Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Am J Med Genet A ; 191(6): 1593-1598, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36866832

RESUMEN

The Notch proteins play key roles in cell fate determination during development. Germline pathogenic variants in NOTCH1 predispose to a spectrum of cardiovascular malformations including Adams-Oliver syndrome and a wide variety of isolated complex and simple congenital heart defects. The intracellular C-terminus of the single-pass transmembrane receptor encoded by NOTCH1 contains a transcriptional activating domain (TAD) required for target gene activation and a PEST domain (a sequence rich in proline, glutamic acid, serine, and threonine), regulating protein stability and turnover. We present a patient with a novel variant encoding a truncated NOTCH1 protein without the TAD and PEST domain (NM_017617.4: c.[6626_6629del];[=], p.(Tyr2209CysfsTer38)) and extensive cardiovascular abnormalities consistent with a NOTCH1-mediated mechanism. This variant fails to promote transcription of target genes as assessed by luciferase reporter assay. Given the roles of the TAD and PEST domains in NOTCH1 function and regulation, we hypothesize that loss of both the TAD and the PEST domain results in a stable, loss-of-function protein that acts as an antimorph through competition with wild-type NOTCH1.


Asunto(s)
Displasia Ectodérmica , Deformidades Congénitas de las Extremidades , Dermatosis del Cuero Cabelludo , Humanos , Receptor Notch1/genética , Displasia Ectodérmica/genética , Dermatosis del Cuero Cabelludo/congénito , Deformidades Congénitas de las Extremidades/genética
2.
NPJ Genom Med ; 8(1): 4, 2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36765070

RESUMEN

Autophagy regulates the degradation of damaged organelles and protein aggregates, and is critical for neuronal development, homeostasis, and maintenance, yet few neurodevelopmental disorders have been associated with pathogenic variants in genes encoding autophagy-related proteins. We report three individuals from two unrelated families with a neurodevelopmental disorder characterized by speech and motor impairment, and similar facial characteristics. Rare, conserved, bi-allelic variants were identified in ATG4D, encoding one of four ATG4 cysteine proteases important for autophagosome biogenesis, a hallmark of autophagy. Autophagosome biogenesis and induction of autophagy were intact in cells from affected individuals. However, studies evaluating the predominant substrate of ATG4D, GABARAPL1, demonstrated that three of the four ATG4D patient variants functionally impair ATG4D activity. GABARAPL1 is cleaved or "primed" by ATG4D and an in vitro GABARAPL1 priming assay revealed decreased priming activity for three of the four ATG4D variants. Furthermore, a rescue experiment performed in an ATG4 tetra knockout cell line, in which all four ATG4 isoforms were knocked out by gene editing, showed decreased GABARAPL1 priming activity for the two ATG4D missense variants located in the cysteine protease domain required for priming, suggesting that these variants impair the function of ATG4D. The clinical, bioinformatic, and functional data suggest that bi-allelic loss-of-function variants in ATG4D contribute to the pathogenesis of this syndromic neurodevelopmental disorder.

3.
Am J Med Genet A ; 188(10): 3089-3095, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35946377

RESUMEN

Alternative use of short distance tandem sites such as NAGNn AG are a common mechanism of alternative splicing; however, single nucleotide variants are rarely reported as likely to generate or to disrupt tandem splice sites. We identify a pathogenic intron 5 STK11 variant (NM_000455.4:c.[735-6A>G];[=]) segregating with the mucocutaneous features but not the hamartomatous polyps of Peutz-Jeghers syndrome in two individuals. By RNAseq analysis of peripheral blood mRNA, this variant was shown to generate a novel and preferentially used tandem proximal splice acceptor (AAGTGAAG). The variant transcript (NM_000455.4:c.734_734 + 1insTGAAG), which encodes a frameshift (p.[Tyr246Glufs*43]) constituted 36%-43% of STK11 transcripts suggesting partial escape from nonsense mediated mRNA decay and translation of a truncated protein. A review of the ClinVar database identified other similar variants. We suggest that nucleotide changes creating or disrupting tandem alternative splice sites are a pertinent disease mechanism and require contextualization for clinical reporting. Additionally, we hypothesize that some pathogenic STK11 variants cause an attenuated phenotype.


Asunto(s)
Síndrome de Peutz-Jeghers , Quinasas de la Proteína-Quinasa Activada por el AMP , Empalme Alternativo , Codón sin Sentido , Humanos , Nucleótidos , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patología
4.
Am J Med Genet A ; 188(1): 332-335, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34558799

RESUMEN

Cerebral cavernous malformations (CCMs) of the central nervous system arise sporadically or secondary to genomic variation. Established genetic etiologies include deleterious variants in KRIT1 (CCM1), malcavernin (CCM2), and PDCD10 (CCM3). KRIT1-related disease has not been described in conjunction with lymphatic defects, although lymphatic defects with abnormal endothelial cell junctions have been observed in mice deficient in HEG1-KRIT1 signaling. We report a proband with CCMs, multiple chylous mesenteric cysts, and chylous ascites with leaky lymphatic vasculature. Clinical short-read exome sequencing detected a disease-associated KRIT1 variant (NM_194456.1:c.[1927C>T];[=], p.(Gln643*)). We postulate an expansion of KRIT1-related disease to include lymphatic malformations and lymphatic endothelial dysfunction.


Asunto(s)
Hemangioma Cavernoso del Sistema Nervioso Central , Linfocele , Quiste Mesentérico , Animales , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Humanos , Proteína KRIT1/genética , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Proto-Oncogénicas/genética , Transducción de Señal
5.
J Cell Mol Med ; 24(1): 418-430, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31730267

RESUMEN

Numerous genetic and epigenetic alterations cause functional changes in cell biology underlying cancer. These hallmark functional changes constitute potentially tissue-independent anticancer therapeutic targets. We hypothesized that RNA-Seq identifies gene expression changes that underly those hallmarks, and thereby defines relevant therapeutic targets. To test this hypothesis, we analysed the publicly available TCGA-TARGET-GTEx gene expression data set from the University of California Santa CruzToil recompute project using WGCNA to delineate co-correlated 'modules' from tumour gene expression profiles and functional enrichment of these modules to hierarchically cluster tumours. This stratified tumours according to T cell activation, NK-cell activation, complement cascade, ATM, Rb, angiogenic, MAPK, ECM receptor and histone modification signalling. These correspond to the cancer hallmarks of avoiding immune destruction, tumour-promoting inflammation, evading growth suppressors, inducing angiogenesis, sustained proliferative signalling, activating invasion and metastasis, and genome instability and mutation. This approach did not detect pathways corresponding to the cancer enabling replicative immortality, resisting cell death or deregulating cellular energetics hallmarks. We conclude that RNA-Seq stratifies tumours along some, but not all, hallmarks of cancer and, therefore, could be used in conjunction with other analyses collectively to inform precision therapy.


Asunto(s)
Neoplasias/genética , RNA-Seq , Regulación Neoplásica de la Expresión Génica , Humanos , Especificidad de Órganos/genética , Transducción de Señal/genética
6.
Am J Hum Genet ; 104(2): 213-228, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30639323

RESUMEN

Primary defects in lung branching morphogenesis, resulting in neonatal lethal pulmonary hypoplasias, are incompletely understood. To elucidate the pathogenetics of human lung development, we studied a unique collection of samples obtained from deceased individuals with clinically and histopathologically diagnosed interstitial neonatal lung disorders: acinar dysplasia (n = 14), congenital alveolar dysplasia (n = 2), and other lethal lung hypoplasias (n = 10). We identified rare heterozygous copy-number variant deletions or single-nucleotide variants (SNVs) involving TBX4 (n = 8 and n = 2, respectively) or FGF10 (n = 2 and n = 2, respectively) in 16/26 (61%) individuals. In addition to TBX4, the overlapping ∼2 Mb recurrent and nonrecurrent deletions at 17q23.1q23.2 identified in seven individuals with lung hypoplasia also remove a lung-specific enhancer region. Individuals with coding variants involving either TBX4 or FGF10 also harbored at least one non-coding SNV in the predicted lung-specific enhancer region, which was absent in 13 control individuals with the overlapping deletions but without any structural lung anomalies. The occurrence of rare coding variants involving TBX4 or FGF10 with the putative hypomorphic non-coding SNVs implies a complex compound inheritance of these pulmonary hypoplasias. Moreover, they support the importance of TBX4-FGF10-FGFR2 epithelial-mesenchymal signaling in human lung organogenesis and help to explain the histopathological continuum observed in these rare lethal developmental disorders of the lung.


Asunto(s)
Factor 10 de Crecimiento de Fibroblastos/genética , Enfermedades del Recién Nacido/genética , Enfermedades del Recién Nacido/mortalidad , Enfermedades Pulmonares/genética , Enfermedades Pulmonares/mortalidad , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Variaciones en el Número de Copia de ADN/genética , Femenino , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Edad Gestacional , Humanos , Recién Nacido , Enfermedades del Recién Nacido/metabolismo , Enfermedades del Recién Nacido/patología , Pulmón/embriología , Pulmón/crecimiento & desarrollo , Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Masculino , Herencia Materna , Organogénesis , Herencia Paterna , Linaje , Polimorfismo de Nucleótido Simple/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas de Dominio T Box/metabolismo
7.
Am J Med Genet A ; 173(12): 3172-3181, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28884888

RESUMEN

The forkhead box (FOX) transcription factors have roles in development, carcinogenesis, metabolism, and immunity. In humans FOXP1 mutations have been associated with language and speech defects, intellectual disability, autism spectrum disorder, facial dysmorphisms, and congenital anomalies of the kidney and urinary tract. In mice, Foxp1 plays critical roles in development of the spinal motor neurons, lymphocytes, cardiomyocytes, foregut, and skeleton. We hypothesized therefore that mutations of FOXP1 affect additional tissues in some humans. Supporting this hypothesis, we describe two individuals with novel variants of FOXP1 (NM_032682.5:c.975-2A>C and NM_032682.5:c.1574G>A) and additional features. One had a lung disease resembling neuroendocrine cell hyperplasia of infancy (NEHI), and the second had a skeletal disorder with undertubulation of the long bones and relapsing-remitting fevers associated with flushing and edema. Although attribution of these traits to mutation of FOXP1 requires ascertainment of additional patients, we hypothesize that the variable expression of these additional features might arise by means of stochastic developmental variation.


Asunto(s)
Trastorno del Espectro Autista/genética , Factores de Transcripción Forkhead/genética , Discapacidad Intelectual/genética , Trastornos del Lenguaje/genética , Enfermedades Pulmonares/genética , Proteínas Represoras/genética , Secuencia de Aminoácidos , Trastorno del Espectro Autista/diagnóstico por imagen , Femenino , Haploinsuficiencia , Humanos , Recién Nacido , Discapacidad Intelectual/diagnóstico por imagen , Trastornos del Lenguaje/diagnóstico por imagen , Pulmón/diagnóstico por imagen , Enfermedades Pulmonares/diagnóstico , Masculino , Modelos Moleculares , Mutación , Fenotipo , Dominios Proteicos , Alineación de Secuencia , Secuenciación del Exoma
8.
Orphanet J Rare Dis ; 11(1): 149, 2016 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-27816064

RESUMEN

BACKGROUND: Schimke immuno-osseous dysplasia (SIOD) is a multisystemic disorder caused by biallelic mutations in the SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily A-like 1 (SMARCAL1) gene. Changes in gene expression underlie the arteriosclerosis and T-cell immunodeficiency of SIOD; therefore, we hypothesized that SMARCAL1 deficiency causes the focal segmental glomerulosclerosis (FSGS) of SIOD by altering renal gene expression. We tested this hypothesis by gene expression analysis of an SIOD patient kidney and verified these findings through immunofluorescent analysis in additional SIOD patients and a genetic interaction analysis in Drosophila. RESULTS: We found increased expression of components and targets of the Wnt and Notch signaling pathways in the SIOD patient kidney, increased levels of unphosphorylated ß-catenin and Notch1 intracellular domain in the glomeruli of most SIOD patient kidneys, and genetic interaction between the Drosophila SMARCAL1 homologue Marcal1 and genes of the Wnt and Notch signaling pathways. CONCLUSIONS: We conclude that increased Wnt and Notch activity result from SMARCAL1 deficiency and, as established causes of FSGS, contribute to the renal disease of most SIOD patients. This further clarifies the pathogenesis of SIOD and will hopefully direct potential therapeutic approaches for SIOD patients.


Asunto(s)
Arteriosclerosis/metabolismo , Glomeruloesclerosis Focal y Segmentaria/metabolismo , Síndromes de Inmunodeficiencia/metabolismo , Enfermedades Renales/metabolismo , Síndrome Nefrótico/metabolismo , Osteocondrodisplasias/metabolismo , Embolia Pulmonar/metabolismo , Receptores Notch/metabolismo , Proteínas Wnt/metabolismo , Animales , Arteriosclerosis/genética , Niño , Preescolar , ADN Helicasas/genética , ADN Helicasas/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Técnica del Anticuerpo Fluorescente Indirecta , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Síndromes de Inmunodeficiencia/genética , Enfermedades Renales/genética , Masculino , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar/genética , Proteínas Wnt/genética
9.
Nucleus ; 7(6): 560-571, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27813696

RESUMEN

Mutations in SMARCAL1, which encodes a DNA annealing helicase with roles in DNA replication fork restart, DNA repair, and gene expression modulation, cause Schimke immuno-osseous dysplasia (SIOD), an autosomal recessive disease characterized by skeletal dysplasia, renal disease, T-cell immunodeficiency, and arteriosclerosis. The clinical features of SIOD arise from pathological changes in gene expression; however, the underlying mechanism for these gene expression alterations remains unclear. We hypothesized that changes of the epigenome alter gene expression in SIOD. To test this, we performed a genetic screen for interaction between Marcal1, the Drosophila melanogaster ortholog of SMARCAL1, and the genes of the trithorax group (trxG) and Polycomb group (PcG), which encode epigenetic regulators. SMARCAL1 and Marcal1 genetically interacted with trxG and PcG members. A homozygous null mutation of Marcal1 suppressed the wing-to-haltere transformation, ectopic Ultrabithorax (Ubx) expression, and ectopic Ubx minigene expression caused by PcG deficiency. The suppression of ectopic Ubx expression correlated with reduced chromatin accessibility of the Ubx promoter. To our knowledge, this is the first in vivo evidence for deficiency of a SMARCAL1 ortholog altering the chromatin structure of a gene.


Asunto(s)
Arteriosclerosis/genética , Arteriosclerosis/metabolismo , Cromatina/metabolismo , ADN Helicasas/deficiencia , Regulación de la Expresión Génica , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/metabolismo , Síndrome Nefrótico/genética , Síndrome Nefrótico/metabolismo , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Embolia Pulmonar/genética , Embolia Pulmonar/metabolismo , Animales , ADN Helicasas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas del Grupo Polycomb/metabolismo , Enfermedades de Inmunodeficiencia Primaria , Homología de Secuencia de Ácido Nucleico , Factores de Transcripción/metabolismo
10.
Proc Natl Acad Sci U S A ; 113(36): 9967-76, 2016 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-27551064

RESUMEN

Somatic copy number amplification and gene overexpression are common features of many cancers. To determine the role of gene overexpression on chromosome instability (CIN), we performed genome-wide screens in the budding yeast for yeast genes that cause CIN when overexpressed, a phenotype we refer to as dosage CIN (dCIN), and identified 245 dCIN genes. This catalog of genes reveals human orthologs known to be recurrently overexpressed and/or amplified in tumors. We show that two genes, TDP1, a tyrosyl-DNA-phosphdiesterase, and TAF12, an RNA polymerase II TATA-box binding factor, cause CIN when overexpressed in human cells. Rhabdomyosarcoma lines with elevated human Tdp1 levels also exhibit CIN that can be partially rescued by siRNA-mediated knockdown of TDP1 Overexpression of dCIN genes represents a genetic vulnerability that could be leveraged for selective killing of cancer cells through targeting of an unlinked synthetic dosage lethal (SDL) partner. Using SDL screens in yeast, we identified a set of genes that when deleted specifically kill cells with high levels of Tdp1. One gene was the histone deacetylase RPD3, for which there are known inhibitors. Both HT1080 cells overexpressing hTDP1 and rhabdomyosarcoma cells with elevated levels of hTdp1 were more sensitive to histone deacetylase inhibitors valproic acid (VPA) and trichostatin A (TSA), recapitulating the SDL interaction in human cells and suggesting VPA and TSA as potential therapeutic agents for tumors with elevated levels of hTdp1. The catalog of dCIN genes presented here provides a candidate list to identify genes that cause CIN when overexpressed in cancer, which can then be leveraged through SDL to selectively target tumors.


Asunto(s)
Inestabilidad Cromosómica/genética , Hidrolasas Diéster Fosfóricas/genética , Rabdomiosarcoma/genética , Proteínas de Saccharomyces cerevisiae/genética , Factores Asociados con la Proteína de Unión a TATA/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 2/genética , Histona Desacetilasas/genética , Humanos , Ácidos Hidroxámicos/administración & dosificación , Mutación , ARN Interferente Pequeño/genética , Rabdomiosarcoma/patología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Ácido Valproico/administración & dosificación
11.
Am J Med Genet A ; 170(9): 2440-4, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27374786

RESUMEN

Mutations in the T-box transcription factor TBX4 gene have been reported in patients with Ischiocoxopodopatellar syndrome (MIM# 147891) and childhood-onset pulmonary arterial hypertension. Whole exome sequencing of DNA from a 1 day old deceased newborn, with severe diffuse developmental lung disorder exhibiting features of acinar dysplasia, and her unaffected parents identified a de novo TBX4 missense mutation p.E86Q (c.256G>C) in the DNA-binding T-box domain. We propose phenotypic expansion of the TBX4-related clinical disease spectrum to include acinar dysplasia of the lungs. The reported mutation is the first identified genetic variant causative for acinar dysplasia. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Estudios de Asociación Genética , Pulmón/anomalías , Mutación , Fenotipo , Proteínas de Dominio T Box/genética , Alelos , Autopsia , Cromosomas Humanos Par 16 , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Resultado Fatal , Femenino , Genotipo , Heterocigoto , Humanos , Recién Nacido , Cariotipo , Pulmón/patología , Linaje , Radiografía Torácica
12.
Clin Immunol ; 161(2): 355-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26499378

RESUMEN

Schimke immuno-osseous dysplasia (SIOD) is an autosomal recessive, fatal childhood disorder associated with skeletal dysplasia, renal dysfunction, and T-cell immunodeficiency. This disease is linked to biallelic loss-of-function mutations of the SMARCAL1 gene. Although recurrent infection, due to T-cell deficiency, is a leading cause of morbidity and mortality, the etiology of the T-cell immunodeficiency is unclear. Here, we demonstrate that the T cells of SIOD patients have undetectable levels of protein and mRNA for the IL-7 receptor alpha chain (IL7Rα) and are unresponsive to stimulation with IL-7, indicating a loss of functional receptor. No pathogenic mutations were detected in the exons of IL7R in these patients; however, CpG sites in the IL7R promoter were hypermethylated in SIOD T cells. We propose therefore that the lack of IL7Rα expression, associated with hypermethylation of the IL7R promoter, in T cells and possibly their earlier progenitors, restricts T-cell development in SIOD patients.


Asunto(s)
Arteriosclerosis/genética , Síndromes de Inmunodeficiencia/genética , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Embolia Pulmonar/genética , Receptores de Interleucina-7/genética , Linfocitos T/metabolismo , Adolescente , Adulto , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Células Cultivadas , Niño , Preescolar , ADN Helicasas/genética , Metilación de ADN , Citometría de Flujo , Expresión Génica , Humanos , Inmunohistoquímica , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Interleucina-17/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Mutación , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Enfermedades de Inmunodeficiencia Primaria , Regiones Promotoras Genéticas/genética , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patología , Receptores de Interleucina-7/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Adulto Joven
13.
Pediatr Res ; 78(6): 609-17, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26309238

RESUMEN

BACKGROUND: Schimke immuno-osseous dysplasia (SIOD) is an autosomal recessive disorder caused by mutations in SMARCAL1. A frequent complication is arteriosclerosis associated with reduced elastin expression; however, the mechanism underlying the reduced elastin expression remains unknown. METHODS: Expression of transcriptional regulators of elastin (ELN) and microRNA (miRNA) regulators of ELN messenger RNA (mRNA), ELN promoter methylation, and ELN mRNA poly(A) tail length were assessed by quantitative RT-PCR, bisulfite Sanger sequencing, and the Poly(A) Tail Length Assay Kit, respectively, in unaffected developing human aortae and in an SIOD aorta. RESULTS: Comparing unaffected fetal and adult aortae, ELN precursor mRNA (pre-mRNA) levels remained nearly constant, whereas mRNA levels declined by ~10(2)-fold. This corresponded with a reduction in poly(A) tail length but not with changes in the other parameters. In contrast, compared to the unaffected fetal aortae, the SIOD aorta had 18-fold less ELN pre-mRNA and 10(4)-fold less mRNA. This corresponded with increased expression of miRNA regulators and shorter ELN mRNA poly(A) tail lengths but not with altered expression of ELN transcriptional regulators or ELN promoter methylation. CONCLUSION: Posttranscriptional mechanisms account for the reduction in ELN mRNA levels in unaffected aortae, whereas transcriptional and posttranscriptional mechanisms reduce elastin expression in SIOD aorta and predispose to arteriosclerosis.


Asunto(s)
Aorta/metabolismo , Arteriosclerosis/genética , Elastina/genética , Síndromes de Inmunodeficiencia/genética , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Embolia Pulmonar/genética , Precursores del ARN/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/genética , Transcripción Genética , Adolescente , Adulto , Aorta/embriología , Aorta/patología , Arteriosclerosis/embriología , Arteriosclerosis/metabolismo , Arteriosclerosis/patología , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Metilación de ADN , Regulación hacia Abajo , Elastina/metabolismo , Femenino , Edad Gestacional , Humanos , Síndromes de Inmunodeficiencia/embriología , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Síndrome Nefrótico/embriología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Osteocondrodisplasias/embriología , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Enfermedades de Inmunodeficiencia Primaria , Regiones Promotoras Genéticas , Embolia Pulmonar/embriología , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patología , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Orphanet J Rare Dis ; 10: 27, 2015 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-25888122

RESUMEN

BACKGROUND: Snyder-Robinson Syndrome (SRS) is an X-linked intellectual disability disorder also characterized by osteoporosis, scoliosis, and dysmorphic facial features. It is caused by mutations in SMS, a ubiquitously expressed gene encoding the polyamine biosynthetic enzyme spermine synthase. We hypothesized that the tissue specificity of SRS arises from differential sensitivity to spermidine toxicity or spermine deficiency. METHODS: We performed detailed clinical, endocrine, histopathologic, and morphometric studies on two affected brothers with a spermine synthase loss of function mutation (NM_004595.4:c.443A > G, p.Gln148Arg). We also measured spermine and spermidine levels in cultured human bone marrow stromal cells (hBMSCs) and fibroblasts using the Biochrom 30 polyamine protocol and assessed the osteogenic potential of hBMSCs. RESULTS: In addition to the known tissue-specific features of SRS, the propositi manifested retinal pigmentary changes, recurrent episodes of hyper- and hypoglycemia, nephrocalcinosis, renal cysts, and frequent respiratory infections. Bone histopathology and morphometry identified a profound depletion of osteoblasts and osteoclasts, absence of a trabecular meshwork, a low bone volume and a thin cortex. Comparison of cultured fibroblasts from affected and unaffected individuals showed relatively small changes in polyamine content, whereas comparison of cultured osteoblasts identified marked differences in spermidine and spermine content. Osteogenic differentiation of the SRS-derived hBMSCs identified a severe deficiency of calcium phosphate mineralization. CONCLUSIONS: Our findings support the hypothesis that cell specific alterations in polyamine metabolism contribute to the tissue specificity of SRS features, and that the low bone density arises from a failure of mineralization.


Asunto(s)
Discapacidad Intelectual Ligada al Cromosoma X/patología , Osteoblastos/patología , Osteoclastos/patología , Osteoporosis/patología , Fibroblastos/metabolismo , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mutación , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Espermidina/metabolismo , Espermina/metabolismo , Espermina Sintasa/genética , Espermina Sintasa/metabolismo
15.
Am J Med Genet A ; 167(6): 1374-80, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25845469

RESUMEN

Intellectual disability (ID) is a heterogeneous condition arising from a variety of environmental and genetic factors. Among these causes are defects in transcriptional regulators. Herein, we report on two brothers in a nonconsanguineous family with novel compound heterozygous, disease-segregating mutations (NM_015979.3: [3656A > G];[4006C > T], NP_057063.2: [H1219R];[R1336X]) in MED23. This gene encodes a subunit of the Mediator complex that modulates the expression of RNA polymerase II-dependent genes. These brothers, who had profound ID, spasticity, congenital heart disease, brain abnormalities, and atypical electroencephalography, represent the first case of MED23-associated ID in a non-consanguineous family. They also expand upon the clinical features previously reported for mutations in this gene.


Asunto(s)
Anomalías Múltiples/genética , Cardiopatías Congénitas/genética , Discapacidad Intelectual/genética , Complejo Mediador/genética , Mutación Missense , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Niño , Preescolar , Exoma , Expresión Génica , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/patología , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Hermanos
16.
Turk J Pediatr ; 57(5): 509-13, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27411420

RESUMEN

Schimke immuno-osseous dysplasia is an autosomal recessive multisystem disorder caused by defects in SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like 1 gene (SMARCAL1). SMARCAL1 product is a helicase that has role in selective cellular proliferation. The disorder is characterized by spondyloepiphyseal dysplasia with short stature, nephropathy, T cell deficiency, neurologic and cutaneous signs. Patients may have hyperpigmented skin lesions similar to café au lait spots. Symptoms and disease severity in Schimke immuno-osseous dysplasia varies from patient to patient. Genetic, epigenetic and environmental factors play role on the severity of the disease. Here we report on a patient with short stature, steroid resistant nephrotic syndrome and recurrent infections. Cutaneous findings and developmental delay helped us to reach the diagnosis of Schimke immuno-osseous dysplasia. A homozygous missense mutation in SMARCAL1 gene confirmed the clinical diagnosis.


Asunto(s)
Arteriosclerosis/diagnóstico , ADN Helicasas/genética , Síndromes de Inmunodeficiencia/diagnóstico , Síndrome Nefrótico/diagnóstico , Osteocondrodisplasias/diagnóstico , Embolia Pulmonar/diagnóstico , Piel/patología , Arteriosclerosis/genética , Preescolar , Homocigoto , Humanos , Síndromes de Inmunodeficiencia/genética , Masculino , Mutación Missense , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar/genética
17.
J Histochem Cytochem ; 63(1): 32-44, 2015 01.
Artículo en Inglés | MEDLINE | ID: mdl-25319549

RESUMEN

Schimke immuno-osseous dysplasia (SIOD) is a pleiotropic disorder caused by mutations in the SWI/SNF2-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a-like-1 (SMARCAL1) gene, with multiple clinical features, notably end-stage renal disease. Here we characterize the renal pathology in SIOD patients. Our analysis of SIOD patient renal biopsies demonstrates the tip and collapsing variants of focal segmental glomerulosclerosis (FSGS). Additionally, electron microscopy revealed numerous glomerular abnormalities most notably in the podocyte and Bowman's capsule. To better understand the role of SMARCAL1 in the pathogenesis of FSGS, we defined SMARCAL1 expression in the developing and mature kidney. In the developing fetal kidney, SMARCAL1 is expressed in the ureteric epithelium, stroma, metanephric mesenchyme, and in all stages of the developing nephron, including the maturing glomerulus. In postnatal kidneys, SMARCAL1 expression is localized to epithelial tubules of the nephron, collecting ducts, and glomerulus (podocytes and endothelial cells). Interestingly, not all cells within the same lineage expressed SMARCAL1. In renal biopsies from SIOD patients, TUNEL analysis detected marked increases in DNA fragmentation. Our results highlight the cells that may contribute to the renal pathogenesis in SIOD. Further, we suggest that disruptions in genomic integrity during fetal kidney development contribute to the pathogenesis of FSGS in SIOD patients.


Asunto(s)
Arteriosclerosis/metabolismo , Arteriosclerosis/patología , ADN Helicasas/metabolismo , Regulación de la Expresión Génica , Síndromes de Inmunodeficiencia/metabolismo , Síndromes de Inmunodeficiencia/patología , Riñón/metabolismo , Riñón/patología , Síndrome Nefrótico/metabolismo , Síndrome Nefrótico/patología , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Embolia Pulmonar/metabolismo , Embolia Pulmonar/patología , Animales , Arteriosclerosis/complicaciones , Arteriosclerosis/genética , Niño , Preescolar , Fragmentación del ADN , Femenino , Glomeruloesclerosis Focal y Segmentaria/complicaciones , Humanos , Síndromes de Inmunodeficiencia/complicaciones , Síndromes de Inmunodeficiencia/genética , Riñón/embriología , Riñón/ultraestructura , Masculino , Ratones , Síndrome Nefrótico/complicaciones , Síndrome Nefrótico/genética , Osteocondrodisplasias/complicaciones , Osteocondrodisplasias/genética , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar/complicaciones , Embolia Pulmonar/genética
18.
Am J Med Genet A ; 167A(1): 231-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25355368

RESUMEN

Chromosome 4q deletion syndrome is a rare intellectual disability disorder caused by a variety of non-recurrent deletions of 4q. We describe the evolution of the phenotypic features of a female patient with a previously unreported deletion of 4q12-4q21.21 (hg 18; 54,711,575-79,601,919). By review reported individuals with interstitial deletions extending telomeric from 4q12 have syndromic intellectual disability with variable piebaldism. We expand the phenotype to include dolichocephaly, pectus excavatum, hip dysplasia, pes planus, myopia, lens opacities, and an absence of spoken language but not of communication through sign. The proposita also did not have piebaldism suggesting again that piebaldism arises from a mechanism more complex than simple haploinsufficiency of KIT. Comparing deletions among affected individuals localizes the critical interval within 4q12-4q13.1, although the absence of molecular boundaries for nearly all reported cases precludes precise delineation and genotype-phenotype correlation.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 4/genética , Estudios de Asociación Genética , Piebaldismo/genética , Proteínas Proto-Oncogénicas c-kit/genética , Niño , Preescolar , Rotura Cromosómica , Femenino , Haploinsuficiencia , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo , Piebaldismo/diagnóstico por imagen , Radiografía
19.
J Biomol Screen ; 19(10): 1372-82, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25117203

RESUMEN

Mutations of DNA repair pathways contribute to tumorigenesis and provide a therapeutic target for synthetic lethal interactions in tumor cells. Given that tyrosyl-DNA phosphodiesterase 1 (Tdp1) repairs stalled topoisomerase-I DNA complexes, we hypothesized that inhibition of Tdp1 has synthetic lethal effects in some cancers. To test this, we screened tumor arrays for Tdp1 expression and observed that Tdp1 is expressed in many tumors, including more than 90% of human breast tumors. Subsequent chemical screening identified putative Tdp1 inhibitors. Treatment of control human mammary epithelial cells and the breast cancer cell line MCF-7 with compound CD00509 preferentially sensitized MCF-7 cells to camptothecin and decreased cell proliferation 25% more than camptothecin treatment alone. This suggests that CD00509 specifically targeted Tdp1 in vitro, and CD00509 increased the sensitivity of wild-type murine embryonic fibroblasts (MEFs) to camptothecin to a degree comparable to that of Tdp1(-/-) MEFs. In addition, consistent with poly ADP-ribose polymerase-1 (PARP-1) collaborating with Tdp1 in DNA repair, combined Tdp1 and PARP-1 inhibition was more detrimental to MCF-7 cells than either treatment alone, whereas the combination was not additively harmful to control mammary cells. We conclude that targeting Tdp1 in anticancer therapy preferentially enhances the sensitivity of some breast cancer cells to camptothecin and may be an effective adjuvant for breast cancer therapy.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento/métodos , Neoplasias/metabolismo , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Piranos/farmacología , Tiobarbitúricos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Simulación por Computador , Daño del ADN/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Histonas/metabolismo , Humanos , Técnicas In Vitro , Células MCF-7/efectos de los fármacos , Ratones , Simulación del Acoplamiento Molecular , Inhibidores de Fosfodiesterasa/química , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/genética , Análisis de Matrices Tisulares , Inhibidores de Topoisomerasa I/farmacología
20.
Am J Med Genet A ; 161A(10): 2609-13, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23950031

RESUMEN

Schimke immuno-osseous dysplasia (SIOD, OMIM 242900) is a rare autosomal recessive multisystem childhood disorder characterized by short stature, renal failure, T-cell immunodeficiency, and hypersensitivity to genotoxic agents. SIOD is associated with biallelic mutations in SMARCAL1 (SWI/SNF-related matrix-associated actin-dependent regulator of chromatin, subfamily a-like 1), which encodes a DNA stress response enzyme with annealing helicase activity. Two features of SIOD causing much morbidity and mortality are bone marrow failure and T-cell deficiency with the consequent opportunistic infections. To address the safety and efficacy of bone marrow transplantation (BMT) in SIOD, we reviewed the outcomes of the only five SIOD patients known to us in whom bone marrow or hematopoietic stem cell transplantation has been attempted. We find that only one patient survived the transplantation procedure and that the existing indicators of a good prognosis for bone marrow transplantation were not predictive in this small cohort. Given these observations, we also discuss some considerations for the poor outcomes.


Asunto(s)
Arteriosclerosis/terapia , Trasplante de Médula Ósea , Síndromes de Inmunodeficiencia/terapia , Síndrome Nefrótico/terapia , Osteocondrodisplasias/terapia , Embolia Pulmonar/terapia , Arteriosclerosis/genética , Trasplante de Médula Ósea/efectos adversos , Niño , Preescolar , ADN Helicasas/genética , Resultado Fatal , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Humanos , Síndromes de Inmunodeficiencia/genética , Masculino , Mutación , Síndrome Nefrótico/genética , Osteocondrodisplasias/genética , Enfermedades de Inmunodeficiencia Primaria , Embolia Pulmonar/genética , Trasplante Homólogo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA