Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
J Inherit Metab Dis ; 44(4): 879-892, 2021 07.
Article in English | MEDLINE | ID: mdl-33739445

ABSTRACT

Prevention of hypertriglyceridemia is one of the biomedical targets in Glycogen Storage Disease type Ia (GSD Ia) patients, yet it is unclear how hypoglycemia links to plasma triglyceride (TG) levels. We analyzed whole-body TG metabolism in normoglycemic (fed) and hypoglycemic (fasted) hepatocyte-specific glucose-6-phosphatase deficient (L-G6pc-/- ) mice. De novo fatty acid synthesis contributed substantially to hepatic TG accumulation in normoglycemic L-G6pc-/- mice. In hypoglycemic conditions, enhanced adipose tissue lipolysis was the main driver of liver steatosis, supported by elevated free fatty acid concentrations in GSD Ia mice and GSD Ia patients. Plasma very-low-density lipoprotein (VLDL) levels were increased in GSD Ia patients and in normoglycemic L-G6pc-/- mice, and further elevated in hypoglycemic L-G6pc-/- mice. VLDL-TG secretion rates were doubled in normo- and hypoglycemic L-G6pc-/- mice, while VLDL-TG catabolism was selectively inhibited in hypoglycemic L-G6pc-/- mice. In conclusion, fasting-induced hypoglycemia in L-G6pc-/- mice promotes adipose tissue lipolysis and arrests VLDL catabolism. This mechanism likely contributes to aggravated liver steatosis and dyslipidemia in GSD Ia patients with poor glycemic control and may explain clinical heterogeneity in hypertriglyceridemia between GSD Ia patients.


Subject(s)
Glucose/metabolism , Glycogen Storage Disease Type I/complications , Hypertriglyceridemia/etiology , Hypoglycemia/etiology , Lipoproteins, VLDL/metabolism , Triglycerides/metabolism , Adult , Aged , Animals , Disease Models, Animal , Fatty Liver/etiology , Female , Glucose-6-Phosphatase/genetics , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Hepatocytes/metabolism , Humans , Hypertriglyceridemia/prevention & control , Hypoglycemia/metabolism , Lipid Metabolism , Male , Mice , Middle Aged
2.
FEBS J ; 288(7): 2257-2277, 2021 04.
Article in English | MEDLINE | ID: mdl-33089625

ABSTRACT

Dietary protein restriction has been demonstrated to improve metabolic health under various conditions. However, the relevance of ageing and age-related decline in metabolic flexibility on the effects of dietary protein restriction has not been addressed. Therefore, we investigated the effect of short-term dietary protein restriction on metabolic health in young and aged mice. Young adult (3 months old) and aged (18 months old) C57Bl/6J mice were subjected to a 3-month dietary protein restriction. Outcome parameters included fibroblast growth factor 21 (FGF21) levels, muscle strength, glucose tolerance, energy expenditure (EE) and transcriptomics of brown and white adipose tissue (WAT). Here, we report that a low-protein diet had beneficial effects in aged mice by reducing some aspects of age-related metabolic decline. These effects were characterized by increased plasma levels of FGF21, browning of subcutaneous WAT, increased body temperature and EE, while no changes were observed in glucose homeostasis and insulin sensitivity. Moreover, the low-protein diet used in this study was well-tolerated in aged mice indicated by the absence of adverse effects on body weight, locomotor activity and muscle performance. In conclusion, our study demonstrates that a short-term reduction in dietary protein intake can impact age-related metabolic health alongside increased FGF21 signalling, without negatively affecting muscle function. These findings highlight the potential of protein restriction as a strategy to induce EE and browning of WAT in aged individuals.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Energy Metabolism/genetics , Fibroblast Growth Factors/genetics , Age Factors , Aging/genetics , Aging/metabolism , Animals , Caloric Restriction , Dietary Proteins/metabolism , Humans , Mice , Signal Transduction
3.
Hepatology ; 72(5): 1638-1653, 2020 11.
Article in English | MEDLINE | ID: mdl-32083759

ABSTRACT

BACKGROUND AND AIMS: Glycogen storage disease (GSD) type 1a is an inborn error of metabolism caused by defective glucose-6-phosphatase catalytic subunit (G6PC) activity. Patients with GSD 1a exhibit severe hepatomegaly due to glycogen and triglyceride (TG) accumulation in the liver. We have shown that the activity of carbohydrate response element binding protein (ChREBP), a key regulator of glycolysis and de novo lipogenesis, is increased in GSD 1a. In the current study, we assessed the contribution of ChREBP to nonalcoholic fatty liver disease (NAFLD) development in a mouse model for hepatic GSD 1a. APPROACH AND RESULTS: Liver-specific G6pc-knockout (L-G6pc-/- ) mice were treated with adeno-associated viruses (AAVs) 2 or 8 directed against short hairpin ChREBP to normalize hepatic ChREBP activity to levels observed in wild-type mice receiving AAV8-scrambled short hairpin RNA (shSCR). Hepatic ChREBP knockdown markedly increased liver weight and hepatocyte size in L-G6pc-/- mice. This was associated with hepatic accumulation of G6P, glycogen, and lipids, whereas the expression of glycolytic and lipogenic genes was reduced. Enzyme activities, flux measurements, hepatic metabolite analysis and very low density lipoprotein (VLDL)-TG secretion assays revealed that hepatic ChREBP knockdown reduced downstream glycolysis and de novo lipogenesis but also strongly suppressed hepatic VLDL lipidation, hence promoting the storage of "old fat." Interestingly, enhanced VLDL-TG secretion in shSCR-treated L-G6pc-/- mice associated with a ChREBP-dependent induction of the VLDL lipidation proteins microsomal TG transfer protein and transmembrane 6 superfamily member 2 (TM6SF2), the latter being confirmed by ChIP-qPCR. CONCLUSIONS: Attenuation of hepatic ChREBP induction in GSD 1a liver aggravates hepatomegaly because of further accumulation of glycogen and lipids as a result of reduced glycolysis and suppressed VLDL-TG secretion. TM6SF2, critical for VLDL formation, was identified as a ChREBP target in mouse liver. Altogether, our data show that enhanced ChREBP activity limits NAFLD development in GSD 1a by balancing hepatic TG production and secretion.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Glycogen Storage Disease Type I/complications , Liver/pathology , Non-alcoholic Fatty Liver Disease/metabolism , Adipose Tissue, White/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Dependovirus/genetics , Disease Models, Animal , Gene Knockdown Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Glucose-6-Phosphatase/genetics , Glycogen/metabolism , Glycogen Storage Disease Type I/genetics , Glycogen Storage Disease Type I/metabolism , Glycolysis , Hepatocytes , Humans , Lipogenesis , Lipoproteins, VLDL/metabolism , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , RNA, Small Interfering/genetics , Triglycerides/metabolism
4.
Hepatology ; 66(6): 2042-2054, 2017 12.
Article in English | MEDLINE | ID: mdl-28727166

ABSTRACT

It is a long-standing enigma how glycogen storage disease (GSD) type I patients retain a limited capacity for endogenous glucose production despite the loss of glucose-6-phosphatase activity. Insight into the source of residual endogenous glucose production is of clinical importance given the risk of sudden death in these patients, but so far contradictory mechanisms have been proposed. We investigated glucose-6-phosphatase-independent endogenous glucose production in hepatocytes isolated from a liver-specific GSD Ia mouse model (L-G6pc-/- mice) and performed real-time analysis of hepatic glucose fluxes and glycogen metabolism in L-G6pc-/- mice using state-of-the-art stable isotope methodologies. Here we show that G6pc-deficient hepatocytes are capable of producing glucose. In vivo analysis of hepatic glucose metabolism revealed that the hepatic glucokinase flux was decreased by 95% in L-G6pc-/- mice. It also showed increased glycogen phosphorylase flux in L-G6pc-/- mice, which is coupled to the release of free glucose through glycogen debranching. Although the ex vivo activities of debranching enzyme and lysosomal acid maltase, two major hepatic α-glucosidases, were unaltered in L-G6pc-/- mice, pharmacological inhibition of α-glucosidase activity almost completely abolished residual glucose production by G6pc-deficient hepatocytes. CONCLUSION: Our data indicate that hepatocytes contribute to residual glucose production in GSD Ia. We show that α-glucosidase activity, i.e. glycogen debranching and/or lysosomal glycogen breakdown, contributes to residual glucose production by GSD Ia hepatocytes. A strong reduction in hepatic GCK flux in L-G6pc-/- mice furthermore limits the phosphorylation of free glucose synthesized by G6pc-deficient hepatocytes, allowing the release of glucose into the circulation. The almost complete abrogation of GCK flux in G6pc-deficient liver also explains the contradictory reports on residual glucose production in GSD Ia patients. (Hepatology 2017;66:2042-2054).


Subject(s)
Glucose/metabolism , Glycogen Storage Disease Type I/metabolism , Hepatocytes/metabolism , Animals , Disease Models, Animal , Galactose/metabolism , Glucose-6-Phosphatase/genetics , Glycerol/metabolism , Male , Mice , alpha-Glucosidases/metabolism
5.
Gastroenterology ; 152(5): 1126-1138.e6, 2017 04.
Article in English | MEDLINE | ID: mdl-28065787

ABSTRACT

BACKGROUND & AIMS: The role of the intestine in the maintenance of cholesterol homeostasis increasingly is recognized. Fecal excretion of cholesterol is the last step in the atheroprotective reverse cholesterol transport pathway, to which biliary and transintestinal cholesterol excretion (TICE) contribute. The mechanisms controlling the flux of cholesterol through the TICE pathway, however, are poorly understood. We aimed to identify mechanisms that regulate and stimulate TICE. METHODS: We performed studies with C57Bl/6J mice, as well as with mice with intestine-specific knockout of the farnesoid X receptor (FXR), mice that express an FXR transgene specifically in the intestine, and ABCG8-knockout mice. Mice were fed a control diet or a diet supplemented with the FXR agonist PX20606, with or without the cholesterol absorption inhibitor ezetimibe. Some mice with intestine-specific knockout of FXR were given daily injections of fibroblast growth factor (FGF)19. To determine fractional cholesterol absorption, mice were given intravenous injections of cholesterol D5 and oral cholesterol D7. Mice were given 13C-acetate in drinking water for measurement of cholesterol synthesis. Bile cannulations were performed and biliary cholesterol secretion rates were assessed. In a separate set of experiments, bile ducts of male Wistar rats were exteriorized, allowing replacement of endogenous bile by a model bile. RESULTS: In mice, we found TICE to be regulated by intestinal FXR via induction of its target gene Fgf15 (FGF19 in rats and human beings). Stimulation of this pathway caused mice to excrete up to 60% of their total cholesterol content each day. PX20606 and FGF19 each increased the ratio of muricholate:cholate in bile, inducing a more hydrophilic bile salt pool. The altered bile salt pool stimulated robust secretion of cholesterol into the intestinal lumen via the sterol-exporting heterodimer adenosine triphosphate binding cassette subfamily G member 5/8 (ABCG5/G8). Of note, the increase in TICE induced by PX20606 was independent of changes in cholesterol absorption. CONCLUSIONS: Hydrophilicity of the bile salt pool, controlled by FXR and FGF15/19, is an important determinant of cholesterol removal via TICE. Strategies that alter bile salt pool composition might be developed for the prevention of cardiovascular disease. Transcript profiling: http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?token=irsrayeohfcntqx&acc=GSE74101.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Bile Acids and Salts/metabolism , Cholesterol/metabolism , Fibroblast Growth Factors/metabolism , Intestinal Elimination/genetics , Intestinal Mucosa/metabolism , Lipoproteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Animals , Anticholesteremic Agents/pharmacology , Benzoates/pharmacology , Bile Ducts , Ezetimibe/pharmacology , Intestinal Elimination/drug effects , Intestinal Mucosa/drug effects , Intestines/drug effects , Isoxazoles/pharmacology , Lipoproteins/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Rats , Rats, Wistar , Receptors, Cytoplasmic and Nuclear/agonists
6.
J Lipid Res ; 57(8): 1455-64, 2016 08.
Article in English | MEDLINE | ID: mdl-27313057

ABSTRACT

Statins are competitive inhibitors of HMG-CoA reductase, the rate-limiting enzyme of cholesterol synthesis. Statins reduce plasma cholesterol levels, but whether this is actually caused by inhibition of de novo cholesterol synthesis has not been clearly established. Using three different statins, we investigated the effects on cholesterol metabolism in mice in detail. Surprisingly, direct measurement of whole body cholesterol synthesis revealed that cholesterol synthesis was robustly increased in statin-treated mice. Measurement of organ-specific cholesterol synthesis demonstrated that the liver is predominantly responsible for the increase in cholesterol synthesis. Excess synthesized cholesterol did not accumulate in the plasma, as plasma cholesterol decreased. However, statin treatment led to an increase in cholesterol removal via the feces. Interestingly, enhanced cholesterol excretion in response to rosuvastatin and lovastatin treatment was mainly mediated via biliary cholesterol secretion, whereas atorvastatin mainly stimulated cholesterol removal via the transintestinal cholesterol excretion pathway. Moreover, we show that plasma cholesterol precursor levels do not reflect cholesterol synthesis rates during statin treatment in mice. In conclusion, cholesterol synthesis is paradoxically increased upon statin treatment in mice. However, statins potently stimulate the excretion of cholesterol from the body, which sheds new light on possible mechanisms underlying the cholesterol-lowering effects of statins.


Subject(s)
Cholesterol/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver/metabolism , Lovastatin/pharmacology , Animals , Cholesterol/blood , Drug Evaluation, Preclinical , Gene Expression/drug effects , Glutarates/metabolism , Hypercholesterolemia/blood , Hypercholesterolemia/drug therapy , Intestinal Elimination/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Liver/drug effects , Male , Mice, Inbred C57BL
7.
J Hepatol ; 63(3): 697-704, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26022694

ABSTRACT

BACKGROUND & AIMS: Regulation of bile acid homeostasis in mammals is a complex process regulated via extensive cross-talk between liver, intestine and intestinal microbiota. Here we studied the effects of gut microbiota on bile acid homeostasis in mice. METHODS: Bile acid homeostasis was assessed in four mouse models. Germfree mice, conventionally-raised mice, Asbt-KO mice and intestinal-specific Gata4-iKO mice were treated with antibiotics (bacitracin, neomycin and vancomycin; 100 mg/kg) for five days and subsequently compared with untreated mice. RESULTS: Attenuation of the bacterial flora by antibiotics strongly reduced fecal excretion and synthesis of bile acids, but increased the expression of the bile acid synthesis enzyme CYP7A1. Similar effects were seen in germfree mice. Intestinal bile acid absorption was increased and accompanied by increases in plasma bile acid levels, biliary bile acid secretion and enterohepatic cycling of bile acids. In the absence of microbiota, the expression of the intestinal bile salt transporter Asbt was strongly increased in the ileum and was also expressed in more proximal parts of the small intestine. Most of the effects of antibiotic treatment on bile acid homeostasis could be prevented by genetic inactivation of either Asbt or the transcription factor Gata4. CONCLUSIONS: Attenuation of gut microbiota alters Gata4-controlled expression of Asbt, increasing absorption and decreasing synthesis of bile acids. Our data support the concept that under physiological conditions microbiota stimulate Gata4, which suppresses Asbt expression, limiting the expression of this transporter to the terminal ileum. Our studies expand current knowledge on the bacterial control of bile acid homeostasis.


Subject(s)
Bile Acids and Salts/metabolism , GATA4 Transcription Factor/physiology , Gastrointestinal Microbiome/physiology , Intestinal Absorption , Organic Anion Transporters, Sodium-Dependent/physiology , Symporters/physiology , Animals , Anti-Bacterial Agents/pharmacology , Cholesterol 7-alpha-Hydroxylase/genetics , Homeostasis , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis
8.
PLoS One ; 9(12): e115028, 2014.
Article in English | MEDLINE | ID: mdl-25506828

ABSTRACT

The nuclear receptor FXR acts as an intracellular bile salt sensor that regulates synthesis and transport of bile salts within their enterohepatic circulation. In addition, FXR is involved in control of a variety of crucial metabolic pathways. Four FXR splice variants are known, i.e. FXRα1-4. Although these isoforms show differences in spatial and temporal expression patterns as well as in transcriptional activity, the physiological relevance hereof has remained elusive. We have evaluated specific roles of hepatic FXRα2 and FXRα4 by stably expressing these isoforms using liver-specific self-complementary adeno-associated viral vectors in total body FXR knock-out mice. The hepatic gene expression profile of the FXR knock-out mice was largely normalized by both isoforms. Yet, differential effects were also apparent; FXRα2 was more effective in reducing elevated HDL levels and transrepressed hepatic expression of Cyp8b1, the regulator of cholate synthesis. The latter coincided with a switch in hydrophobicity of the bile salt pool. Furthermore, FXRα2-transduction caused an increased neutral sterol excretion compared to FXRα4 without affecting intestinal cholesterol absorption. Our data show, for the first time, that hepatic FXRα2 and FXRα4 differentially modulate bile salt and lipoprotein metabolism in mice.


Subject(s)
Bile Acids and Salts/metabolism , Lipoproteins/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Lipid Metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
9.
J Hepatol ; 61(2): 351-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24681341

ABSTRACT

BACKGROUND & AIMS: Glucocorticoids, produced by the adrenal gland under control of the hypothalamic-pituitary-adrenal axis, exert their metabolic actions largely via activation of the glucocorticoid receptor (GR). Synthetic glucocorticoids are widely used as anti-inflammatory and immunosuppressive drugs but their application is hampered by adverse metabolic effects. Recently, it has been shown that GR may regulate several genes involved in murine bile acid (BA) and cholesterol metabolism, yet the physiological relevance hereof is controversial. The aim of this study is to provide a mechanistic basis for effects of prednisolone on BA and cholesterol homeostasis in mice. METHODS: Male BALB/c mice were treated with prednisolone (12.5mg/kg/day) for 7days by subcutaneous implantation of slow-release pellets, followed by extensive metabolic profiling. RESULTS: Sustained prednisolone treatment induced the expression of the apical sodium-dependent bile acid transporter (Asbt) in the ileum, which stimulated BA absorption. This resulted in elevated plasma BA levels and enhanced biliary BA secretion. Concomitantly, both biliary cholesterol and phospholipid secretion rates were increased. Enhanced BA reabsorption suppressed hepatic BA synthesis, as evident from hepatic gene expression, reduced plasma C4 levels and reduced fecal BA loss. Plasma HDL cholesterol levels were elevated in prednisolone-treated mice, which likely contributed to the stimulated flux of cholesterol from intraperitoneally injected macrophage foam cells into feces. CONCLUSIONS: Sustained prednisolone treatment increases enterohepatic recycling of BA, leading to elevated plasma levels and reduced synthesis in the absence of cholestasis. Under these conditions, prednisolone promotes macrophage-derived reverse cholesterol transport.


Subject(s)
Bile Acids and Salts/metabolism , Cholesterol/metabolism , Enterohepatic Circulation , Organic Anion Transporters, Sodium-Dependent/biosynthesis , Prednisolone/pharmacology , Symporters/biosynthesis , Animals , Biological Transport , Homeostasis , Male , Mice , Mice, Inbred BALB C
10.
Am J Physiol Gastrointest Liver Physiol ; 305(7): G474-82, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23868407

ABSTRACT

Polyethylene glycol (PEG) is a frequently used osmotic laxative that accelerates gastrointestinal transit. It has remained unclear, however, whether PEG affects intestinal functions. We aimed to determine the effect of PEG treatment on intestinal sterol metabolism. Rats were treated with PEG in drinking water (7%) for 2 wk or left untreated (controls). We studied the enterohepatic circulation of the major bile salt (BS) cholate with a plasma stable isotope dilution technique and determined BS profiles and concentrations in bile, intestinal lumen contents, and feces. We determined the fecal excretion of cholesterol plus its intestinally formed metabolites. Finally, we determined the cytolytic activity of fecal water (a surrogate marker of colorectal cancer risk) and the amount and composition of fecal microbiota. Compared with control rats, PEG treatment increased the pool size (+51%; P < 0.01) and decreased the fractional turnover of cholate (-32%; P < 0.01). PEG did not affect the cholate synthesis rate, corresponding with an unaffected fecal primary BS excretion. PEG reduced fecal excretion of secondary BS and of cholesterol metabolites (each P < 0.01). PEG decreased the cytolytic activity of fecal water [54 (46-62) vs. 87 (85-92)% erythrocyte potassium release in PEG-treated and control rats, respectively; P < 0.01]. PEG treatment increased the contribution of Verrucomicrobia (P < 0.01) and decreased that of Firmicutes (P < 0.01) in fecal flora. We concluded that PEG treatment changes the intestinal bacterial composition, decreases the bacterial dehydroxylation of primary BS and the metabolism of cholesterol, and increases the pool size of the primary BS cholate in rats.


Subject(s)
Bile Acids and Salts/metabolism , Intestinal Mucosa/metabolism , Intestines/drug effects , Lipid Metabolism/drug effects , Polyethylene Glycols/pharmacology , Animals , Feces , Intestines/microbiology , Laxatives/pharmacology , Polyethylene Glycols/administration & dosage , Random Allocation , Rats , Rats, Wistar
11.
Atherosclerosis ; 228(1): 117-23, 2013 May.
Article in English | MEDLINE | ID: mdl-23497783

ABSTRACT

AIMS: Bile acid sequestrants (BAS) and physical activity (RUN) decrease incidence of cardiovascular events. Both treatments are often prescribed, yet it is not known whether their beneficial effects are additive. We assessed the effects of BAS treatment alone and in combination with RUN on cholesterol metabolism, heart function and atherosclerotic lesion size in hypercholesterolemic mice. METHODS: Male Ldlr-deficient mice remained either sedentary (CONTROL), were treated with Colesevelam HCl (BAS), had access to a running wheel (RUN), or were exposed to BAS and RUN (BAS RUN). All groups were fed a high cholesterol diet for 12 weeks. Then, feces, bile and plasma were collected. Atherosclerotic lesion size was determined in the aortic arch and heart function by echocardiography. RESULTS: BAS RUN ran more than RUN (6.4 ± 1.4 vs. 3.5 ± 1.0 km/day, p < 0.05). BAS and BAS RUN displayed ~3-fold reductions in plasma cholesterol levels (p < 0.001), ~2.5-fold increases in fecal neutral sterol (p < 0.001) and bile acid (p = 0.01) outputs, decreases in biliary secretions of cholesterol (~6-fold, p < 0.0001) and bile acids (~2-fold, p < 0.001) vs. CONTROL while no significant effects were observed in RUN. Compared to CONTROL, lesion size decreased by 78% in both BAS and BAS RUN, (p < 0.0001). CONCLUSION: BAS reduce atherosclerosis in Ldlr-deficient mice, coinciding with a switch from body cholesterol accumulation to cholesterol loss. RUN slightly modulated atherosclerotic lesion formation but the combination of BAS and RUN had no clear additive effects in this respect.


Subject(s)
Allylamine/analogs & derivatives , Atherosclerosis/metabolism , Atherosclerosis/physiopathology , Bile Acids and Salts/metabolism , Hypercholesterolemia , Physical Conditioning, Animal/physiology , Allylamine/pharmacology , Animal Feed , Animals , Anticholesteremic Agents/pharmacology , Aorta/diagnostic imaging , Aorta/metabolism , Atherosclerosis/diagnostic imaging , Cardiac Output/physiology , Cholesterol/biosynthesis , Cholesterol/blood , Colesevelam Hydrochloride , Echocardiography , Feces , Hypercholesterolemia/drug therapy , Hypercholesterolemia/metabolism , Hypercholesterolemia/physiopathology , Liver/metabolism , Male , Mice , Mice, Knockout , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sterols/metabolism
12.
Gastroenterology ; 135(2): 689-98, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18565334

ABSTRACT

BACKGROUND & AIMS: Conversion into bile acids represents an important route to remove excess cholesterol from the body. Rev-erbalpha is a nuclear receptor that participates as one of the clock genes in the control of circadian rhythmicity and plays a regulatory role in lipid metabolism and adipogenesis. Here, we investigate a potential role for Rev-erbalpha in the control of bile acid metabolism via the regulation of the neutral bile acid synthesis pathway. METHODS: Bile acid synthesis and CYP7A1 gene expression were studied in vitro and in vivo in mice deficient for or over expressing Rev-erbalpha. RESULTS: Rev-erbalpha-deficient mice display a lower synthesis rate and an impaired excretion of bile acids into the bile and feces. Expression of CYP7A1, the rate-limiting enzyme of the neutral pathway, is decreased in livers of Rev-erbalpha-deficient mice, whereas adenovirus-mediated hepatic Rev-erbalpha overexpression induces its expression. Moreover, bile acid feeding resulted in a more pronounced suppression of hepatic CYP7A1 expression in Rev-erbalpha-deficient mice. Hepatic expression of E4BP4 and the orphan nuclear receptor small heterodimer partner (SHP), both negative regulators of CYP7A1 expression, is increased in Rev-erbalpha-deficient mice. Promoter analysis and chromatin immunoprecipitation experiments demonstrated that SHP and E4BP4 are direct Rev-erbalpha target genes. Finally, the circadian rhythms of liver CYP7A1, SHP, and E4BP4 messenger RNA levels were perturbed in Rev-erbalpha-deficient mice. CONCLUSIONS: These data identify a role for Rev-erbalpha in the regulatory loop of bile acid synthesis, likely acting by regulating both hepatic SHP and E4BP4 expression.


Subject(s)
Bile Acids and Salts/metabolism , DNA-Binding Proteins/metabolism , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Bile/metabolism , Cell Line, Tumor , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Circadian Rhythm , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Feces/chemistry , Gene Expression Regulation, Enzymologic , Humans , Liver/enzymology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1 , Promoter Regions, Genetic , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/genetics , Time Factors , Transfection
13.
Am J Physiol Gastrointest Liver Physiol ; 293(3): G615-22, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17627970

ABSTRACT

Cholestasis is associated with systemic accumulation of bile salts and with deficiency of bile in the intestinal lumen. During the past years bile salts have been identified as signaling molecules that regulate lipid, glucose, and energy metabolism. Bile salts have also been shown to activate signaling routes leading to proliferation, apoptosis, or differentiation. It is unclear, however, whether cholestasis affects the constitution and absorptive capacity of the intestinal epithelium in vivo. We studied small intestinal morphology, proliferation, apoptosis, expression of intestine-specific genes, and carbohydrate absorption in cholestatic (1 wk bile duct ligation), bile-deficient (1 wk bile diversion), and control (sham) rats. Absorptive capacity was assessed by determination of plasma [(2)H]- and [(13)C]glucose concentrations after intraduodenal administration of [(2)H]glucose and naturally enriched [(13)C]sucrose, respectively. Small intestinal morphology, proliferation, apoptosis, and gene expression of intestinal transcription factors (mRNA levels of Cdx-2, Gata-4, and Hnf-1alpha, and Cdx-2 protein levels) were similar in cholestatic, bile-deficient, and control rats. The (unlabeled) blood glucose response after intraduodenal administration was delayed in cholestatic animals, but the absorption over 180 min was quantitatively similar between the groups. Plasma concentrations of [(2)H]glucose and [(13)C]glucose peaked to similar extents in all groups within 7.5 and 30 min, respectively. Absorption of [(2)H]glucose and [(13)C]glucose in plasma was similar in all groups. The present data indicate that neither accumulation of bile salts in the body, nor their intestinal deficiency, two characteristic features of cholestasis, affect rat small intestinal proliferation, differentiation, apoptosis, or its capacity to digest and absorb carbohydrates.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis, Extrahepatic/metabolism , Dietary Carbohydrates/metabolism , Digestion , Intestinal Absorption , Jejunum/metabolism , Animals , Apoptosis , Bile Acids and Salts/blood , Bile Acids and Salts/deficiency , Bile Ducts/surgery , Blood Glucose/metabolism , CDX2 Transcription Factor , Carbon Isotopes , Cell Differentiation , Cell Proliferation , Cholestasis, Extrahepatic/pathology , Cholestasis, Extrahepatic/physiopathology , Dietary Carbohydrates/administration & dosage , Dietary Sucrose/metabolism , Disease Models, Animal , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Glucose/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intubation, Gastrointestinal , Jejunum/pathology , Jejunum/physiopathology , Ligation , Lipid Metabolism , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism
14.
J Lipid Res ; 48(9): 2058-64, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17595447

ABSTRACT

The phosphatidylethanolamine N-methyltransferase (PEMT) pathway of phosphatidylcholine (PC) biosynthesis is not essential for the highly specific acyl chain composition of biliary PC. We evaluated whether the PEMT pathway is quantitatively important for biliary PC secretion in mice under various experimental conditions. Biliary bile salt and PC secretion were determined in mice in which the gene encoding PEMT was inactivated (Pemt(-/-)) and in wild-type mice under basal conditions, during acute metabolic stress (intravenous infusion of the bile salt tauroursodeoxycholate), and during chronic metabolic stress (feeding a taurocholate-containing diet for 1 week). The activity of CTP:phosphocholine cytidylyltransferase, the rate-limiting enzyme of PC biosynthesis via the CDP-choline pathway, and the abundance of multi-drug-resistant protein 2 (Mdr2; encoded by the Abcb4 gene), the canalicular membrane flippase essential for biliary PC secretion, were determined. Under basal conditions, Pemt(-/-) and wild-type mice exhibited similar biliary secretion rates of bile salt and PC ( approximately 145 and approximately 28 nmol/min/100 g body weight, respectively). During acute or chronic bile salt administration, the biliary PC secretion rates increased similarly in Pemt(-/-) and control mice. Mdr2 mRNA and protein abundance did not differ between Pemt(-/-) and wild-type mice. The cytidylyltransferase activity in hepatic lysates was increased by 20% in Pemt(-/-) mice fed the basal (bile salt-free) diet (P < 0.05). We conclude that the biosynthesis of PC via the PEMT pathway is not quantitatively essential for biliary PC secretion under acute or chronic bile salt administration.


Subject(s)
Bile/metabolism , Phosphatidylcholines/metabolism , Phosphatidylethanolamine N-Methyltransferase/metabolism , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Choline Deficiency/physiopathology , Choline-Phosphate Cytidylyltransferase/metabolism , Liver/metabolism , Metabolic Networks and Pathways/physiology , Mice , Phosphatidylethanolamine N-Methyltransferase/deficiency , ATP-Binding Cassette Sub-Family B Member 4
15.
Liver Int ; 25(3): 604-12, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15910498

ABSTRACT

BACKGROUND/AIMS: Bile flow consists of bile salt-dependent bile flow (BSDF), generated by canalicular secretion of bile salts, and bile salt-independent flow (BSIF), probably of combined canalicular and ductular origin. Bile salt transport proteins have been identified in cholangiocytes, suggesting a role in control of BSDF and/or in control of bile salt synthesis through cholehepatic shunting. METHODS: We studied effects of bile duct proliferation under non-cholestatic conditions in multidrug resistance-2 P-glycoprotein (Abcb4)-deficient multidrug resistance gene-2 (Mdr2(-/-)) mice. BSDF and BSIF were determined in wild-type and Mdr2(-/-) mice during infusion of step-wise increasing dosages of tauroursodeoxycholate (TUDC). Cholate synthesis rate was determined by 2H4-cholate dilution. Results were related to expression of transport proteins in liver and intestine. RESULTS: During TUDC infusion, BSDF was increased by approximately 50% and BSIF by approximately 100% in Mdr2(-/-) mice compared with controls. Cholate synthesis rate was unaffected in Mdr2(-/-) mice. Hepatic expression of the apical sodium-dependent bile salt transporter (Asbt), its truncated form (tAsbt) and the multidrug resistance-related protein 3 were upregulated in Mdr2(-/-) mice. CONCLUSIONS: Bile duct proliferation in Mdr2(-/-) mice enhances cholehepatic shunting of bile salts, which is associated with a disproportionally high bile flow but does not affect bile salt synthesis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , Bile Acids and Salts/metabolism , Bile Duct Diseases/metabolism , Bile Duct Diseases/pathology , Bile Ducts/pathology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Division , Cholates/pharmacokinetics , Deuterium , Gallbladder/metabolism , Intestines/physiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Mutant Strains , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Phospholipids/metabolism , RNA, Messenger/analysis , Symporters/genetics , Symporters/metabolism , ATP-Binding Cassette Sub-Family B Member 4
16.
J Hepatol ; 42(1): 102-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15629514

ABSTRACT

BACKGROUND/AIMS: In sepsis-associated cholestasis, expression of many genes involved in bile acid transport, including Ntcp, is suppressed by cytokines. Kupffer cells (KC) are an important source of cytokines in sepsis. To assess the consequences of KC depletion on hepatic Ntcp expression in endotoxemic rats. METHODS: Sprague-Dawley rats received liposomal clodronate (CLO) or vehicle (PBS) to deplete KC prior to lipopolysaccharide (LPS) exposure. Plasma and liver samples were taken 1 and 16 h after LPS exposure. RESULTS: Complete CLO-depletion of KC by was demonstrated by immunohistochemistry. Hepatic gene expression of IL-1beta and TNFalpha as well as TNFalpha plasma levels in CLO/LPS-injected animals were significantly reduced to a mean of 41, 36 and 23% of controls injected with LPS only. Ntcp RNA- and protein expression was significantly higher whereas plasma bile salt concentration was lower in CLO/LPS animals vs. animals injected with LPS only. Binding activity of transcription factors RXR:RAR and HNF1alpha was decreased in LPS only controls but preserved in CLO/LPS treated animals. CONCLUSIONS: Clodronate-depletion of KC blocks cytokine-mediated Ntcp suppression upon endotoxin exposure. KC may represent pharmacological targets for treatment of sepsis-associated cholestasis.


Subject(s)
Clodronic Acid/administration & dosage , Kupffer Cells/drug effects , Lipopolysaccharides/toxicity , Membrane Transport Proteins/genetics , Animals , Cholestasis/drug therapy , DNA-Binding Proteins/metabolism , Hepatocyte Nuclear Factor 1 , Hepatocyte Nuclear Factor 1-alpha , Interleukin-1/biosynthesis , Kupffer Cells/physiology , Liposomes , Male , Nuclear Proteins/metabolism , Organic Anion Transporters, Sodium-Dependent , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/genetics , Symporters , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
17.
J Biol Chem ; 278(43): 41930-7, 2003 Oct 24.
Article in English | MEDLINE | ID: mdl-12917447

ABSTRACT

The bile salt-activated farnesoid X receptor (FXR; NR1H4) controls expression of several genes considered crucial in maintenance of bile salt homeostasis. We evaluated the physiological consequences of FXR deficiency on bile formation and on the kinetics of the enterohepatic circulation of cholate, the major bile salt species in mice. The pool size, fractional turnover rate, synthesis rate, and intestinal absorption of cholate were determined by stable isotope dilution and were related to expression of relevant transporters in the livers and intestines of FXR-deficient (Fxr-/-) mice. Fxr-/- mice showed only mildly elevated plasma bile salt concentrations associated with a 2.4-fold higher biliary bile salt output, whereas hepatic mRNA levels of the bile salt export pump were decreased. Cholate pool size and total bile salt pool size were increased by 67 and 39%, respectively, in Fxr-/- mice compared with wild-type mice. The cholate synthesis rate was increased by 85% in Fxr-/- mice, coinciding with a 2.5-fold increase in cholesterol 7alpha-hydroxylase (Cyp7a1) and unchanged sterol 12alpha-hydroxylase (Cyp8b1) expression in the liver. Despite a complete absence of ileal bile acid-binding protein mRNA and protein, the fractional turnover rate and cycling time of the cholate pool were not affected. The calculated amount of cholate reabsorbed from the intestine per day was approximately 2-fold higher in Fxr-/- mice than in wild-type mice. Thus, the absence of FXR in mice is associated with defective feedback inhibition of hepatic cholate synthesis, which leads to enlargement of the circulating cholate pool with an unaltered fractional turnover rate. The absence of ileal bile acid-binding protein does not negatively interfere with the enterohepatic circulation of cholate in mice.


Subject(s)
Bile Acids and Salts/metabolism , DNA-Binding Proteins/physiology , Enterohepatic Circulation/physiology , Hydroxysteroid Dehydrogenases , Membrane Glycoproteins , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Animals , Carrier Proteins/analysis , Cholesterol 7-alpha-Hydroxylase/biosynthesis , Cholesterol 7-alpha-Hydroxylase/genetics , Cholic Acid/biosynthesis , Cholic Acid/metabolism , DNA-Binding Proteins/genetics , Feedback, Physiological , Homeostasis , Intestinal Absorption , Mice , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics
18.
Gastroenterology ; 124(1): 160-71, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12512040

ABSTRACT

BACKGROUND & AIMS: Fatty acids are natural ligands of the peroxisome proliferator-activated receptor alpha (PPARalpha). Synthetic ligands of this nuclear receptor, i.e., fibrates, induce the hepatic expression of the multidrug resistance 2 gene (Mdr2), encoding the canalicular phospholipid translocator, and affect hepatobiliary lipid transport. We tested whether fasting-associated fatty acid release from adipose tissues alters hepatic transporter expression and bile formation in a PPARalpha-dependent manner. METHODS: A 24-hour fasting/48-hour refeeding schedule was used in wild-type and Pparalpha((-/-)) mice. Expression of genes involved in the control of bile formation was determined and related to secretion rates of biliary components. RESULTS: Expression of Pparalpha, farnesoid X receptor, and liver X receptor alpha genes encoding nuclear receptors that control hepatic bile salt and sterol metabolism was induced on fasting in wild-type mice only. The expression of Mdr2 was 5-fold increased in fasted wild-type mice and increased only marginally in Pparalpha((-/-)) mice, and it normalized on refeeding. Mdr2 protein levels and maximal biliary phospholipid secretion rates were clearly increased in fasted wild-type mice. Hepatic expression of the liver X receptor target genes ATP binding cassette transporter a1 (Abca1), Abcg5, and Abcg8, implicated in hepatobiliary cholesterol transport, was induced in fasted wild-type mice only. However, the maximal biliary cholesterol secretion rate was reduced by approximately 50%. CONCLUSIONS: Induction of Mdr2 expression and function is part of the PPARalpha-mediated fasting response in mice. Fasting also induces expression of the putative hepatobiliary cholesterol transport genes Abca1, Abcg5, and Abcg8, but, nonetheless, maximal biliary cholesterol excretion is decreased after fasting.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Fasting/physiology , Liver/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/physiology , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/physiology , Animal Feed , Animals , Bile/physiology , Lipid Metabolism , Male , Mice , Mice, Knockout/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Reference Values
19.
Biochem J ; 369(Pt 3): 539-47, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12381268

ABSTRACT

Peroxisome proliferator-activated receptor alpha (PPARalpha) is a nuclear receptor that controls expression of genes involved in lipid metabolism and is activated by fatty acids and hypolipidaemic fibrates. Fibrates induce the hepatic expression of murine multidrug resistance 2 ( Mdr2 ), encoding the canalicular phospholipid translocator. The physiological role of PPARalpha in regulation of Mdr2 and other genes involved in bile formation is unknown. We found no differences in hepatic expression of the ATP binding cassette transporter genes Mdr2, Bsep (bile salt export pump), Mdr1a / 1b, Abca1 and Abcg5 / Abcg8 (implicated in cholesterol transport), the bile salt-uptake systems Ntcp (Na(+)-taurocholate co-transporting polypeptide gene) and Oatp1 (organic anion-transporting polypeptide 1 gene) or in bile formation between wild-type and Ppar alpha((-/-)) mice. Upon treatment of wild-type mice with ciprofibrate (0.05%, w/w, in diet for 2 weeks), the expression of Mdr2 (+3-fold), Mdr1a (+6-fold) and Mdr1b (+11-fold) mRNAs was clearly induced, while that of Oatp1 (-5-fold) was reduced. Mdr2 protein levels were increased, whereas Bsep, Ntcp and Oatp1 were drastically decreased. Exposure of cultured wild-type mouse hepatocytes to PPARalpha agonists specifically induced Mdr2 mRNA levels and did not affect expression of Mdr1a / 1b. Altered transporter expression in fibrate-treated wild-type mice was associated with a approximately 400% increase in bile flow: secretion of phospholipids and cholesterol was increased only during high-bile-salt infusions. No fibrate effects were observed in Ppar alpha((-/-)) mice. In conclusion, our results show that basal bile formation is not affected by PPARalpha deficiency in mice. The induction of Mdr2 mRNA and Mdr2 protein levels by fibrates is mediated by PPARalpha, while the induction of Mdr1a / 1b in vivo probably reflects a secondary phenomenon related to chronic PPARalpha activation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Clofibric Acid/analogs & derivatives , Hepatocytes/physiology , Membrane Transport Proteins , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily B/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/drug effects , Animals , Bile/chemistry , Bile/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Clofibric Acid/pharmacology , Fibric Acids , Lipid Metabolism , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Mutant Strains , Organ Size/drug effects , Organic Anion Transporters, Sodium-Dependent , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Peroxisome Proliferators/pharmacology , Pyrimidines/pharmacology , Receptors, Cytoplasmic and Nuclear/drug effects , Receptors, Cytoplasmic and Nuclear/genetics , Symporters , Taurochenodeoxycholic Acid/administration & dosage , Taurochenodeoxycholic Acid/pharmacology , Transcription Factors/drug effects , Transcription Factors/genetics , ATP-Binding Cassette Sub-Family B Member 4
20.
J Pharmacol Exp Ther ; 304(1): 356-63, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12490612

ABSTRACT

Cyclosporin A (CsA) has been shown to inhibit synthesis and hepatobiliary transport of bile salts. However, effects of CsA on the enterohepatic circulation of bile salts in vivo are largely unknown. We characterized the effects of CsA on the enterohepatic circulation of cholate, with respect to synthesis rate, pool size, cycling time, intestinal absorption, and the expression of relevant transporters in liver and intestine in rats. CsA (1 mg. 100 g(-1). day(-1) s.c.) or its solvent was administered daily to male rats for 10 days. Cholate synthesis rate and pool size were determined by a 2H4-cholate dilution technique. Bile and feces were collected for determination of cholate and total bile salts, respectively. Cycling time and intestinal absorption of cholate were calculated. The mRNA levels and corresponding transporter protein levels in liver and intestine were assessed by real-time polymerase chain reaction and Western analysis, respectively. CsA treatment decreased cholate synthesis rate by 71%, but did not affect pool size or cycling time. CsA reduced the amount of cholate lost per enterohepatic cycle by approximately 70%. Protein levels of the apical sodium-dependent bile salt transporter (Asbt) were 2-fold increased in distal ileum of CsA-treated rats, due to post-transcriptional events. In conclusion, chronic CsA treatment markedly reduces cholate synthesis rate in rats, but does not affect cholate pool size or cycling time. Our results strongly suggest that CsA enhances efficacy of intestinal cholate reabsorption through increased Asbt protein expression in the distal ileum, which contributes to maintenance of cholate pool size in CsA-treated rats.


Subject(s)
Bile Acids and Salts/metabolism , Cholic Acid/biosynthesis , Cyclosporine/pharmacology , Enterohepatic Circulation/drug effects , Immunosuppressive Agents/pharmacology , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Organic Anion Transporters, Sodium-Dependent , Symporters , Animals , Carrier Proteins/biosynthesis , Carrier Proteins/metabolism , Chromatography, Gas , Feces/chemistry , Ileum/drug effects , Ileum/metabolism , Kinetics , Liver Function Tests , Male , Mass Spectrometry , Membranes/chemistry , Membranes/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Radioisotope Dilution Technique , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...