Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Clin Endocrinol Metab ; 106(9): e3473-e3482, 2021 08 18.
Article in English | MEDLINE | ID: mdl-34019660

ABSTRACT

CONTEXT: Severe hypertriglyceridemia (fasting triglycerides [TG] concentration ≥10 mmol/L) can be caused by multifactorial chylomicronemia syndrome (MCS) or familial chylomicronemia syndrome (FCS). Both conditions are associated with an increased risk of acute pancreatitis. The clinical differences between MCS patients with or without a rare variant in TG-related genes have never been studied. OBJECTIVE: To compare the clinical and biochemical characteristics of FCS, positive-MCS patients, and negative-MCS patients, as well as to investigate the predictors of acute pancreatitis in MCS patients. METHODS: All patients referred at the clinic for severe hypertriglyceridemia underwent genetic testing for the 5 canonical genes involved in TG metabolism (LPL, APOC2, GPIHBP1, APOA5, and LMF1) using next-generation sequencing. RESULTS: A total of 53 variant negative-MCS, 22 variant positive-MCS and 28 FCS subjects were included in this retrospective cross-sectional study. A significant difference was observed in the prevalence of pancreatitis (9%, 41%, and 61%) and multiple pancreatitis (6%, 23%, and 46%) in the negative-MCS, the positive-MCS, and the FCS groups, respectively (P < 0.0001). Predictors of pancreatitis among MCS subjects included the presence of a rare variant, lower apolipoprotein B, as well as higher gamma-glutamyl transferase, maximal TG value, and fructose consumption. CONCLUSION: We observed that the MCS individuals who carried a rare variant have an intermediate phenotype between FCS and negative-MCS subjects. Since novel molecules such as the antisense oligonucleotide against APOC3 mRNA showed high efficacy in reducing TG levels in patients with multifactorial chylomicronemia, identification of higher-risk MCS patients who would benefit from additional treatment is essential.


Subject(s)
Hyperlipoproteinemia Type I/epidemiology , Hyperlipoproteinemia Type I/genetics , Pancreatitis/epidemiology , Pancreatitis/genetics , Triglycerides/genetics , Adult , Cross-Sectional Studies , Female , Genetic Testing , Genetic Therapy , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/classification , Hypertriglyceridemia/genetics , Male , Middle Aged , Predictive Value of Tests , Prevalence , Retrospective Studies , Young Adult
2.
Nat Genet ; 53(2): 128-134, 2021 02.
Article in English | MEDLINE | ID: mdl-33495596

ABSTRACT

The heart muscle diseases hypertrophic (HCM) and dilated (DCM) cardiomyopathies are leading causes of sudden death and heart failure in young, otherwise healthy, individuals. We conducted genome-wide association studies and multi-trait analyses in HCM (1,733 cases), DCM (5,521 cases) and nine left ventricular (LV) traits (19,260 UK Biobank participants with structurally normal hearts). We identified 16 loci associated with HCM, 13 with DCM and 23 with LV traits. We show strong genetic correlations between LV traits and cardiomyopathies, with opposing effects in HCM and DCM. Two-sample Mendelian randomization supports a causal association linking increased LV contractility with HCM risk. A polygenic risk score explains a significant portion of phenotypic variability in carriers of HCM-causing rare variants. Our findings thus provide evidence that polygenic risk score may account for variability in Mendelian diseases. More broadly, we provide insights into how genetic pathways may lead to distinct disorders through opposing genetic effects.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Hypertrophic/mortality , Cardiomyopathy, Hypertrophic/physiopathology , Case-Control Studies , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Heart Ventricles/physiopathology , Humans , Kaplan-Meier Estimate , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Ventricular Function, Left/genetics
3.
Am J Med Genet A ; 182(10): 2359-2368, 2020 10.
Article in English | MEDLINE | ID: mdl-32808748

ABSTRACT

Rare loss of function variants in DSP, which codes for the desmosomal protein desmoplakin, have been implicated in dilated and arrhythmogenic right ventricular cardiomyopathies. We present a family with arrhythmogenic cardiomyopathy associated with a novel missense variant in DSP (NM_004415.4): c.877G>A, p.(Glu293Lys). The phenotype is characterized by predominant involvement of the left ventricle with systolic dysfunction, fibrosis, and life-threatening arrhythmias. We performed a systematic review of literature collecting all cardiomyopathy cases with rare missense variants in DSP. We demonstrate that the distribution of missense variants across the protein domains in cardiomyopathy cases differs from that in gnomAD (p = .04), with a case enrichment of rare missense variants in the spectrin repeat domain (36/78 [46%] in cases vs. 449/1495 [30%] in gnomAD; p = .004). Our findings highlight the predominance of cardiac arrhythmia and left ventricular involvement in desmoplakin cardiomyopathy and pinpoint to a potential mutation hotspot in DSP thereby facilitating missense variant interpretation in the diagnostic setting.


Subject(s)
Arrhythmias, Cardiac/genetics , Arrhythmogenic Right Ventricular Dysplasia/genetics , Desmoplakins/genetics , Genetic Predisposition to Disease , Arrhythmias, Cardiac/pathology , Arrhythmogenic Right Ventricular Dysplasia/pathology , Female , Genetic Variation , Heart Ventricles/pathology , Humans , Male , Mutation, Missense/genetics , Phenotype
4.
Pharmacotherapy ; 35(6): 600-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26095008

ABSTRACT

STUDY OBJECTIVE: To evaluate the pharmacokinetic and pharmacodynamic profiles of piperacillin-tazobactam administered as a 4-hour infusion in critically ill patients undergoing continuous renal replacement therapy (CRRT). DESIGN: Prospective, observational, pharmacokinetic study. SETTING: Intensive care unit of a tertiary care hospital in Montréal, Canada. PATIENTS: Twenty critically ill adults who were undergoing continuous venovenous hemodiafiltration and receiving a 4-hour infusion of piperacillin 4 g-tazobactam 0.5 g every 8 hours for a documented or suspected infection. INTERVENTION: Blood samples were collected every hour over an 8-hour dosing interval. Prefilter and postfilter blood samples, and effluent and urine samples were also collected. MEASUREMENTS AND MAIN RESULTS: The primary outcome was the proportion of patients who achieved an unbound piperacillin plasma concentration above a target minimum inhibitory concentration (MIC) of 64 mg/L (MIC that inhibits 90% of isolates for Pseudomonas aeruginosa) for at least 50% of the dosing interval; 18 (90%) of the 20 patients achieved this outcome. In all patients, the free piperacillin concentrations were above the Pseudomonas aeruginosa breakpoint of 16 mg/L for the entire time interval. Regarding piperacillin pharmacokinetic parameters, the median (interquartile range) minimum unbound plasma concentration was 65.15 mg/L (51.30-89.30), maximum unbound plasma concentration was 141.3 mg/L (116.75-173.90), sieving coefficient was 0.809 (0.738-0.938), total clearance was 65.82 ml/minute (53.79-102.87), and renal clearance was 0.16 ml/minute (0.05-3.04). The median CRRT dose was 32.0 ml/kg/h (25.0-39.8). CONCLUSIONS: Administration of a 4-hour infusion of piperacillin-tazobactam was associated with a favorable pharmacodynamic profile in patients undergoing CRRT. Concentrations associated with maximal activity were attained in our patients.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Hemodiafiltration , Penicillanic Acid/analogs & derivatives , Aged , Critical Illness , Female , Humans , Infusions, Intravenous , Male , Microbial Sensitivity Tests , Middle Aged , Penicillanic Acid/pharmacokinetics , Piperacillin/pharmacokinetics , Piperacillin, Tazobactam Drug Combination , Prospective Studies , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/drug effects , Tertiary Care Centers
5.
PLoS One ; 7(4): e36200, 2012.
Article in English | MEDLINE | ID: mdl-22558381

ABSTRACT

BACKGROUND: Type 2 diabetes is characterized by pancreatic ß-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that the signalling cascade activated by lipopolysaccharides (LPS) binding to Toll-Like Receptor 4 (TLR4) exerts deleterious effects on pancreatic ß-cell function; however, the molecular mechanisms of these effects are incompletely understood. In this study, we tested the hypothesis that LPS alters insulin gene expression via TLR4 and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) in islets. METHODOLOGY/PRINCIPAL FINDINGS: A 24-h exposure of isolated human, rat and mouse islets of Langerhans to LPS dose-dependently reduced insulin gene expression. This was associated in mouse and rat islets with decreased mRNA expression of pancreas-duodenum homebox-1 (PDX-1) and mammalian homologue of avian MafA/l-Maf (MafA). Accordingly, LPS exposure also decreased glucose-induced insulin secretion. LPS repression of insulin, PDX-1 and MafA expression, as well as its inhibition of insulin secretion, were not observed in islets from TLR4-deficient mice. LPS inhibition of ß-cell gene expression in rat islets was prevented by inhibition of the NF-κB pathway, but not the p38 mitogen-activated protein kinase (p38 MAPK) pathway. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that LPS inhibit ß-cell gene expression in a TLR4-dependent manner and via NF-κB signaling in pancreatic islets, suggesting a novel mechanism by which the gut microbiota might affect pancreatic ß-cell function.


Subject(s)
Gene Expression Regulation/drug effects , Insulin/genetics , Islets of Langerhans/drug effects , Lipopolysaccharides/pharmacology , NF-kappa B/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Homeodomain Proteins/metabolism , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Male , Mice , NF-kappa B/antagonists & inhibitors , RNA Precursors/genetics , RNA Precursors/metabolism , Rats , Toll-Like Receptor 4/deficiency , Trans-Activators/metabolism
6.
J Mol Endocrinol ; 47(3): 273-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21821716

ABSTRACT

Pancreatic ß-cells have a well-developed endoplasmic reticulum due to their highly specialized secretory function to produce insulin in response to glucose and nutrients. It has been previously reported that overexpression of activating transcription factor 6 (ATF6) reduces insulin gene expression in part via upregulation of small heterodimer partner. In this study, we investigated whether ATF6 directly binds to the insulin gene promoter, and whether its direct binding represses insulin gene promoter activity. A bioinformatics analysis identified a putative ATF6 binding site in the A5/Core region of the rat insulin II gene promoter. Direct binding of ATF6 was confirmed using several approaches. Electrophoretic mobility shift assays in nuclear extracts from MCF7 cells, isolated rat islets and insulin-secreting HIT-T15 cells showed ATF6 binding to the native A5/Core of the rat insulin II gene promoter. Antibody-mediated supershift analyses revealed the presence of both ATF6 isoforms, ATF6α and ATF6ß, in the complex. Chromatin immunoprecipitation assays confirmed the binding of ATF6α and ATF6ß to a region encompassing the A5/Core of the rat insulin II gene promoter in isolated rat islets. Overexpression of the active (cleaved) fragment of ATF6α, but not ATF6ß, inhibited the activity of an insulin promoter-reporter by 50%. However, the inhibitory effect of ATF6α was insensitive to mutational inactivation or deletion of the A5/Core. Therefore, although ATF6 binds directly to the A5/Core of the rat insulin II gene promoter, this direct binding does not appear to contribute to its repressive activity.


Subject(s)
Activating Transcription Factor 6/metabolism , Insulin/genetics , Promoter Regions, Genetic , Transcription, Genetic , Activating Transcription Factor 6/genetics , Animals , Base Sequence , Binding Sites/genetics , Consensus Sequence/genetics , Gene Expression , Gene Expression Regulation/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lactones/pharmacology , Male , Molecular Sequence Data , RNA Precursors/metabolism , RNA, Messenger/metabolism , Rats , Sequence Alignment , Sesquiterpenes/pharmacology
7.
Biochim Biophys Acta ; 1801(3): 289-98, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19715772

ABSTRACT

The concept of glucolipotoxicity refers to the combined, deleterious effects of elevated glucose and fatty acid levels on pancreatic beta-cell function and survival. Significant progress has been made in recent years towards a better understanding of the cellular and molecular basis of glucolipotoxicity in the beta cell. The permissive effect of elevated glucose on the detrimental actions of fatty acids stems from the influence of glucose on intracellular fatty acid metabolism, promoting the synthesis of cellular lipids. The combination of excessive levels of fatty acids and glucose therefore leads to decreased insulin secretion, impaired insulin gene expression, and beta-cell death by apoptosis, all of which probably have distinct underlying mechanisms. Recent studies from our laboratory have identified several pathways implicated in fatty acid inhibition of insulin gene expression, including the extracellular-regulated kinase (ERK1/2) pathway, the metabolic sensor Per-Arnt-Sim kinase (PASK), and the ATF6 branch of the unfolded protein response. We have also confirmed in vivo in rats that the decrease in insulin gene expression is an early defect which precedes any detectable abnormality in insulin secretion. While the role of glucolipotoxicity in humans is still debated, the inhibitory effects of chronically elevated fatty acid levels has been clearly demonstrated in several studies, at least in individuals genetically predisposed to developing type 2 diabetes. It is therefore likely that glucolipotoxicity contributes to beta-cell failure in type 2 diabetes as well as to the decline in beta-cell function observed after the onset of the disease.


Subject(s)
Fatty Acids/metabolism , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Lipid Metabolism Disorders/metabolism , Animals , Female , Humans , Insulin Resistance , Male , Obesity/metabolism
8.
Diabetes ; 57(2): 424-31, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17991758

ABSTRACT

OBJECTIVE: Prolonged exposure of isolated islets of Langerhans to elevated levels of fatty acids, in the presence of high glucose, impairs insulin gene expression via a transcriptional mechanism involving nuclear exclusion of pancreas-duodenum homeobox-1 (Pdx-1) and loss of MafA expression. Whether such a phenomenon also occurs in vivo is unknown. Our objective was therefore to ascertain whether chronic nutrient oversupply inhibits insulin gene expression in vivo. RESEARCH DESIGN AND METHODS: Wistar rats received alternating 4-h infusions of glucose and Intralipid for a total of 72 h. Control groups received alternating infusions of glucose and saline, saline and Intralipid, or saline only. Insulin and C-peptide secretion were measured under hyperglycemic clamps. Insulin secretion and gene expression were assessed in isolated islets, and beta-cell mass was quantified by morphometric analysis. RESULTS: Neither C-peptide secretion nor insulin sensitivity was different among infusion regimens. Insulin content and insulin mRNA levels were lower in islets isolated from rats infused with glucose plus Intralipid. This was associated with reduced Pdx-1 binding to the endogenous insulin promoter, and an increased proportion of Pdx-1 localized in the cytoplasm versus the nucleus. In contrast, MafA mRNA and protein levels and beta-cell mass and proliferation were unchanged. CONCLUSIONS: Cyclical and alternating infusions of glucose and Intralipid in normal rats inhibit insulin gene expression without affecting insulin secretion or beta-cell mass. We conclude that fatty acid inhibition of insulin gene expression, in the presence of high glucose, is an early functional defect that may contribute to beta-cell failure in type 2 diabetes.


Subject(s)
Fat Emulsions, Intravenous/pharmacology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Homeodomain Proteins/metabolism , Insulin/genetics , Trans-Activators/metabolism , Animals , Blood Glucose/metabolism , C-Peptide/drug effects , C-Peptide/metabolism , Fat Emulsions, Intravenous/administration & dosage , Fatty Acids, Nonesterified/blood , Glucose/administration & dosage , Glucose Tolerance Test , Homeodomain Proteins/drug effects , Hyperglycemia , Infusions, Intravenous , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , RNA, Messenger/genetics , Rats , Rats, Wistar , Trans-Activators/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...