Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Elife ; 122023 09 28.
Article in English | MEDLINE | ID: mdl-37769126

ABSTRACT

Intermittent fasting (IF) has been shown to reduce cardiovascular risk factors in both animals and humans, and can protect the heart against ischemic injury in models of myocardial infarction. However, the underlying molecular mechanisms behind these effects remain unclear. To shed light on the molecular and cellular adaptations of the heart to IF, we conducted comprehensive system-wide analyses of the proteome, phosphoproteome, and transcriptome, followed by functional analysis. Using advanced mass spectrometry, we profiled the proteome and phosphoproteome of heart tissues obtained from mice that were maintained on daily 12- or 16 hr fasting, every-other-day fasting, or ad libitum control feeding regimens for 6 months. We also performed RNA sequencing to evaluate whether the observed molecular responses to IF occur at the transcriptional or post-transcriptional levels. Our analyses revealed that IF significantly affected pathways that regulate cyclic GMP signaling, lipid and amino acid metabolism, cell adhesion, cell death, and inflammation. Furthermore, we found that the impact of IF on different metabolic processes varied depending on the length of the fasting regimen. Short IF regimens showed a higher correlation of pathway alteration, while longer IF regimens had an inverse correlation of metabolic processes such as fatty acid oxidation and immune processes. Additionally, functional echocardiographic analyses demonstrated that IF enhances stress-induced cardiac performance. Our systematic multi-omics study provides a molecular framework for understanding how IF impacts the heart's function and its vulnerability to injury and disease.


Subject(s)
Intermittent Fasting , Multiomics , Humans , Mice , Animals , Proteome , Fasting/physiology , Energy Metabolism
2.
J Clin Invest ; 132(21)2022 11 01.
Article in English | MEDLINE | ID: mdl-36107630

ABSTRACT

BACKGROUNDCytochrome P450 family 8 subfamily B member 1 (CYP8B1) generates 12α-hydroxylated bile acids (BAs) that are associated with insulin resistance in humans.METHODSTo determine whether reduced CYP8B1 activity improves insulin sensitivity, we sequenced CYP8B1 in individuals without diabetes and identified carriers of complete loss-of-function (CLOF) mutations utilizing functional assays.RESULTSMutation carriers had lower plasma 12α-hydroxylated/non-12α-hydroxylated BA and cholic acid (CA)/chenodeoxycholic acid (CDCA) ratios compared with age-, sex-, and BMI-matched controls. During insulin clamps, hepatic glucose production was suppressed to a similar magnitude by insulin, but glucose infusion rates to maintain euglycemia were higher in mutation carriers, indicating increased peripheral insulin sensitivity. Consistently, a polymorphic CLOF CYP8B1 mutation associated with lower fasting insulin in the AMP-T2D-GENES study. Exposure of primary human muscle cells to mutation-carrier CA/CDCA ratios demonstrated increased FOXO1 activity, and upregulation of both insulin signaling and glucose uptake, which were mediated by increased CDCA. Inhibition of FOXO1 attenuated the CDCA-mediated increase in muscle insulin signaling and glucose uptake. We found that reduced CYP8B1 activity associates with increased insulin sensitivity in humans.CONCLUSIONOur findings suggest that increased circulatory CDCA due to reduced CYP8B1 activity increases skeletal muscle insulin sensitivity, contributing to increased whole-body insulin sensitization.FUNDINGBiomedical Research Council/National Medical Research Council of Singapore.


Subject(s)
Insulin Resistance , Steroid 12-alpha-Hydroxylase , Humans , Steroid 12-alpha-Hydroxylase/genetics , Insulin Resistance/genetics , Insulin/genetics , Haploinsufficiency , Bile Acids and Salts , Cholic Acid , Glucose
3.
Autophagy ; 18(9): 2150-2160, 2022 09.
Article in English | MEDLINE | ID: mdl-35012409

ABSTRACT

Caffeine is among the most highly consumed substances worldwide, and it has been associated with decreased cardiovascular risk. Although caffeine has been shown to inhibit the proliferation of vascular smooth muscle cells (VSMCs), the mechanism underlying this effect is unknown. Here, we demonstrated that caffeine decreased VSMC proliferation and induced macroautophagy/autophagy in an in vivo vascular injury model of restenosis. Furthermore, we studied the effects of caffeine in primary human and mouse aortic VSMCs and immortalized mouse aortic VSMCs. Caffeine decreased cell proliferation, and induced autophagy flux via inhibition of MTOR signaling in these cells. Genetic deletion of the key autophagy gene Atg5, and the Sqstm1/p62 gene encoding a receptor protein, showed that the anti-proliferative effect by caffeine was dependent upon autophagy. Interestingly, caffeine also decreased WNT-signaling and the expression of two WNT target genes, Axin2 and Ccnd1 (cyclin D1). This effect was mediated by autophagic degradation of a key member of the WNT signaling cascade, DVL2, by caffeine to decrease WNT signaling and cell proliferation. SQSTM1/p62, MAP1LC3B-II and DVL2 were also shown to interact with each other, and the overexpression of DVL2 counteracted the inhibition of cell proliferation by caffeine. Taken together, our in vivo and in vitro findings demonstrated that caffeine reduced VSMC proliferation by inhibiting WNT signaling via stimulation of autophagy, thus reducing the vascular restenosis. Our findings suggest that caffeine and other autophagy-inducing drugs may represent novel cardiovascular therapeutic tools to protect against restenosis after angioplasty and/or stent placement.


Subject(s)
Autophagy , Muscle, Smooth, Vascular , Animals , Autophagy/physiology , Caffeine/metabolism , Caffeine/pharmacology , Cell Proliferation , Cells, Cultured , Humans , Mice , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Sequestosome-1 Protein/metabolism , Wnt Signaling Pathway
4.
J Lipid Res ; 63(1): 100147, 2022 01.
Article in English | MEDLINE | ID: mdl-34752805

ABSTRACT

The myelin sheath, which is wrapped around axons, is a lipid-enriched structure produced by mature oligodendrocytes. Disruption of the myelin sheath is observed in several neurological diseases, such as multiple sclerosis. A crucial component of myelin is sphingomyelin, levels of which can be increased by ABCA8, a member of the ATP-binding cassette transporter family. ABCA8 is highly expressed in the cerebellum, specifically in oligodendroglia. However, whether ABCA8 plays a role in myelination and mechanisms that would underlie this role remain unknown. Here, we found that the absence of Abca8b, a mouse ortholog of ABCA8, led to decreased numbers of cerebellar oligodendrocyte precursor cells (OPCs) and mature oligodendrocytes in mice. We show that in oligodendrocytes, ABCA8 interacts with chondroitin sulfate proteoglycan 4 (CSPG4), a molecule essential for OPC proliferation, migration, and myelination. In the absence of Abca8b, localization of CSPG4 to the plasma membrane was decreased, contributing to reduced cerebellar CSPG4 expression. Cerebellar CSPG4+ OPCs were also diminished, leading to decreased mature myelinating oligodendrocyte numbers and cerebellar myelination levels in Abca8b-/- mice. In addition, electron microscopy analyses showed that the number of nonmyelinated cerebellar axons was increased, whereas cerebellar myelin thickness (g-ratio), myelin sheath periodicity, and axonal diameter were all decreased, indicative of disordered myelin ultrastructure. In line with disrupted cerebellar myelination, Abca8b-/- mice showed lower cerebellar conduction velocity and disturbed locomotion. In summary, ABCA8 modulates cerebellar myelination, in part through functional regulation of the ABCA8-interacting protein CSPG4. Our findings suggest that ABCA8 disruption may contribute to the pathophysiology of myelin disorders.


Subject(s)
Oligodendrocyte Precursor Cells
5.
Sci Rep ; 11(1): 20674, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667238

ABSTRACT

Vascular restenosis remains a major problem in patients with coronary artery disease (CAD) and peripheral artery disease (PAD). Neointimal hyperplasia, defined by post-procedure proliferation and migration of vascular smooth muscle cells (VSMCs) is a key underlying pathology. Here we investigated the role of Interleukin 11 (IL-11) in a mouse model of injury-related plaque development. Apoe-/- mice were fed a hyperlipidaemic diet and subjected to carotid wire injury of the right carotid. Mice were injected with an anti-IL11 antibody (X203), IgG control antibody or buffer. We performed ultrasound analysis to assess vessel wall thickness and blood velocity. Using histology and immunofluorescence approaches, we determined the effects of IL-11 inhibition on VSMC and macrophages phenotypes and fibrosis. Treatment of mice with carotid wire injury using X203 significantly reduced post-endothelial injury vessel wall thickness, and injury-related plaque, when compared to control. Immunofluorescence staining of the injury-related plaque showed that X203 treatment did not reduce macrophage numbers, but reduced the number of VSMCs and lowered matrix metalloproteinase 2 (MMP2) levels and collagen content in comparison to control. X203 treatment was associated with a significant increase in smooth muscle protein 22α (SM22α) positive cells in injury-related plaque compared to control, suggesting preservation of the contractile VSMC phenotype. Interestingly, X203 also reduced the collagen content of uninjured carotid arteries as compared to IgG, showing an additional effect on hyperlipidemia-induced arterial remodeling in the absence of mechanical injury. Therapeutic inhibition of IL-11 reduced vessel wall thickness, attenuated neointimal hyperplasia, and has favorable effects on vascular remodeling following wire-induced endothelial injury. This suggests IL-11 inhibition as a potential novel therapeutic approach to reduce arterial stenosis following revascularization in CAD and PAD patients.


Subject(s)
Antibodies, Neutralizing/pharmacology , Carotid Arteries/drug effects , Carotid Artery Injuries/drug therapy , Hyperplasia/drug therapy , Interleukin-11/metabolism , Animals , Carotid Arteries/metabolism , Carotid Artery Injuries/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen/metabolism , Disease Models, Animal , Hyperplasia/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neointima/drug therapy , Neointima/metabolism , Vascular Remodeling/drug effects
6.
Neuromolecular Med ; 23(1): 99-117, 2021 03.
Article in English | MEDLINE | ID: mdl-33085065

ABSTRACT

Bile acids are signalling hormones involved in the regulation of several metabolic pathways. The ability of bile acids to bind and signal through their receptors is modulated by the gut microbiome, since the microbiome contributes to the regulation and synthesis of bile acids as well to their physiochemical properties. From the gut, bacteria have been shown to send signals to the central nervous system via their metabolites, thus affecting the behaviour and brain function of the host organism. In the last years it has become increasingly evident that bile acids affect brain function, during normal physiological and pathological conditions. Although bile acids may be synthesized locally in the brain, the majority of brain bile acids are taken up from the systemic circulation. Since the composition of the brain bile acid pool may be regulated by the action of intestinal bacteria, it is possible that bile acids function as a communication bridge between the gut microbiome and the brain. However, little is known about the molecular mechanisms and the physiological roles of bile acids in the central nervous system. The possibility that bile acids may be a direct link between the intestinal microbiome and the brain is also an understudied subject. Here we review the influence of gut bacteria on the bile acid pool composition and properties, as well as striking evidence showing the role of bile acids as neuroactive molecules.


Subject(s)
Bile Acids and Salts/metabolism , Brain/metabolism , Gastrointestinal Microbiome , Animals , Cholesterol/metabolism , Cytochrome P-450 Enzyme System/metabolism , Eating , Enterochromaffin Cells/metabolism , Fermentation , Gallbladder/metabolism , Germ-Free Life , Humans , Liver/metabolism , Mice , Neurodegenerative Diseases/metabolism , Neurotransmitter Agents/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Stroke/metabolism , Xanthomatosis, Cerebrotendinous/metabolism
7.
Adv Drug Deliv Rev ; 159: 54-93, 2020.
Article in English | MEDLINE | ID: mdl-32423566

ABSTRACT

Lipids are hydrophobic and amphiphilic molecules involved in diverse functions such as membrane structure, energy metabolism, immunity, and signaling. However, altered intra-cellular lipid levels or composition can lead to metabolic and inflammatory dysfunction, as well as lipotoxicity. Thus, intra-cellular lipid homeostasis is tightly regulated by multiple mechanisms. Since most peripheral cells do not catabolize cholesterol, efflux (extra-cellular transport) of cholesterol is vital for lipid homeostasis. Defective efflux contributes to atherosclerotic plaque development, impaired ß-cell insulin secretion, and neuropathology. Of these, defective lipid efflux in macrophages in the arterial walls leading to foam cell and atherosclerotic plaque formation has been the most well studied, likely because a leading global cause of death is cardiovascular disease. Circulating high density lipoprotein particles play critical roles as acceptors of effluxed cellular lipids, suggesting their importance in disease etiology. We review here mechanisms and pathways that modulate lipid efflux, the role of lipid efflux in disease etiology, and therapeutic options aimed at modulating this critical process.


Subject(s)
Cardiovascular Diseases/metabolism , Lipid Metabolism , Animals , Cardiovascular Diseases/therapy , Humans , Lipoproteins, HDL/metabolism
8.
Nat Commun ; 11(1): 1677, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32245952

ABSTRACT

Human stem cell-derived hepatocyte-like cells (HLCs) offer an attractive platform to study liver biology. Despite their numerous advantages, HLCs lack critical in vivo characteristics, including cell polarity. Here, we report a stem cell differentiation protocol that uses transwell filters to generate columnar polarized HLCs with clearly defined basolateral and apical membranes separated by tight junctions. We show that polarized HLCs secrete cargo directionally: Albumin, urea, and lipoproteins are secreted basolaterally, whereas bile acids are secreted apically. Further, we show that enterically transmitted hepatitis E virus (HEV) progeny particles are secreted basolaterally as quasi-enveloped particles and apically as naked virions, recapitulating essential steps of the natural infectious cycle in vivo. We also provide proof-of-concept that polarized HLCs can be used for pharmacokinetic and drug-drug interaction studies. This novel system provides a powerful tool to study hepatocyte biology, disease mechanisms, genetic variation, and drug metabolism in a more physiologically relevant setting.


Subject(s)
Cell Culture Techniques/methods , Cell Polarity , Hepatocytes/physiology , Induced Pluripotent Stem Cells/physiology , Antiviral Agents/pharmacology , Cell Differentiation , Cells, Cultured , Drug Evaluation, Preclinical/methods , Drug Interactions , Hepatitis A Virus, Human/physiology , Hepatitis E virus/physiology , Hepatocytes/ultrastructure , Hepatocytes/virology , Humans , Liver/cytology , Liver/metabolism , Membrane Transport Proteins/metabolism , Microscopy, Electron, Transmission , Proof of Concept Study , Virion/metabolism , Virus Release , Virus Replication
9.
Arterioscler Thromb Vasc Biol ; 39(3): 387-401, 2019 03.
Article in English | MEDLINE | ID: mdl-30651003

ABSTRACT

Objective- Atherosclerotic coronary artery disease is the leading cause of death worldwide, and current treatment options are insufficient. Using systems-level network cluster analyses on a large coronary artery disease case-control cohort, we previously identified PCSK3 (proprotein convertase subtilisin/kexin family member 3; FURIN) as a member of several coronary artery disease-associated pathways. Thus, our objective is to determine the role of FURIN in atherosclerosis. Approach and Results- In vitro, FURIN inhibitor treatment resulted in reduced monocyte migration and reduced macrophage and vascular endothelial cell inflammatory and cytokine gene expression. In vivo, administration of an irreversible inhibitor of FURIN, α-1-PDX (α1-antitrypsin Portland), to hyperlipidemic Ldlr-/- mice resulted in lower atherosclerotic lesion area and a specific reduction in severe lesions. Significantly lower lesional macrophage and collagen area, as well as systemic inflammatory markers, were observed. MMP2 (matrix metallopeptidase 2), an effector of endothelial function and atherosclerotic lesion progression, and a FURIN substrate was significantly reduced in the aorta of inhibitor-treated mice. To determine FURIN's role in vascular endothelial function, we administered α-1-PDX to Apoe-/- mice harboring a wire injury in the common carotid artery. We observed significantly decreased carotid intimal thickness and lower plaque cellularity, smooth muscle cell, macrophage, and inflammatory marker content, suggesting protection against vascular remodeling. Overexpression of FURIN in this model resulted in a significant 67% increase in intimal plaque thickness, confirming that FURIN levels directly correlate with atherosclerosis. Conclusions- We show that systemic inhibition of FURIN in mice decreases vascular remodeling and atherosclerosis. FURIN-mediated modulation of MMP2 activity may contribute to the atheroprotection observed in these mice.


Subject(s)
Atherosclerosis/prevention & control , Furin/antagonists & inhibitors , Plaque, Atherosclerotic/drug therapy , alpha 1-Antitrypsin/therapeutic use , Animals , Aorta/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Artery, Common , Disease Progression , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Furin/genetics , Furin/physiology , Gene Expression Regulation/drug effects , Macrophages/physiology , Male , Matrix Metalloproteinase 2/analysis , Mice , Mice, Inbred C57BL , Monocytes/physiology , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular Remodeling , alpha 1-Antitrypsin/pharmacology
10.
FASEB J ; 32(7): 3792-3802, 2018 07.
Article in English | MEDLINE | ID: mdl-29481310

ABSTRACT

Bile acids (BAs) are surfactant molecules that regulate the intestinal absorption of lipids. Thus, the modulation of BAs represents a potential therapy for nonalcoholic fatty liver disease (NAFLD), which is characterized by hepatic accumulation of fat and is a major cause of liver disease worldwide. Cyp8b1 is a critical modulator of the hydrophobicity index of the BA pool. As a therapeutic proof of concept, we aimed to determine the impact of Cyp8b1 inhibition in vivo on BA pool composition and as protection against NAFLD. Inhibition of Cyp8b1 expression in mice led to a remodeling of the BA pool, which altered its signaling properties and decreased intestinal fat absorption. In a model of cholesterol-induced NAFLD, Cyp8b1 knockdown significantly decreased steatosis and hepatic lipid content, which has been associated with an increase in fecal lipid and BA excretion. Moreover, inhibition of Cyp8b1 not only decreased hepatic lipid accumulation, but also resulted in the clearance of previously accumulated hepatic cholesterol, which led to a regression in hepatic steatosis. Taken together, our data demonstrate that Cyp8b1 inhibition is a viable therapeutic target of crucial interest for metabolic diseases, such as NAFLD.-Chevre, R., Trigueros-Motos, L., Castaño, D., Chua, T., Corlianò, M., Patankar, J. V., Sng, L., Sim, L., Juin, T. L., Carissimo, G., Ng, L. F. P., Yi, C. N. J., Eliathamby, C. C., Groen, A. K., Hayden, M. R., Singaraja, R. R. Therapeutic modulation of the bile acid pool by Cyp8b1 knockdown protects against nonalcoholic fatty liver disease in mice.


Subject(s)
Bile Acids and Salts/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Steroid 12-alpha-Hydroxylase/genetics , Animals , Female , HEK293 Cells , Humans , Liver/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/therapy , RNAi Therapeutics , Steroid 12-alpha-Hydroxylase/metabolism
11.
Arterioscler Thromb Vasc Biol ; 37(11): 2147-2155, 2017 11.
Article in English | MEDLINE | ID: mdl-28882873

ABSTRACT

OBJECTIVE: High-density lipoproteins (HDL) are considered to protect against atherosclerosis in part by facilitating the removal of cholesterol from peripheral tissues. However, factors regulating lipid efflux are incompletely understood. We previously identified a variant in adenosine triphosphate-binding cassette transporter A8 (ABCA8) in an individual with low HDL cholesterol (HDLc). Here, we investigate the role of ABCA8 in cholesterol efflux and in regulating HDLc levels. APPROACH AND RESULTS: We sequenced ABCA8 in individuals with low and high HDLc and identified, exclusively in low HDLc probands, 3 predicted deleterious heterozygous ABCA8 mutations (p.Pro609Arg [P609R], IVS17-2 A>G and p.Thr741Stop [T741X]). HDLc levels were lower in heterozygous mutation carriers compared with first-degree family controls (0.86±0.34 versus 1.17±0.26 mmol/L; P=0.005). HDLc levels were significantly decreased by 29% (P=0.01) in Abca8b-/- mice on a high-cholesterol diet compared with wild-type mice, whereas hepatic overexpression of human ABCA8 in mice resulted in significant increases in plasma HDLc and the first steps of macrophage-to-feces reverse cholesterol transport. Overexpression of wild-type but not mutant ABCA8 resulted in a significant increase (1.8-fold; P=0.01) of cholesterol efflux to apolipoprotein AI in vitro. ABCA8 colocalizes and interacts with adenosine triphosphate-binding cassette transporter A1 and further potentiates adenosine triphosphate-binding cassette transporter A1-mediated cholesterol efflux. CONCLUSIONS: ABCA8 facilitates cholesterol efflux and modulates HDLc levels in humans and mice.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol, Dietary/blood , Cholesterol, HDL/blood , ATP-Binding Cassette Transporters/deficiency , ATP-Binding Cassette Transporters/genetics , Adult , Aged , Animals , Apolipoprotein A-I/blood , Apolipoprotein B-100/blood , Biological Transport , Biomarkers/blood , COS Cells , Case-Control Studies , Chlorocebus aethiops , DNA Mutational Analysis , Diet, High-Fat , Feces/chemistry , Female , HEK293 Cells , Heredity , Heterozygote , Humans , Liver/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mutation , Pedigree , Phenotype , Transfection
12.
Cell Death Differ ; 24(3): 433-444, 2017 03.
Article in English | MEDLINE | ID: mdl-27911442

ABSTRACT

Caspase-6 (CASP6) has an important role in axonal degeneration during neuronal apoptosis and in the neurodegenerative diseases Alzheimer and Huntington disease. Decreasing CASP6 activity may help to restore neuronal function in these and other diseases such as stroke and ischemia, where increased CASP6 activity has been implicated. The key to finding approaches to decrease CASP6 activity is a deeper understanding of the mechanisms regulating CASP6 activation. We show that CASP6 is posttranslationally palmitoylated by the palmitoyl acyltransferase HIP14 and that the palmitoylation of CASP6 inhibits its activation. Palmitoylation of CASP6 is decreased both in Hip14-/- mice, where HIP14 is absent, and in YAC128 mice, a model of Huntington disease, where HIP14 is dysfunctional and where CASP6 activity is increased. Molecular modeling suggests that palmitoylation of CASP6 may inhibit its activation via steric blockage of the substrate-binding groove and inhibition of CASP6 dimerization, both essential for CASP6 function. Our studies identify palmitoylation as a novel CASP6 modification and as a key regulator of CASP6 activity.


Subject(s)
Acyltransferases/metabolism , Caspase 6/metabolism , Acyltransferases/deficiency , Acyltransferases/genetics , Animals , Brain/metabolism , COS Cells , Caspase 6/genetics , Chlorocebus aethiops , Dimerization , Disease Models, Animal , Huntington Disease/metabolism , Huntington Disease/pathology , Immunoprecipitation , Lipoylation , Mice , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Protein Structure, Tertiary , Substrate Specificity
13.
J Lipid Res ; 56(10): 1993-2001, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26255038

ABSTRACT

A low level of HDL cholesterol (HDL-C) is a common clinical scenario and an important marker for increased cardiovascular risk. Many patients with very low or very high HDL-C have a rare mutation in one of several genes, but identification of the molecular abnormality in patients with extreme HDL-C is rarely performed in clinical practice. We investigated the accuracy and diagnostic yield of a targeted next-generation sequencing (NGS) assay for extreme levels of HDL-C. We developed a targeted NGS panel to capture the exons, intron/exon boundaries, and untranslated regions of 26 genes with highly penetrant effects on plasma lipid levels. We sequenced 141 patients with extreme HDL-C levels and prioritized variants in accordance with medical genetics guidelines. We identified 35 pathogenic and probably pathogenic variants in HDL genes, including 21 novel variants, and performed functional validation on a subset of these. Overall, a molecular diagnosis was established in 35.9% of patients with low HDL-C and 5.2% with high HDL-C, and all prioritized variants identified by NGS were confirmed by Sanger sequencing. Our results suggest that a molecular diagnosis can be identified in a substantial proportion of patients with low HDL-C using targeted NGS.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Cardiovascular Diseases/genetics , Cholesterol, HDL/blood , Cholesterol, HDL/genetics , High-Throughput Nucleotide Sequencing/methods , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , ATP Binding Cassette Transporter 1/blood , Alleles , Cardiovascular Diseases/blood , Cardiovascular Diseases/diagnosis , Exons , Female , Genetic Association Studies , Humans , Introns , Male , Middle Aged , Risk Factors
14.
Dev Biol ; 397(2): 257-66, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25478910

ABSTRACT

Huntington disease (HD) is an adult-onset neurodegenerative disease characterized by motor, cognitive, and psychiatric symptoms that is caused by a CAG expansion in the HTT gene. Palmitoylation is the addition of saturated fatty acids to proteins by DHHC palmitoylacyl transferases. HTT is palmitoylated by huntingtin interacting proteins 14 and 14-like (HIP14 and HIP14L or ZDHHC17 and 13 respectively). Mutant HTT is less palmitoylated and this reduction of palmitoylation accelerates its aggregation and increases cellular toxicity. Mouse models deficient in either Hip14 (Hip14(-/-)) or Hip14l (Hip14l(-/-)) develop HD-like phenotypes. The biological function of HTT palmitoylation and the role that loss of HTT palmitoylation plays in the pathogenesis of HD are unknown. To address these questions mice deficient for both genes were created. Loss of Hip14 and Hip14l leads to early embryonic lethality at day embryonic day 10-11 due to failed chorioallantoic fusion. The chorion is thickened and disorganized and the allantois does not fuse correctly with the chorion and forms a balloon-like shape compared to Hip14l(-/-); Hip14(+/+) littermate control embryos. Interestingly, the Hip14(-/-) ; Hip14(-/-) embryos share many features with the Htt(-/-) embryos, including folding of the yolk sac, a bulb shaped allantois, and a thickened and disorganized chorion. This may be due to a decrease in HTT palmitoylation. In Hip14(-/-); Hip14l(-/-) mouse embryonic fibroblasts show a 25% decrease in HTT palmitoylation compared to wild type cells. This is the first description of a double PAT deficient mouse model where loss of a PAT or multiple PATs results in embryonic lethality in mammals. These results reinforce the physiological importance of palmitoylation during embryogenesis.


Subject(s)
Acyltransferases/metabolism , Chorioallantoic Membrane/embryology , Membrane Fusion/genetics , Placenta/embryology , Serotonin Plasma Membrane Transport Proteins/metabolism , Acyltransferases/genetics , Animals , Blotting, Western , Female , Genotype , In Situ Hybridization , Lipoylation , Membrane Fusion/physiology , Mice , Mice, Knockout , Pregnancy , Real-Time Polymerase Chain Reaction
15.
Diabetes ; 64(4): 1168-79, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25338812

ABSTRACT

Besides their role in facilitating lipid absorption, bile acids are increasingly being recognized as signaling molecules that activate cell-signaling receptors. Targeted disruption of the sterol 12α-hydroxylase gene (Cyp8b1) results in complete absence of cholic acid (CA) and its derivatives. Here we investigate the effect of Cyp8b1 deletion on glucose homeostasis. Absence of Cyp8b1 results in improved glucose tolerance, insulin sensitivity, and ß-cell function, mediated by absence of CA in Cyp8b1(-/-) mice. In addition, we show that reduced intestinal fat absorption in the absence of biliary CA leads to increased free fatty acids reaching the ileal L cells. This correlates with increased secretion of the incretin hormone GLP-1. GLP-1, in turn, increases the biosynthesis and secretion of insulin from ß-cells, leading to the improved glucose tolerance observed in the Cyp8b1(-/-) mice. Thus, our data elucidate the importance of Cyp8b1 inhibition on the regulation of glucose metabolism.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Homeostasis/physiology , Insulin Resistance/physiology , Insulin/metabolism , Steroid 12-alpha-Hydroxylase/metabolism , Animals , Cholic Acid/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/genetics , Insulin-Secreting Cells/metabolism , Mice , Mice, Knockout , Steroid 12-alpha-Hydroxylase/genetics
16.
J Lipid Res ; 55(8): 1693-701, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24891332

ABSTRACT

While genetic determinants strongly influence HDL cholesterol (HDLc) levels, most genetic causes underlying variation in HDLc remain unknown. We aimed to identify novel rare mutations with large effects in candidate genes contributing to extreme HDLc in humans, utilizing family-based Mendelian genetics. We performed next-generation sequencing of 456 candidate HDLc-regulating genes in 200 unrelated probands with extremely low (≤10th percentile) or high (≥90th percentile) HDLc. Probands were excluded if known mutations existed in the established HDLc-regulating genes ABCA1, APOA1, LCAT, cholesteryl ester transfer protein (CETP), endothelial lipase (LIPG), and UDP-N-acetyl-α-D-galactosamine:polypeptide N-acetylgalactosaminyltransferase 2 (GALNT2). We identified 93 novel coding or splice-site variants in 72 candidate genes. Each variant was genotyped in the proband's family. Family-based association analyses were performed for variants with sufficient power to detect significance at P < 0.05 with a total of 627 family members being assessed. Mutations in the genes glucokinase regulatory protein (GCKR), RNase L (RNASEL), leukocyte immunoglobulin-like receptor 3 (LILRA3), and dynein axonemal heavy chain 10 (DNAH10) segregated with elevated HDLc levels in families, while no mutations associated with low HDLc. Taken together, we have identified mutations in four novel genes that may play a role in regulating HDLc levels in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Axonemal Dyneins/genetics , Cholesterol, HDL/blood , Endoribonucleases/genetics , Mutation , Receptors, Immunologic/genetics , ATP Binding Cassette Transporter 1/genetics , Adult , Aged , Apolipoprotein A-I/genetics , Cholesterol Ester Transfer Proteins/genetics , Cholesterol, HDL/genetics , Female , Humans , Lipase/genetics , Male , Middle Aged , N-Acetylgalactosaminyltransferases/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Polypeptide N-acetylgalactosaminyltransferase
17.
Hum Mol Genet ; 23(15): 4142-60, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24705354

ABSTRACT

HIP14 is the most highly conserved of 23 human palmitoyl acyltransferases (PATs) that catalyze the post-translational addition of palmitate to proteins, including huntingtin (HTT). HIP14 is dysfunctional in the presence of mutant HTT (mHTT), the causative gene for Huntington disease (HD), and we hypothesize that reduced palmitoylation of HTT and other HIP14 substrates contributes to the pathogenesis of the disease. Here we describe the yeast two-hybrid (Y2H) interactors of HIP14 in the first comprehensive study of interactors of a mammalian PAT. Unexpectedly, we discovered a highly significant overlap between HIP14 interactors and 370 published interactors of HTT, 4-fold greater than for control proteins (P = 8 × 10(-5)). Nearly half of the 36 shared interactors are already implicated in HD, supporting a direct link between HIP14 and the disease. The HIP14 Y2H interaction set is significantly enriched for palmitoylated proteins that are candidate substrates. We confirmed that three of them, GPM6A, and the Sprouty domain-containing proteins SPRED1 and SPRED3, are indeed palmitoylated by HIP14; the first enzyme known to palmitoylate these proteins. These novel substrates functions might be affected by reduced palmitoylation in HD. We also show that the vesicular cargo adapter optineurin, an established HTT-binding protein, co-immunoprecipitates with HIP14 but is not palmitoylated. mHTT leads to mislocalization of optineurin and aberrant cargo trafficking. Therefore, it is possible that optineurin regulates trafficking of HIP14 to its substrates. Taken together, our data raise the possibility that defective palmitoylation by HIP14 might be an important mechanism that contributes to the pathogenesis of HD.


Subject(s)
Acyltransferases/genetics , Adaptor Proteins, Signal Transducing/genetics , Huntington Disease/genetics , Nerve Tissue Proteins/genetics , Protein Processing, Post-Translational , Acyltransferases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , COS Cells , Cell Cycle Proteins , Chlorocebus aethiops , Gene Regulatory Networks , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipoylation , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Transport Proteins , Molecular Sequence Annotation , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Interaction Mapping , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Transcription Factor TFIIIA/genetics , Transcription Factor TFIIIA/metabolism , Two-Hybrid System Techniques
18.
Proc Natl Acad Sci U S A ; 110(50): 20296-301, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277827

ABSTRACT

Palmitoylation of neurotransmitter receptors and associated scaffold proteins regulates their membrane association in a rapid, reversible, and activity-dependent fashion. This makes palmitoylation an attractive candidate as a key regulator of the fast, reversible, and activity-dependent insertion of synaptic proteins required during the induction and expression of long-term plasticity. Here we describe that the constitutive loss of huntingtin interacting protein 14 (Hip14, also known as DHHC17), a single member of the broad palmitoyl acyltransferase (PAT) family, produces marked alterations in synaptic function in varied brain regions and significantly impairs hippocampal memory and synaptic plasticity. The data presented suggest that, even though the substrate pool is overlapping for the 23 known PAT family members, the function of a single PAT has marked effects upon physiology and cognition. Moreover, an improved understanding of the role of PATs in synaptic modification and maintenance highlights a potential strategy for intervention against early cognitive impairments in neurodegenerative disease.


Subject(s)
Acyltransferases/genetics , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Memory Disorders/genetics , Neuronal Plasticity/genetics , Synapses/genetics , Acyltransferases/metabolism , Analysis of Variance , Animals , Cell Count , Dendrites/ultrastructure , Hippocampus/cytology , Hippocampus/physiology , Lipoylation , Mice , Mice, Knockout , Neuronal Plasticity/physiology , Patch-Clamp Techniques , Synapses/physiology
19.
Chem Biol ; 20(11): 1421-34, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24211138

ABSTRACT

Protein palmitoylation, a reversible lipid modification of proteins, is widely used in the nervous system, with dysregulated palmitoylation being implicated in a variety of neurological disorders. Described below is ABE/SILAM, a proteomic strategy that couples acyl-biotinyl exchange (ABE) purification of palmitoyl-proteins to whole animal stable isotope labeling (SILAM) to provide an accurate tracking of palmitoylation change within rodent disease models. As a first application, we have used ABE/SILAM to look at Huntington's disease (HD), profiling palmitoylation change in two HD-relevant mouse mutants: the transgenic HD model mouse YAC128 and the hypomorphic Hip14-gt mouse, which has sharply reduced expression for HIP14 (Zdhhc17), a palmitoyl-transferase implicated in the HD disease process. Rather than mapping to the degenerating neurons themselves, the biggest disease changes instead map to astrocytes and oligodendrocytes (i.e., the supporting glial cells).


Subject(s)
Brain/metabolism , Brain/pathology , Disease Models, Animal , Huntington Disease/metabolism , Huntington Disease/pathology , Neuroglia/metabolism , Palmitic Acid/metabolism , Acyltransferases/genetics , Acyltransferases/metabolism , Animals , Huntington Disease/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
Hum Mol Genet ; 22(3): 452-65, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23077216

ABSTRACT

Palmitoylation, the dynamic post-translational addition of the lipid, palmitate, to proteins by Asp-His-His-Cys-containing palmitoyl acyltransferase (PAT) enzymes, modulates protein function and localization and plays a key role in the nervous system. Huntingtin-interacting protein 14 (HIP14), a well-characterized neuronal PAT, has been implicated in the pathogenesis of Huntington disease (HD), a fatal neurodegenerative disease associated with motor, psychiatric and cognitive symptoms, caused by a CAG expansion in the huntingtin gene (HTT). Mice deficient for Hip14 expression develop neuropathological and behavioural features similar to HD, and the catalytic activity of HIP14 is impaired in HD mice, most likely due to the reduced interaction of HIP14 with HTT. Huntingtin-interacting protein 14-like (HIP14L) is a paralog of HIP14, with identical domain structure. Together, HIP14 and HIP14L are the major PATs for HTT. Here, we report the characterization of a Hip14l-deficient mouse model, which develops adult-onset, widespread and progressive neuropathology accompanied by early motor deficits in climbing, impaired motor learning and reduced palmitoylation of a novel HIP14L substrate: SNAP25. Although the phenotype resembles that of the Hip14(-/-) mice, a more progressive phenotype, similar to that of the YAC128 transgenic mouse model of HD, is observed. In addition, HIP14L interacts less with mutant HTT than the wild-type protein, suggesting that reduced HIP14L-dependent palmitoylation of neuronal substrates may contribute to the pathogenesis of HD. Thus, both HIP14 and HIP14L may be dysfunctional in the disease.


Subject(s)
Acyltransferases/genetics , Disease Models, Animal , Gene Deletion , Huntington Disease/genetics , Neurons/pathology , Acyltransferases/deficiency , Acyltransferases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Female , Gene Expression Regulation , Huntingtin Protein , Huntington Disease/pathology , Immunoblotting , Learning/physiology , Lipoylation , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Sequence Analysis, DNA , Synaptosomal-Associated Protein 25/genetics , Synaptosomal-Associated Protein 25/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...