Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Alzheimers Dement ; 20(1): 695-708, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37774088

RESUMEN

The Alzheimer's Disease Neuroimaging Initiative (ADNI) Private Partners Scientific Board (PPSB) encompasses members from industry, biotechnology, diagnostic, and non-profit organizations that have until recently been managed by the Foundation for the National Institutes of Health (FNIH) and provided financial and scientific support to ADNI programs. In this article, we review some of the major activities undertaken by the PPSB, focusing on those supporting the most recently completed National Institute on Aging grant, ADNI3, and the impact it has had on streamlining biomarker discovery and validation in Alzheimer's disease. We also provide a perspective on the gaps that may be filled with future PPSB activities as part of ADNI4 and beyond. HIGHLIGHTS: The Private Partners Scientific board (PPSB) continues to play a key role in enabling several Alzheimer's Disease Neuroimaging Initiative (ADNI) activities. PPSB working groups have led landscape assessments to provide valuable feedback on new technologies, platforms, and methods that may be taken up by ADNI in current or future iterations.


Asunto(s)
Enfermedad de Alzheimer , Investigación Biomédica , Factores de Coagulación Sanguínea , Humanos , Enfermedad de Alzheimer/diagnóstico por imagen , Neuroimagen/métodos , Biomarcadores
2.
Blood ; 127(3): 310-3, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26644450

RESUMEN

BRCA1 is critical for maintenance of genomic stability and interacts directly with several proteins that regulate hematopoietic stem cell function and are part of the Fanconi anemia (FA) double-strand break DNA repair pathway. The effects of complete BRCA1 deficiency on bone marrow (BM) function are unknown. To test the hypothesis that Brca1 is essential in hematopoiesis, we developed a conditional mouse model with Mx1-Cre-mediated Brca1 deletion. Mice lacking Brca1 in the BM have baseline cytopenias and develop spontaneous bone marrow failure or diverse hematologic malignancies by 6 months of age. Brca1(-/-) BM cells have a reduced capacity to form hematopoietic colonies in vitro and to reconstitute hematopoiesis in irradiated recipients, consistent with a hematopoietic progenitor functional defect. Brca1(-/-) BM cells also show FA-like hypersensitivity to the DNA crosslinking agent mitomycin C, and karyotypes feature genomic instability. Taken together, our results show that loss of Brca1 in murine BM causes hematopoietic defects similar to those seen in people with FA, which provides strong evidence that Brca1 is critical for normal hematopoiesis and that Brca1 is a bona fide FA-like gene.


Asunto(s)
Proteína BRCA1/deficiencia , Proteína BRCA1/genética , Neoplasias Hematológicas/genética , Hemoglobinuria Paroxística/genética , Anemia Aplásica , Animales , Médula Ósea/patología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Enfermedades de la Médula Ósea , Trastornos de Fallo de la Médula Ósea , Modelos Animales de Enfermedad , Inestabilidad Genómica , Neoplasias Hematológicas/diagnóstico , Hematopoyesis/genética , Hemoglobinuria Paroxística/diagnóstico , Humanos , Infiltración Leucémica , Recuento de Leucocitos , Ratones , Ratones Noqueados , Pancitopenia/genética , Pancitopenia/patología , Fenotipo
3.
Stem Cells ; 33(12): 3643-54, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26417967

RESUMEN

Epigenetic alterations play a central role in the control of normal and malignant blood cell development. We demonstrate here that expression of a truncated DNA methyltransferase 3B isoform DNMT3B7, which has been shown to alter cellular epigenetic patterns, decreases the overall number of hematopoietic stem and progenitor cells (HSPCs), and markedly diminishes blood cell reconstitution within the female hormonal microenvironment. Gene expression profiling of HSPCs isolated from DNMT3B7 transgenic embryos identified Apolipoprotein E (Apoe) as overexpressed. The CpG island controlling Apoe expression had lower levels of modified cytosines in DNMT3B7 transgenic HSPCs, corresponding with the observed increase in gene expression. Furthermore, we observed that spleens and bone marrows of female mice transplanted with DNMT3B7 transgenic HSPCs express very high levels of Apoe. Finally, the introduction of Apoe-overexpressing HSPCs into male recipients decreased bone marrow engraftment, recapitulating our original observations in female recipients. Our work reveals a dynamic interplay between the intrinsic epigenetic changes in HSPCs and extrinsic endocrine factors acting on these cells to regulate the efficiency of HSPC engraftment and reconstitution. We have identified a novel mechanism by which gender-specific hormones modulate HSPC function, which could serve as a target for augmenting hematopoiesis in cases with limited HSC functionality.


Asunto(s)
Apolipoproteínas E/biosíntesis , Islas de CpG/fisiología , Epigénesis Genética/fisiología , Hematopoyesis/fisiología , Caracteres Sexuales , Animales , Apolipoproteínas E/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Femenino , Masculino , Ratones , Ratones Noqueados
4.
Blood ; 121(11): 2059-63, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23315164

RESUMEN

The drivers of abnormal DNA methylation in human cancers include widespread aberrant splicing of the DNMT3B gene, producing abnormal transcripts that encode truncated proteins that may act as dominant negative isoforms. To test whether reduced Dnmt3b dosage can alter tumorigenesis, we bred Dnmt3b(+/-) mice to Eµ-Myc mice, a mouse model susceptible to B-cell lymphomas. Eµ-Myc/Dnmt3b(+/-) mice showed a dramatic acceleration of lymphomagenesis, greater even than that observed in Eµ-Myc mice that express a truncated DNMT3B isoform found in human tumors, DNMT3B7. This finding indicates that Dnmt3b can act as a haploinsufficient tumor suppressor gene. Although reduction in both Dnmt3b dosage and expression of DNMT3B7 within the Eµ-Myc system had similar effects on tumorigenesis and DNA hypermethylation, different molecular mechanisms appear to underlie these changes. This study offers insight into how de novo DNA methyltransferases function as tumor suppressors and the sensitivity of Myc-induced lymphomas to DNA methylation.


Asunto(s)
Transformación Celular Neoplásica/genética , ADN (Citosina-5-)-Metiltransferasas/fisiología , Genes Supresores de Tumor/fisiología , Haploinsuficiencia/fisiología , Linfoma de Células B/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Animales , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Haploinsuficiencia/genética , Linfoma de Células B/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Regiones Promotoras Genéticas/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ADN Metiltransferasa 3B
5.
Nucleic Acids Res ; 41(16): e157, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23861445

RESUMEN

5-hydroxymethylcytosine (5-hmC) is a recently discovered epigenetic modification that is altered in cancers. Genome-wide assays for 5-hmC determination are needed as many of the techniques for 5-methylcytosine (5-mC) determination, including methyl-sensitive restriction digestion and bisulfite sequencing cannot distinguish between 5-mC and 5-hmC. Glycosylation of 5-hmC residues by beta-glucosyl transferase (ß-GT) can make CCGG residues insensitive to digestion by MspI. Restriction digestion by HpaII, MspI or MspI after ß-GT conversion, followed by adapter ligation, massive parallel sequencing and custom bioinformatic analysis allowed us determine distribution of 5-mC and 5-hmC at single base pair resolution at MspI restriction sites. The resulting HpaII tiny fragment Enrichment by Ligation-mediated PCR with ß-GT (HELP-GT) assay identified 5-hmC loci that were validated at global level by liquid chromatography-mass spectrometry (LC-MS) and the locus-specific level by quantitative reverse transcriptase polymerase chain reaction of 5-hmC pull-down DNA. Hydroxymethylation at both promoter and intragenic locations correlated positively with gene expression. Analysis of pancreatic cancer samples revealed striking redistribution of 5-hmC sites in cancer cells and demonstrated enrichment of this modification at many oncogenic promoters such as GATA6. The HELP-GT assay allowed global determination of 5-hmC and 5-mC from low amounts of DNA and with the use of modest sequencing resources. Redistribution of 5-hmC seen in cancer highlights the importance of determination of this modification in conjugation with conventional methylome analysis.


Asunto(s)
Citosina/análogos & derivados , ADN de Neoplasias/química , 5-Metilcitosina/análisis , Animales , Línea Celular Tumoral , Transformación Celular Neoplásica , Citosina/análisis , Citosina/metabolismo , Expresión Génica , Genoma Humano , Genómica/métodos , Glicosiltransferasas/metabolismo , Humanos , Ratones , Neoplasias Pancreáticas/genética , Reacción en Cadena de la Polimerasa
6.
Genome Med ; 16(1): 43, 2024 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-38515211

RESUMEN

BACKGROUND: Limited understanding of the diversity of variants in the cystic fibrosis transmembrane conductance regulator (CFTR) gene across ancestries hampers efforts to advance molecular diagnosis of cystic fibrosis (CF). The consequences pose a risk of delayed diagnoses and subsequently worsened health outcomes for patients. Therefore, characterizing the spectrum of CFTR variants across ancestries is critical for revolutionizing molecular diagnoses of CF. METHODS: We analyzed 454,727 UK Biobank (UKBB) whole-exome sequences to characterize the diversity of CFTR variants across ancestries. Using the PanUKBB classification, the participants were assigned into six major groups: African (AFR), American/American Admixed (AMR), Central South Asia (CSA), East Asian (EAS), European (EUR), and Middle East (MID). We segregated ancestry-specific CFTR variants, including those that are CF-causing or clinically relevant. The ages of certain CF-causing variants were determined and analyzed for selective pressure effects, and curated phenotype analysis was performed for participants with clinically relevant CFTR genotypes. RESULTS: We detected over 4000 CFTR variants, including novel ancestry-specific variants, across six ancestries. Europeans had the most unique CFTR variants [n = 2212], while the American group had the least unique variants [n = 23]. F508del was the most prevalent CF-causing variant found in all ancestries, except in EAS, where V520F was the most prevalent. Common EAS variants such as 3600G > A, V456A, and V520, which appeared approximately 270, 215, and 338 generations ago, respectively, did not show evidence of selective pressure. Sixteen participants had two CF-causing variants, with two being diagnosed with CF. We found 154 participants harboring a CF-causing and varying clinical consequences (VCC) variant. Phenotype analysis performed for participants with multiple clinically relevant variants returned significant associations with CF and its pulmonary phenotypes [Bonferroni-adjusted p < 0.05]. CONCLUSIONS: We leveraged the UKBB database to comprehensively characterize the broad spectrum of CFTR variants across ancestries. The detection of over 4000 CFTR variants, including several ancestry-specific and uncharacterized CFTR variants, warrants the need for further characterization of their functional and clinical relevance. Overall, the presentation of classical CF phenotypes seen in non-CF diagnosed participants with more than one CF-causing variant indicates that they may benefit from current CFTR modulator therapies.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Humanos , Bancos de Muestras Biológicas , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Exoma , Mutación , Biobanco del Reino Unido
7.
Blood ; 118(9): 2551-5, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21734233

RESUMEN

TET2 converts 5-methylcytosine to 5-hydroxymethylcytosine (5-hmC) in DNA and is frequently mutated in myeloid malignancies, including myeloproliferative neoplasms. Here we show that the level of 5-hmC is decreased in granulocyte DNA from myeloproliferative neoplasm patients with TET2 mutations compared with granulocyte DNA from healthy patients. Inhibition of TET2 by RNA interference decreases 5-hmC levels in both human leukemia cell lines and cord blood CD34(+) cells. These results confirm the enzymatic function of TET2 in human hematopoietic cells. Knockdown of TET2 in cord blood CD34(+) cells skews progenitor differentiation toward the granulomonocytic lineage at the expense of lymphoid and erythroid lineages. In addition, by monitoring in vitro granulomonocytic development we found a decreased granulocytic differentiation and an increase in monocytic cells. Our results indicate that TET2 disruption affects 5-hmC levels in human myeloid cells and participates in the pathogenesis of myeloid malignancies through the disturbance of myeloid differentiation.


Asunto(s)
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Metilación de ADN/genética , Proteínas de Unión al ADN/fisiología , Eritropoyesis/genética , Células Madre Hematopoyéticas/citología , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Línea Celular Tumoral , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Citosina/biosíntesis , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Dioxigenasas , Eritropoyesis/fisiología , Sangre Fetal/citología , Vectores Genéticos/genética , Granulocitos/metabolismo , Granulocitos/patología , Humanos , Lentivirus/genética , Monocitos/metabolismo , Monocitos/patología , Mutación , Mielopoyesis/fisiología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/administración & dosificación
8.
Blood ; 118(13): 3559-69, 2011 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-21828137

RESUMEN

The phenotype of germinal center (GC) B cells includes the unique ability to tolerate rapid proliferation and the mutagenic actions of activation induced cytosine deaminase (AICDA). Given the importance of epigenetic patterning in determining cellular phenotypes, we examined DNA methylation and the role of DNA methyltransferases in the formation of GCs. DNA methylation profiling revealed a marked shift in DNA methylation patterning in GC B cells versus resting/naive B cells. This shift included significant differential methylation of 235 genes, with concordant inverse changes in gene expression affecting most notably genes of the NFkB and MAP kinase signaling pathways. GC B cells were predominantly hypomethylated compared with naive B cells and AICDA binding sites were highly overrepresented among hypomethylated loci. GC B cells also exhibited greater DNA methylation heterogeneity than naive B cells. Among DNA methyltransferases (DNMTs), only DNMT1 was significantly up-regulated in GC B cells. Dnmt1 hypomorphic mice displayed deficient GC formation and treatment of mice with the DNA methyltransferase inhibitor decitabine resulted in failure to form GCs after immune stimulation. Notably, the GC B cells of Dnmt1 hypomorphic animals showed evidence of increased DNA damage, suggesting dual roles for DNMT1 in DNA methylation and double strand DNA break repair.


Asunto(s)
Linfocitos B/fisiología , Diferenciación Celular/genética , ADN (Citosina-5-)-Metiltransferasas/fisiología , Metilación de ADN/fisiología , Centro Germinal/inmunología , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Diferenciación Celular/inmunología , Análisis por Conglomerados , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Epigénesis Genética/fisiología , Perfilación de la Expresión Génica , Centro Germinal/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Ovinos , Estudios de Validación como Asunto
9.
J Cheminform ; 15(1): 64, 2023 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-37468968

RESUMEN

The identification of human proteins that are amenable to pharmacologic modulation without significant off-target effects remains an important unsolved challenge. Computational methods have been devised to identify features which distinguish between "druggable" and "undruggable" proteins, finding that protein sequence, tissue and cellular localization, biological role, and position in the protein-protein interaction network are all important discriminant factors. However, many prior efforts to automate the assessment of protein druggability suffer from low performance or poor interpretability. We developed a neural network-based machine learning model capable of generating druggability sub-scores based on each of four distinct categories, combining them to form an overall druggability score. The model achieves an excellent performance in separating drugged and undrugged proteins in the human proteome, with an area under the receiver operating characteristic (AUC) of 0.95. Our use of multiple sub-scores allows the assessment of potential protein targets of interest based on distinct contributors to druggability, leading to a more interpretable and holistic model to identify novel targets.

10.
medRxiv ; 2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36993271

RESUMEN

Determining the genetic architecture of Alzheimer's disease (AD) pathologies can enhance mechanistic understanding and inform precision medicine strategies. Here, we performed a genome-wide association study of cortical tau quantified by positron emission tomography in 3,136 participants from 12 independent studies. The CYP1B1-RMDN2 locus was associated with tau deposition. The most significant signal was at rs2113389, which explained 4.3% of the variation in cortical tau, while APOE4 rs429358 accounted for 3.6%. rs2113389 was associated with higher tau and faster cognitive decline. Additive effects, but no interactions, were observed between rs2113389 and diagnosis, APOE4 , and Aß positivity. CYP1B1 expression was upregulated in AD. rs2113389 was associated with higher CYP1B1 expression and methylation levels. Mouse model studies provided additional functional evidence for a relationship between CYP1B1 and tau deposition but not Aß. These results may provide insight into the genetic basis of cerebral tau and novel pathways for therapeutic development in AD.

11.
Alzheimers Dement (Amst) ; 14(1): e12354, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36187194

RESUMEN

Introduction: The acceleration of biological aging is a risk factor for Alzheimer's disease (AD). Here, we performed weighted gene co-expression network analysis (WGCNA) to identify modules and dysregulated genesinvolved in biological aging in AD. Methods: We performed WGCNA to identify modules associated with biological clocks and hub genes of the module with the highest module significance. In addition, we performed differential expression analysis and association analysis with AD biomarkers. Results: WGCNA identified five modules associated with biological clocks, with the module designated as "purple" showing the strongest association. Functional enrichment analysis revealed that the purple module was related to cell migration and death. Ten genes were identified as hub genes in purple modules, of which CX3CR1 was downregulated in AD and low levels of CX3CR1 expression were associated with AD biomarkers. Conclusion: Network analysis identified genes associated with biological clocks, which suggests the genetic architecture underlying biological aging in AD. Highlights: Examine links between Alzheimer's disease (AD) peripheral transcriptome and biological aging changes.Weighted gene co-expression network analysis (WGCNA) found five modules related to biological aging.Among the hub genes of the module, CX3CR1 was downregulated in AD.The CX3CR1 expression level was associated with cognitive performance and brain atrophy.

12.
Clin Epigenetics ; 13(1): 191, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34654479

RESUMEN

BACKGROUND: Identifying biomarkers associated with Alzheimer's disease (AD) progression may enable patient enrichment and improve clinical trial designs. Epigenome-wide association studies have revealed correlations between DNA methylation at cytosine-phosphate-guanine (CpG) sites and AD pathology and diagnosis. Here, we report relationships between peripheral blood DNA methylation profiles measured using Infinium® MethylationEPIC BeadChip and AD progression in participants from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. RESULTS: The rate of cognitive decline from initial DNA sampling visit to subsequent visits was estimated by the slopes of the modified Preclinical Alzheimer Cognitive Composite (mPACC; mPACCdigit and mPACCtrailsB) and Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) plots using robust linear regression in cognitively normal (CN) participants and patients with mild cognitive impairment (MCI), respectively. In addition, diagnosis conversion status was assessed using a dichotomized endpoint. Two CpG sites were significantly associated with the slope of mPACC in CN participants (P < 5.79 × 10-8 [Bonferroni correction threshold]); cg00386386 was associated with the slope of mPACCdigit, and cg09422696 annotated to RP11-661A12.5 was associated with the slope of CDR-SB. No significant CpG sites associated with diagnosis conversion status were identified. Genes involved in cognition and learning were enriched. A total of 19, 13, and 5 differentially methylated regions (DMRs) associated with the slopes of mPACCtrailsB, mPACCdigit, and CDR-SB, respectively, were identified by both comb-p and DMRcate algorithms; these included DMRs annotated to HOXA4. Furthermore, 5 and 19 DMRs were associated with conversion status in CN and MCI participants, respectively. The most significant DMR was annotated to the AD-associated gene PM20D1 (chr1: 205,818,956 to 205,820,014 [13 probes], Sidak-corrected P = 7.74 × 10-24), which was associated with both the slope of CDR-SB and the MCI conversion status. CONCLUSION: Candidate CpG sites and regions in peripheral blood were identified as associated with the rate of cognitive decline in participants in the ADNI cohort. While we did not identify a single CpG site with sufficient clinical utility to be used by itself due to the observed effect size, a biosignature composed of DNA methylation changes may have utility as a prognostic biomarker for AD progression.


Asunto(s)
Enfermedad de Alzheimer/genética , Metilación de ADN/genética , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/diagnóstico , Biomarcadores/análisis , Biomarcadores/sangre , Disfunción Cognitiva/genética , Estudios de Cohortes , Metilación de ADN/fisiología , Progresión de la Enfermedad , Femenino , Humanos , Masculino
13.
Metallomics ; 12(12): 2186-2198, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33325950

RESUMEN

Iron regulatory proteins (IRPs) are iron-responsive RNA binding proteins that dictate changes in cellular iron metabolism in animal cells by controlling the fate of mRNAs containing iron responsive elements (IREs). IRPs have broader physiological roles as some targeted mRNAs encode proteins with functions beyond iron metabolism suggesting hierarchical regulation of IRP-targeted mRNAs. We observe that the translational regulation of IRP-targeted mRNAs encoding iron storage (L- and H-ferritins) and export (ferroportin) proteins have different set-points of iron responsiveness compared to that for the TCA cycle enzyme mitochondrial aconitase. The ferritins and ferroportin mRNA were largely translationally repressed in the liver of rats fed a normal diet whereas mitochondrial aconitase mRNA is primarily polysome bound. Consequently, acute iron overload increases polysome association of H- and L-ferritin and ferroportin mRNAs while mitochondrial aconitase mRNA showed little stimulation. Conversely, mitochondrial aconitase mRNA is most responsive in iron deficiency. These differences in regulation were associated with a faster off-rate of IRP1 for the IRE of mitochondrial aconitase in comparison to that of L-ferritin. Thus, hierarchical control of mRNA translation by IRPs involves selective control of cellular functions acting at different states of cellular iron status and that are critical for adaptations to iron deficiency or prevention of iron toxicity.


Asunto(s)
Anemia Ferropénica/genética , Sobrecarga de Hierro/genética , Proteínas Reguladoras del Hierro/genética , ARN Mensajero/genética , Animales , Proteínas de Transporte de Catión/genética , Ferritinas/genética , Masculino , Ratones , Biosíntesis de Proteínas , Ratas Sprague-Dawley
14.
Clin Epigenetics ; 12(1): 84, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32539856

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a chronic progressive neurodegenerative disease impacting an estimated 44 million adults worldwide. The causal pathology of AD (accumulation of amyloid-beta and tau), precedes hallmark symptoms of dementia by more than a decade, necessitating development of early diagnostic markers of disease onset, particularly for new drugs that aim to modify disease processes. To evaluate differentially methylated positions (DMPs) as novel blood-based biomarkers of AD, we used a subset of 653 individuals with peripheral blood (PB) samples in the Alzheimer's disease Neuroimaging Initiative (ADNI) consortium. The selected cohort of AD, mild cognitive impairment (MCI), and age-matched healthy controls (CN) all had imaging, genetics, transcriptomics, cerebrospinal protein markers, and comprehensive clinical records, providing a rich resource of concurrent multi-omics and phenotypic information on a well-phenotyped subset of ADNI participants. RESULTS: In this manuscript, we report cross-diagnosis differential peripheral DNA methylation in a cohort of AD, MCI, and age-matched CN individuals with longitudinal DNA methylation measurements. Epigenome-wide association studies (EWAS) were performed using a mixed model with repeated measures over time with a P value cutoff of 1 × 10-5 to test contrasts of pairwise differential peripheral methylation in AD vs CN, AD vs MCI, and MCI vs CN. The most highly significant differentially methylated loci also tracked with Mini Mental State Examination (MMSE) scores. Differentially methylated loci were enriched near brain and neurodegeneration-related genes (e.g., BDNF, BIN1, APOC1) validated using the genotype tissue expression project portal (GTex). CONCLUSIONS: Our work shows that peripheral differential methylation between age-matched subjects with AD relative to healthy controls will provide opportunities to further investigate and validate differential methylation as a surrogate of disease. Given the inaccessibility of brain tissue, the PB-associated methylation marks may help identify the stage of disease and progression phenotype, information that would be central to bringing forward successful drugs for AD.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Disfunción Cognitiva/diagnóstico por imagen , Metilación de ADN/genética , Neuroimagen/métodos , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/sangre , Enfermedad de Alzheimer/líquido cefalorraquídeo , Biomarcadores/sangre , Biomarcadores/líquido cefalorraquídeo , Estudios de Casos y Controles , Disfunción Cognitiva/sangre , Disfunción Cognitiva/líquido cefalorraquídeo , Diagnóstico Diferencial , Progresión de la Enfermedad , Diagnóstico Precoz , Epigenómica/métodos , Femenino , Genotipo , Humanos , Estudios Longitudinales , Masculino , Pruebas de Estado Mental y Demencia/normas , Fenotipo , Transcriptoma/genética
15.
PLoS One ; 13(5): e0198296, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29851985

RESUMEN

BACKGROUND: Genetic variants of inosine triphosphatase (ITPA) that confer reduced ITPase activity are associated with protection against ribavirin(RBV)-induced hemolytic anemia in peginterferon(IFN)/RBV-based treatment of hepatitis C virus (HCV). Patients with reduced ITPase activity showed improved treatment efficacy when treated with IFN/RBV. In addition, a genetic polymorphism near the IL28B gene is associated with an improved response to IFN/RBV treatment. RBV has been an important component of IFN-containing regimens, and is currently recommended in combination with several IFN-free regimens for treatment of harder to cure HCV infections. AIM: To evaluate whether genetic variations that reduce ITPase activity impact RBV-induced anemia in IFN-free/RBV regimens. METHODS: In this study, genetic analyses were conducted in the PEARL-IV trial to investigate the effect of activity-reducing ITPA variants as well as IL28B polymorphism on anemia, platelet (PLT) counts, and virologic response in HCV genotype1a-infected patients treated with the direct-acting antiviral (DAA) regimen of ombitasvir/paritaprevir/ritonavir and dasabuvir±RBV. RESULTS: Reduction in ITPase activity and homozygosity for the IL28Brs12979860 CC genotype protected against RBV-induced anemia. In patients receiving RBV, reduced ITPase activity was associated with reduced plasma RBV concentration and higher PLT counts. ITPase activity had no impact on response to DAA treatment, viral kinetics, or baseline IP-10 levels. CONCLUSIONS: Our study demonstrates that genetics of ITPA and IL28B may help identify patients protected from RBV-induced anemia when treated with IFN-free regimens. Our work demonstrates for the first time that IL28B genetics may also have an impact on RBV-induced anemia. This may be of particular significance in patients with difficult-to-cure HCV infections, such as patients with decompensated cirrhosis where RBV-containing regimens likely will continue to be recommended.


Asunto(s)
Anemia Hemolítica/inducido químicamente , Anemia Hemolítica/genética , Antivirales/efectos adversos , Interleucinas/genética , Polimorfismo Genético , Pirofosfatasas/metabolismo , Ribavirina/efectos adversos , Anemia Hemolítica/enzimología , Femenino , Humanos , Interferones , Masculino , Inosina Trifosfatasa
17.
Cancer Genet ; 208(5): 167-77, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25892122

RESUMEN

Emerging data have demonstrated that 5-methylcytosine (5-mC) and its oxidized products 5-hydroxymethylcytosine (5-hmC), 5-formylcytosine (5-fC), and 5-carboxylcytosine (5-CaC) play unique roles in several biological processes, including the control of gene expression and in the pathogenesis of cancer. In this review, we focus on 5-hmC and the disruption of its distribution in several cancers, including hematological malignancies and solid tumors. We present an outline of how 5-hmC is closely associated with metabolic pathways and may be the missing link connecting epigenetics with metabolism in the context of cancer cells. Finally, we discuss the diagnostic and prognostic importance of 5-mC and 5-hmC patterning, and how we may be able to establish new paradigms in cancer therapy based on these alterations.


Asunto(s)
5-Metilcitosina/metabolismo , Citosina/análogos & derivados , Metilación de ADN , Neoplasias/diagnóstico , Neoplasias/terapia , Citosina/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Humanos , Isocitrato Deshidrogenasa/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas/genética
18.
Cell Rep ; 12(12): 2086-98, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26365193

RESUMEN

Changes in DNA methylation are required for the formation of germinal centers (GCs), but the mechanisms of such changes are poorly understood. Activation-induced cytidine deaminase (AID) has been recently implicated in DNA demethylation through its deaminase activity coupled with DNA repair. We investigated the epigenetic function of AID in vivo in germinal center B cells (GCBs) isolated from wild-type (WT) and AID-deficient (Aicda(-/-)) mice. We determined that the transit of B cells through the GC is associated with marked locus-specific loss of methylation and increased methylation diversity, both of which are lost in Aicda(-/-) animals. Differentially methylated cytosines (DMCs) between GCBs and naive B cells (NBs) are enriched in genes that are targeted for somatic hypermutation (SHM) by AID, and these genes form networks required for B cell development and proliferation. Finally, we observed significant conservation of AID-dependent epigenetic reprogramming between mouse and human B cells.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/metabolismo , Epigénesis Genética , Centro Germinal/metabolismo , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Secuencia Conservada , Citidina Desaminasa/genética , Citidina Desaminasa/inmunología , Citosina/metabolismo , Metilación de ADN , Centro Germinal/citología , Centro Germinal/inmunología , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados
19.
Cancer Res ; 75(14): 2833-43, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25972343

RESUMEN

TET2 enzymatically converts 5-methylcytosine to 5-hydroxymethylcytosine as well as other covalently modified cytosines and its mutations are common in myeloid leukemia. However, the exact mechanism and the extent to which TET2 mutations affect DNA methylation remain in question. Here, we report on DNA methylomes in TET2 wild-type (TET2-WT) and mutant (TET2-MT) cases of chronic myelomonocytic leukemia (CMML). We analyzed 85,134 CpG sites [28,114 sites in CpG islands (CGI) and 57,020 in non-CpG islands (NCGI)]. TET2 mutations do not explain genome-wide differences in DNA methylation in CMML, and we found few and inconsistent differences at CGIs between TET2-WT and TET2-MT cases. In contrast, we identified 409 (0.71%) TET2-specific differentially methylated CpGs (tet2-DMCs) in NCGIs, 86% of which were hypermethylated in TET2-MT cases, suggesting a strikingly different biology of the effects of TET2 mutations at CGIs and NCGIs. DNA methylation of tet2-DMCs at promoters and nonpromoters repressed gene expression. Tet2-DMCs showed significant enrichment at hematopoietic-specific enhancers marked by H3K4me1 and at binding sites for the transcription factor p300. Tet2-DMCs showed significantly lower 5-hydroxymethylcytosine in TET2-MT cases. We conclude that leukemia-associated TET2 mutations affect DNA methylation at NCGI regions containing hematopoietic-specific enhancers and transcription factor-binding sites.


Asunto(s)
Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos/genética , Leucemia Mielomonocítica Crónica/genética , Mutación Missense , Proteínas Proto-Oncogénicas/genética , Factores de Transcripción/metabolismo , Sitios de Unión/genética , Estudios de Casos y Controles , Islas de CpG , Dioxigenasas , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Genoma Humano , Hematopoyesis/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Análisis por Micromatrices , Regiones Promotoras Genéticas/genética
20.
Cell Stem Cell ; 14(4): 421-2, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702991

RESUMEN

Recent studies (Shlush et al., 2014; Corces-Zimmerman et al., 2014) have demonstrated that leukemias develop from hematopoietic stem cells that acquire preleukemic mutations, allowing clonal expansion and subsequent acquisition of mutations leading to cancer. Preleukemic cells survive chemotherapy and serve as reservoirs for disease, generating new clones and leading to relapse.


Asunto(s)
Evolución Clonal/genética , Epigénesis Genética/genética , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Modelos Biológicos , Células Madre Neoplásicas/citología , Preleucemia/genética , Animales , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA