Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Curr Opin Pediatr ; 35(6): 620-630, 2023 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-37791807

RESUMO

PURPOSE OF REVIEW: This review will focus on the current knowledge of the diagnosis and management of overgrowth syndromes with specific focus on mosaic conditions and treatment strategies. RECENT FINDINGS: With the implementation of massively parallel sequencing, the genetic etiology of many classically described overgrowth syndromes have been identified. More recently, the role of mosaic genetic changes has been well described in numerous syndromes. Furthermore, the role of imprinting and methylation, especially of the 11p15 region, has been shown to be instrumental for growth. Perhaps most importantly, many overgrowth syndromes carry an increased risk of neoplasm formation especially in the first 10 years of life and possibly beyond. The systematic approach to the child with overgrowth will aide in timely diagnosis and efficiently align them with appropriate screening strategies. In some cases, precision medical interventions are available to target the perturbed growth signaling pathways. SUMMARY: The systematic approach to the child with overgrowth aids in the standardization of the diagnostic pathway for these young patients, thereby expediting the diagnostic timeline, enabling rigorous monitoring, and delivering tailored therapeutic interventions.


Assuntos
Transdução de Sinais , Criança , Humanos , Síndrome
2.
Am J Med Genet A ; 179(12): 2517-2531, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31639285

RESUMO

The hedgehog (Hh) pathway is highly conserved and required for embryonic patterning and determination. Mutations in the Hh pathway are observed in sporadic tumors as well as under syndromic conditions. Common to these syndromes are the findings of polydactyly/syndactyly and brain overgrowth. The latter is also a finding most commonly observed in the cases of mutations in the PI3K/AKT/mTOR pathway. We have identified novel Hh pathway mutations and structural copy number variations in individuals with somatic overgrowth, macrocephaly, dysmorphic facial features, and developmental delay, which phenotypically closely resemble patients with phosphatase and tensin homolog (PTEN) mutations. We hypothesized that brain overgrowth and phenotypic overlap with syndromic overgrowth syndromes in these cases may be due to crosstalk between the Hh and PI3K/AKT/mTOR pathways. To test this, we modeled disease-associated variants by generating PTCH1 and Suppressor of Fused (SUFU) heterozygote cell lines using the CRISPR/Cas9 system. These cells demonstrate activation of PI3K signaling and increased phosphorylation of its downstream target p4EBP1 as well as a distinct cellular phenotype. To further investigate the mechanism underlying this crosstalk, we treated human neural stem cells with sonic hedgehog (SHH) ligand and performed transcriptional analysis of components of the mTOR pathway. These studies identified decreased expression of a set of mTOR negative regulators, leading to its activation. We conclude that there is a significant crosstalk between the SHH and PI3K/AKT/mTOR. We propose that this crosstalk is responsible for why mutations in PTCH1 and SUFU lead to macrocephaly phenotypes similar to those observed in PTEN hamartoma and other overgrowth syndromes associated with mutations in PI3K/AKT/mTOR pathway genes.


Assuntos
Proteínas Hedgehog/metabolismo , Megalencefalia/genética , Megalencefalia/metabolismo , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular , Pré-Escolar , Feminino , Deleção de Genes , Haploinsuficiência , Humanos , Lactente , Masculino , Megalencefalia/diagnóstico , Modelos Biológicos , Células-Tronco Neurais
3.
Am J Med Genet A ; 173(5): 1319-1327, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28296084

RESUMO

The cohesin complex is an evolutionarily conserved multi-subunit protein complex which regulates sister chromatid cohesion during mitosis and meiosis. Additionally, the cohesin complex regulates DNA replication, DNA repair, and transcription. The core of the complex consists of four subunits: SMC1A, SMC3, RAD21, and STAG1/2. Loss-of-function mutations in many of these proteins have been implicated in human developmental disorders collectively termed "cohesinopathies." Through clinical exome sequencing (CES) of an 8-year-old girl with a clinical history of global developmental delay, microcephaly, microtia with hearing loss, language delay, ADHD, and dysmorphic features, we describe a heterozygous de novo variant (c.205C>T; p.(Arg69*)) in the integral cohesin structural protein, STAG2. This variant is associated with decreased STAG2 protein expression. The analyses of metaphase spreads did not exhibit premature sister chromatid separation; however, delayed sister chromatid cohesion was observed. To further support the pathogenicity of STAG2 variants, we identified two additional female cases from the DECIPHER research database with mutations in STAG2 and phenotypes similar to our patient. Interestingly, the clinical features of these three cases are remarkably similar to those observed in other well-established cohesinopathies. Herein, we suggest that STAG2 is a dosage-sensitive gene and that heterozygous loss-of-function variants lead to a cohesinopathy.


Assuntos
Antígenos Nucleares/genética , Anormalidades Congênitas/genética , Deficiências do Desenvolvimento/genética , Microcefalia/genética , Antígenos Nucleares/biossíntese , Proteínas de Ciclo Celular/genética , Criança , Proteínas Cromossômicas não Histona/genética , Anormalidades Congênitas/fisiopatologia , Deficiências do Desenvolvimento/fisiopatologia , Feminino , Regulação da Expressão Gênica , Heterozigoto , Humanos , Microcefalia/fisiopatologia , Coesinas
4.
Cancers (Basel) ; 15(9)2023 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-37174013

RESUMO

Patients with Beckwith-Wiedemann syndrome (BWS), an epigenetic imprinting disorder involving alterations in genes at the 11p15 chromosomal location, are predisposed to develop hepatoblastomas (HBs), which are rare embryonal liver tumors. Tumors can develop after a BWS diagnosis or, conversely, can be the presenting feature leading to a subsequent diagnosis. While HBs are the cardinal tumors of BWS, not all patients with the BWS spectrum will develop HBs. This observation has led to many hypotheses, including genotype-associated risk, tissue mosaicism, and tumor-specific second hits. To explore these hypotheses, we present the largest cohort of patients with BWS and HBs to date. Our cohort comprised 16 cases, and we broadened our sample size by searching the literature for all cases of BWS with HBs. From these isolated case studies, we amassed another 34 cases, bringing the total number to 50 cases of BWS-HB. We observed that paternal uniparental isodisomy (upd(11)pat) was the most common genotype, representing 38% of cases. The next most common genotype was IC2 LOM, representing 14% of cases. Five patients had clinical BWS without a molecular diagnosis. To investigate the potential mechanism of HBs in BWS, we analyzed normal liver and HB samples from eight cases and isolated tumor samples from another two cases. These samples underwent methylation testing, and 90% of our tumor samples underwent targeted cancer next-generation sequencing (NGS) panels. These matched samples provided novel insights into the oncogenesis of HBs in BWS. We found that 100% of the HBs that underwent NGS panel testing had variants in the CTNNB1 gene. We further identified three distinct groups of BWS-HB patients based on epigenotype. We also demonstrated epigenotype mosaicism, where 11p15 alterations can differ between the blood, HB, and normal liver. In light of this epigenotype mosaicism, tumor risk assessment based on blood profiling may not be accurate. Therefore, universal screening is recommended for all patients with BWS.

6.
Artigo em Inglês | MEDLINE | ID: mdl-34697083

RESUMO

Beckwith-Wiedemann syndrome (BWS) is a rare overgrowth disorder caused by epigenetic alterations on Chromosome 11p15.5. Most molecular changes are sporadic and are thought to occur in a mosaic pattern. Thereby, the distribution of affected cells differs between tissues for each individual, which can complicate genotype-phenotype correlations. In two of the BWS molecular subtypes, tissue mosaicism has been demonstrated; however, mosaicism has not been specifically studied in the most common cause of BWS, loss of methylation (LOM) at KCNQ1OT1:TSS differentially methylated region (DMR) imprinting center 2 (IC2) LOM. The increased prevalence of twinning associated with the IC2 LOM subtype and the discordant phenotypes between the twins previously led to the proposal of diffused epigenetic mosaicism, leading to asymmetric distribution of affected cells during embryonic development. In this study, we evaluated the level of methylation detected in 64 samples collected from 30 individuals with IC2 LOM. We demonstrate that the IC2 LOM defect can occur in mosaic and nonmosaic patterns, and tissues from the same individual can show variable patterns, which suggests that this asymmetric distribution occurs during development. We further suggest that the clinical phenotype in individuals with BWS IC2 LOM is correlated with the epigenetic burden of affected cells in each tissue type. This series is the first report to demonstrate tissue mosaicism within the IC2 LOM epigenotype, and consideration of this mosaicism is necessary to understanding the pathogenesis of BWS.


Assuntos
Síndrome de Beckwith-Wiedemann , Síndrome de Beckwith-Wiedemann/genética , Metilação de DNA , Feminino , Impressão Genômica , Humanos , Mosaicismo , Fenótipo , Gravidez
7.
Sci Rep ; 11(1): 11295, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34050248

RESUMO

MBD5-associated neurodevelopmental disorder (MAND) is an autism spectrum disorder (ASD) characterized by intellectual disability, motor delay, speech impairment and behavioral problems; however, the biological role of methyl-CpG-binding domain 5, MBD5, in neurodevelopment and ASD remains largely undefined. Hence, we created neural progenitor cells (NPC) derived from individuals with chromosome 2q23.1 deletion and conducted RNA-seq to identify differentially expressed genes (DEGs) and the biological processes and pathways altered in MAND. Primary skin fibroblasts from three unrelated individuals with MAND and four unrelated controls were converted into induced pluripotent stem cell (iPSC) lines, followed by directed differentiation of iPSC to NPC. Transcriptome analysis of MAND NPC revealed 468 DEGs (q < 0.05), including 20 ASD-associated genes. Comparison of DEGs in MAND with SFARI syndromic autism genes revealed a striking significant overlap in biological processes commonly altered in neurodevelopmental phenotypes, with TGFß, Hippo signaling, DNA replication, and cell cycle among the top enriched pathways. Overall, these transcriptome deviations provide potential connections to the overlapping neurocognitive and neuropsychiatric phenotypes associated with key high-risk ASD genes, including chromatin modifiers and epigenetic modulators, that play significant roles in these disease states.


Assuntos
Transtorno do Espectro Autista/genética , Proteínas de Ligação a DNA/genética , Transtornos do Neurodesenvolvimento/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Transtorno do Espectro Autista/metabolismo , Transtorno Autístico/genética , Diferenciação Celular/genética , Deleção Cromossômica , Cromossomos Humanos Par 2/genética , Cromossomos Humanos Par 2/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Proteínas de Ligação a DNA/metabolismo , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Células-Tronco Neurais/metabolismo , Transtornos do Neurodesenvolvimento/metabolismo , Fenótipo , Cultura Primária de Células , RNA-Seq , Transdução de Sinais/genética , Transcriptoma/genética
8.
Microrna ; 9(1): 70-80, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31232238

RESUMO

BACKGROUND: We have previously described mosaic mutations in the RNase IIIb domain of DICER1that display global developmental delays, lung cysts, somatic overgrowth, macrocephaly and Wilms tumor. This constellation of phenotypes was classified as GLOW syndrome. Due to the phenotypic overlap between GLOW and syndromes caused by mutations in the PI3K/AKT/mTOR pathway, we hypothesized that alterations in miRNA regulation of this pathway cause its specific constellation of phenotypes. OBJECTIVE: To test the hypothesis that DICER1 "hot spot" mutations associated with GLOW syndrome activate PI3K/AKT/mTOR signaling. METHODS: We developed HEK293T cells with loss of exon 25 in DICER1, a genetic modification that is synonymous with the "hot spot" RNAseIIIb mutations that cause GLOW syndrome. We assayed the cells for activation of the PI3K/AKT/mTOR signaling pathway. RESULTS: We observed activation of the PI3K/AKT/mTOR pathway as demonstrated by increased pS6Kinase, p4EBP1 and pTSC2 levels. Additionally, these cells demonstrate a striking cellular phenotype, with the ability to form spheres when the serum is removed from their growth medium. The cells in these spheres are Oct4 and Sox2 positive and exhibit the property of reversion with the addition of serum. We queried miRNA expression data and identified a population of miRNAs that increase due to these mutations and target negative regulators of the PI3K/AKT/mTOR pathway. CONCLUSION: This work identifies the delicate and essential role for miRNA control of the PI3K/AKT/mTOR pathway. We conclude that the phenotypes observed in the GLOW syndrome are the result of PI3K/AKT/mTOR activation.


Assuntos
RNA Helicases DEAD-box/genética , Neoplasias Renais/genética , Megalencefalia/genética , MicroRNAs/genética , Ribonuclease III/genética , Tumor de Wilms/genética , Linhagem Celular , Células HEK293 , Humanos , Neoplasias Renais/patologia , Megalencefalia/patologia , Fator 3 de Transcrição de Octâmero/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Tumor de Wilms/patologia
9.
Mol Genet Genomic Med ; 7(2): e00501, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30447054

RESUMO

BACKGROUND: The cohesin complex is a multi-subunit protein complex which regulates sister chromatid cohesion and separation during cellular division. In addition, this evolutionarily conserved protein complex plays an integral role in DNA replication, DNA repair, and the regulation of transcription. The core complex is composed of four subunits: RAD21, SMC1A, SMC3, and STAG1/2. Mutations in these proteins have been implicated in human developmental disorders collectively termed "cohesinopathies." METHODS: Using clinical exome sequencing, we have previously identified three female cases with heterozygous STAG2 mutations and overlapping syndromic phenotypes. Subsequently, a familial missense variant was identified in five male family members. RESULTS: We now present the case of a 4-year-old male with developmental delay, failure to thrive, short stature, and polydactyly with a likely pathogenic STAG2 de novo missense hemizygous variant, c.3027A>T, p.Lys1009Asn. Furthermore, we compare the phenotypes of the four previously reported STAG2 variants with our case. CONCLUSION: We conclude that mutations in STAG2 cause a novel constellation of sex-specific cohesinopathy-related phenotypes and are furthermore, essential for neurodevelopment, human growth, and behavioral development.


Assuntos
Antígenos Nucleares/genética , Deficiências do Desenvolvimento/genética , Doenças Genéticas Ligadas ao Cromossomo X/genética , Transtornos do Crescimento/genética , Fenótipo , Polidactilia/genética , Proteínas de Ciclo Celular , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Transtornos do Crescimento/patologia , Humanos , Masculino , Mutação de Sentido Incorreto , Polidactilia/patologia , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA