Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Dev Cell ; 56(9): 1238-1252.e5, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33891899

RESUMEN

The human placenta and its specialized cytotrophoblasts rapidly develop, have a compressed lifespan, govern pregnancy outcomes, and program the offspring's health. Understanding the molecular underpinnings of these behaviors informs development and disease. Profiling the extraembryonic epigenome and transcriptome during the 2nd and 3rd trimesters revealed H3K9 trimethylation overlapping deeply DNA hypomethylated domains with reduced gene expression and compartment-specific patterns that illuminated their functions. Cytotrophoblast DNA methylation increased, and several key histone modifications decreased across the genome as pregnancy advanced. Cytotrophoblasts from severe preeclampsia had substantially increased H3K27 acetylation globally and at genes that are normally downregulated at term but upregulated in this syndrome. In addition, some cases had an immature pattern of H3K27ac peaks, and others showed evidence of accelerated aging, suggesting subtype-specific alterations in severe preeclampsia. Thus, the cytotrophoblast epigenome dramatically reprograms during pregnancy, placental disease is associated with failures in this process, and H3K27 hyperacetylation is a feature of severe preeclampsia.


Asunto(s)
Epigenoma , Enfermedades Placentarias/genética , Enfermedades Placentarias/patología , Trofoblastos/metabolismo , Trofoblastos/patología , Acetilación , Metilación de ADN/genética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Histonas/metabolismo , Humanos , Lisina/metabolismo , Preeclampsia/genética , Embarazo , Procesamiento Proteico-Postraduccional
2.
Nucleic Acids Res ; 47(W1): W158-W165, 2019 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-31165883

RESUMEN

The WashU Epigenome Browser (https://epigenomegateway.wustl.edu/) provides visualization, integration and analysis tools for epigenomic datasets. Since 2010, it has provided the scientific community with data from large consortia including the Roadmap Epigenomics and the ENCODE projects. Recently, we refactored the codebase, redesigned the user interface, and developed various novel features. New features include: (i) visualization using virtual reality (VR), which has implications in biology education and the study of 3D chromatin structure; (ii) expanded public data hubs, including data from the 4DN, ENCODE, Roadmap Epigenomics, TaRGET, IHEC and TCGA consortia; (iii) a more responsive user interface; (iv) a history of interactions, which enables undo and redo; (v) a feature we call Live Browsing, which allows multiple users to collaborate remotely on the same session; (vi) the ability to visualize local tracks and data hubs. Amazon Web Services also hosts the redesign at https://epigenomegateway.org/.


Asunto(s)
Bases de Datos Genéticas , Epigenoma/genética , Programas Informáticos , Navegador Web , Conjuntos de Datos como Asunto , Epigenómica , Genoma Humano , Humanos , Internet , Interfaz Usuario-Computador
3.
Eur J Hum Genet ; 26(10): 1462-1477, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29955172

RESUMEN

Primary familial brain calcification (PFBC) is a rare cerebral microvascular calcifying disorder with a wide spectrum of motor, cognitive, and neuropsychiatric symptoms. It is typically inherited as an autosomal-dominant trait with four causative genes identified so far: SLC20A2, PDGFRB, PDGFB, and XPR1. Our study aimed at screening the coding regions of these genes in a series of 177 unrelated probands that fulfilled the diagnostic criteria for primary brain calcification regardless of their family history. Sequence variants were classified as pathogenic, likely pathogenic, or of uncertain significance (VUS), based on the ACMG-AMP recommendations. We identified 45 probands (25.4%) carrying either pathogenic or likely pathogenic variants (n = 34, 19.2%) or VUS (n = 11, 6.2%). SLC20A2 provided the highest contribution (16.9%), followed by XPR1 and PDGFB (3.4% each), and PDGFRB (1.7%). A total of 81.5% of carriers were symptomatic and the most recurrent symptoms were parkinsonism, cognitive impairment, and psychiatric disturbances (52.3%, 40.9%, and 38.6% of symptomatic individuals, respectively), with a wide range of age at onset (from childhood to 81 years). While the pathogenic and likely pathogenic variants identified in this study can be used for genetic counseling, the VUS will require additional evidence, such as recurrence in unrelated patients, in order to be classified as pathogenic.


Asunto(s)
Encefalopatías/genética , Calcinosis/genética , Disfunción Cognitiva/genética , Variación Genética/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Encefalopatías/fisiopatología , Calcinosis/fisiopatología , Niño , Disfunción Cognitiva/fisiopatología , Femenino , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje , Fenotipo , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Virales/efectos de los fármacos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Receptor de Retrovirus Xenotrópico y Politrópico , Adulto Joven
4.
Hum Mol Genet ; 27(17): 2965-2977, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29790959

RESUMEN

Transcriptional changes in Friedreich's ataxia (FRDA), a rare and debilitating recessive Mendelian neurodegenerative disorder, have been studied in affected but inaccessible tissues-such as dorsal root ganglia, sensory neurons and cerebellum-in animal models or small patient series. However, transcriptional changes induced by FRDA in peripheral blood, a readily accessible tissue, have not been characterized in a large sample. We used differential expression, association with disability stage, network analysis and enrichment analysis to characterize the peripheral blood transcriptome and identify genes that were differentially expressed in FRDA patients (n = 418) compared with both heterozygous expansion carriers (n = 228) and controls (n = 93 739 individuals in total), or were associated with disease progression, resulting in a disease signature for FRDA. We identified a transcriptional signature strongly enriched for an inflammatory innate immune response. Future studies should seek to further characterize the role of peripheral inflammation in FRDA pathology and determine its relevance to overall disease progression.


Asunto(s)
Biomarcadores/sangre , Ataxia de Friedreich/sangre , Ataxia de Friedreich/genética , Redes Reguladoras de Genes , Mediadores de Inflamación/sangre , Inflamación/genética , Transcriptoma , Adulto , Estudios de Casos y Controles , Femenino , Ataxia de Friedreich/patología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad
5.
BMC Genomics ; 18(1): 724, 2017 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-28899353

RESUMEN

BACKGROUND: Uncovering mechanisms of epigenome evolution is an essential step towards understanding the evolution of different cellular phenotypes. While studies have confirmed DNA methylation as a conserved epigenetic mechanism in mammalian development, little is known about the conservation of tissue-specific genome-wide DNA methylation patterns. RESULTS: Using a comparative epigenomics approach, we identified and compared the tissue-specific DNA methylation patterns of rat against those of mouse and human across three shared tissue types. We confirmed that tissue-specific differentially methylated regions are strongly associated with tissue-specific regulatory elements. Comparisons between species revealed that at a minimum 11-37% of tissue-specific DNA methylation patterns are conserved, a phenomenon that we define as epigenetic conservation. Conserved DNA methylation is accompanied by conservation of other epigenetic marks including histone modifications. Although a significant amount of locus-specific methylation is epigenetically conserved, the majority of tissue-specific DNA methylation is not conserved across the species and tissue types that we investigated. Examination of the genetic underpinning of epigenetic conservation suggests that primary sequence conservation is a driving force behind epigenetic conservation. In contrast, evolutionary dynamics of tissue-specific DNA methylation are best explained by the maintenance or turnover of binding sites for important transcription factors. CONCLUSIONS: Our study extends the limited literature of comparative epigenomics and suggests a new paradigm for epigenetic conservation without genetic conservation through analysis of transcription factor binding sites.


Asunto(s)
Secuencia Conservada , Metilación de ADN/genética , Animales , Sitios de Unión , Epigenómica , Evolución Molecular , Humanos , Ratones , Especificidad de Órganos , Ratas , Factores de Transcripción/metabolismo
6.
Neurobiol Aging ; 39: 82-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26923404

RESUMEN

The precise physiologic function of many of the recently discovered Alzheimer's disease risk variants remains unknown. The downstream effects of genetic variants remain largely unexplored. We studied the relationship between the top 10 non-APOE genes with cortical and hippocampal atrophy as markers of neurodegeneration using 1.5T magnetic resonance imaging, 1-million single nucleotide polymorphism Illumina Human Omni-Quad array and Illumina Human BeadChip peripheral blood expression array data on 50 cognitively normal and 98 mild cognitive impairment subjects. After explicit matching of cortical and hippocampal morphology, we computed in 3D, the cortical thickness and hippocampal radial distance measures for each participant. Associations between the top 10 non-APOE genome-wide hits and neurodegeneration were explored using linear regression. Map-wise statistical significance was determined with permutations using threshold of p < 0.01. MS4A6A rs610932 and ABCA7 rs3764650 demonstrated significant associations with cortical and hippocampal atrophy. Exploratory MS4A6A and ABCA7 peripheral blood expression analyses revealed a similar pattern of associations with cortical neurodegeneration. To our knowledge, this is the first report of the effect of ABCA7 and MS4A6A on neurodegeneration.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Corteza Cerebral/patología , Estudios de Asociación Genética , Variación Genética/genética , Hipocampo/patología , Proteínas de la Membrana/genética , Transportadoras de Casetes de Unión a ATP/sangre , Anciano , Anciano de 80 o más Años , Apolipoproteínas E/genética , Atrofia/genética , Femenino , Humanos , Masculino , Proteínas de la Membrana/sangre , Persona de Mediana Edad
7.
Neurocase ; 22(1): 76-83, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26040468

RESUMEN

Patients with frontotemporal lobar degeneration (FTLD) can show superimposed amyloid pathology, though the impact of amyloid on the clinical presentation of FTLD is not well characterized. This cross-sectional case-control study compared clinical features, fluorodeoxyglucose-positron emission tomography metabolism and gray matter volume loss in 30 patients with familial FTLD in whom amyloid status was confirmed with autopsy or Pittsburgh compound B-PET. Compared to the amyloid-negative patients, the amyloid-positive patients performed significantly worse on several cognitive tests and showed hypometabolism and volume loss in more temporoparietal regions. Our results suggest that in FTLD amyloid positivity is associated with a more Alzheimer's disease-like pattern of neurodegeneration.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Amiloide/metabolismo , Encéfalo/patología , Demencia Frontotemporal/patología , Degeneración Lobar Frontotemporal/patología , Sustancia Gris/patología , Anciano , Encéfalo/metabolismo , Estudios de Casos y Controles , Estudios Transversales , Femenino , Demencia Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Sustancia Gris/metabolismo , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad
8.
Nat Genet ; 47(6): 579-81, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25938945

RESUMEN

Primary familial brain calcification (PFBC) is a neurological disease characterized by calcium phosphate deposits in the basal ganglia and other brain regions and has thus far been associated with SLC20A2, PDGFB or PDGFRB mutations. We identified in multiple families with PFBC mutations in XPR1, a gene encoding a retroviral receptor with phosphate export function. These mutations alter phosphate export, implicating XPR1 and phosphate homeostasis in PFBC.


Asunto(s)
Encefalopatías Metabólicas Innatas/genética , Calcinosis/genética , Receptores Acoplados a Proteínas G/genética , Receptores Virales/genética , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Escala de Lod , Masculino , Persona de Mediana Edad , Mutación Missense , Enfermedades Neurodegenerativas/genética , Linaje , Receptor de Retrovirus Xenotrópico y Politrópico
9.
BMC Neurol ; 15: 47, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25880661

RESUMEN

BACKGROUND: Heritability of Alzheimer's disease (AD) is estimated at 74% and genetic contributors have been widely sought. The ε4 allele of apolipoprotein E (APOE) remains the strongest common risk factor for AD, with numerous other common variants contributing only modest risk for disease. Variability in clinical presentation of AD, which is typically amnestic (AmnAD) but can less commonly involve visuospatial, language and/or dysexecutive syndromes (atypical or AtAD), further complicates genetic analyses. Taking a multi-locus approach may increase the ability to identify individuals at highest risk for any AD syndrome. In this study, we sought to develop and investigate the utility of a multi-variant genetic risk assessment on a cohort of phenotypically heterogeneous patients with sporadic AD clinical diagnoses. METHODS: We genotyped 75 variants in our cohort and, using a two-staged study design, we developed a 17-marker AD risk score in a Discovery cohort (n = 59 cases, n = 133 controls) then assessed its utility in a second Validation cohort (n = 126 cases, n = 150 controls). We also performed a data-driven decision tree analysis to identify genetic and/or demographic criteria that are most useful for accurately differentiating all AD cases from controls. RESULTS: We confirmed APOE ε4 as a strong risk factor for AD. A 17-marker risk panel predicted AD significantly better than APOE genotype alone (P < 0.00001) in the Discovery cohort, but not in the Validation cohort. In decision tree analyses, we found that APOE best differentiated cases from controls only in AmnAD but not AtAD. In AtAD, HFE SNP rs1799945 was the strongest predictor of disease; variation in HFE has previously been implicated in AD risk in non-ε4 carriers. CONCLUSIONS: Our study suggests that APOE ε4 remains the best predictor of broad AD risk when compared to multiple other genetic factors with modest effects, that phenotypic heterogeneity in broad AD can complicate simple polygenic risk modeling, and supports the association between HFE and AD risk in individuals without APOE ε4.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Árboles de Decisión , Anciano , Anciano de 80 o más Años , Algoritmos , Alelos , Estudios de Cohortes , Femenino , Marcadores Genéticos/genética , Genotipo , Humanos , Masculino , Examen Neurológico , Fenotipo , Medición de Riesgo , Factores de Riesgo
10.
JAMA Neurol ; 72(4): 414-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25706306

RESUMEN

IMPORTANCE: Previous studies have indicated a heritable component of the etiology of neurodegenerative diseases such as Alzheimer disease (AD), frontotemporal dementia (FTD), and progressive supranuclear palsy (PSP). However, few have examined the contribution of low-frequency coding variants on a genome-wide level. OBJECTIVE: To identify low-frequency coding variants that affect susceptibility to AD, FTD, and PSP. DESIGN, SETTING, AND PARTICIPANTS: We used the Illumina HumanExome BeadChip array to genotype a large number of variants (most of which are low-frequency coding variants) in a cohort of patients with neurodegenerative disease (224 with AD, 168 with FTD, and 48 with PSP) and in 224 control individuals without dementia enrolled between 2005-2012 from multiple centers participating in the Genetic Investigation in Frontotemporal Dementia and Alzheimer's Disease (GIFT) Study. An additional multiancestral replication cohort of 240 patients with AD and 240 controls without dementia was used to validate suggestive findings. Variant-level association testing and gene-based testing were performed. MAIN OUTCOMES AND MEASURES: Statistical association of genetic variants with clinical diagnosis of AD, FTD, and PSP. RESULTS: Genetic variants typed by the exome array explained 44%, 53%, and 57% of the total phenotypic variance of AD, FTD, and PSP, respectively. An association with the known AD gene ABCA7 was replicated in several ancestries (discovery P=.0049, European P=.041, African American P=.043, and Asian P=.027), suggesting that exonic variants within this gene modify AD susceptibility. In addition, 2 suggestive candidate genes, DYSF (P=5.53×10(-5)) and PAXIP1 (P=2.26×10(-4)), were highlighted in patients with AD and differentially expressed in AD brain. Corroborating evidence from other exome array studies and gene expression data points toward potential involvement of these genes in the pathogenesis of AD. CONCLUSIONS AND RELEVANCE: Low-frequency coding variants with intermediate effect size may account for a significant fraction of the genetic susceptibility to AD and FTD. Furthermore, we found evidence that coding variants in the known susceptibility gene ABCA7, as well as candidate genes DYSF and PAXIP1, confer risk for AD.


Asunto(s)
Enfermedad de Alzheimer/genética , Exoma , Demencia Frontotemporal/genética , Predisposición Genética a la Enfermedad , Parálisis Supranuclear Progresiva/genética , Adulto , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/diagnóstico , Femenino , Demencia Frontotemporal/diagnóstico , Pruebas Genéticas/métodos , Variación Genética/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Masculino , Análisis por Micromatrices , Persona de Mediana Edad , Riesgo
12.
PLoS Genet ; 10(3): e1004211, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24603599

RESUMEN

Little is known about how changes in DNA methylation mediate risk for human diseases including dementia. Analysis of genome-wide methylation patterns in patients with two forms of tau-related dementia--progressive supranuclear palsy (PSP) and frontotemporal dementia (FTD)--revealed significant differentially methylated probes (DMPs) in patients versus unaffected controls. Remarkably, DMPs in PSP were clustered within the 17q21.31 region, previously known to harbor the major genetic risk factor for PSP. We identified and replicated a dose-dependent effect of the risk-associated H1 haplotype on methylation levels within the region in blood and brain. These data reveal that the H1 haplotype increases risk for tauopathy via differential methylation at that locus, indicating a mediating role for methylation in dementia pathophysiology.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética , Demencia Frontotemporal/genética , Parálisis Supranuclear Progresiva/genética , Tauopatías/genética , Encéfalo/metabolismo , Encéfalo/patología , Cromosomas Humanos Par 17 , Demencia Frontotemporal/patología , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Haplotipos , Humanos , Enfermedades Neurodegenerativas , Factores de Riesgo , Parálisis Supranuclear Progresiva/patología , Tauopatías/etiología , Tauopatías/patología , Proteínas tau/genética
13.
Nat Genet ; 45(9): 1077-82, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23913003

RESUMEN

Calcifications in the basal ganglia are a common incidental finding and are sometimes inherited as an autosomal dominant trait (idiopathic basal ganglia calcification (IBGC)). Recently, mutations in the PDGFRB gene coding for the platelet-derived growth factor receptor ß (PDGF-Rß) were linked to IBGC. Here we identify six families of different ancestry with nonsense and missense mutations in the gene encoding PDGF-B, the main ligand for PDGF-Rß. We also show that mice carrying hypomorphic Pdgfb alleles develop brain calcifications that show age-related expansion. The occurrence of these calcium depositions depends on the loss of endothelial PDGF-B and correlates with the degree of pericyte and blood-brain barrier deficiency. Thus, our data present a clear link between Pdgfb mutations and brain calcifications in mice, as well as between PDGFB mutations and IBGC in humans.


Asunto(s)
Enfermedades de los Ganglios Basales/genética , Enfermedades de los Ganglios Basales/patología , Calcinosis/genética , Mutación , Proteínas Proto-Oncogénicas c-sis/genética , Sustitución de Aminoácidos , Animales , Enfermedades de los Ganglios Basales/diagnóstico , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Femenino , Orden Génico , Humanos , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Linaje , Tomografía Computarizada por Rayos X
14.
Neurogenetics ; 14(1): 11-22, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23334463

RESUMEN

Familial idiopathic basal ganglia calcification (IBGC) or Fahr's disease is a rare neurodegenerative disorder characterized by calcium deposits in the basal ganglia and other brain regions, which is associated with neuropsychiatric and motor symptoms. Familial IBGC is genetically heterogeneous and typically transmitted in an autosomal dominant fashion. We performed a mutational analysis of SLC20A2, the first gene found to cause IBGC, to assess its genetic contribution to familial IBGC. We recruited 218 subjects from 29 IBGC-affected families of varied ancestry and collected medical history, neurological exam, and head CT scans to characterize each patient's disease status. We screened our patient cohort for mutations in SLC20A2. Twelve novel (nonsense, deletions, missense, and splice site) potentially pathogenic variants, one synonymous variant, and one previously reported mutation were identified in 13 families. Variants predicted to be deleterious cosegregated with disease in five families. Three families showed nonsegregation with clinical disease of such variants, but retrospective review of clinical and neuroimaging data strongly suggested previous misclassification. Overall, mutations in SLC20A2 account for as many as 41% of our familial IBGC cases. Our screen in a large series expands the catalog of SLC20A2 mutations identified to date and demonstrates that mutations in SLC20A2 are a major cause of familial IBGC. Non-perfect segregation patterns of predicted deleterious variants highlight the challenges of phenotypic assessment in this condition with highly variable clinical presentation.


Asunto(s)
Enfermedades de los Ganglios Basales/genética , Calcinosis/genética , Mutación , Enfermedades Neurodegenerativas/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Adulto , Anciano , Secuencia de Aminoácidos , Estudios de Cohortes , Análisis Mutacional de ADN , Familia , Femenino , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Modelos Biológicos , Datos de Secuencia Molecular , Mutación/fisiología , Estudios Retrospectivos
15.
Hum Mol Genet ; 21(15): 3500-12, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22556362

RESUMEN

Rare mutations in the gene encoding for tau (MAPT, microtubule-associated protein tau) cause frontotemporal dementia-spectrum (FTD-s) disorders, including FTD, progressive supranuclear palsy (PSP) and corticobasal syndrome, and a common extended haplotype spanning across the MAPT locus is associated with increased risk of PSP and Parkinson's disease. We identified a rare tau variant (p.A152T) in a patient with a clinical diagnosis of PSP and assessed its frequency in multiple independent series of patients with neurodegenerative conditions and controls, in a total of 15 369 subjects. Tau p.A152T significantly increases the risk for both FTD-s (n = 2139, OR = 3.0, CI: 1.6-5.6, P = 0.0005) and Alzheimer's disease (AD) (n = 3345, OR = 2.3, CI: 1.3-4.2, P = 0.004) compared with 9047 controls. Functionally, p.A152T (i) decreases the binding of tau to microtubules and therefore promotes microtubule assembly less efficiently; and (ii) reduces the tendency to form abnormal fibers. However, there is a pronounced increase in the formation of tau oligomers. Importantly, these findings suggest that other regions of the tau protein may be crucial in regulating normal function, as the p.A152 residue is distal to the domains considered responsible for microtubule interactions or aggregation. These data provide both the first genetic evidence and functional studies supporting the role of MAPT p.A152T as a rare risk factor for both FTD-s and AD and the concept that rare variants can increase the risk for relatively common, complex neurodegenerative diseases, but since no clear significance threshold for rare genetic variation has been established, some caution is warranted until the findings are further replicated.


Asunto(s)
Enfermedad de Alzheimer/genética , Demencia Frontotemporal/genética , Variación Genética , Proteínas tau/genética , Anciano , Enfermedad de Alzheimer/epidemiología , Demencia Frontotemporal/epidemiología , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Humanos , Persona de Mediana Edad , Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...