Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Infect Dis ; 22(1): 917, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36482326

RESUMEN

BACKGROUND: Despite the development and application of vaccines against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) around the world, the scientific community is still trying to find some therapies to avoid or ameliorate the fatal evolution of the Coronavirus disease 2019 (COVID-19). Since the publication of the potential use of ivermectin as a treatment against the disease, a pleiad of information about it has been published. However, the evidence is not strong or weak enough to conclude its usefulness in the clinical evolution of patients infected with SARS-CoV-2. We evaluate the efficacy and safety of ivermectin in the treatment of Mexican patients with asymptomatic and mild COVID-19 in a three-day administration in comparison to placebo. METHODS: A randomized, double-blind, placebo-controlled trial was carried out in 66 adults with asymptomatic and mild COVID-19. Patients were randomly assigned 1:1 ratio to ivermectin plus acetaminophen or placebo plus acetaminophen. The primary endpoint was the proportion of subjects without a disease progression to severity according to COVID-19 guidelines by the National Institutes of Health (NIH) since randomization to 14 days. RESULTS: None of the participants presented progression to a severe state in either group. Viral load was measured on Days 1, 5, and 14. No significant differences were observed in baseline or 14-day between groups (p = 0.720 and 0.362, respectively). However, on Day 5, a significant difference in viral load was observed between groups (p = 0.039). The frequency of symptoms was similar between groups, and no significant differences were observed. The most frequent symptom was cough. One severe adverse event associated with SARS-CoV-2 infection was observed in the ivermectin group. CONCLUSIONS: At standard doses, ivermectin is not effective to prevent progression to a severe state or reducing symptoms in adults with asymptomatic and mild COVID-19. Trial registration The study was registered with ClinicalTrial.gov (NCT04407507) on May 29, 2020.


Asunto(s)
COVID-19 , Ivermectina , Humanos , Progresión de la Enfermedad , Ivermectina/uso terapéutico , SARS-CoV-2 , Estados Unidos
2.
Cells ; 11(6)2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35326508

RESUMEN

Apart from the known associations between arachidonic acid (AA), weight gain, and neurological and immune function, AA exposure leads to alterations in global and gene-specific DNA methylation (DNAm) and fatty acid (FA) content in human cultured cells. However, it is unknown as to whether the latter effects occur in vivo and are maintained over extended periods of time and across generations. To address this issue, we asked whether AA supplementation for three consecutive generations (prior to coitus in sires or in utero in dams) affected offspring growth phenotypes, in addition to liver DNAm and FA profiles in mice. Twelve-week-old BALB/c mice were exposed daily to AA dissolved in soybean oil (vehicle, VH), or VH only, for 10 days prior to mating or during the entire pregnancy (20 days). On average, 15 mice were supplemented per generation, followed by analysis of offspring body weight and liver traits (x average = 36 and 10 per generation, respectively). Body weight cumulatively increased in F2 and F3 offspring generations and positively correlated with milligrams of paternal or maternal offspring AA exposure. A concomitant increase in liver weight was observed. Notably, akin to AA-challenged cultured cells, global DNAm and cis-7-hexadecenoic acid (16:1n-9), an anti-inflammatory FA that is dependent on stearoyl-CoA desaturase 1 (SCD1) activity, increased with milligrams of AA exposure. In accordance, liver Scd1 promoter methylation decreased with milligrams of germline AA exposure and was negatively correlated with liver weight. Our results show that mice retain cellular memories of AA exposure across generations that could potentially be beneficial to the innate immune system.


Asunto(s)
Suplementos Dietéticos , Aumento de Peso , Animales , Ácido Araquidónico , Epigénesis Genética , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Embarazo
3.
Thromb Haemost ; 121(11): 1541-1553, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33677828

RESUMEN

In addition to genetic and epigenetic inheritance, somatic variation may contribute to cardiovascular disease (CVD) risk. CVD-associated somatic mutations have been reported in human clonal hematopoiesis, but evidence in the atheroma is lacking. To probe for somatic variation in atherosclerosis, we sought single-nucleotide private variants (PVs) in whole-exome sequencing (WES) data of aorta, liver, and skeletal muscle of two C57BL/6J coisogenic male ApoE null/wild-type (WT) sibling pairs, and RNA-seq data of one of the two pairs. Relative to the C57BL/6 reference genome, we identified 9 and 11 ApoE null aorta- and liver-specific PVs that were shared by all WES and RNA-seq datasets. Corresponding PVs in WT sibling aorta and liver were 1 and 0, respectively, and not overlapping with ApoE null PVs. Pyrosequencing analysis of 4 representative PVs in 17 ApoE null aortas and livers confirmed tissue-specific shifts toward the alternative allele, in addition to significant deviations from mendelian allele ratios. Notably, all aorta and liver PVs were present in the dbSNP database and were predominantly transition mutations within atherosclerosis-related genes. The majority of PVs were in discrete clusters approximately 3 Mb and 65 to 73 Mb away from hypermutable immunoglobin loci in chromosome 6. These features were largely shared with previously reported CVD-associated somatic mutations in human clonal hematopoiesis. The observation that SNPs exhibit tissue-specific somatic DNA mosaicism in ApoE null mice is potentially relevant for genetic association study design. The proximity of PVs to hypermutable loci suggests testable mechanistic hypotheses.


Asunto(s)
Enfermedades de la Aorta/genética , Aterosclerosis/genética , Mosaicismo , Polimorfismo de Nucleótido Simple , Animales , Aorta/patología , Enfermedades de la Aorta/patología , Aterosclerosis/patología , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Predisposición Genética a la Enfermedad , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Fenotipo , RNA-Seq , Secuenciación del Exoma
4.
Curr Atheroscler Rep ; 22(10): 62, 2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32844297

RESUMEN

PURPOSE OF REVIEW: A detailed understanding of the epigenome of cardiovascular disease (CVD) should broaden current insights into mechanisms of atherogenesis and help identify suitable biomarkers for disease risk and progression. This review addresses the question whether a consensus has been reached on identifying the main aberrant DNA methylation profile in CVD. Additionally, it presents advances and setbacks in the search for specific CVD biomarkers. RECENT FINDINGS: Although the literature points to DNA hypermethylation as an epigenetic landmark of CVD, inconsistencies are significant. In particular, the DNA methylomes of peripheral blood cells and the vascular wall do not show a consistent direction of change in all studies. An additional significant hurdle is the relatively low study-to-study reproducibility and the difficulty to assess specificity for CVD. Nonetheless, a number of biologically plausible markers have been proposed that warrant further studies. An integrated model for dynamic changes of DNA methylation during the natural history of atherosclerosis predisposition and progression is presented, that might reconcile conflicting findings. Cohort design and technical criteria for DNA methylation analysis need to be further homogenized to allow for meaningful validation. As stable DNA methylation profiles are likely determined by genetic variants, many of which might control a range of diseases, it is anticipated that CVD biomarker discovery will be a delicate balancing act between reproducibility and specificity.


Asunto(s)
Aterosclerosis/genética , Metilación de ADN , Animales , Aterosclerosis/sangre , Biomarcadores , Islas de CpG/genética , Progresión de la Enfermedad , Epigénesis Genética , Humanos , Ratones
5.
J Am Heart Assoc ; 7(3)2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386205

RESUMEN

BACKGROUND: The signals that determine atherosclerosis-specific DNA methylation profiles are only partially known. We previously identified a 29-bp DNA motif (differential methylation motif [DMM]) proximal to CpG islands (CGIs) that undergo demethylation in advanced human atheromas. Those data hinted that the DMM docks modifiers of DNA methylation and transcription. METHODS AND RESULTS: We sought to functionally characterize the DMM. We showed that the DMM overlaps with the RNA polymerase III-binding B box of Alu short interspersed nuclear elements and contains a DR2 nuclear receptor response element. Pointing to a possible functional role for an Alu DMM, CGIs proximal (<100 bp) to near-intact DMM-harboring Alu are significantly less methylated relative to CGIs proximal to degenerate DMM-harboring Alu or to DMM-devoid mammalian-wide interspersed repeat short interspersed nuclear elements in human arteries. As for DMM-binding factors, LXRB (liver X receptor ß) binds the DMM in a DR2-dependent fashion, and LXR (liver X receptor) agonists induce significant hypermethylation of the bulk of Alu in THP-1 cells. Furthermore, we describe 3 intergenic long noncoding RNAs that harbor a DMM, are under transcriptional control by LXR agonists, and are differentially expressed between normal and atherosclerotic human aortas. Notably, CGIs adjacent to those long noncoding RNAs tend to be hypomethylated in symptomatic relative to stable human atheromas. CONCLUSIONS: Collectively, the data suggest that a DMM is associated with 2 distinct methylation states: relatively low methylation of in cis CGIs and Alu element hypermethylation. Based on the known atheroprotective role of LXRs, we propose that LXR agonist-induced Alu hypermethylation, a landmark of atherosclerosis, is a compensatory rather than proatherogenic response.


Asunto(s)
Elementos Alu , Aterosclerosis/genética , Islas de CpG , Metilación de ADN , Epigénesis Genética , Receptores X del Hígado/metabolismo , Motivos de Nucleótidos , Aterosclerosis/metabolismo , Benzoatos/farmacología , Bencilaminas/farmacología , Sitios de Unión , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Receptores X del Hígado/agonistas , Receptores X del Hígado/genética , Unión Proteica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células THP-1 , Técnicas del Sistema de Dos Híbridos
6.
Sci Rep ; 6: 25867, 2016 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-27181711

RESUMEN

Fatty acids (FA) modify DNA methylation in vitro, but limited information is available on whether corresponding associations exist in vivo and reflect any short-term effect of the diet. Associations between global DNA methylation and FAs were sought in blood from lactating infants (LI; n = 49) and adult males (AMM; n = 12) equally distributed across the three conventional BMI classes. AMM provided multiple samples at 2-hour intervals during 8 hours after either a single Western diet-representative meal (post-prandial samples) or no meal (fasting samples). Lipid/glucose profile, HDAC4 promoter and PDK4 5'UTR methylation were determined in AMM. Multiple regression analysis revealed that global (in LI) and both global and PDK4-specific DNA methylation (in AMM) were positively associated with eicosapentaenoic and arachidonic acid. HDAC4 methylation was inversely associated with arachidonic acid post-prandially in AMM. Global DNA methylation did not show any defined within-day pattern that would suggest a short-term response to the diet. Nonetheless, global DNA methylation was higher in normal weight subjects both post-prandially and in fasting and coincided with higher polyunsaturated relative to monounsaturated and saturated FAs. We show for the first time strong associations of DNA methylation with specific FAs in two human cohorts of distinct age, diet and postnatal development stage.


Asunto(s)
Metilación de ADN , Ayuno/sangre , Ácidos Grasos/sangre , Histona Desacetilasas/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Represoras/genética , Regiones no Traducidas 5' , Adulto , Ácido Araquidónico/sangre , Dieta Occidental/efectos adversos , Ácido Eicosapentaenoico/sangre , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Recién Nacido , Lactancia , Masculino , Periodo Posprandial , Regiones Promotoras Genéticas , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Análisis de Regresión , Células THP-1
7.
BMC Med Genomics ; 8: 7, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25881171

RESUMEN

BACKGROUND: Atherosclerosis severity-independent alterations in DNA methylation, a reversible and highly regulated DNA modification, have been detected in aortic atheromas, thus supporting the hypothesis that epigenetic mechanisms participate in the pathogenesis of atherosclerosis. One yet unaddressed issue is whether the progression of atherosclerosis is associated with an increase in DNA methylation drift in the vascular tissue. The purpose of the study was to identify CpG methylation profiles that vary with the progression of atherosclerosis in the human aorta. METHODS: We interrogated a set of donor-matched atherosclerotic and normal aortic samples ranging from histological grade III to VII, with a high-density (>450,000 CpG sites) DNA methylation microarray. RESULTS: We detected a correlation between histological grade and intra-pair differential methylation for 1,985 autosomal CpGs, the vast majority of which drifted towards hypermethylation with lesion progression. The identified CpG loci map to genes that are regulated by known critical transcription factors involved in atherosclerosis and participate in inflammatory and immune responses. Functional relevance was corroborated by crossing the DNA methylation profiles with expression data obtained in the same human aorta sample set, by a transcriptome-wide analysis of murine atherosclerotic aortas and from available public databases. CONCLUSIONS: Our work identifies for the first time atherosclerosis progression-specific DNA methylation profiles in the vascular tissue. These findings provide potential novel markers of lesion severity and targets to counteract the progression of the atheroma.


Asunto(s)
Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Metilación de ADN , Animales , Análisis por Conglomerados , Islas de CpG , Bases de Datos Genéticas , Progresión de la Enfermedad , Epigénesis Genética , Humanos , Sistema Inmunológico , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Placa Aterosclerótica/patología , Factores de Transcripción/metabolismo
8.
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...