Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oral Dis ; 23(5): 551-558, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27259978

RESUMEN

Human hearing loss is a common neurosensory disorder about which many basic research and clinically relevant questions are unresolved. This review on hereditary deafness focuses on three examples considered at first glance to be uncomplicated, however, upon inspection, are enigmatic and ripe for future research efforts. The three examples of clinical and genetic complexities are drawn from studies of (i) Pendred syndrome/DFNB4 (PDS, OMIM 274600), (ii) Perrault syndrome (deafness and infertility) due to mutations of CLPP (PRTLS3, OMIM 614129), and (iii) the unexplained extensive clinical variability associated with TBC1D24 mutations. At present, it is unknown how different mutations of TBC1D24 cause non-syndromic deafness (DFNB86, OMIM 614617), epilepsy (OMIM 605021), epilepsy with deafness, or DOORS syndrome (OMIM 220500) that is characterized by deafness, onychodystrophy (alteration of toenail or fingernail morphology), osteodystrophy (defective development of bone), mental retardation, and seizures. A comprehensive understanding of the multifaceted roles of each gene associated with human deafness is expected to provide future opportunities for restoration as well as preservation of normal hearing.


Asunto(s)
Proteínas Portadoras/genética , Sordera/genética , Bocio Nodular/genética , Disgenesia Gonadal 46 XX/genética , Pérdida Auditiva Sensorineural/genética , Anomalías Craneofaciales/genética , Endopeptidasa Clp/genética , Proteínas Activadoras de GTPasa , Deformidades Congénitas de la Mano/genética , Humanos , Discapacidad Intelectual/genética , Proteínas de la Membrana , Proteínas de Transporte de Membrana/genética , Uñas Malformadas/genética , Proteínas del Tejido Nervioso , Transportadores de Sulfato
2.
Clin Genet ; 91(2): 328-332, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27629923

RESUMEN

Perrault syndrome (PS) is a genetically heterogeneous disorder characterized by primary ovarian insufficiency (POI) in females and sensorineural hearing loss in males and females. In many PS subjects, causative variants have not been found in the five reported PS genes. The objective of this study was to identify the genetic cause of PS in an extended consanguineous family with six deaf individuals. Whole exome sequencing (WES) was completed on four affected members of a large family, and variants and co-segregation was confirmed by Sanger sequencing. All hearing impaired individuals, including the proband, are homozygous for a pathogenic variant of CLDN14, but this only explains the deafness. The PS proband is also homozygous for a frameshift variant (c.1453_1454delGA, p.(Glu485Lysfs*5)) in exon 7 of SGO2 encoding shugoshin 2, which is the likely cause of her concurrent ovarian insufficiency. In mouse, Sgol2a encoding shugoshin-like 2a is necessary during meiosis in both sexes to maintain the integrity of the cohesin complex that tethers sister chromatids. Human SGO2 has not previously been implicated in any disorder, but in this case of POI and perhaps others, it is a candidate for unexplained infertility.


Asunto(s)
Proteínas de Ciclo Celular/genética , Claudinas/genética , Disgenesia Gonadal 46 XX/genética , Pérdida Auditiva Sensorineural/genética , Animales , Consanguinidad , Exoma/genética , Femenino , Disgenesia Gonadal 46 XX/patología , Pérdida Auditiva Sensorineural/patología , Homocigoto , Humanos , Masculino , Ratones , Mutación , Linaje
4.
Clin Genet ; 76(3): 270-5, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19650862

RESUMEN

Autosomal recessive nonsyndromic hearing impairment (ARNSHI) segregating in three unrelated, large consanguineous Pakistani families (PKDF528, PKDF859 and PKDF326) is linked to markers on chromosome 12q14.2-q15. This novel locus is designated DFNB74. Maximum two-point limit of detection (LOD) scores of 5.6, 5.7 and 2.6 were estimated for markers D12S313,D12S83 and D12S75 at theta = 0 for recessive deafness segregating in these three families. Haplotype analyses identified a critical linkage interval of 5.35 cM (5.36 Mb) defined by D12S329 at 74.58 cM and D12S313 at 79.93 cM. DFNB74 is the second ARNSHI locus mapped to chromosome 12, but the physical intervals do not overlap with one another. A locus contributing to the early onset, rapidly progressing hearing loss of A/J mice (ahl4, age-related hearing loss 4) was reported to map to chromosome 10 in a region of conserved synteny to DFNB74, suggesting that ahl4 and DFNB74 may be due to mutations of the same gene in these two species.


Asunto(s)
Cromosomas Humanos Par 12/genética , Genes Recesivos , Sitios Genéticos , Pérdida Auditiva/genética , Audiometría de Tonos Puros , Segregación Cromosómica/genética , Familia , Femenino , Humanos , Escala de Lod , Masculino , Linaje
5.
Clin Genet ; 75(3): 237-43, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19250381

RESUMEN

Mutations in OTOF, encoding otoferlin, cause non-syndromic recessive hearing loss. The goal of our study was to define the identities and frequencies of OTOF mutations in a model population. We screened a cohort of 557 large consanguineous Pakistani families segregating recessive, severe-to-profound, prelingual-onset deafness for linkage to DFNB9. There were 13 families segregating deafness consistent with linkage to markers for DFNB9. We analyzed the genomic nucleotide sequence of OTOF and detected probable pathogenic sequence variants among all 13 families. These include the previously reported nonsense mutation p.R708X and 10 novel variants: 3 nonsense mutations (p.R425X, p.W536X, and p.Y1603X), 1 frameshift (c.1103_1104delinsC), 1 single amino acid deletion (p.E766del) and 5 missense substitutions of conserved residues (p.L573R, p.A1090E, p.E1733K, p.R1856Q and p.R1939W). OTOF mutations thus account for deafness in 13 (2.3%) of 557 Pakistani families. This overall prevalence is similar, but the mutation spectrum is different from those for Western populations. In addition, we demonstrate the existence of an alternative splice isoform of OTOF expressed in the human cochlea. This isoform must be required for human hearing because it encodes a unique alternative C-terminus affected by some DFNB9 mutations.


Asunto(s)
Sordera/genética , Frecuencia de los Genes/genética , Proteínas de la Membrana/genética , Empalme Alternativo , Secuencia de Aminoácidos , Cóclea/metabolismo , Exones , Genes Recesivos , Variación Genética , Humanos , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Mutación , Pakistán , Linaje , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
6.
Clin Genet ; 75(1): 86-91, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18505454

RESUMEN

Usher syndrome (USH) is a hereditary disorder associated with sensorineural hearing impairment, progressive loss of vision attributable to retinitis pigmentosa (RP) and variable vestibular function. Three clinical types have been described with type I (USH1) being the most severe. To date, six USH1 loci have been reported. We ascertained two large Pakistani consanguineous families segregating profound hearing loss, vestibular dysfunction, and RP, the defining features of USH1. In these families, we excluded linkage of USH to the 11 known USH loci and subsequently performed a genome-wide linkage screen. We found a novel USH1 locus designated USH1H that mapped to chromosome 15q22-23 in a 4.92-cM interval. This locus overlaps the non-syndromic deafness locus DFNB48 raising the possibility that the two disorders may be caused by allelic mutations.


Asunto(s)
Cromosomas Humanos Par 15/genética , Síndromes de Usher/genética , Adolescente , Adulto , Anciano , Niño , Mapeo Cromosómico , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Mutación , Linaje , Adulto Joven
7.
Clin Genet ; 73(4): 367-72, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18279434

RESUMEN

We ascertained a large North American family, LMG2, segregating progressive, non-syndromic, sensorineural hearing loss. A genome-wide scan identified significant evidence for linkage (maximum logarithm of the odds (LOD) score = 4.67 at theta = 0 for D4S398) to markers in a 5.7-cM interval on chromosome 4q12-13.1. The DFNA27 interval spans 8.85 Mb and includes at least 61 predicted and 8 known genes. We sequenced eight genes and excluded them as candidates for the DFNA27 gene.


Asunto(s)
Cromosomas Humanos Par 4/genética , Pérdida Auditiva Sensorineural/genética , Adulto , Anciano , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Linaje
8.
Clin Genet ; 73(1): 50-4, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18028453

RESUMEN

We ascertained a large North American family, LMG309, with matrilineal transmission of non-syndromic, progressive sensorineural hearing loss (SNHL). There was no history of aminoglycoside exposure, and penetrance was complete. We sequenced the entire mitochondrial genome and identified the previously reported 7510T>C transition in the tRNA(Ser(UCN)) gene. The 7510T>C was homoplasmic in all affected members. The LMG309 mitochondrial sequence belongs to an unnamed subgroup of mitochondrial haplogroup H. We demonstrate that the previously reported Spanish family S258 carries 7510T>C on a different mitochondrial sub-haplogroup, H1. We did not detect 7510T>C among 79 Caucasian haplogroup H control samples, including 11 from sub-haplogroup H1 and one from the same sub-haplogroup as LMG309. Our results provide strong genetic evidence that 7510T>C is a pathogenic mutation that causes non-syndromic SNHL.


Asunto(s)
ADN Mitocondrial/genética , Haplotipos , Pérdida Auditiva Sensorineural/genética , Mutación Puntual , ARN de Transferencia de Serina/genética , Salud de la Familia , Genoma Mitocondrial , América del Norte , Linaje
9.
Clin Genet ; 72(6): 546-50, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17877751

RESUMEN

Non-syndromic deafness is genetically heterogeneous. We previously reported that mutations of transmembrane channel-like gene 1 (TMC1) cause non-syndromic recessive deafness at the DFNB7/B11 locus on chromosome 9q13-q21 in nine Pakistani families. The goal of this study was to define the identities, origins and frequencies of TMC1 mutations in an expanded cohort of 557 large Pakistani families segregating recessive deafness. We screened affected family members for homozygosity at short-tandem repeats flanking known autosomal recessive (DFNB) deafness loci, followed by TMC1 sequence analysis in families segregating deafness linked to DFNB7/B11. We identified 10 new families segregating DFNB7/B11 deafness and TMC1 mutations, including three novel alleles. Overall, 9 different TMC1 mutations account for deafness in 19 (3.4%) of the 557 Pakistani families. A single mutation, p.R34X, causes deafness in 10 (1.8%) of the families. Genotype analysis of p.R34X-linked markers indicates that it arose from a common founder. We also detected p.R34X among normal control samples of African-American and northern European origins, raising the possibility that p.R34X and other mutations of TMC1 are prevalent contributors to the genetic load of deafness across a variety of populations and continents.


Asunto(s)
Sordera/genética , Proteínas de la Membrana/genética , Mutación , Secuencia de Aminoácidos , Cromosomas Humanos Par 9/genética , Codón sin Sentido , Femenino , Frecuencia de los Genes , Genes Recesivos , Humanos , Masculino , Datos de Secuencia Molecular , Pakistán , Linaje , Homología de Secuencia de Aminoácido
10.
Clin Genet ; 71(2): 148-52, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17250663

RESUMEN

We ascertained a North American Caucasian family (LMG248) segregating autosomal dominant, non-syndromic, post-lingual, progressive sensorineural hearing loss. The hearing loss begins in the second decade of life and initially affects high frequencies. It progresses to profound deafness at all frequencies by the fourth or fifth decade. The phenotype co-segregates with short-tandem repeat markers flanking the TMC1 gene at the DFNA36 locus on chromosome 9q31-q21. The affected individuals carry a novel missense substitution, p.D572H (c.G1714C), of the TMC1 gene. This mutation is at the same nucleotide and amino acid position as the only other reported DFNA36 mutation, p.D572N (c.G1714A). Our observations implicate a critical function for amino acid-572 for wild-type TMC1 function or the pathogenesis of DFNA36 hearing loss. The slower progression of hearing loss associated with p.D572H, in comparison with that caused by p.D572N, may reflect a correlation of DFNA36 phenotype with TMC1 genotype.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Mutación Missense , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Secuencia de Bases , Niño , ADN/genética , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Homología de Secuencia de Aminoácido
11.
J Med Genet ; 43(8): 634-40, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16459341

RESUMEN

BACKGROUND: Approximately half the cases of prelingual hearing loss are caused by genetic factors. Identification of genes causing deafness is a crucial first step in understanding the normal function of these genes in the auditory system. Recently, a mutant allele of Tmhs was reported to be associated with deafness and circling behaviour in the hurry-scurry mouse. Tmhs encodes a predicted tetraspan protein of unknown function, which is expressed in inner ear hair cells. The human homologue of Tmhs is located on chromosome 6p. OBJECTIVE: To determine the cause of deafness in four consanguineous families segregating recessive deafness linked to markers on chromosome 6p21.1-p22.3 defining a novel DFNB locus. RESULTS: A novel locus for non-syndromic deafness DFNB67 was mapped in an interval of approximately 28.51 cM on human chromosome 6p21.1-p22.3. DNA sequence analysis of TMHS revealed a homozygous frameshift mutation (246delC) and a missense mutation (Y127C) in affected individuals of two families segregating non-syndromic deafness, one of which showed significant evidence of linkage to markers in the DFNB67 interval. The localisation of mTMHS in developing mouse inner ear hair cells was refined and found to be expressed briefly from E16.5 to P3. CONCLUSIONS: These findings establish the importance of TMHS for normal sound transduction in humans.


Asunto(s)
Genes Recesivos/genética , Pérdida Auditiva/genética , Patrón de Herencia , Proteínas de la Membrana/genética , Mutación Missense/genética , Alelos , Secuencia de Aminoácidos , Animales , Cromosomas Humanos Par 6/genética , Análisis Mutacional de ADN , Epitelio/metabolismo , Ligamiento Genético , Marcadores Genéticos , Células Ciliadas Auditivas Internas/citología , Células Ciliadas Auditivas Internas/metabolismo , Humanos , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Linaje , Mapeo Físico de Cromosoma , Alineación de Secuencia
12.
J Med Genet ; 41(8): 591-5, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15286153

RESUMEN

We mapped a human deafness locus DFNB36 to chromosome 1p36.3 in two consanguineous families segregating recessively inherited deafness and vestibular areflexia. This phenotype co-segregates with either of two frameshift mutations, 1988delAGAG and 2469delGTCA, in ESPN, which encodes a calcium-insensitive actin-bundling protein called espin. A recessive mutation of ESPN is known to cause hearing loss and vestibular dysfunction in the jerker mouse. Our results establish espin as an essential protein for hearing and vestibular function in humans. The abnormal vestibular phenotype associated with ESPN mutations will be a useful clinical marker for refining the differential diagnosis of non-syndromic deafness.


Asunto(s)
Sordera/genética , Mutación del Sistema de Lectura/genética , Genes Recesivos/genética , Proteínas de Microfilamentos/genética , Enfermedades Vestibulares/genética , Adolescente , Adulto , Secuencia de Aminoácidos/genética , Animales , Niño , Cromosomas Humanos Par 1/genética , Femenino , Humanos , Masculino , Ratones , Proteínas de Microfilamentos/fisiología , Datos de Secuencia Molecular , Linaje , Ratas , Alineación de Secuencia
16.
Am J Hum Genet ; 71(2): 262-75, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12075507

RESUMEN

Usher syndrome type I is characterized by congenital hearing loss, retinitis pigmentosa (RP), and variable vestibular areflexia. Usher syndrome type ID, one of seven Usher syndrome type I genetic localizations, have been mapped to a chromosomal interval that overlaps with a nonsyndromic-deafness localization, DFNB12. Mutations in CDH23, a gene that encodes a putative cell-adhesion protein with multiple cadherin-like domains, are responsible for both Usher syndrome and DFNB12 nonsyndromic deafness. Specific CDH23 mutational defects have been identified that differentiate these two phenotypes. Only missense mutations of CDH23 have been observed in families with nonsyndromic deafness, whereas nonsense, frameshift, splice-site, and missense mutations have been identified in families with Usher syndrome. In the present study, a panel of 69 probands with Usher syndrome and 38 probands with recessive nonsyndromic deafness were screened for the presence of mutations in the entire coding region of CDH23, by heteroduplex, single-strand conformation polymorphism, and direct sequence analyses. A total of 36 different CDH23 mutations were detected in 45 families; 33 of these mutations were novel, including 18 missense, 3 nonsense, 5 splicing defects, 5 microdeletions, and 2 insertions. A total of seven mutations were common to more than one family. Numerous exonic and intronic polymorphisms also were detected. Results of ophthalmologic examinations of the patients with nonsyndromic deafness have found asymptomatic RP-like manifestations, indicating that missense mutations may have a subtle effect in the retina. Furthermore, patients with mutations in CDH23 display a wide range of hearing loss and RP phenotypes, differing in severity, age at onset, type, and the presence or absence of vestibular areflexia.


Asunto(s)
Cadherinas/genética , Sordera/genética , Mutación , Retinitis Pigmentosa/genética , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Proteínas Relacionadas con las Cadherinas , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Heterogeneidad Genética , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Fenotipo , Alineación de Secuencia , Síndrome , Pruebas de Función Vestibular
17.
Hum Genet ; 109(5): 535-41, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11735029

RESUMEN

Mutations in myosin XVA are responsible for the shaker 2 ( sh2) phenotype in mice and nonsyndromic autosomal recessive profound hearing loss DFNB3 on chromosome 17p11.2. We have ascertained seven families with profound congenital hearing loss from Pakistan and India with evidence of linkage to DFNB3 at 17p11.2. We report three novel homozygous mutations in MYO15A segregating in three of these families. In addition, one hemizygous missense mutation of MYO15A was found in one of eight Smith-Magenis syndrome (del(17)p11.2) patients from North America who had moderately severe sensorineural hearing loss.


Asunto(s)
Anomalías Múltiples/genética , Consanguinidad , Sordera/genética , Mutación Missense , Miosinas/genética , Animales , Secuencia de Bases , Cromosomas Humanos Par 17 , Cartilla de ADN , Femenino , Ligamiento Genético , Homocigoto , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Linaje , Síndrome
18.
Am J Pathol ; 159(4): 1375-82, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11583965

RESUMEN

The myosin superfamily of molecular motor proteins includes conventional myosins and several classes of unconventional myosins. Recent studies have characterized the human and mouse unconventional myosin XVA, which has a role in the formation and/or maintenance of the unique actin-rich structures of inner ear sensory hair cells. Myosin XVA is also highly expressed in human anterior pituitary cells. In this study we examined the distribution of myosin XVA protein and mRNA in normal and neoplastic human pituitaries and other neuroendocrine cells and tumors. Myosin XVA was expressed in all types of normal anterior pituitary cells and pituitary tumors and in other neuroendocrine cells and tumors including those of the adrenal medulla, parathyroid, and pancreatic islets. Most nonneuroendocrine tissues examined including liver cells were negative for myosin XVA protein and mRNA, although the distal and proximal tubules of normal kidneys showed moderate immunoreactivity for myosin XVA. Ultrastructural immunohistochemistry localized myosin XVA in association with secretory granules of human anterior pituitary cells and human pituitary tumors. These data suggest that in neuroendocrine cells myosin XVA may have a role in secretory granule movement and/or secretion.


Asunto(s)
Neoplasias de las Glándulas Endocrinas/metabolismo , Miosinas/metabolismo , Neoplasias del Sistema Nervioso/metabolismo , Sistemas Neurosecretores/metabolismo , Hipófisis/metabolismo , Neoplasias de las Glándulas Endocrinas/ultraestructura , Humanos , Inmunohistoquímica , Hibridación in Situ , Microscopía Electrónica , Datos de Secuencia Molecular , Neoplasias del Sistema Nervioso/ultraestructura , Sistemas Neurosecretores/ultraestructura , Hipófisis/ultraestructura
19.
Ear Hear ; 22(4): 279-88, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11527035

RESUMEN

OBJECTIVE: The purpose of this research was to identify the gene responsible for a novel form of nonsyndromic, late-onset, bilateral, progressive, sensorineural hearing loss in a Michigan family of English descent. This report describes the audiologic aspects of the search. DESIGN: Fifty-eight members of the family served as subjects for the study. Family pedigree information was gathered from family interviews, family records, birth and death registration records and census data. Audiologic evaluation was used to describe the hearing loss (phenotype) and classify family members as affected or unaffected based on hearing status. These data then were used in a linkage analysis, a process in which the inheritance of a trait is compared with the inheritance of genetic markers and statistically significant associations are sought. RESULTS: The team mapped the hearing loss to the long arm of chromosome 17 at band 17q25. The pattern of inheritance is autosomal dominant. The search for the gene is continuing using a candidate gene approach. CONCLUSIONS: The hearing loss demonstrated by this mid-Michigan family is a novel form of nonsyndromic, genetic, late-onset, bilateral, progressive, sensorineural hearing loss. The locus of the gene, the 20th for autosomal dominant hearing loss, is at band 17q25 of chromosome 17.


Asunto(s)
Proteínas Portadoras/genética , Expresión Génica/genética , Pérdida Auditiva Sensorineural/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Audiometría de Tonos Puros , Umbral Auditivo/fisiología , Conducción Ósea/fisiología , Niño , Preescolar , Cromosomas Humanos Par 17/genética , Cóclea/fisiopatología , Femenino , Ligamiento Genético , Pérdida Auditiva Sensorineural/epidemiología , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Lactante , Masculino , Persona de Mediana Edad , Emisiones Otoacústicas Espontáneas/fisiología , Linaje , Mutación Puntual/genética
20.
Am J Hum Genet ; 69(1): 25-34, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11398101

RESUMEN

Human chromosome 10q21-22 harbors USH1F in a region of conserved synteny to mouse chromosome 10. This region of mouse chromosome 10 contains Pcdh15, encoding a protocadherin gene that is mutated in ames waltzer and causes deafness and vestibular dysfunction. Here we report two mutations of protocadherin 15 (PCDH15) found in two families segregating Usher syndrome type 1F. A Northern blot probed with the PCDH15 cytoplasmic domain showed expression in the retina, consistent with its pathogenetic role in the retinitis pigmentosa associated with USH1F.


Asunto(s)
Cadherinas/genética , Cromosomas Humanos Par 10/genética , Sordera/genética , Mutación/genética , Precursores de Proteínas/genética , Retinitis Pigmentosa/genética , Anciano , Alelos , Animales , Secuencia de Bases , Proteínas Relacionadas con las Cadherinas , Cadherinas/química , Análisis Mutacional de ADN , Femenino , Haplotipos , Humanos , Escala de Lod , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Pakistán , Linaje , Fenotipo , Mapeo Físico de Cromosoma , Precursores de Proteínas/química , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Mensajero/metabolismo , Retina/metabolismo , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA