Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Hypertension ; 79(11): 2493-2504, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36043416

RESUMO

BACKGROUND: Natural killer (NK) cell impairment is a feature of pulmonary arterial hypertension (PAH) and contributes to vascular remodeling in animal models of disease. Although mutations in BMPR2, the gene encoding the BMP (bone morphogenetic protein) type-II receptor, are strongly associated with PAH, the contribution of BMPR2 loss to NK cell impairment remains unknown. We explored the impairment of IL (interleukin)-15 signaling, a central mediator of NK cell homeostasis, as both a downstream target of BMPR2 loss and a contributor to the pathogenesis of PAH. METHODS: The expression, trafficking, and secretion of IL-15 and IL-15Rα (interleukin 15 α-type receptor) were assessed in human pulmonary artery endothelial cells, with or without BMPR2 silencing. NK cell development and IL-15/IL-15Rα levels were quantified in mice bearing a heterozygous knock-in of the R899X-BMPR2 mutation (bmpr2+/R899X). NK-deficient Il15-/- rats were exposed to the Sugen/hypoxia and monocrotaline models of PAH to assess the impact of impaired IL-15 signaling on disease severity. RESULTS: BMPR2 loss reduced IL-15Rα surface presentation and secretion in human pulmonary artery endothelial cells via impaired trafficking through the trans-Golgi network. bmpr2+/R899X mice exhibited a decrease in NK cells, which was not attributable to impaired hematopoietic development but was instead associated with reduced IL-15/IL-15Rα levels in these animals. Il15-/- rats of both sexes exhibited enhanced disease severity in the Sugen/hypoxia model, with only male Il15-/- rats developing more severe PAH in response to monocrotaline. CONCLUSIONS: This work identifies the loss of IL-15 signaling as a novel BMPR2-dependent contributor to NK cell impairment and pulmonary vascular disease.


Assuntos
Deficiência de GATA2 , Hipertensão Pulmonar , Hipertensão Arterial Pulmonar , Humanos , Feminino , Masculino , Ratos , Camundongos , Animais , Hipertensão Pulmonar/etiologia , Interleucina-15/genética , Interleucina-15/metabolismo , Monocrotalina , Células Endoteliais/metabolismo , Deficiência de GATA2/complicações , Deficiência de GATA2/metabolismo , Deficiência de GATA2/patologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/genética , Receptores de Proteínas Morfogenéticas Ósseas Tipo II/metabolismo , Artéria Pulmonar/metabolismo , Hipóxia/metabolismo
3.
Mutagenesis ; 35(5): 381-389, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33511998

RESUMO

Severe congenital neutropenia (SCN) is a rare blood disorder characterised by abnormally low levels of circulating neutrophils. The most common recurrent mutations that cause SCN involve neutrophil elastase (ELANE). The treatment of choice for SCN is the administration of granulocyte-colony stimulating factor (G-CSF), which increases the neutrophil number and improves the survival and quality of life. Long-term survival is however linked to the development of myelodysplastic syndrome/acute myeloid leukemia (MDS/AML). About 70% of MDS/AML patients acquire nonsense mutations affecting the cytoplasmic domain of CSF3R (the G-CSF receptor). About 70% of SCN patients with AML harbour additional mutations in RUNX1. We hypothesised that this coding region of CSF3R constitutes a hotspot vulnerable to mutations resulting from excessive oxidative stress or endoplasmic reticulum (ER) stress. We used the murine Ba/F3 cell line to measure the effect of induced oxidative or ER stress on the mutation rate in our hypothesised hotspot of the exogenous human CSF3R, the corresponding region in the endogenous Csf3r, and Runx1. Ba/F3 cells transduced with the cDNA for partial C-terminal of CSF3R fused in-frame with a green fluorescent protein (GFP) tag were subjected to stress-inducing treatment for 30 days (~51 doubling times). The amplicon-based targeted deep sequencing data for days 15 and 30 samples show that although there was increased mutagenesis observed in all the three genes of interest (partial CSF3R, Csf3r and Runx1), there were more mutations in the GFP region compared with the partial CSF3R region. Our findings also indicate that there is no correlation between the stress-inducing chemical treatments and mutagenesis in Ba/F3 cells. Our data suggest that oxidative or ER stress induction does not promote genomic instability, affecting partial C-terminal of the transduced CSF3R, the endogenous Csf3R and the endogenous Runx1 in Ba/F3 cells that could account for these targets to being mutational hotspots. We conclude that other mechanisms to acquire mutations of CSF3R that help drive the evolution of SCN to MDS/AML.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Deficiência de GATA2/tratamento farmacológico , Receptores de Fator Estimulador de Colônias/genética , Animais , Síndrome Congênita de Insuficiência da Medula Óssea/complicações , Síndrome Congênita de Insuficiência da Medula Óssea/tratamento farmacológico , Síndrome Congênita de Insuficiência da Medula Óssea/genética , Síndrome Congênita de Insuficiência da Medula Óssea/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Deficiência de GATA2/complicações , Deficiência de GATA2/genética , Deficiência de GATA2/patologia , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Mutagênese/efeitos dos fármacos , Mutagênese/genética , Mutação/efeitos dos fármacos , Taxa de Mutação , Neutropenia/complicações , Neutropenia/congênito , Neutropenia/tratamento farmacológico , Neutropenia/genética , Neutropenia/patologia , Neutrófilos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Receptores de Fator Estimulador de Colônias/administração & dosagem , Transdução de Sinais , Resposta a Proteínas não Dobradas/efeitos dos fármacos
5.
Blood Adv ; 2(23): 3553-3565, 2018 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-30538114

RESUMO

GATA2 deficiency is an inherited or sporadic genetic disorder characterized by distinct cellular deficiency, bone marrow failure, various infections, lymphedema, pulmonary alveolar proteinosis, and predisposition to myeloid malignancies resulting from heterozygous loss-of-function mutations in the GATA2 gene. How heterozygous GATA2 mutations affect human hematopoietic development or cause characteristic cellular deficiency and eventual hypoplastic myelodysplastic syndrome or leukemia is not fully understood. We used induced pluripotent stem cells (iPSCs) to study hematopoietic development in the setting of GATA2 deficiency. We performed hematopoietic differentiation using iPSC derived from patients with GATA2 deficiency and examined their ability to commit to mesoderm, hemogenic endothelial precursors (HEPs), hematopoietic stem progenitor cells, and natural killer (NK) cells. Patient-derived iPSC, either derived from fibroblasts/marrow stromal cells or peripheral blood mononuclear cells, did not show significant defects in committing to mesoderm, HEP, hematopoietic stem progenitor, or NK cells. However, HEP derived from GATA2-mutant iPSC showed impaired maturation toward hematopoietic lineages. Hematopoietic differentiation was nearly abolished from homozygous GATA2 knockout (KO) iPSC lines and markedly reduced in heterozygous KO lines compared with isogenic controls. On the other hand, correction of the mutated GATA2 allele in patient-specific iPSC did not alter hematopoietic development consistently in our model. GATA2 deficiency usually manifests within the first decade of life. Newborn and infant hematopoiesis appears to be grossly intact; therefore, our iPSC model indeed may resemble the disease phenotype, suggesting that other genetic, epigenetic, or environmental factors may contribute to bone marrow failure in these patients following birth. However, heterogeneity of PSC-based models and limitations of in vitro differentiation protocol may limit the possibility to detect subtle cellular phenotypes.


Assuntos
Deficiência de GATA2/patologia , Fator de Transcrição GATA2/genética , Hematopoese , Células-Tronco Pluripotentes Induzidas/metabolismo , Adulto , Antígenos CD34/metabolismo , Diferenciação Celular , Feminino , Deficiência de GATA2/genética , Edição de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Heterozigoto , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Antígenos Comuns de Leucócito/metabolismo , Masculino , Mesoderma/citologia , Mesoderma/metabolismo , Pessoa de Meia-Idade , Mutação
7.
Hematol Oncol Clin North Am ; 32(4): 713-728, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30047422

RESUMO

GATA2 deficiency is an immunodeficiency and bone marrow failure disorder caused by pathogenic variants in GATA2. It is inherited in an autosomal-dominant pattern or can be due to de novo sporadic germline mutation. Patients commonly have B-cell, dendritic cell, natural killer cell, and monocytopenias, and are predisposed to myelodysplastic syndrome, acute myeloid leukemia, and chronic myelomonocytic leukemia. Patients may suffer from disseminated human papilloma virus and mycobacterial infections, pulmonary alveolar proteinosis, and lymphedema. The bone marrow eventually takes on a characteristic hypocellular myelodysplasia with loss of monocytes and hematogones, megakaryocytes with separated nuclear lobes, micromegakaryocytes, and megakaryocytes with hypolobated nuclei.


Assuntos
Anemia Aplástica , Doenças da Medula Óssea , Deficiência de GATA2 , Fator de Transcrição GATA2/genética , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Hemoglobinúria Paroxística , Anemia Aplástica/genética , Anemia Aplástica/metabolismo , Anemia Aplástica/patologia , Doenças da Medula Óssea/genética , Doenças da Medula Óssea/metabolismo , Doenças da Medula Óssea/patologia , Transtornos da Insuficiência da Medula Óssea , Deficiência de GATA2/genética , Deficiência de GATA2/metabolismo , Deficiência de GATA2/patologia , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/metabolismo , Hemoglobinúria Paroxística/patologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/patologia
8.
J Pediatr Hematol Oncol ; 40(4): e225-e228, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29620682

RESUMO

GATA2 deficiency is an inherited bone marrow failure syndrome that can manifest with myelodysplasia (myelodysplastic syndrome) with chromosomal aberrations and high risk of evolution to leukemia (particularly, acute myeloid leukemia); immunodeficiency with opportunistic infections; and/or lymphedema. It can be transmitted in families in autosomal-dominant fashion, or present de novo as sporadic disease in adults or children. The authors report a case of an adolescent male with features of GATA2 deficiency resulting from a complete monoallelic deletion, review chromosomal anomalies associated with this disorder, and discuss the management implications.


Assuntos
Aberrações Cromossômicas , Deficiência de GATA2/genética , Fator de Transcrição GATA2/genética , Deleção de Genes , Síndromes Mielodisplásicas/genética , Adolescente , Deficiência de GATA2/patologia , Humanos , Masculino , Síndromes Mielodisplásicas/patologia
9.
Mol Cell Biol ; 38(12)2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29581184

RESUMO

The transcription factor GATA2 is required for expansion and differentiation of hematopoietic stem cells (HSCs). In mesenchymal stem cells (MSCs), GATA2 blocks adipogenesis, but its biological relevance and underlying genomic events are unknown. We report a dual function of GATA2 in bone homeostasis. GATA2 in MSCs binds near genes involved in skeletal system development and colocalizes with motifs for FOX and HOX transcription factors, known regulators of skeletal development. Ectopic GATA2 blocks osteoblastogenesis by interfering with SMAD1/5/8 activation. MSC-specific deletion of GATA2 in mice increases the numbers and differentiation capacity of bone-derived precursors, resulting in elevated bone formation. Surprisingly, MSC-specific GATA2 deficiency impairs the trabecularization and mechanical strength of bone, involving reduced MSC expression of the osteoclast inhibitor osteoprotegerin and increased osteoclast numbers. Thus, GATA2 affects bone turnover via MSC-autonomous and indirect effects. By regulating bone trabecularization, GATA2 expression in the osteogenic lineage may contribute to the anatomical and cellular microenvironment of the HSC niche required for hematopoiesis.


Assuntos
Osso e Ossos/metabolismo , Fator de Transcrição GATA2/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese/genética , Células 3T3 , Animais , Sítios de Ligação/genética , Células da Medula Óssea/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Microambiente Celular/genética , Fraturas Ósseas/genética , Deficiência de GATA2/genética , Deficiência de GATA2/patologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Proteínas Nucleares/metabolismo , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo , Fatores de Transcrição/metabolismo
11.
Nat Rev Clin Oncol ; 15(4): 219-233, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29311715

RESUMO

Several haematological malignancies, including multiple myeloma (MM) and acute myeloid leukaemia (AML), have well-defined precursor states that precede the development of overt cancer. MM is almost always preceded by monoclonal gammopathy of undetermined significance (MGUS), and at least a quarter of all patients with myelodysplastic syndromes (MDS) have disease that evolves into AML. In turn, MDS are frequently anteceded by clonal haematopoiesis of indeterminate potential (CHIP). The acquisition of additional genetic and epigenetic alterations over time clearly influences the increasingly unstable and aggressive behaviour of neoplastic haematopoietic clones; however, perturbations in the bone-marrow microenvironment are increasingly recognized to have key roles in initiating and supporting oncogenesis. In this Review, we focus on the concept that the haematopoietic neoplasia-microenvironment relationship is an intimate rapport between two partners, provide an overview of the evidence supporting a role for the bone-marrow niche in promoting neoplasia, and discuss the potential for niche-specific therapeutic targets.


Assuntos
Deficiência de GATA2/genética , Leucemia Mieloide Aguda/genética , Gamopatia Monoclonal de Significância Indeterminada/genética , Mieloma Múltiplo/genética , Células da Medula Óssea/patologia , Evolução Clonal/genética , Deficiência de GATA2/patologia , Hematopoese/genética , Humanos , Leucemia Mieloide Aguda/patologia , Gamopatia Monoclonal de Significância Indeterminada/patologia , Mieloma Múltiplo/patologia , Células-Tronco Neoplásicas/patologia , Nicho de Células-Tronco/genética , Microambiente Tumoral/genética
12.
J Pediatr Hematol Oncol ; 40(6): e383-e388, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29189513

RESUMO

Emberger syndrome with underlying guanine-adenine-thymine-adenine 2 (GATA2) mutation is a rare disorder and very few successful nonmyeloablative allogeneic hematopoietic stem cell transplants (HSCTs) have been reported. We report a case of Emberger syndrome with GATA2 mutation in a 9-year-old girl who presented with congenital sensorineural deafness, warts, lymphedema, and Myelodysplastic syndrome. Her sister had died of a similar illness. She underwent a nonmyeloablative matched related donor peripheral blood HSCT with rabbit antithymoglobulin (5 mg/kg), fludarabine (160 mg/m), cyclophophamide (29 mg/kg), and total body irradiation (2 Gray). Graft versus host disease prophylaxis consisted of tacrolimus and mycophenolate moefetil. She had neutrophil engraftment on day+15 and fully donor chimerism by day+30. She developed limited chronic skin graft versus host disease on tapering off immunosuppression. She is disease free on day+475. The review of literature showed a total of 28 patients with GATA2 mutation have undergone HSCT mostly nonmyeloablative and overall survival is 75%. Nonmyeloablatove HSCT is feasible and safe for the patients with GATA2 mutation.


Assuntos
Deficiência de GATA2/terapia , Fator de Transcrição GATA2/genética , Mutação , Transplante de Células-Tronco de Sangue Periférico , Condicionamento Pré-Transplante , Aloenxertos , Soro Antilinfocitário/administração & dosagem , Criança , Ciclofosfamida/administração & dosagem , Feminino , Deficiência de GATA2/genética , Deficiência de GATA2/patologia , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Ácido Micofenólico/administração & dosagem , Dermatopatias/genética , Dermatopatias/prevenção & controle , Tacrolimo/administração & dosagem , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados , Irradiação Corporal Total
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA