Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 384
Filter
1.
Wiad Lek ; 77(2): 262-267, 2024.
Article in English | MEDLINE | ID: mdl-38592987

ABSTRACT

OBJECTIVE: Aim: The current study was designed to investigate the role of ABCG5 and ABCG5 in serum with normal and expected cardiac complaints with CVDs as individual early diagnostic tools. PATIENTS AND METHODS: Materials and Methods: Data was collected in paper form and recorded from 100 healthy personals and 100 personals suspected with CVS after take the case history and clinical signs in private clinical hospital and the serum was collected for measurements the activity of ABCG5 and ABCG5 by used ELISA reader and the results illustrated that activity of ABCG5 and ABCG5 in all aged groups. RESULTS: Results: Activity of ABCG5 and ABCG5 in all aged groups periods in patient person male and female significant decrease as compared with same age in same period of live, so that the researched depicted that can used the serum activity of ABCG5 and ABCG5 as a diagnostics tools for atherosclerotic cardiovascular disease. CONCLUSION: Conclusions: We identified areas of further exploration on cholesterol transport related with CVD risk and concluded that changes in the Adenosine Triphosphate Binding Cassette transporters mainly G5 and G8 early diagnostic tools for cardiovascular disease in Human. We correlated areas of farther disquisition on nutrient cholesterol and CVD threat, in the included trials, healthy grown-ups consumed high doses of dietary cholesterol.


Subject(s)
Cardiovascular Diseases , Lipoproteins , Humans , Male , Female , Aged , Lipoproteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Cardiovascular Diseases/diagnosis , Adenosine Triphosphate/metabolism , ATP-Binding Cassette Transporters/metabolism , Cholesterol/metabolism
2.
Hum Genomics ; 18(1): 19, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347599

ABSTRACT

The causal relationships between plasma metabolites and cholelithiasis/cholecystitis risks remain elusive. Using two-sample Mendelian randomization, we found that genetic proxied plasma campesterol level showed negative correlation with the risk of both cholelithiasis and cholecystitis. Furthermore, the increased risk of cholelithiasis is correlating with the increased level of plasma campesterol. Lastly, genetic colocalization study showed that the leading SNP, rs4299376, which residing at the ABCG5/ABCG8 gene loci, was shared by plasma campesterol level and cholelithiasis, indicating that the aberrant transportation of plant sterol/cholesterol from the blood stream to the bile duct/gut lumen might be the key in preventing cholesterol gallstone formation.


Subject(s)
Cholecystitis , Cholesterol/analogs & derivatives , Gallstones , Phytosterols , Humans , Lipoproteins/genetics , Mendelian Randomization Analysis , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Cholecystitis/epidemiology , Cholecystitis/genetics , Gallstones/epidemiology , Gallstones/genetics , Gallstones/metabolism
3.
Hepatology ; 79(5): 986-1004, 2024 May 01.
Article in English | MEDLINE | ID: mdl-37976384

ABSTRACT

BACKGROUND AND AIMS: Parenteral nutrition-associated cholestasis (PNAC) is an important complication in patients with intestinal failure with reduced LRH-1 expression. Here, we hypothesized that LRH-1 activation by its agonist, dilauroylphosphatidylcholine (DLPC), would trigger signal transducer and activator of transcription 6 (STAT6) signaling and hepatic macrophage polarization that would mediate hepatic protection in PNAC. APPROACH AND RESULTS: PNAC mouse model (oral DSSx4d followed by PNx14d; DSS-PN) was treated with LRH-1 agonist DLPC (30 mg/kg/day) intravenously. DLPC treatment prevented liver injury and cholestasis while inducing hepatic mRNA expression of Nr5a2 (nuclear receptor subfamily 5 group A member 2), Abcb11 (ATP binding cassette subfamily B member 11), Abcg5 (ATP-binding cassette [ABC] transporters subfamily G member 5), Abcg8 (ATP-binding cassette [ABC] transporters subfamily G member 8), nuclear receptor subfamily 0, and ATP-binding cassette subfamily C member 2 ( Abcc2) mRNA, all of which were reduced in PNAC mice. To determine the mechanism of the DLPC effect, we performed RNA-sequencing analysis of the liver from Chow, DSS-PN, and DSS-PN/DLPC mice, which revealed DLPC upregulation of the anti-inflammatory STAT6 pathway. In intrahepatic mononuclear cells or bone-marrow derived macrophages (BMDM) from PNAC mice, DLPC treatment prevented upregulation of pro-inflammatory (M1) genes, suppressed activation of NFκB and induced phosphorylation of STAT6 and its target genes, indicating M2 macrophage polarization. In vitro, incubation of DLPC with cultured macrophages showed that the increased Il-1b and Tnf induced by exposure to lipopolysaccharides or phytosterols was reduced significantly, which was associated with increased STAT6 binding to promoters of its target genes. Suppression of STAT6 expression by siRNA in THP-1 cells exposed to lipopolysaccharides, phytosterols, or both resulted in enhanced elevation of IL-1B mRNA expression. Furthermore, the protective effect of DLPC in THP-1 cells was abrogated by STAT6 siRNA. CONCLUSIONS: These results indicate that activation of LRH-1 by DLPC may protect from PNAC liver injury through STAT6-mediated macrophage polarization.


Subject(s)
Cholestasis , Phosphatidylcholines , Phytosterols , Humans , Mice , Animals , Lipoproteins/metabolism , STAT6 Transcription Factor/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Cholestasis/etiology , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Kupffer Cells/metabolism , RNA, Small Interfering , RNA, Messenger/metabolism , Parenteral Nutrition/adverse effects , Adenosine Triphosphate
4.
PLoS One ; 18(6): e0287146, 2023.
Article in English | MEDLINE | ID: mdl-37310967

ABSTRACT

Macrophage inhibitory cytokine 1 (MIC-1), which is overproduced in various human cancers and associated with cachexia, acts on the hypothalamus to suppress appetite and reduce body weight. We investigated the mechanisms through which MIC-1 affects bile acid metabolism and gallstone formation, which are poorly understood. Over 6 weeks, male C57BL/6 mice fed either standard chow or a lithogenic diet were intraperitoneally injected with phosphate-buffered saline (PBS) or MIC-1 (200 µg/kg/week). Among lithogenic diet-fed mice, MIC-1 treatment resulted in increased gallstone formation compared with PBS treatment. Compared with PBS treatment, MIC-1 treatment decreased hepatic cholesterol and bile acid levels and reduced expression of HMG-CoA reductase (HMGCR), the master cholesterol metabolism regulator sterol regulatory element-binding protein 2, cholesterol 7α-hydroxylase (CYP7A1), mitochondrial sterol 27-hydroxylase, and oxysterol 7α-hydroxylase. Compared with PBS treatment, MIC-1 treatment had no effect on small heterodimer partner, farnesoid X receptor, or pregnane X receptor expression, and extracellular signal-related kinase and c-Jun N-terminal kinase phosphorylation decreased, suggesting that these factors do not contribute to the MIC-1-induced reduction in CYP7A1 expression. Compared with PBS treatment, MIC-1 treatment increased AMP-activated protein kinase (AMPK) phosphorylation. Treatment with the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) reduced CYP7A1 and HMGCR expression, whereas the AMPK inhibitor Compound C reversed MIC-1-induced reductions in CYP7A1 and HMGCR expression. Furthermore, in MIC-1-treated mice, total biliary cholesterol levels increased together with increased ATP-binding cassette subfamily G (ABCG)5 and ABCG8 expression. Compared with PBS treatment, MIC-1 treatment did not affect expression of liver X receptors α and ß, liver receptor homolog 1, hepatocyte nuclear factor 4α, or NR1I3 (also known as constitutive androstane receptor), which are upstream of ABCG5/8; however, MIC-1 treatment increased ABCG5/8 expression and promoter activities. Our study indicates that MIC-1 influences gallstone formation by increasing AMPK phosphorylation, reducing CYP7A1 and HMGCR expression, and increasing ABCG5 and ABCG8 expression.


Subject(s)
Gallstones , Humans , Male , Animals , Mice , Mice, Inbred C57BL , Growth Differentiation Factor 15 , AMP-Activated Protein Kinases , Diet , Macrophages , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Lipoproteins
5.
Asian J Surg ; 46(9): 3560-3567, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37344314

ABSTRACT

BACKGROUND: The overexpression of the cholesterol transporter: ATP-binding cassette transporter (ABCG8) due to the effect of ABCG8 genetic variant (rs11887534) leads to the precipitation of cholesterol crystals and gallstone disease (GSD). OBJECTIVE: To evaluate the chemical composition of gallstones and the role of ABCG8 (rs11887534) in the susceptibility to GSD. METHODS: We enrolled 77 patients with GSD treated with standard laparoscopic or open cholecystectomy and 75 age and sex-matched healthy controls. Chemical analysis of the extracted gallstones was performed. Analysis of the rs11887534 was performed by real-time PCR TaqMan technique for both cases and controls. RESULTS: Pure cholesterol stones were the main type of stones in GSD patients. The CC genotype carriers of rs11887534 were more prone to gallstone formation than other genotypes. The CC genotype carriers were 100 folds at increased risk to develop pure cholesterol stones. CONCLUSION: The most prevalent type of gallbladder stones is pure cholesterol stone. ABCG8 (rs11887534) could be associated with increased risk for cholesterol gallstones formation, this risk was more pronounced in female patients.


Subject(s)
Gallstones , Humans , Female , Genetic Predisposition to Disease , Egypt , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Cholesterol
6.
Genes (Basel) ; 14(3)2023 03 20.
Article in English | MEDLINE | ID: mdl-36981027

ABSTRACT

ABCG5 and ABCG8 are two key adenosine triphosphate-binding cassette (ABC) proteins that regulate whole-body sterol trafficking. This study aimed to elucidate the association between ABCG5/G8 gene region variants and lipid profile, cardiometabolic traits, and gallstone disease history in Taiwan. A total of 1494 Taiwan Biobank participants with whole-genome sequencing data and 117,679 participants with Axiom Genome-Wide CHB Array data were enrolled for analysis. Using genotype-phenotype and stepwise linear regression analyses, we found independent associations of four Asian-specific ABCG5 variants, rs119480069, rs199984328, rs560839317, and rs748096191, with total, low-density lipoprotein (LDL), and non-high-density lipoprotein (HDL) cholesterol levels (all p ≤ 0.0002). Four other variants, which were in nearly complete linkage disequilibrium, exhibited genome-wide significant associations with gallstone disease history, and the ABCG8 rs11887534 variant showed a trend of superiority for gallstone disease history in a nested logistic regression model (p = 0.074). Through regional association analysis of various other cardiometabolic traits, two variants of the PLEKHH2, approximately 50 kb from the ABCG5/G8 region, exhibited significant associations with blood pressure status (p < 10-6). In conclusion, differential effects of ABCG5/G8 region variants were noted for lipid profile, blood pressure status, and gallstone disease history in Taiwan. These results indicate the crucial role of individualized assessment of ABCG5/G8 variants for different cardiometabolic phenotypes.


Subject(s)
Cardiovascular Diseases , Gallstones , Humans , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , Blood Pressure/genetics , Taiwan , Lipoproteins/genetics , Gallstones/genetics , Cholesterol
7.
Nutrients ; 15(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36839356

ABSTRACT

ATP-binding cassette protein G5 (ABCG5)/ABCG8 heterodimer exports cholesterol from cells, while Niemann-Pick C1-like 1 (NPC1L1) imports cholesterol and vitamin K. We examined whether ABCG5/ABCG8 transports vitamin K similar to NPC1L1. Since high concentrations of vitamin K3 show cytotoxicity, the cytoprotective effects of ABCG5/ABCG8 were examined. BHK cells expressing ABCG5/ABCG8 were more resistant to vitamin K3 cytotoxicity than control cells, suggesting that ABCG5/ABCG8 transports vitamin K3 out of cells. The addition of vitamin K1 reversed the effects of ABCG5/ABCG8, suggesting that vitamin K1 competitively inhibits the transport of vitamin K3. To examine the transport of vitamin K1 by ABCG5/ABCG8, vitamin K1 levels in the medium and cells were measured. Vitamin K1 levels in cells expressing ABCG5/ABCG8 were lower than those in control cells, while vitamin K1 efflux increased in cells expressing ABCG5/ABCG8. Furthermore, the biliary vitamin K1 concentration in Abcg5/Abcg8-deficient mice was lower than that in wild-type mice, although serum vitamin K1 levels were not affected by the presence of Abcg5/Abcg8. These findings suggest that ABCG5 and ABCG8 are involved in the transport of sterols and vitamin K. ABCG5/ABCG8 and NPC1L1 might play important roles in the regulation of vitamin K absorption and excretion.


Subject(s)
ATP-Binding Cassette Transporters , Lipoproteins , Mice , Animals , Lipoproteins/metabolism , ATP-Binding Cassette Transporters/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Vitamin K , Cholesterol/metabolism
8.
Adv Clin Chem ; 110: 145-169, 2022.
Article in English | MEDLINE | ID: mdl-36210074

ABSTRACT

Sitosterolemia is an inherited metabolic disorder characterized by increased levels of plant sterols, such as sitosterol. This disease is caused by loss-of-function genetic mutations in the ATP-binding cassette (ABC) subfamily G member 5 or member 8 (ABCG5 or ABCG8, respectively), both of which play important roles in the selective excretion of plant sterols from the liver and intestine, leading to a failure to excrete plant sterols. Sitosterolemia, which is currently considered a rare genetic disorder, has been described as a phenocopy of homozygous familial hypercholesterolemia (FH). Typical phenotypes of sitosterolemia, including elevated low-density lipoprotein (LDL) cholesterol, tendon xanthomas, and premature coronary artery disease, overlap those of homozygous FH; however, there are substantial differences between these two diseases in terms of treatments and prognoses. Moreover, it is of note that sitosterolemia appears to be quite underdiagnosed, although accurate diagnosis and appropriate interventions will likely to lead to better prognoses compared with homozygous FH. Unlike cases of homozygous FH, dietary counseling is quite effective in reducing the LDL cholesterol as well as sitosterol of patients with sitosterolemia. In this chapter, we summarize the current understandings of this disease and provide useful tips for the diagnosis as well as better treatment of patients with sitosterolemia.


Subject(s)
Phytosterols , Sitosterols , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adenosine Triphosphate , Cholesterol/metabolism , Cholesterol, LDL/metabolism , Humans , Hypercholesterolemia , Intestinal Diseases , Lipid Metabolism, Inborn Errors , Lipoproteins/genetics , Phytosterols/adverse effects , Phytosterols/genetics , Sitosterols/chemistry
9.
J Mol Biol ; 434(20): 167795, 2022 10 30.
Article in English | MEDLINE | ID: mdl-35988751

ABSTRACT

The ATP-binding cassette (ABC) sterol transporters are responsible for maintaining cholesterol homeostasis in mammals by participating in reverse cholesterol transport (RCT) or transintestinal cholesterol efflux (TICE). The heterodimeric ABCG5/G8 carries out selective sterol excretion, preventing the abnormal accumulation of plant sterols in human bodies, while homodimeric ABCG1 contributes to the biogenesis and metabolism of high-density lipoproteins. A sterol-binding site on ABCG5/G8 was proposed at the interface of the transmembrane domain and the core of lipid bilayers. In this study, we have determined the crystal structure of ABCG5/G8 in a cholesterol-bound state. The structure combined with amino acid sequence analysis shows that in the proximity of the sterol-binding site, a highly conserved phenylalanine array supports functional implications for ABCG cholesterol/sterol transporters. Lastly, in silico docking analysis of cholesterol and stigmasterol (a plant sterol) suggests sterol-binding selectivity on ABCG5/G8, but not ABCG1. Together, our results provide a structural basis for cholesterol binding on ABCG5/G8 and the sterol selectivity by ABCG transporters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Cholesterol , ATP Binding Cassette Transporter, Subfamily G, Member 5/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/chemistry , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Cholesterol/chemistry , Cholesterol/metabolism , Cryoelectron Microscopy , Humans , Lipid Bilayers/chemistry , Lipoproteins, HDL/metabolism , Phenylalanine/metabolism , Phytosterols/metabolism , Protein Binding , Protein Conformation , Stigmasterol/metabolism
10.
Nature ; 608(7922): 413-420, 2022 08.
Article in English | MEDLINE | ID: mdl-35922515

ABSTRACT

High cholesterol is a major risk factor for cardiovascular disease1. Currently, no drug lowers cholesterol through directly promoting cholesterol excretion. Human genetic studies have identified that the loss-of-function Asialoglycoprotein receptor 1 (ASGR1) variants associate with low cholesterol and a reduced risk of cardiovascular disease2. ASGR1 is exclusively expressed in liver and mediates internalization and lysosomal degradation of blood asialoglycoproteins3. The mechanism by which ASGR1 affects cholesterol metabolism is unknown. Here, we find that Asgr1 deficiency decreases lipid levels in serum and liver by stabilizing LXRα. LXRα upregulates ABCA1 and ABCG5/G8, which promotes cholesterol transport to high-density lipoprotein and excretion to bile and faeces4, respectively. ASGR1 deficiency blocks endocytosis and lysosomal degradation of glycoproteins, reduces amino-acid levels in lysosomes, and thereby inhibits mTORC1 and activates AMPK. On one hand, AMPK increases LXRα by decreasing its ubiquitin ligases BRCA1/BARD1. On the other hand, AMPK suppresses SREBP1 that controls lipogenesis. Anti-ASGR1 neutralizing antibody lowers lipid levels by increasing cholesterol excretion, and shows synergistic beneficial effects with atorvastatin or ezetimibe, two widely used hypocholesterolaemic drugs. In summary, this study demonstrates that targeting ASGR1 upregulates LXRα, ABCA1 and ABCG5/G8, inhibits SREBP1 and lipogenesis, and therefore promotes cholesterol excretion and decreases lipid levels.


Subject(s)
Asialoglycoprotein Receptor , Cholesterol , Lipid Metabolism , AMP-Activated Protein Kinases/metabolism , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Asialoglycoprotein Receptor/antagonists & inhibitors , Asialoglycoprotein Receptor/deficiency , Asialoglycoprotein Receptor/genetics , Asialoglycoprotein Receptor/metabolism , Asialoglycoproteins/metabolism , Atorvastatin/pharmacology , BRCA1 Protein , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cholesterol/metabolism , Drug Synergism , Endocytosis , Ezetimibe/pharmacology , Humans , Lipids/analysis , Lipids/blood , Liver/metabolism , Liver X Receptors/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1 , Ubiquitin-Protein Ligases/metabolism
11.
Circ Genom Precis Med ; 15(3): e003390, 2022 06.
Article in English | MEDLINE | ID: mdl-35549507

ABSTRACT

BACKGROUND: Sitosterolemia is a rare autosomal recessive disorder caused by homozygous or compound heterozygous variants in ABCG5/ABCG8. The disease is characterized by increased plasma plant sterols. Small case series suggest that patients with sitosterolemia have wide phenotypic heterogeneity with great variability on either plasma cholesterol levels or development of atherosclerotic cardiovascular disease. The present study aims to characterize the prevalence and clinical features of sitosterolemia participating in a familial hypercholesterolemia genetic cascade screening program. METHODS: From 443 familial hypercholesterolemia index cases, 260 were negative for familial hypercholesterolemia genes and were sequenced for the ABCG5/8 genes. Clinical and laboratory characteristics of affected individuals were determined. RESULTS: Eight (3.1%) index cases were found to be homozygous or compound heterozygous variant for ABCG5/ABCG8 genes, confirming the genetic diagnosis of sitosterolemia. Screening their relatives led to the identification of 6 additional confirmed sitosterolemia cases (3 homozygous and 3 compound heterozygous variant) and 18 carriers (heterozygous). The mean age of identified sitosterolemia cases (n=14) was 37.2±19.8 years, 50% were females, and 78.6% (all adults) presented either clinical or subclinical atherosclerotic cardiovascular disease. As expected, affected individuals presented elevated plasma plant sterol levels (mean ß-Sitosterol and campesterol, respectively, 160.3±107.1 and 32.0±19.6 µg/mL) and the highest plasma LDL (low-density lipoprotein)-cholesterol was 269.0±120.0 mg/dL (range: 122-521 mg/dL). LDL-cholesterol mean reduction with therapy among cases was 65%. Eighty-three percent (83%) of identified sitosterolemia patients presented hematologic abnormalities. CONCLUSIONS: Testing genes associated with sitosterolemia in the molecular routine workflow of a familial hypercholesterolemia cascade screening program allowed the precise diagnosis of sitosterolemia in a substantial number of patients with varying LDL-C levels and high incidence of early atherosclerotic cardiovascular disease and hematologic abnormalities.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , Cardiovascular Diseases , Hypercholesterolemia , Hyperlipoproteinemia Type II , Intestinal Diseases , Lipid Metabolism, Inborn Errors , Phytosterols , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adolescent , Adult , Cardiovascular Diseases/genetics , Cholesterol , Cholesterol, LDL , Female , Humans , Hypercholesterolemia/genetics , Hyperlipoproteinemia Type II/genetics , Intestinal Diseases/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipoproteins/genetics , Male , Middle Aged , Phytosterols/adverse effects , Young Adult
12.
Protein Sci ; 31(5): e4297, 2022 05.
Article in English | MEDLINE | ID: mdl-35481657

ABSTRACT

ATP-binding cassette (ABC) systems, characterized by ABC-type nucleotide-binding domains (NBDs), play crucial roles in various aspects of human physiology. Human ABCG5 and ABCG8 form a heterodimeric transporter that functions in the efflux of sterols. We used sequence similarity search, multiple sequence alignment, phylogenetic analysis, and structure comparison to study the evolutionary origin and sequence signatures of ABCG5 and ABCG8. Orthologs of ABCG5 and ABCG8, supported by phylogenetic analysis and signature residues, were identified in bilaterian animals, Filasterea, Fungi, and Amoebozoa. Such a phylogenetic distribution suggests that ABCG5 and ABCG8 could have originated in the last common ancestor of Amorphea (the unikonts), the eukaryotic group including Amoebozoa and Opisthokonta. ABCG5 and ABCG8 were missing in genomes of various lineages such as snakes, jawless vertebrates, non-vertebrate chordates, echinoderms, and basal metazoan groups. Amino-acid changes in key positions in ABCG8 Walker A motif and/or ABCG5 C-loop were observed in most tetrapod organisms, likely resulted in the loss of ATPase activity at one nucleotide-binding site. ABCG5 and ABCG8 in Ecdysozoa (such as insects) exhibit elevated evolutionary rates and accumulate various changes in their NBD functional motifs. Alignment inspection revealed several residue positions that show different amino-acid usages in ABCG5/ABCG8 compared to other ABCG subfamily proteins. These residues were mapped to the structural cores of transmembrane domains (TMDs), the NBD-TMD interface, and the interface between TMDs. They serve as sequence signatures to differentiate ABCG5/ABCG8 from other ABCG subfamily proteins, and some of them may contribute to substrate specificity of the ABCG5/ABCG8 transporter.


Subject(s)
ATP-Binding Cassette Transporters , Lipoproteins , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Lipoproteins/chemistry , Nucleotides/metabolism , Phylogeny
13.
Clin Chim Acta ; 530: 39-44, 2022 May 01.
Article in English | MEDLINE | ID: mdl-35248527

ABSTRACT

BACKGROUND AND AIM: Clinical manifestations and genetic backgrounds of Japanese patients with sitosterolemia have been unclear. MATERIALS AND METHODS: We searched PubMed for studies using the keywords "sitosterolemia" or "phytosterolemia" and "Japan". Moreover, we added information from the members of the Committee on Primary Dyslipidemia under the Research Program on Rare and Intractable Disease of the Ministry of Health, Labour and Welfare (MHLW) of Japan. RESULTS: We identified 36 patients with sitosterolemia caused by biallelic pathogenic mutations in the ATP-binding cassette subfamily G member 5 (ABCG5) or ATP-binding cassette subfamily G member 8 (ABCG8) from 31 families in Japan. The diagnosed age ranged from 0 to 64 years (median 13 years). The median sitosterol and LDL cholesterol levels were 100 µg/ml (IQR: 50-183), and 193 mg/dl (IQR: 108-295), respectively. All the patients exhibited cutaneous and/or tendon xanthomas, up to 9 (25%) patients exhibited premature coronary artery disease, 5 (16%) patients exhibited arthritis, and 8 (22%) patients exhibited blood abnormalities. Ezetimibe was administered to all the patients, including infantile cases, while statins, colestimide, evolocumab, probucol, and LDL apheresis were also used. CONCLUSION: We are providing a demographic overview of the clinical and genetic backgrounds of Japanese patients with sitosterolemia.


Subject(s)
Intestinal Diseases , Lipid Metabolism, Inborn Errors , Phytosterols , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adenosine Triphosphate , Adolescent , Adult , Child , Child, Preschool , Humans , Hypercholesterolemia , Infant , Infant, Newborn , Intestinal Diseases/diagnosis , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Japan , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Middle Aged , Phytosterols/adverse effects , Phytosterols/genetics , Young Adult
14.
Liver Int ; 42(7): 1585-1592, 2022 07.
Article in English | MEDLINE | ID: mdl-35129276

ABSTRACT

INTRODUCTION: Gallstones are increasingly common in children. Genetic analyses of adult cohorts demonstrated that the sterol transporter ABCG8 p.D19H and Gilbert UGT1A1*28 variants enhance the odds of developing gallstones. The genetic background of common lithiasis in children remains unknown. METHODS: Overall, 214 children with gallstone disease (1 month-17 years, 107 boys) were inclueded. The control cohorts comprised 214 children (age 6-17 years, 115 boys) and 172 adults (age 40-92 years, 70 men) without gallstones. The ABCG8 p.D19H and UGT1A1*28 polymorphisms as well as ABCB4 (c.504C>T rs1202283, c.711A>T rs2109505) and NPC1L1 variants (p.V1296V rs217434, c.-18C>A rs41279633) were genotyped using TaqMan assays. Serum concentrations of plant sterols and cholesterol precursors were measured by gas chromatography/mass spectrometry. RESULTS: The ABCG8 risk allele was associated with an increased risk of stones (OR = 1.82, p = .03). Children carrying the p.19H allele presented with lower serum concentrations of surrogate markers of intestinal cholesterol absorption and decreased ratios of phytosterols to the cholesterol precursor desmosterol. Carriers of the common NPC1L1 rs217434 allele had an increased gallstone risk compared with stone-free adults (OR 1.90, p < .01). This variant also affected the ratio of phytosterols to cholesterol precursors (p = .03). Other tested variants were not associated with gallstone risk. CONCLUSIONS: The p.D19H ABCG8 and, to a lesser extent, NPC1L1 rs217434 variants increase the risk of early-onset gallstone formation. These results point to the presence of a common lithogenic pathway in children and adults.


Subject(s)
Gallstones , Phytosterols , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Child , Cholesterol , Gallstones/genetics , Gallstones/metabolism , Genetic Predisposition to Disease , Humans , Male , Membrane Transport Proteins/genetics , Middle Aged , Phytosterols/adverse effects , Phytosterols/genetics , Sterols/metabolism
15.
Lipids Health Dis ; 21(1): 11, 2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35042526

ABSTRACT

BACKGROUND: Sitosterolemia is a lipid disorder characterized by the accumulation of phytosterols in plasma and organs, caused by mutations in the ABCG5 and/or ABCG8 genes. The disease is frequently misdiagnosed and mistreated as familial hypercholesterolemia (FH). To gain a better understanding of the disease, the current status of diagnosis and treatment of Chinese patients with sitosterolemia was reviewed and summarized. METHOD: Literature search was performed. The clinical features and molecular characteristics of Chinese patients with sitosterolemia were analysed. Four children with sitosterolemia and the treatment experience were described. RESULTS: Fifty-five patients with sitosterolemia have been reported in China. These patients were aged from 3 months to 67 years at diagnosis, and the median was 8 years of age. Several complications, such as xanthomas in 47 patients (85%), thrombocytopenia in 17 patients (31%), anemia in 14 patients (25%), and cardiovascular damage in 12 patients (22%), were observed. Thirty-nine patients (71%) exhibited mutations in the ABCG5 gene, 15 patients (27%) showed mutations in ABCG8, and variations in both genes occurred in one patient (2%). A patient with two clinically rare diseases, namely, sitosterolemia and glycogen storage disease type VI (GSD VI)), is reported here for the first time. The four reported patients were treated with low cholesterol and phytosterol-limited diet alone or combined with cholestyramine. Even though decreases were observed for total plasma cholesterol (TC) and low-density-lipoprotein cholesterol (LDL-C), and these levels were as low as normal in some patients, the levels of plant sterols remained above the normal range. However, TC, LDL-C and plant sterol levels remained at high levels in patients treated with a control diet control only. CONCLUSIONS: The analysis reveals that different from Caucasians carrying mainly variations in ABCG8, most Chinese patients have mutations in the ABCG5 gene, and Arg446Ter, Gln251Ter, anArg389His might be hot-spot mutations in Chinese patients. The current survey provides clinical data to enable the development of a standardized protocol for the diagnosis and treatment of sitosterolemia in China.


Subject(s)
Hypercholesterolemia/diagnosis , Intestinal Diseases/diagnosis , Lipid Metabolism, Inborn Errors/diagnosis , Phytosterols/adverse effects , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , China , Female , Humans , Hypercholesterolemia/complications , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Infant , Intestinal Diseases/complications , Intestinal Diseases/genetics , Intestinal Diseases/pathology , Lipid Metabolism, Inborn Errors/complications , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Lipoproteins/genetics , Male , Middle Aged , Mutation/genetics , Phytosterols/genetics , Young Adult
16.
Nat Commun ; 13(1): 143, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013273

ABSTRACT

Phytosterol serum concentrations are under tight genetic control. The relationship between phytosterols and coronary artery disease (CAD) is controversially discussed. We perform a genome-wide meta-analysis of 32 phytosterol traits reflecting resorption, cholesterol synthesis and esterification in six studies with up to 9758 subjects and detect ten independent genome-wide significant SNPs at seven genomic loci. We confirm previously established associations at ABCG5/8 and ABO and demonstrate an extended locus heterogeneity at ABCG5/8 with different functional mechanisms. New loci comprise HMGCR, NPC1L1, PNLIPRP2, SCARB1 and APOE. Based on these results, we perform Mendelian Randomization analyses (MR) revealing a risk-increasing causal relationship of sitosterol serum concentrations and CAD, which is partly mediated by cholesterol. Here we report that phytosterols are polygenic traits. MR add evidence of both, direct and indirect causal effects of sitosterol on CAD.


Subject(s)
Cholesterol/blood , Coronary Artery Disease/genetics , Genetic Loci , Lipid Metabolism/genetics , Phytosterols/blood , ABO Blood-Group System/blood , ABO Blood-Group System/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/blood , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/blood , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Adult , Apolipoproteins E/blood , Apolipoproteins E/genetics , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Hydroxymethylglutaryl CoA Reductases/blood , Hydroxymethylglutaryl CoA Reductases/genetics , Lipase/blood , Lipase/genetics , Lipoproteins/blood , Lipoproteins/genetics , Male , Membrane Transport Proteins/blood , Membrane Transport Proteins/genetics , Mendelian Randomization Analysis , Multifactorial Inheritance , Polymorphism, Single Nucleotide , Scavenger Receptors, Class B/blood , Scavenger Receptors, Class B/genetics
17.
Int J Mol Sci ; 24(1)2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36613930

ABSTRACT

The subfamily-G ATP-binding cassette (ABCG) transporters play important roles in regulating cholesterol homeostasis. Recent progress in the structural data of ABCG1 and ABCG5/G8 disclose putative sterol binding sites that suggest the possible cholesterol translocation pathway. ABCG1 and ABCG5/G8 share high similarity in the overall molecular architecture, and both transporters appear to use several unique structural motifs to facilitate cholesterol transport along this pathway, including the phenylalanine highway and the hydrophobic valve. Interestingly, ABCG5/G8 is known to transport cholesterol and phytosterols, whereas ABCG1 seems to exclusively transport cholesterol. Ligand docking analysis indeed suggests a difference in recruiting sterol molecules to the known sterol-binding sites. Here, we further discuss how the different and shared structural features are relevant to their physiological functions, and finally provide our perspective on future studies in ABCG cholesterol transporters.


Subject(s)
Cholesterol , Lipoproteins , Lipoproteins/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Cholesterol/metabolism , Sterols/metabolism
18.
Biochem Genet ; 60(2): 822-841, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34505223

ABSTRACT

Several proteins are involved in cholesterol homeostasis, as scavenger receptor class B type I and ATP-binding cassette (ABC) transporters including ABCA1, ABCG1, ABCG5, and ABCG8. This study aimed to determine the effects of single nucleotide variants (SNVs) rs2275543 (ABCA1), rs1893590 (ABCG1), rs6720173 (ABCG5), rs6544718 (ABCG8), and rs5888 (SCARB1) on plasma lipids, lipoproteins, and adiposity markers in an asymptomatic population and its sex-specific effects. Volunteers (n = 590) were selected and plasma lipids, lipoproteins, and adiposity markers (waist-to-hip and waist-to-height ratios, lipid accumulation product and body adiposity index) were measured. Genomic DNA was isolated from peripheral blood cells according to the method adapted from Gross-Bellard. SNVs were detected in the TaqMan® OpenArray® Real-Time polymerase chain reaction platform and data analyses were performed using the TaqMan® Genotyper Software. The rs2275543*C point to an increase of high-density lipoprotein size in females while in males very-low-density lipoprotein, cholesterol, and triglycerides were statistically lower (P value < 0.05). The rs1893590*C was statistically associated with lower apolipoprotein A-I levels and higher activities of paraoxonase-1 and cholesteryl ester transfer protein (P value < 0.05). The rs6720173 was statistically associated with an increase in cholesterol and low-density lipoprotein cholesterol in males; moreover, rs6544718*T reduced adiposity markers in females (P value < 0.05). Regarding the rs5888, a decreased adiposity marker in the total population and in females occurred (P value < 0.05). Multivariate analysis of variance showed that SNVs could influence components of high-density lipoprotein metabolism, mainly through ABCG1 (P value < 0.05). The ABCA1 and ABCG5 variants showed sex-specific effects on lipids and lipoproteins, while SCARB1 and ABCG8 variants might influence adiposity markers in females. Our data indicate a possible role of ABCG1 on HDL metabolism.


Subject(s)
Adiposity , Lipoproteins , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 5/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adiposity/genetics , Cholesterol/metabolism , Female , Humans , Lipoproteins/genetics , Lipoproteins/metabolism , Lipoproteins, HDL/genetics , Male , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
19.
Hepatology ; 75(2): 252-265, 2022 02.
Article in English | MEDLINE | ID: mdl-34387888

ABSTRACT

BACKGROUND AND AIMS: Parenteral nutrition (PN)-associated cholestasis (PNAC) complicates the care of patients with intestinal failure. In PNAC, phytosterol containing PN synergizes with intestinal injury and IL-1ß derived from activated hepatic macrophages to suppress hepatocyte farnesoid X receptor (FXR) signaling and promote PNAC. We hypothesized that pharmacological activation of FXR would prevent PNAC in a mouse model. APPROACH AND RESULTS: To induce PNAC, male C57BL/6 mice were subjected to intestinal injury (2% dextran sulfate sodium [DSS] for 4 days) followed by central venous catheterization and 14-day infusion of PN with or without the FXR agonist GW4064. Following sacrifice, hepatocellular injury, inflammation, and biliary and sterol transporter expression were determined. GW4064 (30 mg/kg/day) added to PN on days 4-14 prevented hepatic injury and cholestasis; reversed the suppressed mRNA expression of nuclear receptor subfamily 1, group H, member 4 (Nr1h4)/FXR, ATP-binding cassette subfamily B member 11 (Abcb11)/bile salt export pump, ATP-binding cassette subfamily C member 2 (Abcc2), ATP binding cassette subfamily B member 4(Abcb4), and ATP-binding cassette subfamily G members 5/8(Abcg5/8); and normalized serum bile acids. Chromatin immunoprecipitation of liver showed that GW4064 increased FXR binding to the Abcb11 promoter. Furthermore, GW4064 prevented DSS-PN-induced hepatic macrophage accumulation, hepatic expression of genes associated with macrophage recruitment and activation (ll-1b, C-C motif chemokine receptor 2, integrin subunit alpha M, lymphocyte antigen 6 complex locus C), and hepatic macrophage cytokine transcription in response to lipopolysaccharide in vitro. In primary mouse hepatocytes, GW4064 activated transcription of FXR canonical targets, irrespective of IL-1ß exposure. Intestinal inflammation and ileal mRNAs (Nr1h4, Fgf15, and organic solute transporter alpha) were not different among groups, supporting a liver-specific effect of GW4064 in this model. CONCLUSIONS: GW4064 prevents PNAC in mice through restoration of hepatic FXR signaling, resulting in increased expression of canalicular bile and of sterol and phospholipid transporters and suppression of macrophage recruitment and activation. These data support augmenting FXR activity as a therapeutic strategy to alleviate or prevent PNAC.


Subject(s)
Cholestasis/prevention & control , Gene Expression/drug effects , Isoxazoles/pharmacology , Parenteral Nutrition/adverse effects , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Animals , Bile Acids and Salts/blood , Cholestasis/etiology , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Interleukin-1beta/pharmacology , Intestinal Diseases/chemically induced , Intestinal Diseases/therapy , Isoxazoles/therapeutic use , Lipoproteins/genetics , Liver Diseases/etiology , Liver Diseases/pathology , Liver Diseases/prevention & control , Macrophage Activation/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Multidrug Resistance-Associated Protein 2/genetics , Multidrug Resistance-Associated Proteins/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction/drug effects
20.
J Steroid Biochem Mol Biol ; 216: 106039, 2022 02.
Article in English | MEDLINE | ID: mdl-34861389

ABSTRACT

We evaluated oxyphytosterol (OPS) concentrations in plasma and various tissues of two genetically modified mouse models with either increased cholesterol (apoE KO mice) or increased cholesterol and plant sterol (PS) concentrations (apoExABCG8 dKO mice). Sixteen female apoE KO and 16 dKO mice followed the same standard, low OPS-chow diet. Animals were euthanized at 36 weeks to measure PS and OPS concentrations in plasma, brain, liver and aortic tissue. Cholesterol and oxysterol (OS) concentrations were analyzed as reference for sterol oxidation in general. Plasma campesterol (24.1 ± 4.3 vs. 11.8 ± 3.0 mg/dL) and sitosterol (67.4 ± 12.7 vs. 4.9 ± 1.1 mg/dL) concentrations were severely elevated in the dKO compared to the apoE KO mice (p < 0.001). Also, in aortic and brain tissue, PS levels were significantly elevated in dKO. However, plasma, aortic and brain OPS concentrations were comparable or even lower in the dKO mice. In contrast, in liver tissue, both PS and OPS concentrations were severely elevated in the dKO compared to apoE KO mice (sum OPS: 7.4 ± 1.6 vs. 4.1 ± 0.8 ng/mg, p < 0.001). OS concentrations followed cholesterol concentrations in plasma and all tissues suggesting ubiquitous oxidation. Despite severely elevated PS concentrations, OPS concentrations were only elevated in liver tissue, suggesting that OPS are primarily formed in the liver and plasma concentrations originate from hepatic spill-over into the circulation.


Subject(s)
Liver/metabolism , Oxysterols/blood , Phytosterols/blood , ATP Binding Cassette Transporter, Subfamily G, Member 8/genetics , Animals , Apolipoproteins E/genetics , Cholesterol/analogs & derivatives , Cholesterol/blood , Cholesterol/metabolism , Female , Lipid Metabolism/genetics , Lipoproteins/genetics , Mice , Mice, Knockout , Oxidation-Reduction , Oxysterols/metabolism , Phytosterols/metabolism , Sitosterols/blood , Sitosterols/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...