Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Blood Adv ; 7(20): 6351-6363, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37595058

RESUMO

Mutations in the transcription factor GATA2 can cause MonoMAC syndrome, a GATA2 deficiency disease characterized by several findings, including disseminated nontuberculous mycobacterial infections, severe deficiencies of monocytes, natural killer cells, and B lymphocytes, and myelodysplastic syndrome. GATA2 mutations are found in âˆ¼90% of patients with a GATA2 deficiency phenotype and are largely missense mutations in the conserved second zinc-finger domain. Mutations in an intron 5 regulatory enhancer element are also well described in GATA2 deficiency. Here, we present a multigeneration kindred with the clinical features of GATA2 deficiency but lacking an apparent GATA2 mutation. Whole genome sequencing revealed a unique adenine-to-thymine variant in the GATA2 -110 enhancer 116,855 bp upstream of the GATA2 ATG start site. The mutation creates a new E-box consensus in position with an existing GATA-box to generate a new hematopoietic regulatory composite element. The mutation segregates with the disease in several generations of the family. Cell type-specific allelic imbalance of GATA2 expression was observed in the bone marrow of a patient with higher expression from the mutant-linked allele. Allele-specific overexpression of GATA2 was observed in CRISPR/Cas9-modified HL-60 cells and in luciferase assays with the enhancer mutation. This study demonstrates overexpression of GATA2 resulting from a single nucleotide change in an upstream enhancer element in patients with MonoMAC syndrome. Patients in this study were enrolled in the National Institute of Allergy and Infectious Diseases clinical trial and the National Cancer Institute clinical trial (both trials were registered at www.clinicaltrials.gov as #NCT01905826 and #NCT01861106, respectively).


Assuntos
Deficiência de GATA2 , Síndromes Mielodisplásicas , Humanos , Deficiência de GATA2/genética , Sequências Reguladoras de Ácido Nucleico , Síndromes Mielodisplásicas/genética , Mutação , Regulação da Expressão Gênica , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo
2.
Blood Adv ; 6(3): 793-807, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-34529785

RESUMO

Patients with GATA2 deficiencyharbor de novo or inherited germline mutations in the GATA2 transcription factor gene, predisposing them to myeloid malignancies. There is considerable variation in disease progression, even among family members with the same mutation in GATA2. We investigated somatic mutations in 106 patients with GATA2 deficiency to identify acquired mutations that are associated with myeloid malignancies. Myelodysplastic syndrome (MDS) was the most common diagnosis (∼44%), followed by GATA2 bone marrow immunodeficiency disorder (G2BMID; ∼37%). Thirteen percent of the cohort had GATA2 mutations but displayed no disease manifestations. There were no correlations between age or sex with disease progression or survival. Cytogenetic analyses showed a high incidence of abnormalities (∼43%), notably trisomy 8 (∼23%) and monosomy 7 (∼12%), but the changes did not correlate with lower survival. Somatic mutations in ASXL1 and STAG2 were detected in ∼25% of patients, although the mutations were rarely concomitant. Mutations in DNMT3A were found in ∼10% of patients. These somatic mutations were found similarly in G2BMID and MDS, suggesting clonal hematopoiesis in early stages of disease, before the onset of MDS. ASXL1 mutations conferred a lower survival probability and were more prevalent in female patients. STAG2 mutations also conferred a lower survival probability, but did not show a statistically significant sex bias. There was a conspicuous absence of many commonly mutated genes associated with myeloid malignancies, including TET2, IDH1/2, and the splicing factor genes. Notably, somatic mutations in chromatin-related genes and cohesin genes characterized disease progression in GATA2 deficiency.


Assuntos
Deficiência de GATA2 , Síndromes Mielodisplásicas , Transtornos Mieloproliferativos , Neoplasias , Proteínas de Ciclo Celular/genética , Progressão da Doença , Feminino , Deficiência de GATA2/complicações , Deficiência de GATA2/genética , Fator de Transcrição GATA2/genética , Humanos , Mutação , Síndromes Mielodisplásicas/patologia , Proteínas Repressoras/genética
3.
Zebrafish ; 12(3): 221-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25798815

RESUMO

The zebrafish (Danio rerio) represents an important animal model for analyzing genetic contributors to carcinogenesis. To assess the role for mutationally activated Ras in ovarian cancer, we developed a transgenic zebrafish model using the putative promoter for zebrafish insulin-like growth factor 3 (igf3) to drive expression of the human oncogene KRAS(G12V) fused to EGFP. A member of the IGF family, igf3 is unique to teleosts and reportedly exhibits gonad-specific expression in fish species. In contrast to previous studies, we observed igf3 expression in wild-type zebrafish gills in addition to gonads, indicating that igf3 expression is not necessarily gonad specific. In transgenic zebrafish, expression of EGFP-KRAS(G12V) driven by the igf3 promoter occurred only in the gills and resulted in proliferation of a putative progenitor cell population, chondroid hyperplasia, and localized inflammation. KRAS(G12V)-transformed cells in transgenic zebrafish showed activation of the ERK-MAP kinase pathway and expressed the zebrafish homologue for human CXCL8, a cytokine produced by mammalian Ras-transformed cells in tumor-associated inflammatory lesions. These findings indicate that KRAS(G12V)-transformed cells in zebrafish recruit inflammatory cells, but may require additional mutational events for neoplastic transformation. The conserved role for mutationally activated KRAS in leukocyte recruitment indicates that zebrafish can provide a valuable comparative model for Ras-associated inflammation.


Assuntos
Inflamação/etiologia , Interleucina-8/metabolismo , Neoplasias Experimentais/etiologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Somatomedinas/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Animais Geneticamente Modificados , Feminino , Brânquias/metabolismo , Brânquias/patologia , Proteínas de Fluorescência Verde , Hiperplasia , Inflamação/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Neoplasias Experimentais/metabolismo , Oncogenes , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Transcrição SOX9/metabolismo , Peixe-Zebra
4.
PLoS One ; 9(1): e87177, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24489863

RESUMO

Germline mutations in the tumor suppressor genes BRCA2 and TP53 significantly influence human cancer risk, and cancers from humans who inherit one mutant allele for BRCA2 or TP53 often display loss of the wildtype allele. In addition, BRCA2-associated cancers often exhibit mutations in TP53. To determine the relationship between germline heterozygous mutation (haploinsufficiency) and somatic loss of heterozygosity (LOH) for BRCA2 and TP53 in carcinogenesis, we analyzed zebrafish with heritable mutations in these two genes. Tumor-bearing zebrafish were examined by histology, and normal and neoplastic tissues were collected by laser-capture microdissection for LOH analyses. Zebrafish on a heterozygous tp53(M214K) background had a high incidence of malignant tumors. The brca2(Q658X) mutation status determined both the incidence of LOH and the malignant tumor phenotype. LOH for tp53 occurred in the majority of malignant tumors from brca2 wildtype and heterozygous mutant zebrafish, and most of these were malignant peripheral nerve sheath tumors. Malignant tumors in zebrafish with heterozygous mutations in both brca2 and tp53 frequently displayed LOH for both genes. In contrast, LOH for tp53 was uncommon in malignant tumors from brca2 homozygotes, and these tumors were primarily undifferentiated sarcomas. Thus, carcinogenesis in zebrafish with combined mutations in tp53 and brca2 typically requires biallelic mutation or loss of at least one of these genes, and the specific combination of inherited mutations influences the development of LOH and the tumor phenotype. These results provide insight into cancer development associated with heritable BRCA2 and TP53 mutations.


Assuntos
Proteína BRCA2/genética , Carcinogênese/genética , Proteína Supressora de Tumor p53/genética , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/genética , Animais , Modelos Animais de Doenças , Feminino , Estudos de Associação Genética , Haploinsuficiência , Humanos , Masculino , Mutação de Sentido Incorreto , Neoplasias de Bainha Neural/genética , Sarcoma/genética
5.
Proc Natl Acad Sci U S A ; 107(45): 19350-5, 2010 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-20974951

RESUMO

Humans with inherited mutations in BRCA2 are at increased risk for developing breast and ovarian cancer; however, the relationship between BRCA2 mutation and these cancers is not understood. Studies of Brca2 mutation by gene targeting in mice are limited, given that homozygous Brca2 mutation typically leads to early embryonic lethality. We established a zebrafish line with a nonsense mutation in brca2 exon 11 (brca2(Q658X)), a mutation similar in location and type to BRCA2 mutations found in humans with hereditary breast and ovarian cancer. brca2(Q658X) homozygous zebrafish are viable and survive to adulthood; however, juvenile homozygotes fail to develop ovaries during sexual differentiation. Instead, brca2(Q658X) homozygotes develop as infertile males with meiotic arrest in spermatocytes. Germ cell migration to the embryonic gonadal ridge is unimpaired in brca2(Q658X) homozygotes; thus, failure of ovarian development is not due to defects in early establishment of the embryonic gonad. Homozygous tp53 mutation rescues ovarian development in brca2(Q658X) homozygous zebrafish, reflecting the importance of germ cell apoptosis in gonad morphogenesis. Adult brca2(Q658X) homozygous zebrafish are predisposed to testicular neoplasias. In addition, tumorigenesis in multiple tissues is significantly accelerated in combination with homozygous tp53 mutation in both brca2(Q658X) homozygous and brca2(Q658X) heterozygous zebrafish. These studies reveal critical roles for brca2 in ovarian development and tumorigenesis in reproductive tissues.


Assuntos
Proteína BRCA2/genética , Códon sem Sentido , Genes BRCA2 , Neoplasias/etiologia , Ovário/crescimento & desenvolvimento , Espermatogênese/genética , Animais , Éxons , Feminino , Células Germinativas , Homozigoto , Infertilidade/genética , Masculino , Neoplasias/genética , Neoplasias Testiculares/etiologia , Neoplasias Testiculares/genética , Proteína Supressora de Tumor p53/genética , Peixe-Zebra
6.
Cancer Res ; 69(10): 4363-71, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19417137

RESUMO

The mechanism whereby the fusion of EWSR1 with the ETS transcription factor FLI1 contributes to malignant transformation in Ewing sarcoma remains unclear. We show that injection of human or zebrafish EWSR1/FLI1 mRNA into developing zebrafish embryos leads to mitotic defects with multipolar and disorganized mitotic spindles. Expression of human EWSR1/FLI1 in HeLa cells also results in mitotic defects, along with mislocalization of Aurora kinase B, a key regulator of mitotic progression. Because these mitotic abnormalities mimic those observed with the knockdown of EWSR1 in zebrafish embryos and HeLa cells, we investigated whether EWSR1/FLI1 interacts with EWSR1 and interferes with its function. EWSR1 coimmunoprecipitates with EWSR1/FLI1, and overexpression of EWSR1 rescues the mitotic defects in EWSR1/FLI1-transfected HeLa cells. This interaction between EWSR1/FLI1 and EWSR1 in Ewing sarcoma may induce mitotic defects leading to genomic instability and subsequent malignant transformation.


Assuntos
Proteínas de Ligação a Calmodulina/genética , Proteína Proto-Oncogênica c-fli-1/genética , Proteínas de Ligação a RNA/genética , Sarcoma de Ewing/genética , Sarcoma de Ewing/patologia , Proteínas de Peixe-Zebra/genética , Animais , Proteínas de Ligação a Calmodulina/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Embrião não Mamífero/patologia , Instabilidade Genômica , Células HeLa , Humanos , Mitose/genética , Proteína Proto-Oncogênica c-fli-1/metabolismo , RNA Mensageiro/genética , Proteína EWS de Ligação a RNA , Proteínas de Ligação a RNA/metabolismo , Sarcoma de Ewing/veterinária , Transfecção , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
7.
PLoS One ; 2(10): e979, 2007 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-17912356

RESUMO

BACKGROUND: The Ewing sarcoma breakpoint region 1 gene (EWSR1), also known as EWS, is fused to a number of different partner genes as a result of chromosomal translocation in diverse sarcomas. Despite the involvement of EWSR1 in these diverse sarcomas, the in vivo function of wild type EWSR1 remains unclear. PRINCIPAL FINDINGS: We identified two zebrafish EWSR1 orthologues, ewsr1a and ewsr1b, and demonstrate that both genes are expressed maternally, and are expressed ubiquitously throughout zebrafish embryonic development. Morpholino induced knockdown of both zebrafish ewsr1 genes led to mitotic defects with multipolar or otherwise abnormal mitotic spindles starting from the bud stage (10 hour post-fertilization (hpf)). The abnormalities in mitotic spindles were followed by p53-mediated apoptosis in the developing central nervous system (CNS) leading to a reduction in the number of proneural cells, disorganization of neuronal networks, and embryonic lethality by 5 days post-fertilization. siRNA silencing of EWSR1 in Hela cells resulted in mitotic defects accompanied by apoptotic cell death, indicating that the role of EWSR1 is conserved between zebrafish and human. CONCLUSIONS: Ewsr1 maintains mitotic integrity and proneural cell survival in early zebrafish development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Mitose , Neurônios/metabolismo , Proteína EWS de Ligação a RNA/fisiologia , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/embriologia , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Apoptose , Sobrevivência Celular , Feminino , Modelos Biológicos , Dados de Sequência Molecular , Ligação Proteica , Proteína EWS de Ligação a RNA/genética , Homologia de Sequência de Aminoácidos
8.
Proc Natl Acad Sci U S A ; 103(41): 15166-71, 2006 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-17015828

RESUMO

Acute lymphoblastic leukemia (ALL) is a clonal disease that evolves through the accrual of genetic rearrangements and/or mutations within the dominant clone. The TEL-AML1 (ETV6-RUNX1) fusion in precursor-B (pre-B) ALL is the most common genetic rearrangement in childhood cancer; however, the cellular origin and the molecular pathogenesis of TEL-AML1-induced leukemia have not been identified. To study the origin of TEL-AML1-induced ALL, we generated transgenic zebrafish expressing TEL-AML1 either ubiquitously or in lymphoid progenitors. TEL-AML1 expression in all lineages, but not lymphoid-restricted expression, led to progenitor cell expansion that evolved into oligoclonal B-lineage ALL in 3% of the transgenic zebrafish. This leukemia was transplantable to conditioned wild-type recipients. We demonstrate that TEL-AML1 induces a B cell differentiation arrest, and that leukemia development is associated with loss of TEL expression and elevated Bcl2/Bax ratio. The TEL-AML1 transgenic zebrafish models human pre-B ALL, identifies the molecular pathways associated with leukemia development, and serves as the foundation for subsequent genetic screens to identify modifiers and leukemia therapeutic targets.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Modelos Animais de Doenças , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular/genética , Criança , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 2 de Fator de Ligação ao Core/biossíntese , Inativação Gênica , Células-Tronco Hematopoéticas/patologia , Humanos , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/biossíntese , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Regulação para Cima/genética , Proteína X Associada a bcl-2/biossíntese
9.
Blood ; 103(9): 3582-9, 2004 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-14715622

RESUMO

Children with the severe phenotype of the genetic immunodeficiency disease leukocyte adhesion deficiency or LAD experience life-threatening bacterial infections because of molecular defects in the leukocyte integrin CD18 molecule and the resultant failure to express the CD11/CD18 adhesion molecules on the leukocyte surface. Hematopoietic stem cell transplantation remains the only definitive therapy for LAD; however, the degree of donor chimerism and particularly the number of CD18(+) donor-derived neutrophils required to reverse the disease phenotype are not known. We performed nonmyeloablative hematopoietic stem cell transplantations from healthy matched littermates in 9 dogs with the canine form of LAD known as CLAD and demonstrate that in the 3 dogs with the lowest level of donor chimerism, less than 500 CD18(+) donor-derived neutrophils/microL in the peripheral blood of the CLAD recipients resulted in reversal of the CLAD disease phenotype. These results demonstrate the value of a disease-specific, large-animal model for identifying the lowest therapeutic level required for successful cellular and gene therapy.


Assuntos
Antígenos CD18/análise , Transplante de Células-Tronco Hematopoéticas , Síndrome da Aderência Leucocítica Deficitária/terapia , Neutrófilos/citologia , Animais , DNA/análise , Doenças do Cão , Cães , Citometria de Fluxo , Contagem de Leucócitos , Síndrome da Aderência Leucocítica Deficitária/sangue , Fenótipo , Quimeras de Transplante , Transplante Homólogo , Resultado do Tratamento
10.
Vet Immunol Immunopathol ; 95(3-4): 113-21, 2003 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12963272

RESUMO

The genetic disease canine leukocyte adhesion deficiency (CLAD) is characterized by recurrent, severe bacterial infections, typically culminating in death by 6 months of age. CLAD is due to a mutation in the leukocyte integrin CD18 subunit, which prevents surface expression of the CD11/CD18 leukocyte integrin complex. We demonstrate that stable mixed donor:host hematopoietic chimerism, achieved by a non-myeloablative bone marrow transplant from a histocompatible littermate, reverses the disease phenotype in CLAD. Donor chimerism following the transplant was demonstrated both by flow cytometric detection of donor-derived CD18-positive leukocytes in the peripheral blood of the recipient, and by the demonstration of donor-derived DNA microsatellite repeats in the peripheral blood leukocytes of the recipient. These results indicate that mixed hematopoietic chimerism reverses the clinical phenotype in CLAD and represents a potential therapeutic approach for the human disease leukocyte adhesion deficiency.


Assuntos
Doenças do Cão/terapia , Transplante de Células-Tronco Hematopoéticas/veterinária , Síndrome da Aderência Leucocítica Deficitária/veterinária , Quimeras de Transplante/imunologia , Animais , Antígenos CD34/imunologia , Antígenos CD11/imunologia , Antígenos CD18/imunologia , DNA/química , DNA/genética , Doenças do Cão/imunologia , Doenças do Cão/patologia , Cães , Citometria de Fluxo/veterinária , Transplante de Células-Tronco Hematopoéticas/métodos , Contagem de Leucócitos/veterinária , Síndrome da Aderência Leucocítica Deficitária/imunologia , Síndrome da Aderência Leucocítica Deficitária/patologia , Síndrome da Aderência Leucocítica Deficitária/terapia , Reação em Cadeia da Polimerase/veterinária
11.
Vet Immunol Immunopathol ; 94(1-2): 11-22, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12842608

RESUMO

The genetic immunodeficiency disease canine leukocyte adhesion deficiency (CLAD) was originally described in juvenile Irish Setters with severe, recurrent bacterial infections. CLAD was subsequently shown to result from a mutation in the leukocyte integrin CD18 subunit which prevents leukocyte surface expression of the CD11/CD18 complex. We describe the development of a mixed-breed CLAD colony with clinical features that closely parallel those described in Irish Setters. We demonstrate that the early identification of CLAD heterozygotes and CLAD-affected dogs by a combination of flow cytometry and DNA sequencing allows the CLAD-affected animals to receive life-saving antibiotic therapy. The distinct clinical phenotype in CLAD, the ability to detect CD18 on the leukocyte surface by flow cytometry, and the history of the canine model in marrow transplantation, enable CLAD to serve as an attractive large-animal model for the investigation of novel hematopoietic stem cell and gene therapy strategies.


Assuntos
Doenças do Cão/genética , Cães/genética , Síndrome da Aderência Leucocítica Deficitária/genética , Síndrome da Aderência Leucocítica Deficitária/veterinária , Animais , Cruzamento , Antígenos CD18/análise , Doenças do Cão/patologia , Doenças do Cão/terapia , Feminino , Genótipo , Heterozigoto , Síndrome da Aderência Leucocítica Deficitária/patologia , Síndrome da Aderência Leucocítica Deficitária/terapia , Masculino , Repetições Minissatélites/genética , Mutação/genética , Linhagem , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA