Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Med Genet A ; 188(10): 3089-3095, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35946377

RESUMO

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.


Assuntos
Síndrome de Peutz-Jeghers , Quinases Proteína-Quinases Ativadas por AMP , Processamento Alternativo , Códon sem Sentido , Humanos , Nucleotídeos , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patologia
2.
Blood ; 140(17): 1858-1874, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-35789258

RESUMO

The discovery of humans with monogenic disorders has a rich history of generating new insights into biology. Here we report the first human identified with complete deficiency of nuclear factor of activated T cells 1 (NFAT1). NFAT1, encoded by NFATC2, mediates calcium-calcineurin signals that drive cell activation, proliferation, and survival. The patient is homozygous for a damaging germline NFATC2 variant (c.2023_2026delTACC; p.Tyr675Thrfs∗18) and presented with joint contractures, osteochondromas, and recurrent B-cell lymphoma. Absence of NFAT1 protein in chondrocytes caused enrichment in prosurvival and inflammatory genes. Systematic single-cell-omic analyses in PBMCs revealed an environment that promotes lymphomagenesis with accumulation of naïve B cells (enriched for oncogenic signatures MYC and JAK1), exhausted CD4+ T cells, impaired T follicular helper cells, and aberrant CD8+ T cells. This work highlights the pleiotropic role of human NFAT1, will empower the diagnosis of additional patients with NFAT1 deficiency, and further defines the detrimental effects associated with long-term use of calcineurin inhibitors.


Assuntos
Contratura , Leucemia de Células B , Osteocondroma , Humanos , Calcineurina/genética , Leucemia de Células B/genética , Leucemia de Células B/metabolismo , Recidiva Local de Neoplasia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Linfoma de Células B/genética , Linfoma de Células B/metabolismo
3.
Front Immunol ; 12: 788278, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34887873

RESUMO

B-cell lymphoma/leukemia 11B (BCL11B) is a C2H2 zinc finger transcription factor that is critically important for regulating the development and function of a variety of systems including the central nervous system, the skin, and the immune system. Germline heterozygous variants are associated with a spectrum of clinical disorders, including severe combined immunodeficiency as well as neurological, craniofacial, and dermal defects. Of these individuals, ~50% present with severe allergic disease. Here, we report the detailed clinical and laboratory workup of one of the most severe BCL11B-dependent atopic cases to date. Leveraging a zebrafish model, we were able to confirm a strong T-cell defect in the patient. Based on these data, we classify germline BCL11B-dependent atopic disease as a novel primary atopic disorder.


Assuntos
Mutação em Linhagem Germinativa , Hipersensibilidade/genética , Doenças da Imunodeficiência Primária/genética , Proteínas Repressoras/genética , Linfócitos T/imunologia , Proteínas Supressoras de Tumor/genética , Adolescente , Animais , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Heterozigoto , Humanos , Hipersensibilidade/diagnóstico , Hipersensibilidade/imunologia , Fenótipo , Doenças da Imunodeficiência Primária/diagnóstico , Doenças da Imunodeficiência Primária/imunologia , Doenças da Imunodeficiência Primária/metabolismo , Proteínas Repressoras/metabolismo , Índice de Gravidade de Doença , Linfócitos T/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
4.
Allergy Asthma Clin Immunol ; 17(1): 9, 2021 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-33446255

RESUMO

X-linked hypohidrotic ectodermal dysplasia (XLHED) is the most common form of ectodermal dysplasia. Clinical and genetic heterogeneity between different ectodermal dysplasia types and evidence of incomplete penetrance and variable expressivity increase the potential for misdiagnosis. We describe a family with X-linked hypohidrotic ectodermal dysplasia (XLHED) presenting with variable expressivity of symptoms between affected siblings. In addition to the classical signs of hypohidrosis, hypotrichosis and hypodontia, the index patient-a 5 year old boy, also presented with a severe atopy phenotype that was not observed in the other two affected brothers. Exome sequencing in the index and the mother identified a pathogenic nonsense variant in EDA (NM_001399.4: c.766 C>T; p. Gln256Ter). This study highlights how exome sequencing was crucial in establishing a precise molecular diagnosis of XLHED by enabling us to rule out other differential diagnoses including NEMO deficiency syndrome, that was initially presented as a clinical diagnosis to the family.

5.
BMC Pediatr ; 21(1): 45, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33472608

RESUMO

BACKGROUND: KRAS (KRAS proto-oncogene, GTPase; OMIM: 190,070) encodes one of three small guanosine triphosphatase proteins belonging to the RAS family. This group of proteins is responsible for cell proliferation, differentiation and inhibition of apoptosis. Gain-of-function variants in KRAS are commonly found in human cancers. Non-malignant somatic KRAS variants underlie a subset of RAS-associated autoimmune leukoproliferative disorders (RALD). RALD is characterized by splenomegaly, persistent monocytosis, hypergammaglobulinemia and cytopenia, but can also include autoimmune features and lymphadenopathy. In this report, we describe a non-malignant somatic variant in KRAS with prominent clinical features of massive splenomegaly, thrombocytopenia and lymphopenia. CASE PRESENTATION: A now-11-year-old girl presented in early childhood with easy bruising and bleeding, but had an otherwise unremarkable medical history. After consulting for the first time at 5 years of age, she was discovered to have massive splenomegaly. Clinical follow-up revealed thrombocytopenia, lymphopenia and increased polyclonal immunoglobulins and C-reactive protein. The patient had an unremarkable bone marrow biopsy, flow cytometry showed no indication of expanded double negative T-cells, while malignancy and storage disorders were also excluded. When the patient was 8 years old, whole exome sequencing performed on DNA derived from whole blood revealed a heterozygous gain-of-function variant in KRAS (NM_004985.5:c.37G > T; (p.G13C)). The variant was absent from DNA derived from a buccal swab and was thus determined to be somatic. CONCLUSIONS: This case of idiopathic splenomegaly in childhood due to a somatic variant in KRAS expands our understanding of the clinical spectrum of RAS-associated autoimmune leukoproliferative disorder and emphasizes the value of securing a molecular diagnosis in children with unusual early-onset presentations with a suspected monogenic origin.


Assuntos
Transtornos Linfoproliferativos , Esplenomegalia , Biópsia , Criança , Pré-Escolar , Feminino , Citometria de Fluxo , Humanos , Mutação , Proto-Oncogene Mas , Esplenomegalia/etiologia
6.
Clin Immunol ; 175: 143-146, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28043923

RESUMO

OBJECTIVES: Clinicians need to be aware of the growing list of defined monogenic etiologies of autoimmune diseases. This is particularly relevant when evaluating children, as these rare monogenic forms of autoimmunity tend to present very early in life. METHODS AND RESULTS: By harnessing the transformative power of next generation sequencing, we made the unifying diagnosis of RAS-associated autoimmune leukoproliferative disease (RALD), caused by the somatic gain-of-function p.G13C KRAS mutation, in a boy with the seemingly unrelated immune dysregulatory conditions of Rosai-Dorfman and systemic lupus erythematosus (SLE). CONCLUSIONS: This case expands our understanding of the clinical phenotypes associated with the extremely rare condition of RALD, and emphasizes the importance of always considering the possibility of a monogenic cause for autoimmunity, particularly when the disease manifestations begin early in life and do not follow a typical clinical course.


Assuntos
Autoimunidade/genética , Histiocitose Sinusal/genética , Lúpus Eritematoso Sistêmico/genética , Mutação/genética , Mutação/imunologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Adolescente , Autoimunidade/imunologia , Histiocitose Sinusal/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Síndrome
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA