Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 312
Filtrar
1.
Circulation ; 141(1): 67-79, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31779484

RESUMEN

BACKGROUND: Mutations in low-density lipoprotein (LDL) receptor (LDLR) are one of the main causes of familial hypercholesterolemia, which induces atherosclerosis and has a high lifetime risk of cardiovascular disease. The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system is an effective tool for gene editing to correct gene mutations and thus to ameliorate disease. METHODS: The goal of this work was to determine whether in vivo somatic cell gene editing through the CRISPR/Cas9 system delivered by adeno-associated virus (AAV) could treat familial hypercholesterolemia caused by the Ldlr mutant in a mouse model. We generated a nonsense point mutation mouse line, LdlrE208X, based on a relevant familial hypercholesterolemia-related gene mutation. The AAV-CRISPR/Cas9 was designed to correct the point mutation in the Ldlr gene in hepatocytes and was delivered subcutaneously into LdlrE208X mice. RESULTS: We found that homogeneous LdlrE208X mice (n=6) exhibited severe atherosclerotic phenotypes after a high-fat diet regimen and that the Ldlr mutation was corrected in a subset of hepatocytes after AAV-CRISPR/Cas9 treatment, with LDLR protein expression partially restored (n=6). Compared with the control groups (n=6 each group), the AAV-CRISPR/Cas9 with targeted single guide RNA group (n=6) had significant reductions in total cholesterol, total triglycerides, and LDL cholesterol in the serum, whereas the aorta had smaller atherosclerotic plaques and a lower degree of macrophage infiltration. CONCLUSIONS: Our work shows that in vivo AAV-CRISPR/Cas9-mediated Ldlr gene correction can partially rescue LDLR expression and effectively ameliorate atherosclerosis phenotypes in Ldlr mutants, providing a potential therapeutic approach for the treatment of patients with familial hypercholesterolemia.


Asunto(s)
Aterosclerosis , Sistemas CRISPR-Cas , Dependovirus , Edición Génica , Hiperlipoproteinemia Tipo II , Animales , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Aterosclerosis/terapia , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/metabolismo , Hiperlipoproteinemia Tipo II/patología , Hiperlipoproteinemia Tipo II/terapia , Ratones , Ratones Transgénicos , Mutación Missense , Receptores de LDL/genética , Receptores de LDL/metabolismo
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Curr Opin Lipidol ; 31(3): 111-118, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32332432

RESUMEN

PURPOSE OF REVIEW: This review explores the concepts of monogenic and the so-called polygenic familial hypercholesterolemia and how the identification of familial hypercholesterolemia as a monogenic condition and its separation from polygenic primary hypercholesterolemia may have implications for clinical practice. RECENT FINDINGS: Through genetic testing, a mutation in any of the three known autosomal dominant familial hypercholesterolemia-causing genes is found in 60-80% of cases with a clinical diagnosis of definite familial hypercholesterolemia. As individuals with a polygenic basis for their hypercholesterolemia do not follow the same inheritance pattern observed in monogenic familial hypercholesterolemia, the use of family-based cascade screening in individuals with a polygenic origin is not recommend, as only 30% of relatives have an elevated LDL-C compared to the 50% in monogenic families. The presence of a causative monogenic mutation associates the highest cardiovascular risk vs. not having a mutation or having a polygenic background, providing prognostic information independent of LDL-C. It may also help assess intensity of interventions. Treatment adherence also seems to be higher after monogenic confirmation of hypercholesterolemia. SUMMARY: Knowledge about the genetic status of an individual with clinical familial hypercholesterolemia (monogenic vs. polygenic) can provide a more informed understanding to evaluating risk, managing disease and opportunities for screening strategies.


Asunto(s)
LDL-Colesterol/genética , Predisposición Genética a la Enfermedad , Hiperlipoproteinemia Tipo II/genética , Errores Innatos del Metabolismo Lipídico/genética , Femenino , Pruebas Genéticas , Humanos , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/patología , Errores Innatos del Metabolismo Lipídico/patología , Masculino , Herencia Multifactorial/genética , Mutación/genética , Fenotipo , Factores de Riesgo
8.
IUBMB Life ; 72(4): 782-789, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31633867

RESUMEN

Familial hypercholesterolemia (FH) is an autosomal dominant disorder of lipoprotein metabolism that mainly occurs due to mutations in the low-density lipoprotein receptor gene and is characterized by increased levels of low-density lipoprotein cholesterol, leading to accelerated atherogenesis and premature coronary heart disease. Both innate and adaptive immune responses, which mainly include monocytes, macrophages, neutrophils, T lymphocytes, and B lymphocytes, have been shown to play a key role for the initiation and progression of atherogenesis in the general population. In FH patients, these immune cells have been suggested to play specific pro-atherosclerotic activities, from the initial leukocyte recruitment to plaque rupture. In fact, the accumulation of cholesterol crystals and oxLDL in the vessels in FH patients is particularly high, with consequent abnormal mobilization of immune cells and secretion of various pro-inflammatory and chemokines. In addition, cholesterol accumulation in immune cells is exaggerated with chronic exposure to relevant pro-atherosclerotic triggers. The topics considered in this review may provide a more specific focus on the immune system alterations in FH and open new insights toward immune cells as potential therapeutic targets in FH.


Asunto(s)
Hiperlipoproteinemia Tipo II/inmunología , Hiperlipoproteinemia Tipo II/patología , Aterosclerosis , Linfocitos B/inmunología , Linfocitos B/patología , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Macrófagos/metabolismo , Macrófagos/patología , Neutrófilos/inmunología , Neutrófilos/patología , Linfocitos T/inmunología , Linfocitos T/patología
9.
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
10.
Clin Genet ; 97(3): 457-466, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31893465

RESUMEN

Familial hypercholesterolaemia (FH) is a monogenic disorder characterised by high low-density lipoprotein cholesterol (LDL-C) concentrations and increased cardiovascular risk. However, in clinically defined FH cohorts worldwide, an FH-causing variant is only found in 40%-50% of the cases. The aim of this work was to characterise the genetic cause of the FH phenotype in Portuguese clinical FH patients. Between 1999 and 2017, 731 index patients (311 children and 420 adults) who met the Simon Broome diagnostic criteria had been referred to our laboratory. LDLR, APOB, PCSK9, APOE, LIPA, LDLRAP1, ABCG5/8 genes were analysed by polymerase chain reaction amplification and Sanger sequencing. The 6-SNP LDL-C genetic risk score (GRS) for polygenic hypercholesterolaemia was validated in the Portuguese population and cases with a GRS over the 25th percentile were considered to have a high likelihood of polygenic hypercholesterolaemia. An FH-causing mutation was found in 39% of patients (94% in LDLR, 5% APOB and 1% PCSK9), while at least 29% have polygenic hypercholesterolaemia and 1% have other lipid disorders. A genetic cause for the FH phenotype was found in 503 patients (69%). All known causes of the FH phenotype should be investigated in FH cohorts to ensure accurate diagnosis and appropriate management.


Asunto(s)
LDL-Colesterol/genética , Predisposición Genética a la Enfermedad , Hiperlipoproteinemia Tipo II/genética , Errores Innatos del Metabolismo Lipídico/genética , Adolescente , Adulto , Apolipoproteína B-100/genética , Niño , Femenino , Humanos , Hiperlipoproteinemia Tipo II/patología , Errores Innatos del Metabolismo Lipídico/patología , Masculino , Persona de Mediana Edad , Herencia Multifactorial/genética , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Proproteína Convertasa 9/genética , Receptores de LDL/genética , Adulto Joven
11.
Arterioscler Thromb Vasc Biol ; 39(11): 2338-2352, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31554418

RESUMEN

OBJECTIVE: In an adult porcine model of familial hypercholesterolemia (FH), coronary plaque development was characterized. To elucidate the underlying mechanisms of the observed inter-individual variation in disease severity, detailed lipoprotein profiles were determined. Approach and Results: FH pigs (3 years old, homozygous LDLR R84C mutation) received an atherogenic diet for 12 months. Coronary atherosclerosis development was monitored using serial invasive imaging and histology. A pronounced difference was observed between mildly diseased pigs which exclusively developed early lesions (maximal plaque burden, 25% [23%-34%]; n=5) and advanced-diseased pigs (n=5) which developed human-like, lumen intruding plaques (maximal plaque burden, 69% [57%-77%]) with large necrotic cores, intraplaque hemorrhage, and calcifications. Advanced-diseased pigs and mildly diseased pigs displayed no differences in conventional risk factors. Additional plasma lipoprotein profiling by size-exclusion chromatography revealed 2 different LDL (low-density lipoprotein) subtypes: regular and larger LDL. Cholesterol, sphingosine-1-phosphate, ceramide, and sphingomyelin levels were determined in these LDL-subfractions using standard laboratory techniques and high-pressure liquid chromatography mass-spectrometry analyses, respectively. At 3 months of diet, regular LDL of advanced-diseased pigs contained relatively more cholesterol (LDL-C; regular/larger LDL-C ratio 1.7 [1.3-1.9] versus 0.8 [0.6-0.9]; P=0.008) than mildly diseased pigs, while larger LDL contained more sphingosine-1-phosphate, ceramides, and sphingomyelins. Larger and regular LDL was also found in plasma of 3 patients with homozygous FH with varying LDL-C ratios. CONCLUSIONS: In our adult FH pig model, inter-individual differences in atherosclerotic disease severity were directly related to the distribution of cholesterol and sphingolipids over a distinct LDL profile with regular and larger LDL shortly after the diet start. A similar LDL profile was detected in patients with homozygous FH.


Asunto(s)
LDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/patología , Placa Aterosclerótica/sangre , Placa Aterosclerótica/patología , Animales , LDL-Colesterol/clasificación , Dieta Aterogénica , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico por imagen , Masculino , Placa Aterosclerótica/diagnóstico por imagen , Índice de Severidad de la Enfermedad , Esfingolípidos/sangre , Porcinos
12.
Arterioscler Thromb Vasc Biol ; 39(5): 945-955, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30866660

RESUMEN

Objective- Heterozygous familial hypercholesterolemia (FH) is the most common genetic disorder associated with premature atherosclerotic cardiovascular disease. Circulating microvesicles (cMV) are released when cells are activated. We investigated whether cMV could provide information on coronary calcification and atherosclerosis in FH patients. Approach and Results- Eighty-two patients (mean of 44±9 years old) with molecular diagnosis of heterozygous FH and asymptomatic cardiovascular disease were investigated. Atherosclerotic plaque characterization was performed by computed tomography angiography, and Agatston coronary calcium score and plaque composition sum were calculated. cMV were quantified by flow cytometry using AV (annexin V) and cell surface-specific antibodies. Of the 82 FH patients, 48 presented atherosclerotic plaque. Patients with atherosclerosis were men and older in a higher percentage than patients without atherosclerotic plaque. FH patients with atherosclerotic plaque showed higher levels of total AV+ cMV, cMV AV+ from platelet origin, from granulocytes and neutrophils, and cMV AV+/- from endothelial cells than FH-patients without atherosclerotic plaque. Plaque composition sum correlated with platelet- and endothelial-derived cMV, and Agatston coronary calcium score correlated with granulocyte-, platelet-, and endothelial-derived cMV. Receiver operating characteristic curve analyses indicated that the cluster of platelet-, granulocyte-, neutrophil, and endothelial-derived cMV considered together, added significant predictive value to the specific SAFEHEART (Spanish Familial Hypercholesterolaemia Cohort Study) risk equation for plaque presence (area under the curve=0.866, 95% CI, 0.775-0.958; P<0.0001, P=0.030 for the increment of the area under the curve). Conclusions- Endothelial-, granulocyte-, neutrophil- and platelet-derived cMV discriminate and map coronary atherosclerotic plaque and calcification in asymptomatic patients with FH. Liquid biopsy of cMV may be a surrogate biomarker of coronary atherosclerotic plaque burden in FH patients.


Asunto(s)
Enfermedad de la Arteria Coronaria/patología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Hiperlipoproteinemia Tipo II/patología , Calcificación Vascular/diagnóstico por imagen , Adulto , Área Bajo la Curva , Enfermedades Asintomáticas , Estudios de Cohortes , Enfermedad de la Arteria Coronaria/diagnóstico por imagen , Femenino , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico por imagen , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Biopsia Líquida , Masculino , Persona de Mediana Edad , Placa Aterosclerótica/diagnóstico por imagen , Placa Aterosclerótica/patología , Pronóstico , Curva ROC , Estudios Retrospectivos , Medición de Riesgo , Análisis de Supervivencia , Calcificación Vascular/patología
13.
Arterioscler Thromb Vasc Biol ; 39(6): 1172-1181, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31070479

RESUMEN

Objective- Circulating microvesicles (cMVs) exert regulatory roles in atherothrombosis. Patients with familial hypercholesterolemia (FH) that are at high risk for premature cardiovascular events (CVEs) have previously shown high levels of cMVs related to disease severity. However, much remains unknown about their value as markers of CVE. We sought to investigate the prognostic cMV signature for future major CVE presentation in patients with FH. Approach and Results- Liquid biopsies from genetically characterized patients with FH from the SAFEHEART (Spanish Familial Hypercholesterolemia Cohort Study)-cohort without clinical manifestation of disease at entry that were going to suffer a CVE within a mean period of 3.3±2.6 years postsampling (CVE, N=92) and from age/cardiovascular risk factor/treatment-matched patients with FH that did not suffer an event within the same time-period (non-CVE, N=48) were investigated. cMVs were phenotyped by flow cytometry to identify activated parental cells. Patients with CVE had higher number of overall procoagulant annexin V+-cMVs than non-CVE ( P<0.05). Pan-leukocyte-derived and neutrophil-derived cMVs, as well as activated platelet-derived cMVs, were significantly higher in patients with CVE. Baseline number of cMVs derived from lymphocytes, neutrophils, and activated platelets were positively associated with mortality at follow-up ( P<0.05). Patient-risk calculated by classical cardiovascular risk-factor scores did not correlate with cMVs. Inclusion of the cMV signature into the SAFEHEART risk model for patients with FH for the prediction of ischemic events increased the area under the curve from 0.603±0.050 to 0.768±0.042 ( P<0.005). Conclusions- Patients with FH who are going to suffer a CVE within a mean period of 3.3 years, despite being treated according to guidelines, have ongoing innate immune cell and platelet activation. The proposed cMV signature is a prognostic marker for accelerated atherosclerosis and clinical event presentation in patients with FH.


Asunto(s)
Plaquetas/patología , Isquemia Encefálica/patología , Micropartículas Derivadas de Células/patología , Hiperlipoproteinemia Tipo II/patología , Leucocitos/patología , Biopsia Líquida , Isquemia Miocárdica/patología , Accidente Cerebrovascular/patología , Biomarcadores/sangre , Plaquetas/metabolismo , Isquemia Encefálica/sangre , Isquemia Encefálica/genética , Isquemia Encefálica/mortalidad , Micropartículas Derivadas de Células/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/mortalidad , Leucocitos/metabolismo , Lípidos/sangre , Masculino , Persona de Mediana Edad , Mutación , Isquemia Miocárdica/sangre , Isquemia Miocárdica/genética , Isquemia Miocárdica/mortalidad , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Medición de Riesgo , Factores de Riesgo , España , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/mortalidad , Factores de Tiempo
14.
Nano Lett ; 19(4): 2562-2567, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30848605

RESUMEN

The fundamental task of lipoprotein particles is extracellular transport of cholesterol, lipids, and fatty acids. Besides, cholesterol-rich apoB-containing lipoprotein particles (i.e., low density lipoprotein LDL) are key players in progression of atherosclerotic cardiovascular disease and are associated with familial hypercholesterolemia (FH). So far, lipoprotein particle binding to the cell membrane and subsequent cargo transfer is directly linked to the lipoprotein receptors on the target cell surface. However, our observations showed that lipoprotein particle cargo transport takes place even in the absence of the receptor. This finding suggests that an alternative mechanism for lipoprotein-particle/membrane interaction, besides the receptor-mediated one, exists. Here, we combined several complementary biophysical techniques to obtain a comprehensive view on the nonreceptor mediated LDL-particle/membrane. We applied a combination of atomic force and single-molecule-sensitive fluorescence microscopy (AFM and SMFM) to investigate the LDL particle interaction with membranes of increasing complexity. We observed direct transfer of fluorescently labeled amphiphilic lipid molecules from LDL particles into the pure lipid bilayer. We further confirmed cargo transfer by fluorescence cross-correlation spectroscopy (FCCS) and spectral imaging of environment-sensitive probes. Moreover, the integration of the LDL particle into the membranes was directly visualized by high-speed atomic force microscopy (HS-AFM) and cryo-electron microscopy (cryo-EM). Overall, our data show that lipoprotein particles are able to incorporate into lipid membranes upon contact to transfer their cargo in the absence of specific receptors.


Asunto(s)
Membrana Celular/ultraestructura , Enfermedad de la Arteria Coronaria/patología , Hiperlipoproteinemia Tipo II/metabolismo , Lipoproteínas LDL/química , Apolipoproteínas B/química , Fenómenos Biofísicos , Membrana Celular/química , Membrana Celular/efectos de los fármacos , Enfermedad de la Arteria Coronaria/metabolismo , Microscopía por Crioelectrón , Progresión de la Enfermedad , Colorantes Fluorescentes/química , Colorantes Fluorescentes/farmacología , Humanos , Hiperlipoproteinemia Tipo II/patología , Membrana Dobles de Lípidos/química , Lipoproteínas LDL/farmacología , Lipoproteínas LDL/ultraestructura , Microscopía de Fuerza Atómica
15.
J Biol Chem ; 293(24): 9176-9187, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29712723

RESUMEN

Cardiovascular disease risk depends on high-density lipoprotein (HDL) function, not HDL-cholesterol. Isolevuglandins (IsoLGs) are lipid dicarbonyls that react with lysine residues of proteins and phosphatidylethanolamine. IsoLG adducts are elevated in atherosclerosis. The consequences of IsoLG modification of HDL have not been studied. We hypothesized that IsoLG modification of apoA-I deleteriously alters HDL function. We determined the effect of IsoLG on HDL structure-function and whether pentylpyridoxamine (PPM), a dicarbonyl scavenger, can preserve HDL function. IsoLG adducts in HDL derived from patients with familial hypercholesterolemia (n = 10, 233.4 ± 158.3 ng/mg) were found to be significantly higher than in healthy controls (n = 7, 90.1 ± 33.4 pg/mg protein). Further, HDL exposed to myeloperoxidase had elevated IsoLG-lysine adducts (5.7 ng/mg protein) compared with unexposed HDL (0.5 ng/mg protein). Preincubation with PPM reduced IsoLG-lysine adducts by 67%, whereas its inactive analogue pentylpyridoxine did not. The addition of IsoLG produced apoA-I and apoA-II cross-links beginning at 0.3 molar eq of IsoLG/mol of apoA-I (0.3 eq), whereas succinylaldehyde and 4-hydroxynonenal required 10 and 30 eq. IsoLG increased HDL size, generating a subpopulation of 16-23 nm. 1 eq of IsoLG decreased HDL-mediated [3H]cholesterol efflux from macrophages via ABCA1, which corresponded to a decrease in HDL-apoA-I exchange from 47.4% to only 24.8%. This suggests that IsoLG inhibits apoA-I from disassociating from HDL to interact with ABCA1. The addition of 0.3 eq of IsoLG ablated HDL's ability to inhibit LPS-stimulated cytokine expression by macrophages and increased IL-1ß expression by 3.5-fold. The structural-functional effects were partially rescued with PPM scavenging.


Asunto(s)
Hiperlipoproteinemia Tipo II/metabolismo , Lipoproteínas HDL/metabolismo , Aldehídos/metabolismo , Animales , Apolipoproteína A-I/metabolismo , Apolipoproteína A-II/metabolismo , Células Cultivadas , Colesterol/metabolismo , Femenino , Humanos , Hiperlipoproteinemia Tipo II/patología , Cetonas/metabolismo , Metabolismo de los Lípidos , Lípidos , Lipoproteínas HDL/química , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fosfatidiletanolaminas/metabolismo
16.
Microcirculation ; 26(6): e12539, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30821858

RESUMEN

OBJECTIVE: Swine with familial hypercholesterolemia (FH) exhibit attenuated exercise-induced systemic vasodilation that is restored by phosphodiesterase 5 (PDE5) inhibition. Whether the impacts of FH and PDE5 inhibition to impair and restore exercise-induced vasodilation, respectively, results from tissue-specific or generalized effects remains unclear. Thus, we hypothesized that FH induces generalized impairment of skeletal muscle vasodilation that would be alleviated by PDE5 inhibition. METHODS: Systemic vascular responses to exercise were assessed in chronically instrumented normal and FH swine before and after PDE5 inhibition with EMD360527. Skeletal muscle and organ blood flows and conductances were determined via the microsphere technique. RESULTS: As previously reported, vs normal swine, FH swine have pronounced elevation of total cholesterol and impaired exercise-induced vasodilation that is restored by PDE5 inhibition. Blood flows to several, not all, skeletal muscle vascular beds were severely impaired by FH associated with reduced blood flow to many visceral organs. PDE5 inhibition differentially impacted skeletal muscle and organ blood flows in normal and FH swine. CONCLUSIONS: These data indicate that FH induces regional, not generalized, vasomotor dysfunction and that FH and normal swine exhibit unique tissue blood flow responses to PDE5 inhibition thereby adding to accumulating evidence of vascular bed-specific dysfunction in co-morbid conditions.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hiperlipoproteinemia Tipo II , Músculo Esquelético , Inhibidores de Fosfodiesterasa 5/farmacología , Condicionamiento Físico Animal , Vasodilatación/efectos de los fármacos , Animales , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Hiperlipoproteinemia Tipo II/enzimología , Hiperlipoproteinemia Tipo II/patología , Hiperlipoproteinemia Tipo II/fisiopatología , Masculino , Músculo Esquelético/irrigación sanguínea , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Porcinos
17.
Clin Transplant ; 33(12): e13730, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31626710

RESUMEN

BACKGROUND: Familial hypercholesterolemia (FH) is an autosomal dominant genetic disorder characterized by premature mortal cardiovascular complications. Liver transplantation (LT) is the only curative treatment option. In this study, the long-term clinical follow-up data of 8 patients who underwent LT with a diagnosis of FH in our center are presented. MATERIALS AND METHODS: A total of 638 LT were performed between December 1985 and June 2019 at Baskent University, of which 8 patients underwent LT with a diagnosis of FH and were evaluated retrospectively. RESULTS: Of the 8 patients, 4 underwent deceased donor and 4 living donor transplantation. Five patients had preoperative cardiovascular disease and consequent interventional operations. There was significant reduction in postoperative LDL-C and TC levels starting from the first week, and stabilizing at the first month and first year. The median survival time of patients was 5 years (2-12 years). All patients are still alive. None of the complications of patients with preoperative cardiovascular complications had progressed. CONCLUSION: Liver transplantation is the preferred curative treatment for the pathophysiology of FH. In our study, LDL-C levels were brought under control with LT performed on patients with FH. Median 5-year follow-up of patients showed that the progression of cardiac complications was abated.


Asunto(s)
Hiperlipoproteinemia Tipo II/cirugía , Trasplante de Hígado/métodos , Donantes de Tejidos/estadística & datos numéricos , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Hiperlipoproteinemia Tipo II/patología , Masculino , Estudios Retrospectivos , Resultado del Tratamiento , Adulto Joven
18.
Clin Chem Lab Med ; 57(7): 1102-1110, 2019 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-30710474

RESUMEN

Background Familial hypercholesterolemia (FH) is a genetic disorder caused by mutations in genes involved in low-density lipoprotein (LDL) uptake (LDLR, APOB and PCSK9). Genetic diagnosis is particularly useful in asymptomatic children allowing for the detection of definite FH patients. Furthermore, defining their genetic status may be of considerable importance as the compound heterozygous status is much more severe than the heterozygous one. Our study aims at depicting the genetic background of an Italian pediatric population with FH focusing on the correlation between lipid profile and genetic status. Methods Out of 196 patients with clinically suspected FH (LDL-cholesterol [LDL-C] levels above 3.37 mmol/L, cholesterol level above 6.46 mmol/L in a first-degree relative or the presence of premature cardiovascular acute disease in a first/second-degree relative), we screened 164 index cases for mutations in the LDLR, APOB and PCSK9 genes. Results Patients with mutations (129/164) showed increased levels of LDL-C, 95th percentile-adjusted LDL-C and LDL/high-density lipoprotein (HDL) ratio and decreased levels of HDL-C, adjusted HDL-C. The association of the LDL/HDL ratio with the presence of mutations was assessed independently of age, (body mass index) BMI, parental hypercholesterolemia, premature coronary artery disease (CAD), triglycerides by multivariate logistic regression (odds ratio [OR]=1.701 [1.103-2.621], p=0.016). The LDL/HDL ratio gradually increased from patients without mutations to patients with missense mutations, null mutations and compound heterozygotes. Conclusions In conclusion, the LDL/HDL ratio proved to be a better parameter than LDL-C for discriminating patients with from patients without mutations across different genetic statuses.


Asunto(s)
HDL-Colesterol/sangre , LDL-Colesterol/sangre , Hiperlipoproteinemia Tipo II/genética , Lípidos/sangre , Adolescente , Apolipoproteínas B/genética , Niño , Femenino , Heterocigoto , Humanos , Hiperlipoproteinemia Tipo II/patología , Modelos Logísticos , Mutación con Pérdida de Función , Masculino , Mutación Missense , Oportunidad Relativa , Proproteína Convertasa 9/genética , Receptores de LDL/genética
19.
J Endocrinol Invest ; 42(5): 591-598, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30276670

RESUMEN

PURPOSE: Though type 2 diabetes mellitus (T2DM) is an important and independent risk factor for coronary artery disease (CAD) in the general population, the impact of T2DM on CAD in patients with familial hypercholesterolemia (FH) is less understood. Thus, the current study aimed to examine the features of FH patients with T2DM and explore the effects of T2DM on CAD in FH. METHODS: A total of 289 clinical heterozygous FH (HeFH) patients diagnosed with Dutch Lipid Clinic Criteria were consecutively recruited and divided into a T2DM group (n = 58) and non-T2DM group (n = 231). Clinical characteristics and laboratory findings were compared between the two groups. Target exome sequencing was used for gene mutation analysis. RESULTS: HeFH patients with T2DM had significantly higher levels of triglycerides, body mass index and free fatty acids than did non-T2DM patients; moreover, patients with T2DM more frequently exhibited hypertension. However, the spectrum of FH-causing mutations was not significantly different (p = 0.061). Notably, patients with T2DM had higher prevalence of CAD (p = 0.012) and higher Gensini Score (p = 0.002). The regression analysis confirmed that HbA1c was an independent risk factor for both the presence and severity of CAD [OR 2.321 (1.098-4.904), p = 0.027; OR 1.349 (1.032-1.762), p = 0.028, respectively] in patients with HeFH. CONCLUSIONS: Although there were not many differences in the clinical, lipid and genetic aspects of HeFH patients with and without T2DM, T2DM and HbA1c were associated with worse coronary lesions, suggesting that diabetes and the degree of blood glucose control are also important determinants of cardiovascular disease in these patients.


Asunto(s)
Biomarcadores/análisis , Enfermedad de la Arteria Coronaria/etiología , Diabetes Mellitus Tipo 2/complicaciones , Hiperlipoproteinemia Tipo II/complicaciones , Glucemia/análisis , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Femenino , Estudios de Seguimiento , Hemoglobina Glucada/análisis , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Lípidos/análisis , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Riesgo , Triglicéridos/metabolismo
20.
Lipids Health Dis ; 18(1): 80, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30935416

RESUMEN

BACKGROUND: Evidence regarding the correlation between lipoproteins and telomere length in US adults is limited. We aimed to investigate whether lipoproteins was associated with telomere length using US National Health and Nutrition Examination Survey (NHANES) database. METHODS: A total of 6468 selected participants were identified in the NHANES Data Base (1999-2002). The independent and dependent variables were lipoproteins and telomere length, respectively. The covariates included demographic data, dietary data, physical examination data, and comorbidities. RESULTS: In fully-adjusted model, we found that 0.1 differences of telomere length were positively associated with HDL-C [0.19 (95% CI 0.07, 0.31)], while the associations between LDL-C [0.19 (95% CI -0.27, 0.65)], TG [- 1.00 (95% CI -2.09, 0.07) and telomere length were not detected. By nonlinearity test, only the relationship between HDL-C and telomere length was nonlinear. The inflection point we got was 1.25. On the left side of the inflection point (telomere length ≤ 1.25), a difference in 0.1 of telomere length was associated with 0.50 difference in HDL-C. CONCLUSION: After adjusting for demographic data, dietary data, physical examination data, and comorbidities, telomere length is not associated with LDL-C and TG, but is positively associated with HDL-C when telomere length is less than 1.25.


Asunto(s)
Hiperlipoproteinemia Tipo II/epidemiología , Lipoproteínas/genética , Homeostasis del Telómero/genética , Telómero/genética , Adulto , Anciano , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta , Femenino , Humanos , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/patología , Leucocitos/citología , Leucocitos/metabolismo , Lipoproteínas/sangre , Masculino , Persona de Mediana Edad , Encuestas Nutricionales , Factores de Riesgo , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA