Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Intern Med ; 292(2): 296-307, 2022 08.
Article in English | MEDLINE | ID: mdl-34982494

ABSTRACT

BACKGROUND: Sterol O-acyltransferase 2 (Soat2) encodes acyl-coenzyme A:cholesterol acyltransferase 2 (ACAT2), which synthesizes cholesteryl esters in hepatocytes and enterocytes fated either to storage or to secretion into nascent triglyceride-rich lipoproteins. OBJECTIVES: We aimed to unravel the molecular mechanisms leading to reduced hepatic steatosis when Soat2 is depleted in mice. METHODS: Soat2-/- and wild-type mice were fed a high-fat, a high-carbohydrate, or a chow diet, and parameters of lipid and glucose metabolism were assessed. RESULTS: Glucose, insulin, homeostatic model assessment for insulin resistance (HOMA-IR), oral glucose tolerance (OGTT), and insulin tolerance tests significantly improved in Soat2-/- mice, irrespective of the dietary regimes (2-way ANOVA). The significant positive correlations between area under the curve (AUC) OGTT (r = 0.66, p < 0.05), serum fasting insulin (r = 0.86, p < 0.05), HOMA-IR (r = 0.86, p < 0.05), Adipo-IR (0.87, p < 0.05), hepatic triglycerides (TGs) (r = 0.89, p < 0.05), very-low-density lipoprotein (VLDL)-TG (r = 0.87, p < 0.05) and the hepatic cholesteryl esters in wild-type mice disappeared in Soat2-/- mice. Genetic depletion of Soat2 also increased whole-body oxidation by 30% (p < 0.05) compared to wild-type mice. CONCLUSION: Our data demonstrate that ACAT2-generated cholesteryl esters negatively affect the metabolic control by retaining TG in the liver and that genetic inhibition of Soat2 improves liver steatosis via partitioning of lipids into secretory (VLDL-TG) and oxidative (fatty acids) pathways.


Subject(s)
Fatty Liver , Insulins , Sterol O-Acyltransferase , Animals , Cholesterol Esters/metabolism , Fatty Liver/metabolism , Glucose/metabolism , Insulins/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Triglycerides , Sterol O-Acyltransferase 2
2.
BMC Cancer ; 21(1): 544, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33985458

ABSTRACT

BACKGROUND: One reason for the often late diagnosis of lung cancer (LC) may be that potentially-indicative sensations and symptoms are often diffuse, and may not be considered serious or urgent, making their interpretation complicated. However, with only a few exceptions, efforts to use people's own in-depth knowledge about prodromal bodily experiences has been a missing link in efforts to facilitate early LC diagnosis. In this study, we describe and discuss facilitators and challenges in our process of developing and initial testing an interactive, self-completion e-questionnaire based on patient descriptions of experienced prodromal sensations and symptoms, to support early identification of lung cancer (LC). METHODS: E-questionnaire items were derived from in-depth, detailed explorative interviews with individuals undergoing investigation for suspected LC. The descriptors of sensations/symptoms and the background items obtained were the basis for developing an interactive, individualized instrument, PEX-LC, which was refined for usability through think-aloud and other interviews with patients, members of the public, and clinical staff. RESULTS: Major challenges in the process of developing PEX-LC related to collaboration among many actors, and design/user interface problems including technical issues. Most problems identified through the think-aloud interviews related to design/user interface problems and technical issues rather than content, for example we re-ordered questions to be in line with patients' chronological, rather than retrospective, descriptions of their experiences. PEX-LC was developed into a final e-questionnaire on a touch-screen smart tablet with one background module covering sociodemographic characteristics, 10 interactive, individualized modules covering early sensations and symptoms, and a 12th assessing current symptoms. CONCLUSIONS: Close collaboration with patients throughout the process was intrinsic for developing PEX-LC. Similarly, we recognized the extent to which clinicians and technical experts were also important in this process. Similar endeavors should assure all necessary competence is included in the core research team, to facilitate timely progress. Our experiences developing PEX-LC combined with new empirical research suggest that this individualized, interactive e-questionnaire, developed through systematizing patients' own formulations of their prodromal symptom experiences, is both feasible for use and has potential value in the intended group.


Subject(s)
Early Detection of Cancer/methods , Lung Neoplasms/diagnosis , Surveys and Questionnaires , Humans , Internet , Intersectoral Collaboration , Knowledge , User-Computer Interface
3.
Article in English | MEDLINE | ID: mdl-32058035

ABSTRACT

In contrast to human hepatocytes in vivo, which solely express acyl-coenzyme A:cholesterol acyltransferase (ACAT) 2, both ACAT1 and ACAT2 (encoded by SOAT1 and SOAT2) are expressed in primary human hepatocytes and in human hepatoma cell lines. Here, we aimed to create hepatocyte-like cells expressing the ACAT2, but not the ACAT1, protein to generate a model that - at least in this regard - resembles the human condition in vivo and to assess the effects on lipid metabolism. Using the Clustered Regularly Interspaced Short Palindromic Repeats technology, we knocked out SOAT1 in HepG2 and Huh7.5 cells. The wild type and SOAT2-only-cells were cultured with fetal bovine or human serum and the effects on lipoprotein and lipid metabolism were studied. In SOAT2-only-HepG2 cells, increased levels of cholesterol, triglycerides, apolipoprotein B and lipoprotein(a) in the cell media were detected; this was likely dependent of the increased expression of key genes involved in lipid metabolism (e.g. MTP, APOB, HMGCR, LDLR, ACACA, and DGAT2). Opposite effects were observed in SOAT2-only-Huh7.5 cells. Our study shows that the expression of SOAT1 in hepatocyte-like cells contributes to the distorted phenotype observed in HepG2 and Huh7.5 cells. As not only parameters of lipoprotein and lipid metabolism but also some markers of differentiation/maturation increase in the SOAT2-only-HepG2 cells cultured with HS, this cellular model represent an improved model for studies of lipid metabolism.


Subject(s)
Hepatocytes/enzymology , Lipid Metabolism/physiology , Lipoproteins/metabolism , Sterol O-Acyltransferase/genetics , Sterol O-Acyltransferase/metabolism , Cell Culture Techniques , Cell Differentiation/physiology , Gene Knockdown Techniques , Hep G2 Cells , Humans , Sterol O-Acyltransferase 2
4.
Sci Rep ; 9(1): 16504, 2019 11 11.
Article in English | MEDLINE | ID: mdl-31712735

ABSTRACT

The aim of this study was to identify a combination of early predictive symptoms/sensations attributable to primary lung cancer (LC). An interactive e-questionnaire comprised of pre-diagnostic descriptors of first symptoms/sensations was administered to patients referred for suspected LC. Respondents were included in the present analysis only if they later received a primary LC diagnosis or had no cancer; and inclusion of each descriptor required ≥4 observations. Fully-completed data from 506/670 individuals later diagnosed with primary LC (n = 311) or no cancer (n = 195) were modelled with orthogonal projections to latent structures (OPLS). After analysing 145/285 descriptors, meeting inclusion criteria, through randomised seven-fold cross-validation (six-fold training set: n = 433; test set: n = 73), 63 provided best LC prediction. The most-significant LC-positive descriptors included a cough that varied over the day, back pain/aches/discomfort, early satiety, appetite loss, and having less strength. Upon combining the descriptors with the background variables current smoking, a cold/flu or pneumonia within the past two years, female sex, older age, a history of COPD (positive LC-association); antibiotics within the past two years, and a history of pneumonia (negative LC-association); the resulting 70-variable model had accurate cross-validated test set performance: area under the ROC curve = 0.767 (descriptors only: 0.736/background predictors only: 0.652), sensitivity = 84.8% (73.9/76.1%, respectively), specificity = 55.6% (66.7/51.9%, respectively). In conclusion, accurate prediction of LC was found through 63 early symptoms/sensations and seven background factors. Further research and precision in this model may lead to a tool for referral and LC diagnostic decision-making.


Subject(s)
Lung Neoplasms/diagnosis , Machine Learning , Sensation , Symptom Assessment , Aged , Female , Humans , Male , Middle Aged , Models, Theoretical , Prognosis , ROC Curve , Reproducibility of Results
6.
J Steroid Biochem Mol Biol ; 169: 10-21, 2017 05.
Article in English | MEDLINE | ID: mdl-26851362

ABSTRACT

27-Hydroxycholesterol (27OH) is a strong suppressor of cholesterol synthesis and a weak activator of LXR in vitro. The regulatory importance of 27OH in vivo is controversial. Here we utilized male mice with increased levels of 27OH either due to increased production (CYP27A1 transgenic mice) or reduced metabolism (Cyp7b1-/- mice). We also used mice lacking 27OH due to a knockout of Cyp27a1. The latter mice were treated with cholic acid to compensate for reduced bile acid synthesis. The effects of the different levels of 27OH on Srebp- and other LXR-regulated genes in the liver were investigated. In the liver of CYP27tg mice we found a modest increase of the mRNA levels corresponding to the LXR target genes Cyp7b1 and Abca1. A number of other LXR-regulated genes were not affected. The effect on Abca1 mRNA was not seen in the liver of Cyp7b1-/- mice. There were little or no effects on cholesterol synthesis. In the liver of the Cyp27-/- mice treated with 0.025% cholic acid there was no significant effect of the knockout on the LXR target genes. In a previous work triple-knockout mice deficient in the biosynthesis of 24S-hydroxycholesterol, 25-hydroxycholesterol and 27OH were shown to have impaired response to dietary cholesterol, suggesting side-chain oxidized oxysterols to be mediators in cholesterol-induced effects on LXR target genes at a transcriptional level (Chen W. et al., Cell Metab. 5 (2007) 73-79). The hydroxylated oxysterol responsible for the effect was not defined. We show here that treatment of wildtype mice with dietary cholesterol under the same conditions as in the above study induced the LXR target genes Lpl, Abcg8 and Srebp1c in wild type mice but failed to activate the same genes in mice lacking 27-hydroxycholesterol due to a knockout of Cyp27. We failed to demonstrate the above effects at the protein level (Abcg8) or at the activity level (Lpl). The results suggest that 27OH is not an important regulator of Srebp- or LXR regulated genes under basal conditions in mouse liver. On the other hand 27OH appears to mediate cholesterol-induced effects on some LXR target genes at a transcriptional level under some in vivo conditions.


Subject(s)
Hydroxycholesterols/metabolism , Liver/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 8/metabolism , Animals , Cholestanetriol 26-Monooxygenase/genetics , Cytochrome P450 Family 7/genetics , Gene Expression Profiling , Lipoprotein Lipase/metabolism , Lipoproteins/metabolism , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cytoplasmic and Nuclear/metabolism , Steroid Hydroxylases/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription, Genetic
7.
J Biol Chem ; 289(34): 23712-22, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24973215

ABSTRACT

The presence of the blood-brain barrier (BBB) is critical for cholesterol metabolism in the brain, preventing uptake of lipoprotein-bound cholesterol from the circulation. The metabolic consequences of a leaking BBB for cholesterol metabolism have not been studied previously. Here we used a pericyte-deficient mouse model, Pdgfb(ret/ret), shown to have increased permeability of the BBB to a range of low-molecular mass and high-molecular mass tracers. There was a significant accumulation of plant sterols in the brains of the Pdgfb(ret/ret) mice. By dietary treatment with 0.3% deuterium-labeled cholesterol, we could demonstrate a significant flux of cholesterol from the circulation into the brains of the mutant mice roughly corresponding to about half of the measured turnover of cholesterol in the brain. We expected the cholesterol flux into the brain to cause a down-regulation of cholesterol synthesis. Instead, cholesterol synthesis was increased by about 60%. The levels of 24(S)-hydroxycholesterol (24S-OHC) were significantly reduced in the brains of the pericyte-deficient mice but increased in the circulation. After treatment with 1% cholesterol in diet, the difference in cholesterol synthesis between mutants and controls disappeared. The findings are consistent with increased leakage of 24S-OHC from the brain into the circulation in the pericyte-deficient mice. This oxysterol is an efficient suppressor of cholesterol synthesis, and the results are consistent with a regulatory role of 24S-OHC in the brain. To our knowledge, this is the first demonstration that a defective BBB may lead to increased flux of a lipophilic compound out from the brain. The relevance of the findings for the human situation is discussed.


Subject(s)
Blood-Brain Barrier , Brain/metabolism , Cholesterol/metabolism , Homeostasis , Animals , Base Sequence , Cholesterol/biosynthesis , DNA Primers , Genes, sis , Homeostasis/genetics , Mice , Mice, Transgenic , Plants/metabolism , Real-Time Polymerase Chain Reaction , Sterols/metabolism
8.
PLoS One ; 8(7): e68534, 2013.
Article in English | MEDLINE | ID: mdl-23874659

ABSTRACT

We previously described a heterozygous mouse model overexpressing human HA-tagged 24S-hydroxylase (CYP46A1) utilizing a ubiquitous expression vector. In this study, we generated homozygotes of these mice with circulating levels of 24OH 30-60% higher than the heterozygotes. Female homozygous CYP46A1 transgenic mice, aged 15 months, showed an improvement in spatial memory in the Morris water maze test as compared to the wild type mice. The levels of N-Methyl-D-Aspartate receptor 1, phosphorylated-N-Methyl-D-Aspartate receptor 2A, postsynaptic density 95, synapsin-1 and synapthophysin were significantly increased in the hippocampus of the CYP46A1 transgenic mice as compared to the controls. The levels of lanosterol in the brain of the CYP46A1 transgenic mice were significantly increased, consistent with a higher synthesis of cholesterol. Our results are discussed in relation to the hypothesis that the flux in the mevalonate pathway in the brain is of importance in cognitive functions.


Subject(s)
Brain/enzymology , Brain/physiology , Memory/physiology , Steroid Hydroxylases/metabolism , Animals , Cholesterol 24-Hydroxylase , Female , Humans , Mice , Mice, Transgenic , Steroid Hydroxylases/genetics
9.
PLoS One ; 8(7): e68031, 2013.
Article in English | MEDLINE | ID: mdl-23844150

ABSTRACT

24S- and 27-hydroxycholesterol (24OHC and 27OHC) are potent regulators of different biochemical systems in vitro and are the major circulating oxysterols. A small fraction of these oxysterols has been reported to be sulphated but there are no detailed studies. We considered the possibility that sulphatation is a protective mechanism preventing accumulation of free oxysterols. Using an accurate assay we found the sulphated fraction of 24OHC and 27OHC in circulation of adults to be less than 15% of total. In two patients with a mutation in CYP7B1 and markedly increased levels of 27OHC the sulphated fraction was 8% and 10% respectively. Infants with severe neonatal cholestasis had however markedly increased sulphate fraction of the above oxysterols. In untreated mice the degree of sulphatation of 24OHC and 27OHC in serum varied between 0 and 16%. Similar degree of sulphatation was found in two mouse models with markedly increased levels of 27OHC and 24OHC respectively. Bile duct ligated mice had higher levels of oxysterols than sham-operated controls but the sulphate fraction was not increased. We conclude that a primary increase in the levels of the oxysterols due to increased synthesis or reduced metabolism in adults and mice does not induce increased sulphatation.


Subject(s)
Cholesterol/metabolism , Adult , Aged , Animals , Cholesterol/blood , Female , Humans , Hydroxycholesterols/blood , Male , Mice , Middle Aged , Oxidation-Reduction
10.
J Lipid Res ; 54(4): 1033-43, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23284090

ABSTRACT

The two oxysterols, 27-hydroxycholesterol (27OH) and 24S-hydroxycholesterol (24OH), are both inhibitors of cholesterol synthesis and activators of the liver X receptor (LXR) in vitro. Their role as physiological regulators under in vivo conditions is controversial, however. In the present work, we utilized a previously described mouse model with overexpressed human sterol 27-hydroxylase (CYP27A1). The levels of 27OH were increased about 12-fold in the brain. The brain levels of HMG-CoA reductase mRNA and HMG-CoA synthase mRNA levels were increased. In accordance with increased cholesterol synthesis, most of the cholesterol precursors were also increased. The level of 24OH, the dominating oxysterol in the brain, was decreased by about 25%, most probably due to increased metabolism by CYP27A1. The LXR target genes were unaffected or slightly changed in a direction opposite to that expected for LXR activation. In the brain of Cyp27(-/-) mice, cholesterol synthesis was slightly increased, with increased levels of cholesterol precursors but normal mRNA levels of HMG-CoA reductase and HMG-CoA synthase. The mRNA levels corresponding to LXR target genes were not affected. The results are consistent with the possibility that both 24OH and 27OH are physiological suppressors of cholesterol synthesis in the brain. The results do not support the contention that 27OH is a general activator of LXR target genes in this organ.


Subject(s)
Cholestanetriol 26-Monooxygenase/metabolism , Hydroxycholesterols/metabolism , Animals , Brain , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/metabolism , Female , Humans , Hydroxymethylglutaryl-CoA Synthase/metabolism , Liver X Receptors , Male , Mice , Mice, Knockout , Mice, Transgenic , Orphan Nuclear Receptors/metabolism
11.
Neurochem Int ; 60(2): 192-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22185844

ABSTRACT

Cholesterol metabolism in the brain is distinct from that in other tissues due to the fact that cholesterol itself is unable to pass across the blood-brain barrier. Elimination of brain cholesterol is mainly dependent on a neuronal-specific cytochrome P450, CYP46A1, catalyzing the conversion of cholesterol into 24(S)-hydroxycholesterol (24OHC), which is able to pass the blood-brain barrier. A suitable model for studying this elimination from human neuronal cells has not been described previously. It is shown here that differentiated Ntera2/clone D1 (NT2) cells express the key genes involved in brain cholesterol homeostasis including CYP46A1, and that the expression profiles of the genes observed during neuronal differentiation are those expected to occur in vivo. Thus there was a decrease in the mRNA levels corresponding to cholesterol synthesis enzymes and a marked increase in the mRNA level of CYP46A1. The latter increase was associated with increased levels of CYP46A1 protein and increased production of 24OHC. The magnitude of the secretion of 24OHC from the differentiated NT2 cells into the medium was similar to that expected to occur under in vivo conditions. An alternative to elimination of cholesterol by the CYP46A1 mechanism is elimination by CYP27A1, and the product of this enzyme, 27-hydroxycholesterol (27OHC), is also known to pass the blood-brain barrier. The CYP27A1 protein level decreased during the differentiation of the NT2 cells in parallel with decreased production of 27OHC. The ratio between 24OHC and 27OHC in the medium from the cultured cells increased, by a factor of 13, during the differentiation process. The results suggest that progenitor cells eliminate cholesterol in the form of 27OHC while neurogenesis induces a change to the CYP46A1 dependent pathway. Furthermore this study demonstrates that differentiated NT2 cells are suitable for studies of cholesterol homeostasis in human neurons.


Subject(s)
Cell Differentiation/physiology , Cholestanetriol 26-Monooxygenase/physiology , Neurogenesis/physiology , Steroid Hydroxylases/physiology , Cell Differentiation/genetics , Cell Line, Tumor , Cholestanetriol 26-Monooxygenase/biosynthesis , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol 24-Hydroxylase , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Homeostasis/genetics , Humans , Neurogenesis/genetics , RNA, Messenger/biosynthesis , Steroid Hydroxylases/biosynthesis , Steroid Hydroxylases/genetics
12.
J Lipid Res ; 52(5): 1004-10, 2011 May.
Article in English | MEDLINE | ID: mdl-21335619

ABSTRACT

There is a significant flux of the neurotoxic oxysterol 27-hydroxycholesterol (27OHC) from the circulation across the blood-brain barrier. Because there is a correlation between 27OHC and cholesterol in the circulation and lipoprotein-bound cholesterol does not pass the blood-brain barrier, we have suggested that 27OHC may mediate the effects of hypercholesterolemia on the brain. We previously demonstrated a modest accumulation of 27OHC in brains of patients with sporadic Alzheimer's disease (AD), consistent with a role of 27OHC as a primary pathogenetic factor. We show here that there is a 4-fold accumulation of 27OHC in different regions of the cortexes of patients carrying the Swedish amyloid precursor protein (APPswe) 670/671 mutation. The brain levels of sitosterol and campesterol were not significantly different in the AD patients compared with the controls, suggesting that the blood-brain barrier was intact in the AD patients. We conclude that accumulation of 27OHC is likely to be secondary to neurodegeneration, possibly a result of reduced activity of CYP7B1, the neuronal enzyme responsible for metabolism of 27OHC. We discuss the possibility of a vicious circle in the brains of the patients with familial AD whereby neurodegenerative changes cause an accumulation of 27OHC that further accelerates neurodegeneration.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Brain/metabolism , Hydroxycholesterols/metabolism , Aged , Alzheimer Disease/genetics , Blotting, Western , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Female , Humans , Male , Middle Aged , Mutation , Phytosterols/metabolism , Sitosterols/metabolism
13.
J Lipid Res ; 51(9): 2722-30, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20511491

ABSTRACT

The rare disease cerebrotendinous xanthomatosis (CTX) is due to a lack of sterol 27-hydroxylase (CYP27A1) and is characterized by cholestanol-containing xanthomas in brain and tendons. Mice with the same defect do not develop xanthomas. The driving force in the development of the xanthomas is likely to be conversion of a bile acid precursor into cholestanol. The mechanism behind the xanthomas in the brain has not been clarified. We demonstrate here that female cyp27a1(-/-) mice have an increase of cholestanol of about 2.5- fold in plasma, 6-fold in tendons, and 12-fold in brain. Treatment of cyp27a1(-/-) mice with 0.05% cholic acid normalized the cholestanol levels in tendons and plasma and reduced the content in the brain. The above changes occurred in parallel with changes in plasma levels of 7alpha-hydroxy-4-cholesten-3-one, a precursor both to bile acids and cholestanol. Injection of a cyp27a1(-/-) mouse with (2)H(7)-labeled 7alpha-hydroxy-4-cholesten-3-one resulted in a significant incorporation of (2)H(7)-cholestanol in the brain. The results are consistent with a concentration-dependent flux of 7alpha-hydroxy-4-cholesten-3-one across the blood-brain barrier in cyp27a1(-/-) mice and subsequent formation of cholestanol. It is suggested that the same mechanism is responsible for accumulation of cholestanol in the brain of patients with CTX.


Subject(s)
Brain/metabolism , Cholestanetriol 26-Monooxygenase/metabolism , Cholestanol/metabolism , Animals , Brain Chemistry , Cholestanetriol 26-Monooxygenase/genetics , Cholestanol/chemistry , Cholestenones/metabolism , Cholestyramine Resin , Cholic Acid/metabolism , Female , Humans , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Knockout , Molecular Structure , Tendons/chemistry , Tendons/metabolism , Xanthomatosis, Cerebrotendinous/enzymology , Xanthomatosis, Cerebrotendinous/pathology
14.
Brain Res ; 1344: 185-91, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20450896

ABSTRACT

Variations in genes associated with cholesterol homeostasis have been reported to modify the risk of developing Alzheimer disease (AD). To date there have been few investigations into variations in genes directly involved in cholesterol biosynthesis and AD. We investigated the influence of the -911C>A polymorphism (rs3761740) in the hydroxy-methyl-glutaryl CoA reductase (HMGCR) gene promoter on basal and regulated transcription, plasma cholesterol levels and the association with AD. Under in vitro conditions the A allele was found to be significantly more responsive to SREBP-2 mediated regulation than the C allele. In an age and sex matched case-control study, the genotype distribution and allele frequency of this polymorphism were not associated with AD (OR=1.03; 95% CI=0.72-1.48). However, we did find evidence supporting an interaction between the HMGCR A allele, the APOE E4 allele and an altered risk of AD (OR=2.41; 95% CI=0.93-6.22).


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Hydroxymethylglutaryl CoA Reductases/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Aged , Aged, 80 and over , Alzheimer Disease/epidemiology , Apolipoprotein E4/genetics , Case-Control Studies , Cell Line, Transformed , Cholesterol/metabolism , Female , Gene Frequency , Genotype , Humans , Male , Risk Factors , Sweden/epidemiology , Transfection/methods
15.
Biochim Biophys Acta ; 1801(6): 674-81, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20298807

ABSTRACT

Two diets simulating the recommendations of the American Heart Association to increase the intake of n-3 polyunsaturated fatty acids (n-3 PUFAs) were tested on Golden Syrian hamsters and compared to the diet simulating the current estimated consumption of fat in the United States. N-3 PUFAs were evaluated for their effects on serum and brain lipids and on the three cytochrome P450 enzymes (CYPs 7A1, 27A1, and 46A1) that play key roles in cholesterol elimination from different organs. Hamsters on the highest concentration of n-3 PUFAs had a statistically significant decrease in LDL and HDL cholesterol and no change in serum total cholesterol and triglycerides levels. CYP27A1 and CYP46A1 mRNA levels were increased in the liver and brain, respectively, whereas possible effects on CYP7A1 were obscured by a marked intergroup variability at mRNA, protein, and sterol product levels. Increased levels of CYP46A1 mRNA in the brain did not lead to significant changes in the levels of lathosterol, 24S-hydroxycholesterol or cholesterol in this organ. The data obtained are discussed in relation to inconsistent effects of n-3 PUFAs on serum lipids in human trials and reported positive effects of fish oil on cognitive function.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fatty Acids, Omega-3/pharmacology , Isoenzymes/metabolism , Liver/enzymology , Microsomes, Liver/enzymology , Animals , Base Sequence , Cloning, Molecular , Cricetinae , Cytochrome P-450 Enzyme System/genetics , DNA Primers , Gas Chromatography-Mass Spectrometry , Isoenzymes/genetics , Male , Mesocricetus
16.
Arterioscler Thromb Vasc Biol ; 27(10): 2191-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17761942

ABSTRACT

OBJECTIVE: Characterization of cholesterol homeostasis in male mice with a genetic inactivation of 3beta-hydroxysteroid-delta24-reductase, causing replacement of almost all cholesterol with desmosterol. METHODS AND RESULTS: There was an increase in hepatic sterol synthesis and markedly increased fecal loss of neutral sterols. Fecal excretion of bile acids was similar in knockout mice and in controls. The composition of bile acids was changed, with reduced formation of cholic acid. It was shown that both Cyp7a1 and Cyp27a1 are active toward desmosterol, consistent with the formation of normal bile acids from this steroid. The levels of plant sterols were markedly reduced. Hepatic mRNA levels of 3-hydroxy-3-methylglutaryl (HMG) coenzyme A (CoA) reductase, Srebp-1c, Srebp-2, Cyp7a1, Abcg5, Abcg8, and Fas were all significantly increased. CONCLUSIONS: The changes in hepatic mRNA levels in combination with increased biliary and fecal excretion of neutral steroids, reduced tissue levels of plant sterols, increased plasma levels of triglyceride-rich VLDL, are consistent with a strong activation of LXR-targeted genes. The markedly increased fecal loss of neutral sterols may explain the fact that the Dhcr24-/- mice do not accumulate dietary cholesterol. The study illustrates the importance of the integrity of the cholesterol structure--presence of a double bond in the steroid side-chain is compatible with life but is associated with serious disturbances in sterol homeostasis.


Subject(s)
Cholesterol/metabolism , DNA-Binding Proteins/metabolism , Desmosterol/metabolism , Gene Expression Regulation , Lipid Metabolism , Liver/metabolism , Nerve Tissue Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Bile/metabolism , Bile Acids and Salts/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholestanetriol 26-Monooxygenase/metabolism , Cholesterol/analogs & derivatives , Cholesterol/blood , Cholesterol/deficiency , Cholesterol 7-alpha-Hydroxylase/genetics , Cholesterol 7-alpha-Hydroxylase/metabolism , DNA-Binding Proteins/genetics , Desmosterol/blood , Feces/chemistry , Homeostasis , Hydroxymethylglutaryl CoA Reductases/genetics , Hydroxymethylglutaryl CoA Reductases/metabolism , Lipid Metabolism/genetics , Lipids/blood , Lipoproteins/blood , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/enzymology , Liver X Receptors , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Orphan Nuclear Receptors , Oxidoreductases Acting on CH-CH Group Donors/genetics , Phytosterols/blood , Phytosterols/metabolism , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Substrate Specificity , Time Factors
17.
Saudi Med J ; 28(7): 1113-8, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17603722

ABSTRACT

A Saudi Arabian family is described in which there were 2 siblings with typical features of cerebral xanthomatosis CTX including premature cataracts, xanthomata of the Achilles tendons, neuro-psychiatric disturbances, and atherosclerosis. The 2 patients were homozygous for a point mutation in the mitochondrial 27-hydroxylase gene CYP27A1, OMIM 606530 located in the splice site of intron 6, where G was exchanged for A IVS6+1G>A. Their parents were cousins, 5 siblings were healthy, 2 were heterozygous for the mutation, and one showed the wild-type genotype. The father was heterozygous for the mutation, while the other family members were not tested. The progress of the 2 CTX patients over 14 years is described; firstly when they were receiving treatment with chenodeoxycholic acid; when this medication was not available, and later when it was restored. A hereditary hyperlipidemia was also present in this family. It is suggested that when this occurs with CTX, a more serious illness results that merits more aggressive dual therapy.


Subject(s)
Xanthomatosis, Cerebrotendinous/genetics , Adolescent , Adult , Child , Child, Preschool , Cholestanetriol 26-Monooxygenase/genetics , Female , Follow-Up Studies , Genotype , Humans , Male , Saudi Arabia
18.
Histochem Cell Biol ; 127(3): 253-61, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17237956

ABSTRACT

The synthesis of primary bile acids is confined to the hepatocytes. This study aimed to evaluate the expression pattern within the liver architecture of the rate-limiting enzyme of the neutral pathway, cholesterol 7alpha-hydroxylase (Cyp7a1), and sterol 12alpha-hydroxylase (Cyp8b1), the enzyme necessary for the synthesis of cholic acid. Specific Cyp8b1 and Cyp7a1 peptide antiserums were used for immunohistochemical staining of livers from wild type and Cyp8b1 null mice, the latter instead expressing beta-galactosidase (beta-Gal) as a replacement reporter gene. Cyp8b1 was mainly expressed in the hepatocytes in a zonal pattern surrounding the central vein while the areas surrounding the portal zones showed much lower levels. The zonation was maintained in cholic acid-depleted mice using beta-Gal as a reporter protein. Cyp7a1 expression in wild type mice also showed a zonal distribution pattern, although less distinct, with a maximal expression within a 1-2 cell thick layer of hepatocytes surrounding the central vein. In Cyp8b1 null mice, a more intense staining was obtained, in accordance with the higher expression level of Cyp7a1, although the overall expression pattern was maintained. Our results in mice indicate possible differences in the regulation of the cellular zonation of Cyp7a1 and Cyp8b1. Also, cholic acid affects the set-point of Cyp7a1 expression but not its zonal distribution.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Steroid 12-alpha-Hydroxylase/metabolism , Animals , Cholesterol 7-alpha-Hydroxylase/genetics , Gene Expression Regulation, Enzymologic , Genes, Reporter , Immunohistochemistry , Male , Mice , Mice, Knockout , Steroid 12-alpha-Hydroxylase/genetics , beta-Galactosidase/metabolism
19.
J Biol Chem ; 281(7): 3810-20, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16321981

ABSTRACT

Mammalian CNS contains a disproportionally large and remarkably stable pool of cholesterol. Despite an efficient recycling there is some requirement for elimination of brain cholesterol. Conversion of cholesterol into 24S-hydroxycholesterol by the cholesterol 24-hydroxylase (CYP46A1) is the quantitatively most important mechanism. Based on the protein expression and plasma levels of 24S-hydroxycholesterol, CYP46A1 activity appears to be highly stable in adults. Here we have made a structural and functional characterization of the promoter of the human CYP46A1 gene. No canonical TATA or CAAT boxes were found in the promoter region. Moreover this region had a high GC content, a feature often found in genes considered to have a largely housekeeping function. A broad spectrum of regulatory axes using a variety of promoter constructs did not result in a significant transcriptional regulation. Oxidative stress caused a significant increase in transcriptional activity. The possibility of a substrate-dependent transcriptional regulation was explored in vivo in a sterol-deficient mouse model (Dhcr24 null) in which almost all cholesterol had been replaced with desmosterol, which is not a substrate for CYP46A1. Compared with heterozygous littermates there was no statistically significant difference in the mRNA levels of Cyp46a1. During the first 2 weeks of life in the wild-type mouse, however, a significant increase of Cyp46a1 mRNA levels was found, in parallel with an increase in 24S-hydroxycholesterol level and a reduction of cholesterol synthesis. The failure to demonstrate a significant transcriptional regulation under most conditions is discussed in relation to the turnover of brain and neuronal cholesterol.


Subject(s)
Steroid Hydroxylases/genetics , Transcription, Genetic , Alzheimer Disease/enzymology , Animals , Base Sequence , Brain/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Cholesterol 24-Hydroxylase , Humans , Mice , Molecular Sequence Data , Promoter Regions, Genetic , RNA, Messenger/analysis , Transcription Initiation Site
20.
J Pharmacol Exp Ther ; 316(3): 1146-52, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16282522

ABSTRACT

On a global scale, there is an increasing tendency for a more aggressive treatment of hypercholesterolemia. Minor effects of statins on brain cholesterol metabolism have been reported in some in vivo animal studies, and it seems that this is due to a local effect of the drug. We treated male mice of the inbred strain C57/BL6 with a high daily dose of lipophilic simvastatin (100 mg/kg b.wt.) or hydrophilic pravastatin (200 mg/kg b.wt.) or vehicle (controls) by oral gavage for 3 days. To compare the impact of both statins on brain cholesterol synthesis and degradation, levels of cholesterol, its precursor lathosterol, and its brain metabolite 24(S)-hydroxycholesterol as well as statin concentrations were determined in whole-brain lipid extracts using mass spectrometry. The expression of 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase mRNA and of other target genes were evaluated using real-time reverse transcription-polymerase chain reaction. In addition, analysis of liver and serum samples was performed. Similar levels of simvastatin and pravastatin were detected in whole-brain homogenates. Cholesterol contents in the brain, liver, and serum were not affected by high-dose statin treatment. Whereas brain cholesterol precursor levels were reduced in simvastatin-treated animals only, no effect was observed on the formation of the brain cholesterol metabolite, 24(S)-hydroxycholesterol. Polymerase chain reaction analysis revealed that mRNA expression of HMG-CoA reductase and ATP-binding cassette transporter A1 in the brain was significantly up-regulated in simvastatin-treated animals compared with pravastatin-treated or control animals. We conclude that, under the present experimental conditions, brain cholesterol synthesis is significantly affected by short-term treatment with high doses of lipophilic simvastatin, whereas whole-brain cholesterol turnover is not disturbed.


Subject(s)
Brain/metabolism , Cholesterol/biosynthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Pravastatin/pharmacology , Simvastatin/pharmacology , Animals , Hydroxymethylglutaryl CoA Reductases/metabolism , Liver/metabolism , Male , Mice , Phytosterols/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...