Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34916285

RESUMEN

Spina bifida (SB) is a debilitating birth defect caused by multiple gene and environment interactions. Though SB shows non-Mendelian inheritance, genetic factors contribute to an estimated 70% of cases. Nevertheless, identifying human mutations conferring SB risk is challenging due to its relative rarity, genetic heterogeneity, incomplete penetrance, and environmental influences that hamper genome-wide association studies approaches to untargeted discovery. Thus, SB genetic studies may suffer from population substructure and/or selection bias introduced by typical candidate gene searches. We report a population based, ancestry-matched whole-genome sequence analysis of SB genetic predisposition using a systems biology strategy to interrogate 298 case-control subject genomes (149 pairs). Genes that were enriched in likely gene disrupting (LGD), rare protein-coding variants were subjected to machine learning analysis to identify genes in which LGD variants occur with a different frequency in cases versus controls and so discriminate between these groups. Those genes with high discriminatory potential for SB significantly enriched pathways pertaining to carbon metabolism, inflammation, innate immunity, cytoskeletal regulation, and essential transcriptional regulation consistent with their having impact on the pathogenesis of human SB. Additionally, an interrogation of conserved noncoding sequences identified robust variant enrichment in regulatory regions of several transcription factors critical to embryonic development. This genome-wide perspective offers an effective approach to the interrogation of coding and noncoding sequence variant contributions to rare complex genetic disorders.


Asunto(s)
Genoma Humano , Disrafia Espinal/genética , Estudios de Casos y Controles , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Biología de Sistemas , Factores de Transcripción/genética
2.
Neuromuscul Disord ; 30(6): 457-471, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32444167

RESUMEN

Congenital LAMA2 related muscular dystrophy (LAMA2-RD), the most commonly recognized type of congenital muscular dystrophies, has been described in patients' cohorts from Europe and the UK but not from Middle-Eastern. This study aimed to reveal the prevalence, clinical and genomic characteristics of congenital LAMA2-RD in a patient's cohort of 17 families (21 patients) from the Gulf and Middle East. Affected subjects exhibited the classic phenotype of generalized hypotonia, developmental delay, and progressive muscular weakness. Despite the homogeneous background of most of our patients, clinical variability was evident; however, none of our patients was able to achieve independent ambulation. The associated features of nephrocalcinosis, infantile-onset osteopenia, and cardiac arrest were first described in this study. LAMA2 mutations constituted 48% of the genetic causes underlying congenital muscular dystrophies (CMDs) in our patients. We estimated a point prevalence of 0.8 in 100.000 for LAMA2-RD in Qatar, relatively higher compared to that described in Europe's studies. The founder mutation and high rate of consanguinity are potential contributors. This study identified five LAMA2 truncating variants, two novel and three recurrent, of which the c.6488delA-frameshift that was found in 12 unrelated Qatari families, highlighting a founder mutation in Qatari patients. The two novel variants involved an acceptor splice site and N-terminus deletion that removes the LAMA2 promoter, exon1, and part of intron1. The "residual" expression of LAMA2 transcript and protein associated with this large N-terminus deletion suggested an alternative promoter that, while seems to be activated, acts less efficiently.


Asunto(s)
Laminina/genética , Distrofias Musculares/genética , Distrofias Musculares/patología , Distrofias Musculares/fisiopatología , Adolescente , Niño , Preescolar , Consanguinidad , Femenino , Efecto Fundador , Mutación del Sistema de Lectura , Humanos , Lactante , Masculino , Linaje , Qatar
3.
BMC Med Genet ; 18(1): 33, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28327087

RESUMEN

BACKGROUND: Hereditary Spastic Paraplegia (HSP) is a genetically heterogeneous group of neurodegenerative diseases. Thin Corpus Callosum (TCC) associated HSP is a distinguished subgroup of complex forms. Purines and pyrimidine, the basic DNA and RNA components, are regulating the cell metabolism, having roles in signal transduction, energy preservation and cellular repair. Genetic defects in nucleotide metabolism related genes have been only recently implicated in brain and neurodegenerative diseases' pathogenesis. CASE PRESENTATION: We present a consanguineous Qatari family with two brothers, 9 and 3 years, who displayed a characteristic phenotype of early onset and markedly-severe spasticity with tiptoe walking, delayed dysarthric speech, persistent truncal hypotonia, and multiple variable-sized areas of brownish skin discoloration appearing at different places on the body. A clinical diagnosis suggestive of complex hereditary spastic paraplegia (HSP) was set after the family had the second affected child. Whole genome sequencing identified a novel homozygous NT5C2 splice site mutation (NM_012229.4/NM_001134373.2: c.1159 + 1G > T) that recessively segregated in family members. Brain MRI revealed dysgenic and thin corpus callosum (TCC) with peri-trigonal white matter cystic changes in both affected boys, whereas a well-developed corpus callosum with normal white matter was shown in their apparently normal brother, who found to be a carrier for the mutant variant. This mutation led to skipping of exon 14 with removal of 58 amino acid residues at the C-terminal half. The aberrantly spliced NT5C2 showed substantial reduction in expression level in the in-vitro study, indicating marked instability of the mutant NT5C2 protein. CONCLUSION: The present report expands the phenotypic spectrum of SPG45 and confirms NT5C2-SPG45 as a member of the rare TCC SPG-subtypes. Homozygous alteration in NT5C2 seems essential to produce central white matter developmental defects. The study highlights the importance of cytosolic II 5'-nucleotidase (NT5C2) in maintaining the normal balance of purines' pool in the brain, which seems to play a pivotal role in the normal development of central white matter structures.


Asunto(s)
5'-Nucleotidasa/genética , Fenotipo , Paraplejía Espástica Hereditaria/genética , 5'-Nucleotidasa/metabolismo , Niño , Preescolar , Cuerpo Calloso/diagnóstico por imagen , ADN/química , ADN/aislamiento & purificación , ADN/metabolismo , Expresión Génica , Células HEK293 , Homocigoto , Humanos , Imagen por Resonancia Magnética , Masculino , Linaje , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Qatar , Sitios de Empalme de ARN , Análisis de Secuencia de ADN , Paraplejía Espástica Hereditaria/diagnóstico
4.
Ann Neurol ; 81(1): 68-78, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27863452

RESUMEN

OBJECTIVE: Exome sequences account for only 2% of the genome and may overlook mutations causing disease. To obtain a more complete view, whole genome sequencing (WGS) was analyzed in a large consanguineous family in which members displayed autosomal recessively inherited cerebellar ataxia manifesting before 2 years of age. METHODS: WGS from blood-derived genomic DNA was used for homozygosity mapping and a rare variant search. RNA from isolated blood leukocytes was used for quantitative polymerase chain reaction (PCR), RNA sequencing, and comparison of the transcriptomes of affected and unaffected family members. RESULTS: WGS revealed a point mutation in noncoding RNA RNU12 that was associated with early onset cerebellar ataxia. The U12-dependent minor spliceosome edits 879 known transcripts. Reverse transcriptase PCR demonstrated minor intron retention in all of 9 randomly selected RNAs from this group, and RNAseq showed splicing disruption specific to all U12-type introns detected in blood monocytes from affected individuals. Moreover, 144 minor intron-containing RNAs were differentially expressed, including transcripts for 3 genes previously associated with cerebellar neurodegeneration. INTERPRETATION: Interference with particular spliceosome components, including small nuclear RNAs, cause reproducible uniquely distributed phenotypic and transcript-specific effects, making this an important category of disease-associated mutation. Our approach to differential expression analysis of minor intron-containing genes is applicable to other diseases involving altered transcriptome processing. ANN NEUROL 2017;81:68-78.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , ARN Nuclear Pequeño/genética , ARN no Traducido/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Mutación Puntual , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ARN , Adulto Joven
5.
BMC Res Notes ; 7: 747, 2014 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-25339461

RESUMEN

BACKGROUND: With diminishing costs of next generation sequencing (NGS), whole genome analysis becomes a standard tool for identifying genetic causes of inherited diseases. Commercial NGS service providers in general not only provide raw genomic reads, but further deliver SNP calls to their clients. However, the question for the user arises whether to use the SNP data as is, or process the raw sequencing data further through more sophisticated SNP calling pipelines with more advanced algorithms. RESULTS: Here we report a detailed comparison of SNPs called using the popular GATK multiple-sample calling protocol to SNPs delivered as part of a 40x whole genome sequencing project by Illumina Inc of 171 human genomes of Arab descent (108 unrelated Qatari genomes, 19 trios, and 2 families with rare diseases) and compare them to variants provided by the Illumina CASAVA pipeline. GATK multi-sample calling identifies more variants than the CASAVA pipeline. The additional variants from GATK are robust for Mendelian consistencies but weak in terms of statistical parameters such as TsTv ratio. However, these additional variants do not make a difference in detecting the causative variants in the studied phenotype. CONCLUSION: Both pipelines, GATK multi-sample calling and Illumina CASAVA single sample calling, have highly similar performance in SNP calling at the level of putatively causative variants.


Asunto(s)
Algoritmos , Árabes/genética , Diabetes Mellitus/genética , Genoma Humano , Estudio de Asociación del Genoma Completo/métodos , Herencia , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Obesidad/genética , Polimorfismo de Nucleótido Simple , Enfermedades Raras/genética , Bases de Datos Genéticas , Diabetes Mellitus/etnología , Predisposición Genética a la Enfermedad , Humanos , Modelos Genéticos , Obesidad/etnología , Linaje , Fenotipo , Enfermedades Raras/etnología , Reproducibilidad de los Resultados , Programas Informáticos
7.
Nature ; 488(7409): 43-8, 2012 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-22722829

RESUMEN

Medulloblastoma is a malignant childhood brain tumour comprising four discrete subgroups. Here, to identify mutations that drive medulloblastoma, we sequenced the entire genomes of 37 tumours and matched normal blood. One-hundred and thirty-six genes harbouring somatic mutations in this discovery set were sequenced in an additional 56 medulloblastomas. Recurrent mutations were detected in 41 genes not yet implicated in medulloblastoma; several target distinct components of the epigenetic machinery in different disease subgroups, such as regulators of H3K27 and H3K4 trimethylation in subgroups 3 and 4 (for example, KDM6A and ZMYM3), and CTNNB1-associated chromatin re-modellers in WNT-subgroup tumours (for example, SMARCA4 and CREBBP). Modelling of mutations in mouse lower rhombic lip progenitors that generate WNT-subgroup tumours identified genes that maintain this cell lineage (DDX3X), as well as mutated genes that initiate (CDH1) or cooperate (PIK3CA) in tumorigenesis. These data provide important new insights into the pathogenesis of medulloblastoma subgroups and highlight targets for therapeutic development.


Asunto(s)
Neoplasias Cerebelosas/clasificación , Neoplasias Cerebelosas/genética , Meduloblastoma/clasificación , Meduloblastoma/genética , Mutación/genética , Animales , Antígenos CD , Proteína de Unión a CREB/genética , Cadherinas/genética , Proteínas Cdh1 , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Linaje de la Célula , Neoplasias Cerebelosas/patología , Niño , Fosfatidilinositol 3-Quinasa Clase I , ARN Helicasas DEAD-box/genética , Variaciones en el Número de Copia de ADN , ADN Helicasas/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Genoma Humano/genética , Genómica , Proteínas Hedgehog/metabolismo , Histona Demetilasas/genética , Histonas/metabolismo , Humanos , Meduloblastoma/patología , Metilación , Ratones , Proteínas Nucleares/genética , Fosfatidilinositol 3-Quinasas/genética , Factores de Transcripción/genética , Proteínas Wnt/metabolismo , beta Catenina/genética
8.
Genes Dev ; 23(14): 1619-24, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19605683

RESUMEN

Loss of PTEN causes unregulated activation of downstream components of phosphatidylinositol 3-kinase (PI3K) signaling, including PDK1, and disrupts normal nervous system development and homeostasis. We tested the contribution of Pdk1 to the abnormalities induced by Pten deletion in the brain. Conditional deletion of Pdk1 caused microcephaly. Combined deletion of Pdk1 and Pten rescued hypertrophy, but not migration defects of Pten-deficient neurons. Pdk1 inactivation induced strikingly different effects on the regulation of phosphorylated Akt in glia versus neurons. Our results show Pdk1-dependent and Pdk1-independent abnormalities in Pten-deficient brains, and demonstrate cell type specific differences in feedback regulation of the ubiquitous PI3K pathway.


Asunto(s)
Encéfalo/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Movimiento Celular , Núcleo Celular/metabolismo , Cerebelo/metabolismo , Fosfatidilinositol 3-Quinasa Clase I , Síndrome de Deleción Distal 11q de Jacobsen , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(6): 1880-5, 2009 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-19164512

RESUMEN

Inactivation of homologous recombination (HR) or nonhomologous end-joining (NHEJ) predisposes to a spectrum of tumor types. Here, we inactivated DNA double-strand break repair (DSBR) proteins, DNA Ligase IV (Lig4), Xrcc2, and Brca2, or combined Lig4/Xrcc2 during neural development using Nestin-cre. In all cases, inactivation of these repair factors, together with p53 loss, led to rapid medulloblastoma formation. Genomic analysis of these tumors showed recurring chromosome 13 alterations via chromosomal loss or translocations involving regions containing Ptch1. Sequence analysis of the remaining Ptch1 allele showed a variety of inactivating mutations in all tumors analyzed, highlighting the critical tumor suppressor function of this hedgehog-signaling regulator. We also observed genomic amplification or up-regulation of either N-Myc or cyclin D2 in all medulloblastomas. Additionally, chromosome 19, which contains Pten, was also selectively deleted in medulloblastoma arising after disruption of HR. Thus, our data highlight the preeminence of Ptch1 as a tumor suppressor in cerebellar granule cells and reveal other genomic events central to the genesis of medulloblastoma.


Asunto(s)
Trastornos por Deficiencias en la Reparación del ADN/etiología , Inestabilidad Genómica , Meduloblastoma/genética , Receptores de Superficie Celular/fisiología , Animales , Proteína BRCA2/genética , Aberraciones Cromosómicas , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP) , ADN Ligasas/genética , Reparación del ADN , Proteínas de Unión al ADN/genética , Meduloblastoma/etiología , Meduloblastoma/patología , Ratones , Ratones Noqueados , Receptores Patched , Receptor Patched-1 , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor
10.
Annu Rev Pathol ; 4: 127-50, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18767981

RESUMEN

PI3-kinase and PTEN are major positive and negative regulators, respectively, of the PI3-kinase pathway, which regulates growth, survival, and proliferation. These key signaling components are two of the most frequently mutated proteins in human cancers, resulting in unregulated activation of PI3K signaling and providing irrefutable genetic evidence of the central role of this pathway in tumorigenesis. PTEN regulates PI3K signaling by dephosphorylating the lipid signaling intermediate PIP(3), but PTEN may have additional phosphatase-independent activities, as well as other functions in the nucleus. In this review, we highlight current work showing cancer-relevant complexities in the regulation of PTEN and PI3K activity, potential novel functions for PTEN, and feedback regulation within the pathway. The significance and complexity of PI3K signaling make it an important but challenging therapeutic target for cancer.


Asunto(s)
Neoplasias/enzimología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales , Antineoplásicos/farmacología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Terapia Genética , Humanos , Ratones , Modelos Animales , Mutación , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
11.
Brain Res ; 1100(1): 32-41, 2006 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-16777079

RESUMEN

The tumor suppressor PTEN (phosphatase and tensin homolog) plays a critical role in the development and maintenance of the mammalian nervous system. Effects of inherited mutation of PTEN are highly variable and include macrocephaly, Lhermitte-Duclos disease (LDD) caused by a hamartomatous enlargement of the cerebellum, ataxia, seizures and autism, in addition to cancer predisposition. In the mouse, selective inactivation of Pten in post-mitotic granule neurons of the cerebellum and dentate gyrus showed that Pten was required for proper regulation of neuronal nuclear and soma size. Hypertrophy of Pten-deficient neurons required the activity of the serine-threonine kinase mTor. mTor is a master regulator of cell and organ growth which can trigger a cascade of downstream signaling pathways involving, in part, components of the translational machinery, including S6k1 and its substrate the ribosomal protein S6. Deletion of S6k1 in mice results in decreased size. Therefore, to determine the relative contribution of S6k1 to Pten-deficient neuronal hypertrophy in vivo, we crossed Pten brain-conditional knockouts with S6k1 null mice. Double mutant mice show no reversion or improvement in their Pten-related size and neurological defects including enlarged cerebella and dentate gyri with increased size of neuronal nuclei and somata, ataxia, and premature death. The hypertrophic Pten/S6k1-deficient neurons contained high levels of phosphorylated S6, similar to Pten-deficient neurons, suggesting that the mTor/S6k/S6 branch of the pathway was still active. Thus, we conclude that S6k1 is not required to cause hypertrophy of Pten-deficient neurons. This study reveals a cell type-dependent role for S6k1 in PI3K-dependent hypertrophy.


Asunto(s)
Neuronas/fisiología , Fosfohidrolasa PTEN/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Animales , Peso Corporal/genética , Encéfalo/anatomía & histología , Encéfalo/citología , Recuento de Células , Núcleo Celular/ultraestructura , Tamaño de la Célula , Cerebelo/citología , Cerebelo/ultraestructura , Giro Dentado/citología , Giro Dentado/ultraestructura , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Noqueados , Mutación/genética , Mutación/fisiología , Tamaño de los Órganos/genética , Fosfohidrolasa PTEN/deficiencia , Fosfohidrolasa PTEN/fisiología , Fenotipo , Proteínas Quinasas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/fisiología , Serina-Treonina Quinasas TOR
12.
J Bone Miner Res ; 19(7): 1194-9, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15177004

RESUMEN

Infantile malignant autosomal recessive osteopetrosis is a genetically heterogeneous disease caused by the inability of OCLs to resorb and remodel bone, resulting in generalized osteosclerosis and obliteration of marrow spaces and cranial foramina. The classical clinical features are pathological fractures, visual impairment, and bone marrow failure. Two human genes have been described as the cause of this form of osteopetrosis: the T-cell immune-regulator-1 (TCIRG1) gene, which is mutated in >50% of the patients, and the chloride channel 7 (ClCN7) gene, which accounts for approximately 10% of cases. We report the clinical, radiographic, and histopathologic findings of the first human osteopetrosis case caused by a mutation in the grey-lethal (GL) gene. The patient, a 9-day-old male infant, presented with a very severe osteopetrotic phenotype including substantial hepatosplenomegaly since birth, cytopenia, and progressive major liver failure. Skeletal radiographs revealed a generalized increase in bone density with loss of corticomedullary differentiation. Histopathologic bone examination showed the typical osteopetrotic changes, with absence of resorptive activity, and osteoclasts, slightly decreased in number, with evident morphological alterations.


Asunto(s)
Osteopetrosis/genética , Mutación Puntual/genética , Huesos/diagnóstico por imagen , Huesos/patología , Encéfalo/patología , Humanos , Recién Nacido , Hígado/patología , Masculino , Proteínas de la Membrana/genética , Osteopetrosis/diagnóstico por imagen , Osteopetrosis/patología , Radiografía , Ubiquitina-Proteína Ligasas
13.
Nat Med ; 9(4): 399-406, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12627228

RESUMEN

The spontaneous mouse grey-lethal (gl) mutation is responsible for a coat color defect and for the development of the most severe autosomal recessive form of osteopetrosis. Using a positional cloning approach, we have mapped and isolated the gl locus from a approximately 1.5 cM genetic interval. The gl locus was identified in a bacterial artificial chromosome (BAC) contig by functional genetic complementation in transgenic mice. Genomic sequence analysis revealed that the gl mutation is a deletion resulting in complete loss of function. The unique approximately 3 kb wild-type transcript is expressed primarily in osteoclasts and melanocytes as well as in brain, kidney, thymus and spleen. The gl gene is predicted to encode a 338-amino acid type I transmembrane protein that localizes to the intracellular compartment. Mutation in the human GL gene leads to severe recessive osteopetrosis. Our studies show that mouse Gl protein function is absolutely required for osteoclast and melanocyte maturation and function.


Asunto(s)
Genes Letales , Genes Recesivos , Proteínas de la Membrana/genética , Osteopetrosis/genética , Trastornos de la Pigmentación/genética , Secuencia de Aminoácidos , Animales , Cromosomas Artificiales Bacterianos , Eliminación de Gen , Humanos , Líquido Intracelular/metabolismo , Melanocitos/fisiología , Proteínas de la Membrana/química , Ratones , Datos de Secuencia Molecular , Osteoclastos/fisiología , Osteopetrosis/patología , Mapeo Físico de Cromosoma , Homología de Secuencia de Aminoácido
14.
Ann Genet ; 45(1): 39-44, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11934389

RESUMEN

We genotyped 19 neurofibromatosis type 1 (NF1) families from French Canadians of the Quebec population with four intragenic microsatellites (IVS26-2.3, IVS27AC28.4, IVS27AC33.1, and IVS38GT53.0). Linkage analysis of the four microsatellite markers among the 19 NF1 families indicates that the four microsatellites are strongly linked with NF1 disease (LOD = 2.76-3.64). The four markers are associated (P = 0-0.077) except marker pair IVS26-2.3/IVS27AC33.1 (P = 0.18 or 0.17). However, perhaps due to the high mutation rate of the NF1 gene, no founder effect for NF1 was detected in the Quebec French Canadians.


Asunto(s)
Efecto Fundador , Genes de Neurofibromatosis 1 , Desequilibrio de Ligamiento , Frecuencia de los Genes , Marcadores Genéticos , Haplotipos , Humanos , Repeticiones de Microsatélite , Quebec
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA