Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 496
Filter
1.
Cells ; 10(10)2021 10 05.
Article in English | MEDLINE | ID: mdl-34685636

ABSTRACT

Ambient temperature is an important determinant of both the alternative bile acid synthesis pathway controlled by oxysterol 7-α hydroxylase (CYP7B1) and the progression of metabolic-associated fatty liver disease (MAFLD). Here, we investigated whether CYP7B1 is involved in the etiology of MAFLD under conditions of low and high energy expenditure. For this, Cyp7b1-/- and wild type (WT) mice were fed a choline-deficient high-fat diet and housed either at 30 °C (thermoneutrality) or at 22 °C (mild cold). To study disease phenotype and underlying mechanisms, plasma and organ samples were analyzed to determine metabolic parameters, immune cell infiltration by immunohistology and flow cytometry, lipid species including hydroxycholesterols, bile acids and structural lipids. In WT and Cyp7b1-/- mice, thermoneutral housing promoted MAFLD, an effect that was more pronounced in CYP7B1-deficient mice. In these mice, we found higher plasma alanine aminotransferase activity, hyperlipidemia, hepatic accumulation of potentially harmful lipid species, aggravated liver fibrosis, increased inflammation and immune cell infiltration. Bile acids and hydroxycholesterols did not correlate with aggravated MAFLD in Cyp7b1-/- mice housed at thermoneutrality. Notably, an up-regulation of lipoprotein receptors was detected at 22 °C but not at 30 °C in livers of Cyp7b1-/- mice, suggesting that accelerated metabolism of lipoproteins carrying lipotoxic molecules counteracts MAFLD progression.


Subject(s)
Cytochrome P450 Family 7/metabolism , Fatty Liver/enzymology , Fatty Liver/metabolism , Steroid Hydroxylases/metabolism , Temperature , Animals , Biomarkers/metabolism , Cytochrome P450 Family 7/deficiency , Inflammation/pathology , Lipid Metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Phenotype , Receptors, Lipoprotein/metabolism , Spleen/immunology , Steroid Hydroxylases/deficiency , Up-Regulation
2.
PLoS One ; 16(9): e0257576, 2021.
Article in English | MEDLINE | ID: mdl-34551004

ABSTRACT

Exaggerated inflammatory response results in pathogenesis of various inflammatory diseases. Tumor Necrosis Factor-alpha (TNF) is a multi-functional pro-inflammatory cytokine regulating a wide spectrum of physiological, biological, and cellular processes. TNF induces Focal Adhesion Kinase (FAK) for various activities including induction of pro-inflammatory response. The mechanism of FAK activation by TNF is unknown and the involvement of cell surface integrins in modulating TNF response has not been determined. In the current study, we have identified an oxysterol 25-hydroxycholesterol (25HC) as a soluble extracellular lipid amplifying TNF mediated innate immune pro-inflammatory response. Our results demonstrated that 25HC-integrin-FAK pathway amplifies and optimizes TNF-mediated pro-inflammatory response. 25HC generating enzyme cholesterol 25-hydroxylase (C25H) was induced by TNF via NFκB and MAPK pathways. Specifically, chromatin immunoprecipitation assay identified binding of AP-1 (Activator Protein-1) transcription factor ATF2 (Activating Transcription Factor 2) to the C25H promoter following TNF stimulation. Furthermore, loss of C25H, FAK and α5 integrin expression and inhibition of FAK and α5ß1 integrin with inhibitor and blocking antibody, respectively, led to diminished TNF-mediated pro-inflammatory response. Thus, our studies show extracellular 25HC linking TNF pathway with integrin-FAK signaling for optimal pro-inflammatory activity and MAPK/NFκB-C25H-25HC-integrin-FAK signaling network playing an essential role to amplify TNF dependent pro-inflammatory response. Thus, we have identified 25HC as the key factor involved in FAK activation during TNF mediated response and further demonstrated a role of cell surface integrins in positively regulating TNF dependent pro-inflammatory response.


Subject(s)
Signal Transduction/drug effects , Steroid Hydroxylases/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Activating Transcription Factor 2/metabolism , Animals , Cells, Cultured , Chemokine CCL3/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Hydroxycholesterols/metabolism , Integrin alpha5/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Protein Binding , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Up-Regulation/drug effects
3.
J Lipid Res ; 61(1): 54-69, 2020 01.
Article in English | MEDLINE | ID: mdl-31645370

ABSTRACT

The bile acid (BA) composition in mice is substantially different from that in humans. Chenodeoxycholic acid (CDCA) is an end product in the human liver; however, mouse Cyp2c70 metabolizes CDCA to hydrophilic muricholic acids (MCAs). Moreover, in humans, the gut microbiota converts the primary BAs, cholic acid and CDCA, into deoxycholic acid (DCA) and lithocholic acid (LCA), respectively. In contrast, the mouse Cyp2a12 reverts this action and converts these secondary BAs to primary BAs. Here, we generated Cyp2a12 KO, Cyp2c70 KO, and Cyp2a12/Cyp2c70 double KO (DKO) mice using the CRISPR-Cas9 system to study the regulation of BA metabolism under hydrophobic BA composition. Cyp2a12 KO mice showed the accumulation of DCAs, whereas Cyp2c70 KO mice lacked MCAs and exhibited markedly increased hepatobiliary proportions of CDCA. In DKO mice, not only DCAs or CDCAs but also DCAs, CDCAs, and LCAs were all elevated. In Cyp2c70 KO and DKO mice, chronic liver inflammation was observed depending on the hepatic unconjugated CDCA concentrations. The BA pool was markedly reduced in Cyp2c70 KO and DKO mice, but the FXR was not activated. It was suggested that the cytokine/c-Jun N-terminal kinase signaling pathway and the pregnane X receptor-mediated pathway are the predominant mechanisms, preferred over the FXR/small heterodimer partner and FXR/fibroblast growth factor 15 pathways, for controlling BA synthesis under hydrophobic BA composition. From our results, we hypothesize that these KO mice can be novel and useful models for investigating the roles of hydrophobic BAs in various human diseases.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Bile Acids and Salts/metabolism , Cytochrome P-450 Enzyme System/genetics , Disease Models, Animal , Steroid Hydroxylases/genetics , Animals , Aryl Hydrocarbon Hydroxylases/deficiency , Aryl Hydrocarbon Hydroxylases/metabolism , Bile Acids and Salts/chemistry , Chenodeoxycholic Acid/chemistry , Chenodeoxycholic Acid/metabolism , Cytochrome P-450 Enzyme System/deficiency , Cytochrome P-450 Enzyme System/metabolism , Female , Hydrophobic and Hydrophilic Interactions , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Rats , Rats, Sprague-Dawley , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/metabolism
4.
J Crohns Colitis ; 13(9): 1186-1200, 2019 Sep 19.
Article in English | MEDLINE | ID: mdl-31220227

ABSTRACT

Intestinal fibrosis and stenosis are common complications of Crohn's disease [CD], frequently requiring surgery. Anti-inflammatory strategies can only partially prevent fibrosis; hence, anti-fibrotic therapies remain an unmet clinical need. Oxysterols are oxidised cholesterol derivatives with important roles in various biological processes. The enzyme cholesterol 25-hydroxylase [CH25H] converts cholesterol to 25-hydroxycholesterol [25-HC], which modulates immune responses and oxidative stress. In human intestinal samples from CD patients, we found a strong correlation of CH25H mRNA expression with the expression of fibrosis markers. We demonstrate reduced intestinal fibrosis in mice deficient for the CH25H enzyme, using the sodium dextran sulphate [DSS]-induced chronic colitis model. Additionally, using a heterotopic transplantation model of intestinal fibrosis, we demonstrate reduced collagen deposition and lower concentrations of hydroxyproline in CH25H knockouts. In the heterotopic transplant model, CH25H was expressed in fibroblasts. Taken together, our findings indicate an involvement of oxysterol synthesis in the pathogenesis of intestinal fibrosis.


Subject(s)
Intestines/pathology , Oxysterols/metabolism , Steroid Hydroxylases/metabolism , Adult , Aged , Aged, 80 and over , Animals , Colitis/chemically induced , Colitis/enzymology , Crohn Disease/complications , Crohn Disease/pathology , Dextran Sulfate/pharmacology , Disease Models, Animal , Female , Fibrosis , Humans , Intestines/enzymology , Intestines/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Steroid Hydroxylases/deficiency
5.
Biomolecules ; 9(4)2019 04 13.
Article in English | MEDLINE | ID: mdl-31013940

ABSTRACT

Deficiency in cytochrome P450 (CYP) 7B1, also known as oxysterol 7α-hydroxylase, in humans leads to hereditary spastic paraplegia type 5 (SPG5) and in some cases in infants to liver disease. SPG5 is medically characterized by loss of motor neurons in the corticospinal tract. In an effort to gain a better understanding of the fundamental biochemistry of this disorder, we have extended our previous profiling of the oxysterol content of brain and plasma of Cyp7b1 knockout (-/-) mice to include, amongst other sterols, 25-hydroxylated cholesterol metabolites. Although brain cholesterol levels do not differ between wild-type (wt) and knockout mice, we find, using a charge-tagging methodology in combination with liquid chromatography-mass spectrometry (LC-MS) and multistage fragmentation (MSn), that there is a build-up of the CYP7B1 substrate 25-hydroxycholesterol (25-HC) in Cyp7b1-/- mouse brain and plasma. As reported earlier, levels of (25R)26-hydroxycholesterol (26-HC), 3ß-hydroxycholest-5-en-(25R)26-oic acid and 24S,25-epoxycholesterol (24S,25-EC) are similarly elevated in brain and plasma. Side-chain oxysterols including 25-HC, 26-HC and 24S,25-EC are known to bind to INSIG (insulin-induced gene) and inhibit the processing of SREBP-2 (sterol regulatory element-binding protein-2) to its active form as a master regulator of cholesterol biosynthesis. We suggest the concentration of cholesterol in brain of the Cyp7b1-/- mouse is maintained by balancing reduced metabolism, as a consequence of a loss in CYP7B1, with reduced biosynthesis. The Cyp7b1-/- mouse does not show a motor defect; whether the defect in humans is a consequence of less efficient homeostasis of cholesterol in brain has yet to be uncovered.


Subject(s)
Brain/metabolism , Cytochrome P450 Family 7/genetics , Hydroxycholesterols/metabolism , Spastic Paraplegia, Hereditary/metabolism , Steroid Hydroxylases/genetics , Animals , Cytochrome P450 Family 7/deficiency , Hydroxycholesterols/blood , Male , Mice , Spastic Paraplegia, Hereditary/blood , Spastic Paraplegia, Hereditary/genetics , Steroid Hydroxylases/deficiency
6.
Nature ; 566(7743): 254-258, 2019 02.
Article in English | MEDLINE | ID: mdl-30728500

ABSTRACT

Osteoarthritis-the most common form of age-related degenerative whole-joint disease1-is primarily characterized by cartilage destruction, as well as by synovial inflammation, osteophyte formation and subchondral bone remodelling2,3. However, the molecular mechanisms that underlie the pathogenesis of osteoarthritis are largely unknown. Although osteoarthritis is currently considered to be associated with metabolic disorders, direct evidence for this is lacking, and the role of cholesterol metabolism in the pathogenesis of osteoarthritis has not been fully investigated4-6. Various types of cholesterol hydroxylases contribute to cholesterol metabolism in extrahepatic tissues by converting cellular cholesterol to circulating oxysterols, which regulate diverse biological processes7,8. Here we show that the CH25H-CYP7B1-RORα axis of cholesterol metabolism in chondrocytes is a crucial catabolic regulator of the pathogenesis of osteoarthritis. Osteoarthritic chondrocytes had increased levels of cholesterol because of enhanced uptake, upregulation of cholesterol hydroxylases (CH25H and CYP7B1) and increased production of oxysterol metabolites. Adenoviral overexpression of CH25H or CYP7B1 in mouse joint tissues caused experimental osteoarthritis, whereas knockout or knockdown of these hydroxylases abrogated the pathogenesis of osteoarthritis. Moreover, retinoic acid-related orphan receptor alpha (RORα) was found to mediate the induction of osteoarthritis by alterations in cholesterol metabolism. These results indicate that osteoarthritis is a disease associated with metabolic disorders and suggest that targeting the CH25H-CYP7B1-RORα axis of cholesterol metabolism may provide a therapeutic avenue for treating osteoarthritis.


Subject(s)
Cholesterol/metabolism , Cytochrome P450 Family 7/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Osteoarthritis/metabolism , Steroid Hydroxylases/metabolism , Animals , Biological Transport , Chondrocytes/enzymology , Chondrocytes/metabolism , Male , Mice , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Osteoarthritis/enzymology , Osteoarthritis/pathology , Oxysterols/metabolism , Steroid Hydroxylases/deficiency , Up-Regulation
7.
Clin Liver Dis ; 22(4): 671-687, 2018 11.
Article in English | MEDLINE | ID: mdl-30266156

ABSTRACT

Inborn errors of bile acid metabolism are rare causes of neonatal cholestasis and liver disease in older children and adults. The diagnosis should be considered in the context of hyperbilirubinemia with normal serum bile acids and made by urinary liquid secondary ionization mass spectrometry or DNA testing. Cholic acid is an effective treatment of most single-enzyme defects and patients with Zellweger spectrum disorder with liver disease.


Subject(s)
Bile Acids and Salts/metabolism , Cholestasis/etiology , Liver Diseases/etiology , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/genetics , 3-Oxo-5-alpha-Steroid 4-Dehydrogenase/deficiency , Acyl-CoA Oxidase/deficiency , Adrenal Hyperplasia, Congenital/complications , Adrenal Hyperplasia, Congenital/genetics , Amino-Acid N-Acetyltransferase/deficiency , Cholic Acid/therapeutic use , Genetic Testing , Humans , Liver Diseases/pathology , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/drug therapy , Racemases and Epimerases/deficiency , Steroid Hydroxylases/deficiency , Xanthomatosis, Cerebrotendinous/complications , Xanthomatosis, Cerebrotendinous/genetics
8.
Cell Rep ; 23(13): 3750-3758, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29949760

ABSTRACT

The intestinal tract is constantly exposed to various stimuli. Group 3 innate lymphoid cells (ILC3s) reside in lymphoid organs and in the intestinal tract and are required for immunity to enteric bacterial infection. However, the mechanisms that regulate the ILC3s in vivo remain incompletely defined. Here, we show that GPR183, a chemotactic receptor expressed on murine and human ILC3s, regulates ILC3 migration toward its ligand 7α,25-dihydroxycholesterol (7α,25-OHC) in vitro, and GPR183 deficiency in vivo leads to a disorganized distribution of ILC3s in mesenteric lymph nodes and decreased ILC3 accumulation in the intestine. GPR183 functions intrinsically in ILC3s, and GPR183-deficient mice are more susceptible to enteric bacterial infection. Together, these results reveal a role for the GPR183-7α,25-OHC pathway in regulating the accumulation, distribution, and anti-microbial and tissue-protective functions of ILC3s and define a critical role for this pathway in promoting innate immunity to enteric bacterial infection.


Subject(s)
Lymphoid Tissue/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Movement , Citrobacter rodentium/pathogenicity , Cytochrome P450 Family 7/metabolism , Enterobacteriaceae Infections/pathology , Enterobacteriaceae Infections/prevention & control , Enterobacteriaceae Infections/veterinary , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/metabolism , Immunity, Innate , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Ligands , Lymphoid Tissue/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/cytology , Mucous Membrane/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism
9.
J Pharm Biomed Anal ; 150: 396-405, 2018 Feb 20.
Article in English | MEDLINE | ID: mdl-29288965

ABSTRACT

The potential use of cholesterol esterases was tested to avoid alkaline hydrolysis for cleavage of plasma esterified oxysterols. The enzymatic hydrolysis was optimized by testing two sources of enzyme-Pseudomonas and bovine pancreas, presence of surfactants, incubation time and amount of enzyme. Free forms of 4ß-, 7-, 24-, 25- and 27-hydroxycholesterol (HC) as well 7-ketocholesterol (7-KC) were analyzed by liquid chromatography and mass-spectrometry using the deuterated internal standard, 25-HC(d6). Enzymatic hydrolysis was more effective using the Pseudomonas enzyme and in presence of surfactants. Compared to alkaline hydrolysis, it generated a cleaner chromatographic baseline and better recovery of the internal standard. Oxysterols were assayed with detection limits between 7 and 31 pg/mL. Interassay coefficients of variation were lower than 10% and extraction recovery efficiencies, higher than 90%. The procedure was used to characterize plasma levels of Cyp7b1-deficient rat, where it showed increased plasma levels of 7, 24 and 25-HC. Due to the low volume of sample required, it may be used in other animal models, particularly rodents, as well as in pediatric samples where sample amount is always a problem. Thus, the proposed new method offers mild enzymatic processing that greatly facilitates oxysterol determinations to delineate their role in physiopathology.


Subject(s)
Bacterial Proteins/chemistry , Chromatography, Liquid , Oxysterols/blood , Solid Phase Extraction , Sterol Esterase/chemistry , Tandem Mass Spectrometry , Animals , Animals, Genetically Modified , Calibration , Chromatography, Liquid/standards , Cytochrome P450 Family 7/deficiency , Cytochrome P450 Family 7/genetics , Humans , Hydrolysis , Male , Pseudomonas aeruginosa/enzymology , Rats, Inbred F344 , Reference Standards , Reproducibility of Results , Solid Phase Extraction/standards , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Tandem Mass Spectrometry/standards
10.
Drug Metab Dispos ; 45(8): 977-981, 2017 08.
Article in English | MEDLINE | ID: mdl-28546505

ABSTRACT

Phenobarbital (PB) promotes liver tumorigenesis in rodents, in part through activation of the constitutive androstane receptor (CAR) and the consequent changes in hepatic gene expression and increases in hepatocyte proliferation. A typical effect of CAR activation by PB is a marked induction of Cyp2b10 expression in the liver; the latter has been suspected to be vital for PB-induced hepatocellular proliferation. This hypothesis was tested here by using a Cyp2a(4/5)bgs-null (null) mouse model in which all Cyp2b genes are deleted. Adult male and female wild-type (WT) and null mice were treated intraperitoneally with PB at 50 mg/kg once daily for 5 successive days and tested on day 6. The liver-to-body weight ratio, an indicator of liver hypertrophy, was increased by 47% in male WT mice, but by only 22% in male Cyp2a(4/5)bgs-null mice, by the PB treatment. The fractions of bromodeoxyuridine-positive hepatocyte nuclei, assessed as a measure of the rate of hepatocyte proliferation, were also significantly lower in PB-treated male null mice compared with PB-treated male WT mice. However, whereas few proliferating hepatocytes were detected in saline-treated mice, many proliferating hepatocytes were still detected in PB-treated male null mice. In contrast, female WT mice were much less sensitive than male WT mice to PB-induced hepatocyte proliferation, and PB-treated female WT and PB-treated female null mice did not show significant difference in rates of hepatocyte proliferation. These results indicate that CYP2B induction plays a significant, but partial, role in PB-induced hepatocyte proliferation in male mice.


Subject(s)
Aryl Hydrocarbon Hydroxylases/physiology , Cytochrome P450 Family 2/physiology , Hepatocytes/drug effects , Hepatocytes/enzymology , Phenobarbital/pharmacology , Steroid Hydroxylases/physiology , Animals , Aryl Hydrocarbon Hydroxylases/deficiency , Aryl Hydrocarbon Hydroxylases/genetics , Cell Proliferation/drug effects , Cell Proliferation/physiology , Enzyme Induction/drug effects , Enzyme Induction/physiology , Female , Hypnotics and Sedatives/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics
11.
Nat Commun ; 7: 13129, 2016 10 25.
Article in English | MEDLINE | ID: mdl-27779191

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD), caused by an ABCD1 mutation, is a progressive neurodegenerative disorder associated with the accumulation of very long-chain fatty acids (VLCFA). Cerebral inflammatory demyelination is the major feature of childhood cerebral ALD (CCALD), the most severe form of ALD, but its underlying mechanism remains poorly understood. Here, we identify the aberrant production of cholesterol 25-hydroxylase (CH25H) and 25-hydroxycholesterol (25-HC) in the cellular context of CCALD based on the analysis of ALD patient-derived induced pluripotent stem cells and ex vivo fibroblasts. Intriguingly, 25-HC, but not VLCFA, promotes robust NLRP3 inflammasome assembly and activation via potassium efflux-, mitochondrial reactive oxygen species (ROS)- and liver X receptor (LXR)-mediated pathways. Furthermore, stereotaxic injection of 25-HC into the corpus callosum of mouse brains induces microglial recruitment, interleukin-1ß production, and oligodendrocyte cell death in an NLRP3 inflammasome-dependent manner. Collectively, our results indicate that 25-HC mediates the neuroinflammation of X-ALD via activation of the NLRP3 inflammasome.


Subject(s)
Adrenoleukodystrophy/metabolism , Corpus Callosum/drug effects , Hydroxycholesterols/pharmacology , Induced Pluripotent Stem Cells/drug effects , Inflammasomes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/metabolism , Adrenoleukodystrophy/chemically induced , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/pathology , Animals , Corpus Callosum/metabolism , Corpus Callosum/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Hydroxycholesterols/metabolism , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Inflammasomes/metabolism , Inflammation , Injections, Intraventricular , Liver X Receptors/genetics , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/pathology , Primary Cell Culture , Reactive Oxygen Species/metabolism , Stereotaxic Techniques , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , White Matter/drug effects , White Matter/metabolism , White Matter/pathology
12.
Mol Neurodegener ; 10: 29, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26169917

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) 42 has been implicated as the initiating molecule in the pathogenesis of Alzheimer's disease (AD); thus, therapeutic strategies that target Aß42 are of great interest. γ-Secretase modulators (GSMs) are small molecules that selectively decrease Aß42. We have previously reported that many acidic steroids are GSMs with potencies ranging in the low to mid micromolar concentration with 5ß-cholanic acid being the most potent steroid identified GSM with half maximal effective concentration (EC50) of 5.7 µM. RESULTS: We find that the endogenous cholesterol metabolite, 3ß-hydroxy-5-cholestenoic acid (CA), is a steroid GSM with enhanced potency (EC50 of 250 nM) relative to 5ß-cholanic acid. CA i) is found in human plasma at ~100-300 nM concentrations ii) has the typical acidic GSM signature of decreasing Aß42 and increasing Aß38 levels iii) is active in in vitro γ-secretase assay iv) is made in the brain. To test if CA acts as an endogenous GSM, we used Cyp27a1 knockout (Cyp27a1-/-) and Cyp7b1 knockout (Cyp7b1-/-) mice to investigate if manipulation of cholesterol metabolism pathways relevant to CA formation would affect brain Aß42 levels. Our data show that Cyp27a1-/- had increased brain Aß42, whereas Cyp7b1-/- mice had decreased brain Aß42 levels; however, peripheral dosing of up to 100 mg/kg CA did not affect brain Aß levels. Structure-activity relationship (SAR) studies with multiple known and novel CA analogs studies failed to reveal CA analogs with increased potency. CONCLUSION: These data suggest that CA may act as an endogenous GSM within the brain. Although it is conceptually attractive to try and increase the levels of CA in the brain for prevention of AD, our data suggest that this will not be easily accomplished.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cholesterol/analogs & derivatives , Peptide Fragments/metabolism , Animals , Blood-Brain Barrier , CHO Cells , Cells, Cultured , Cholestanetriol 26-Monooxygenase/deficiency , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/chemistry , Cholesterol/metabolism , Cholesterol/pharmacology , Cholic Acids/pharmacology , Coculture Techniques , Cricetinae , Cricetulus , Cytochrome P450 Family 7 , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Structure , Neuroglia/metabolism , Neurons/metabolism , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Structure-Activity Relationship
13.
J Autoimmun ; 56: 45-55, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25456971

ABSTRACT

Perturbation of steroids pathways is linked to inflammation and chronic diseases, however the underlying mechanism remains unclear. Oxysterols, oxidized forms of cholesterol, are not only essential for bile synthesis and sterol transportation but have recently been shown to contribute to the immune response. In addition, serum oxysterols levels have been proposed as suitable candidate biomarkers for neurological diseases such as multiple sclerosis (MS). However how oxysterols modulate adaptive immunity is unknown and their functions in autoimmunity have not been investigated. The enzyme cholesterol 25 hydroxylase (Ch25h) is the rate limiting step to synthesize the oxysterol 7α,25-dihydroxycholesterol (7α,25-OHC) from cholesterol. We here report, using the MS murine model experimental autoimmune encephalomyelitis (EAE), that Ch25h deletion significantly attenuated EAE disease course by limiting trafficking of pathogenic CD4(+) T lymphocytes to the central nervous system (CNS). Mechanistically, we show a critical involvement for oxysterols in recruiting leukocytes into inflamed tissues and propose that 7α,25-OHC preferentially promotes the migration of activated CD44(+)CD4(+) T cells by binding the G protein-coupled receptor called Epstein-Barr virus induced gene 2 (EBI2). Collectively, our results support a pro-inflammatory role for oxysterols during EAE and identify oxysterols as a potential therapeutic target to treat autoimmune diseases.


Subject(s)
Autoimmunity , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Movement/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Hydroxycholesterols/pharmacology , Animals , Antigens/immunology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Hyaluronan Receptors/metabolism , Interleukin-17/biosynthesis , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/drug effects , Mice , Mice, Knockout , Monocytes/cytology , Receptors, G-Protein-Coupled/metabolism , Severity of Illness Index , Signal Transduction , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics
14.
PLoS One ; 9(9): e108382, 2014.
Article in English | MEDLINE | ID: mdl-25268137

ABSTRACT

AIM: Altered vitamin D signaling is associated with cardiac dysfunction, but the pathogenic mechanism is not clearly understood. We examine the mechanism and the role of vitamin D signaling in the development of cardiac dysfunction. METHODS AND RESULTS: We analyzed 1α-hydroxylase (1α-OHase) knockout (1α-OHase-/-) mice, which lack 1α-OH enzymes that convert the inactive form to hormonally active form of vitamin D. 1α-OHase-/- mice showed modest cardiac hypertrophy at baseline. Induction of pressure overload by transverse aortic constriction (TAC) demonstrated exaggerated cardiac dysfunction in 1α-OHase-/- mice compared to their WT littermates with a significant increase in fibrosis and expression of inflammatory cytokines. Analysis of calcium (Ca2+) transient demonstrated profound Ca2+ handling abnormalities in 1α-OHase-/- mouse cardiomyocytes (CMs), and treatment with paricalcitol (PC), an activated vitamin D3 analog, significantly attenuated defective Ca2+ handling in 1α-OHase-/- CMs. We further delineated the effect of vitamin D deficiency condition to TAC by first correcting the vitamin D deficiency in 1α-OHase-/- mice, followed then by either a daily maintenance dose of vitamin D or vehicle (to achieve vitamin D deficiency) at the time of sham or TAC. In mice treated with vitamin D, there was a significant attenuation of TAC-induced cardiac hypertrophy, interstitial fibrosis, inflammatory markers, Ca2+ handling abnormalities and cardiac function compared to the vehicle treated animals. CONCLUSIONS: Our results provide insight into the mechanism of cardiac dysfunction, which is associated with severely defective Ca2+ handling and defective vitamin D signaling in 1α-OHase-/- mice.


Subject(s)
Aortic Valve Stenosis/metabolism , Calcium/metabolism , Cardiomegaly/metabolism , Signal Transduction , Steroid Hydroxylases/genetics , Vitamin D/metabolism , Animals , Aortic Valve Stenosis/diet therapy , Aortic Valve Stenosis/genetics , Aortic Valve Stenosis/pathology , Cardiomegaly/diet therapy , Cardiomegaly/genetics , Cardiomegaly/pathology , Ergocalciferols/pharmacology , Fibrosis , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Primary Cell Culture , Steroid Hydroxylases/deficiency , Vitamin D/administration & dosage
15.
Am J Pathol ; 184(9): 2403-19, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25065682

ABSTRACT

Cholesterol elimination from nonhepatic cells involves metabolism to side-chain oxysterols, which serve as transport forms of cholesterol and bioactive molecules modulating a variety of cellular processes. Cholesterol metabolism is tissue specific, and its significance has not yet been established for the retina, where cytochromes P450 (CYP27A1 and CYP46A1) are the major cholesterol-metabolizing enzymes. We generated Cyp27a1(-/-)Cyp46a1(-/-) mice, which were lean and had normal serum cholesterol and glucose levels. These animals, however, had changes in the retinal vasculature, retina, and several nonocular organs (lungs, liver, and spleen). Changes in the retinal vasculature included structural abnormalities (retinal-choroidal anastomoses, arteriovenous shunts, increased permeability, dilation, nonperfusion, and capillary degeneration) and cholesterol deposition and oxidation in the vascular wall, which also exhibited increased adhesion of leukocytes and activation of the complement pathway. Changes in the retina included increased content of cholesterol and its metabolite, cholestanol, which were focally deposited at the apical and basal sides of the retinal pigment epithelium. Retinal macrophages of Cyp27a1(-/-)Cyp46a1(-/-) mice were activated, and oxidative stress was noted in their photoreceptor inner segments. Our findings demonstrate the importance of retinal cholesterol metabolism for maintenance of the normal retina, and suggest new targets for diseases affecting the retinal vasculature.


Subject(s)
Cholestanetriol 26-Monooxygenase/deficiency , Cholesterol/metabolism , Retina/metabolism , Retina/pathology , Steroid Hydroxylases/deficiency , Animals , Cholesterol 24-Hydroxylase , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Spleen/pathology
16.
Cell Metab ; 20(1): 172-82, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24954418

ABSTRACT

Oxysterols are cholesterol metabolites that serve multiple functions in lipid metabolism, including as liver X receptor (LXR) ligands. 27-hydroxycholesterol (27HC) is an abundant oxysterol metabolized by CYP7B1. How 27HC impacts vascular health is unknown. We show that elevations in 27HC via cyp7b1 deletion promote atherosclerosis in apoe(-/-) mice without altering lipid status; furthermore, estrogen-related atheroprotection is attenuated. In wild-type mice, leukocyte-endothelial cell adhesion is increased by 27HC via estrogen receptor (ER)-dependent processes. In monocytes/macrophages, 27HC upregulates proinflammatory genes and increases adhesion via ERα. In endothelial cells, 27HC is also proadhesive via ERα, and in contrast to estrogen, which blunts NF-κB activation, 27HC stimulates NF-κB activation via Erk1,2 and JNK-dependent IκBα degradation. Whereas 27HC administration to apoe(-/-) mice increases atherosclerosis, apoe(-/-);erα(-/-) are unaffected. Thus, 27HC promotes atherosclerosis via proinflammatory processes mediated by ERα, and it attenuates estrogen-related atheroprotection. Strategies to lower 27HC may complement approaches targeting cholesterol to prevent vascular disease.


Subject(s)
Cholesterol/metabolism , Estrogen Receptor alpha/metabolism , Hydroxycholesterols/pharmacology , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Adhesion/drug effects , Cell Line , Cytochrome P450 Family 7 , Cytokines/genetics , Cytokines/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Estrogen Receptor alpha/genetics , Female , Hydroxycholesterols/metabolism , I-kappa B Proteins/metabolism , Inflammation , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism
17.
Biochem Biophys Res Commun ; 449(2): 229-34, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24832732

ABSTRACT

24S,25-Epoxycholesterol is formed in a shunt of the mevalonate pathway that produces cholesterol. It is one of the most potent known activators of the liver X receptors and can inhibit sterol regulatory element-binding protein processing. Until recently analysis of 24S,25-epoxycholesterol at high sensitivity has been precluded by its thermal lability and lack of a strong chromophore. Here we report on the analysis of 24S,25-epoxycholesterol in rodent brain where its level was determined to be of the order of 0.4-1.4µg/g wet weight in both adult mouse and rat. For comparison the level of 24S-hydroxycholesterol in brain of both rodents was of the order of 20µg/g, while that of cholesterol in mouse was 10-20mg/g. By exploiting knockout mice for the enzyme oxysterol 7α-hydroxylase (Cyp7b1) we show that this enzymes is important for the subsequent metabolism of the 24S,25-epoxide.


Subject(s)
Brain/metabolism , Cholesterol/analogs & derivatives , Animals , Cholesterol/metabolism , Cytochrome P450 Family 7 , Female , Male , Metabolic Networks and Pathways , Mevalonic Acid/metabolism , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Sterols/metabolism
18.
J Inherit Metab Dis ; 37(5): 851-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24658845

ABSTRACT

A child of consanguineous parents of Pakistani origin developed jaundice at 5 weeks and then, at 3 months, irritability, a prolonged prothrombin time, a low albumin, and episodes of hypoglycaemia. Investigation showed an elevated alanine aminotransferase with a normal γ-glutamyl-transpeptidase. Analysis of urine by electrospray ionisation tandem mass spectrometry (ESI-MS/MS) showed that the major peaks were m/z 480 (taurine-conjugated 3ß-hydroxy-5-cholenoic acid) and m/z 453 (sulphated 3ß-hydroxy-5-cholenoic acid). Analysis of plasma by gas chromatography-mass spectrometry (GC-MS) showed increased concentrations of 3ß-hydroxy-5-cholenoic acid, 3ß-hydroxy-5-cholestenoic acid and 27-hydroxycholesterol, indicating oxysterol 7 α-hydroxylase deficiency. The patient was homozygous for a mutation (c.1249C>T) in CYP7B1 that alters a highly conserved residue in oxysterol 7 α-hydroxylase (p.R417C) - previously reported in a family with hereditary spastic paraplegia type 5. On treatment with ursodeoxycholic acid (UDCA), his condition was worsening, but on chenodeoxycholic acid (CDCA), 15 mg/kg/d, he improved rapidly. A biopsy (after 2 weeks on CDCA), showed a giant cell hepatitis, an evolving micronodular cirrhosis, and steatosis. The improvement in liver function on CDCA was associated with a drop in the plasma concentrations and urinary excretions of the 3ß-hydroxy-Δ5 bile acids which are considered hepatotoxic. At age 5 years (on CDCA, 6 mg/kg/d), he was thriving with normal liver function. Neurological development was normal apart from a tendency to trip. Examination revealed pes cavus but no upper motor neuron signs. The findings in this case suggest that CDCA can reduce the activity of cholesterol 27-hydroxylase - the first step in the acidic pathway for bile acid synthesis.


Subject(s)
Chenodeoxycholic Acid/therapeutic use , Liver Diseases/drug therapy , Liver Diseases/genetics , Steroid Hydroxylases/deficiency , Steroid Hydroxylases/genetics , Bile Acids and Salts/blood , Bile Acids and Salts/urine , Consanguinity , Cytochrome P450 Family 7 , Humans , Infant , Liver/pathology , Liver Diseases/enzymology , Male , Metabolism, Inborn Errors/drug therapy , Metabolism, Inborn Errors/genetics
19.
Clin J Am Soc Nephrol ; 8(4): 649-57, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23293122

ABSTRACT

BACKGROUND AND OBJECTIVES: Elevated serum vitamin D with hypercalciuria can result in nephrocalcinosis and nephrolithiasis. This study evaluated the cause of excess 1,25-dihydroxycholecalciferol (1α,25(OH)2D3) in the development of those disorders in two individuals. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Two patients with elevated vitamin D levels and nephrocalcinosis or nephrolithiasis were investigated at the National Institutes of Health (NIH) Clinical Center and the NIH Undiagnosed Diseases Program, by measuring calcium, phosphate, and vitamin D metabolites, and by performing CYP24A1 mutation analysis. RESULTS: Both patients exhibited hypercalciuria, hypercalcemia, low parathyroid hormone, elevated vitamin D (1α,25(OH)2D3), normal 25-OHD3, decreased 24,25(OH)2D, and undetectable activity of 1,25(OH)2D-24-hydroxylase (CYP24A1), the enzyme that inactivates 1α,25(OH)2D3. Both patients had bi-allelic mutations in CYP24A1 leading to loss of function of this enzyme. On the basis of dbSNP data, the frequency of predicted deleterious bi-allelic CYP24A1 variants in the general population is estimated to be as high as 4%-20%. CONCLUSIONS: The results of this study show that 1,25(OH)2D-24-hydroxylase deficiency due to bi-allelic mutations in CYP24A1 causes elevated serum vitamin D, hypercalciuria, nephrocalcinosis, and renal stones.


Subject(s)
Nephrocalcinosis/genetics , Nephrolithiasis/genetics , Steroid Hydroxylases/genetics , Adult , Calcium/blood , Child , Family Health , Female , Humans , Hypercalciuria/etiology , Hypercalciuria/genetics , Hypercalciuria/metabolism , Male , Nephrocalcinosis/etiology , Nephrocalcinosis/metabolism , Nephrolithiasis/etiology , Nephrolithiasis/metabolism , Pedigree , Phosphates/blood , Primary Cell Culture , Steroid Hydroxylases/deficiency , Vitamin D/blood , Vitamin D3 24-Hydroxylase
20.
Clin Chim Acta ; 413(19-20): 1700-4, 2012 Oct 09.
Article in English | MEDLINE | ID: mdl-22652365

ABSTRACT

BACKGROUND: Inborn errors of bile acid synthesis are rare genetic disorders that can present with cholestatic liver disease. Recently we encountered 3 infants with neonatal cholestasis and excessive 3ß-monohydroxy-Δ5-C24 bile acids in serum and urine. We investigated whether identification of 3ß-hydroxy-5-cholestenoic acid and 27-hydroxycholesterol in serum and urine of cholestatic patients is necessary for diagnosis of primary oxysterol 7α-hydroxylase deficiency. METHODS: These 3 patients initially led us to suspected oxysterol 7α-hydroxylase deficiency. However, sequence analysis of genomic DNA resulted in diagnosis of 2 patients with oxysterol 7α-hydroxylase deficiency and 1 patient with 3ß-hydroxy-Δ5-C27-steroid dehydrogenase/isomerase deficiency. We examined identification of 3ß-hydroxy-5-cholestenoic acid and 27-hydroxycholesterol by gas chromatography-mass spectrometry after diagnosis. RESULTS: Interestingly, we detected a peak for 3ß-hydroxy-5-cholestenoic acid in serum and 27-hydroxycholesterol of the neutral sterol in urine from 2 patients who were diagnosed with primary oxysterol 7α-hydroxylase deficiency. CONCLUSION: In evaluating infants with cholestasis and excessive 3ß-monohydroxy-Δ5-C24 bile acids in infancy, one needs to conduct C24 bile acid analysis serially. Results can guide performance and interpretation of genomic DNA analysis. Moreover, identification of 3ß-hydroxy-5-cholestenoic acid in serum and 27-hydroxycholesterol in urine is highly important for diagnosis of oxysterol 7α-hydroxylase deficiency as is genomic DNA analysis.


Subject(s)
3-Hydroxysteroid Dehydrogenases/deficiency , Bile Acids and Salts/blood , Bile Acids and Salts/urine , Cholestasis/diagnosis , Isomerases/deficiency , Steroid Hydroxylases/deficiency , 3-Hydroxysteroid Dehydrogenases/genetics , Asian People , Bile Acids and Salts/biosynthesis , Cholestasis/genetics , Cholestasis/metabolism , DNA/chemistry , DNA/genetics , Female , Gas Chromatography-Mass Spectrometry , Humans , Hydroxycholesterols/urine , Infant , Isomerases/genetics , Male , Sequence Analysis, DNA , Steroid Hydroxylases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...