Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38871496

RESUMEN

AIMS: Familial Hypercholesterolemia (FH) is a genetic disorder of lipoprotein metabolism that causes an increased risk of premature atherosclerotic cardiovascular disease (ASCVD). Although early diagnosis and treatment of FH can significantly improve the cardiovascular prognosis, this disorder is underdiagnosed and undertreated. For these reasons the Italian Society for the Study of Atherosclerosis (SISA) assembled a Consensus Panel with the task to provide guidelines for FH diagnosis and treatment. DATA SYNTHESIS: Our guidelines include: i) an overview of the genetic complexity of FH and the role of candidate genes involved in LDL metabolism; ii) the prevalence of FH in the population; iii) the clinical criteria adopted for the diagnosis of FH; iv) the screening for ASCVD and the role of cardiovascular imaging techniques; v) the role of molecular diagnosis in establishing the genetic bases of the disorder; vi) the current therapeutic options in both heterozygous and homozygous FH. Treatment strategies and targets are currently based on low-density lipoprotein cholesterol (LDL-C) levels, as the prognosis of FH largely depends on the magnitude of LDL-C reduction achieved by lipid-lowering therapies. Statins with or without ezetimibe are the mainstay of treatment. Addition of novel medications like PCSK9 inhibitors, ANGPTL3 inhibitors or lomitapide in homozygous FH results in a further reduction of LDL-C levels. LDL apheresis is indicated in FH patients with inadequate response to cholesterol-lowering therapies. CONCLUSION: FH is a common, treatable genetic disorder and, although our understanding of this disease has improved, many challenges still remain with regard to its identification and management.

2.
Curr Opin Lipidol ; 28(3): 267-272, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28323660

RESUMEN

PURPOSE OF REVIEW: The systematic analysis of the major candidate genes in autosomal dominant hypercholesterolemia (ADH) and the use of next-generation sequencing (NGS) technology have made possible the discovery of several rare gene variants whose pathogenic effect in most cases remains poorly defined. RECENT FINDINGS: One major advance in the field has been the adoption of a set of international guidelines for the assignment of pathogenicity to low-density lipoprotein receptor (LDLR) gene variants based on the use of softwares, complemented with data available from literature and public databases. The clinical impact of several novel rare variants in LDLR, APOB, PCSK9, APOE genes have been reported in large studies describing patients with ADH found to be homozygotes/compound heterozygotes, double heterozygotes, or simple heterozygotes. In-vitro functional studies have been conducted to clarify the effect of some rare ApoB variants on LDL binding to LDLR and the impact of a rare ApoE variant on the uptake of VLDL and LDL by hepatocytes. SUMMARY: The update of the ADH gene variants database and the classification of variants in categories of pathogenicity is a major advance in the understanding the pathophysiology of ADH and in the management of this disorder. The studies of molecularly characterized patients with ADH have emphasized the impact of a specific variant and the variable clinical expression of different genotypes. The functional studies of some variants have increased our understanding of the molecular bases of some forms of ADH.


Asunto(s)
Variación Genética , Hiperlipoproteinemia Tipo II/genética , Apolipoproteínas B/genética , Apolipoproteínas E/genética , Humanos , Proproteína Convertasa 9/genética , Receptores de LDL/genética
3.
Biochim Biophys Acta ; 1861(11): 1623-1633, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27487388

RESUMEN

We describe two new hypolipidemic patients with very low plasma triglyceride and apolipoprotein B (apoB) levels with plasma lipid profiles similar to abetalipoproteinemia (ABL) patients. In these patients, we identified two previously uncharacterized missense mutations in the microsomal triglyceride transfer protein (MTP) gene, R46G and D361Y, and studied their functional effects. We also characterized three missense mutations (H297Q, D384A, and G661A) reported earlier in a familial hypobetalipoproteinemia patient. R46G had no effect on MTP expression or function and supported apoB secretion. H297Q, D384A, and G661A mutants also supported apoB secretion similarly to WT MTP. Contrary to these four missense mutations, D361Y was unable to support apoB secretion. Functional analysis revealed that this mutant was unable to bind protein disulfide isomerase (PDI) or transfer lipids. The negative charge at residue 361 was critical for MTP function as D361E was able to support apoB secretion and transfer lipids. D361Y most likely disrupts the tightly packed middle α-helical region of MTP, mitigates PDI binding, abolishes lipid transfer activity, and causes ABL. On the other hand, the hypolipidemia in the other two patients was not due to MTP dysfunction. Thus, in this study of five missense mutations spread throughout MTP's three structural domains found in three hypolipidemic patients, we found that four of the mutations did not affect MTP function. Thus, novel mutations that cause severe hypolipidemia probably exist in other genes in these patients, and their recognition may identify novel proteins involved in the synthesis and/or catabolism of plasma lipoproteins.


Asunto(s)
Abetalipoproteinemia/genética , Proteínas Portadoras/química , Proteínas Portadoras/genética , Hipobetalipoproteinemias/genética , Mutación Missense/genética , Abetalipoproteinemia/sangre , Secuencia de Aminoácidos , Animales , Apolipoproteínas B/metabolismo , Células COS , Niño , Chlorocebus aethiops , Simulación por Computador , Retículo Endoplásmico/metabolismo , Regulación de la Expresión Génica , Humanos , Hipobetalipoproteinemias/sangre , Lactante , Metabolismo de los Lípidos/genética , Masculino , Fenotipo , Unión Proteica , Proteína Disulfuro Isomerasas/metabolismo , Alineación de Secuencia , Relación Estructura-Actividad , Triglicéridos/metabolismo , Vitaminas/sangre , Adulto Joven
4.
Biochim Biophys Acta ; 1861(4): 371-9, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26825690

RESUMEN

We have previously identified a deletion mutant of human apoB [apoB (Thr26_Tyr27del)] in a subject with primary hypobetalipoproteinemia. The present study determined the effect of Thr26_Tyr27del mutation on apoB secretion using transfected McA-RH7777 cells. Transient or stable transfection of apoB-48 containing the Thr26_Tyr27del mutation showed drastically reduced secretion of the mutant as compared to wild-type apoB-48. No lipoproteins containing the mutant apoB-48 were secreted into the medium. Incubation of transfected cells in a lipid-rich medium in the presence of cycloheximide showed rapid turnover of cell-associated mutant apoB-48 as compared to that of wild-type apoB-48. Immunofluorescence experiments showed that the mutant apoB-48 was mostly localized in the endoplasmic reticulum. Treatment with the proteasomal inhibitor MG132 markedly attenuated the turnover of cell-associated mutant apoB-48, whereas treatment with inhibitors of autophagosomal/lysosomal function (e.g. 3-MA or ammonium chloride) had no effect. Taken together, these results indicated that the defective secretion of the Thr26_Tyr27del mutant was associated with increased intracellular degradation of apoB through the proteasome-dependent pathway.


Asunto(s)
Apolipoproteína B-100/genética , Apolipoproteína B-48/genética , Hipobetalipoproteinemias/genética , Eliminación de Secuencia , Apolipoproteína B-100/metabolismo , Apolipoproteína B-48/metabolismo , Línea Celular , Análisis Mutacional de ADN , Retículo Endoplásmico/metabolismo , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Hipobetalipoproteinemias/metabolismo , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología , Proteolisis , Factores de Tiempo , Transfección
5.
J Biol Chem ; 290(43): 25863-75, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26350457

RESUMEN

Sphingolipids, a large family of bioactive lipids, are implicated in stress responses, differentiation, proliferation, apoptosis, and other physiological processes. Aberrant plasma levels of sphingolipids contribute to metabolic disease, atherosclerosis, and insulin resistance. They are fairly evenly distributed in high density and apoB-containing lipoproteins (B-lps). Mechanisms involved in the transport of sphingolipids to the plasma are unknown. Here, we investigated the role of microsomal triglyceride transfer protein (MTP), required for B-lp assembly and secretion, in sphingolipid transport to the plasma. Abetalipoproteinemia patients with deleterious mutations in MTP and absence of B-lps had significantly lower plasma ceramide and sphingomyelin but normal hexosylceramide, lactosylceramide, and different sphingosines compared with unaffected controls. Furthermore, similar differential effects on plasma sphingolipids were seen in liver- and intestine-specific MTP knock-out (L,I-Mttp(-/-)) mice, suggesting that MTP specifically plays a role in the regulation of plasma ceramide and sphingomyelin. We hypothesized that MTP deficiency may affect either their synthesis or secretion. MTP deficiency had no effect on ceramide and sphingomyelin synthesis but reduced secretion from primary hepatocytes and hepatoma cells. Therefore, MTP is involved in ceramide and sphingomyelin secretion but not in their synthesis. We also found that MTP transferred these lipids between vesicles in vitro. Therefore, we propose that MTP might regulate plasma ceramide and sphingomyelin levels by transferring these lipids to B-lps in the liver and intestine and facilitating their secretion.


Asunto(s)
Proteínas Portadoras/fisiología , Ceramidas/sangre , Esfingomielinas/sangre , Animales , Línea Celular , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
6.
Arterioscler Thromb Vasc Biol ; 33(8): 2021-5, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23723369

RESUMEN

OBJECTIVE: In familial hypobetalipoproteinemia, fatty liver is a characteristic feature, and there are several reports of associated cirrhosis and hepatocarcinoma. We investigated a large kindred in which low-density lipoprotein cholesterol, fatty liver, and hepatocarcinoma displayed an autosomal dominant pattern of inheritance. APPROACH AND RESULTS: The proband was a 25-year-old female with low plasma cholesterol and hepatic steatosis. Low plasma levels of total cholesterol and fatty liver were observed in 10 more family members; 1 member was affected by liver cirrhosis, and 4 more subjects died of either hepatocarcinoma or carcinoma on cirrhosis. To identify the causal mutation in this family, we performed exome sequencing in 2 participants with hypocholesterolemia and fatty liver. Approximately 22 400 single nucleotide variants were identified in each sample. After variant filtering, 300 novel shared variants remained. A nonsense variant, p.K2240X, attributable to an A>T mutation in exon 26 of APOB (c.6718A>T) was identified, and this variant was confirmed by Sanger sequencing. The gentotypic analysis of 16 family members in total showed that this mutation segregated with the low cholesterol trait. In addition, genotyping of the PNPLA3 p.I148M did not show significant frequency differences between carriers and noncarriers of the c.6718A>T APOB gene mutation. CONCLUSIONS: We used exome sequencing to discover a novel nonsense mutation in exon 26 of APOB (p.K2240X) responsible for low cholesterol and fatty liver in a large kindred. This mutation may also be responsible for cirrhosis and liver cancer in this family.


Asunto(s)
Apolipoproteínas B/genética , Codón sin Sentido , Hígado Graso/genética , Hipobetalipoproteinemias/genética , Neoplasias Hepáticas/genética , Adulto , Apolipoproteínas B/sangre , Colesterol/sangre , Colesterol/genética , Exoma/genética , Salud de la Familia , Hígado Graso/sangre , Femenino , Humanos , Hipobetalipoproteinemias/sangre , Neoplasias Hepáticas/sangre , Masculino , Persona de Mediana Edad , Linaje , Adulto Joven
7.
Eur J Prev Cardiol ; 31(8): 1038-1047, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38374534

RESUMEN

AIMS: The availability of novel lipid-lowering therapies (LLTs) has remarkably changed the clinical management of homozygous familial hypercholesterolaemia (HoFH). The impact of these advances was evaluated in a cohort of 139 HoFH patients followed in a real-world clinical setting. METHODS AND RESULTS: The clinical characteristics of 139 HoFH patients, along with information about LLTs and low-density lipoprotein cholesterol (LDL-C) levels at baseline and after a median follow-up of 5 years, were retrospectively retrieved from the records of patients enrolled in the LIPid transport disorders Italian GEnetic Network-Familial Hypercholesterolaemia (LIPIGEN-FH) Registry. The annual rates of major atherosclerotic cardiovascular events (MACE-plus) during follow-up were compared before and after baseline. Additionally, the lifelong survival free from MACE-plus was compared with that of the historical LIPIGEN HoFH cohort. At baseline, LDL-C level was 332 ± 138 mg/dL. During follow-up, the potency of LLTs was enhanced and, at the last visit, 15.8% of patients were taking quadruple therapy. Consistently, LDL-C decreased to an average value of 124 mg/dL corresponding to a 58.3% reduction (Pt < 0.001), with the lowest value (∼90 mg/dL) reached in patients receiving proprotein convertase subtilisin/kexin type 9 inhibitors and lomitapide and/or evinacumab as add-on therapies. The average annual MACE-plus rate in the 5-year follow-up was significantly lower than that observed during the 5 years before baseline visit (21.7 vs. 56.5 per 1000 patients/year; P = 0.0016). CONCLUSION: Our findings indicate that the combination of novel and conventional LLTs significantly improved LDL-C control with a signal of better cardiovascular prognosis in HoFH patients. Overall, these results advocate the use of intensive, multidrug LLTs to effectively manage HoFH.


Contemporary real-world data from the Italian cohort of patients affected by homozygous familial hypercholesterolaemia demonstrated that the addition of novel, low-density lipoprotein receptor (LDLR)-independent medications to conventional therapies allowed the achievement of unprecedented low-density lipoprotein cholesterol (LDL-C) values with a trend towards a reduction of cardiovascular risk.


Asunto(s)
LDL-Colesterol , Hiperlipoproteinemia Tipo II , Sistema de Registros , Humanos , Hiperlipoproteinemia Tipo II/tratamiento farmacológico , Hiperlipoproteinemia Tipo II/sangre , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/diagnóstico , Masculino , Femenino , Italia/epidemiología , LDL-Colesterol/sangre , Estudios Retrospectivos , Adulto , Persona de Mediana Edad , Homocigoto , Resultado del Tratamiento , Enfermedades Cardiovasculares/prevención & control , Enfermedades Cardiovasculares/epidemiología , Medición de Riesgo , Factores de Tiempo , Biomarcadores/sangre , Factores de Riesgo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Anticolesterolemiantes/uso terapéutico , Predisposición Genética a la Enfermedad , Inhibidores de PCSK9/uso terapéutico , Hipolipemiantes/uso terapéutico
8.
J Lipid Res ; 54(12): 3481-90, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24058201

RESUMEN

Angiopoietin-like 3 (ANGPTL3) regulates lipoprotein metabolism by modulating extracellular lipases. Loss-of function mutations in ANGPTL3 gene cause familial combined hypolipidemia (FHBL2). The mode of inheritance and hepatic and vascular consequences of FHBL2 have not been fully elucidated. To get further insights on these aspects, we reevaluated the clinical and the biochemical characteristics of all reported cases of FHBL2. One hundred fifteen FHBL2 individuals carrying 13 different mutations in the ANGPTL3 gene (14 homozygotes, 8 compound heterozygotes, and 93 heterozygotes) and 402 controls were considered. Carriers of two mutant alleles had undetectable plasma levels of ANGPTL3 protein, whereas heterozygotes showed a reduction ranging from 34% to 88%, according to genotype. Compared with controls, homozygotes as well as heterozygotes showed a significant reduction of all plasma lipoproteins, while no difference in lipoprotein(a) [Lp(a)] levels was detected between groups. The prevalence of fatty liver was not different in FHBL2 subjects compared with controls. Notably, diabetes mellitus and cardiovascular disease were absent among homozygotes. FHBL2 trait is inherited in a codominant manner, and the lipid-lowering effect of two ANGPTL3 mutant alleles was more than four times larger than that of one mutant allele. No changes in Lp(a) were detected in FHBL2. Furthermore, our analysis confirmed that FHBL2 is not associated with adverse clinical sequelae. The possibility that FHBL2 confers lower risk of diabetes and cardiovascular disease warrants more detailed investigation.


Asunto(s)
Hipobetalipoproteinemias/sangre , Hipobetalipoproteinemias/genética , Lípidos/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/sangre , Angiopoyetinas/genética , Enfermedades Cardiovasculares/genética , Niño , Estudios de Cohortes , Hígado Graso/genética , Regulación de la Expresión Génica , Heterocigoto , Homocigoto , Humanos , Lipoproteína(a)/sangre , Persona de Mediana Edad , Mutación , Adulto Joven
9.
Arterioscler Thromb Vasc Biol ; 32(3): 805-9, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22247256

RESUMEN

OBJECTIVE: Mutations of the ANGPTL3 gene have been associated with a novel form of primary hypobetalipoproteinemia, the combined hypolipidemia (cHLP), characterized by low total cholesterol and low HDL-cholesterol levels. The aim of this work is to define the role of ANGPTL3 gene as determinant of the combined hypolipidemia phenotype in 2 large cohorts of 913 among American and Italian subjects with primary hypobetalipoproteinemia (total cholesterol<5th percentile). METHODS AND RESULTS: The combined hypolipidemia cut-offs were chosen according to total cholesterol and HDL-cholesterol levels reported in the ANGPTL3 kindred described to date: total cholesterol levels, <2nd percentile and HDL-cholesterol, levels<2nd decile. Seventy-eight subjects with combined hypolipidemia were analyzed for ANGPTL3 and APOB genes. We identified nonsense and/or missense mutations in ANGPTL3 gene in 8 subjects; no mutations of the APOB gene were found. Mutated ANGPTL3 homozygous/compound heterozygous subjects showed a more severe biochemical phenotype compared to heterozygous or ANGPTL3 negative subjects, although ANGPTL3 heterozygotes did not differ from ANGPTL3 negative subjects. CONCLUSION: These results demonstrated that in a cohort of subjects with severe primary hypobetalipoproteinemia the prevalence of ANGPTL3 gene mutations responsible for a combined hypolipidemia phenotype is about 10%, whereas mutations of APOB gene are absent.


Asunto(s)
Angiopoyetinas/genética , Apolipoproteínas B/genética , Codón sin Sentido , Hipobetalipoproteinemias/genética , Mutación Missense , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Biomarcadores/sangre , Colesterol/sangre , HDL-Colesterol/sangre , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Heterocigoto , Homocigoto , Humanos , Hipobetalipoproteinemias/sangre , Hipobetalipoproteinemias/epidemiología , Italia/epidemiología , Masculino , Persona de Mediana Edad , Missouri/epidemiología , Datos de Secuencia Molecular , Fenotipo , Prevalencia , Índice de Severidad de la Enfermedad , Adulto Joven
10.
J Am Heart Assoc ; 12(10): e029223, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37183858

RESUMEN

Background Evidence suggests that LPA risk genotypes are a possible contributor to the clinical diagnosis of familial hypercholesterolemia (FH). This study aimed at determining the prevalence of LPA risk variants in adult individuals with FH enrolled in the Italian LIPIGEN (Lipid Transport Disorders Italian Genetic Network) study, with (FH/M+) or without (FH/M-) a causative genetic variant. Methods and Results An lp(a) [lipoprotein(a)] genetic score was calculated by summing the number risk-increasing alleles inherited at rs3798220 and rs10455872 variants. Overall, in the 4.6% of 1695 patients with clinically diagnosed FH, the phenotype was not explained by a monogenic or polygenic cause but by genotype associated with high lp(a) levels. Among 765 subjects with FH/M- and 930 subjects with FH/M+, 133 (17.4%) and 95 (10.2%) were characterized by 1 copy of either rs10455872 or rs3798220 or 2 copies of either rs10455872 or rs3798220 (lp(a) score ≥1). Subjects with FH/M- also had lower mean levels of pretreatment low-density lipoprotein cholesterol than individuals with FH/M+ (t test for difference in means between FH/M- and FH/M+ groups <0.0001); however, subjects with FH/M- and lp(a) score ≥1 had higher mean (SD) pretreatment low-density lipoprotein cholesterol levels (223.47 [50.40] mg/dL) compared with subjects with FH/M- and lp(a) score=0 (219.38 [54.54] mg/dL for), although not statistically significant. The adjustment of low-density lipoprotein cholesterol levels based on lp(a) concentration reduced from 68% to 42% the proportion of subjects with low-density lipoprotein cholesterol level ≥190 mg/dL (or from 68% to 50%, considering a more conservative formula). Conclusions Our study supports the importance of measuring lp(a) to perform the diagnosis of FH appropriately and to exclude that the observed phenotype is driven by elevated levels of lp(a) before performing the genetic test for FH.


Asunto(s)
Hiperlipoproteinemia Tipo II , Lipoproteína(a) , Humanos , Lipoproteína(a)/genética , Redes Reguladoras de Genes , Factores de Riesgo , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/complicaciones , Genotipo , LDL-Colesterol
11.
Nutrients ; 15(15)2023 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-37571405

RESUMEN

Detection and treatment of patients with familial hypercholesterolemia (FH) starting from childhood is fundamental to reduce morbidity and mortality. The activity of National realities such as the LIPIGEN (LIpid transPort disorders Italian GEnetic Network) Paediatric Group, founded in 2018, is a milestone in this context. The aim of this exploratory survey, conducted in October 2021 among Italian lipid clinics included in the LIPIGEN Paediatric Group, was to investigate the current clinical approach in the management and treatment of paediatric patients with suspected FH. A digital questionnaire composed of 20 questions investigating nutritional treatment and nutraceutical and pharmacological therapy for children and adolescents with FH was proposed to the principal investigators of 30 LIPIGEN centres. Twenty-four centres responded to the section referring to children aged < 10 years and 30 to that referring to adolescents. Overall, 66.7% of children and 73.3% of adolescents were given lipid-lowering nutritional treatment as the first intervention level for at least 3-4 months (29.2% and 23.3%) or 6-12 months (58.3% and 53.3%). Nutraceuticals were considered in 41.7% (regarding children) and 50.0% (regarding adolescents) of the centres as a supplementary approach to diet. Lipid-lowering drug therapy initiation was mainly recommended (91.7% and 80.0%). In 83.3% of children and 96.7% of adolescents, statins were the most frequently prescribed drug. We highlighted several differences in the treatment of paediatric patients with suspected FH among Italian centres; however, the overall approach is in line with the European Atherosclerosis Society (EAS) recommendations for FH children and adolescents. We consider this survey as a starting point to reinforce collaboration between LIPIGEN centres and to elaborate in the near future a consensus document on the management of paediatric patients with suspected FH so as to improve and uniform detection, management, and treatment of these patients in our country.


Asunto(s)
Anticolesterolemiantes , Dieta , Suplementos Dietéticos , Hiperlipoproteinemia Tipo II , Humanos , Masculino , Femenino , Niño , Adolescente , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/terapia , Anticolesterolemiantes/uso terapéutico
12.
Curr Opin Lipidol ; 22(2): 93-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21157333

RESUMEN

PURPOSE OF REVIEW: To review recent publications concerning the functional assessment on pre-mRNA splicing of genomic variants found in some monogenic dyslipidemias. Examples are derived from familial hypercholesterolemia, familial HDL deficiency/Tangier disease and familial hypobetalipoproteinemia. RECENT FINDINGS: About 5-10% of genomic variants found in familial hypercholesterolemia, FHD/Tangier disease and familial hypobetalipoproteinemia are located in the introns of the candidate genes and are classified as splicing mutations. Although variants affecting highly conserved GT/AG dinucleotides at the splice sites are likely to be pathogenic, it is difficult to predict the effects of variants located deep in the introns. Algorithms were developed to predict the effect of these variants and to provide the rationale for functional studies. Combined in-silico and wet bench analysis revealed that some intronic variants classified as pathogenic have no effect, whereas others generated abnormal transcripts. Nucleotide substitutions at the 5' and the 3' of exons might change the splice site consensus sequence, causing splicing defects. Rare silent mutations were identified which create new splice sites within exons, with the consequent production of abnormal transcripts. SUMMARY: Intronic variants, even if located deep in introns, as well as exonic variants could affect splicing with the formation of abnormal transcripts encoding structurally abnormal proteins.


Asunto(s)
Errores Innatos del Metabolismo Lipídico/fisiopatología , Empalme del ARN/genética , Variación Genética , Humanos , Errores Innatos del Metabolismo Lipídico/genética
13.
J Clin Lipidol ; 16(5): 591-595, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35945124

RESUMEN

Familial chylomicronemia syndrome (FCS) is a rare and severe genetic disorder, characterized by marked elevation of plasma triglycerides, often diagnosed in infancy. We describe the long-term follow-up (almost 60 years), the diagnostic assessment and the management of two siblings with severe hypertriglyceridemia and a history of pancreatitis who also developed cardiovascular complications later in life. We recently disclosed that the surviving index case was homozygous for a pathogenic LPL gene variant (c.984 G>T, p.M328I). The same variant was also found in two apparently unrelated siblings with FCS living in the same geographical area as the index case.


Asunto(s)
Hiperlipoproteinemia Tipo I , Hipertrigliceridemia , Humanos , Hiperlipoproteinemia Tipo I/diagnóstico , Hiperlipoproteinemia Tipo I/genética , Hermanos , Estudios de Seguimiento , Hipertrigliceridemia/genética , Lipoproteína Lipasa/genética
14.
Biofactors ; 48(3): 707-717, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35579277

RESUMEN

High-density lipoproteins (HDL) are well known for their atheroprotective function, mainly due to their ability to remove cell cholesterol and to exert antioxidant and anti-inflammatory activities. Through the same mechanisms HDL could also affect the development and progression of tumors. Cancer cells need cholesterol to proliferate, especially in hormone-dependent tumors, as prostate cancer (PCa). Aim of the study was to investigate the ability of HDL to modulate cholesterol content and metabolism in androgen receptor (AR)-positive and AR-null PCa cell lines and the consequences on cell proliferation. HDL inhibited colony formation of LNCaP and PC3 cells. HDL reduced cell cholesterol content and proliferation of LNCaP cells loaded with low-density lipoproteins but were not effective on PC3 cells. Here, the expression of the ATP-binding cassette transporter A1 (ABCA1) was markedly reduced due to proteasome degradation. Bortezomib, a proteasome inhibitor, restored ABCA1 expression and HDL ability to promote cholesterol removal from PC3; consequently, HDL inhibited the proliferation of PC3 cells induced by LDL only after bortezomib pre-treatment. In conclusion, the antiproliferative activity of HDL on AR-positive and AR-null PCa cells also rely on cholesterol removal, a process in which the ABCA1 transporter plays a key role.


Asunto(s)
Colesterol , Lipoproteínas HDL , Neoplasias de la Próstata , Complejo de la Endopetidasa Proteasomal , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/farmacología , Bortezomib/farmacología , Proliferación Celular , Colesterol/metabolismo , Humanos , Lipoproteínas HDL/metabolismo , Masculino , Células PC-3 , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma/farmacología
15.
Atherosclerosis ; 349: 233-239, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35562202

RESUMEN

BACKGROUND AND AIMS: Little is known about the role of Lp(a) in the assessment of cardiovascular risk in the paediatric population. Trying to clarify the clinical relevance of Lp(a) in risk stratification, the aim of the study is to evaluate the association between Lp(a) plasma levels in children with familial hypercholesterolaemia (FH) and positive family history for premature cardiovascular disease (pCVD) in first- and second-degree relatives. METHODS: 653 Caucasian children and adolescents (334 females and 319 males), aged 2-17 years, with diagnosis of FH from a paediatric cohort included in the LIPIGEN Network, were selected. We compared family history of pCVD, lipid and genetic profile in two groups based on Lp(a) levels below or above 30 mg/dL. To determine the independent predictors of pCVD, a multivariate logistic regression was used, with all clinical characteristics and blood measurements as predictors. RESULTS: Subjects with Lp(a) > 30 mg/dl more frequently reported positive family history of pCVD compared to subjects with Lp(a)≤30 mg/dl (69.90% vs 36.66%, p < 0.0001), while did not show differences in terms of median [interquartile range] LDL-cholesterol level (153.00 [88.00 vs 164.50 [90.25] mg/dL, p = 0.3105). In the regression analysis, Lp(a) > 30 mg/dl was an independent predictor of family history of pCVD. Comparing subjects with or without family history of pCVD, we reported significant differences for Lp(a) > 30 mg/dl (46.25% vs 17.65%, p < 0.0001), FH genetic mutation (50.48% vs 40.75%, p = 0,0157), as well as for LDL-cholesterol (p = 0.0013) and total cholesterol (p = 0.0101). CONCLUSIONS: Children/adolescents with FH and Lp(a) > 30 mg/dl where more likely to have a positive family history of pCVD. Lp(a) screening in children and adolescents with FH may enhance risk assessment and help identify those subjects, children and relatives, at increased pCVD risk.


Asunto(s)
Enfermedades Cardiovasculares , Hiperlipoproteinemia Tipo II , Adolescente , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Niño , LDL-Colesterol , Femenino , Factores de Riesgo de Enfermedad Cardiaca , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/genética , Lipoproteína(a)/genética , Masculino , Factores de Riesgo
16.
J Clin Lipidol ; 16(4): 530-537, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35589500

RESUMEN

BACKGROUND: Primary Hypobetalipoproteinemias (HBL) are a group of dominant and recessive monogenic genetic disorders caused by mutations in APOB, PCSK9, ANGPTL3, MTTP, Sar1b genes and characterized by plasma levels of total cholesterol (TC), low density lipoprotein-cholesterol (LDL-C) and apolipoprotein B (apoB) below the 5th percentile of the distribution in a given population. Mutations in the candidate genes account only for a small proportion of subjects with HBL suggesting a role for a polygenic contribution to the low cholesterol phenotype. OBJECTIVE: To explore the complex genetic architecture of HBL we compared two polygenic risk scores in order to assess the role of the polygenic burden and the differences in the clinical phenotype between monogenic and polygenic HBL; we studied a cohort of 170 subjects with primary HBL referred over a 25-year period to 2 Italian reference centers have been studied. METHODS: The genetic analyses have been based on: Sanger sequencing, in-house NGS customized panel and two scores, PRS1 and PRS2 for the polygenic burden. RESULTS: Sixty 60 (35%) and 63 (37%) subjects had a monogenic and polygenic HBL respectively. LDL-C plasma levels were significantly lower in monogenic HBL (30.87 ± 3.12 mg/dl) compared with the non-monogenic HBL (42.80 ± 2.18 mg/dl) (p<0.002) with no differences in the percentage of fatty liver. CONCLUSION: Only PRS1 is effective in detecting polygenic HBL while PRS2 does not improve the polygenic diagnosis.


Asunto(s)
Hipobetalipoproteinemias , Trastornos del Metabolismo de los Lípidos , Proteína 3 Similar a la Angiopoyetina/genética , Apolipoproteínas B/genética , LDL-Colesterol/sangre , Humanos , Hipobetalipoproteinemias/genética , Trastornos del Metabolismo de los Lípidos/genética , Proteínas de Unión al GTP Monoméricas/genética , Herencia Multifactorial , Mutación , Proproteína Convertasa 9/genética , Factores de Riesgo
17.
J Clin Lipidol ; 16(6): 797-812, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36243606

RESUMEN

The Abetalipoproteinemia and Related Disorders Foundation was established in 2019 to provide guidance and support for the life-long management of inherited hypocholesterolemia disorders. Our mission is "to improve the lives of individuals and families affected by abetalipoproteinemia and related disorders". This review explains the molecular mechanisms behind the monogenic hypobetalipoproteinemia disorders and details their specific pathophysiology, clinical presentation and management throughout the lifespan. In this review, we focus on abetalipoproteinemia, homozygous hypobetalipoproteinemia and chylomicron retention disease; rare genetic conditions that manifest early in life and cause severe complications without appropriate treatment. Absent to low plasma lipid levels, in particular cholesterol and triglyceride, along with malabsorption of fat and fat-soluble vitamins are characteristic features of these diseases. We summarize the genetic basis of these disorders, provide guidance in their diagnosis and suggest treatment regimens including high dose fat-soluble vitamins as therapeutics. A section on preconception counseling and other special considerations pertaining to pregnancy is included. This information may be useful for patients, caregivers, physicians and insurance agencies involved in the management and support of affected individuals.


Asunto(s)
Abetalipoproteinemia , Hipobetalipoproteinemias , Trastornos del Metabolismo de los Lípidos , Humanos , Abetalipoproteinemia/diagnóstico , Abetalipoproteinemia/genética , Abetalipoproteinemia/terapia , Hipobetalipoproteinemias/diagnóstico , Hipobetalipoproteinemias/genética , Hipobetalipoproteinemias/terapia , Homocigoto , Vitaminas
18.
J Lipid Res ; 52(11): 1885-926, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21862702

RESUMEN

This review integrates historical biochemical and modern genetic findings that underpin our understanding of the low-density lipoprotein (LDL) dyslipidemias that bear on human disease. These range from life-threatening conditions of infancy through severe coronary heart disease of young adulthood, to indolent disorders of middle- and old-age. We particularly focus on the biological aspects of those gene mutations and variants that impact on sterol absorption and hepatobiliary excretion via specific membrane transporter systems (NPC1L1, ABCG5/8); the incorporation of dietary sterols (MTP) and of de novo synthesized lipids (HMGCR, TRIB1) into apoB-containing lipoproteins (APOB) and their release into the circulation (ANGPTL3, SARA2, SORT1); and receptor-mediated uptake of LDL and of intestinal and hepatic-derived lipoprotein remnants (LDLR, APOB, APOE, LDLRAP1, PCSK9, IDOL). The insights gained from integrating the wealth of genetic data with biological processes have important implications for the classification of clinical and presymptomatic diagnoses of traditional LDL dyslipidemias, sitosterolemia, and newly emerging phenotypes, as well as their management through both nutritional and pharmaceutical means.


Asunto(s)
Enfermedad/genética , Predisposición Genética a la Enfermedad , Lipoproteínas LDL/sangre , Esteroles/metabolismo , Absorción , Secuencia de Aminoácidos , Animales , Apolipoproteínas B/genética , Apolipoproteínas B/metabolismo , Humanos , Datos de Secuencia Molecular
19.
J Biol Chem ; 285(9): 6453-64, 2010 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-20032471

RESUMEN

Five nontruncating missense APOB mutations, namely A31P, G275S, L324M, G912D, and G945S, were identified in heterozygous carriers of familial hypobetalipoproteinemia (FHBL) in the Italian population. To test that the FHBL phenotype was a result of impaired hepatic secretion of mutant apoB proteins, we performed transfection studies using McA-RH7777 cells stably expressing wild type or mutant forms of human apolipoprotein B-48 (apoB-48). All mutant proteins displayed varied impairment in secretion, with G912D the least affected and A31P barely secreted. Although some A31P was degraded by proteasomes, a significant proportion of it (although inappropriately glycosylated) escaped endoplasmic reticulum (ER) quality control and presented in the Golgi compartment. Degradation of the post-ER A31P was achieved by autophagy. Expression of A31P also decreased secretion of endogenous apoB and triglycerides, yet the impaired lipoprotein secretion did not lead to lipid accumulation in the cells or ER stress. Rather, expression of genes involved in lipogenesis was down-regulated, including liver X receptor alpha, sterol regulator element-binding protein 1c, fatty acid synthase, acetyl-CoA carboxylase 1, stearoyl-CoA desaturase 1, and lipin-1. These results suggest that feedback inhibition of hepatic lipogenesis in conjunction with post-ER degradation of misfolded apoB proteins can contribute to reduce fat accumulation in the FHBL liver.


Asunto(s)
Apolipoproteínas B/genética , Hipobetalipoproteinemia Familiar por Apolipoproteína B/metabolismo , Lipogénesis/genética , Mutación Missense , Apolipoproteínas B/metabolismo , Autofagia , Regulación hacia Abajo , Retículo Endoplásmico , Retroalimentación Fisiológica , Aparato de Golgi , Heterocigoto , Humanos , Hipobetalipoproteinemia Familiar por Apolipoproteína B/genética , Italia , Hígado/metabolismo , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Pliegue de Proteína
20.
Arterioscler Thromb Vasc Biol ; 29(12): 2191-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19762784

RESUMEN

OBJECTIVE: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a central player in the regulation of cholesterol homeostasis, increasing the low-density lipoprotein (LDL) receptor degradation. Our study aimed at exploring the pathogenic consequences in vivo and in vitro of a PCSK9 prodomain mutation found in a family with hypobetalipoproteinemia (FHBL). METHODS AND RESULTS: A white 49-year-old diabetic man had profound FBHL (LDLC: 16 mg/dL) whereas his daughter and sister displayed a milder phenotype (LDLC 44 mg/dL and 57 mg/dL, respectively), all otherwise healthy with a normal liver function. A monoallelic PCSK9 double-mutant R104C/V114A cosegregated with FBHL, with no mutation found at other FHBL-causing loci. A dose-effect was also found in FBHL relatives for plasma APOB and PCSK9 (very-low to undetectable in proband, approximately 50% decreased in sister and daughter) and LDL catabolic rate (256% and 88% increased in proband and daughter). Transient transfection in hepatocytes showed severely impaired processing and secretion of the double mutant which acted as a dominant negative over secretion of wild-type PCSK9. CONCLUSIONS: These results show that heterozygous PCSK9 missense mutations may associate with profound hypobetalipoproteinemia and constitute the first direct evidence in human that decrease of plasma LDLC concentrations associated to PCSK9 LOF mutations are attributable to an increased clearance rate of LDL.


Asunto(s)
LDL-Colesterol/sangre , Hipobetalipoproteinemias/enzimología , Hipobetalipoproteinemias/genética , Mutación Missense , Serina Endopeptidasas/sangre , Serina Endopeptidasas/genética , Adulto , Sustitución de Aminoácidos , Apolipoproteínas B/sangre , Femenino , Genes Dominantes , Hepatocitos/enzimología , Heterocigoto , Humanos , Hipobetalipoproteinemia Familiar por Apolipoproteína B/sangre , Hipobetalipoproteinemia Familiar por Apolipoproteína B/enzimología , Hipobetalipoproteinemia Familiar por Apolipoproteína B/genética , Hipobetalipoproteinemias/sangre , Cinética , Masculino , Persona de Mediana Edad , Linaje , Proproteína Convertasa 9 , Proproteína Convertasas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidasas/deficiencia , Transfección
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda