrays of human colon cancer specimens, gene expression data, and clinical features of cancer patients were analyzed. Intestinal-specific Papss2 knockout mice (Papss2ΔIE) were created and subjected to dextran sodium sulfate-induced colitis and colonic carcinogenesis induced by a combined treatment of azoxymethane and dextran sodium sulfate or azoxymethane alone. RESULTS: The expression of PAPSS2 is decreased in the colon cancers of mice and humans. The lower expression of PAPSS2 in colon cancer patients is correlated with worse survival. Papss2ΔIE mice showed heightened sensitivity to colitis and colon cancer by damaging the intestinal mucosal barrier, increasing intestinal permeability and bacteria infiltration, and worsening the intestinal tumor microenvironment. Mechanistically, the Papss2ΔIE mice exhibited reduced intestinal sulfomucin content. Metabolomic analyses revealed the accumulation of bile acids, including the Farnesoid X receptor antagonist bile acid tauro-ß-muricholic acid, and deficiency in the formation of bile acid sulfates in the colon of Papss2ΔIE mice. CONCLUSIONS: We have uncovered an important role of PAPSS2-mediated sulfation in colitis and colonic carcinogenesis. Intestinal sulfation may represent a potential diagnostic marker and PAPSS2 may serve as a potential therapeutic target for inflammatory bowel disease and colon cancer.


Subject(s)
Colitis-Associated Neoplasms/prevention & control , Colitis/prevention & control , Colon/enzymology , Intestinal Mucosa/enzymology , Mucins/metabolism , Multienzyme Complexes/metabolism , Sulfate Adenylyltransferase/metabolism , Animals , Bile Acids and Salts/metabolism , Colitis/enzymology , Colitis/genetics , Colitis/pathology , Colitis-Associated Neoplasms/enzymology , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/pathology , Colon/pathology , Databases, Genetic , Disease Models, Animal , Humans , Intestinal Mucosa/pathology , Metabolome , Metabolomics , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes/genetics , Prognosis , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfate Adenylyltransferase/genetics
4.
J Am Soc Nephrol ; 31(7): 1496-1508, 2020 07.
Article in English | MEDLINE | ID: mdl-32424001

ABSTRACT

BACKGROUND: Studies have suggested that estrogens may protect mice from AKI. Estrogen sulfotransferase (SULT1E1, or EST) plays an important role in estrogen homeostasis by sulfonating and deactivating estrogens, but studies on the role of SULT1E1 in AKI are lacking. METHODS: We used the renal ischemia-reperfusion model to investigate the role of SULT1E1 in AKI. We subjected wild-type mice, Sult1e1 knockout mice, and Sult1e1 knockout mice with liver-specific reconstitution of SULT1E1 expression to bilateral renal ischemia-reperfusion or sham surgery, either in the absence or presence of gonadectomy. We assessed relevant biochemical, histologic, and gene expression markers of kidney injury. We also used wild-type mice treated with the SULT1E1 inhibitor triclosan to determine the effect of pharmacologic inhibition of SULT1E1 on AKI. RESULTS: AKI induced the expression of Sult1e1 in a tissue-specific and sex-specific manner. It induced expression of Sult1e1 in the liver in both male and female mice, but Sult1e1 induction in the kidney occurred only in male mice. Genetic knockout or pharmacologic inhibition of Sult1e1 protected mice of both sexes from AKI, independent of the presence of sex hormones. Instead, a gene profiling analysis indicated that the renoprotective effect was associated with increased vitamin D receptor signaling. Liver-specific transgenic reconstitution of SULT1E1 in Sult1e1 knockout mice abolished the protection in male mice but not in female mice, indicating that Sult1e1's effect on AKI was also tissue-specific and sex-specific. CONCLUSIONS: SULT1E1 appears to have a novel function in the pathogenesis of AKI. Our findings suggest that inhibitors of SULT1E1 might have therapeutic utility in the clinical management of AKI.


Subject(s)
Acute Kidney Injury/metabolism , Acute Kidney Injury/prevention & control , Liver/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Acute Kidney Injury/etiology , Animals , Calcitriol/pharmacology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Gene Expression , Gene Expression Profiling , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Male , Mice , Mice, Knockout , Orchiectomy , Ovariectomy , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Reperfusion Injury/complications , Sex Factors , Signal Transduction , Sulfotransferases/antagonists & inhibitors , Triclosan/pharmacology
5.
J Biol Chem ; 294(32): 12112-12121, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31217279

ABSTRACT

Conjugated equine estrogens (CEEs), whose brand name is Premarin, are widely used as a hormone-replacement therapy (HRT) drug to manage postmenopausal symptoms in women. Extracted from pregnant mare urine, CEEs are composed of nearly a dozen estrogens existing in an inactive sulfated form. To determine whether the hepatic steroid sulfatase (STS) is a key contributor to the efficacy of CEEs in HRT, we performed estrogen-responsive element (ERE) reporter gene assay, real-time PCR, and UPLC-MS/MS to assess the STS-dependent and inflammation-responsive estrogenic activity of CEEs in HepG2 cells and human primary hepatocytes. Using liver-specific STS-expressing transgenic mice, we also evaluated the effect of STS on the estrogenic activity of CEEs in vivo We observed that CEEs induce activity of the ERE reporter gene in an STS-dependent manner and that genetic or pharmacological inhibition of STS attenuates CEE estrogenic activity. In hepatocytes, inflammation enhanced CEE estrogenic activity by inducing STS gene expression. The inflammation-responsive estrogenic activity of CEEs, in turn, attenuated inflammation through the anti-inflammatory activity of the active estrogens. In vivo, transgenic mice with liver-specific STS expression exhibited markedly increased sensitivity to CEE-induced estrogenic activity in the uterus resulting from increased levels of liver-derived and circulating estrogens. Our results reveal a critical role of hepatic STS in mediating the hormone-replacing activity of CEEs. We propose that caution needs to be applied when Premarin is used in patients with chronic inflammatory liver diseases because such patients may have heightened sensitivity to CEEs due to the inflammatory induction of STS activity.


Subject(s)
Estrogens, Conjugated (USP)/metabolism , Steryl-Sulfatase/metabolism , Animals , Cells, Cultured , Chromatography, High Pressure Liquid , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens, Conjugated (USP)/analysis , Estrogens, Conjugated (USP)/pharmacology , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Horses , Humans , Inflammation/metabolism , Inflammation/pathology , Liver/drug effects , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA Interference , RNA, Small Interfering/metabolism , Steryl-Sulfatase/antagonists & inhibitors , Steryl-Sulfatase/genetics , Tandem Mass Spectrometry , Uterus/drug effects , Uterus/metabolism , Uterus/pathology
6.
Gastroenterology ; 157(3): 793-806.e14, 2019 09.
Article in English | MEDLINE | ID: mdl-31170413

ABSTRACT

BACKGROUND & AIMS: The role of aryl hydrocarbon receptor (AhR) in liver fibrosis is controversial because loss and gain of AhR activity both lead to liver fibrosis. The goal of this study was to investigate how the expression of AhR by different liver cell types, hepatic stellate cells (HSCs) in particular, affects liver fibrosis in mice. METHODS: We studied the effects of AhR on primary mouse and human HSCs, measuring their activation and stimulation of fibrogenesis using RNA-sequencing analysis. C57BL/6J mice were given the AhR agonists 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) or 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE); were given carbon tetrachloride (CCl4); or underwent bile duct ligation. We also performed studies in mice with disruption of Ahr specifically in HSCs, hepatocytes, or Kupffer cells. Liver tissues were collected from mice and analyzed by histology, immunohistochemistry, and immunoblotting. RESULTS: AhR was expressed at high levels in quiescent HSCs, but the expression decreased with HSC activation. Activation of HSCs from AhR-knockout mice was accelerated compared with HSCs from wild-type mice. In contrast, TCDD or ITE inhibited spontaneous and transforming growth factor ß-induced activation of HSCs. Mice with disruption of Ahr in HSCs, but not hepatocytes or Kupffer cells, developed more severe fibrosis after administration of CCl4 or bile duct ligation. C57BL/6J mice given ITE did not develop CCl4-induced liver fibrosis, whereas mice without HSC AhR given ITE did develop CCl4-induced liver fibrosis. In studies of mouse and human HSCs, we found that AhR prevents transforming growth factor ß-induced fibrogenesis by disrupting the interaction of Smad3 with ß-catenin, which prevents the expression of genes that mediate fibrogenesis. CONCLUSIONS: In studies of human and mouse HSCs, we found that AhR prevents HSC activation and expression of genes required for liver fibrogenesis. Development of nontoxic AhR agonists or strategies to activate AhR signaling in HSCs might be developed to prevent or treat liver fibrosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cellular Senescence , Chemical and Drug Induced Liver Injury/prevention & control , Hepatic Stellate Cells/metabolism , Liver Cirrhosis, Experimental/prevention & control , Liver/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Cells, Cultured , Cellular Senescence/drug effects , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Gene Expression Regulation , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Indoles/pharmacology , Liver/drug effects , Liver/pathology , Liver Cirrhosis, Experimental/genetics , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Aryl Hydrocarbon/agonists , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Smad3 Protein/metabolism , Thiazoles/pharmacology , beta Catenin/metabolism
7.
Hepatology ; 70(3): 995-1010, 2019 09.
Article in English | MEDLINE | ID: mdl-31038762

ABSTRACT

Hemorrhagic shock (HS) is a life-threatening condition associated with tissue hypoperfusion and often leads to injury of multiple organs including the liver. Pregnane X receptor (PXR) is a species-specific xenobiotic receptor that regulates the expression of drug-metabolizing enzymes (DMEs) such as the cytochrome P450 (CYP) 3A. Many clinical drugs, including those often prescribed to trauma patients, are known to activate PXR and induce CYP3A. The goal of this study is to determine whether PXR plays a role in the regulation of DMEs in the setting of HS and whether activation of PXR is beneficial or detrimental to HS-induced hepatic injury. PXR transgenic, knockout, and humanized mice were subject to HS, and the liver injury was assessed histologically and biochemically. The expression and/or activity of PXR and CYP3A were manipulated genetically or pharmacologically in order to determine their effects on HS-induced liver injury. Our results showed that genetic or pharmacological activation of PXR sensitized wild-type and hPXR/CYP3A4 humanized mice to HS-induced hepatic injury, whereas knockout of PXR protected mice from HS-induced liver injury. Mechanistically, the sensitizing effect of PXR activation was accounted for by PXR-responsive induction of CYP3A and increased oxidative stress in the liver. The sensitizing effect of PXR was attenuated by ablation or pharmacological inhibition of CYP3A, treatment with the antioxidant N-acetylcysteine amide, or treatment with a PXR antagonist. Conclusion: We have uncovered a function of PXR in HS-induced hepatic injury. Our results suggest that the unavoidable use of PXR-activating drugs in trauma patients has the potential to exacerbate HS-induced hepatic injury, which can be mitigated by the coadministration of antioxidative agents, CYP3A inhibitors, or PXR antagonists.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors/pharmacology , Cytochrome P-450 CYP3A/metabolism , Hepatic Insufficiency/pathology , Pregnane X Receptor/genetics , Shock, Hemorrhagic/genetics , Animals , Disease Models, Animal , Gene Expression Regulation , Hepatic Insufficiency/etiology , Hepatic Insufficiency/genetics , Humans , Mice , Mice, Knockout , Mice, Transgenic , Prognosis , Random Allocation , Risk Assessment , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/drug therapy , Survival Rate , Treatment Outcome , Up-Regulation
8.
Mol Pharmacol ; 95(6): 597-605, 2019 06.
Article in English | MEDLINE | ID: mdl-30944208

ABSTRACT

Overdose of acetaminophen (APAP) is the leading cause of acute liver failure (ALF) in the United States. The sulfotransferase-mediated sulfation of APAP is widely believed to be a protective mechanism to attenuate the hepatotoxicity of APAP. The cholesterol sulfotransferase SULT2B1b is best known for its activity in catalyzing the sulfoconjugation of cholesterol to synthesize cholesterol sulfate. SULT2B1b can be transcriptionally and positively regulated by the hepatic nuclear factor 4α (HNF4α). In this study, we uncovered an unexpected role for SULT2B1b in APAP toxicity. Hepatic overexpression of SULT2B1b sensitized mice to APAP-induced liver injury, whereas ablation of the Sult2B1b gene in mice conferred resistance to the APAP hepatotoxicity. Consistent with the notion that Sult2B1b is a transcriptional target of HNF4α, overexpression of HNF4α sensitized mice or primary hepatocytes to APAP-induced hepatotoxicity in a Sult2B1b-dependent manner. We conclude that the HNF4α-SULT2B1b axis has a unique role in APAP-induced acute liver injury, and SULT2B1b induction might be a risk factor for APAP hepatotoxicity.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Drug Overdose/complications , Hepatocyte Nuclear Factor 4/metabolism , Sulfotransferases/genetics , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury, Chronic/genetics , Disease Models, Animal , Drug Overdose/etiology , Drug Overdose/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Mice , Sulfotransferases/metabolism
9.
Mol Pharmacol ; 94(4): 1145-1154, 2018 10.
Article in English | MEDLINE | ID: mdl-30045953

ABSTRACT

Cholesterol is essential for numerous biologic functions and processes, but an excess of intracellular cholesterol can be toxic. Intestinal cholesterol absorption is a major determinant of plasma cholesterol level. The liver X receptor (LXR) is a nuclear receptor known for its activity in cholesterol efflux and reverse cholesterol transport. In this study, we uncovered a surprising function of LXR in intestinal cholesterol absorption and toxicity. Genetic or pharmacologic activation of LXRα-sensitized mice to a high-cholesterol diet (HCD) induced intestinal toxicity and tissue damage, including the disruption of enterocyte tight junctions, whereas the same HCD caused little toxicity in the absence of LXR activation. The gut toxicity in HCD-fed LXR-KI mice may have been accounted for by the increased intestinal cholesterol absorption and elevation of enterocyte and systemic levels of free cholesterol. The increased intestinal cholesterol absorption preceded the gut toxicity, suggesting that the increased absorption was not secondary to tissue damage. The heightened sensitivity to HCD in the HCD-fed LXRα-activated mice appeared to be intestine-specific because the liver was not affected despite activation of the same receptor in this tissue. Moreover, heightened sensitivity to HCD cannot be reversed by ezetimibe, a Niemann-Pick C1-like 1 inhibitor that inhibits intestinal cholesterol absorption, suggesting that the increased cholesterol absorption in LXR-activated intestine is mediated by a mechanism that has yet to be defined.


Subject(s)
Cholesterol/adverse effects , Diet/adverse effects , Intestinal Mucosa/metabolism , Liver X Receptors/metabolism , Liver/metabolism , Animals , Intestinal Absorption/physiology , Intestines , Mice , Mice, Inbred C57BL
10.
Mol Pharmacol ; 93(3): 239-250, 2018 03.
Article in English | MEDLINE | ID: mdl-29351922

ABSTRACT

Acute kidney injury (AKI) is associate with high mortality. Despite evidence of AKI-induced distant organ injury, a relationship between AKI and liver injury has not been clearly established. The goal of this study is to investigate whether renal ischemia-reperfusion (IR) can affect liver pathophysiology. We showed that renal IR in mice induced fatty liver and compromised liver function through the downregulation of constitutive androstane receptor (CAR; -90.4%) and inhibition of hepatic very-low-density lipoprotein triglyceride (VLDL-TG) secretion (-28.4%). Treatment of mice with the CAR agonist 1,4-bis[2-(3,5 dichloropyridyloxy)] benzene (TCPOBOP) prevented the development of AKI-induced fatty liver and liver injury, which was associated with the attenuation of AKI-induced inhibition of VLDL-TG secretion. The hepatoprotective effect of TCPOBOP was abolished in CAR-/- mice. Interestingly, alleviation of fatty liver by TCPOBOP also improved the kidney function, whereas CAR ablation sensitized mice to AKI-induced kidney injury and lethality. The serum concentrations of interleukin-6 (IL-6) were elevated by 27-fold after renal IR, but were normalized in TCPOBOP-treated AKI mice, suggesting that the increased release of IL-6 from the kidney may have mediated the AKI responsive liver injury. Taken together, our results revealed an interesting kidney-liver organ cross-talk in response to AKI. Given the importance of CAR in the pathogenesis of renal IR-induced fatty liver and impaired kidney function, fatty liver can be considered as an important risk factor for kidney injury, and a timely management of hepatic steatosis by CAR activation may help to restore kidney function in patients with AKI or kidney transplant.


Subject(s)
Acute Kidney Injury/drug therapy , Fatty Liver/drug therapy , Liver/physiopathology , Pyridines/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Reperfusion Injury/complications , Acute Kidney Injury/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Animals , Constitutive Androstane Receptor , Down-Regulation/drug effects , Fatty Liver/etiology , Fatty Liver/metabolism , Fatty Liver/physiopathology , Gene Expression Regulation, Neoplastic/drug effects , Interleukin-6/metabolism , Lipoproteins, VLDL/metabolism , Liver/drug effects , Liver/metabolism , Liver Function Tests , Mice , Pyridines/pharmacology , Reperfusion Injury/metabolism
11.
Am J Pathol ; 187(5): 1059-1067, 2017 May.
Article in English | MEDLINE | ID: mdl-28279656

ABSTRACT

Sepsis is defined as the host's deleterious systemic inflammatory response to microbial infections. Herein, we report an essential role of the fatty acid binding protein 4 (FABP4; alias adipocyte protein 2 or aP2), a lipid-binding chaperone, in sepsis response. Bioinformatic analysis of the Gene Expression Omnibus data sets showed the level of FABP4 was higher in the nonsurvival sepsis patients' whole blood compared to the survival cohorts. The expression of Fabp4 was induced in a liver-specific manner in cecal ligation and puncture (CLP) and lipopolysaccharide treatment models of sepsis. The induction of Fabp4 may have played a pathogenic role, because ectopic expression of Fabp4 in the liver sensitized mice to CLP-induced inflammatory response and worsened the animal's survival. In contrast, pharmacological inhibition of Fabp4 markedly alleviated the CLP responsive inflammation and tissue damage and improved survival. We conclude that FABP4 is an important mediator of the sepsis response. Early intervention by pharmacological inhibition of FABP4 may help to manage sepsis in the clinic.


Subject(s)
Fatty Acid-Binding Proteins/metabolism , Liver/metabolism , Sepsis/etiology , Adipose Tissue , Animals , Cecum , Disease Models, Animal , Escherichia coli Infections/physiopathology , Fatty Acid-Binding Proteins/antagonists & inhibitors , Female , Hepatocytes/microbiology , Hepatocytes/physiology , Humans , Interleukin-6/metabolism , Kupffer Cells/physiology , Ligation , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Pimozide/pharmacology , Punctures , Sepsis/mortality
12.
Hepatology ; 65(3): 893-906, 2017 03.
Article in English | MEDLINE | ID: mdl-28102638

ABSTRACT

Chronic hepatitis B virus infection is a major risk factor for hepatocellular carcinoma (HCC). Hepatitis B virus X protein (HBx) is a hepatitis B virus protein that has multiple cellular functions, but its role in HCC pathogenesis has been controversial. Farnesoid X receptor (FXR) is a nuclear receptor with activities in anti-inflammation and inhibition of hepatocarcinogenesis. However, whether or how FXR can impact hepatitis B virus/HBx-induced hepatocarcinogenesis remains unclear. In this study, we showed that HBx can interact with FXR and function as a coactivator of FXR. Expression of HBx in vivo enhanced FXR-responsive gene regulation. HBx also increased the transcriptional activity of FXR in a luciferase reporter gene assay. The HBx-FXR interaction was confirmed by coimmunoprecipitation and glutathione S-transferase pull-down assays, and the FXR activation function 1 domain was mapped to bind to the third α helix in the C terminus of HBx. We also found that the C-terminally truncated variants of HBx, which were found in clinical HCC, were not effective at transactivating FXR. Interestingly, recruitment of the full-length HBx, but not the C-terminally truncated HBx, enhanced the binding of FXR to its response element. In vivo, FXR ablation markedly sensitized mice to HBx-induced hepatocarcinogenesis. CONCLUSIONS: We propose that transactivation of FXR by full-length HBx may represent a protective mechanism to inhibit HCC and that this inhibition may be compromised upon the appearance of C-terminally truncated HBx or when the expression and/or activity of FXR is decreased. (Hepatology 2017;65:893-906).


Subject(s)
Carcinoma, Hepatocellular/genetics , Hepatitis C/complications , RNA-Binding Proteins/metabolism , Trans-Activators/genetics , Transcriptional Activation/genetics , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Silencing , Hepatitis B virus/genetics , Hepatitis C/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic , Random Allocation , Viral Regulatory and Accessory Proteins
13.
Mol Pharmacol ; 92(4): 425-436, 2017 10.
Article in English | MEDLINE | ID: mdl-28739572

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a prevalent chronic liver disease. The incidence of NAFLD has increased steadily due to its close association with the global epidemic of obesity and type 2 diabetes. However, there is no effective pharmacological therapy approved for NAFLD. Farnesoid X receptor (FXR), a member of the nuclear receptor subfamily, plays important roles in maintaining the homeostasis of bile acids, glucose, and lipids. FXR agonists have shown promise for the treatment of NAFLD. In this study, we report altenusin (2076A), a natural nonsteroidal fungal metabolite, as a novel selective agonist of FXR with an EC50 value of 3.2 ± 0.2 µM. Administration of 2076A protected mice from high-fat diet (HFD)-induced obesity by reducing the body weight and fat mass by 22.9% and 50.0%, respectively. Administration of 2076A also decreased the blood glucose level from 178.3 ± 12.4 mg/dl to 116.2 ± 4.1 mg/dl and the serum insulin level from 1.4 ± 0.6 ng/dl to 0.4 ± 0.1 ng/dl. Moreover, 2076A treatment nearly reversed HFD-induced hepatic lipid droplet accumulation and macrovesicular steatosis. These metabolic effects were abolished in FXR knockout mice. Mechanistically, the metabolic benefits of 2076A might have been accounted for by the increased insulin sensitivity and suppression of genes that are involved in hepatic gluconeogenesis and lipogenesis. In summary, we have uncovered a new class of nonsteroidal FXR agonist that shows promise in treating NAFLD and the associated metabolic syndrome.


Subject(s)
Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Biphenyl Compounds/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Docking Simulation/methods , Protein Structure, Secondary , Receptors, Cytoplasmic and Nuclear/chemistry
14.
Am J Pathol ; 186(10): 2614-22, 2016 10.
Article in English | MEDLINE | ID: mdl-27520356

ABSTRACT

Liver X receptors (LXRs) were identified as receptors that sense oxidized cholesterol derivatives. LXRs are best known for their hepatic functions in regulating cholesterol metabolism and triglyceride synthesis, but whether and how LXRs play a role in the lung diseases is less understood. To study the function of LXRs in acute respiratory distress syndrome (ARDS), we applied the oleic acid (OA) model of ARDS to mice whose LXR was genetically or pharmacologically activated. The VP-LXRα knock-in (LXR-KI) mice, in which a constitutively activated LXRα (VP-LXRα) was inserted into the mouse LXRα locus, were used as the genetic gain-of-function model. We showed that the OA-induced lung damages, including the cytokine levels and total cell numbers and neutrophil numbers in the bronchoalveolar lavage fluid, the wet/dry weight ratio, and morphological abnormalities were reduced in the LXR-KI mice and wild-type mice treated with the LXR agonist GW3965. The pulmonoprotective effect of GW3965 was abolished in the LXR-null mice. Consistent with the pulmonoprotective effect of LXR and the induction of antioxidant enzymes by LXR, the OA-induced suppression of superoxide dismutase and catalase was attenuated in LXR-KI mice and GW3965-treated wild-type mice. Taken together, our results demonstrate that activation of LXRs can alleviate OA-induced ARDS by attenuating the inflammatory response and enhancing antioxidant capacity.


Subject(s)
Benzoates/pharmacology , Benzylamines/pharmacology , Cholesterol/metabolism , Liver X Receptors/metabolism , Oleic Acid/adverse effects , Respiratory Distress Syndrome/prevention & control , Animals , Antioxidants/metabolism , Cytokines/metabolism , Female , Liver X Receptors/genetics , Lung/drug effects , Lung/immunology , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Respiratory Distress Syndrome/chemically induced , Respiratory Distress Syndrome/immunology
15.
J Biol Chem ; 290(23): 14754-64, 2015 Jun 05.
Article in English | MEDLINE | ID: mdl-25922074

ABSTRACT

Estrogen sulfotransferase (EST) regulates estrogen homeostasis by sulfonating and deactivating estrogens. Liver ischemia and reperfusion (I/R) involves both hypoxia during the ischemic phase and oxidative damage during the reperfusion phase. In this report, we showed that the expression of EST was markedly induced by I/R. Mechanistically, oxidative stress-induced activation of Nrf2 was responsible for the EST induction, which was abolished in Nrf2(-/-) mice. EST is a direct transcriptional target of Nrf2. In female mice, the I/R-responsive induction of EST compromised estrogen activity. EST ablation attenuated I/R injury as a result of decreased estrogen deprivation, whereas this benefit was abolished upon ovariectomy. The effect of EST ablation was sex-specific because the EST(-/-) males showed heightened I/R injury. Reciprocally, both estrogens and EST regulate the expression and activity of Nrf2. Estrogen deprivation by ovariectomy abolished the I/R-responsive Nrf2 accumulation, whereas the compromised estrogen deprivation in EST(-/-) mice was associated with increased Nrf2 accumulation. Our results suggested a novel I/R-responsive feedback mechanism to limit the activity of Nrf2 in which Nrf2 induces the expression of EST, which subsequently increases estrogen deactivation and limits the estrogen-responsive activation of Nrf2. Inhibition of EST, at least in females, may represent an effective approach to manage hepatic I/R injury.


Subject(s)
Liver/pathology , Oxidative Stress , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Sulfotransferases/genetics , Animals , Cells, Cultured , Estrogens/metabolism , Female , Gene Deletion , Hep G2 Cells , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Reperfusion Injury/metabolism , Sex Factors , Sulfotransferases/metabolism , Up-Regulation
16.
Hepatology ; 61(6): 1908-19, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25614121

ABSTRACT

UNLABELLED: The aryl hydrocarbon receptor (AHR), also known as the dioxin receptor, was originally characterized as a xenobiotic receptor that senses xenotoxicants. We investigated the endobiotic and hepatic role of AHR in fatty liver and energy metabolism and identified the endocrine factor that mediates the metabolic function of AHR. Wild-type and liver-specific constitutively activated human AHR transgenic mice were used to investigate the role of AHR in fatty liver and energy homeostasis. Adenovirus expressing short hairpin RNA targeting fibroblast growth factor 21 (FGF21) were used to determine the involvement of FGF21 in the metabolic effect of AHR. We showed that, despite their severe fatty liver, the transgenic mice were protected from diet-induced obesity and type 2 diabetes. We identified the endocrine hormone FGF21 as a mediator for the metabolic benefit of AHR and established FGF21 as a direct transcriptional target of AHR. Interestingly, the transactivation of FGF21 by AHR contributed to both hepatic steatosis and systemic insulin hypersensitivity, both of which were largely abolished upon FGF21 knockdown. CONCLUSIONS: The AHR-FGF21 endocrine signaling pathway establishes AHR as a pivotal environmental modifier that integrates signals from chemical exposure in the regulation of lipid and energy metabolism.


Subject(s)
Fatty Liver/metabolism , Fibroblast Growth Factors/metabolism , Insulin Resistance , Receptors, Aryl Hydrocarbon/metabolism , Animals , Diet, High-Fat/adverse effects , Fatty Liver/etiology , Gene Knockdown Techniques , Genetic Pleiotropy , Humans , Mice, Transgenic , Obesity/etiology
17.
J Biol Chem ; 289(12): 8086-97, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24497646

ABSTRACT

The steroid sulfatase (STS)-mediated desulfation is a critical metabolic mechanism that regulates the chemical and functional homeostasis of endogenous and exogenous molecules. In this report, we first showed that the liver expression of Sts was induced in both the high fat diet (HFD) and ob/ob models of obesity and type 2 diabetes and during the fed to fasting transition. In defining the functional relevance of STS induction in metabolic disease, we showed that overexpression of STS in the liver of transgenic mice alleviated HFD and ob/ob models of obesity and type 2 diabetes, including reduced body weight, improved insulin sensitivity, and decreased hepatic steatosis and inflammation. Interestingly, STS exerted its metabolic benefit through sex-specific mechanisms. In female mice, STS may have increased hepatic estrogen activity by converting biologically inactive estrogen sulfates to active estrogens and consequently improved the metabolic functions, whereas ovariectomy abolished this protective effect. In contrast, the metabolic benefit of STS in males may have been accounted for by the male-specific decrease of inflammation in white adipose tissue and skeletal muscle as well as a pattern of skeletal muscle gene expression that favors energy expenditure. The metabolic benefit in male STS transgenic mice was retained after castration. Treatment with the STS substrate estrone sulfate also improved metabolic functions in both the HFD and ob/ob models. Our results have uncovered a novel function of STS in energy metabolism and type 2 diabetes. Liver-specific STS induction or estrogen/estrogen sulfate delivery may represent a novel approach to manage metabolic syndrome.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Liver/enzymology , Obesity/genetics , Steryl-Sulfatase/genetics , Up-Regulation , Animals , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Energy Metabolism , Estrogens/metabolism , Fatty Liver/enzymology , Fatty Liver/genetics , Fatty Liver/pathology , Female , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Transgenic , Obesity/enzymology , Obesity/pathology , Steryl-Sulfatase/metabolism
18.
J Hepatol ; 63(4): 855-62, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26070408

ABSTRACT

BACKGROUND & AIMS: Fatty acid binding protein 4 (FABP4) has been known as a mediator of inflammatory response in the macrophages and adipose tissue, but its hepatic function is poorly understood. The goal of this study is to investigate the role of FABP4 in liver ischemia/reperfusion (I/R), a clinical condition that involves both hypoxia and inflammation. METHODS: To examine the I/R regulation of FABP4, mice were subjected to I/R surgery before being measured for FABP4 gene expression. Both loss-of-function (by using a pharmacological FABP4 inhibitor) and gain-of-function (by adenoviral overexpression of FABP4) were used to determine the functional relevance of FABP4 expression and its regulation during I/R. To determine the hypoxia responsive regulation of FABP4, primary mouse hepatocytes were exposed to hypoxia. The FABP4 gene promoter was cloned and its regulation by hypoxia inducible factor 1α (HIF-1α) was characterized by luciferase reporter gene, electrophoretic mobility shift, and chromatin immunoprecipitation assays. RESULTS: We found that the hepatic expression of FABP4 was markedly induced by I/R. At the functional level, pharmacological inhibition of FABP4 alleviated the I/R injury, whereas adenoviral overexpression of FABP4 sensitized mice to I/R injury. We also showed that exposure of primary hepatocytes to hypoxia or transgenic overexpression of HIF-1α in the mouse liver was sufficient to induce the expression of FABP4. Our promoter analysis established FABP4 as a novel transcriptional target of HIF-1α. CONCLUSIONS: FABP4 is a hypoxia inducible gene that sensitizes mice to liver I/R injury. FABP4 may represent a novel therapeutic target, and FABP4 inhibitors may be used as therapeutic agents to manage hepatic I/R injury.


Subject(s)
DNA/genetics , Fatty Acid-Binding Proteins/genetics , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Ischemia/genetics , Liver/blood supply , Reperfusion Injury/genetics , Animals , Blotting, Western , Cell Hypoxia , Cells, Cultured , Chromatin Immunoprecipitation , Disease Models, Animal , Fatty Acid-Binding Proteins/biosynthesis , Female , Hepatocytes/metabolism , Hepatocytes/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Ischemia/etiology , Ischemia/metabolism , Liver/pathology , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Reperfusion Injury/complications , Reperfusion Injury/metabolism
19.
Drug Metab Dispos ; 42(10): 1684-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25028566

ABSTRACT

The nuclear receptor liver X receptor (LXR) plays an important role in the metabolism and homeostasis of cholesterol, lipids, bile acids, and steroid hormones. In this study, we uncovered a function of LXRα (NR1H3) in regulating the human hydroxysteroid sulfotransferase SULT2A1, a phase II conjugating enzyme known to sulfonate bile acids, hydroxysteroid dehydroepiandrosterone, and related androgens. We showed that activation of LXR induced the expression of SULT2A1 at mRNA, protein, and enzymatic levels. A combination of promoter reporter gene and chromatin immunoprecipitation assays showed that LXRα transactivated the SULT2A1 gene promoter through its specific binding to the -500- to -258-base pair region of the SULT2A1 gene promoter. LXR small interfering RNA knockdown experiments suggested that LXRα, but not LXRß, played a dominant role in regulating SULT2A1. In primary human hepatocytes, we found a positive correlation between the expression of SULT2A1 and LXRα, which further supported the regulation of SULT2A1 by LXRα. In summary, our results established human SULT2A1 as a novel LXRα target gene. The expression of LXRα is a potential predictor for the expression of SULT2A1 in human liver.


Subject(s)
Gene Expression Regulation , Orphan Nuclear Receptors/metabolism , Sulfotransferases/genetics , Transcription, Genetic , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Humans , Liver X Receptors , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/pharmacology , Transcription, Genetic/drug effects
20.
Acta Pharm Sin B ; 14(3): 1241-1256, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38487006

ABSTRACT

Sulfation is a crucial and prevalent conjugation reaction involved in cellular processes and mammalian physiology. 3'-Phosphoadenosine 5'-phosphosulfate (PAPS) synthase 2 (PAPSS2) is the primary enzyme to generate the universal sulfonate donor PAPS. The involvement of PAPSS2-mediated sulfation in adenomatous polyposis coli (APC) mutation-promoted colonic carcinogenesis has not been reported. Here, we showed that the expression of PAPSS2 was decreased in human colon tumors along with cancer stages, and the lower expression of PAPSS2 was correlated with poor prognosis in advanced colon cancer. Gut epithelial-specific heterozygous Apc deficient and Papss2-knockout (ApcΔgut-HetPapss2Δgut) mice were created, and the phenotypes were compared to the spontaneous intestinal tumorigenesis of ApcΔgut-Het mice. ApcΔgut-HetPapss2Δgut mice were more sensitive to gut tumorigenesis, which was mechanistically accounted for by the activation of Wnt/ß-catenin signaling pathway due to the suppression of chondroitin sulfation and inhibition of the farnesoid X receptor (FXR)-transducin-like enhancer of split 3 (TLE3) gene regulatory axis. Chondroitin sulfate supplementation in ApcΔgut-HetPapss2Δgut mice alleviated intestinal tumorigenesis. In summary, we have uncovered the protective role of PAPSS2-mediated chondroitin sulfation and bile acids-FXR-TLE3 activation in the prevention of gut carcinogenesis via the antagonization of Wnt/ß-catenin signaling. Chondroitin sulfate may be explored as a therapeutic agent for Papss2 deficiency-associated colonic carcinogenesis.

SELECTION OF CITATIONS
SEARCH DETAIL