Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 40(8): 1935-1941, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32580631

RESUMEN

OBJECTIVE: Genetic determinants of severe hypertriglyceridemia include both common variants with small effects (assessed using polygenic risk scores) plus heterozygous and homozygous rare variants in canonical genes directly affecting triglyceride metabolism. Here, we broadened our scope to detect associations with rare loss-of-function variants in genes affecting noncanonical pathways, including those known to affect triglyceride metabolism indirectly. Approach and Results: From targeted next-generation sequencing of 69 metabolism-related genes in 265 patients of European descent with severe hypertriglyceridemia (≥10 mmol/L or ≥885 mg/dL) and 477 normolipidemic controls, we focused on the association of rare heterozygous loss-of-function variants in individual genes. We observed that compared with controls, severe hypertriglyceridemia patients were 20.2× (95% CI, 1.11-366.1; P=0.03) more likely than controls to carry a rare loss-of-function variant in CREB3L3, which encodes a transcription factor that regulates several target genes with roles in triglyceride metabolism. CONCLUSIONS: Our findings indicate that rare variants in a noncanonical gene for triglyceride metabolism, namely CREB3L3, contribute significantly to severe hypertriglyceridemia. Secondary genes and pathways should be considered when evaluating the genetic architecture of this complex trait.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Hipertrigliceridemia/genética , Adulto , Anciano , Apolipoproteína A-V/genética , Femenino , Heterocigoto , Humanos , Lipoproteína Lipasa/genética , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Triglicéridos/metabolismo
2.
Europace ; 23(6): 844-850, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33682005

RESUMEN

AIMS: Atrial fibrillation (AF) is a complex heritable disease whose genetic underpinnings remain largely unexplained, though recent work has suggested that the arrhythmia may develop secondary to an underlying atrial cardiomyopathy. We sought to evaluate for enrichment of loss-of-function (LOF) and copy number variants (CNVs) in genes implicated in ventricular cardiomyopathy in 'lone' AF. METHODS AND RESULTS: Whole-exome sequencing was performed in 255 early onset 'lone' AF cases, defined as arrhythmia onset prior to 60 years of age in the absence of known clinical risk factors. Subsequent evaluations were restricted to 195 cases of European genetic ancestry, as defined by principal component analysis, and focused on a pre-defined set of 43 genes previously implicated in ventricular cardiomyopathy. Bioinformatic analysis identified 6 LOF variants (3.1%), including 3 within the TTN gene, among cases in comparison with 4 of 503 (0.80%) controls [odds ratio: 3.96; 95% confidence interval (CI): 1.11-14.2; P = 0.033]. Further, two AF cases possessed a novel heterozygous 8521 base pair TTN deletion, confirmed with Sanger sequencing and breakpoint validation, which was absent from 4958 controls (P = 0.0014). Subsequent cascade screening in two families revealed evidence of co-segregation of a LOF variant with 'lone' AF. CONCLUSION: 'Lone' AF cases are enriched in rare LOF variants from cardiomyopathy genes, findings primarily driven by TTN, and a novel TTN deletion, providing additional evidence to implicate atrial cardiomyopathy as an AF genetic sub-phenotype. Our results also highlight that AF may develop in the context of these variants in the absence of a discernable ventricular cardiomyopathy.


Asunto(s)
Fibrilación Atrial , Cardiomiopatías , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/genética , Cardiomiopatías/diagnóstico , Cardiomiopatías/genética , Variaciones en el Número de Copia de ADN , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Fenotipo
3.
J Lipid Res ; 60(11): 1953-1958, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31519763

RESUMEN

Severe hypertriglyceridemia (HTG) is a relatively common form of dyslipidemia with a complex pathophysiology and serious health complications. HTG can develop in the presence of rare genetic factors disrupting genes involved in the triglyceride (TG) metabolic pathway, including large-scale copy-number variants (CNVs). Improvements in next-generation sequencing technologies and bioinformatic analyses have better allowed assessment of CNVs as possible causes of or contributors to severe HTG. We screened targeted sequencing data of 632 patients with severe HTG and identified partial deletions of the LPL gene, encoding the central enzyme involved in the metabolism of TG-rich lipoproteins, in four individuals (0.63%). We confirmed the genomic breakpoints in each patient with Sanger sequencing. Three patients carried an identical heterozygous deletion spanning the 5' untranslated region (UTR) to LPL exon 2, and one patient carried a heterozygous deletion spanning the 5'UTR to LPL exon 1. All four heterozygous CNV carriers were determined to have multifactorial severe HTG. The predicted null nature of our identified LPL deletions may contribute to relatively higher TG levels and a more severe clinical phenotype than other forms of genetic variation associated with the disease, particularly in the polygenic state. The identification of novel CNVs in patients with severe HTG suggests that methods for CNV detection should be included in the diagnostic workup and molecular genetic evaluation of patients with high TG levels.


Asunto(s)
Variaciones en el Número de Copia de ADN , Eliminación de Gen , Hipertrigliceridemia/genética , Lipoproteína Lipasa/genética , Biología Computacional , Análisis Mutacional de ADN , Exones , Humanos , Hipertrigliceridemia/metabolismo , Lipoproteína Lipasa/deficiencia , Lipoproteína Lipasa/metabolismo
4.
Can J Neurol Sci ; 46(5): 491-498, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31217043

RESUMEN

BACKGROUND/OBJECTIVE: Apolipoprotein E (APOE) E4 is the main genetic risk factor for Alzheimer's disease (AD). Due to the consistent association, there is interest as to whether E4 influences the risk of other neurodegenerative diseases. Further, there is a constant search for other genetic biomarkers contributing to these phenotypes, such as microtubule-associated protein tau (MAPT) haplotypes. Here, participants from the Ontario Neurodegenerative Disease Research Initiative were genotyped to investigate whether the APOE E4 allele or MAPT H1 haplotype are associated with five neurodegenerative diseases: (1) AD and mild cognitive impairment (MCI), (2) amyotrophic lateral sclerosis, (3) frontotemporal dementia (FTD), (4) Parkinson's disease, and (5) vascular cognitive impairment. METHODS: Genotypes were defined for their respective APOE allele and MAPT haplotype calls for each participant, and logistic regression analyses were performed to identify the associations with the presentations of neurodegenerative diseases. RESULTS: Our work confirmed the association of the E4 allele with a dose-dependent increased presentation of AD, and an association between the E4 allele alone and MCI; however, the other four diseases were not associated with E4. Further, the APOE E2 allele was associated with decreased presentation of both AD and MCI. No associations were identified between MAPT haplotype and the neurodegenerative disease cohorts; but following subtyping of the FTD cohort, the H1 haplotype was significantly associated with progressive supranuclear palsy. CONCLUSION: This is the first study to concurrently analyze the association of APOE isoforms and MAPT haplotypes with five neurodegenerative diseases using consistent enrollment criteria and broad phenotypic analysis.


Étude de variance génétique dans le cadre de l'initiative de recherche sur les maladies neurodégénératives en Ontario. Contexte/Objectif : L'apolipoprotéine E4 (ApoE4) constitue le principal facteur de risque génétique de la maladie d'Alzheimer. En raison de cette association systématique, il existe un intérêt certain à savoir dans quelle mesure cette classe d'apolipoprotéines peut influencer le risque d'autres maladies neurodégénératives. En outre, le milieu de la recherche n'a de cesse d'identifier d'autres biomarqueurs génétiques, par exemple les haplotypes H1 de la protéine tau associée aux microtubules, qui contribuent à certains phénotypes, Dans le cadre de cette étude, des participants à l'initiative de recherche sur les maladies neurodégénératives en Ontario ont été « génotypés ¼ afin de déterminer si l'ApoE4 ou l'haplotype H1 mentionné ci-dessus peuvent être associés à cinq maladies neurodégénératives : 1) la maladie d'Alzheimer et d'autres troubles cognitifs légers ; 2) la sclérose latérale amyotrophique ; 3) la démence fronto-temporale ; 4) la maladie de Parkinson ; 5) et finalement les déficits cognitifs d'origine vasculaire. Méthodes : Pour chaque participant, la cartographie des génotypes a été établie en fonction de leur ApoE4 respectif et de la présence d'haplotypes H1 de la protéine tau associée aux microtubules. Des analyses de régression logistique ont été ensuite effectuées dans le but d'identifier de possibles liens avec ces maladies neurodégénératives. Résultats : Nos travaux ont confirmé l'association entre l'ApoE4 et une plus grande occurrence de cas d'Alzheimer, et ce, en tenant compte de l'effet d'une dose de médicament. Ils ont aussi montré une association entre la seule ApoE4 et des troubles cognitifs légers. Cela dit, il convient de préciser que les quatre autres maladies n'ont pas été associées à cet allèle. Plus encore, nous avons trouvé que l'allèle E2 de l'apolipoprotéine était associé à une occurrence plus faible de cas d'Alzheimer et de troubles cognitifs légers. Fait à souligner, aucune association n'a été détectée entre l'haplotype H1 de la protéine tau associée aux microtubules et nos cohortes atteintes de maladies neurodégénératives. Toutefois, à la suite du sous-typage de la cohorte de participants atteints de démence fronto-temporale, il s'est avéré que l'haplotype H1 était associé de façon notable à la paralysie supra-nucléaire progressive. Conclusion : Il s'agit de la première étude à analyser simultanément, au moyen de critères de participation cohérents et d'une analyse phénotypique élargie, les associations entre les isoformes de l'ApoE, l'haplotype H1 de la protéine tau associée aux microtubules et cinq maladies neurodégénératives.


Asunto(s)
Apolipoproteínas E/genética , Predisposición Genética a la Enfermedad/genética , Enfermedades Neurodegenerativas/genética , Proteínas tau/genética , Anciano , Apolipoproteína E4/genética , Femenino , Variación Genética , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Ontario
5.
Lipids Health Dis ; 17(1): 48, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540175

RESUMEN

BACKGROUND: Abetalipoproteinemia and homozygous hypobetalipoproteinemia are classical Mendelian autosomal recessive and co-dominant conditions, respectively, which are phenotypically similar and are usually caused by bi-allelic mutations in MTTP and APOB genes, respectively. Instances of more complex patterns of genomic variants resulting in this distinct phenotype have not been reported. METHODS: A 43 year-old male had a longstanding severe deficiency of apolipoprotein (apo) B-containing lipoproteins and circulating fat soluble vitamins consistent with either abetalipoproteinemia or homozygous familial hypobetalipoproteinemia (FHBL). He also had acanthocytosis, a long term history of fat malabsorption, and mild retinopathy, but was free from coagulopathy, myopathy and neuropathy. He had taken high dose oral fat soluble vitamins since childhood. RESULTS: Targeted next generation DNA sequencing revealed several rare heterozygous missense variants in both MTTP and APOB genes known or predicted to be deleterious, in addition to a novel heterozygous missense variant in SAR1B, which encodes the gene causing chylomicron retention disease. Evaluation of first degree relatives with mild FHBL clarified the segregation of variants. CONCLUSIONS: The proband's characteristic phenotype likely resulted from an oligogenic interaction involving multiple rare variants in MTTP and APOB, and related genes, each of which individually was associated with a milder or minimal clinical and biochemical phenotype.


Asunto(s)
Apolipoproteína B-100/genética , Proteínas Portadoras/genética , Proteínas de Unión al GTP Monoméricas/genética , Adulto , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Hipobetalipoproteinemias , Masculino , Mutación Missense/genética
6.
J Lipid Res ; 58(11): 2202-2209, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28874442

RESUMEN

Familial hypercholesterolemia (FH) is a heritable condition of severely elevated LDL cholesterol, caused predominantly by autosomal codominant mutations in the LDL receptor gene (LDLR). In providing a molecular diagnosis for FH, the current procedure often includes targeted next-generation sequencing (NGS) panels for the detection of small-scale DNA variants, followed by multiplex ligation-dependent probe amplification (MLPA) in LDLR for the detection of whole-exon copy number variants (CNVs). The latter is essential because ∼10% of FH cases are attributed to CNVs in LDLR; accounting for them decreases false negative findings. Here, we determined the potential of replacing MLPA with bioinformatic analysis applied to NGS data, which uses depth-of-coverage analysis as its principal method to identify whole-exon CNV events. In analysis of 388 FH patient samples, there was 100% concordance in LDLR CNV detection between these two methods: 38 reported CNVs identified by MLPA were also successfully detected by our NGS method, while 350 samples negative for CNVs by MLPA were also negative by NGS. This result suggests that MLPA can be removed from the routine diagnostic screening for FH, significantly reducing associated costs, resources, and analysis time, while promoting more widespread assessment of this important class of mutations across diagnostic laboratories.


Asunto(s)
Variaciones en el Número de Copia de ADN , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hiperlipoproteinemia Tipo II/genética , Receptores de LDL/genética , Biología Computacional , Femenino , Humanos , Masculino , Persona de Mediana Edad
7.
J Lipid Res ; 58(11): 2162-2170, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28870971

RESUMEN

HDL cholesterol (HDL-C) remains a superior biochemical predictor of CVD risk, but its genetic basis is incompletely defined. In patients with extreme HDL-C concentrations, we concurrently evaluated the contributions of multiple large- and small-effect genetic variants. In a discovery cohort of 255 unrelated lipid clinic patients with extreme HDL-C levels, we used a targeted next-generation sequencing panel to evaluate rare variants in known HDL metabolism genes, simultaneously with common variants bundled into a polygenic trait score. Two additional cohorts were used for validation and included 1,746 individuals from the Montréal Heart Institute Biobank and 1,048 individuals from the University of Pennsylvania. Findings were consistent between cohorts: we found rare heterozygous large-effect variants in 18.7% and 10.9% of low- and high-HDL-C patients, respectively. We also found common variant accumulation, indicated by extreme polygenic trait scores, in an additional 12.8% and 19.3% of overall cases of low- and high-HDL-C extremes, respectively. Thus, the genetic basis of extreme HDL-C concentrations encountered clinically is frequently polygenic, with contributions from both rare large-effect and common small-effect variants. Multiple types of genetic variants should be considered as contributing factors in patients with extreme dyslipidemia.


Asunto(s)
HDL-Colesterol/sangre , HDL-Colesterol/genética , Genotipo , Adulto , Anciano , Femenino , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad
8.
J Lipid Res ; 58(11): 2188-2196, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28887372

RESUMEN

Recent cell culture and animal studies have suggested that expression of human apo C-III in the liver has a profound impact on the triacylglycerol (TAG)-rich VLDL1 production under lipid-rich conditions. The apoC-III Gln38Lys variant was identified in subjects of Mexican origin with moderate hypertriglyceridemia. We postulated that Gln38Lys (C3QK), being a gain-of-function mutation, promotes hepatic VLDL1 assembly/secretion. To test this hypothesis, we expressed C3QK in McA-RH7777 cells and apoc3-null mice to contrast its effect with WT apoC-III (C3WT). In both model systems, C3QK expression increased the secretion of VLDL1-TAG (by 230%) under lipid-rich conditions. Metabolic labeling experiments with C3QK cells showed an increase in de novo lipogenesis (DNL). Fasting plasma concentration of TAG, cholesterol, cholesteryl ester, and FA were increased in C3QK mice as compared with C3WT mice. Liver of C3QK mice also displayed an increase in DNL and expression of lipogenic genes as compared with that in C3WT mice. These results suggest that C3QK variant is a gain-of-function mutation that can stimulate VLDL1 production, through enhanced DNL.


Asunto(s)
Apolipoproteína C-III/genética , Mutación con Ganancia de Función , Hipertrigliceridemia/genética , Animales , Apolipoproteína C-III/deficiencia , Línea Celular , Eliminación de Gen , Regulación de la Expresión Génica , Humanos , Hipertrigliceridemia/metabolismo , Lipogénesis/genética , Lipoproteínas HDL/metabolismo , Masculino , Ratones
10.
Arterioscler Thromb Vasc Biol ; 36(12): 2439-2445, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27765764

RESUMEN

OBJECTIVE: Next-generation sequencing technology is transforming our understanding of heterozygous familial hypercholesterolemia, including revision of prevalence estimates and attribution of polygenic effects. Here, we examined the contributions of monogenic and polygenic factors in patients with severe hypercholesterolemia referred to a specialty clinic. APPROACH AND RESULTS: We applied targeted next-generation sequencing with custom annotation, coupled with evaluation of large-scale copy number variation and polygenic scores for raised low-density lipoprotein cholesterol in a cohort of 313 individuals with severe hypercholesterolemia, defined as low-density lipoprotein cholesterol >5.0 mmol/L (>194 mg/dL). We found that (1) monogenic familial hypercholesterolemia-causing mutations detected by targeted next-generation sequencing were present in 47.3% of individuals; (2) the percentage of individuals with monogenic mutations increased to 53.7% when copy number variations were included; (3) the percentage further increased to 67.1% when individuals with extreme polygenic scores were included; and (4) the percentage of individuals with an identified genetic component increased from 57.0% to 92.0% as low-density lipoprotein cholesterol level increased from 5.0 to >8.0 mmol/L (194 to >310 mg/dL). CONCLUSIONS: In a clinically ascertained sample with severe hypercholesterolemia, we found that most patients had a discrete genetic basis detected using a comprehensive screening approach that includes targeted next-generation sequencing, an assay for copy number variations, and polygenic trait scores.


Asunto(s)
Hiperlipoproteinemia Tipo II/genética , Herencia Multifactorial , Mutación , Adulto , Anciano , Biomarcadores/sangre , LDL-Colesterol/sangre , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Femenino , Dosificación de Gen , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Herencia , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/diagnóstico , Masculino , Persona de Mediana Edad , Ontario , Fenotipo , Valor Predictivo de las Pruebas , Factores de Riesgo , Índice de Severidad de la Enfermedad
12.
Arterioscler Thromb Vasc Biol ; 35(1): 213-21, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25395619

RESUMEN

OBJECTIVE: Estrogen deficiency is linked with increased low-density lipoprotein (LDL) cholesterol. The hormone receptor mediating this effect is unknown. G-protein estrogen receptor (GPER) is a recently recognized G-protein-coupled receptor that is activated by estrogens. We recently identified a common hypofunctional missense variant of GPER, namely P16L. However, the role of GPER in LDL metabolism is unknown. Therefore, we examined the association of the P16L genotype with plasma LDL cholesterol level. Furthermore, we studied the role of GPER in regulating expression of the LDL receptor and proprotein convertase subtilisin kexin type 9. APPROACH AND RESULTS: Our discovery cohort was a genetically isolated population of Northern European descent, and our validation cohort consisted of normal, healthy women aged 18 to 56 years from London, Ontario. In addition, we examined the effect of GPER on the regulation of proprotein convertase subtilisin kexin type 9 and LDL receptor expression by the treatment with the GPER agonist, G1. In the discovery cohort, GPER P16L genotype was associated with a significant increase in LDL cholesterol (mean±SEM): 3.18±0.05, 3.25±0.08, and 4.25±0.33 mmol/L, respectively, in subjects with CC (homozygous for P16), CT (heterozygotes), and TT (homozygous for L16) genotypes (P<0.05). In the validation cohort (n=339), the GPER P16L genotype was associated with a similar increase in LDL cholesterol: 2.17±0.05, 2.34±0.06, and 2.42±0.16 mmol/L, respectively, in subjects with CC, CT, and TT genotypes (P<0.05). In the human hepatic carcinoma cell line, the GPER agonist, G1, mediated a concentration-dependent increase in LDL receptor expression, blocked by either pretreatment with the GPER antagonist G15 or by shRNA-mediated GPER downregulation. G1 also mediated a GPER- and concentration-dependent decrease in proprotein convertase subtilisin kexin type 9 expression. CONCLUSIONS: GPER activation upregulates LDL receptor expression, probably at least, in part, via proprotein convertase subtilisin kexin type 9 downregulation. Furthermore, humans carrying the hypofunctional P16L genetic variant of GPER have increased plasma LDL cholesterol. In aggregate, these data suggest an important role of GPER in the regulation of LDL receptor expression and consequently LDL metabolism.


Asunto(s)
LDL-Colesterol/sangre , Mutación Missense , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Adolescente , Adulto , Anciano , Canadá , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica , Frecuencia de los Genes , Genética de Población , Células Hep G2 , Heterocigoto , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Proproteína Convertasa 9 , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Interferencia de ARN , Receptores de Estrógenos/efectos de los fármacos , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Transfección , Población Blanca/genética , Adulto Joven
13.
Curr Opin Lipidol ; 26(2): 103-13, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25692347

RESUMEN

PURPOSE OF REVIEW: To evaluate the potential clinical translation of high-throughput next-generation sequencing (NGS) methods in diagnosis and management of dyslipidemia. RECENT FINDINGS: Recent NGS experiments indicate that most causative genes for monogenic dyslipidemias are already known. Thus, monogenic dyslipidemias can now be diagnosed using targeted NGS. Targeting of dyslipidemia genes can be achieved by either: designing custom reagents for a dyslipidemia-specific NGS panel; or performing genome-wide NGS and focusing on genes of interest. Advantages of the former approach are lower cost and limited potential to detect incidental pathogenic variants unrelated to dyslipidemia. However, the latter approach is more flexible because masking criteria can be altered as knowledge advances, with no need for re-design of reagents or follow-up sequencing runs. Also, the cost of genome-wide analysis is decreasing and ethical concerns can likely be mitigated. DNA-based diagnosis is already part of the clinical diagnostic algorithms for familial hypercholesterolemia. Furthermore, DNA-based diagnosis is supplanting traditional biochemical methods to diagnose chylomicronemia caused by deficiency of lipoprotein lipase or its co-factors. SUMMARY: The increasing availability and decreasing cost of clinical NGS for dyslipidemia means that its potential benefits can now be evaluated on a larger scale.


Asunto(s)
Dislipidemias/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Dislipidemias/diagnóstico , Exoma , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Técnicas de Diagnóstico Molecular , Análisis de Secuencia de ADN
14.
Am J Hum Genet ; 90(1): 49-60, 2012 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-22226083

RESUMEN

The molecular basis for primary hereditary hypertriglyceridemia has been identified in fewer than 5% of cases. Investigation of monogenic dyslipidemias has the potential to expose key metabolic pathways. We describe a hitherto unreported disease in ten individuals manifesting as moderate to severe transient childhood hypertriglyceridemia and fatty liver followed by hepatic fibrosis and the identification of the mutated gene responsible for this condition. We performed SNP array-based homozygosity mapping and found a single large continuous segment of homozygosity on chromosomal region 12q13.12. The candidate region contained 35 genes that are listed in Online Mendelian Inheritance in Man (OMIM) and 27 other genes. We performed candidate gene sequencing and screened both clinically affected individuals (children and adults with hypertriglyceridemia) and also a healthy cohort for mutations in GPD1, which encodes glycerol-3-phosphate dehydrogenase 1. Mutation analysis revealed a homozygous splicing mutation, c.361-1G>C, which resulted in an aberrantly spliced mRNA in the ten affected individuals. This mutation is predicted to result in a truncated protein lacking essential conserved residues, including a functional site responsible for initial substrate recognition. Functional consequences of the mutation were evaluated by measuring intracellular concentrations of cholesterol and triglyceride as well as triglyceride secretion in HepG2 (hepatocellular carcinoma) human cells lines overexpressing normal and mutant GPD1 cDNA. Overexpression of mutant GPD1 in HepG2 cells, in comparison to overexpression of wild-type GPD1, resulted in increased secretion of triglycerides (p = 0.01). This finding supports the pathogenicity of the identified mutation.


Asunto(s)
Hígado Graso/genética , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/genética , Hipertrigliceridemia/genética , Cirrosis Hepática/genética , Mutación , Adolescente , Empalme Alternativo/genética , Secuencia de Bases , Niño , Preescolar , Colesterol/análisis , Cromosomas Humanos Par 12/genética , Estudios de Cohortes , Análisis Mutacional de ADN , Femenino , Células Hep G2 , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Índice de Severidad de la Enfermedad , Triglicéridos/análisis , Triglicéridos/metabolismo
15.
CMAJ ; 187(2): 102-107, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25452324

RESUMEN

BACKGROUND: Congenital sucrase-isomaltase deficiency is a rare hereditary cause of chronic diarrhea in children. People with this condition lack the intestinal brush-border enzyme required for digestion of di- and oligosaccharides, including sucrose and isomaltose, leading to malabsorption. Although the condition is known to be highly prevalent (about 5%-10%) in several Inuit populations, the genetic basis for this has not been described. We sought to identify a common mutation for congenital sucrase-isomaltase deficiency in the Inuit population. METHODS: We sequenced the sucrase-isomaltase gene, SI, in a single Inuit proband with congenital sucrase-isomaltase deficiency who had severe fermentative diarrhea and failure to thrive. We then genotyped a further 128 anonymized Inuit controls from a variety of locales in the Canadian Arctic to assess for a possible founder effect. RESULTS: In the proband, we identified a novel, homozygous frameshift mutation, c.273_274delAG (p.Gly92Leufs*8), predicted to result in complete absence of a functional protein product. This change was very common among the Inuit controls, with an observed allele frequency of 17.2% (95% confidence interval [CI] 12.6%-21.8%). The predicted Hardy-Weinberg prevalence of congenital sucrase-isomaltase deficiency in Inuit people, based on this single founder allele, is 3.0% (95% CI 1.4%-4.5%), which is comparable with previous estimates. INTERPRETATION: We found a common mutation, SI c.273_274delAG, to be responsible for the high prevalence of congenital sucrase-isomaltase deficiency among Inuit people. Targeted mutation testing for this allele should afford a simple and minimally invasive means of diagnosing this condition in Inuit patients with chronic diarrhea.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/etnología , Errores Innatos del Metabolismo de los Carbohidratos/genética , Efecto Fundador , Inuk/genética , Mutación/genética , Complejo Sacarasa-Isomaltasa/deficiencia , Complejo Sacarasa-Isomaltasa/genética , Canadá/epidemiología , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Estudios de Casos y Controles , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Recién Nacido
16.
J Lipid Res ; 55(4): 765-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24503134

RESUMEN

We report the design of a targeted resequencing panel for monogenic dyslipidemias, LipidSeq, for the purpose of replacing Sanger sequencing in the clinical detection of dyslipidemia-causing variants. We also evaluate the performance of the LipidSeq approach versus Sanger sequencing in 84 patients with a range of phenotypes including extreme blood lipid concentrations as well as additional dyslipidemias and related metabolic disorders. The panel performs well, with high concordance (95.2%) in samples with known mutations based on Sanger sequencing and a high detection rate (57.9%) of mutations likely to be causative for disease in samples not previously sequenced. Clinical implementation of LipidSeq has the potential to aid in the molecular diagnosis of patients with monogenic dyslipidemias with a high degree of speed and accuracy and at lower cost than either Sanger sequencing or whole exome sequencing. Furthermore, LipidSeq will help to provide a more focused picture of monogenic and polygenic contributors that underlie dyslipidemia while excluding the discovery of incidental pathogenic clinically actionable variants in nonmetabolism-related genes, such as oncogenes, that would otherwise be identified by a whole exome approach, thus minimizing potential ethical issues.


Asunto(s)
Dislipidemias/genética , Técnicas de Diagnóstico Molecular , Análisis Mutacional de ADN , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Anotación de Secuencia Molecular , Mutación
18.
Br J Clin Pharmacol ; 78(6): 1441-52, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25039431

RESUMEN

AIMS: Activation of vascular GPER has been linked to vasodepressor effects in animals. However, the significance of GPER regulation on chronic blood pressure control in humans is unknown. METHODS: To examine this question we determined the functional significance of expression of a common missense single nucleotide variant of GPER, P16L in vascular smooth muscle cells, and its association with blood pressure in humans. Further, to validate the importance of carrying GPER P16L in the development of hypertension we assessed allele frequency in a cohort of hard-to-treat hypertensive patients referred to a tertiary care clinic. RESULTS: Expression of the GPER P16L variant (V) vs. wild type (WT) in rat aortic vascular smooth muscle cells, was associated with a significant decrease in G1 (1 µm, a GPER agonist)-mediated ERK phosphorylation (slope of the function of G1-stimulated ERK phosphorylation: GPER content WT: 16.2, 95% CI 9.9, 22.6; V: 5.0, 95% CI 1.0, 9.0; P < 0.005) and apoptosis (slope of the function of G1-stimulated apoptosis: GPER content: WT: 4.4, 95% CI: 3.4, 5.4; V: 2.5, 95% CI 1.6, 2.3 P < 0.005). Normotensive female subjects, but not male subjects, carrying this hypofunctional variant (allele frequency 22%) have increased blood pressure [mean arterial pressure: P16/P16: 80 ± 1 mmHg (n = 204) vs. P16L carriers: 82 ± 1 mmHg (n = 127), 95% CI for difference: 0.6, 4.0 mmHg, P < 0.05], [systolic blood pressure: P16/P16: 105 ± 1 mmHg vs. P16L carriers: 108 ± 1 mmHg, 95% CI for difference:1.0, 5.1 mmHg, P < 0.05], [diastolic blood pressure: P16/P16: 66 ± 0.5 mmHg vs. P16L carriers 68 ± 0.7, 95% CI for difference: 0.2, 3.6 mmHg, P < 0.05]. Further, the P16L allele frequency was almost two-fold higher in female vs. male hypertensive patients (31% vs. 16%, allele ratio 0.5, 95% CI 0.32, 0.76, P < 0.05). CONCLUSIONS: The common genetic variant, GPER P16L, is hypofunctional and female carriers of this allele have increased blood pressure. There was an increased prevalence in a population of hard-to-treat hypertensive female patients. Cumulatively, these data suggest that in females, impaired GPER function might be associated with increased blood pressure and risk of hypertension.


Asunto(s)
Hipertensión/genética , Receptores de Estrógenos/genética , Receptores Acoplados a Proteínas G/genética , Adolescente , Adulto , Animales , Apoptosis , Células Cultivadas , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Frecuencia de los Genes , Variación Genética , Humanos , Hipertensión/etiología , Masculino , Fosforilación , Ratas , Caracteres Sexuales
19.
Paediatr Child Health ; 19(10): 515-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25587224

RESUMEN

The melanocortin-4-receptor gene (MC4R) is a key regulator of energy homeostasis, food intake and body weight. MC4R gene mutations are associated with early-onset severe obesity. Most patients are heterozygotes, with some reports of homozygotes and compound heterozygotes. The authors report a case involving an eight-year-old girl with progressive weight gain from infancy, body mass index 44 kg/m(2) (>97th percentile), hyperphagia, hyperinsulinemia and increased linear growth. There was no phenotype of morbid obesity in the parents or sibling. Coding regions and intron-exon boundaries of the genes encoding leptin, leptin receptor, pro-opiomelanocortin and MC4R were analyzed. Two heterozygous coding mutations in the MCR4 gene (S94N and C293R) were detected, of which the second has not been previously reported. The mutations were on opposite chromosomes, confirming compound heterozygosity. The molecular findings and clinical features associated with this novel MC4R mutation are described. The authors emphasize that rare mutations can be found in some patients with severe childhood-onset obesity.


Le gène récepteur de la mélanocortine 4 (MC4R) joue un rôle clé dans le maintien de l'homéostasie énergétique, de la prise alimentaire et du poids corporel. Les mutations du gène MC4R s'associent à une grave obésité à apparition précoce. La plupart des patients sont hétérozygotes, mais il y a certaines descriptions de patients homozygotes et d'hétérozygotes composés. Les auteurs présentent le cas d'une fillette de huit ans ayant une prise de poids progressive depuis la première enfance, un indice de masse corporel de 44 kg/m2 (>97e percentile), une hyperphagie, une hyperinsulinémie et une augmentation de la croissance linéaire. Les parents et la fratrie ne possédaient pas de phénotype d'obésité morbide. Les auteurs ont analysé les régions codantes et les limites intron-exon des gènes codant la leptine, son récepteur, la pro-opiomélanocortine et le MC4R. Ils ont décelé deux mutations en région codante du gène MCR4 (S94N et C293R), la deuxième n'ayant jamais été déclarée auparavant. Les mutations se situaient sur des chromosomes opposés, ce qui confirme leur hétérozygotie composée. Les auteurs décrivent les observations moléculaires et les caractéristiques cliniques associées à cette nouvelle mutation MC4R. Ils soulignent que certains patients ayant une importante obésité qui se manifeste pendant l'enfance peuvent présenter des mutations rares.

20.
CJC Open ; 6(1): 40-46, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38313344

RESUMEN

Background: Low density lipoprotein (LDL) and Lipoprotein (Lp)(a) are proatherogenic apolipoprotein (apo) B-containing members of the non-high-density lipoprotein (non-HDL) family of particles. Elevated plasma levels of LDL cholesterol (C), non-HDL-C, and apo B are defining features of heterozygous familial hypercholesterolemia (HeFH), but reports of elevated plasma Lp(a) concentration are inconsistent. Methods: We performed retrospective chart reviews of 256 genetically characterized patients with hypercholesterolemia and 272 control subjects from the Lipid Genetics Clinic at University Hospital in London, Ontario. We evaluated pairwise correlations between plasma levels of Lp(a) and those of LDL-C, non-HDL-C and apo B. Results: Mean Lp(a) levels were not different between individuals with hypercholesterolemia and control subjects. No correlations were found between Lp(a) and LDL-C or non-HDL-C levels in controls or patients with hypercholesterolemia; all r values < 0.079 and all P values > 0.193. Borderline weak correlations between Lp(a) and apo B were identified in patients r = 0.103; P = 0.112) and controls (r = 0.175; P = 0.005). Results were similar across genotypic subgroups. Conclusions: Lp(a) levels are independent of LDL-C and non-HDL-C; in particular Lp(a) levels are not increased in patients with hypercholesterolemia and molecularly proven HeFH. Apo B was only weakly associated with Lp(a). Elevated Lp(a) does not cause FH in our clinic patients. Genetic variants causing HeFH that raise LDL-C do not affect Lp(a), confirming that these lipoproteins are metabolically distinct. Lp(a) cannot be predicted from LDL-C and must be determined separately to evaluate its amplifying effect on atherosclerotic risk in patients with hypercholesterolemia.


Introduction: Les lipoprotéines de faible densité (LDL) et les lipoprotéines (Lp)(a) sont des éléments contenant des apolipoprotéines (apo)B proathérogènes de la famille des particules non à lipoprotéines de haute densité (non-HDL). Des concentrations plasmatiques élevées de cholestérol LDL, de cholestérol non-HDL et d'apoB sont des caractéristiques déterminantes de l'hypercholestérolémie familiale de type hétérozygote (HFHe), mais les documents qui portent sur les concentrations plasmatiques élevées de Lp(a) sont contradictoires. Méthodes: Nous avons effectué des revues rétrospectives de dossiers de 256 patients ayant les caractéristiques génétiques de l'hypercholestérolémie, et 272 témoins de la clinique de dyslipidémies génétiques de l'Hôpital universitaire de London, en Ontario. Nous avons évalué les corrélations par paires entre les concentrations plasmatiques de Lp(a) et celles du cholestérol LDL, du cholestérol non-HDL et de l'apoB. Résultats: Les concentrations moyennes de Lp(a) n'étaient pas différentes entre les individus atteints d'hypercholestérolémie et les témoins. Aucune corrélation n'a été observée entre les concentrations de Lp(a) et de cholestérol LDL ou de cholestérol non­HDL des témoins ou des patients atteints d'hypercholestérolémie ; toutes valeurs r < 0,079 et toutes valeurs P > 0,193. Nous avons relevé des corrélations limites faibles entre les concentrations de Lp(a) et d'apoB chez les patients r = 0,103 ; P = 0,112) et les témoins (r = 0,175 ; P = 0,005). Les résultats étaient similaires dans tous les sous-groupes génotypiques. Conclusions: Les concentrations de Lp(a) sont indépendantes des concentrations de cholestérol LDL et de cholestérol non­HDL ; notamment les concentrations de Lp(a) n'augmentent pas chez les patients atteints d'hypercholestérolémie et de HFHe moléculairement prouvée. L'apoB n'était que faiblement associée aux concentrations de Lp(a). Les concentrations élevées de Lp(a) ne causaient pas la HF chez les patients de notre clinique. Les variants génétiques causant la HFHe qui font augmenter les concentrations de cholestérol LDL n'affectent pas les concentrations de Lp(a), ce qui confirme que ces lipoprotéines sont distinctes sur le plan métabolique. Puisqu'on ne peut prédire les concentrations de Lp(a) à partir des concentrations de cholestérol LDL, elles doivent être déterminées séparément pour évaluer leur effet amplificateur sur le risque athérosclérotique des patients atteints d'hypercholestérolémie.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA