Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 564
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(10): 1741-1752, 2023 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-36715614

RESUMEN

Tandem cytosine-adenine-guanine (CAG) repeat sizes of 36 or more in the huntingtin gene (HTT) cause Huntington's disease (HD). Apart from neuropsychiatric complications, the disease is also accompanied by metabolic dysregulation and weight loss, which contribute to a progressive functional decline. Recent studies also reported an association between repeats below the pathogenic threshold (<36) for HD and body mass index (BMI), suggesting that HTT repeat sizes in the non-pathogenic range are associated with metabolic dysregulation. In this study, we hypothesized that HTT repeat sizes < 36 are associated with metabolite levels, possibly mediated through reduced BMI. We pooled data from three European cohorts (n = 10 228) with genotyped HTT CAG repeat size and metabolomic measurements. All 145 metabolites were measured on the same targeted platform in all studies. Multilevel mixed-effects analysis using the CAG repeat size in HTT identified 67 repeat size metabolite associations. Overall, the metabolomic profile associated with larger CAG repeat sizes in HTT were unfavorable-similar to those of higher risk of coronary artery disease and type 2 diabetes-and included elevated levels of amino acids, fatty acids, low-density lipoprotein (LDL)-, very low-density lipoprotein- and intermediate density lipoprotein (IDL)-related metabolites while with decreased levels of very large high-density lipoprotein (HDL)-related metabolites. Furthermore, the associations of 50 metabolites, in particular, specific very large HDL-related metabolites, were mediated by lower BMI. However, no mediation effect was found for 17 metabolites related to LDL and IDL. In conclusion, our findings indicate that large non-pathogenic CAG repeat sizes in HTT are associated with an unfavorable metabolomic profile despite their association with a lower BMI.


Asunto(s)
Diabetes Mellitus Tipo 2 , Enfermedad de Huntington , Humanos , Índice de Masa Corporal , Diabetes Mellitus Tipo 2/genética , Valores de Referencia , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Lipoproteínas , Lipoproteínas LDL/genética , Expansión de Repetición de Trinucleótido/genética
2.
Lipids Health Dis ; 23(1): 85, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38515137

RESUMEN

BACKGROUND: Familial hypercholesterolemia (FH) is a prevalent hereditary disease that can cause aberrant cholesterol metabolism. In this study, we confirmed that c.415G > A in low-density lipoprotein receptor (LDLR), an FH-related gene, is a pathogenic variant in FH by in silico analysis and functional experiments. METHODS: The proband and his family were evaluated using the diagnostic criteria of the Dutch Lipid Clinic Network. Whole-exome and Sanger sequencing were used to explore and validate FH-related variants. In silico analyses were used to evaluate the pathogenicity of the candidate variant and its impact on protein stability. Molecular and biochemical methods were performed to examine the effects of the LDLR c.415G > A variant in vitro. RESULTS: Four of six participants had a diagnosis of FH. It was estimated that the LDLR c.415G > A variant in this family was likely pathogenic. Western blotting and qPCR suggested that LDLR c.415G > A does not affect protein expression. Functional studies showed that this variant may lead to dyslipidemia by impairing the binding and absorption of LDLR to low-density lipoprotein ( LDL). CONCLUSION: LDLR c.415G > A is a pathogenic variant in FH; it causes a significant reduction in LDLR's capacity to bind LDL, resulting in impaired LDL uptake. These findings expand the spectrum of variants associated with FH.


Asunto(s)
Hiperlipoproteinemia Tipo II , Humanos , Fenotipo , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/diagnóstico , Receptores de LDL/genética , Receptores de LDL/metabolismo , Lipoproteínas LDL/genética , Mutación , Proproteína Convertasa 9/genética
3.
PLoS Genet ; 17(1): e1009285, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33513160

RESUMEN

Hypercholesterolemia is a causal and modifiable risk factor for atherosclerotic cardiovascular disease. A critical pathway regulating cholesterol homeostasis involves the receptor-mediated endocytosis of low-density lipoproteins into hepatocytes, mediated by the LDL receptor. We applied genome-scale CRISPR screening to query the genetic determinants of cellular LDL uptake in HuH7 cells cultured under either lipoprotein-rich or lipoprotein-starved conditions. Candidate LDL uptake regulators were validated through the synthesis and secondary screening of a customized library of gRNA at greater depth of coverage. This secondary screen yielded significantly improved performance relative to the primary genome-wide screen, with better discrimination of internal positive controls, no identification of negative controls, and improved concordance between screen hits at both the gene and gRNA level. We then applied our customized gRNA library to orthogonal screens that tested for the specificity of each candidate regulator for LDL versus transferrin endocytosis, the presence or absence of genetic epistasis with LDLR deletion, the impact of each perturbation on LDLR expression and trafficking, and the generalizability of LDL uptake modifiers across multiple cell types. These findings identified several previously unrecognized genes with putative roles in LDL uptake and suggest mechanisms for their functional interaction with LDLR.


Asunto(s)
Aterosclerosis/genética , Colesterol/genética , Lipoproteínas LDL/genética , Receptores de LDL/genética , Aterosclerosis/patología , Sistemas CRISPR-Cas/genética , Colesterol/metabolismo , Endocitosis/genética , Regulación de la Expresión Génica/genética , Genoma Humano/genética , Células Hep G2 , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Lipoproteínas LDL/metabolismo , ARN Guía de Kinetoplastida/genética
4.
Nano Lett ; 22(24): 10025-10033, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-36521071

RESUMEN

Lipid nanoparticles (LNPs) have delivered therapeutic RNA to hepatocytes in humans. Adsorption of apolipoprotein E (ApoE) onto these clinical LNP-mRNA drugs has been shown to facilitate hepatocyte entry via the low-density lipoprotein receptor (LDLR). Since ApoE-LDLR trafficking is conserved in mice, non-human primates, and humans, characterizing this mechanism eased clinical transition. Recently, LNPs have delivered mRNA to non-hepatocytes in mice and non-human primates, suggesting they can target new cell types via ApoE- and LDLR-independent pathways. To test this hypothesis, we quantified how 60 LNPs delivered mRNA with cell type resolution in wild-type mice and three knockout mouse strains related to lipid trafficking: ApoE-/-, LDLR-/-, and PCSK9-/-. These data suggest that the hydrophobic tail length of diketopiperazine-based lipids can be changed to drive ApoE- and LDLR-independent delivery in vivo. More broadly, the results support the hypothesis that endogenous LNP trafficking can be tuned by modifying lipid chemistry.


Asunto(s)
Apolipoproteínas E , Lipoproteínas LDL , Nanopartículas , Animales , Ratones , Apolipoproteínas E/genética , Lipoproteínas LDL/genética , Ratones Noqueados , Nanopartículas/química , ARN Mensajero/química
5.
Int J Mol Sci ; 24(13)2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37446330

RESUMEN

We previously described the role of low-density lipoprotein (LDL) in aggressiveness in papillary thyroid cancer (PTC). Moreover, the MAPK signaling pathway in the presence of BRAF V600E mutation is associated with more aggressive PTC. Although the link between MAPK cascade and LDL receptor (LDLR) expression has been previously described, it is unknown whether LDL can potentiate the adverse effects of PTC through it. We aimed to investigate whether the presence of LDL might accelerate the oncogenic processes through MAPK pathway in presence or absence of BRAF V600E in two thyroid cell lines: TPC1 and BCPAP (wild-type and BRAF V600E, respectively). LDLR, PI3K-AKT and RAS/RAF/MAPK (MEK)/ERK were analyzed via Western blot; cell proliferation was measured via MTT assay, cell migration was studied through wound-healing assay and LDL uptake was analyzed by fluorometric and confocal analysis. TPC1 demonstrated a time-specific downregulation of the LDLR, while BCPAP resulted in a receptor deregulation after LDL exposition. LDL uptake was increased in BCPAP over-time, as well as cell proliferation (20% higher) in comparison to TPC1. Both cell lines differed in migration pattern with a wound closure of 83.5 ± 9.7% after LDL coculture in TPC1, while a loss in the adhesion capacity was detected in BCPAP. The siRNA knockdown of LDLR in LDL-treated BCPAP cells resulted in a p-ERK expression downregulation and cell proliferation modulation, demonstrating a link between LDLR and MAPK pathway. The modulation of BRAF-V600E using vemurafenib-impaired LDLR expression decreased cellular proliferation. Our results suggest that LDLR regulation is cell line-specific, regulating the RAS/RAF/MAPK (MEK)/ERK pathway in the LDL-signaling cascade and where BRAF V600E can play a critical role. In conclusion, targeting LDLR and this downstream signaling cascade, could be a new therapeutic strategy for PTC with more aggressive behavior, especially in those harboring BRAF V600E.


Asunto(s)
Proteínas Proto-Oncogénicas B-raf , Neoplasias de la Tiroides , Humanos , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Mutación , Neoplasias de la Tiroides/patología , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/patología , Receptores de LDL/genética , Lipoproteínas LDL/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Línea Celular Tumoral
6.
Hum Mol Genet ; 29(8): 1229-1238, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-31600776

RESUMEN

The cell-surface low-density lipoprotein receptor (LDLR) internalizes low-density lipoprotein (LDL) by receptor-mediated endocytosis and plays a key role in the regulation of plasma cholesterol levels. The ligand-binding domain of the LDLR contains seven ligand-binding repeats of approximately 40 residues each. Between ligand-binding repeats 4 and 5, there is a 10-residue linker region that is subject to enzymatic cleavage. The cleaved LDLR is unable to bind LDL. In this study, we have screened a series of enzyme inhibitors in order to identify the enzyme that cleaves the linker region. These studies have identified bone morphogenetic protein 1 (BMP1) as being the cleavage enzyme. This conclusion is based upon the use of the specific BMP1 inhibitor UK 383367, silencing of the BMP1 gene by the use of siRNA or CRISPR/Cas9 technology and overexpression of wild-type BMP1 or the loss-of-function mutant E214A-BMP1. We have also shown that the propeptide of BMP1 has to be cleaved at RSRR120↓ by furin-like proprotein convertases for BMP1 to have an activity towards the LDLR. Targeting BMP1 could represent a novel strategy to increase the number of functioning LDLRs in order to lower plasma LDL cholesterol levels. However, a concern by using BMP1 inhibitors as cholesterol-lowering drugs could be the risk of side effects based on the important role of BMP1 in collagen assembly.


Asunto(s)
Proteína Morfogenética Ósea 1/genética , LDL-Colesterol/genética , Colesterol/genética , Furina/genética , Receptores de LDL/genética , Animales , Proteína Morfogenética Ósea 1/antagonistas & inhibidores , Células CHO , Sistemas CRISPR-Cas/genética , LDL-Colesterol/antagonistas & inhibidores , LDL-Colesterol/sangre , Cricetulus , Endocitosis/genética , Inhibidores Enzimáticos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hidrazinas/farmacología , Ligandos , Lipoproteínas LDL/genética , Proproteína Convertasas/genética , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/genética , Receptores de LDL/antagonistas & inhibidores , Tiourea/análogos & derivados , Tiourea/farmacología
7.
Genet Med ; 24(10): 2103-2111, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35913489

RESUMEN

PURPOSE: In Europe, >2 million individuals with familial hypercholesterolemia (FH) are currently undiagnosed. Effective screening strategies for FH diagnosis in childhood are urgently needed. We assessed the overall performances of 2 different FH screening programs in children: universal screening program with opt-out and opt-in type participation. METHODS: We analyzed the data from 2 independent populations based on >166,000 individuals screened for hypercholesterolemia. Genetic analyses of FH-related genes were finalized in 945 children and 99 parents. RESULTS: A total of 305 (32.3%) children were genotyped as positive or with a variant of uncertain significance in FH-related genes. For low-density lipoprotein cholesterol levels of 3.5 mmol L (135.3 mg/dL), the overall sensitivity and specificity for confirming FH were 90.5% and 55.3%, respectively. As part of child-parent screening, in >90% of the families, the parent with reported higher cholesterol levels was positive for the familial genetic variant. The cohort-based prevalence of FH from the opt-out universal screening program was estimated to be 1 in 431 individuals (95% CI = 1/391-1/472). CONCLUSION: Universal 3-step FH screening approach in children enabled detection of most children and their parents in every generation screened at reasonable costs. Opt-out screening strategy might be preferable over opt-in screening strategy.


Asunto(s)
Hiperlipoproteinemia Tipo II , Colesterol , Pruebas Genéticas , Humanos , Hiperlipoproteinemia Tipo II/diagnóstico , Hiperlipoproteinemia Tipo II/epidemiología , Hiperlipoproteinemia Tipo II/genética , Lipoproteínas LDL/genética , Tamizaje Masivo
8.
FASEB J ; 35(2): e21309, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33423309

RESUMEN

Cryptochromes are photoreceptors that mediate the circadian entrainment by light in plants and animals. They are also involved in magnetic field sensing in some animals. Recent studies suggest that cryptochromes play an essential role in metabolism and cardiovascular disease. However, the tissue-specific function of cryptochromes in atherosclerosis is unknown. We transplanted bone marrow from wild-type (WT) and cryptochrome 1/2 knockout (Cry1/2 KO) mice into irradiated recipient low-density lipoprotein receptor knockout (LDLR-/- ) mice and induced atherosclerosis with a high cholesterol diet for 12 weeks. There was a reduction in atherosclerotic plaques and macrophage accumulation in the aorta of LDLR-/- mice that received Cry1/2 KO bone marrow compared to mice that received WT bone marrow. Bone marrow-derived macrophages (BMDMs) from Cry1/2 KO mice exhibited impaired uptake of low-density lipoprotein, and subsequently, impaired foam cell formation. Analysis of macrophage mRNA circadian oscillations revealed that the circadian rhythm of the LDLR mRNAs was lost in Cry1/2 KO BMDMs. Reinstalling the circadian oscillatory LDLR mRNAs using adenovirus into the BMDMs was able to rescue the lipid uptake and foam cell formation function. However, the noncircadian oscillatory LDLR mRNAs exhibited reduced ability to rescue the macrophage functions. These findings indicate that cryptochromes in bone marrow-derived cells are critical mediators of atherosclerosis through regulation of the LDLR mRNA circadian rhythm. Therapeutic measures targeting cryptochromes in the macrophage may have important implications for atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Células de la Médula Ósea/metabolismo , Criptocromos/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo , Animales , Aterosclerosis/genética , Células Cultivadas , Colesterol/sangre , Criptocromos/genética , Femenino , Inmunohistoquímica , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Noqueados , Placa Aterosclerótica/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de LDL/genética
9.
Immunol Invest ; 51(5): 1162-1169, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33941028

RESUMEN

In this study, we determined to interpret the effects of the interleukin (IL)1B gene rs1143634 C/T polymorphism on myocardial infarction (MI) risk. This study, conducted in a Chinese Han population, recruited 369 MI patients and 465 controls. The variant of IL1B gene (rs1143634 C/T polymorphism) was genotyped by PCR-RFLP method. In this study, a significant link was shown between the IL1B rs1143634 C/T polymorphism and MI risk. We found that the IL1B rs1143634 C/T polymorphism enhanced the risk of MI in this population. Subgroup analysis detected that the IL1B rs1143634 C/T polymorphism associated with MI susceptibility in males, smokers, and individuals with diabetes mellitus. In addition, the IL1B rs1143634 C/T polymorphism was related with the levels of blood lipids including low-density lipoprotein (LDL), and total cholesterol (TC). This study uncovers that the IL1B rs1143634 C/T polymorphism may associate with the risk and blood lipid levels of MI in an Eastern Chinese Han population.Abbreviations: MI: myocardial infarction; IL-1: Interleukin-1; SNP: single nucleotide polymorphism; BMI: Body Mass Index; HDL: high-density lipoprotein; TC: total cholesterol; TG: triglyceride; LDL: low-density lipoprotein; PCR: polymerase chain reaction; 95% CI: 95% confidence interval; OR: odds ratio.


Asunto(s)
Interleucina-1beta , Infarto del Miocardio , China/epidemiología , Colesterol/sangre , Colesterol/genética , Femenino , Variación Genética/genética , Humanos , Interleucina-1/genética , Interleucina-1beta/genética , Lípidos/sangre , Lípidos/genética , Lipoproteínas LDL/sangre , Lipoproteínas LDL/genética , Masculino , Infarto del Miocardio/sangre , Infarto del Miocardio/epidemiología , Infarto del Miocardio/genética , Polimorfismo de Nucleótido Simple
10.
J Biochem Mol Toxicol ; 36(1): e22939, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34719845

RESUMEN

The anti-senescence function of genistein is related to inhibiting oxidative stress, however, the mechanism has not been clarified. The present study aimed to explore the effects of genistein on oxidized low-density lipoprotein (ox-LDL)-induced endothelial senescence and the role of the sirtuin-1 (SIRT1)-66-kDa Src homology 2 domain-containing protein (p66Shc)-forkhead box protein O3 (Foxo3a) pathways in the process. In this paper, human umbilical vein endothelial cells were pretreated with 1000 nM genistein for 30 min and then incubated with 50 mg/L ox-LDL for another 12 h; meanwhile, the functions of adenovirus-mediated overexpression of p66shc and small interfering RNA-mediated silencing of SIRT1 were investigated. Results showed that genistein pretreatment alleviated ox-LDL-induced mitochondrial reactive oxygen species, the levels of oxidatively modified DNA (8-OHdG) and pai-1, and the activity of SA-ß-gal, which was associated with mitigating p66shc. Further studies indicated the inhibitory effect of genistein on p66shc was correlated with suppressing the acetylation and phosphorylation of p66shc, and ameliorating its mitochondrial translocation by activating SIRT1. Moreover, the inactivated p66shc could enhance the activity of Foxo3a via restraining the phosphorylation and triggering nucleus accumulation. The study demonstrates genistein could prevent ox-LDL-induced mitochondrial oxidative stress and senescence through the SIRT1-p66shc-Foxo3a pathways.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Genisteína/farmacología , Lipoproteínas LDL/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Proteína Forkhead Box O3/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipoproteínas LDL/genética , Sirtuina 1/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética
11.
J Biochem Mol Toxicol ; 36(11): e23198, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35993694

RESUMEN

Atherosclerosis (AS) is a chronic inflammatory disease characterized by the formation of atherosclerotic plaque in the intima of arteries. Among the known regulators of atherosclerosis, microRNAs (miRNAs) have been reported to play critical roles in lipoprotein homeostasis and plaque formation. But the roles of microRNA-125a-3p (miR-125a-3p) in the pathogenesis of AS remain unknown. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to construct the vascular injury model of AS pathogenesis in vitro. miR-125a-3p and BMP and activin membrane-bound inhibitor (BAMBI) expression levels in HUVECs were then measured by quantitative real-time polymerase chain reaction and western blot. The viability and apoptosis of HUVECs were analyzed by Cell Counting Kit-8 assay, TUNEL assay, and flow cytometry, respectively. The relationship between BAMBI 3'-untranslated region and miR-125a-3p was validated by dual luciferase reporter gene assay. miR-125a-3p expression was raised in HUVECs induced with ox-LDL. In HUVECs, miR-125a-3p enhanced the effects of ox-LDL treatment on repressing the viability and promoting the apoptosis of cells. Additionally, BAMBI was confirmed as a direct target of miR-125a-3p and BAMBI overexpression reversed the effects of miR-125a-3p on HUVECs. miR-125a-3p aggravates the dysfunction of HUVECs induced by ox-LDL via BAMBI, which implies that miR-125a-3p is involved in the pathogenesis of AS.


Asunto(s)
Aterosclerosis , Células Endoteliales de la Vena Umbilical Humana , Lipoproteínas LDL , Proteínas de la Membrana , MicroARNs , Humanos , Apoptosis/genética , Apoptosis/fisiología , Aterosclerosis/genética , Aterosclerosis/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/patología , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , MicroARNs/genética , MicroARNs/metabolismo
12.
Mol Ther ; 29(6): 2019-2029, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-33609733

RESUMEN

Gene disruption via programmable, sequence-specific nucleases represents a promising gene therapy strategy in which the reduction of specific protein levels provides a therapeutic benefit. Proprotein convertase subtilisin/kexin type 9 (PCSK9), an antagonist of the low-density lipoprotein (LDL) receptor, is a suitable target for nuclease-mediated gene disruption as an approach to treat hypercholesterolemia. We sought to determine the long-term durability and safety of PCSK9 knockdown in non-human primate (NHP) liver by adeno-associated virus (AAV)-delivered meganuclease following our initial report on the feasibility of this strategy. Six previously treated NHPs and additional NHPs administered AAV-meganuclease in combination with corticosteroid treatment or an alternative AAV serotype were monitored for a period of up to 3 years. The treated NHPs exhibited a sustained reduction in circulating PCSK9 and LDL cholesterol (LDL-c) through the course of the study concomitant with stable gene editing of the PCSK9 locus. Low-frequency off-target editing remained stable, and no obvious adverse changes in histopathology of the liver were detected. We demonstrate similar on-target nuclease activity in primary human hepatocytes using a chimeric liver-humanized mouse model. These studies demonstrate that targeted in vivo gene disruption exerts a lasting therapeutic effect and provide pivotal data for safety considerations, which support clinical translation.


Asunto(s)
Edición Génica , Lipoproteínas LDL/metabolismo , Proproteína Convertasa 9/genética , Animales , Sistemas CRISPR-Cas , Dependovirus/genética , Modelos Animales de Enfermedad , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Lipoproteínas LDL/genética , Hígado/metabolismo , Ratones , Ratones Noqueados , Primates , Proproteína Convertasa 9/metabolismo
13.
J Hum Nutr Diet ; 35(4): 651-662, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34908197

RESUMEN

BACKGROUND: Type 2 diabetes mellitus (T2DM) is a multidimensional consequence of environmental and genetic factors. Cholesteryl ester transfer protein (CETP) Taq1B polymorphism has been reported as a main predictor of dyslipidaemia, comprising an important complication in persons with T2DM. However, diet could affect T2DM patients metabolic health. METHODS: We investigated the combination of gene-diet effects on some metabolic biomarkers. In our cross-sectional study, blood samples of 220 patients were collected. Dietary indices (healthy eating index, dietary quality index and dietary phytochemical index) were obtained from a validated semi-quantitative food frequency questionnaire. CETP Taq1B polymorphism was genotyped by a polymerase chain reaction-restriction fragment polymorphism method. Data were analysed by analysis of covariance. RESULTS: The interaction between the CETP Taq1B polymorphism and dietary indices on low density lipoprotein/high density lipoprotein was significant (p < 0.001 both crude and adjusted models). In addition, the interaction between polymorphism and dietary quality index on total antioxidant capacity (p = 0.004 crude model, p = 0.005 after adjusting) and pentraxin 3 (p = 0.01 both crude and adjusted models) was significant. Also, the interaction between polymorphism and healthy eating index on waist circumference (p = 0.005 both crude and adjusted models) and dietary phytochemical index on interleukin-18 (p = 0.03 crude model) was significant. CONCLUSIONS: Our results indicated the effect of CETP Taq1B polymorphism on some inflammatory and anthropometrics markers (total antioxidant capacity, pentraxin 3, interleukin-18, low density lipoprotein/high density lipoprotein and waist circumference) with high and low adherence to dietary incides.


Asunto(s)
Proteínas de Transferencia de Ésteres de Colesterol , Diabetes Mellitus Tipo 2 , Antioxidantes , Biomarcadores , Proteínas de Transferencia de Ésteres de Colesterol/genética , Estudios Transversales , Diabetes Mellitus Tipo 2/genética , Genotipo , Humanos , Interleucina-18/genética , Lipoproteínas HDL/genética , Lipoproteínas LDL/genética
14.
Curr Opin Lipidol ; 32(6): 355-362, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34653074

RESUMEN

PURPOSE OF REVIEW: Antisense oligomers (ASOs) have been available for decades: however, only recently have these molecules been applied clinically. This review aims to discuss the possible development of antisense-mediated splice correction therapies as precision medicines for familial hypercholesterolemic patients carrying mutations that compromise normal splicing of the low-density lipoprotein receptor (LDLR) gene transcript. RECENT FINDINGS: Three antisense drugs are currently being assessed in ongoing clinical trials for dyslipidemias, aiming to lower the plasma concentrations of lipoproteins that lead to end-organ damage, principally coronary artery disease. Although a handful of drugs may be applicable to many patients with familial hypercholesterolemia (FH), mutation-specific personalised antisense drugs may be even more effective in selected patients. Currently, there is no therapy that effectively addresses mutations in the LDLR, the major cause of FH. Many mutations in the LDLR that disrupt normal pre-mRNA processing could be applicable to splice correction therapy to restore receptor activity. SUMMARY: Precision medicine could provide long-term economic and social benefits if they can be implemented effectively and sustainably. Many mutations found in the LDLR gene could be amendable to therapeutic splice correction and we should consider developing a therapeutic ASO platform for these mutations.


Asunto(s)
Hiperlipoproteinemia Tipo II , Receptores de LDL , Humanos , Hiperlipoproteinemia Tipo II/genética , Hiperlipoproteinemia Tipo II/terapia , Lipoproteínas LDL/genética , Mutación , Receptores de LDL/genética
15.
J Biol Chem ; 295(22): 7697-7709, 2020 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-32332094

RESUMEN

Lysine N-pyrrolation converts lysine residues to Nϵ-pyrrole-l-lysine (pyrK) in a covalent modification reaction that significantly affects the chemical properties of proteins, causing them to mimic DNA. pyrK in proteins has been detected in vivo, indicating that pyrrolation occurs as an endogenous reaction. However, the source of pyrK remains unknown. In this study, on the basis of our observation in vitro that pyrK is present in oxidized low-density lipoprotein and in modified proteins with oxidized polyunsaturated fatty acids, we used LC-electrospray ionization-MS/MS coupled with a stable isotope dilution method to perform activity-guided separation of active molecules in oxidized lipids and identified glycolaldehyde (GA) as a pyrK source. The results from mechanistic experiments to study GA-mediated lysine N-pyrrolation suggested that the reactions might include GA oxidation, generating the dialdehyde glyoxal, followed by condensation reactions of lysine amino groups with GA and glyoxal. We also studied the functional significance of GA-mediated lysine N-pyrrolation in proteins and found that GA-modified proteins are recognized by apolipoprotein E, a binding target of pyrrolated proteins. Moreover, GA-modified proteins triggered an immune response to pyrrolated proteins, and monoclonal antibodies generated from mice immunized with GA-modified proteins specifically recognized pyrrolated proteins. These findings reveal that GA is an endogenous source of DNA-mimicking pyrrolated proteins and may provide mechanistic insights relevant for innate and autoimmune responses associated with glucose metabolism and oxidative stress.


Asunto(s)
Acetaldehído/análogos & derivados , Glucosa/metabolismo , Lipoproteínas LDL/metabolismo , Estrés Oxidativo , Procesamiento Proteico-Postraduccional , Acetaldehído/metabolismo , Animales , Glucosa/genética , Lipoproteínas LDL/genética , Masculino , Ratones , Ratones Noqueados para ApoE
16.
J Biol Chem ; 295(47): 15870-15882, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-32913121

RESUMEN

Ascorbic acid, a water-soluble antioxidant, regulates various biological processes and is thought to influence cholesterol. However, little is known about the mechanisms underpinning ascorbic acid-mediated cholesterol metabolism. Here, we determined if ascorbic acid can regulate expression of proprotein convertase subtilisin/kexin 9 (PCSK9), which binds low-density lipoprotein receptor (LDLR) leading to its intracellular degradation, to influence low-density lipoprotein (LDL) metabolism. At cellular levels, ascorbic acid inhibited PCSK9 expression in HepG2 and Huh7 cell lines. Consequently, LDLR expression and cellular LDL uptake were enhanced. Similar effects of ascorbic acid on PCSK9 and LDLR expression were observed in mouse primary hepatocytes. Mechanistically, ascorbic acid suppressed PCSK9 expression in a forkhead box O3-dependent manner. In addition, ascorbic acid increased LDLR transcription by regulating sterol regulatory element-binding protein 2. In vivo, administration of ascorbic acid reduced serum PCSK9 levels and enhanced liver LDLR expression in C57BL/6J mice. Reciprocally, lack of ascorbic acid supplementation in L-gulono-γ-lactone oxidase deficient (Gulo-/-) mice increased circulating PCSK9 and LDL levels, and decreased liver LDLR expression, whereas ascorbic acid supplementation decreased PCSK9 and increased LDLR expression, ameliorating LDL levels in Gulo-/- mice fed a high fat diet. Moreover, ascorbic acid levels were negatively correlated to PCSK9, total and LDL levels in human serum samples. Taken together, these findings suggest that ascorbic acid reduces PCSK9 expression, leading to increased LDLR expression and cellular LDL uptake. Thus, supplementation of ascorbic acid may ameliorate lipid profiles in ascorbic acid-deficient species.


Asunto(s)
Ácido Ascórbico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proproteína Convertasa 9/biosíntesis , Receptores de LDL/biosíntesis , Animales , Células Hep G2 , Humanos , L-Gulonolactona Oxidasa/genética , L-Gulonolactona Oxidasa/metabolismo , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Proproteína Convertasa 9/genética , Receptores de LDL/genética
17.
Hum Mol Genet ; 28(4): 688-697, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445611

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a late-onset fatal neurodegenerative disorder that is predicted to increase across the globe by ~70% in the following decades. Understanding the disease causal mechanism underlying ALS and identifying modifiable risks factors for ALS hold the key for the development of effective preventative and treatment strategies. Here, we investigate the causal effects of four blood lipid traits that include high-density lipoprotein, low-density lipoprotein (LDL), total cholesterol and triglycerides on the risk of ALS. By leveraging instrument variables from multiple large-scale genome-wide association studies in both European and East Asian populations, we carry out one of the largest and most comprehensive Mendelian randomization analyses performed to date on the causal relationship between lipids and ALS. Among the four lipids, we found that only LDL is causally associated with ALS and that higher LDL level increases the risk of ALS in both the European and East Asian populations. Specifically, the odds ratio of ALS per 1 standard deviation (i.e. 39.0 mg/dL) increase of LDL is estimated to be 1.14 [95% confidence interval (CI), 1.05-1.24; P = 1.38E-3] in the European population and 1.06 (95% CI, 1.00-1.12; P = 0.044) in the East Asian population. The identified causal relationship between LDL and ALS is robust with respect to the choice of statistical methods and is validated through extensive sensitivity analyses that guard against various model assumption violations. Our study provides important evidence supporting the causal role of higher LDL on increasing the risk of ALS, paving ways for the development of preventative strategies for reducing the disease burden of ALS across multiple nations.


Asunto(s)
Esclerosis Amiotrófica Lateral/sangre , Esclerosis Amiotrófica Lateral/genética , Lípidos/genética , Lipoproteínas LDL/genética , Esclerosis Amiotrófica Lateral/fisiopatología , Colesterol/sangre , Colesterol/genética , Femenino , Humanos , Lípidos/sangre , Lipoproteínas HDL/sangre , Lipoproteínas HDL/genética , Lipoproteínas LDL/sangre , Masculino , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple/genética , Triglicéridos/sangre , Triglicéridos/genética
18.
Hum Mol Genet ; 28(22): 3734-3741, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31332430

RESUMEN

A main strategy for lowering plasma low-density lipoprotein (LDL) cholesterol levels is to increase the number of cell-surface LDL receptors (LDLRs). This can be achieved by increasing the synthesis or preventing the degradation of the LDLR. One mechanism by which an LDLR becomes non-functional is enzymatic cleavage within the 10 residue linker region between ligand-binding repeats 4 and 5. The cleaved LDLR has only three ligand-binding repeats and is unable to bind LDL. In this study, we have performed cell culture experiments to identify strategies to prevent this cleavage. As a part of these studies, we found that Asp193 within the linker region is critical for cleavage to occur. Moreover, both 14-mer synthetic peptides and antibodies directed against the linker region prevented cleavage. As a consequence, more functional LDLRs were observed on the cell surface. The observation that the cleaved LDLR was present in extracts from the human adrenal gland indicates that cleavage of the linker region takes place in vivo. Thus, preventing cleavage of the LDLR by pharmacological measures could represent a novel lipid-lowering strategy.


Asunto(s)
Lipoproteínas LDL/metabolismo , Receptores de LDL/genética , Receptores de LDL/fisiología , Animales , Anticuerpos/inmunología , Células CHO , Membrana Celular/metabolismo , LDL-Colesterol/genética , LDL-Colesterol/metabolismo , Cricetulus , Humanos , Ligandos , Metabolismo de los Lípidos/genética , Lipoproteínas LDL/genética , Péptidos/metabolismo , Unión Proteica/genética , Unión Proteica/fisiología
19.
Int J Mol Sci ; 22(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203120

RESUMEN

Activated alpha-2 Macroglobulin (α2M*) is specifically recognized by the cluster I/II of LRP1 (Low-density lipoprotein Receptor-related Protein-1). LRP1 is a scaffold protein for insulin receptor involved in the insulin-induced glucose transporter type 4 (GLUT4) translocation to plasma membrane and glucose uptake in different types of cells. Moreover, the cluster II of LRP1 plays a critical role in the internalization of atherogenic lipoproteins, such as aggregated Low-density Lipoproteins (aggLDL), promoting intracellular cholesteryl ester (CE) accumulation mainly in arterial intima and myocardium. The aggLDL uptake by LRP1 impairs GLUT4 traffic and the insulin response in cardiomyocytes. However, the link between CE accumulation, insulin action, and cardiac dysfunction are largely unknown. Here, we found that α2M* increased GLUT4 expression on cell surface by Rab4, Rab8A, and Rab10-mediated recycling through PI3K/Akt and MAPK/ERK signaling activation. Moreover, α2M* enhanced the insulin response increasing insulin-induced glucose uptake rate in the myocardium under normal conditions. On the other hand, α2M* blocked the intracellular CE accumulation, improved the insulin response and reduced cardiac damage in HL-1 cardiomyocytes exposed to aggLDL. In conclusion, α2M* by its agonist action on LRP1, counteracts the deleterious effects of aggLDL in cardiomyocytes, which may have therapeutic implications in cardiovascular diseases associated with hypercholesterolemia.


Asunto(s)
Membrana Celular/metabolismo , Insulina/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Macroglobulinas/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Western Blotting , Línea Celular , Glucosa/metabolismo , Insulina/farmacología , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Microscopía Confocal , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
20.
Curr Opin Lipidol ; 31(3): 125-131, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32304380

RESUMEN

PURPOSE OF REVIEW: To summarize the recent studies directly comparing LDL and lipoprotein(a) as causal factors for cardiovascular disease and mortality. RECENT FINDINGS: In approximately 100,000 individuals from the Copenhagen General Population Study for risk of myocardial infarction, in observational analyses per 39 mg/dl (1 mmol/l) cholesterol increase, the hazard ratio was 1.3 (95% confidence interval: 1.2-1.3) for LDL cholesterol and 1.6 (1.4-1.9) for lipoprotein(a) cholesterol. In corresponding genetic analyses, the causal risk ratio was 2.1 (1.3-3.4) for LDL and 2.0 (1.6-2.6) for lipoprotein(a). Also, a 15 mg/dl (0.39 mmol/l) cholesterol increase was associated with a hazard ratio for cardiovascular mortality of 1.05 (1.04-1.07) for LDL cholesterol and 1.18 (1.12-1.25) for lipoprotein(a) cholesterol. Corresponding values for all-cause mortality were 1.01 (1.00-1.01) for LDL cholesterol and 1.07 (1.04-1.10) for lipoprotein(a) cholesterol. In genetic, causal analyses, the mortality increases for elevated lipoprotein(a) appeared to be through apolipoprotein(a) kringle IV-2 rather than through lipoprotein(a) levels per se. SUMMARY: On cholesterol scales, lipoprotein(a) and LDL appeared equal as causal factors for myocardial infarction; however, lipoprotein(a) was most important for mortality. Lipoprotein(a) effects may not only be due to cholesterol content but could also be due to the structure of lipoprotein(a) resembling plasminogen.


Asunto(s)
Enfermedades Cardiovasculares/genética , Lipoproteína(a)/genética , Lipoproteínas LDL/genética , Infarto del Miocardio/genética , Enfermedades Cardiovasculares/mortalidad , Enfermedades Cardiovasculares/patología , Colesterol/genética , LDL-Colesterol/genética , Humanos , Infarto del Miocardio/mortalidad , Infarto del Miocardio/patología , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA