Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 111(7): 1271-1281, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38843839

RESUMEN

There is mounting evidence of the value of clinical genome sequencing (cGS) in individuals with suspected rare genetic disease (RGD), but cGS performance and impact on clinical care in a diverse population drawn from both high-income countries (HICs) and low- and middle-income countries (LMICs) has not been investigated. The iHope program, a philanthropic cGS initiative, established a network of 24 clinical sites in eight countries through which it provided cGS to individuals with signs or symptoms of an RGD and constrained access to molecular testing. A total of 1,004 individuals (median age, 6.5 years; 53.5% male) with diverse ancestral backgrounds (51.8% non-majority European) were assessed from June 2016 to September 2021. The diagnostic yield of cGS was 41.4% (416/1,004), with individuals from LMIC sites 1.7 times more likely to receive a positive test result compared to HIC sites (LMIC 56.5% [195/345] vs. HIC 33.5% [221/659], OR 2.6, 95% CI 1.9-3.4, p < 0.0001). A change in diagnostic evaluation occurred in 76.9% (514/668) of individuals. Change of management, inclusive of specialty referrals, imaging and testing, therapeutic interventions, and palliative care, was reported in 41.4% (285/694) of individuals, which increased to 69.2% (480/694) when genetic counseling and avoidance of additional testing were also included. Individuals from LMIC sites were as likely as their HIC counterparts to experience a change in diagnostic evaluation (OR 6.1, 95% CI 1.1-∞, p = 0.05) and change of management (OR 0.9, 95% CI 0.5-1.3, p = 0.49). Increased access to genomic testing may support diagnostic equity and the reduction of global health care disparities.


Asunto(s)
Pruebas Genéticas , Enfermedades Raras , Secuenciación Completa del Genoma , Humanos , Masculino , Enfermedades Raras/genética , Enfermedades Raras/diagnóstico , Femenino , Niño , Pruebas Genéticas/métodos , Preescolar , Adolescente , Adulto , Lactante , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/diagnóstico
2.
Immunity ; 45(5): 1038-1051, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27836432

RESUMEN

Tissue regeneration requires inflammatory and reparatory activity of macrophages. Macrophages detect and eliminate the damaged tissue and subsequently promote regeneration. This dichotomy requires the switch of effector functions of macrophages coordinated with other cell types inside the injured tissue. The gene regulatory events supporting the sensory and effector functions of macrophages involved in tissue repair are not well understood. Here we show that the lipid activated transcription factor, PPARγ, is required for proper skeletal muscle regeneration, acting in repair macrophages. PPARγ controls the expression of the transforming growth factor-ß (TGF-ß) family member, GDF3, which in turn regulates the restoration of skeletal muscle integrity by promoting muscle progenitor cell fusion. This work establishes PPARγ as a required metabolic sensor and transcriptional regulator of repair macrophages. Moreover, this work also establishes GDF3 as a secreted extrinsic effector protein acting on myoblasts and serving as an exclusively macrophage-derived regeneration factor in tissue repair.


Asunto(s)
Factor 3 de Diferenciación de Crecimiento/metabolismo , Músculo Esquelético/fisiología , Mioblastos/metabolismo , PPAR gamma/metabolismo , Regeneración/fisiología , Animales , Western Blotting , Separación Celular , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/lesiones , Análisis de Secuencia por Matrices de Oligonucleótidos , Cicatrización de Heridas/fisiología
3.
Nature ; 550(7674): 119-123, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28953873

RESUMEN

Catecholamine-induced lipolysis, the first step in the generation of energy substrates by the hydrolysis of triglycerides, declines with age. The defect in the mobilization of free fatty acids in the elderly is accompanied by increased visceral adiposity, lower exercise capacity, failure to maintain core body temperature during cold stress, and reduced ability to survive starvation. Although catecholamine signalling in adipocytes is normal in the elderly, how lipolysis is impaired in ageing remains unknown. Here we show that adipose tissue macrophages regulate the age-related reduction in adipocyte lipolysis in mice by lowering the bioavailability of noradrenaline. Unexpectedly, unbiased whole-transcriptome analyses of adipose macrophages revealed that ageing upregulates genes that control catecholamine degradation in an NLRP3 inflammasome-dependent manner. Deletion of NLRP3 in ageing restored catecholamine-induced lipolysis by downregulating growth differentiation factor-3 (GDF3) and monoamine oxidase A (MAOA) that is known to degrade noradrenaline. Consistent with this, deletion of GDF3 in inflammasome-activated macrophages improved lipolysis by decreasing levels of MAOA and caspase-1. Furthermore, inhibition of MAOA reversed the age-related reduction in noradrenaline concentration in adipose tissue, and restored lipolysis with increased levels of the key lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone sensitive lipase (HSL). Our study reveals that targeting neuro-immunometabolic signalling between the sympathetic nervous system and macrophages may offer new approaches to mitigate chronic inflammation-induced metabolic impairment and functional decline.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Envejecimiento/metabolismo , Catecolaminas/metabolismo , Inflamasomas/metabolismo , Lipólisis , Macrófagos/metabolismo , Tejido Adiposo/citología , Tejido Adiposo/efectos de los fármacos , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Animales , Caspasa 1/metabolismo , Catecolaminas/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Factor 3 de Diferenciación de Crecimiento/deficiencia , Factor 3 de Diferenciación de Crecimiento/genética , Factor 3 de Diferenciación de Crecimiento/metabolismo , Lipasa/metabolismo , Lipólisis/efectos de los fármacos , Lipólisis/genética , Ratones , Monoaminooxidasa/metabolismo , Inhibidores de la Monoaminooxidasa/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Norepinefrina/metabolismo , Esterol Esterasa/metabolismo
4.
FASEB J ; 35(12): e22018, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34731499

RESUMEN

Adipose tissue is the primary site of energy storage, playing important roles in health. While adipose research largely focuses on obesity, fat also has other critical functions, producing adipocytokines and contributing to normal nutrient metabolism, which in turn play important roles in satiety and total energy homeostasis. SMAD2/3 proteins are downstream mediators of activin signaling, which regulate critical preadipocyte and mature adipocyte functions. Smad2 global knockout mice exhibit embryonic lethality, whereas global loss of Smad3 protects mice against diet-induced obesity. The direct contributions of Smad2 and Smad3 in adipose tissues, however, are unknown. Here, we sought to determine the primary effects of adipocyte-selective reduction of Smad2 or Smad3 on diet-induced adiposity using Smad2 or Smad3 "floxed" mice intercrossed with Adiponectin-Cre mice. Additionally, we examined visceral and subcutaneous preadipocyte differentiation efficiency in vitro. Almost all wild type subcutaneous preadipocytes differentiated into mature adipocytes. In contrast, visceral preadipocytes differentiated poorly. Exogenous activin A suppressed differentiation of preadipocytes from both depots. Smad2 conditional knockout (Smad2cKO) mice did not exhibit significant effects on weight gain, irrespective of diet, whereas Smad3 conditional knockout (Smad3cKO) male mice displayed a trend of reduced body weight on high-fat diet. On both diets, Smad3cKO mice displayed an adipose depot-selective phenotype, with a significant reduction in subcutaneous fat mass but not visceral fat mass. Our data suggest that Smad3 is an important contributor to the maintenance of subcutaneous white adipose tissue in a sex-selective fashion. These findings have implications for understanding SMAD-mediated, depot selective regulation of adipocyte growth and differentiation.


Asunto(s)
Adipogénesis , Tejido Adiposo Blanco/citología , Adiposidad , Grasa Intraabdominal/citología , Proteína Smad2/fisiología , Proteína smad3/fisiología , Grasa Subcutánea/citología , Activinas/genética , Activinas/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular , Dieta Alta en Grasa , Femenino , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Grasa Subcutánea/metabolismo
5.
Am J Hum Genet ; 102(5): 731-743, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29706352

RESUMEN

Large-scale, population-based genomic studies have provided a context for modern medical genetics. Among such studies, however, African populations have remained relatively underrepresented. The breadth of genetic diversity across the African continent argues for an exploration of local genomic context to facilitate burgeoning disease mapping studies in Africa. We sought to characterize genetic variation and to assess population substructure within a cohort of HIV-positive children from Botswana-a Southern African country that is regionally underrepresented in genomic databases. Using whole-exome sequencing data from 164 Batswana and comparisons with 150 similarly sequenced HIV-positive Ugandan children, we found that 13%-25% of variation observed among Batswana was not captured by public databases. Uncaptured variants were significantly enriched (p = 2.2 × 10-16) for coding variants with minor allele frequencies between 1% and 5% and included predicted-damaging non-synonymous variants. Among variants found in public databases, corresponding allele frequencies varied widely, with Botswana having significantly higher allele frequencies among rare (<1%) pathogenic and damaging variants. Batswana clustered with other Southern African populations, but distinctly from 1000 Genomes African populations, and had limited evidence for admixture with extra-continental ancestries. We also observed a surprising lack of genetic substructure in Botswana, despite multiple tribal ethnicities and language groups, alongside a higher degree of relatedness than purported founder populations from the 1000 Genomes project. Our observations reveal a complex, but distinct, ancestral history and genomic architecture among Batswana and suggest that disease mapping within similar Southern African populations will require a deeper repository of genetic variation and allelic dependencies than presently exists.


Asunto(s)
Población Negra/genética , Secuenciación del Exoma , Variación Genética , Botswana , Estudios de Cohortes , Pool de Genes , Genética de Población , Genoma Humano , Geografía , Humanos , Filogenia , Análisis de Componente Principal
6.
Am J Hum Genet ; 103(1): 154-162, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29961569

RESUMEN

TRAF7 is a multi-functional protein involved in diverse signaling pathways and cellular processes. The phenotypic consequence of germline TRAF7 variants remains unclear. Here we report missense variants in TRAF7 in seven unrelated individuals referred for clinical exome sequencing. The seven individuals share substantial phenotypic overlap, with developmental delay, congenital heart defects, limb and digital anomalies, and dysmorphic features emerging as key unifying features. The identified variants are de novo in six individuals and comprise four distinct missense changes, including a c.1964G>A (p.Arg655Gln) variant that is recurrent in four individuals. These variants affect evolutionarily conserved amino acids and are located in key functional domains. Gene-specific mutation rate analysis showed that the occurrence of the de novo variants in TRAF7 (p = 2.6 × 10-3) and the recurrent de novo c.1964G>A (p.Arg655Gln) variant (p = 1.9 × 10-8) in our exome cohort was unlikely to have occurred by chance. In vitro analyses of the observed TRAF7 mutations showed reduced ERK1/2 phosphorylation. Our findings suggest that missense mutations in TRAF7 are associated with a multisystem disorder and provide evidence of a role for TRAF7 in human development.


Asunto(s)
Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Mutación Missense/genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Adulto , Aminoácidos/genética , Niño , Preescolar , Exoma/genética , Femenino , Cardiopatías Congénitas/genética , Humanos , Lactante , Recién Nacido , Sistema de Señalización de MAP Quinasas/genética , Masculino , Anomalías Musculoesqueléticas/genética , Fenotipo
7.
Am J Hum Genet ; 101(4): 503-515, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28942966

RESUMEN

Bromodomain PHD finger transcription factor (BPTF) is the largest subunit of nucleosome remodeling factor (NURF), a member of the ISWI chromatin-remodeling complex. However, the clinical consequences of disruption of this complex remain largely uncharacterized. BPTF is required for anterior-posterior axis formation of the mouse embryo and was shown to promote posterior neuroectodermal fate by enhancing Smad2-activated wnt8 expression in zebrafish. Here, we report eight loss-of-function and two missense variants (eight de novo and two of unknown origin) in BPTF on 17q24.2. The BPTF variants were found in unrelated individuals aged between 2.1 and 13 years, who manifest variable degrees of developmental delay/intellectual disability (10/10), speech delay (10/10), postnatal microcephaly (7/9), and dysmorphic features (9/10). Using CRISPR-Cas9 genome editing of bptf in zebrafish to induce a loss of gene function, we observed a significant reduction in head size of F0 mutants compared to control larvae. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and phospho-histone H3 (PH3) staining to assess apoptosis and cell proliferation, respectively, showed a significant increase in cell death in F0 mutants compared to controls. Additionally, we observed a substantial increase of the ceratohyal angle of the craniofacial skeleton in bptf F0 mutants, indicating abnormal craniofacial patterning. Taken together, our data demonstrate the pathogenic role of BPTF haploinsufficiency in syndromic neurodevelopmental anomalies and extend the clinical spectrum of human disorders caused by ablation of chromatin remodeling complexes.


Asunto(s)
Anomalías Múltiples/genética , Antígenos Nucleares/genética , Anomalías Craneofaciales/genética , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia/genética , Trastornos del Desarrollo del Lenguaje/genética , Microcefalia/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética , Anomalías Múltiples/patología , Adolescente , Animales , Antígenos Nucleares/metabolismo , Sistemas CRISPR-Cas , Proliferación Celular , Células Cultivadas , Niño , Preescolar , Ensamble y Desensamble de Cromatina , Estudios de Cohortes , Anomalías Craneofaciales/patología , Femenino , Edición Génica , Haploinsuficiencia/fisiología , Humanos , Trastornos del Desarrollo del Lenguaje/patología , Larva/genética , Larva/crecimiento & desarrollo , Masculino , Microcefalia/patología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
8.
Genet Med ; 22(8): 1338-1347, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32424177

RESUMEN

PURPOSE: Genitopatellar syndrome and Say-Barber-Biesecker-Young-Simpson syndrome are caused by variants in the KAT6B gene and are part of a broad clinical spectrum called KAT6B disorders, whose variable expressivity is increasingly being recognized. METHODS: We herein present the phenotypes of 32 previously unreported individuals with a molecularly confirmed diagnosis of a KAT6B disorder, report 24 new pathogenic KAT6B variants, and review phenotypic information available on all published individuals with this condition. We also suggest a classification of clinical subtypes within the KAT6B disorder spectrum. RESULTS: We demonstrate that cerebral anomalies, optic nerve hypoplasia, neurobehavioral difficulties, and distal limb anomalies other than long thumbs and great toes, such as polydactyly, are more frequently observed than initially reported. Intestinal malrotation and its serious consequences can be present in affected individuals. Additionally, we identified four children with Pierre Robin sequence, four individuals who had increased nuchal translucency/cystic hygroma prenatally, and two fetuses with severe renal anomalies leading to renal failure. We also report an individual in which a pathogenic variant was inherited from a mildly affected parent. CONCLUSION: Our work provides a comprehensive review and expansion of the genotypic and phenotypic spectrum of KAT6B disorders that will assist clinicians in the assessment, counseling, and management of affected individuals.


Asunto(s)
Blefarofimosis , Discapacidad Intelectual , Blefarofimosis/genética , Exones , Histona Acetiltransferasas/genética , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Mutación
9.
Am J Med Genet A ; 182(1): 38-52, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31782611

RESUMEN

White-Sutton syndrome (WHSUS) is a recently-identified genetic disorder resulting from de novo heterozygous pathogenic variants in POGZ. Thus far, over 50 individuals have been reported worldwide, however phenotypic characterization and data regarding the natural history are still incomplete. Here we report the clinical features of 22 individuals with 21 unique loss of function POGZ variants. We observed a broad spectrum of intellectual disability and/or developmental delay with or without autism, and speech delay in all individuals. Other common problems included ocular abnormalities, hearing loss and gait abnormalities. A validated sleep disordered breathing questionnaire identified symptoms of obstructive sleep apnea in 4/12 (33%) individuals. A higher-than-expected proportion of cases also had gastrointestinal phenotypes, both functional and anatomical, as well as genitourinary anomalies. In line with previous publications, we observed an increased body mass index (BMI) z-score compared to the general population (mean 0.59, median 0.9; p 0.0253). Common facial features included microcephaly, broad forehead, midface hypoplasia, triangular mouth, broad nasal root and flat nasal bridge. Analysis of the Baylor Genetics clinical laboratory database revealed that POGZ variants were implicated in approximately 0.14% of cases who underwent clinical exome sequencing for neurological indications with or without involvement of other body systems. This study describes a greater allelic series and expands the phenotypic spectrum of this new syndromic form of intellectual disability and autism.


Asunto(s)
Trastorno Autístico/genética , Discapacidad Intelectual/genética , Trastornos del Desarrollo del Lenguaje/genética , Transposasas/genética , Adolescente , Adulto , Trastorno Autístico/patología , Niño , Preescolar , Exoma/genética , Femenino , Heterocigoto , Humanos , Lactante , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/patología , Trastornos del Desarrollo del Lenguaje/patología , Masculino , Microcefalia/genética , Microcefalia/patología , Persona de Mediana Edad , Mutación/genética , Fenotipo , Secuenciación del Exoma , Adulto Joven
10.
Am J Hum Genet ; 96(4): 555-64, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25799105

RESUMEN

We investigated complex genomic rearrangements (CGRs) consisting of triplication copy-number variants (CNVs) that were accompanied by extended regions of copy-number-neutral absence of heterozygosity (AOH) in subjects with multiple congenital abnormalities. Molecular analyses provided observational evidence that in humans, post-zygotically generated CGRs can lead to regional uniparental disomy (UPD) due to template switches between homologs versus sister chromatids by using microhomology to prime DNA replication-a prediction of the replicative repair model, MMBIR. Our findings suggest that replication-based mechanisms might underlie the formation of diverse types of genomic alterations (CGRs and AOH) implicated in constitutional disorders.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Reordenamiento Génico/genética , Pérdida de Heterocigocidad/genética , Modelos Genéticos , Disomía Uniparental/genética , Secuencia de Bases , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular , Países Bajos , Reacción en Cadena de la Polimerasa , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN
11.
Hum Mutat ; 38(10): 1365-1371, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28649782

RESUMEN

Pathogenic variants in genes encoding components of the BRG1-associated factor (BAF) chromatin remodeling complex have been associated with intellectual disability syndromes. We identified heterozygous, novel variants in ACTL6A, a gene encoding a component of the BAF complex, in three subjects with varying degrees of intellectual disability. Two subjects have missense variants affecting highly conserved amino acid residues within the actin-like domain. Missense mutations in the homologous region in yeast actin were previously reported to be dominant lethal and were associated with impaired binding of the human ACTL6A to ß-actin and BRG1. A third subject has a splicing variant that creates an in-frame deletion. Our findings suggest that the variants identified in our subjects may have a deleterious effect on the function of the protein by disturbing the integrity of the BAF complex. Thus, ACTL6A gene mutation analysis should be considered in patients with intellectual disability, learning disabilities, or developmental language disorder.


Asunto(s)
Actinas/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Mutación Missense/genética , Adolescente , Niño , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , Exoma , Cara , Femenino , Deformidades Congénitas de la Mano/fisiopatología , Heterocigoto , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Micrognatismo/genética , Micrognatismo/fisiopatología , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Unión Proteica , Factores de Transcripción/genética
12.
Am J Hum Genet ; 95(5): 565-78, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439725

RESUMEN

The 17p13.1 microdeletion syndrome is a recently described genomic disorder with a core clinical phenotype of intellectual disability, poor to absent speech, dysmorphic features, and a constellation of more variable clinical features, most prominently microcephaly. We identified five subjects with copy-number variants (CNVs) on 17p13.1 for whom we performed detailed clinical and molecular studies. Breakpoint mapping and retrospective analysis of published cases refined the smallest region of overlap (SRO) for microcephaly to a genomic interval containing nine genes. Dissection of this phenotype in zebrafish embryos revealed a complex genetic architecture: dosage perturbation of four genes (ASGR1, ACADVL, DVL2, and GABARAP) impeded neurodevelopment and decreased dosage of the same loci caused a reduced mitotic index in vitro. Moreover, epistatic analyses in vivo showed that dosage perturbations of discrete gene pairings induce microcephaly. Taken together, these studies support a model in which concomitant dosage perturbation of multiple genes within the CNV drive the microcephaly and possibly other neurodevelopmental phenotypes associated with rearrangements in the 17p13.1 SRO.


Asunto(s)
Anomalías Múltiples/genética , Dosificación de Gen/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis , Receptor de Asialoglicoproteína/genética , Secuencia de Bases , Línea Celular , Puntos de Rotura del Cromosoma , Deleción Cromosómica , Cromosomas Humanos Par 17/genética , Proteínas Dishevelled , Citometría de Flujo , Humanos , Inmunohistoquímica , Proteínas Asociadas a Microtúbulos/genética , Datos de Secuencia Molecular , Fosfoproteínas/genética , Estudios Retrospectivos , Análisis de Secuencia de ADN , Síndrome de Smith-Magenis , Síndrome , Pez Cebra
13.
Am J Hum Genet ; 95(5): 579-83, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25439098

RESUMEN

5q31.3 microdeletion syndrome is characterized by neonatal hypotonia, encephalopathy with or without epilepsy, and severe developmental delay, and the minimal critical deletion interval harbors three genes. We describe 11 individuals with clinical features of 5q31.3 microdeletion syndrome and de novo mutations in PURA, encoding transcriptional activator protein Pur-α, within the critical region. These data implicate causative PURA mutations responsible for the severe neurological phenotypes observed in this syndrome.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Proteínas de Unión al ADN/genética , Hipotonía Muscular/genética , Convulsiones/genética , Factores de Transcripción/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Caenorhabditis elegans/genética , Mapeo Cromosómico , Humanos , Datos de Secuencia Molecular , Mutación/genética , Análisis de Secuencia de ADN , Síndrome
14.
Development ; 141(12): 2414-28, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24917498

RESUMEN

In the embryonic CNS, development of myelin-forming oligodendrocytes is limited by bone morphogenetic proteins, which constitute one arm of the transforming growth factor-ß (Tgfß) family and signal canonically via Smads 1/5/8. Tgfß ligands and Activins comprise the other arm and signal via Smads 2/3, but their roles in oligodendrocyte development are incompletely characterized. Here, we report that Tgfß ligands and activin B (ActB) act in concert in the mammalian spinal cord to promote oligodendrocyte generation and myelination. In mouse neural tube, newly specified oligodendrocyte progenitors (OLPs) are first exposed to Tgfß ligands in isolation, then later in combination with ActB during maturation. In primary OLP cultures, Tgfß1 and ActB differentially activate canonical Smad3 and non-canonical MAP kinase signaling. Both ligands enhance viability, and Tgfß1 promotes proliferation while ActB supports maturation. Importantly, co-treatment strongly activates both signaling pathways, producing an additive effect on viability and enhancing both proliferation and differentiation such that mature oligodendrocyte numbers are substantially increased. Co-treatment promotes myelination in OLP-neuron co-cultures, and maturing oligodendrocytes in spinal cord white matter display strong Smad3 and MAP kinase activation. In spinal cords of ActB-deficient Inhbb(-/-) embryos, apoptosis in the oligodendrocyte lineage is increased and OLP numbers transiently reduced, but numbers, maturation and myelination recover during the first postnatal week. Smad3(-/-) mice display a more severe phenotype, including diminished viability and proliferation, persistently reduced mature and immature cell numbers, and delayed myelination. Collectively, these findings suggest that, in mammalian spinal cord, Tgfß ligands and ActB together support oligodendrocyte development and myelin formation.


Asunto(s)
Activinas/metabolismo , Sistema Nervioso Central/embriología , Regulación del Desarrollo de la Expresión Génica , Oligodendroglía/citología , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Adhesión Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Perfilación de la Expresión Génica , Humanos , Ligandos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Proteína smad3/genética , Médula Espinal/embriología
15.
Genet Med ; 19(7): 826-833, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28383545

RESUMEN

PURPOSE: The Collaborative African Genomics Network (CAfGEN) aims to establish sustainable genomics research programs in Botswana and Uganda through long-term training of PhD students from these countries at Baylor College of Medicine. Here, we present an overview of the CAfGEN PhD training program alongside trainees' perspectives on their involvement. BACKGROUND: Historically, collaborations between high-income countries (HICs) and low- and middle-income countries (LMICs), or North-South collaborations, have been criticized for the lack of a mutually beneficial distribution of resources and research findings, often undermining LMICs. CAfGEN plans to address this imbalance in the genomics field through a program of technology and expertise transfer to the participating LMICs. METHODS: An overview of the training program is presented. Trainees from the CAfGEN project summarized their experiences, looking specifically at the training model, benefits of the program, challenges encountered relating to the cultural transition, and program outcomes after the first 2 years. CONCLUSION: Collaborative training programs like CAfGEN will not only help establish sustainable long-term research initiatives in LMICs but also foster stronger North-South and South-South networks. The CAfGEN model offers a framework for the development of training programs aimed at genomics education for those for whom genomics is not their "first language." Genet Med advance online publication 06 April 2017.


Asunto(s)
Educación de Postgrado/métodos , Educación/métodos , Genómica/educación , Investigación Biomédica/educación , Investigación Biomédica/métodos , Botswana , Biología Computacional/educación , Curriculum , Femenino , Humanos , Cooperación Internacional , Masculino , Estudiantes , Uganda , Universidades
17.
Genet Med ; 17(10): 831-5, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25569436

RESUMEN

PURPOSE: We applied whole-genome sequencing (WGS) to children diagnosed with neoplasms and found to carry apparently balanced constitutional translocations to discover novel genic disruptions. METHODS: We applied the structural variation (SV) calling programs CREST, BreakDancer, SV-STAT, and CGAP-CNV, and we developed an annotative filtering strategy to achieve nucleotide resolution at the translocations. RESULTS: We identified the breakpoints for t(6;12)(p21.1;q24.31), disrupting HNF1A in a patient diagnosed with hepatic adenomas and maturity-onset diabetes of the young (MODY). Translocation as the disruptive event of HNF1A, a gene known to be involved in MODY3, has not been previously reported. In a subject with Hodgkin lymphoma and subsequent low-grade glioma, we identified t(5;18)(q35.1;q21.2), disrupting both SLIT3 and DCC, genes previously implicated in both glioma and lymphoma. CONCLUSION: These examples suggest that implementing clinical WGS in the diagnostic workup of patients with novel but apparently balanced translocations may reveal unanticipated disruption of disease-associated genes and aid in prediction of the clinical phenotype.


Asunto(s)
Estudios de Asociación Genética , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias/genética , Translocación Genética , Adenoma/diagnóstico , Adenoma/genética , Adulto , Factores de Edad , Secuencia de Bases , Niño , Preescolar , Puntos de Rotura del Cromosoma , Cromosomas Humanos Par 12 , Cromosomas Humanos Par 18 , Cromosomas Humanos Par 5 , Cromosomas Humanos Par 6 , Receptor DCC , Variaciones en el Número de Copia de ADN , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Femenino , Genómica , Humanos , Mutación INDEL , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Masculino , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , Neoplasias/diagnóstico , Polimorfismo de Nucleótido Simple , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/genética
18.
Brain Sci ; 14(2)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38391753

RESUMEN

Recently, a recurrent de novo dominant mutation in UBTF (c.628G>A, p.Glu210Lys; UBTF E210K) was identified as the cause of a neurological disorder which has been named UBTF Neuroregression Syndrome (UNS), or Childhood-Onset Neurodegeneration with Brain Atrophy (CONDBA). To date, only 17 cases have been reported worldwide. The molecular etiology is a pathogenic variant, E210K, within the HMG-box 2 of Upstream Binding Transcription Factor (UBTF). UBTF, a nucleolar protein, plays an important role in ribosomal RNA (rRNA) synthesis, nucleolar integrity, and cell survival. This variant causes unstable preinitiation complexes to form, resulting in altered rDNA chromatin structures, rRNA dysregulation, DNA damage, and ultimately, neurodegeneration. Defining clinical characteristics of the disorder include but are not limited to developmental regression beginning at approximately three years of age, progressive motor dysfunction, declining cognition, ambulatory loss, and behavioral problems. Histological and neuroimaging abnormalities include cortical atrophy, white matter deficits, and enlarged ventricles. Herein, we present a detailed overview of all published cases as well as the functional roles of UBTF to better understand the pathophysiology. Bringing undiagnosed cases to the attention of clinicians and researchers by making them aware of the clinical features will improve research and support the development of therapeutic interventions.

19.
Int J Infect Dis ; 139: 132-140, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38036259

RESUMEN

OBJECTIVES: We utilize a large retrospective study cohort derived from electronic medical records to estimate the prevalence of long-term non-progression (LTNP) and determine the factors associated with progression among children infected with HIV in Botswana and Uganda. METHODS: Electronic medical records from large tertiary HIV clinical centers in Botswana and Uganda were queried to identify LTNP children 0-18 years enrolled between June 2003 and May 2014 and extract demographic and nutritional parameters. Multivariate subdistribution hazard analyses were used to examine demographic factors and nutritional status in progression in the pre-antiretroviral therapy era. RESULTS: Between the two countries, 14,246 antiretroviral therapy-naïve children infected with HIV were enrolled into clinical care. The overall proportion of LTNP was 6.3% (9.5% in Botswana vs 5.9% in Uganda). The median progression-free survival for the cohort was 6.3 years, although this was lower in Botswana than in Uganda (6.6 vs 8.8 years; P <0.001). At baseline, the adjusted subdistribution hazard ratio (aHRsd) of progression was increased among underweight children (aHRsd 1.42; 95% confidence interval [CI]: 1.32-1.53), enrolled after 2010 (aHRsd 1.32; 95% CI 1.22-1.42), and those from Botswana (aHRsd 2; 95% CI 1.91-2.10). CONCLUSIONS: In our study, the prevalence of pediatric LTNP was lower than that observed among adult populations, but progression-free survival was higher than expected. Underweight, year of enrollment into care, and country of origin are independent predictors of progression among children.


Asunto(s)
Infecciones por VIH , Delgadez , Adulto , Humanos , Niño , Estudios Retrospectivos , Delgadez/complicaciones , Botswana/epidemiología , Uganda/epidemiología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología , Infecciones por VIH/complicaciones , Factores de Riesgo , Progresión de la Enfermedad
20.
Hum Mutat ; 34(10): 1415-23, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23878096

RESUMEN

We describe the molecular and clinical characterization of nine individuals with recurrent, 3.4-Mb, de novo deletions of 3q13.2-q13.31 detected by chromosomal microarray analysis. All individuals have hypotonia and language and motor delays; they variably express mild to moderate cognitive delays (8/9), abnormal behavior (7/9), and autism spectrum disorders (3/9). Common facial features include downslanting palpebral fissures with epicanthal folds, a slightly bulbous nose, and relative macrocephaly. Twenty-eight genes map to the deleted region, including four strong candidate genes, DRD3, ZBTB20, GAP43, and BOC, with important roles in neural and/or muscular development. Analysis of the breakpoint regions based on array data revealed directly oriented human endogenous retrovirus (HERV-H) elements of ~5 kb in size and of >95% DNA sequence identity flanking the deletion. Subsequent DNA sequencing revealed different deletion breakpoints and suggested nonallelic homologous recombination (NAHR) between HERV-H elements as a mechanism of deletion formation, analogous to HERV-I-flanked and NAHR-mediated AZFa deletions. We propose that similar HERV elements may also mediate other recurrent deletion and duplication events on a genome-wide scale. Observation of rare recurrent chromosomal events such as these deletions helps to further the understanding of mechanisms behind naturally occurring variation in the human genome and its contribution to genetic disease.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 3/genética , Trastornos del Conocimiento/genética , Discapacidades del Desarrollo/genética , Retrovirus Endógenos/genética , Hipotonía Muscular/genética , Adolescente , Adulto , Secuencia de Bases , Niño , Preescolar , Puntos de Rotura del Cromosoma , Trastornos del Conocimiento/diagnóstico , Hibridación Genómica Comparativa , Discapacidades del Desarrollo/diagnóstico , Facies , Femenino , Orden Génico , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Hipotonía Muscular/diagnóstico , Fenotipo , Alineación de Secuencia , Síndrome , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA