Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Am J Hum Genet ; 108(11): 2195-2204, 2021 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-34715011

RESUMEN

Human mitochondrial RNase P (mt-RNase P) is responsible for 5' end processing of mitochondrial precursor tRNAs, a vital step in mitochondrial RNA maturation, and is comprised of three protein subunits: TRMT10C, SDR5C1 (HSD10), and PRORP. Pathogenic variants in TRMT10C and SDR5C1 are associated with distinct recessive or x-linked infantile onset disorders, resulting from defects in mitochondrial RNA processing. We report four unrelated families with multisystem disease associated with bi-allelic variants in PRORP, the metallonuclease subunit of mt-RNase P. Affected individuals presented with variable phenotypes comprising sensorineural hearing loss, primary ovarian insufficiency, developmental delay, and brain white matter changes. Fibroblasts from affected individuals in two families demonstrated decreased steady state levels of PRORP, an accumulation of unprocessed mitochondrial transcripts, and decreased steady state levels of mitochondrial-encoded proteins, which were rescued by introduction of the wild-type PRORP cDNA. In mt-tRNA processing assays performed with recombinant mt-RNase P proteins, the disease-associated variants resulted in diminished mitochondrial tRNA processing. Identification of disease-causing variants in PRORP indicates that pathogenic variants in all three subunits of mt-RNase P can cause mitochondrial dysfunction, each with distinct pleiotropic clinical presentations.


Asunto(s)
Alelos , Pleiotropía Genética , Mitocondrias/enzimología , ARN Mitocondrial/genética , ARN de Transferencia/genética , Ribonucleasa P/genética , Adulto , Femenino , Humanos , Masculino , Linaje
2.
Am J Med Genet A ; 185(1): 15-25, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33029936

RESUMEN

Biallelic mutations in SNORD118, encoding the small nucleolar RNA U8, cause leukoencephalopathy with calcifications and cysts (LCC). Given the difficulty in interpreting the functional consequences of variants in nonprotein encoding genes, and the high allelic polymorphism across SNORD118 in controls, we set out to provide a description of the molecular pathology and clinical spectrum observed in a cohort of patients with LCC. We identified 64 affected individuals from 56 families. Age at presentation varied from 3 weeks to 67 years, with disease onset after age 40 years in eight patients. Ten patients had died. We recorded 44 distinct, likely pathogenic, variants in SNORD118. Fifty two of 56 probands were compound heterozygotes, with parental consanguinity reported in only three families. Forty nine of 56 probands were either heterozygous (46) or homozygous (three) for a mutation involving one of seven nucleotides that facilitate a novel intramolecular interaction between the 5' end and 3' extension of precursor-U8. There was no obvious genotype-phenotype correlation to explain the marked variability in age at onset. Complementing recently published functional analyses in a zebrafish model, these data suggest that LCC most often occurs due to combinatorial severe and milder mutations, with the latter mostly affecting 3' end processing of precursor-U8.


Asunto(s)
Calcinosis/genética , Estudios de Asociación Genética , Leucoencefalopatías/genética , ARN Nucleolar Pequeño/genética , Adolescente , Adulto , Anciano , Animales , Calcinosis/complicaciones , Calcinosis/patología , Niño , Preescolar , Consanguinidad , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Leucoencefalopatías/complicaciones , Leucoencefalopatías/patología , Masculino , Persona de Mediana Edad , Patología Molecular , Adulto Joven , Pez Cebra/genética
3.
Am J Hum Genet ; 106(5): 694-706, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32359472

RESUMEN

How mutations in the non-coding U8 snoRNA cause the neurological disorder leukoencephalopathy with calcifications and cysts (LCC) is poorly understood. Here, we report the generation of a mutant U8 animal model for interrogating LCC-associated pathology. Mutant U8 zebrafish exhibit defective central nervous system development, a disturbance of ribosomal RNA (rRNA) biogenesis and tp53 activation, which monitors ribosome biogenesis. Further, we demonstrate that fibroblasts from individuals with LCC are defective in rRNA processing. Human precursor-U8 (pre-U8) containing a 3' extension rescued mutant U8 zebrafish, and this result indicates conserved biological function. Analysis of LCC-associated U8 mutations in zebrafish revealed that one null and one functional allele contribute to LCC. We show that mutations in three nucleotides at the 5' end of pre-U8 alter the processing of the 3' extension, and we identify a previously unknown base-pairing interaction between the 5' end and the 3' extension of human pre-U8. Indeed, LCC-associated mutations in any one of seven nucleotides in the 5' end and 3' extension alter the processing of pre-U8, and these mutations are present on a single allele in almost all individuals with LCC identified to date. Given genetic data indicating that bi-allelic null U8 alleles are likely incompatible with human development, and that LCC is not caused by haploinsufficiency, the identification of hypomorphic misprocessing mutations that mediate viable embryogenesis furthers our understanding of LCC molecular pathology and cerebral vascular homeostasis.


Asunto(s)
Alelos , Calcinosis/genética , Quistes del Sistema Nervioso Central/genética , Quistes/genética , Leucoencefalopatías/genética , Mutación , ARN Nucleolar Pequeño/genética , Pez Cebra/genética , Animales , Secuencia de Bases , Calcinosis/patología , Quistes del Sistema Nervioso Central/patología , Secuencia Conservada , Modelos Animales de Enfermedad , Desarrollo Embrionario/genética , Humanos , Leucoencefalopatías/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/embriología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
4.
Ann Clin Transl Neurol ; 7(1): 144-152, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31912665

RESUMEN

Genetic white matter disorders have heterogeneous etiologies and overlapping clinical presentations. We performed a study of the diagnostic efficacy of genome sequencing in 41 unsolved cases with prior exome sequencing, resolving an additional 14 from an historical cohort (n = 191). Reanalysis in the context of novel disease-associated genes and improved variant curation and annotation resolved 64% of cases. The remaining diagnoses were directly attributable to genome sequencing, including cases with small and large copy number variants (CNVs) and variants in deep intronic and technically difficult regions. Genome sequencing, in combination with other methodologies, achieved a diagnostic yield of 85% in this retrospective cohort.


Asunto(s)
Leucoencefalopatías/diagnóstico , Leucoencefalopatías/genética , Sistema de Registros , Secuenciación Completa del Genoma , Adolescente , Niño , Preescolar , Femenino , Humanos , Leucoencefalopatías/patología , Masculino , Linaje
6.
J Allergy Clin Immunol ; 140(2): 543-552.e5, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28087229

RESUMEN

BACKGROUND: Gain-of-function mutations in transmembrane protein 173 (TMEM173) encoding stimulator of interferon genes (STING) underlie a recently described type I interferonopathy called STING-associated vasculopathy with onset in infancy (SAVI). OBJECTIVES: We sought to define the molecular and cellular pathology relating to 3 individuals variably exhibiting the core features of the SAVI phenotype including systemic inflammation, destructive skin lesions, and interstitial lung disease. METHODS: Genetic analysis, conformational studies, in vitro assays and ex vivo flow-cytometry were performed. RESULTS: Molecular and in vitro data demonstrate that the pathology in these patients is due to amino acid substitutions at positions 206, 281, and 284 of the human STING protein. These mutations confer cGAMP-independent constitutive activation of type I interferon signaling through TBK1 (TANK-binding kinase), independent from the alternative STING pathway triggered by membrane fusion of enveloped RNA viruses. This constitutive activation was abrogated by ex vivo treatment with the janus kinase 1/2 inhibitor ruxolitinib. CONCLUSIONS: Structural analysis indicates that the 3 disease-associated mutations at positions 206, 281, and 284 of the STING protein define a novel cluster of amino acids with functional importance in the regulation of type I interferon signaling.


Asunto(s)
Inflamación/genética , Interferón Tipo I/genética , Proteínas de la Membrana/genética , Adolescente , Adulto , Sustitución de Aminoácidos , Niño , Femenino , Células HEK293 , Humanos , Interferón Tipo I/metabolismo , Masculino , Mutación , Factor de Transcripción STAT1/metabolismo , Transducción de Señal
7.
Nat Genet ; 48(10): 1185-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27571260

RESUMEN

Although ribosomes are ubiquitous and essential for life, recent data indicate that monogenic causes of ribosomal dysfunction can confer a remarkable degree of specificity in terms of human disease phenotype. Box C/D small nucleolar RNAs (snoRNAs) are evolutionarily conserved non-protein-coding RNAs involved in ribosome biogenesis. Here we show that biallelic mutations in the gene SNORD118, encoding the box C/D snoRNA U8, cause the cerebral microangiopathy leukoencephalopathy with calcifications and cysts (LCC), presenting at any age from early childhood to late adulthood. These mutations affect U8 expression, processing and protein binding and thus implicate U8 as essential in cerebral vascular homeostasis.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales/genética , Leucoencefalopatías/genética , Mutación , ARN Nucleolar Pequeño/genética , Adolescente , Adulto , Calcinosis/genética , Calcinosis/patología , Línea Celular , Enfermedades de los Pequeños Vasos Cerebrales/patología , Niño , Preescolar , Cromosomas Humanos Par 17 , Estudios de Cohortes , Quistes/genética , Quistes/patología , Exoma , Femenino , Ligamiento Genético , Genoma Humano , Humanos , Lactante , Leucoencefalopatías/patología , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Adulto Joven
8.
Eur J Paediatr Neurol ; 20(4): 604-10, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27091087

RESUMEN

BACKGROUND: Cystic leukoencephalopathy without megalencephaly is a disorder related in some cases to RNASET2 mutations and characterized by bilateral anterior temporal subcortical cysts and multifocal lobar white matter lesions with sparing of central white matter structures. This phenotype significantly overlaps with the sequelae of in utero cytomegalovirus (CMV) infection, including the presence of intracranial calcification in some cases. Aicardi-Goutières syndrome (AGS) is another inherited leukodystrophy with cerebral calcification mimicking congenital infection. Clinical, radiological and biochemical criteria for the diagnosis of AGS have been established, although the breadth of phenotype associated with mutations in the AGS-related genes is much greater than previously envisaged. PATIENTS AND METHODS: We describe the clinical, biochemical and radiological findings of five patients demonstrating a phenotype reminiscent of AGS. RESULTS: All patients were found to carry biallelic mutations of RNASET2. CONCLUSIONS: Our patients illustrate the clinical and radiological overlap that can be seen between RNASET2-related leukodystrophy and AGS in some cases. Our data highlight the need to include both disorders in the same differential diagnosis, and hint at possible shared pathomechanisms related to auto-inflammation which are worthy of further investigation.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/fisiopatología , Encéfalo/fisiopatología , Quistes/fisiopatología , Leucoencefalopatías/fisiopatología , Malformaciones del Sistema Nervioso/fisiopatología , Adolescente , Enfermedades Autoinmunes del Sistema Nervioso/diagnóstico por imagen , Enfermedades Autoinmunes del Sistema Nervioso/genética , Encéfalo/diagnóstico por imagen , Calcinosis/diagnóstico por imagen , Niño , Preescolar , Quistes/diagnóstico por imagen , Quistes/genética , Infecciones por Citomegalovirus/congénito , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/genética , Imagen por Resonancia Magnética , Masculino , Mutación , Malformaciones del Sistema Nervioso/diagnóstico por imagen , Malformaciones del Sistema Nervioso/genética , Fenotipo , Ribonucleasas/genética , Tomografía Computarizada por Rayos X , Proteínas Supresoras de Tumor/genética , Adulto Joven
9.
Hum Mutat ; 36(5): 489-95, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25726928

RESUMEN

Primary familial brain calcification (PFBC) is a heterogeneous neuropsychiatric disorder, with affected individuals presenting a wide variety of motor and cognitive impairments, such as migraine, parkinsonism, psychosis, dementia, and mood swings. Calcifications are usually symmetrical, bilateral, and found predominantly in the basal ganglia, thalamus, and cerebellum. So far, variants in three genes have been linked to PFBC: SLC20A2, PDGFRB, and PDGFB. Variants in SLC20A2 are responsible for most cases identified so far and, therefore, the present review is a comprehensive worldwide summary of all reported variants to date. SLC20A2 encodes an inorganic phosphate transporter, PiT-2, widely expressed in various tissues, including brain, and is part of a major family of solute carrier membrane transporters. Fifty variants reported in 55 unrelated patients so far have been identified in families of diverse ethnicities and only few are recurrent. Various types of variants were detected (missense, nonsense, frameshift) including full or partial SLC20A2 deletions. The recently reported SLC20A2 knockout mouse will enhance our understanding of disease mechanism and allow for screening of therapeutic compounds. In the present review, we also discuss the implications of these recent exciting findings and consider the possibility of treatments based on manipulation of inorganic phosphate homeostasis.


Asunto(s)
Encefalopatías/genética , Encefalopatías/patología , Calcinosis/genética , Mutación , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/genética , Alelos , Sustitución de Aminoácidos , Encefalopatías/diagnóstico , Análisis Mutacional de ADN , Exones , Estudios de Asociación Genética , Variación Genética , Humanos , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo III/metabolismo
10.
J Immunol ; 194(6): 2819-25, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25672750

RESUMEN

In humans, loss of function mutations in the SAMHD1 (AGS5) gene cause a severe form of Aicardi-Goutières syndrome (AGS), an inherited inflammatory-mediated encephalopathy characterized by increased type I IFN activity and upregulation of IFN-stimulated genes (ISGs). In particular, SAMHD1-related AGS is associated with a distinctive cerebrovascular pathology that commonly leads to stroke. Although inflammatory responses are observed in immune cells cultured from Samhd1 null mouse models, these mice are physically healthy, specifically lacking a brain phenotype. We have investigated the use of zebrafish as an alternative system for generating a clinically relevant model of SAMHD1-related AGS. Using temporal gene knockdown of zebrafish samhd1, we observe hindbrain ventricular swelling and brain hemorrhage. Furthermore, loss of samhd1 or of another AGS-associated gene, adar, leads to a significant upregulation of innate immune-related genes and an increase in the number of cells expressing the zebrafish type I IFN ifnphi1. To our knowledge, this is the first example of an in vivo model of AGS that recapitulates features of both the innate immune and neurological characteristics of the disease. The phenotypes associated with loss of samhd1 and adar suggest a function of these genes in controlling innate immune processes conserved to zebrafish, thereby also contributing to our understanding of antiviral signaling in this model organism.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Técnicas de Silenciamiento del Gen , Interferón Tipo I/genética , Malformaciones del Sistema Nervioso/genética , Proteínas de Pez Cebra/genética , Ácido Anhídrido Hidrolasas/metabolismo , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Secuencia de Aminoácidos , Animales , Animales Modificados Genéticamente , Enfermedades Autoinmunes del Sistema Nervioso/embriología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Western Blotting , Ventrículos Cerebrales/anomalías , Ventrículos Cerebrales/metabolismo , Modelos Animales de Enfermedad , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunidad Innata/genética , Interferón Tipo I/metabolismo , Interferones/genética , Interferones/metabolismo , Hemorragias Intracraneales/embriología , Hemorragias Intracraneales/genética , Hemorragias Intracraneales/metabolismo , Microscopía Fluorescente , Datos de Secuencia Molecular , Malformaciones del Sistema Nervioso/embriología , Malformaciones del Sistema Nervioso/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rombencéfalo/anomalías , Rombencéfalo/metabolismo , Proteína 1 que Contiene Dominios SAM y HD , Homología de Secuencia de Aminoácido , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
11.
Nat Genet ; 46(5): 503-509, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24686847

RESUMEN

The type I interferon system is integral to human antiviral immunity. However, inappropriate stimulation or defective negative regulation of this system can lead to inflammatory disease. We sought to determine the molecular basis of genetically uncharacterized cases of the type I interferonopathy Aicardi-Goutières syndrome and of other undefined neurological and immunological phenotypes also demonstrating an upregulated type I interferon response. We found that heterozygous mutations in the cytosolic double-stranded RNA receptor gene IFIH1 (also called MDA5) cause a spectrum of neuroimmunological features consistently associated with an enhanced interferon state. Cellular and biochemical assays indicate that these mutations confer gain of function such that mutant IFIH1 binds RNA more avidly, leading to increased baseline and ligand-induced interferon signaling. Our results demonstrate that aberrant sensing of nucleic acids can cause immune upregulation.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/genética , ARN Helicasas DEAD-box/genética , Interferón Tipo I/inmunología , Modelos Moleculares , Mutación/genética , Malformaciones del Sistema Nervioso/genética , Fenotipo , Transducción de Señal/genética , Análisis de Varianza , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Secuencia de Bases , ARN Helicasas DEAD-box/química , Ensayo de Cambio de Movilidad Electroforética , Exoma/genética , Células HEK293 , Humanos , Helicasa Inducida por Interferón IFIH1 , Repeticiones de Microsatélite/genética , Datos de Secuencia Molecular , Malformaciones del Sistema Nervioso/inmunología , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Análisis Espectral
12.
J Med Genet ; 51(2): 76-82, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24262145

RESUMEN

BACKGROUND: We recently observed mutations in ADAR1 to cause a phenotype of bilateral striatal necrosis (BSN) in a child with the type I interferonopathy Aicardi-Goutières syndrome (AGS). We therefore decided to screen patients with apparently non-syndromic BSN for ADAR1 mutations, and for an upregulation of interferon-stimulated genes (ISGs). METHODS: We performed Sanger sequencing of ADAR1 in a series of patients with BSN presenting to us during our routine clinical practice. We then undertook detailed clinical and neuroradiological phenotyping in nine mutation-positive children. We also measured the expression of ISGs in peripheral blood from these patients, and in children with BSN who did not have ADAR1 mutations. RESULTS: Nine ADAR1 mutation-positive patients from seven families demonstrated an acute (five cases) or subacute (four cases) onset of refractory, four-limb dystonia starting between 8 months and 5 years of age. Eight patients were developmentally normal at initial presentation. In seven cases, the disease was inherited as an autosomal recessive trait, while two related patients were found to have a heterozygous (dominant) ADAR1 mutation. All seven mutation-positive patients assayed showed an upregulation of ISGs (median: 12.50, IQR: 6.43-36.36) compared to controls (median: 0.93, IQR: 0.57-1.30), a so-called interferon signature, present many years after disease onset. No interferon signature was present in four children with BSN negative for mutations in ADAR1 (median: 0.63, IQR: 0.47-1.10). CONCLUSIONS: ADAR1-related disease should be considered in the differential diagnosis of apparently non-syndromic BSN with severe dystonia of varying evolution. The finding of an interferon signature provides a useful screening test for the presence of ADAR1 mutations in this context, and may suggest novel treatment approaches.


Asunto(s)
Adenosina Desaminasa/genética , Interferón Tipo I/fisiología , Degeneración Estriatonigral/congénito , Estudios de Casos y Controles , Preescolar , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Lactante , Masculino , Técnicas de Diagnóstico Molecular , Mutación Missense , Proteínas de Unión al ARN , Degeneración Estriatonigral/enzimología , Degeneración Estriatonigral/genética
13.
Am J Hum Genet ; 92(4): 605-13, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23541340

RESUMEN

Perrault syndrome is a genetically and clinically heterogeneous autosomal-recessive condition characterized by sensorineural hearing loss and ovarian failure. By a combination of linkage analysis, homozygosity mapping, and exome sequencing in three families, we identified mutations in CLPP as the likely cause of this phenotype. In each family, affected individuals were homozygous for a different pathogenic CLPP allele: c.433A>C (p.Thr145Pro), c.440G>C (p.Cys147Ser), or an experimentally demonstrated splice-donor-site mutation, c.270+4A>G. CLPP, a component of a mitochondrial ATP-dependent proteolytic complex, is a highly conserved endopeptidase encoded by CLPP and forms an element of the evolutionarily ancient mitochondrial unfolded-protein response (UPR(mt)) stress signaling pathway. Crystal-structure modeling suggests that both substitutions would alter the structure of the CLPP barrel chamber that captures unfolded proteins and exposes them to proteolysis. Together with the previous identification of mutations in HARS2, encoding mitochondrial histidyl-tRNA synthetase, mutations in CLPP expose dysfunction of mitochondrial protein homeostasis as a cause of Perrault syndrome.


Asunto(s)
Proteasas ATP-Dependientes/genética , Endopeptidasa Clp/genética , Exoma/genética , Genes Recesivos , Disgenesia Gonadal 46 XX/etiología , Pérdida Auditiva Sensorineural/etiología , Mitocondrias/enzimología , Mutación/genética , Proteasas ATP-Dependientes/metabolismo , Adenosina Trifosfato/metabolismo , Adolescente , Adulto , Femenino , Homocigoto , Humanos , Hibridación in Situ , Masculino , Mitocondrias/genética , Linaje , Fenotipo , Adulto Joven
14.
Nat Genet ; 44(11): 1243-8, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23001123

RESUMEN

Adenosine deaminases acting on RNA (ADARs) catalyze the hydrolytic deamination of adenosine to inosine in double-stranded RNA (dsRNA) and thereby potentially alter the information content and structure of cellular RNAs. Notably, although the overwhelming majority of such editing events occur in transcripts derived from Alu repeat elements, the biological function of non-coding RNA editing remains uncertain. Here, we show that mutations in ADAR1 (also known as ADAR) cause the autoimmune disorder Aicardi-Goutières syndrome (AGS). As in Adar1-null mice, the human disease state is associated with upregulation of interferon-stimulated genes, indicating a possible role for ADAR1 as a suppressor of type I interferon signaling. Considering recent insights derived from the study of other AGS-related proteins, we speculate that ADAR1 may limit the cytoplasmic accumulation of the dsRNA generated from genomic repetitive elements.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Interferón Tipo I , Malformaciones del Sistema Nervioso/genética , ARN Bicatenario/metabolismo , Elementos Alu/genética , Animales , Exoma , Expresión Génica , Humanos , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Ratones , Mutación , Conformación Proteica , ARN Bicatenario/genética , Proteínas de Unión al ARN , Análisis de Secuencia de ADN , Transducción de Señal , Relación Estructura-Actividad
15.
Nat Genet ; 44(3): 338-42, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22267198

RESUMEN

Coats plus is a highly pleiotropic disorder particularly affecting the eye, brain, bone and gastrointestinal tract. Here, we show that Coats plus results from mutations in CTC1, encoding conserved telomere maintenance component 1, a member of the mammalian homolog of the yeast heterotrimeric CST telomeric capping complex. Consistent with the observation of shortened telomeres in an Arabidopsis CTC1 mutant and the phenotypic overlap of Coats plus with the telomeric maintenance disorders comprising dyskeratosis congenita, we observed shortened telomeres in three individuals with Coats plus and an increase in spontaneous γH2AX-positive cells in cell lines derived from two affected individuals. CTC1 is also a subunit of the α-accessory factor (AAF) complex, stimulating the activity of DNA polymerase-α primase, the only enzyme known to initiate DNA replication in eukaryotic cells. Thus, CTC1 may have a function in DNA metabolism that is necessary for but not specific to telomeric integrity.


Asunto(s)
Anomalías Múltiples/genética , Predisposición Genética a la Enfermedad/genética , Telangiectasia Retiniana/genética , Proteínas de Unión a Telómeros/genética , Telómero/patología , Secuencia de Bases , Citometría de Flujo , Histonas/metabolismo , Datos de Secuencia Molecular , Telangiectasia Retiniana/patología , Análisis de Secuencia de ADN/métodos
17.
Am J Med Genet A ; 155A(12): 2910-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22002932

RESUMEN

We present a newly recognized, likely autosomal recessive, pleiotropic disorder seen in four individuals (three siblings and their nephew) from a consanguineous family of Pakistani origin. The condition is characterized by hypogonadotropic hypogonadism, severe microcephaly, sensorineural deafness, moderate learning disability, and distinctive facial dysmorphic features. Autozygosity mapping using SNP array genotyping defined a single, large autozygous region of 13.1 Mb on chromosome 3p21 common to the affected individuals. The critical region contains 227 genes and initial sequence analysis of a functional candidate gene has not identified causative mutations.


Asunto(s)
Cromosomas Humanos Par 3 , Anomalías Craneofaciales/genética , Genes Recesivos , Pérdida Auditiva Sensorineural/genética , Hipogonadismo/genética , Microcefalia/genética , Adulto , Encéfalo/patología , Niño , Mapeo Cromosómico , Consanguinidad , Anomalías Craneofaciales/diagnóstico , Femenino , Pérdida Auditiva Sensorineural/diagnóstico , Homocigoto , Humanos , Hipogonadismo/diagnóstico , Imagen por Resonancia Magnética , Masculino , Microcefalia/diagnóstico , Linaje , Fenotipo , Polimorfismo de Nucleótido Simple , Síndrome , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...