Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 315
Filtrar
1.
Stem Cell Res ; 60: 102702, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35152178

RESUMEN

The development of cellular models for familial hypercholesterolemia (FH) is an important direction for creating new approaches to atherosclerosis treatment. Pathogenic mutations in the LDLR gene are the main FH source. We generated an iPSC line from peripheral blood mononuclear cells of the patient with compound heterozygous c.1246C > T/c.940 + 3_940 + 6del LDLR mutation. The resulting iPSC line with confirmed patient-specific mutations maintains a normal karyotype and a typical undifferentiated state, including morphology, pluripotent gene expression, and in vitro differentiation potential. This iPSC line can be further differentiated toward relevant cells to better understand FH pathogenesis.


Asunto(s)
Hiperlipoproteinemia Tipo II , Células Madre Pluripotentes Inducidas , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Hiperlipoproteinemia Tipo II/patología , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Mutación/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo
2.
Stem Cell Res ; 60: 102703, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35152179

RESUMEN

Familial hypercholesterolemia (FH) is an autosomal dominant disorder increasing premature cardiovascular diseases risk due to atherosclerosis. Pathogenic mutations in the LDLR gene cause most FH cases. Available treatments are effective not for all LDLR mutations. Testing drugs on FH cell models help develop new efficient treatments. We obtained an iPSC line from peripheral blood mononuclear cells of the patient with heterozygous p.Trp443Arg LDLR mutation. The iPSCs with confirmed patient-specific mutations express pluripotency markers, spontaneously differentiate into three germ layers and demonstrate normal karyotype. Patient-specific iPSCs-derived hepatocyte-like and endothelial cells are promising to develop new targeted therapies for FH.


Asunto(s)
Hiperlipoproteinemia Tipo II , Células Madre Pluripotentes Inducidas , Células Endoteliales/metabolismo , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Células Madre Pluripotentes Inducidas/metabolismo , Leucocitos Mononucleares/metabolismo , Mutación/genética , Receptores de LDL/genética , Receptores de LDL/metabolismo
3.
Genes (Basel) ; 13(1)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-35052492

RESUMEN

Familial hypercholesterolemia (FH) is one of the most common autosomal, dominantly inherited diseases affecting cholesterol metabolism, which, in the absence of treatment, leads to the development of cardiovascular complications. The disease is still underdiagnosed, even though an early diagnosis would be of great importance for the patient to receive proper treatment and to prevent further complications. No studies are available describing the genetic background of Hungarian FH patients. In this work, we present the clinical and molecular data of 44 unrelated individuals with suspected FH. Sequencing of five FH-causing genes (LDLR, APOB, PCSK9, LDLRAP1 and STAP1) has been performed by next-generation sequencing (NGS). In cases where a copy number variation (CNV) has been detected by NGS, confirmation by multiplex ligation-dependent probe amplification (MLPA) has also been performed. We identified 47 causal or potentially causal (including variants of uncertain significance) LDLR and APOB variants in 44 index patients. The most common variant in the APOB gene was the c.10580G>A p.(Arg3527Gln) missense alteration, this being in accordance with literature data. Several missense variants in the LDLR gene were detected in more than one index patient. LDLR variants in the Hungarian population largely overlap with variants detected in neighboring countries.


Asunto(s)
Variaciones en el Número de Copia de ADN , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Hiperlipoproteinemia Tipo II/patología , Mutación , Adolescente , Adulto , Anciano , Apolipoproteína B-100/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Hungría/epidemiología , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/genética , Masculino , Persona de Mediana Edad , Fenotipo , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Adulto Joven
4.
São Paulo; s.n; s.n; 2022. 145 p. tab, graf.
Tesis en Portugués | LILACS | ID: biblio-1416663

RESUMEN

A Hipercolesterolemia Familial (HF) é uma doença hereditária do metabolismo lipídico que causa aos portadores alta incidência de aterosclerose prematura. A HF pode ser diagnosticada clínica e geneticamente, entretanto, apenas cerca de 40% podem ter confirmados pelo diagnostico molecular. Assim, outros sistemas de diagnóstico devem ser avaliados. Ultimamente devido a estabilidade em fluidos biológicos, os exossomos circulantes apresentam grande potencial, pois carreiam um número variado de compostos e são considerados veículos de intercomunicação entre os tecidos. Sabe-se que vários RNAs são carreados nos exossomos, incluindo miRNAs, lncRNA e uma variedade de proteínas. Estes componentes podem ser marcadores de diagnóstico para várias doenças inclusive a HF e suas complicações cardiovasculares. Foram utilizadas amostras de exossomos plasmáticos provenientes de 54 pacientes HF sem uso de estatina por, no mínimo, seis semanas, e 38 indivíduos normolipidêmicos para sequenciamento de miRNAs e estudo da proteômica. Os exossomos foram isolados utilizando dois métodos precipitação química e cromatográfica de exclusão de tamanho e caraterizados utilizando: dispersão de luz dinâmica, Western blotting, rastreamento de nanopartículas (NanoSight), imunomarcação e microscopia eletrônica de transmissão. Os miRNAs e proteínas foram extraídos dos exossomos e analisados por sequenciamento de nova geração e espectrometria de massa, respectivamente. Os dados clínicos, biodemográficos e laboratoriais dos pacientes HF e controles indicaram diferenças significativas esperadas entre os grupos, indicando que foram selecionados adequadamente. A caracterização físico-química dos exossomos mostrou resultados com tamanho de ˜90nm e imunorreação positiva para tetraspaninas. O resultado do sequenciamento identificou acima 2000 miRNAs. Os miR-122- 5p e miR-21-5p apresentaram expressão aumentada no grupo HF (log2FC=1,79 e log2FC=1,27, respectivamente), e o miR-122-5p pós normalização em relação ao controle manteve significativo comparados ao controle (p=0,034). A análise comparativa entre exossomos e plasma total mostrou diferença significativa, pois foram identificadas 239 proteínas (p <0,05) diferentes entre exossomos e plasma. Em exossomos, 17 proteínas foram aumentadas e 21 diminuídas em pacientes com HF em comparação com o controle (p <0,05). Destas, seis proteínas foram mais abundantes em HF e sete proteínas foram menos abundantes em exossomos de pacientes com HF em comparação com o controle. A análise de enriquecimento por bioinformática mostrou que a maior parte dessas moléculas (miRNAs e proteínas) foram relacionadas com metabolismo lipídico, dislipidemia, aterosclerose, doença arterial coronariana, adipogênese. Assim, na busca de novos alvos como potenciais biomarcadores de diagnóstico da HF, nossos resultados da análise integrativa entre os miRNAs e as proteínas exossomais abre novas frentes de pesquisa mais bem direcionadas, para a validação desses miRNAs e proteínas exossomais


Familial Hypercholesterolemia (FH) is an inherited disease of lipid metabolism that causes a high incidence of premature atherosclerosis in patients. FH can be diagnosed clinically and genetically, however, only about 40% can be confirmed by molecular diagnosis. Thus, other diagnostic systems should be evaluated. Lately, due to stability in biological fluids, circulating exosomes have great potential, as they carry a varied number of compounds and are considered vehicles of intercommunication between tissues. Several RNAs are known to be carried on exosomes, including miRNAs, lncRNA, and a variety of proteins. These components can be diagnostic markers for several diseases including FH and its cardiovascular complications. Plasma exosome samples from 54 FH patients without statin use for at least six weeks and 38 normolipidemic individuals were used for miRNA sequencing and proteomics studies. Exosomes were isolated using two methods chemical precipitation and size exclusion chromatography and characterized using: dynamic light scattering, Western blotting, nanoparticle tracking (NanoSight), immunostaining and transmission electron microscopy. MiRNAs and proteins were extracted from exosomes and analyzed by next-generation sequencing and mass spectrometry, respectively. Clinical, biodemographic and laboratory data of FH patients and controls indicated significant expected differences between the groups, indicating that they were appropriately selected. The physicochemical characterization of exosomes showed results with a size of ˜90nm and positive immunoreaction for tetraspanins. The sequencing result identified above 2000 miRNAs. miR-122-5p and miR-21-5p showed increased expression in the FH group (log2FC=1.79 and log2FC=1.27, respectively), and miR122-5p after normalization in relation to the control remained significant compared to the control (p=0.034). The comparative analysis between exosomes and total plasma showed a significant difference, as 239 different proteins (p < 0.05) were identified between exosome and plasma. In exosomes, 17 proteins were increased and 21 decreased in FH patients compared to control (p < 0.05). Of these, six proteins were more abundant in FH and seven proteins were less abundant in exosomes from patients with FH compared to the control. Bioinformatics enrichment analysis showed that most of these molecules (miRNAs and proteins) were related to lipid metabolism, dyslipidemia, atherosclerosis, coronary artery disease, adipogenesis. Thus, in the search for new targets as potential diagnostic biomarkers of FH, our results of the integrative analysis between miRNAs and exosomal proteins opens new and better-directed research fronts for the validation of these miRNAs and exosomal proteins


Asunto(s)
Proteínas , MicroARNs/análisis , Exosomas/clasificación , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Hiperlipoproteinemia Tipo II/patología , Espectrometría de Masas/métodos , Química Física
5.
Genes (Basel) ; 12(10)2021 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-34680994

RESUMEN

Familial hypercholesterolemia (FH), is an autosomal dominant disorder caused by mutations in the LDLR, APOB, PCSK9, and APOE genes and is characterized by high plasma levels of total and low-density lipoprotein (LDL) cholesterol. Our study aimed to analyze the influences of two different therapies on a wide spectrum of plasma protein biomarkers of cardiovascular diseases. Plasma from FH patients under hypolipidemic therapy (N = 18; men = 8, age 55.4 ± 13.1 years) and patients under combined long-term LDL apheresis/hypolipidemic therapy (N = 14; men = 7; age 58.0 ± 13.6 years) were analyzed in our study. We measured a profile of 184 cardiovascular disease (CVD) associated proteins using a proximity extension assay (PEA). Hypolipidemic therapy significantly (all p < 0.01) influenced 10 plasma proteins (TM, DKK1, CCL3, CD4, PDGF subunit B, AGRP, IL18, THPO, and LOX1 decreased; ST2 increased). Under combined apheresis/hypolipidemic treatment, 18 plasma proteins (LDLR, PCSK9, MMP-3, GDF2, CTRC, SORT1, VEGFD, IL27, CCL24, and KIM1 decreased; OPN, COL1A1, KLK6, IL4RA, PLC, TNFR1, GLO1, and PTX3 increased) were significantly affected (all p < 0.006). Hypolipidemic treatment mainly affected biomarkers involved in vascular endothelial maintenance. Combined therapy influenced proteins that participate in cholesterol metabolism and inflammation.


Asunto(s)
Biomarcadores/sangre , Proteínas Sanguíneas/genética , Enfermedades Cardiovasculares/sangre , Colesterol/sangre , Hiperlipoproteinemia Tipo II/sangre , Adulto , Anciano , Anticolesterolemiantes/uso terapéutico , Proteínas Sanguíneas/clasificación , Proteínas Sanguíneas/aislamiento & purificación , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Colesterol/metabolismo , Femenino , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Inflamación/sangre , Inflamación/genética , Inflamación/metabolismo , Masculino , Persona de Mediana Edad
6.
Lipids Health Dis ; 20(1): 101, 2021 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-34511120

RESUMEN

BACKGROUND: As an autosomal dominant disorder, familial hypercholesterolemia (FH) is mainly attributed to disease-causing variants in the low-density lipoprotein receptor (LDLR) gene. The aim of this study was to explore the molecular mechanism of LDLR c.501C>A variant in FH and assess the efficacy of proprotein convertase subtilisin kexin type 9 (PCSK9) inhibitor treatment for FH patients. METHODS: The whole-exome sequencing was performed on two families to identify disease-causing variants, which were verified by Sanger sequencing. The function of LDLR variant was further explored in HEK293 cells by Western Blot and confocal microscopy. Besides, the therapeutic effects of PCSK9 inhibitor treatment for two probands were assessed for 3 months. RESULTS: All members of the two families with the LDLR c.501C>A variant showed high levels of LDLC. The relationship between the clinical phenotype and LDLR variants was confirmed in the current study. Both in silico and in vitro analyses showed that LDLR c.501C>A variant decreased LDLR expression and LDL uptake. PCSK9 inhibitor treatment lowered the lipid level in proband 1 by 24.91%. However, the treatment was ineffective for proband 2. A follow-up study revealed that the PCSK9 inhibitor treatment had low ability of lipid-lowering effect in the patients. CONCLUSIONS: LDLR c.501C>A variant might be pathogenic for FH. The PCSK9 inhibitor therapy is not a highly effective option for treatment of FH patients with LDLR c.501C>A variant.


Asunto(s)
Hiperlipoproteinemia Tipo II/genética , Mutación Puntual , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Adolescente , Adulto , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Familia , Femenino , Regulación de la Expresión Génica , Células HEK293 , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/patología , Hipolipemiantes/uso terapéutico , Masculino , Persona de Mediana Edad , Inhibidores de PCSK9/uso terapéutico , Linaje , Proproteína Convertasa 9/metabolismo , Receptores de LDL/deficiencia , Insuficiencia del Tratamiento , Triglicéridos/sangre , Secuenciación del Exoma
7.
Int J Mol Sci ; 22(10)2021 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-34066088

RESUMEN

Familial hypercholesterolemia (FH) is increasingly associated with inflammation, a phenotype that persists despite treatment with lipid lowering therapies. The alternative C3 complement system (C3), as a key inflammatory mediator, seems to be involved in the atherosclerotic process; however, the relationship between C3 and lipids during plaque progression remains unknown. The aim of the study was to investigate by a systems biology approach the role of C3 in relation to lipoprotein levels during atherosclerosis (AT) progression and to gain a better understanding on the effects of C3 products on the phenotype and function of human lipid-loaded vascular smooth muscle cells (VSMCs). By mass spectrometry and differential proteomics, we found the extracellular matrix (ECM) of human aortas to be enriched in active components of the C3 complement system, with a significantly different proteomic signature in AT segments. Thus, C3 products were more abundant in AT-ECM than in macroscopically normal segments. Furthermore, circulating C3 levels were significantly elevated in FH patients with subclinical coronary AT, evidenced by computed tomographic angiography. However, no correlation was identified between circulating C3 levels and the increase in plaque burden, indicating a local regulation of the C3 in AT arteries. In cell culture studies of human VSMCs, we evidenced the expression of C3, C3aR (anaphylatoxin receptor) and the integrin αMß2 receptor for C3b/iC3b (RT-PCR and Western blot). C3mRNA was up-regulated in lipid-loaded human VSMCs, and C3 protein significantly increased in cell culture supernatants, indicating that the C3 products in the AT-ECM have a local vessel-wall niche. Interestingly, C3a and iC3b (C3 active fragments) have functional effects on VSMCs, significantly reversing the inhibition of VSMC migration induced by aggregated LDL and stimulating cell spreading, organization of F-actin stress fibers and attachment during the adhesion of lipid-loaded human VSMCs. This study, by using a systems biology approach, identified molecular processes involving the C3 complement system in vascular remodeling and in the progression of advanced human atherosclerotic lesions.


Asunto(s)
Aterosclerosis/patología , Complemento C3/metabolismo , Hiperlipoproteinemia Tipo II/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Proteoma/metabolismo , Adulto , Aterosclerosis/inmunología , Aterosclerosis/metabolismo , Estudios de Casos y Controles , Adhesión Celular , Células Cultivadas , Femenino , Humanos , Hiperlipoproteinemia Tipo II/inmunología , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Persona de Mediana Edad , Músculo Liso Vascular/inmunología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/metabolismo , Proteoma/análisis , Remodelación Vascular , Cicatrización de Heridas , Adulto Joven
8.
J Hum Genet ; 66(11): 1079-1087, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33967275

RESUMEN

Familial hypercholesterolemia (FH) is an autosomal dominant monogenic disorder characterized by elevated levels of low-density lipoprotein cholesterol (LDL-C) and an increased risk of premature coronary artery disease (CAD). Recently, it has been shown that a high polygenic risk score (PRS) could be an independent risk factor for CAD in FH patients of European ancestry. However, it is uncertain whether PRS is also useful for risk stratification of FH patients in East Asia. We recruited and genotyped clinically diagnosed FH (CDFH) patients from the Kanazawa University Mendelian Disease FH registry and controls from the Shikamachi Health Improvement Practice genome cohort in Japan. We calculated PRS from 3.6 million variants of each participant (imputed from the 1000 Genome phase 3 Asian dataset) for LDL-C (PRSLDLC) using a genome-wide association study summary statistic from the BioBank Japan Project. We assessed the association of PRSLDLC with LDL-C and CAD among and within monogenic FH, mutation negative CDFH, and controls. We tested a total of 1223 participants (376 FH patients, including 173 with monogenic FH and 203 with mutation negative CDFH, and 847 controls) for the analyses. PRSLDLC was significantly higher in mutation negative CDFH patients than in controls (p = 3.1 × 10-13). PRSLDLC was also significantly linked to LDL-C in controls (p trend = 3.6 × 10-4) but not in FH patients. Moreover, we could not detect any association between PRSLDLC and CAD in any of the groups. In conclusion, mutation negative CDFH patients demonstrated significantly higher PRSLDLC than controls. However, PRSLDLC may have little additional effect on LDL-C and CAD among FH patients.


Asunto(s)
LDL-Colesterol/genética , Enfermedad de la Arteria Coronaria/genética , Hiperlipoproteinemia Tipo II/genética , Herencia Multifactorial/genética , Adulto , LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/etiología , Enfermedad de la Arteria Coronaria/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/complicaciones , Hiperlipoproteinemia Tipo II/patología , Japón/epidemiología , Masculino , Persona de Mediana Edad , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
9.
J Hum Genet ; 66(10): 983-993, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33864011

RESUMEN

Familial Hypercholesterolemia (FH) is an autosomal, dominant, inherited disorder characterized by severely elevated LDL-cholesterol (LDL-C) levels with high risk for Coronary Artery Disease (CAD). There are limited genetic studies especially on genes other than Low Density Lipoprotein receptor (LDLR) conducted in Indian population. Thus, our aim was to screen the entire Proprotein Convertase Subtilisin/Kexin type 9 gene (PCSK9) gene & hotspot exons 3, 4 and 9 of LDLR gene in FH cases and controls. 50 FH cases were categorized into definite, probable and possible cases according to Dutch Lipid Network Criteria (DLNC) who were gender matched with 50 healthy controls. All 12 exons of PCSK9, and hotspot exons 3, 4 & 9 of LDLR gene were screened through High Resolution Melt (HRM) curve analysis. Enzyme linked immunosorbent assay was performed to measure circulating PCSK9 levels. Total cholesterol and LDL-C were significantly high in all three groups of cases. Total 8 nonpathogenic variants in exon 1, 5, 7 and 9 of the PCSK9 gene were detected. In LDLR gene, 3 known pathogenic and 1 benign variant were found in exon 3 & 4. In FH cases, PCSK9 levels were significantly high compared to controls (P = 0.0001), and were directly correlated to LDL-C (P = 0.0001) and Total Cholesterol (P = 0.0001). Our study is first to screen the entire PCSK9 gene in western part of India. Since no pathogenic variants were identified, it is possible that PCSK9 variants are clinically less relevant. However, 3 known pathogenic variants were found in the LDLR gene. These findings support our understanding of the genetic spectrum of FH in India.


Asunto(s)
Predisposición Genética a la Enfermedad , Hiperlipoproteinemia Tipo II/genética , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Adulto , Pueblo Asiatico/genética , LDL-Colesterol , Exones/genética , Femenino , Variación Genética/genética , Humanos , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/patología , India/epidemiología , Masculino , Persona de Mediana Edad , Mutación/genética , Fenotipo
10.
Sci Rep ; 11(1): 3801, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589716

RESUMEN

Familial hypercholesterolaemia increases circulating LDL-C levels and leads to premature cardiovascular disease when undiagnosed or untreated. Current guidelines support genetic testing in patients complying with clinical diagnostic criteria and cascade screening of their family members. However, most of hyperlipidaemic subjects do not present pathogenic variants in the known disease genes, and most likely suffer from polygenic hypercholesterolaemia, which translates into a relatively low yield of genetic screening programs. This study aims to identify new biomarkers and develop new approaches to improve the identification of individuals carrying monogenic causative variants. Using a machine-learning approach in a paediatric dataset of individuals, tested for disease causative genes and with an extended lipid profile, we developed new models able to classify familial hypercholesterolaemia patients with a much higher specificity than currently used methods. The best performing models incorporated parameters absent from the most common FH clinical criteria, namely apoB/apoA-I, TG/apoB and LDL1. These parameters were found to contribute to an improved identification of monogenic individuals. Furthermore, models using only TC and LDL-C levels presented a higher specificity of classification when compared to simple cut-offs. Our results can be applied towards the improvement of the yield of genetic screening programs and corresponding costs.


Asunto(s)
Biomarcadores/sangre , Dislipidemias/diagnóstico , Hiperlipoproteinemia Tipo II/diagnóstico , Lípidos/sangre , Adulto , Anciano , Apolipoproteína A-I/sangre , Apolipoproteínas B/sangre , Diagnóstico Diferencial , Dislipidemias/sangre , Dislipidemias/patología , Femenino , Pruebas Genéticas , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Lipoproteínas LDL/sangre , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Modelos Biológicos , Triglicéridos/sangre
11.
Circ Genom Precis Med ; 14(1): e003106, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33440130

RESUMEN

BACKGROUND: The clinical implications of a polygenic risk score (PRS) for LDL-C (low-density lipoprotein cholesterol) are not well understood, both within the general population and individuals with familial hypercholesterolemia (FH). METHODS: We developed the LDL-C PRS using Least Absolute Shrinkage and Selection Operator regression in 377 286 White British participants from UK Biobank and tested its association with LDL-C according to FH variant carrier status in another 41 748 whole-exome sequenced individuals. Next, we tested for an enrichment of FH variant carriers among individuals with severe hypercholesterolemia and low LDL-C PRS. Last, we contrasted the effect of the LDL-C PRS, measured LDL-C and FH variant carrier status on risk of ischemic heart disease among 3010 cases and 38 738 controls. RESULTS: Among the 41 748 whole-exome sequenced White British individuals, 1-SD increase in the LDL-C PRS was associated with elevated LDL-C among both FH variant carriers (0.34 [95% CI, 0.22-0.47] mmol/L) and noncarriers (0.42 [95% CI, 0.42-0.43] mmol/L). Among individuals with severe hypercholesterolemia, FH variant carriers were enriched in those with a low LDL-C PRS (odds ratio, 2.20 [95% CI, 1.66-2.71] per SD). Each SD increase in the LDL-C PRS was associated with risk of ischemic heart disease to the comparable magnitude as measured LDL-C (odds ratio, 1.24 [95% CI, 1.20-1.29] and odds ratio, 1.15 [95% CI, 1.09-1.23], respectively). The LDL-C PRS was not strongly associated with other traditional ischemic heart disease risk factors. CONCLUSIONS: An LDL-C PRS could be used to identify individuals with a higher probability of harboring FH variants. The association between ischemic heart disease and the LDL-C PRS was comparable to measured LDL-C, likely because the PRS reflects lifetime exposure to LDL-C levels.


Asunto(s)
LDL-Colesterol/sangre , Hiperlipoproteinemia Tipo II/genética , Isquemia Miocárdica/genética , Anciano , Bases de Datos Genéticas , Femenino , Variación Genética , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/patología , Masculino , Persona de Mediana Edad , Isquemia Miocárdica/patología , Factores de Riesgo , Secuenciación Completa del Genoma
12.
Circ Genom Precis Med ; 14(1): e003029, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33315477

RESUMEN

BACKGROUND: Loss-of-function mutations in the LDL (low-density lipoprotein) receptor gene (LDLR) cause elevated levels of LDL cholesterol and premature cardiovascular disease. To date, a gain-of-function mutation in LDLR with a large effect on LDL cholesterol levels has not been described. Here, we searched for sequence variants in LDLR that have a large effect on LDL cholesterol levels. METHODS: We analyzed whole-genome sequencing data from 43 202 Icelanders. Single-nucleotide polymorphisms and structural variants including deletions, insertions, and duplications were genotyped using whole-genome sequencing-based data. LDL cholesterol associations were carried out in a sample of >100 000 Icelanders with genetic information (imputed or whole-genome sequencing). Molecular analyses were performed using RNA sequencing and protein expression assays in Epstein-Barr virus-transformed lymphocytes. RESULTS: We discovered a 2.5-kb deletion (del2.5) overlapping the 3' untranslated region of LDLR in 7 heterozygous carriers from a single family. Mean level of LDL cholesterol was 74% lower in del2.5 carriers than in 101 851 noncarriers, a difference of 2.48 mmol/L (96 mg/dL; P=8.4×10-8). Del2.5 results in production of an alternative mRNA isoform with a truncated 3' untranslated region. The truncation leads to a loss of target sites for microRNAs known to repress translation of LDLR. In Epstein-Barr virus-transformed lymphocytes derived from del2.5 carriers, expression of alternative mRNA isoform was 1.84-fold higher than the wild-type isoform (P=0.0013), and there was 1.79-fold higher surface expression of the LDL receptor than in noncarriers (P=0.0086). We did not find a highly penetrant detrimental impact of lifelong very low levels of LDL cholesterol due to del2.5 on health of the carriers. CONCLUSIONS: Del2.5 is the first reported gain-of-function mutation in LDLR causing a large reduction in LDL cholesterol. These data point to a role for alternative polyadenylation of LDLR mRNA as a potent regulator of LDL receptor expression in humans.


Asunto(s)
LDL-Colesterol/sangre , Receptores de LDL/genética , Regiones no Traducidas 3' , Empalme Alternativo , Mutación con Ganancia de Función , Eliminación de Gen , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Herpesvirus Humano 4/genética , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Islandia , Linfocitos/citología , Linfocitos/metabolismo , MicroARNs/metabolismo , Linaje , Isoformas de Proteínas/genética , ARN Mensajero/metabolismo
14.
Nat Commun ; 11(1): 4084, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796843

RESUMEN

Lipid peroxidation generates reactive dicarbonyls including isolevuglandins (IsoLGs) and malondialdehyde (MDA) that covalently modify proteins. Humans with familial hypercholesterolemia (FH) have increased lipoprotein dicarbonyl adducts and dysfunctional HDL. We investigate the impact of the dicarbonyl scavenger, 2-hydroxybenzylamine (2-HOBA) on HDL function and atherosclerosis in Ldlr-/- mice, a model of FH. Compared to hypercholesterolemic Ldlr-/- mice treated with vehicle or 4-HOBA, a nonreactive analogue, 2-HOBA decreases atherosclerosis by 60% in en face aortas, without changing plasma cholesterol. Ldlr-/- mice treated with 2-HOBA have reduced MDA-LDL and MDA-HDL levels, and their HDL display increased capacity to reduce macrophage cholesterol. Importantly, 2-HOBA reduces the MDA- and IsoLG-lysyl content in atherosclerotic aortas versus 4-HOBA. Furthermore, 2-HOBA reduces inflammation and plaque apoptotic cells and promotes efferocytosis and features of stable plaques. Dicarbonyl scavenging with 2-HOBA has multiple atheroprotective effects in a murine FH model, supporting its potential as a therapeutic approach for atherosclerotic cardiovascular disease.


Asunto(s)
Aterosclerosis/metabolismo , Bencilaminas/metabolismo , Bencilaminas/farmacología , Bencilaminas/uso terapéutico , Hiperlipoproteinemia Tipo II/metabolismo , Receptores de LDL/genética , Animales , Aorta , Apolipoproteínas E , Aterosclerosis/tratamiento farmacológico , Colesterol/sangre , Colesterol/metabolismo , Femenino , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/patología , Inflamación/tratamiento farmacológico , Peroxidación de Lípido , Lipoproteínas HDL/metabolismo , Lipoproteínas IDL/sangre , Lipoproteínas IDL/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fragmentos de Péptidos
15.
J Clin Lipidol ; 14(5): 636-638, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32828708

RESUMEN

Autosomal dominant familial hypercholesterolemia (FH) is characterized by elevated low-density lipoprotein cholesterol levels and an increased risk for atherosclerotic cardiovascular disease. Although rare pathogenic variants in genes encoding the low-density lipoprotein receptor, apolipoprotein B, proprotein convertase subtilisin/kexin 9 are found in more than 80% of molecularly defined patients with FH, a few rare minor causative genes have been proposed, including the gene encoding signal-transducing adaptor family member 1 (STAP1). Here, we describe a patient with hypercholesterolemia and the rare heterozygous missense variant p.D207N in STAP1. However, extending the pedigree showed failure of the variant to cosegregate with hypercholesterolemia, as both his sons were carriers of the variant and both were also normolipidemic. The findings add to the evidence against STAP1 as a genetic locus for FH.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Hiperlipoproteinemia Tipo II/genética , Mutación Missense , Adolescente , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hiperlipoproteinemia Tipo II/patología , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Pronóstico
16.
J Clin Lipidol ; 14(5): 639-645, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32800790

RESUMEN

Severe familial hypercholesterolemia (SFH) is characterized by markedly elevated low-density lipoprotein cholesterol (LDL-C) and severe early-onset cardiovascular disease if left untreated. We report on the decade-long therapeutic journey of a 15-year-old boy with SFH due to a severe compound heterozygous genotype. He presented at the age of 5 years with widespread xanthomas and LDL-C of 17.4 mmol/L. He was diagnosed with SFH, initially treated with colestyramine that was subsequently combined with simvastatin. At the age of 12 years, he was diagnosed to have supravalvular aortic stenosis and ezetimibe/atorvastatin was introduced in place of colestyramine/simvastatin. At the age of 14 years, he received triple therapy with evolocumab, initially at the recommended dose of 420 mg monthly and then reduced to 140 mg biweekly. Currently at the age of 15 years, he is on atorvastatin 40 mg ON, ezetimibe 10 mg OM, and evolocumab 140 mg biweekly, achieving LDL-C levels of 2.4 mmol/L. Genetic testing identified compound heterozygous mutations in the LDL receptor genes [c.(940 + 1_941-1) (1845 + 1_1846-1)dup] and exon 12, nucleotide c.1747 C > T, amino acid p.(His583Tyr). Medical management without lipoprotein apheresis can achieve target LDL-C in children with SFH. Our patient, who developed supravalvular aortic stenosis at the age of 12 years, needed early aggressive treatment when SFH guidelines and newer drugs for young children were unavailable. Our patient demonstrated that 140 mg biweekly of evolocumab has the same cholesterol-lowering effect as the recommended 420 mg monthly dose.


Asunto(s)
Anticolesterolemiantes/uso terapéutico , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/genética , Mutación , Receptores de LDL/genética , Adolescente , LDL-Colesterol/sangre , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Masculino , Pronóstico
17.
Clin Genet ; 98(5): 457-467, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32770674

RESUMEN

Autosomal-dominant familial hypercholesterolemia (FH) is characterized by increased plasma concentrations of low-density lipoprotein cholesterol (LDL-C) and a substantial risk to develop cardiovascular disease. Causative mutations in three major genes are known: the LDL receptor gene (LDLR), the apolipoprotein B gene (APOB) and the proprotein convertase subtilisin/kexin 9 gene (PCSK9). We clinically characterized 336 patients suspected to have FH and screened them for disease causing mutations in LDLR, APOB, and PCSK9. We genotyped six single nucleotide polymorphisms (SNPs) to calculate a polygenic risk score for the patients and 1985 controls. The 117 patients had a causative variant in one of the analyzed genes. Most variants were found in the LDLR gene (84.9%) with 11 novel mutations. The mean polygenic risk score was significantly higher in FH mutation negative subjects than in FH mutation positive patients (P < .05) and healthy controls (P < .001), whereas the score of the two latter groups did not differ significantly. However, the score explained only about 3% of the baseline LDL-C variance. We verified the previously described clinical and genetic variability of FH for German hypercholesterolemic patients. Evaluation of a six-SNP polygenic score recently proposed for clinical use suggests that it is not a reliable tool to classify hypercholesterolemic patients.


Asunto(s)
Apolipoproteína B-100/genética , LDL-Colesterol/genética , Hiperlipoproteinemia Tipo II/genética , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Enfermedades Cardiovasculares/sangre , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , LDL-Colesterol/sangre , Femenino , Genotipo , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Masculino , Persona de Mediana Edad , Herencia Multifactorial/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
18.
J Clin Lipidol ; 14(5): 631-635, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32654994

RESUMEN

A 22-year-old woman presented with elevated low-density lipoprotein (LDL) cholesterol and clinically suspected familial hypercholesterolemia. Initial genetic analysis by Sanger sequencing found no causal variants in LDLR or other familial hypercholesterolemia genes. More than a decade later, her 9-year-old daughter was also found to have elevated LDL cholesterol. Re-analysis using current genetic methodology detected a novel whole-gene duplication of APOB in both individuals, which was tentatively assumed to explain their elevated LDL cholesterol based mainly on biological plausibility. However, on further assessment with cascade screening and cosegregation analysis involving multiple family members, the APOB duplication was eventually discounted as being causative. This case illustrates the risk of assuming pathogenicity of a novel genetic variant without undertaking corroborative diagnostic measures. It further highlights the time and skill required for accurate variant analysis and emphasizes the challenges faced by clinicians who are increasingly expected to rapidly interpret such results without sufficient time or resources to pursue supportive or corroborating evidence.


Asunto(s)
Apolipoproteína B-100/genética , Duplicación de Gen , Hiperlipoproteinemia Tipo II/genética , Adulto , Anciano , Apolipoproteína B-100/sangre , Niño , Preescolar , LDL-Colesterol/sangre , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Pronóstico , Adulto Joven
19.
Nat Med ; 26(8): 1235-1239, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32719484

RESUMEN

Three inherited autosomal dominant conditions-BRCA-related hereditary breast and ovarian cancer (HBOC), Lynch syndrome (LS) and familial hypercholesterolemia (FH)-have been termed the Centers for Disease Control and Prevention Tier 1 (CDCT1) genetic conditions, for which early identification and intervention have a meaningful potential for clinical actionability and a positive impact on public health1. In typical medical practice, genetic testing for these conditions is based on personal or family history, ethnic background or other demographic characteristics2. In this study of a cohort of 26,906 participants in the Healthy Nevada Project (HNP), we first evaluated whether population screening could efficiently identify carriers of these genetic conditions and, second, we evaluated the impact of genetic risk on health outcomes for these participants. We found a 1.33% combined carrier rate for pathogenic and likely pathogenic (P/LP) genetic variants for HBOC, LS and FH. Of these carriers, 21.9% of participants had clinically relevant disease, among whom 70% had been diagnosed with relevant disease before age 65. Moreover, 90% of the risk carriers had not been previously identified, and less than 19.8% of these had documentation in their medical records of inherited genetic disease risk, including family history. In a direct follow-up survey with all carriers, only 25.2% of individuals reported a family history of relevant disease. Our experience with the HNP suggests that genetic screening in patients could identify at-risk carriers, who would not be otherwise identified in routine care.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Pruebas Genéticas , Genética de Población , Síndrome de Cáncer de Mama y Ovario Hereditario/genética , Hiperlipoproteinemia Tipo II/genética , Adolescente , Adulto , Anciano , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Femenino , Tamización de Portadores Genéticos/métodos , Síndrome de Cáncer de Mama y Ovario Hereditario/diagnóstico , Síndrome de Cáncer de Mama y Ovario Hereditario/patología , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/patología , Persona de Mediana Edad
20.
Curr Opin Lipidol ; 31(3): 119-124, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32332430

RESUMEN

PURPOSE OF REVIEW: Homozygous familial hypercholesterolemia (HoFH) is a rare disorder associated with early atherosclerotic disease due to impairment of the LDL receptor (LDLR) pathway. Because of their molecular defect, current treatment options have limited success in bringing HoFH patient to LDL-C target and morbidity and mortality remain high. We review current and upcoming therapies directed at HoFH, including gene therapy. RECENT FINDINGS: Recent real-world studies have confirmed the strength in lomitapide as a treatment adjunct to statins and other lipid-lowering therapies in HoFH patients. The approval of proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor monoclonal antibodies has also been a welcome addition to the treatment armamentarium offering an additional average reduction in LDL-C levels of 24% when added to background lipid-lowering therapies in this population. Although achieving adequate LDL-C levels in this population is difficult, there are several therapies on the horizon that may help more patients reach goal. Evinacumab, a monoclonal antibody against ANGPTL3, has been shown to substantially reduce LDL-C of an average of 49%, independently of residual LDLR activity. RNA interference targeting PCSK9 and ANGPTL3 shows promise in clinical trials. Adeno-associated virus-mediated gene transfer and gene editing techniques are in early clinical and preclinical development. SUMMARY: LDL-C lowering in HoFH patients remains very challenging. However, novel treatment options are emerging. Upcoming therapies directed at PCSK9 and ANPTL3 may offer additional LDL-C reduction, to help patients achieve adequate LDL-C levels. Gene therapy and gene editing techniques, if proven effective, may offer a unique opportunity to treat patients with a one-time treatment.


Asunto(s)
Proteínas Similares a la Angiopoyetina/genética , LDL-Colesterol/genética , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Proproteína Convertasa 9/genética , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina/antagonistas & inhibidores , Anticuerpos Monoclonales/uso terapéutico , Bencimidazoles/uso terapéutico , LDL-Colesterol/análisis , Homocigoto , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Hipolipemiantes/uso terapéutico , Terapia Molecular Dirigida , Inhibidores de PCSK9 , Receptores de LDL/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...