Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Sci Rep ; 14(1): 110, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167633

ABSTRACT

Non-alcoholic fatty liver disease is a common liver disease worldwide, and is associated with dysregulation of lipid metabolism, leading to inflammation and fibrosis. Acanthopanax senticosus Harms (ASH) is widely used in traditional medicine as an adaptogen food. We examined the effect of ASH on steatohepatitis using a high-fat diet mouse model. Mice were fed a choline-deficient, L-amino acid-defined, high-fat diet with ASH extract (ASHE). After 6 weeks, liver RNA transcriptome sequencing (RNA-Seq) was performed, followed by Ingenuity Pathway Analysis (IPA). Our findings revealed that mice fed a high-fat diet with 5% ASHE exhibited significantly reduced liver steatosis. These mice also demonstrated alleviated inflammation and reduced fibrosis in the liver. IPA of RNA-Seq indicated that hepatocyte nuclear factor 4 alpha (HNF4 alpha), a transcription factor, was the activated upstream regulator (P-value 0.00155, z score = 2.413) in the liver of ASHE-fed mice. Adenosine triphosphate binding cassette transporter 8 and carboxylesterase 2, downstream targets of HNF4 alpha pathway, were upregulated. Finally, ASHE-treated HepG2 cells exposed to palmitate exhibited significantly decreased lipid droplet contents. Our study provides that ASHE can activate HNF4 alpha pathway and promote fat secretion from hepatocytes, thereby serving as a prophylactic treatment for steatohepatitis in mice.


Subject(s)
Eleutherococcus , Non-alcoholic Fatty Liver Disease , Animals , Mice , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Eleutherococcus/chemistry , Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Inflammation/pathology , Disease Models, Animal , Fibrosis , Mice, Inbred C57BL , Diet, High-Fat/adverse effects
2.
J Ethnopharmacol ; 267: 113471, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33075440

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Vinegar-baked Radix Bupleuri (VBRB) is a processed form of Bupleurum chinense DC. As a well-known meridian-guiding drug, it is traditionally used as a component of traditional Chinese medicine formulations indicated for the treatment of liver diseases. However, the liver targeting component in VBRB remains unclear. Therefore, this study aims to explore the efficacy and mechanism of PSS (polysaccharides in Vinegar-baked Radix Bupleuri) in enhancing liver targeting. MATERIALS AND METHODS: Drug distribution of OM alone or combined with PSS was investigated in vivo. Relative uptake efficiency (RUE) and relative targeting efficiency (RTE) were calculated to evaluate liver targeting efficiency. The mRNA and protein expression of organic cation transporter 1 (OCT1), multi-drug resistance protein 2 (Mrp2), and hepatocyte nuclear factor 4α (HNF4α) in the liver were determined by q-PCR and Western blot. Then, AZT, the inhibitor of OCT1 and BI6015, the inhibitor of HNF4α were used to investigate regulatory mechanisms involved in the uptake of OM in the cell. At last, the role of PSS in the anti-hepatitis B virus (HBV) was explored on HepG2.2.15. RESULTS: PSS increased the AUC of OM in the liver and increase the RUE and RTE in the liver which indicated a liver targeting enhancing effect. The mRNA and protein expression of OCT1 was increased while Mrp2 and HNF4α decreased. PSS could increase the uptake of OM in HepG2 by increasing the protein expression of HNF4α and OCT1, while inhibited Mrp2. Moreover, PSS combined with OM could enhance the anti-HBV effect of OM. CONCLUSION: PSS enhanced the liver targeting efficiency and the underlying mechanism related to up-regulating the expression of OCT1 and HNF4α, while down-regulating of Mrp2. These results suggest that PSS may become a potential excipient and provide a new direction for new targeted research.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Acetic Acid/chemistry , Alkaloids/pharmacology , Alkaloids/pharmacokinetics , Catecholamine Plasma Membrane Transport Proteins/metabolism , Cooking , Hepatocyte Nuclear Factor 4/metabolism , Liver/drug effects , Multidrug Resistance-Associated Proteins/metabolism , Plant Extracts/pharmacology , Polysaccharides/pharmacology , Quinolizines/pharmacology , Quinolizines/pharmacokinetics , ATP-Binding Cassette Transporters/genetics , Animals , Bupleurum/chemistry , Catecholamine Plasma Membrane Transport Proteins/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Hot Temperature , Humans , Liver/metabolism , Male , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Plant Extracts/chemistry , Polysaccharides/chemistry , Rats, Sprague-Dawley , Tissue Distribution
3.
Int J Mol Sci ; 21(21)2020 Oct 24.
Article in English | MEDLINE | ID: mdl-33114319

ABSTRACT

Hepatocyte nuclear factor 4α (HNF4α) is a ligand-sensing transcription factor and presents as a potential drug target in metabolic diseases and cancer. In humans, mutations in the HNF4α gene cause maturity-onset diabetes of the young (MODY), and the elevated activity of this protein has been associated with gastrointestinal cancers. Despite the high therapeutic potential, available ligands and structure-activity relationship knowledge for this nuclear receptor are scarce. Here, we disclose a chemically diverse collection of orthogonally validated fragment-like activators as well as inverse agonists, which modulate HNF4α activity in a low micromolar range. These compounds demonstrate the druggability of HNF4α and thus provide a starting point for medicinal chemistry as well as an early tool for chemogenomics.


Subject(s)
Hepatocyte Nuclear Factor 4/chemistry , Hepatocyte Nuclear Factor 4/metabolism , Small Molecule Libraries/pharmacology , Calorimetry , Drug Discovery , Drug Evaluation, Preclinical , Fructose-Bisphosphatase/genetics , Gene Expression Regulation/drug effects , Hep G2 Cells , Hepatocyte Nuclear Factor 4/genetics , Humans , Ligands , Molecular Structure , Small Molecule Libraries/chemistry , Structure-Activity Relationship
4.
Am J Chin Med ; 48(6): 1435-1454, 2020.
Article in English | MEDLINE | ID: mdl-32907363

ABSTRACT

Endoplasmic reticulum stress (ER stress) plays a main role in pancreatic [Formula: see text]-cell dysfunction and death because of intracellular Ca[Formula: see text] turbulence and inflammation activation. Although several drugs are targeting pancreatic [Formula: see text]-cell to improve [Formula: see text]-cell function, there still lacks agents to alleviate [Formula: see text]-cell ER stress conditions. Therefore we used thapsigargin (THAP) or high glucose (HG) to induce ER stress in [Formula: see text]-cell and aimed to screen natural molecules against ER stress-induced [Formula: see text]-cell dysfunction. Through screening the Traditional Chinese drug library ([Formula: see text] molecules), luteolin was finally discovered to improve [Formula: see text]-cell function. Cellular viability results indicated luteolin reduced the THAP or HG-induced [Formula: see text]-cell death and apoptosis through MTT and flow cytometry assay. Moreover, luteolin improved [Formula: see text]-cell insulin secretion ability under ER stress conditions. Also ER stress-induced intracellular Ca[Formula: see text] turbulence and inflammation activation were inhibited by luteolin treatment. Mechanically, luteolin inhibited HNF4[Formula: see text] signaling, which was induced by ER stress. Moreover, luteolin reduced the transcriptional level of HNF4[Formula: see text] downstream gene, such as Asnk4b and HNF1[Formula: see text]. Conversely HNF4[Formula: see text] knockdown abolished the effect of luteolin on [Formula: see text]-cell using siRNA. These results suggested the protective effect of luteolin on [Formula: see text]-cell was through HNF4[Formula: see text]/Asnk4b pathway. In conclusion, our study discovered that luteolin improved [Formula: see text]-cell function and disclosed the underlying mechanism of luteolin on [Formula: see text]-cell, suggesting luteolin is a promising agent against pancreatic dysfunction.


Subject(s)
Cell Survival/drug effects , Drugs, Chinese Herbal/chemistry , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Luteolin/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Thapsigargin/adverse effects , Apoptosis/drug effects , Calcium/metabolism , Cell Death/drug effects , Cells, Cultured , Drug Evaluation, Preclinical , Endoplasmic Reticulum Stress/physiology , Glucose/adverse effects , Insulin-Secreting Cells/metabolism , Luteolin/isolation & purification
5.
Chem Res Toxicol ; 33(7): 1798-1808, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32538071

ABSTRACT

Emodin is the main toxic component in Chinese medicinal herbs such as rhubarb. Our previous studies demonstrated that genetic polymorphisms of UDP-glucuronosyltransferase 2B7 (UGT2B7) had an effect on the glucuronidation and detoxification of emodin. This study aimed to reveal the transcriptional regulation mechanism of UGT2B7 on emodin glucuronidation and its effect on toxicity. Emodin glucuronic activity and genome and transcriptome data were obtained from 36 clinical human kidney tissues. The genome-wide association studies (GWAS) identified that four single nucleotide polymorphisms (SNPs) (rs6093966, rs2868094, rs2071197, and rs6073433), which were located on the hepatocyte nuclear factor 4α (HNF4A) gene, were significantly associated with the emodin glucuronidation (p < 0.05). Notably, rs2071197 was significantly associated with the gene expression of HNF4A and UGT2B7 and the glucuronidation of emodin. The gene expression of HNF4A showed a high correlation with UGT2B7 (R2 = 0.721, p = 5.83 × 10-11). The luciferase activity was increased 7.68-fold in 293T cells and 2.03-fold in HepG2 cells, confirming a significant transcriptional activation of UGT2B7 promoter by HNF4A. The knockdown of HNF4A in HepG2 cells (36.6%) led to a significant decrease of UGT2B7 (19.8%) and higher cytotoxicity (p < 0.05). The overexpression of HNF4A in HepG2 cells (31.2%) led to a significant increase of UGT2B7 (24.4%) and improved cell viability (p < 0.05). Besides, HNF4A and UGT2B7 were both decreased in HepG2 cells and rats after treatment with emodin. In conclusion, emodin used long term or in high doses could inhibit the expression of HNF4A, thereby reducing the expression of UGT2B7 and causing hepatotoxicity.


Subject(s)
Emodin/pharmacokinetics , Glucuronides/metabolism , Glucuronosyltransferase/genetics , Hepatocyte Nuclear Factor 4/genetics , Animals , Cell Line , Emodin/pharmacology , Genome-Wide Association Study , Glucuronosyltransferase/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Humans , Kidney/metabolism , Male , Polymorphism, Single Nucleotide , Rats, Sprague-Dawley
6.
Pharmacol Res ; 157: 104840, 2020 07.
Article in English | MEDLINE | ID: mdl-32353589

ABSTRACT

Hepatocyte nuclear factor 4 alpha (HNF4α) regulates the expression of essential genes involved in very-low-density lipoprotein (VLDL) homeostasis and gluconeogenesis. 18ß-glycyrrhetinic acid (GA) is an active ingredient of Glycyrrhiza uralensis an herbal medicine used for treating liver aliments. In this study, we established that GA functions as a partial antagonist of HNF4α through HNF4α-driven reporter luciferase assay and co-immunoprecipitation experiments with co-activator PGC1α. By virtual docking and site-directed mutagenesis analysis, we confirmed that serine 190 and arginine 235 of HNF4α are both essential for GA to exert its antagonistic action on HNF4α. Importantly, GA suppressed the expression of HNF4α target genes such as apolipoprotein B (ApoB), microsomal triglyceride transfer protein (MTP) and phospholipase A2 G12B (PLA2G12B) modulating hepatic VLDL secretion in mice fed on a high fat diet. In addition, GA also suppressed gluconeogenesis and ameliorated glucose intolerance via down-regulating the expression of HNF4α target genes glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase (Pepck). Furthermore, GA significantly lowered blood glucose and improved insulin resistance in db/db mice. In all, we established that GA acts as a partial HNF4α antagonist modulating lipid and carbohydrate metabolism.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Glycyrrhetinic Acid/analogs & derivatives , Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Lipids/blood , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Blood Glucose/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Disease Models, Animal , Gene Expression Regulation , Gluconeogenesis/drug effects , Glycyrrhetinic Acid/pharmacology , HEK293 Cells , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Insulin Resistance , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Signal Transduction
7.
J Agric Food Chem ; 68(16): 4641-4649, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32249565

ABSTRACT

The intestinal epithelium barrier functions to protect human bodies from damages such as harmful microorganisms, antigens, and toxins. In this study, we evaluated the protective effect and molecular mechanism of a dominant polymethoxyflavone nobiletin (NOB) from tangerine peels on intestinal epithelial integrity. The results from transepithelial electrical resistance (TEER) suggested that NOB pretreatment counteracts epithelial injury induced by inflammatory cytokines (TEER value in 48 h: vehicle, 135.6 ± 3.9 Ω/cm2; TNF-α + IL-1ß, 90.7 ± 0.5 Ω/cm2; 10 µM NOB + TNF-α + IL-1ß, 126.1 ± 0.8 Ω/cm2; 100 µM NOB + TNF-α + IL-1ß, 125.3 ± 0.5 Ω/cm2. P < 0.001). Clinical and pathological test results suggested that administration of NOB effectively alleviates intestinal barrier injury induced by dextran sulfate sodium (DSS) as evidenced by the length of colon villi on day 7 (control, 253.7 ± 4.8 µm, DSS 131.6 ± 4.6 µm, NOB + DSS, 234.5 ± 5.1 µm. P < 0.001). Interestingly, when screening tight junction molecules for intestinal barrier integrity, we observed that independent treatment with NOB sharply increased claudin-7 levels (ratio of claudin-7 over GAPDH: control, 1.0 ± 0.06; DSS, 0.02 ± 0.001; NOB + DSS, 0.3 ± 0.07. P < 0.001), which was previously suppressed upon DSS stimulation. Furthermore, hepatocyte nuclear factor 4α (HNF-4α) transcriptional regulation of claudin-7 contributed to intestinal barrier homeostasis. Therefore, our study suggests potential intestinal protective strategies based on polymethoxyflavones of aged tangerine peels.


Subject(s)
Claudins/metabolism , Colitis/drug therapy , Drugs, Chinese Herbal/administration & dosage , Flavones/administration & dosage , Hepatocyte Nuclear Factor 4/metabolism , Intestinal Mucosa/drug effects , Animals , Caco-2 Cells , Claudins/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Cytokines/genetics , Cytokines/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Hepatocyte Nuclear Factor 4/genetics , Humans , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
8.
J Biol Chem ; 295(12): 3891-3905, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32051143

ABSTRACT

Epidemiological studies have suggested a link between vitamin D deficiency and increased risk for nonalcoholic fatty liver disease (NAFLD); however, the underlying mechanisms have remained unclear. Here, using both clinical samples and experimental rodent models along with several biochemical approaches, we explored the specific effects and mechanisms of vitamin D deficiency in NAFLD pathology. Serum vitamin D levels were significantly lower in individuals with NAFLD and in high-fat diet (HFD)-fed mice than in healthy controls and chow-fed mice, respectively. Vitamin D supplementation ameliorated HFD-induced hepatic steatosis and insulin resistance in mice. Hepatic expression of vitamin D receptor (VDR) was up-regulated in three models of NAFLD, including HFD-fed mice, methionine/choline-deficient diet (MCD)-fed mice, and genetically obese (ob/ob) mice. Liver-specific VDR deletion significantly exacerbated HFD- or MCD-induced hepatic steatosis and insulin resistance and also diminished the protective effect of vitamin D supplementation on NAFLD. Mechanistic experiments revealed that VDR interacted with hepatocyte nuclear factor 4 α (HNF4α) and that overexpression of HNF4α improved HFD-induced NAFLD and metabolic abnormalities in liver-specific VDR-knockout mice. These results suggest that vitamin D ameliorates NAFLD and metabolic abnormalities by activating hepatic VDR, leading to its interaction with HNF4α. Our findings highlight a potential value of using vitamin D for preventing and managing NAFLD by targeting VDR.


Subject(s)
Hepatocyte Nuclear Factor 4/metabolism , Non-alcoholic Fatty Liver Disease/prevention & control , Protective Agents/administration & dosage , Receptors, Calcitriol/metabolism , Vitamin D/administration & dosage , Animals , Diet, High-Fat , Disease Models, Animal , Glucose Tolerance Test , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Obesity/metabolism , Obesity/pathology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/genetics , Up-Regulation , Vitamin D/blood
9.
Biomed Pharmacother ; 105: 144-150, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29852391

ABSTRACT

The association between chronic alcohol consumption and the development of alcpholic liver disease is a very well known phenomenon, but the precise underlying molecular mediators involved in ethanol-induced liver disease remain elusive. This study aimed to characterize the lipid metabolism alterations and the molecular mediators which are related to lipid metabolism in liver under the heavy ethanol exposure alone or combined with ginger extract. Twenty-four male wistar rats were assigned into three groups, namely control, ethanol, and ginger extract treated ethanol (GETE) groups. Six weeks after the treatment, the ethanol group showed a significant increase in fatty acid translocase (FAT)/CD36, protein tyrosine phosphatase 1B (PTP1B) and decrease hepatocyte nuclear factor 4 Alpha (HNF4A) genes expressions compared to the control group. The ethanol administration also significantly increased plasma LDL, cholesterol, triglyceride, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) compared to the control group. Moreover, compared to the control group, the ethanol group showed liver histhological changes, such as fibrosis, focal microvesicular steatosis, some apoptotic hepatocytes, spotty necrosis, portal lymphocytic inflammation, mallory-denk bodies, giant mitochondria, piecemeal necrosis. Consumption of ginger extract along with ethanol, partially ameliorated gene expression alteration and histological changes, improved undesirable lipid profile and liver enzymes changes compare to those in the ethanol group. These findings indicate that ethanol-induced liver abnormalities may in part be associated with lipid homeostasis changes mediated by overexpression of FAT/CD36, PTP1B and downexpressionof HNF4A genes. It also show that these effects can be reduced by using ginger extract as an antioxidant and anti-inflammatory agent.


Subject(s)
CD36 Antigens/metabolism , Dyslipidemias/drug therapy , Hepatocyte Nuclear Factor 4/metabolism , Liver Diseases, Alcoholic/drug therapy , Plant Extracts/therapeutic use , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Zingiber officinale/chemistry , Animals , CD36 Antigens/genetics , Dyslipidemias/complications , Dyslipidemias/metabolism , Gene Expression/drug effects , Hepatocyte Nuclear Factor 4/genetics , Lipid Metabolism/drug effects , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver Diseases, Alcoholic/complications , Liver Diseases, Alcoholic/metabolism , Liver Function Tests , Male , Plant Extracts/isolation & purification , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Rats, Wistar
10.
Life Sci ; 180: 68-74, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28504115

ABSTRACT

AIMS: Protocatechuic acid (PCA) is a phenolic compound found in many antiviral Chinese herbal medicines. HNF4α and HNF1α, the members of hepatocyte nuclear factor (HNF) family, play an important regulatory role in the gene transcription of hepatitis B virus (HBV). Previous studies found that PCA inhibited HBV antigen secretion and HBV DNA replication in HepG2.2.15 cells, but its anti-HBV mechanism has not been fully understood. We aim to illustrate the anti-HBV mechanism of PCA. MATERIALS AND METHODS: MTT was used to estimate cytotoxicity. The content of HBsAg or HBeAg was detected using an enzyme-linked immunosorbent assay kit. HBV DNA in cell-free culture media was detected by PCR kit. HNF1α and HNF4α mRNA expression was detected by real-time PCR. HNF1α, HNF4α and ERK1/2 protein expression was detected by western blotting and HBV promoter activity was tested by luciferase reporter assay. KEY FINDINGS: Our results demonstrated that PCA inhibited the gene transcription and protein translation of HNF1α and HNF4α in Huh7 and HepG2.2.15 cells, as well as the promoter activities of HBV X and preS1 in Huh7 cells transfected with the luciferase reporter plasmid of HBV promoter. Further study suggested that PCA induced the phosphorylation of extracellular-signal-related kinase (ERK) 1/2, and thereby inhibited HNF4α and HNF1α expression in HepG2.2.15 cells to exert its antiviral activity. SIGNIFICANCE: To our knowledge, this study is the first to reveal the anti-HBV mechanism of PCA. Our results demonstrate that PCA inhibits HBV replication by activating ERK1/2 pathway and subsequently down-regulating HNF4α and HNF1α in HepG2.2.15 cells.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/drug effects , Hydroxybenzoates/pharmacology , Virus Replication/drug effects , Blotting, Western , Cell Line, Tumor , DNA, Viral , Down-Regulation/drug effects , Enzyme-Linked Immunosorbent Assay , Hep G2 Cells , Hepatitis B Surface Antigens/metabolism , Hepatitis B e Antigens/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Humans , MAP Kinase Signaling System/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transfection
11.
Best Pract Res Clin Endocrinol Metab ; 30(3): 455-67, 2016 06.
Article in English | MEDLINE | ID: mdl-27432078

ABSTRACT

It is increasingly recognised that diabetes in young adults has a wide differential diagnosis. There are many monogenic causes, including monogenic beta-cell dysfunction, mitochondrial diabetes and severe insulin resistance. Type 2 diabetes in the young is becoming more prevalent, particularly after adolescence. It's important to understand the clinical features and diagnostic tools available to classify the different forms of young adult diabetes. Classic type 1 diabetes is characterised by positive ß-cell antibodies and absence of endogenous insulin secretion. Young type 2 diabetes is accompanied by metabolic syndrome with obesity, hypertension and dyslipidaemia. Monogenic ß-cell dysfunction is characterised by non-autoimmune, C-peptide positive diabetes with a strong family history, while mitochondrial diabetes features deafness and other neurological involvement. Severe insulin resistance involves a young-onset metabolic syndrome often with a disproportionately low BMI. A suspected diagnosis of monogenic diabetes is confirmed with genetic testing, which is widely available in specialist centres across the world. Treatment of young adult diabetes is similarly diverse. Mutations in the transcription factors HNF1A and HNF4A and in the ß-cell potassium ATP channel components cause diabetes which responds to low dose and high dose sulfonylurea agents, respectively, while glucokinase mutations require no treatment. Monogenic insulin resistance and young-onset type 2 diabetes are both challenging to treat, but first line management involves insulin sensitisers and aggressive management of cardiovascular risk. Outcomes are poor in young-onset type 2 diabetes compared to both older onset type 2 and type 1 diabetes diagnosed at a similar age. The evidence base for treatments in monogenic and young-onset type 2 diabetes relies on studies of moderate quality at best and largely on extrapolation from work conducted in older type 2 diabetes subjects. Better quality, larger studies, particularly of newer agents would improve treatment prospects for young adults with diabetes.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Hypoglycemic Agents/therapeutic use , Mutation , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Humans , KATP Channels/genetics , Precision Medicine
12.
Drug Metab Rev ; 47(4): 470-519, 2015.
Article in English | MEDLINE | ID: mdl-26574146

ABSTRACT

With only 1.3-4.3% in total hepatic CYP content, human CYP2D6 can metabolize more than 160 drugs. It is a highly polymorphic enzyme and subject to marked inhibition by a number of drugs, causing a large interindividual variability in drug clearance and drug response and drug-drug interactions. The expression and activity of CYP2D6 are regulated by a number of physiological, pathological and environmental factors at transcriptional, post-transcriptional, translational and epigenetic levels. DNA hypermethylation and histone modifications can repress the expression of CYP2D6. Hepatocyte nuclear factor-4α binds to a directly repeated element in the promoter of CYP2D6 and thus regulates the expression of CYP2D6. Small heterodimer partner represses hepatocyte nuclear factor-4α-mediated transactivation of CYP2D6. GW4064, a farnesoid X receptor agonist, decreases hepatic CYP2D6 expression and activity while increasing small heterodimer partner expression and its recruitment to the CYP2D6 promoter. The genotypes are key determinants of interindividual variability in CYP2D6 expression and activity. Recent genome-wide association studies have identified a large number of genes that can regulate CYP2D6. Pregnancy induces CYP2D6 via unknown mechanisms. Renal or liver diseases, smoking and alcohol use have minor to moderate effects only on CYP2D6 activity. Unlike CYP1 and 3 and other CYP2 members, CYP2D6 is resistant to typical inducers such as rifampin, phenobarbital and dexamethasone. Post-translational modifications such as phosphorylation of CYP2D6 Ser135 have been observed, but the functional impact is unknown. Further functional and validation studies are needed to clarify the role of nuclear receptors, epigenetic factors and other factors in the regulation of CYP2D6.


Subject(s)
Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Gene Expression Regulation, Enzymologic , Precision Medicine/methods , Protein Processing, Post-Translational , Alzheimer Disease/enzymology , Animals , Arthritis, Rheumatoid/enzymology , Cytochrome P-450 CYP2D6/biosynthesis , Diabetes Mellitus/enzymology , Epigenomics , Gene Expression Regulation, Enzymologic/drug effects , Genome-Wide Association Study , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Humans , Inflammation/enzymology , Kidney Failure, Chronic/enzymology , Liver Cirrhosis, Alcoholic/enzymology , Liver Diseases/enzymology , Parkinson Disease/enzymology , Plant Preparations/pharmacology , Polymorphism, Genetic , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , Substrate Specificity
13.
PLoS One ; 10(3): e0119021, 2015.
Article in English | MEDLINE | ID: mdl-25774879

ABSTRACT

Major depressive disorder (MDD) is a common psychiatric disorder that involves marked disabilities in global functioning, anorexia, and severe medical comorbidities. MDD is associated with not only psychological and sociocultural problems, but also pervasive physical dysfunctions such as metabolic, neurobiological and immunological abnormalities. Nevertheless, the mechanisms underlying the interactions between these factors have yet to be determined in detail. The aim of the present study was to identify the molecular mechanisms responsible for the interactions between MDD and dysregulation of physiological homeostasis, including immunological function as well as lipid metabolism, coagulation, and hormonal activity in the brain. We generated depression-like behavior in mice using chronic mild stress (CMS) as a model of depression. We compared the gene expression profiles in the prefrontal cortex (PFC) of CMS and control mice using microarrays. We subsequently categorized genes using two web-based bioinformatics applications: Ingenuity Pathway Analysis and The Database for Annotation, Visualization, and Integrated Discovery. We then confirmed significant group-differences by analyzing mRNA and protein expression levels not only in the PFC, but also in the thalamus and hippocampus. These web tools revealed that hepatocyte nuclear factor 4 alpha (Hnf4a) may exert direct effects on various genes specifically associated with amine synthesis, such as genes involved in serotonin metabolism and related immunological functions. Moreover, these genes may influence lipid metabolism, coagulation, and hormonal activity. We also confirmed the significant effects of Hnf4a on both mRNA and protein expression levels in the brain. These results suggest that Hnf4a may have a critical influence on physiological homeostasis under depressive states, and may be associated with the mechanisms responsible for the interactions between MDD and the dysregulation of physiological homeostasis in humans.


Subject(s)
Depression/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hippocampus/metabolism , Prefrontal Cortex/metabolism , Thalamus/metabolism , Animals , Cells, Cultured , Depression/genetics , Depression/psychology , Disease Models, Animal , Gene Expression Profiling , Homeostasis , Male , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis
14.
PLoS One ; 9(8): e104133, 2014.
Article in English | MEDLINE | ID: mdl-25137413

ABSTRACT

AIM: To investigate the differentiation potential of human umbilical mesenchymal stem cells (HuMSCs) and the key factors that facilitate hepatic differentiation. METHODS: HuMSCs were induced to become hepatocyte-like cells according to a previously published protocol. The differentiation status of the hepatocyte-like cells was examined by observing the morphological changes under an inverted microscope and by immunofluorescence analysis. Hepatocyte nuclear factor 4 alpha (HNF4α) overexpression was achieved by plasmid transfection of the hepatocyte-like cells. The expression of proteins and genes of interest was then examined by Western blotting and reverse transcription-polymerase chain reaction (RT-PCR) or real-time RT-PCR methods. RESULTS: Our results demonstrated that HuMSCs can easily be induced into hepatocyte-like cells using a published differentiation protocol. The overexpression of HNF4α in the induced HuMSCs significantly enhanced the expression levels of hepatic-specific proteins and genes. HNF4α overexpression may be associated with liver-enriched transcription factor networks and the Wnt/ß-Catenin pathway. CONCLUSION: The overexpression of HNF4α improves the hepatic differentiation of HuMSCs and is a simple way to improve cellular sources for clinical applications.


Subject(s)
Fetal Blood/cytology , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/cytology , Mesenchymal Stem Cells/cytology , Activins/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/drug effects , Chondrocytes/metabolism , Epidermal Growth Factor/pharmacology , Fetal Blood/drug effects , Fetal Blood/metabolism , Gene Expression , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Insulin/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Niacinamide/pharmacology , Plasmids , Selenium/pharmacology , Signal Transduction , Transfection , Transferrin/pharmacology , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
15.
J Med Food ; 14(9): 930-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21812644

ABSTRACT

Tea has many beneficial effects. We have previously reported that green tea and a catechin-rich green tea beverage modulated the gene expression of the gluconeogenic enzymes glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) in the normal murine liver. In the present study, we examined the effects of oral administration of oolong tea on the hepatic expression of gluconeogenesis-related genes in the mouse. The intake of oolong tea for 4 weeks reduced the hepatic expression of G6Pase and PEPCK together with that of the transcription factor hepatocyte nuclear factor (HNF) 4α. When rat hepatoma H4IIE cells were incubated in the presence of oolong tea, the expression of these genes was repressed in accordance with the findings in vivo. The reduced protein expression of PEPCK and HNF4α was also demonstrated. We then fractionated oolong tea by sequential extraction with three organic solvents to give three fractions and the residual fraction (Fraction IV). In addition to organic fractions, Fraction IV, which was devoid of low-molecular-weight catechins such as (-)-epigallocatechin gallate (EGCG), had effects similar to those of oolong tea on H4IIE cells. Fraction IV repressed the gene expression of insulin-like growth factor binding protein 1, as insulin did. This activity was different from that of EGCG. The present findings suggest that drinking oolong tea may help to prevent diabetes and that oolong tea contains a component or components with insulin-like activity distinguishable from EGCG. Identification of such component(s) may open the way to developing a new drug for diabetes.


Subject(s)
Gene Expression Regulation, Enzymologic , Gluconeogenesis , Hypoglycemic Agents , Liver/enzymology , Tea , Animals , Cell Line, Tumor , Down-Regulation/drug effects , Drug Discovery , Gene Expression Regulation, Enzymologic/drug effects , Gluconeogenesis/drug effects , Gluconeogenesis/ethics , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hypoglycemic Agents/isolation & purification , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Insulin-Like Growth Factor Binding Protein 1/genetics , Insulin-Like Growth Factor Binding Protein 1/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , RNA, Messenger/metabolism , Rats , Tea/chemistry , Tea/metabolism
16.
Biomed Res ; 32(2): 119-25, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21551947

ABSTRACT

Many biological activities of green tea have been attributed to a major constituent, (-)-epigallocatechin gallate (EGCG). We previously reported that EGCG and a catechin-rich green tea beverage modulated the gene expression of gluconeogenic enzymes, glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK), in the mouse liver. However, it remains to be examined whether or not a constituent other than EGCG contributes to the change in gene expression of these enzymes. In this study, we separated the hot water infusion of green tea leaves (GT) into an ethanol-soluble fraction (GT-E) and an EGCG-free water-soluble fraction (GT-W), and examined their effects using rat hepatoma H4IIE cells. The inclusion of GT, GT-E, and GT-W in the culture medium reduced the gene expression of G6Pase and PEPCK. GT-W caused a decrease in expression of the transcription factor HNF4α. Reduced levels of PEPCK and HNF4α proteins were demonstrated in the cells treated with GT-W. GT-W showed an activity similar to insulin, but different from EGCG. Administration of GT-W to mice for 4 weeks reduced the hepatic expression of G6Pase, PEPCK, and HNF4α. These results suggest that green tea contains some component(s) with insulin-like activity distinguishable from EGCG and that drinking green tea may help to prevent diabetes.


Subject(s)
Antioxidants/pharmacology , Carcinoma, Hepatocellular/enzymology , Gluconeogenesis/drug effects , Glucose-6-Phosphatase/metabolism , Liver/drug effects , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , Plant Extracts/pharmacology , Animals , Carcinoma, Hepatocellular/genetics , Catechin/pharmacology , Cell Line, Tumor , Diabetes Mellitus, Type 2/prevention & control , Gene Expression , Gluconeogenesis/genetics , Glucose-6-Phosphatase/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Insulin/metabolism , Liver/enzymology , Male , Mice , Phosphoenolpyruvate Carboxykinase (ATP)/genetics , Plant Leaves/chemistry , RNA, Messenger/analysis , Rats , Tea/chemistry
17.
Biochem Biophys Res Commun ; 401(2): 213-8, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20846506

ABSTRACT

Suppression of the growth hormone/insulin-like growth factor-I pathway in Ames dwarf (DF) mice, and caloric restriction (CR) in normal mice extends lifespan and delays the onset of age-related disorders. In combination, these interventions have an additive effect on lifespan in Ames DF mice. Therefore, common signaling pathways regulated by DF and CR could have additive effects on longevity. In this study, we tried to identity the signaling mechanism and develop a system to assess pro-longevity status in cells and mice. We previously identified genes up-regulated in the liver of DF and CR mice by DNA microarray analysis. Motif analysis of the upstream sequences of those genes revealed four major consensus sequence motifs, which have been named dwarfism and calorie restriction-responsive elements (DFCR-REs). One of the synthesized sequences bound to hepatocyte nuclear factor-4α (HNF-4α), an important transcription factor involved in liver metabolism. Furthermore, using this sequence information, we developed a highly sensitive bioassay to identify chemicals mimicking the anti-aging effects of CR. When the reporter construct, containing an element upstream of a secreted alkaline phosphatase (SEAP) gene, was co-transfected with HNF-4α and its regulator peroxisome proliferator-activated receptor (PPAR) γ coactivator-1α (PGC-1α), SEAP activity was increased compared with untransfected controls. Moreover, transient transgenic mice established using this construct showed increased SEAP activity in CR mice compared with ad libitum-fed mice. These data suggest that because of its rapidity, ease of use, and specificity, our bioassay will be more useful than the systems currently employed to screen for CR mimetics, which mimic the beneficial effects of CR. Our system will be particularly useful for high-throughput screening of natural and synthetic candidate molecules.


Subject(s)
Biological Assay , Caloric Restriction , Longevity/drug effects , Alkaline Phosphatase/genetics , Animals , Base Sequence , Drug Evaluation, Preclinical , Dwarfism/genetics , Genes, Reporter , Hepatocyte Nuclear Factor 4/genetics , Insulin-Like Growth Factor I/genetics , Mice , Mice, Inbred ICR , Mice, Inbred Strains , Mice, Transgenic , Promoter Regions, Genetic
18.
Cell Transplant ; 18(12): 1319-40, 2009.
Article in English | MEDLINE | ID: mdl-19660180

ABSTRACT

Adipose tissue contains a mesenchymal stem cell (MSC) population known as adipose-derived stem cells (ASCs) capable of differentiating into different cell types. Our aim was to induce hepatic transdifferentiation of ASCs by sequential exposure to several combinations of cytokines, growth factors, and hormones. The most efficient hepatogenic protocol includes fibroblastic growth factors (FGF) 2 and 4 and epidermal growth factor (EGF) (step 1), hepatocyte growth factor (HGF), FGF2, FGF4, and nicotinamide (Nic) (step 2), and oncostatin M (OSM), dexamethasone (Dex), and insulin-tranferrin-selenium (step 3). This protocol activated transcription factors [GATA6, Hex, CCAAT/enhancer binding protein alpha and beta (CEBPalpha and beta), peroxisome proliferator-activated receptor-gamma, coactivator 1 alpha (PGC1alpha), and hepatocyte nuclear factor 4 alpha (HNF4alpha)], which promoted a characteristic hepatic phenotype, as assessed by new informative markers for the step-by-step hepatic transdifferentiation of hMSC [early markers: albumin (ALB), alpha-2-macroglobuline (alpha2M), complement protein C3 (C3), and selenoprotein P1 (SEPP1); late markers: cytochrome P450 3A4 (CYP3A4), apolipoprotein E (APOE), acyl-CoA synthetase long-chain family member 1 (ACSL1), and angiotensin II receptor, type 1 (AGTR1)]. The loss of adipose adult stem cell phenotype was detected by losing expression of Thy1 and inhibitor of DNA binding 3 (Id3). The reexpression of phosphoenolpyruvate corboxykinase (PEPCK), apolipoprotein C3 (APOCIII), aldolase B (ALDOB), and cytochrome P450 1A2 (CYP1A2) was achieved by transduction with a recombinant adenovirus for HNF4alpha and finally hepatic functionality was also assessed by analyzing specific biochemical markers. We conclude that ASCs could represent an alternative tool in clinical therapy for liver dysfunction and regenerative medicine.


Subject(s)
Adipose Tissue/cytology , Hepatocytes/cytology , Mesenchymal Stem Cells/metabolism , Transcription Factors/metabolism , Cell Transdifferentiation , Cells, Cultured , Dexamethasone/pharmacology , Fibroblast Growth Factors/pharmacology , Flow Cytometry , Gene Expression Profiling , Hep G2 Cells , Hepatocyte Growth Factor/pharmacology , Hepatocyte Nuclear Factor 4/genetics , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , Humans , Insulin/pharmacology , Mesenchymal Stem Cells/cytology , Niacinamide/pharmacology , Oncostatin M/pharmacology , Selenium/pharmacology , Signal Transduction , Transcription Factors/genetics , Transferrin/pharmacology
19.
Biochim Biophys Acta ; 1791(12): 1181-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19699314

ABSTRACT

Dietary n-3 polyunsaturated fatty acids (PUFA) suppress the secretion of very low density lipoprotein (VLDL) directly when delivered to the liver in chylomicron remnants (CMR). The role of sterol regulatory element-binding proteins (SREBPs) and hepatic nuclear factor-4alpha (HNF-4alpha) in the regulation of this effect was investigated. Chylomicron remnant-like particles (CRLPs) containing triacylglycerol (TG) from palm (rich in saturated fatty acids (SFA)) or fish (rich in n-3 PUFA) oil were incubated with cultured rat hepatocytes (24h) and the expression of protein and mRNA for SREBP-1, SREBP-2 and HNF-4alpha, and levels of mRNA for their target genes were determined. SREBP-1 and -2 protein expression in the membrane and nuclear fractions was unaffected by either type of CRLPs. mRNA abundance for SREBP-1c and -2 was also unchanged by CRLP-treatment, as were levels of mRNA for target genes of SREBP-1, including steroyl CoA desaturase, acetyl CoA carboxylase, fatty acid synthase and ATP citrate lyase, and SREBP-2 (3-hydroxy-3-methylglutaryl CoA reductase). In contrast, HNF-4alpha protein and mRNA levels were significantly decreased by CRLPs enriched in n-3 PUFA, but not SFA, and the expression of mRNA for HNF-4alpha target genes, including HNF-1alpha, apolipoprotein B and the microsomal TG transfer protein, was also lowered by n-3 PUFA-, but not SFA-enriched CRLPs. These findings suggest that the direct suppression of VLDL secretion by dietary n-3 PUFA delivered to the liver in CMR is mediated via decreased expression of HNF-4alpha.


Subject(s)
Chylomicron Remnants/pharmacology , Fatty Acids, Omega-3/pharmacology , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Lipoproteins, VLDL/metabolism , Animals , Apolipoproteins E/analysis , Cells, Cultured , Fish Oils/chemistry , Fish Oils/pharmacology , Gene Expression Regulation/drug effects , Hepatocyte Nuclear Factor 4/genetics , Male , Palm Oil , Plant Oils/chemistry , Plant Oils/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sterol Regulatory Element Binding Proteins/genetics , Sterol Regulatory Element Binding Proteins/metabolism
20.
J Med Food ; 12(2): 345-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19459736

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs), members of the nuclear hormone receptor family, are key regulators of various metabolic pathways related to lipid and glucose metabolism as well as inflammation. We examined the effect of zingerone, a major ingredient of ginger, on PPAR, hepatic nuclear factor-4 (HNF-4), and nuclear factor-kappaB (NF-kappaB) expression in 21-month-old male Sprague-Dawley rats. Two experimental groups receiving doses of either 2 or 8 mg/kg/day zingerone for 10 days were compared with young rats (6 months old) and an age-matched control group. For molecular work, the endothelial cell line YPEN-1 was used. Both the 2 and 8 mg/kg/day dose of zingerone significantly increased DNA binding activities of PPARs (2.8-fold). Expression of HNF-4 was also increased in the group receiving the 8 mg/kg/day dose. We further showed that zingerone partially prevented the age-related decline in PPAR expression. In vitro experiments revealed zingerone (10 microM) increased PPAR expression (2.5-fold) to a similar extent as the PPAR agonist fibrate (5 microM) and suppressed pro-inflammatory transcription factor NF-kappaB activity. Collectively, our findings suggest that zingerone exerts its potent anti-inflammatory action by increasing HNF-4 and PPAR activities, while suppressing NF-kappaB activity.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Guaiacol/analogs & derivatives , Hepatocyte Nuclear Factor 4/metabolism , NF-kappa B/antagonists & inhibitors , Peroxisome Proliferator-Activated Receptors/metabolism , Plant Extracts/pharmacology , Zingiber officinale/chemistry , Aging/metabolism , Animals , Cell Line , DNA/metabolism , Down-Regulation , Gene Expression , Guaiacol/pharmacology , Hepatocyte Nuclear Factor 4/genetics , Male , NF-kappa B/genetics , Peroxisome Proliferator-Activated Receptors/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL