Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hepatology ; 70(1): 276-293, 2019 07.
Article in English | MEDLINE | ID: mdl-30983011

ABSTRACT

Pregnancy is associated with progressive hypercholanemia, hypercholesterolemia, and hypertriglyceridemia, which can result in metabolic disease in susceptible women. Gut signals modify hepatic homeostatic pathways, linking intestinal content to metabolic activity. We sought to identify whether enteric endocrine signals contribute to raised serum bile acids observed in human and murine pregnancies, by measuring fibroblast growth factor (FGF) 19/15 protein and mRNA levels, and 7α-hydroxy-4-cholesten-3-one. Terminal ileal farnesoid X receptor (FXR)-mediated gene expression and apical sodium bile acid transporter (ASBT) protein concentration were measured by qPCR and western blotting. Shotgun whole-genome sequencing and ultra-performance liquid chromatography tandem mass spectrometry were used to determine the cecal microbiome and metabonome. Targeted and untargeted pathway analyses were performed to predict the systemic effects of the altered metagenome and metabolite profiles. Dietary CA supplementation was used to determine whether the observed alterations could be overcome by intestinal bile acids functioning as FXR agonists. Human and murine pregnancy were associated with reduced intestinal FXR signaling, with lower FGF19/15 and resultant increased hepatic bile acid synthesis. Terminal ileal ASBT protein was reduced in murine pregnancy. Cecal bile acid conjugation was reduced in pregnancy because of elevated bile salt hydrolase-producing Bacteroidetes. CA supplementation induced intestinal FXR signaling, which was not abrogated by pregnancy, with strikingly similar changes to the microbiota and metabonome as identified in pregnancy. Conclusion: The altered intestinal microbiota of pregnancy enhance bile acid deconjugation, reducing ileal bile acid uptake and lowering FXR induction in enterocytes. This exacerbates the effects mediated by reduced bile acid uptake transporters in pregnancy. Thus, in pregnant women and mice, there is reduced FGF19/15-mediated hepatic repression of hepatic bile acid synthesis, resulting in hypercholanemia.


Subject(s)
Cholic Acids/blood , Gastrointestinal Microbiome , Intestinal Reabsorption , Pregnancy/blood , Receptors, Cytoplasmic and Nuclear/metabolism , Amidohydrolases/genetics , Animals , Bacteroides/isolation & purification , Cecum/drug effects , Cecum/microbiology , Cholic Acids/pharmacology , Enterocytes/drug effects , Female , Humans , Mice, Inbred C57BL , Receptors, Cytoplasmic and Nuclear/agonists
2.
FASEB J ; 32(2): 601-612, 2018 02.
Article in English | MEDLINE | ID: mdl-29457550

ABSTRACT

Familial hypercholesterolemia (FH) conveys a high risk of premature atherosclerosis as a result of lifelong exposure to high LDL cholesterol levels that are not fully reduced by standard-of-care lipid-lowering treatment. Inflammatory mediators have played a role in the progression of atherosclerotic lesions. Here, we investigated whether innate immunity cells in patients with FH have a specific proinflammatory phenotype that is distinct from that of cells in normal participants. To this end, miR-505-3p-a microRNA related to chronic inflammation-and its target genes were investigated in monocyte-derived macrophages (MACs) of patients with FH (FH-MACs) and non-FH controls (co-MACs). On the basis of the profiler PCR array analysis of agomiR-505-3p-transfected MACs, we identified the chemokine receptors, CCR3, CCR4, and CXCR1, as genes that are regulated by miR-505-3p via the transcription factor, RUNX1. miR-505-3p was significantly down-regulated, whereas CCR3, CCR4, CXCR, and RUNX1 were increased in FH-MAC compared with co-MAC, with the increase being more evident in the proinflammatory M1-like FH-MAC. Chemokine receptor levels were unrelated to LDL plasma levels at entry, but correlated with age in patients with FH, not in controls. In summary, we demonstrate for first time to our knowledge that MACs from FH-MACs have an inflammatory phenotype that is characterized by the up-regulation of CCR3, CCR4, and CXCR1 under the control of miR-505-3p. These results suggest a chronic inflammatory condition in FH innate immunity cells that is not reverted by standard lipid-lowering treatment.-Escate, R., Mata, P., Cepeda, J. M., Padró, T., Badimon, L. miR-505-3p controls chemokine receptor up-regulation in macrophages: role in familial hypercholesterolemia.


Subject(s)
Cholic Acids/blood , Macrophages/metabolism , MicroRNAs/metabolism , Receptors, Chemokine/biosynthesis , Steroid Metabolism, Inborn Errors/metabolism , Up-Regulation , Cholic Acids/immunology , Cholic Acids/metabolism , Female , Humans , Macrophages/immunology , Macrophages/pathology , Male , MicroRNAs/immunology , Receptors, Chemokine/immunology , Steroid Metabolism, Inborn Errors/immunology , Steroid Metabolism, Inborn Errors/pathology , Steroid Metabolism, Inborn Errors/therapy
3.
J Intern Med ; 284(6): 674-684, 2018 12.
Article in English | MEDLINE | ID: mdl-29974534

ABSTRACT

BACKGROUND: Familial hypercholesterolemia could be prevalent among patients with acute coronary syndrome. OBJECTIVE: To investigate both the frequency of causative mutations for familial hypercholesterolemia (FH) and the optimal selection of patients for genetic testing among patients with an acute coronary syndrome (ACS). METHODS: One hundred and sixteen patients with an ACS during 2009-2015 were identified through the SWEDEHEART registry. Patients who had either a high total cholesterol level ≥7 mmol L-1 combined with a triglyceride level ≤2.6 mmol L-1 , or were treated with lipid-lowering medication and had a total cholesterol level >4.9 mmol L-1 and a triglyceride level ≤2.6 mmol L-1 were included. Genetic testing was performed first with a regionally designed FH mutation panel (118 mutations), followed by testing with a commercially available FH genetic analysis (Progenika Biopharma). RESULTS: A total of 6.9% (8/116) patients had a FH-causative mutation, all in the LDL-receptor. Five patients were detected on the panel, and further testing of the remaining 111 patients detected an additional 3 FH-causative mutations. Baseline characteristics were similar in FH-positive and FH-negative patients with respect to age, gender, prior ACS and diabetes. Patients with a FH-causative mutation had higher Dutch Lipid Clinical Network (DLCN) score (5.5 (5.0-6.5) vs 3.0 (2.0-5.0), P < 0.001) and a higher low-density lipoprotein level (5.7 (4.7-6.5) vs 4.9 (3.5-5.4), P = 0.030). The Dutch Lipid Clinical Network (DLCN) score had a good discrimination with an area under the curve of 0.856 (95% CI 0.763-0.949). CONCLUSION: Genetic testing for FH should be considered in patients with ACS and high DLCN score.


Subject(s)
Acute Coronary Syndrome/genetics , Cholic Acids/blood , Genetic Testing , Steroid Metabolism, Inborn Errors/genetics , Acute Coronary Syndrome/etiology , Aged , Cholesterol/blood , Cholic Acids/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Steroid Metabolism, Inborn Errors/complications , Survivors , Triglycerides/blood
4.
Surg Endosc ; 32(2): 805-812, 2018 02.
Article in English | MEDLINE | ID: mdl-28779240

ABSTRACT

BACKGROUND: Bile acids (BAs) are post-prandial hormones that play an important role in glucose and lipid homeostasis as well as energy expenditure. Total and glycine-amidated BAs increase after sleeve gastrectomy (SG) and correlate to improved metabolic disease. No specific bile acid subtype has been shown conclusively to mediate the weight loss effect. Therefore, the objective of this study was to prospectively evaluate the comprehensive changes in meal-stimulated BAs after SG and determine if a specific change in the BA profile correlates to the early weight loss response. METHODS: Patients were prospectively enrolled at the University of Nebraska Medical Center who were undergoing a SG for treatment of morbid obesity. Primary and secondary plasma bile acids and their amidated (glycine, G-, or taurine, T-) subtypes were measured at fasting, 30 and 60 min after a liquid meal performed pre-op, and at 6 and 12 weeks post-op. Area under the curve (AUC) was calculated for the hour meal test for each bile acid subtype. BAs that were significantly increased post-op were correlated to body mass index (BMI) loss. RESULTS: Total BA AUC was significantly increased at 6 (p < 0.01) and 12 weeks post-op (p < 0.01) compared to pre-operative values. The increase in total BA AUC was due to a statistically significant increase in G-BAs. Nine different BA AUC subtypes were significantly increased at both 6 and 12 weeks post-op. Increased total and G-chenodeoxycholic acid AUC was significantly correlated to the 6 week BMI loss (p = 0.03). Increased G-hyocholic acid was significantly correlated to increased weight loss at both 6 (p = 0.05) and 12 weeks (p = 0.006). CONCLUSIONS: SG induced an early and persistent post-prandial surge in multiple bile acid subtypes. Increased G-hyocholic consistently correlated with greater early BMI loss. This study provides evidence for a role of BAs in the surgical weight loss response after SG.


Subject(s)
Cholic Acids/blood , Gastrectomy , Weight Loss , Bile Acids and Salts/blood , Body Mass Index , Fasting , Female , Humans , Male , Middle Aged , Postprandial Period , Prospective Studies
5.
J Hepatol ; 67(2): 321-327, 2017 08.
Article in English | MEDLINE | ID: mdl-28249726

ABSTRACT

BACKGROUND & AIMS: Hepatobiliary secretion of bile acids is an important liver function. Here, we quantified the hepatic transport kinetics of conjugated bile acids using the bile acid tracer [N-methyl-11C]cholylsarcosine (11C-CSar) and positron emission tomography (PET). METHODS: Nine healthy participants and eight patients with varying degrees of cholestasis were examined with 11C-CSar PET and measurement of arterial and hepatic venous blood concentrations of 11C-CSar. RESULTS: Results are presented as median (range). The hepatic intrinsic clearance was 1.50 (1.20-1.76) ml blood/min/ml liver tissue in healthy participants and 0.46 (0.13-0.91) in patients. In healthy participants, the rate constant for secretion of 11C-CSar from hepatocytes to bile was 0.36 (0.30-0.62)min-1, 20 times higher than the rate constant for backflux from hepatocytes to blood (0.02, 0.005-0.07min-1). In the patients, rate constant for transport from hepatocyte to bile was reduced to 0.12 (0.006-0.27)min-1, 2.3times higher than the rate constant for backflux to blood (0.05, 0.04-0.09). The increased backflux did not fully normalize exposure of the hepatocyte to bile acids as mean hepatocyte residence time of 11C-CSar was 2.5 (1.6-3.1)min in healthy participants and 6.4 (3.1-23.7)min in patients. The rate constant for transport of 11C-CSar from intrahepatic to extrahepatic bile was 0.057 (0.023-0.11)min-1 in healthy participants and only slightly reduced in patients 0.039 (0.017-0.066). CONCLUSIONS: This first in vivo quantification of individual steps involved in the hepatobiliary secretion of a conjugated bile acid in humans provided new insight into cholestatic disease. LAY SUMMARY: Positron emission tomography (PET) using the radiolabelled bile acid (11C-CSar) enabled quantification of the individual steps of the hepatic transport of bile acids from blood to bile in man. Cholestasis reduced uptake and secretion and increased backflux to blood. These findings improve our understanding of cholestatic liver diseases and may support therapeutic decisions. CLINICAL TRIAL REGISTRATION NUMBER: The trial is registered at ClinicalTrials.gov (NCT01879735).


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/metabolism , Cholic Acids/pharmacokinetics , Sarcosine/analogs & derivatives , Aged , Bile/metabolism , Biological Transport, Active , Carbon Radioisotopes , Case-Control Studies , Cholestasis/blood , Cholestasis/diagnostic imaging , Cholic Acids/blood , Female , Humans , Kinetics , Liver/diagnostic imaging , Liver/metabolism , Liver Circulation , Male , Middle Aged , Positron-Emission Tomography , Sarcosine/blood , Sarcosine/pharmacokinetics , Young Adult
6.
Hepatology ; 61(1): 260-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24867799

ABSTRACT

UNLABELLED: The enterohepatic circulation of bile salts is an important physiological route to recycle bile salts and ensure intestinal absorption of dietary lipids. The Na(+)-taurocholate cotransporting polypeptide SLC10A1 (NTCP) plays a key role in this process as the major transporter of conjugated bile salts from the plasma compartment into the hepatocyte. Here we present the first patient with NTCP deficiency, who was clinically characterized by mild hypotonia, growth retardation, and delayed motor milestones. Total bile salts in plasma were extremely elevated (up to 1,500 µM, ref. <16.3) but there were no clinical signs of cholestatic jaundice, pruritis, or liver dysfunction. Bile salt synthesis and intestinal bile salt signaling were not affected, as evidenced by normal plasma 7α-hydroxy-4-cholesten-3-one (C4) and FGF19 levels. Importantly, the presence of secondary bile salts in the circulation suggested residual enterohepatic cycling of bile salts. Sequencing of the SLC10A1 gene revealed a single homozygous nonsynonymous point mutation in the coding sequence of the gene, resulting in an arginine to histidine substitution at position 252. Functional studies showed that this mutation resulted in a markedly reduced uptake activity of taurocholic acid. Immunofluorescence studies and surface biotinylation experiments demonstrated that the mutant protein is virtually absent from the plasma membrane. CONCLUSION: We describe the identification of NTCP deficiency as a new inborn error of metabolism with a relatively mild clinical phenotype. The identification of NTCP deficiency confirms that this transporter is the main import system for conjugated bile salts into the liver but also indicates that auxiliary transporters are able to sustain the enterohepatic cycle in its absence.


Subject(s)
Cholic Acids/blood , Organic Anion Transporters, Sodium-Dependent/deficiency , Steroid Metabolism, Inborn Errors/genetics , Symporters/deficiency , Amino Acid Sequence , Cholic Acids/genetics , Female , Humans , Infant , Molecular Sequence Data , Organic Anion Transporters, Sodium-Dependent/genetics , Phenotype , Point Mutation , Protein Transport/genetics , Symporters/genetics
7.
Mol Pharmacol ; 88(6): 1085-92, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26399598

ABSTRACT

Biliary excretion of organic anions, such as bile acids (BAs), is the main osmotic driving force for bile formation, and its impairment induces intrahepatic cholestasis. We investigated the involvement of Atp11c in the hepatic transport of organic anions using Atp11c mutant mice, which exhibit hypercholanemia and hyperbilirubinemia. Pharmacokinetic analysis following a constant intravenous infusion in Atp11c mutant mice showed decreased hepatic sinusoidal uptake and intact biliary secretion of [(3)H]17ß estradiol 17ß-d-glucuronide. Consistent with this result, compared with cells and membranes from control mice, isolated hepatocytes, and liver plasma membranes from Atp11c mutant mice had a much lower uptake of [(3)H]17ß estradiol 17ß-d-glucuronide and expression of organic anion-transporting polypeptides, which are transporters responsible for hepatic uptake of unconjugated BAs and organic anions, including bilirubin glucuronides. Uptake of [(3)H]TC into hepatocytes and expression of Na(+)-taurocholate cotransporting polypeptide in liver plasma membranes, which mediates hepatic uptake of conjugated BAs, was also lower in the Atp11c mutant mice. Bile flow rate, biliary BA concentration, and expression of hepatobiliary transporters did not differ between Atp11c mutant mice and control mice. These results suggest that Atp11c mediates the transport of BAs and organic anions across the sinusoidal membrane, but not the canalicular membrane, by regulating the abundance of transporters. Atp11c is a candidate gene for genetically undiagnosed cases of hypercholanemia and hyperbilirubinemia, but not of intrahepatic cholestasis. This gene may influence the pharmacological and adverse effect of drugs because organic anion-transporting polypeptides regulate their systemic exposure.


Subject(s)
Adenosine Triphosphatases/genetics , Cholic Acids/blood , Hyperbilirubinemia/genetics , Hyperbilirubinemia/metabolism , Liver/metabolism , Membrane Transport Proteins/genetics , Organic Anion Transporters/metabolism , Steroid Metabolism, Inborn Errors/genetics , Steroid Metabolism, Inborn Errors/metabolism , Animals , Bile Acids and Salts/metabolism , Cholic Acids/genetics , Cholic Acids/metabolism , Hepatocytes/metabolism , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics
8.
Dig Dis ; 33(3): 314-8, 2015.
Article in English | MEDLINE | ID: mdl-26045263

ABSTRACT

P4 ATPases are lipid flippases and transport phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes. Lipid flipping is important for the biogenesis of transport vesicles. Recently it was shown that loss of the P4 ATPases ATP8B1 and ATP11C are associated with severe Cholestatic liver disease. Mutation of ATP8B1 cause progressive familial Intrahepatic Cholestasis type 1 (PFIC1)and benign recurrent intrahepatic cholestasis type 1 (BRIC 1). From our observations we hypothesized that ATP8B1 deficiency causes a phospholipids randomization at the canalicular membrane, which results in extraction of cholesterol due to increase sensitivity of the canalicular membrane. Deficiency of ATP11C causes conjugated hyperbilirubinemia. In our preliminary result we observed accumulation of unconjugated bile salts in Atp11c deficient mice probably because of regulation in the expression or function of OATP1B2. Similar to ATP8B1, ATP11C have regulation on membrane transporters.


Subject(s)
Adenosine Triphosphatases/genetics , Cholestasis, Intrahepatic/genetics , Phospholipid Transfer Proteins/genetics , Adenosine Triphosphatases/deficiency , Animals , Cholic Acids/blood , Cholic Acids/genetics , Hepatocytes/physiology , Humans , Hyperbilirubinemia/genetics , Mice , Mutation , Phospholipid Transfer Proteins/deficiency , Steroid Metabolism, Inborn Errors/genetics
9.
Dig Dis ; 33(3): 300-6, 2015.
Article in English | MEDLINE | ID: mdl-26045261

ABSTRACT

Sulfated progesterone metabolites rise 100-fold in the third trimester of human pregnancy and have been shown to be elevated further in the gestational disorder intrahepatic cholestasis of pregnancy (ICP). Typical concentrations of progesterone sulfates range from 1 to 10 µmol/L in an uncomplicated pregnancy and rise to approximately 40 µmol/L in ICP. At this level they can influence bile acid and lipid metabolism. Studies using human and rodent specimens have shown that sulfated metabolites of progesterone competitively inhibit bile acid homeostasis pathways by functioning as partial agonists of farnesoid X receptor (FXR). This explains the loss of induction of FXR target genes in ICP, and may explain susceptibility to hypercholanaemia and dyslipidaemia in the second half of human pregnancy. Furthermore, progesterone sulfates are competitive inhibitors of biliary influx (NTCP) and efflux (BSEP) transport proteins, actions likely to further exacerbate hypercholanaemia and cholestasis.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis, Intrahepatic/metabolism , Cholic Acids/blood , Pregnancy Complications/metabolism , Progesterone/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/metabolism , Cholestasis, Intrahepatic/etiology , Female , Homeostasis , Humans , Organic Anion Transporters, Sodium-Dependent/metabolism , Pregnancy , Pregnancy Complications/etiology , Pregnanolone/analogs & derivatives , Pregnanolone/metabolism , Symporters/metabolism
10.
J Hum Genet ; 59(2): 88-94, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24284361

ABSTRACT

The LDL receptor has an essential role in regulating plasma LDL-C levels. Genetic variation in the LDLR gene can be associated with either lower or moderately raised plasma levels of LDL-C, or may cause familial hypercholesterolemia. The prevalence of single-nucleotide polymorphisms (SNPs) in the LDLR in the black South African population is not known and therefore, we aimed to determine the genotypic variation of the LDLR in the study population as well as to define the association of the different genotypes with plasma LDL-C levels. A random selection of 1860 apparently healthy black South African volunteers aged 35-60 years was made in a cross-sectional study. Novel SNPs were identified in a subset of 30 individuals by means of automated sequencing before screening the entire cohort by means of the Illumina VeraCode GoldenGate Genotyping Assay on a BeadXpress Reader system. Twenty-five SNPs were genotyped, two of which were novel. A very rare SNP, rs17249141, in the promoter region was significantly associated with lower levels of LDL-C. Four other SNPs (rs2738447, rs14158, rs2738465 and rs3180023) were significantly associated with increased levels of LDL-C. We can conclude that some of the various SNPs identified do indeed associate with LDL-C levels.


Subject(s)
Black People/genetics , Cholesterol, LDL/blood , Cholic Acids/blood , Polymorphism, Single Nucleotide , Receptors, LDL/genetics , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/genetics , Adult , Cholesterol, LDL/genetics , Cholic Acids/genetics , Cross-Sectional Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Receptors, LDL/metabolism , South Africa
11.
Biochim Biophys Acta ; 1822(9): 1397-410, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22465940

ABSTRACT

The importance of peroxisomes in lipid metabolism is now well established and peroxisomes contain approximately 60 enzymes involved in these lipid metabolic pathways. Several acyl-CoA thioesterase enzymes (ACOTs) have been identified in peroxisomes that catalyze the hydrolysis of acyl-CoAs (short-, medium-, long- and very long-chain), bile acid-CoAs, and methyl branched-CoAs, to the free fatty acid and coenzyme A. A number of acyltransferase enzymes, which are structurally and functionally related to ACOTs, have also been identified in peroxisomes, which conjugate (or amidate) bile acid-CoAs and acyl-CoAs to amino acids, resulting in the production of amidated bile acids and fatty acids. The function of ACOTs is to act as auxiliary enzymes in the α- and ß-oxidation of various lipids in peroxisomes. Human peroxisomes contain at least two ACOTs (ACOT4 and ACOT8) whereas mouse peroxisomes contain six ACOTs (ACOT3, 4, 5, 6, 8 and 12). Similarly, human peroxisomes contain one bile acid-CoA:amino acid N-acyltransferase (BAAT), whereas mouse peroxisomes contain three acyltransferases (BAAT and acyl-CoA:amino acid N-acyltransferases 1 and 2: ACNAT1 and ACNAT2). This review will focus on the human and mouse peroxisomal ACOT and acyltransferase enzymes identified to date and discuss their cellular localizations, emerging structural information and functions as auxiliary enzymes in peroxisomal metabolic pathways.


Subject(s)
Acyltransferases/physiology , Lipid Metabolism , Palmitoyl-CoA Hydrolase/physiology , Peroxisomes/enzymology , Acyl Coenzyme A/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Bile Acids and Salts/metabolism , Cholic Acids/blood , Cholic Acids/genetics , Humans , Hydrolysis , Models, Molecular , Palmitoyl-CoA Hydrolase/chemistry , Palmitoyl-CoA Hydrolase/metabolism , Peroxisomes/metabolism , Protein Conformation , Steroid Metabolism, Inborn Errors/enzymology , Steroid Metabolism, Inborn Errors/genetics
12.
Toxicol Appl Pharmacol ; 273(3): 680-90, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24183703

ABSTRACT

Previous studies showed glucose and insulin signaling can regulate bile acid (BA) metabolism during fasting or feeding. However, limited knowledge is available on the effect of calorie restriction (CR), a well-known anti-aging intervention, on BA homeostasis. To address this, the present study utilized a "dose-response" model of CR, where male C57BL/6 mice were fed 0, 15, 30, or 40% CR diets for one month, followed by BA profiling in various compartments of the enterohepatic circulation by UPLC-MS/MS technique. This study showed that 40% CR increased the BA pool size (162%) as well as total BAs in serum, gallbladder, and small intestinal contents. In addition, CR "dose-dependently" increased the concentrations of tauro-cholic acid (TCA) and many secondary BAs (produced by intestinal bacteria) in serum, such as tauro-deoxycholic acid (TDCA), DCA, lithocholic acid, ω-muricholic acid (ωMCA), and hyodeoxycholic acid. Notably, 40% CR increased TDCA by over 1000% (serum, liver, and gallbladder). Interestingly, 40% CR increased the proportion of 12α-hydroxylated BAs (CA and DCA), which correlated with improved glucose tolerance and lipid parameters. The CR-induced increase in BAs correlated with increased expression of BA-synthetic (Cyp7a1) and conjugating enzymes (BAL), and the ileal BA-binding protein (Ibabp). These results suggest that CR increases BAs in male mice possibly through orchestrated increases in BA synthesis and conjugation in liver as well as intracellular transport in ileum.


Subject(s)
Bile Acids and Salts/blood , Caloric Restriction , Enterohepatic Circulation/drug effects , Animals , Bile Acids and Salts/biosynthesis , Blood Glucose/metabolism , Body Weight , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , Cholic Acids/blood , Deoxycholic Acid/blood , Dose-Response Relationship, Drug , Homeostasis , Intestines/drug effects , Intestines/enzymology , Lipids/blood , Liver/drug effects , Liver/enzymology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tandem Mass Spectrometry
13.
Heart Lung Circ ; 21(9): 543-50, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22633397

ABSTRACT

Familial hypercholesterolaemia (FH) is a co-dominantly inherited disorder that causes marked elevation in plasma cholesterol and premature coronary heart disease. There are at least 45,000 people with FH in Australia and New Zealand, but most remain undiagnosed and undertreated. To bridge this gap in coronary prevention the FH Australasia Network has developed a model of care for FH. We present the executive summary, with a commentary contrasting the recommendations with other international guidelines and highlighting the role of the cardiologist.


Subject(s)
Coronary Disease/therapy , Steroid Metabolism, Inborn Errors/therapy , Australia , Cardiology , Cholesterol/blood , Cholic Acids/blood , Coronary Disease/blood , Coronary Disease/etiology , Humans , New Zealand , Practice Guidelines as Topic , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/complications
14.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166269, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34537368

ABSTRACT

OBJECTIVE: To explore the clinical features, fetal outcomes and serum bile acids (BAs) metabolism in asymptomatic hypercholanemia of pregnancy (AHP), as well as the comparison with those in intrahepatic cholestasis of pregnancy (ICP) and normal pregnancies. METHODS: A study containing 676 pregnant women was performed to investigate the clinical informations, routine biochemical features and obstetric outcomes of AHP by the comparison with ICP and normal pregnancies. Within the study subjects, 203 pregnant women received prospective determination for 55 serum individual BAs based on a validated UPLC-QTOF-MS/MS method. The differences in clinical features and serum BAs metabolism among the three groups were then investigated. RESULTS: The risk of adverse fetal outcomes in AHP (28.3%) was significantly higher than that in normal pregnancies (8.9%, p < 0.001), but lower than that in ICP group (52.1%, p < 0.001). Multivariate statistics analysis indicated a distinctive serum BAs metabolic profiling among the three groups (PLS-DA, R2Y = 0.580, Q2 = 0.537). Levels of serum BAs especially for deoxycholic acid species were found remarkably elevated in AHP as compared to those in ICP. CONCLUSIONS: AHP group had distinguished clinical features and serum BAs metabolism as compared to ICP group and normal pregnancies.


Subject(s)
Bile Acids and Salts/blood , Cholestasis, Intrahepatic/metabolism , Cholic Acids/blood , Lipid Metabolism , Pregnancy Complications/metabolism , Steroid Metabolism, Inborn Errors/metabolism , Adult , Asymptomatic Diseases/epidemiology , Cholestasis, Intrahepatic/blood , Cholestasis, Intrahepatic/epidemiology , Cholestasis, Intrahepatic/pathology , Cholic Acids/metabolism , Female , Fetus , Humans , Metabolomics/standards , Multivariate Analysis , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/epidemiology , Pregnancy Complications/pathology , Steroid Metabolism, Inborn Errors/blood , Steroid Metabolism, Inborn Errors/epidemiology , Steroid Metabolism, Inborn Errors/pathology , Tandem Mass Spectrometry
16.
Nat Commun ; 12(1): 1487, 2021 03 05.
Article in English | MEDLINE | ID: mdl-33674561

ABSTRACT

Hyocholic acid (HCA) is a major bile acid (BA) species in the BA pool of pigs, a species known for its exceptional resistance to spontaneous development of diabetic phenotypes. HCA and its derivatives are also present in human blood and urine. We investigate whether human HCA profiles can predict the development of metabolic disorders. We find in the first cohort (n = 1107) that both obesity and diabetes are associated with lower serum concentrations of HCA species. A separate cohort study (n = 91) validates this finding and further reveals that individuals with pre-diabetes are associated with lower levels of HCA species in feces. Serum HCA levels increase in the patients after gastric bypass surgery (n = 38) and can predict the remission of diabetes two years after surgery. The results are replicated in two independent, prospective cohorts (n = 132 and n = 207), where serum HCA species are found to be strong predictors for metabolic disorders in 5 and 10 years, respectively. These findings underscore the association of HCA species with diabetes, and demonstrate the feasibility of using HCA profiles to assess the future risk of developing metabolic abnormalities.


Subject(s)
Biomarkers/blood , Cholic Acids/blood , Cholic Acids/urine , Metabolic Diseases/diagnosis , Adult , Cohort Studies , Cross-Sectional Studies , Diabetes Mellitus, Type 2/metabolism , Feces/chemistry , Female , Humans , Longitudinal Studies , Male , Middle Aged , Obesity/metabolism , Overweight/metabolism , Prediabetic State/diagnosis , Prospective Studies
17.
Sci Rep ; 11(1): 13252, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168163

ABSTRACT

Knowledge about in vivo effects of human circulating C-6 hydroxylated bile acids (BAs), also called muricholic acids, is sparse. It is unsettled if the gut microbiome might contribute to their biosynthesis. Here, we measured a range of serum BAs and related them to markers of human metabolic health and the gut microbiome. We examined 283 non-obese and obese Danish adults from the MetaHit study. Fasting concentrations of serum BAs were quantified using ultra-performance liquid chromatography-tandem mass-spectrometry. The gut microbiome was characterized with shotgun metagenomic sequencing and genome-scale metabolic modeling. We find that tauro- and glycohyocholic acid correlated inversely with body mass index (P = 4.1e-03, P = 1.9e-05, respectively), waist circumference (P = 0.017, P = 1.1e-04, respectively), body fat percentage (P = 2.5e-03, P = 2.3e-06, respectively), insulin resistance (P = 0.051, P = 4.6e-4, respectively), fasting concentrations of triglycerides (P = 0.06, P = 9.2e-4, respectively) and leptin (P = 0.067, P = 9.2e-4). Tauro- and glycohyocholic acids, and tauro-a-muricholic acid were directly linked with a distinct gut microbial community primarily composed of Clostridia species (P = 0.037, P = 0.013, P = 0.027, respectively). We conclude that serum conjugated C-6-hydroxylated BAs associate with measures of human metabolic health and gut communities of Clostridia species. The findings merit preclinical interventions and human feasibility studies to explore the therapeutic potential of these BAs in obesity and type 2 diabetes.


Subject(s)
Bile Acids and Salts/blood , Clostridium/metabolism , Gastrointestinal Microbiome , Adiposity , Body Mass Index , Cholic Acids/blood , Chromatography, High Pressure Liquid , Clostridium/genetics , Deoxycholic Acid/blood , Female , Gastrointestinal Microbiome/genetics , Humans , Logistic Models , Male , Metagenomics , Middle Aged , Obesity/blood , Obesity/microbiology , Tandem Mass Spectrometry , Taurocholic Acid/blood , Waist Circumference
18.
Cell Metab ; 33(4): 791-803.e7, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33338411

ABSTRACT

Hyocholic acid (HCA) and its derivatives are found in trace amounts in human blood but constitute approximately 76% of the bile acid (BA) pool in pigs, a species known for its exceptional resistance to type 2 diabetes. Here, we show that BA depletion in pigs suppressed secretion of glucagon-like peptide-1 (GLP-1) and increased blood glucose levels. HCA administration in diabetic mouse models improved serum fasting GLP-1 secretion and glucose homeostasis to a greater extent than tauroursodeoxycholic acid. HCA upregulated GLP-1 production and secretion in enteroendocrine cells via simultaneously activating G-protein-coupled BA receptor, TGR5, and inhibiting farnesoid X receptor (FXR), a unique mechanism that is not found in other BA species. We verified the findings in TGR5 knockout, intestinal FXR activation, and GLP-1 receptor inhibition mouse models. Finally, we confirmed in a clinical cohort, that lower serum concentrations of HCA species were associated with diabetes and closely related to glycemic markers.


Subject(s)
Cholic Acids/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Glucose/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Blood Glucose/analysis , Cell Line , Cholic Acids/blood , Cholic Acids/chemistry , Cholic Acids/pharmacology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Isoxazoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Swine
19.
Ann Saudi Med ; 40(2): 81-86, 2020.
Article in English | MEDLINE | ID: mdl-32241166

ABSTRACT

BACKGROUND: The relationship between breast arterial calcification (BAC) and angiographic coronary artery disease (CAD) is uncertain. Some studies have shown a positive association between BAC and angiographically proven CAD, while other studies have shown no association. OBJECTIVE: Examine the association between visually detected BAC on mammography and CAD found on invasive coronary angiography (ICA) in women and compare the frequency of risk factors for CAD between women with normal and abnormal ICA. DESIGN: Retrospective. SETTING: Single tertiary care center. PATIENTS AND METHODS: A review of the radiology databases was performed for female patients who underwent both ICA and mammography within six months of each other. Cases were excluded if there was a history of CAD, such as coronary artery bypass graft or prior percutaneous coronary intervention. MAIN OUTCOME MEASURES: BAC as a predictor of obstructive CAD on ICA. SAMPLE SIZE: 203 Saudi women RESULTS: The association between age at catheterization and ICA was statistically significant ( P=.01). There was no association between BAC and abnormal ICA ( P=.108). Women with abnormal ICA were older than women with a normal ICA ( P=.01). There was a higher frequency of CAD risk factors among the patients with abnormal ICA, except for smoking. In the multiple logistic regression model, ICA was associated with age, a family history of CAD, diabetes mellitus, hypertension and hypercholesterolemia. BAC-positive women were older than BAC-negative women ( P=.0001). BAC was associated with age, diabetes, hypertension, and chronic kidney disease in the multiple logistic regression model. CONCLUSIONS: BAC on mammography did not predict angiographically proven CAD. There was a strong association between BAC and age and many other conventional CAD risk factors. LIMITATIONS: Relatively small sample, single-center retrospective study. CONFLICT OF INTEREST: None.


Subject(s)
Breast/blood supply , Coronary Artery Disease/epidemiology , Coronary Stenosis/epidemiology , Vascular Calcification/epidemiology , Age Factors , Aged , Breast/diagnostic imaging , Cardiac Catheterization , Cholic Acids/blood , Coronary Angiography , Coronary Artery Disease/diagnosis , Coronary Stenosis/diagnosis , Diabetes Mellitus/epidemiology , Female , Humans , Hypertension/epidemiology , Logistic Models , Mammography , Middle Aged , Saudi Arabia/epidemiology , Steroid Metabolism, Inborn Errors/epidemiology , Vascular Calcification/diagnostic imaging
20.
Curr Diabetes Rev ; 16(8): 900-909, 2020.
Article in English | MEDLINE | ID: mdl-32013849

ABSTRACT

BACKGROUND: Recent studies have suggested that hyperglycaemia influences the bile acid profile and concentrations of secondary bile acids in the gut. INTRODUCTION: This study aimed to measure changes in the bile acid profile in the gut, tissues, and faeces in type 1 Diabetes (T1D) and Type 2 Diabetes (T2D). METHODS: T1D and T2D were established in a mouse model. Twenty-one seven-weeks old balb/c mice were randomly divided into three equal groups, healthy, T1D and T2D. Blood, tissue, urine and faeces samples were collected for bile acid measurements. RESULTS: Compared with healthy mice, T1D and T2D mice showed lower levels of the primary bile acid, chenodeoxycholic acid, in the plasma, intestine, and brain, and higher levels of the secondary bile acid, lithocholic acid, in the plasma and pancreas. Levels of the bile acid ursodeoxycholic acid were undetected in healthy mice but were found to be elevated in T1D and T2D mice. CONCLUSION: Bile acid profiles in other organs were variably influenced by T1D and T2D development, which suggests similarity in effects of T1D and T2D on the bile acid profile, but these effects were not always consistent among all organs, possibly since feedback mechanisms controlling enterohepatic recirculation and bile acid profiles and biotransformation are different in T1D and T2D.


Subject(s)
Cholic Acids/analysis , Diabetes Mellitus, Type 1 , Diabetes Mellitus, Type 2 , Animals , Bile Acids and Salts/analysis , Bile Acids and Salts/blood , Bile Acids and Salts/urine , Blood Glucose/analysis , Brain Chemistry , Cholic Acids/blood , Cholic Acids/urine , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/urine , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/urine , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/urine , Disease Models, Animal , Feces/chemistry , Gastrointestinal Tract/chemistry , Hyperglycemia/blood , Hyperglycemia/urine , Male , Mice , Mice, Inbred BALB C , Muscles/chemistry , Random Allocation
SELECTION OF CITATIONS
SEARCH DETAIL