Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Semin Immunol ; 54: 101515, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-34772606

RESUMO

A considerable amount of continuous proliferation and differentiation is required to produce daily a billion new neutrophils in an adult human. Of the few cytokines and factors known to control neutrophil production, G-CSF is the guardian of granulopoiesis. G-CSF/CSF3R signaling involves the recruitment of non-receptor protein tyrosine kinases and their dependent signaling pathways of serine/threonine kinases, tyrosine phosphatases, and lipid second messengers. These pathways converge to activate the families of STAT and C/EBP transcription factors. CSF3R mutations are associated with human disorders of neutrophil production, including severe congenital neutropenia, neutrophilia, and myeloid malignancies. More than three decades after their identification, cloning, and characterization of G-CSF and G-CSF receptor, fundamental questions remain about their physiology.


Assuntos
Fator Estimulador de Colônias de Granulócitos , Neutropenia , Adulto , Síndrome Congênita de Insuficiência da Medula Óssea , Fator Estimulador de Colônias de Granulócitos/metabolismo , Hematopoese , Humanos , Neutropenia/congênito , Neutropenia/genética , Neutropenia/patologia , Neutrófilos/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/genética , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo
2.
Br J Haematol ; 204(1): 45-55, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38049194

RESUMO

Neutrophils are the shortest-lived blood cells, which requires a prodigious degree of proliferation and differentiation to sustain physiologically sufficient numbers and be poised to respond quickly to infectious emergencies. More than 107 neutrophils are produced every minute in an adult bone marrow-a process that is tightly regulated by a small group of cytokines and chemical mediators and dependent on nutrients and energy. Like granulocyte colony-stimulating factor, the primary growth factor for granulopoiesis, they stimulate signalling pathways, some affecting metabolism. Nutrient or energy deficiency stresses the survival, proliferation, and differentiation of neutrophils and their precursors. Thus, it is not surprising that monogenic disorders related to metabolism exist that result in neutropenia. Among these are pathogenic mutations in HAX1, G6PC3, SLC37A4, TAFAZZIN, SBDS, EFL1 and the mitochondrial disorders. These mutations perturb carbohydrate, lipid and/or protein metabolism. We hypothesize that metabolic disturbances may drive the pathogenesis of a subset of inherited neutropenias just as defects in DNA damage response do in Fanconi anaemia, telomere maintenance in dyskeratosis congenita and ribosome formation in Diamond-Blackfan anaemia. Greater understanding of metabolic pathways in granulopoiesis will identify points of vulnerability in production and may point to new strategies for the treatment of neutropenias.


Assuntos
Doenças da Medula Óssea , Anemia de Fanconi , Neutropenia , Adulto , Humanos , Doenças da Medula Óssea/genética , Anemia de Fanconi/genética , Medula Óssea/patologia , Transtornos da Insuficiência da Medula Óssea , Neutropenia/patologia , Proteínas Adaptadoras de Transdução de Sinal , Proteínas de Transporte de Monossacarídeos , Antiporters
3.
Br J Haematol ; 204(1): 292-305, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37876306

RESUMO

Shwachman-Diamond syndrome (SDS) is characterized by neutropenia, exocrine pancreatic insufficiency and skeletal abnormalities. SDS bone marrow haematopoietic progenitors show increased apoptosis and impairment in granulocytic differentiation. Loss of Shwachman-Bodian-Diamond syndrome (SBDS) expression results in reduced eukaryotic 80S ribosome maturation. Biallelic mutations in the SBDS gene are found in ~90% of SDS patients, ~55% of whom carry the c.183-184TA>CT nonsense mutation. Several translational readthrough-inducing drugs aimed at suppressing nonsense mutations have been developed. One of these, ataluren, has received approval in Europe for the treatment of Duchenne muscular dystrophy. We previously showed that ataluren can restore full-length SBDS protein synthesis in SDS-derived bone marrow cells. Here, we extend our preclinical study to assess the functional restoration of SBDS capabilities in vitro and ex vivo. Ataluren improved 80S ribosome assembly and total protein synthesis in SDS-derived cells, restored myelopoiesis in myeloid progenitors, improved neutrophil chemotaxis in vitro and reduced neutrophil dysplastic markers ex vivo. Ataluren also restored full-length SBDS synthesis in primary osteoblasts, suggesting that its beneficial role may go beyond the myeloid compartment. Altogether, our results strengthened the rationale for a Phase I/II clinical trial of ataluren in SDS patients who harbour the nonsense mutation.


Assuntos
Doenças da Medula Óssea , Insuficiência Pancreática Exócrina , Lipomatose , Humanos , Síndrome de Shwachman-Diamond , Proteína Supressora de Tumor p53/genética , Lipomatose/genética , Códon sem Sentido , Mielopoese , Neutrófilos/metabolismo , Quimiotaxia , Doenças da Medula Óssea/genética , Doenças da Medula Óssea/terapia , Insuficiência Pancreática Exócrina/genética , Ribossomos/metabolismo
4.
Haematologica ; 108(10): 2594-2605, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37226705

RESUMO

Shwachman-Diamond syndrome is a rare inherited bone marrow failure syndrome characterized by neutropenia, exocrine pancreatic insufficiency, and skeletal abnormalities. In 10-30% of cases, transformation to a myeloid neoplasm occurs. Approximately 90% of patients have biallelic pathogenic variants in the SBDS gene located on human chromosome 7q11. Over the past several years, pathogenic variants in three other genes have been identified to cause similar phenotypes; these are DNAJC21, EFL1, and SRP54. Clinical manifestations involve multiple organ systems and those classically associated with the Shwachman-Diamond syndrome (bone, blood, and pancreas). Neurocognitive, dermatologic, and retinal changes may also be found. There are specific gene-phenotype differences. To date, SBDS, DNAJC21, and SRP54 variants have been associated with myeloid neoplasia. Common to SBDS, EFL1, DNAJC21, and SRP54 is their involvement in ribosome biogenesis or early protein synthesis. These four genes constitute a common biochemical pathway conserved from yeast to humans that involve early stages of protein synthesis and demonstrate the importance of this synthetic pathway in myelopoiesis.


Assuntos
Doenças da Medula Óssea , Insuficiência Pancreática Exócrina , Lipomatose , Humanos , Síndrome de Shwachman-Diamond , Lipomatose/genética , Lipomatose/metabolismo , Lipomatose/patologia , Doenças da Medula Óssea/genética , Doenças da Medula Óssea/patologia , Mutação , Insuficiência Pancreática Exócrina/genética , Insuficiência Pancreática Exócrina/metabolismo , Insuficiência Pancreática Exócrina/patologia , Partícula de Reconhecimento de Sinal/genética
5.
J Biol Chem ; 295(21): 7492-7500, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32299910

RESUMO

Severe congenital neutropenia (SCN) is characterized by a near absence of neutrophils, rendering individuals with this disorder vulnerable to recurrent life-threatening infections. The majority of SCN cases arise because of germline mutations in the gene elastase, neutrophil-expressed (ELANE) encoding the neutrophil granule serine protease neutrophil elastase. Treatment with a high dose of granulocyte colony-stimulating factor increases neutrophil production and reduces infection risk. How ELANE mutations produce SCN remains unknown. The currently proposed mechanism is that ELANE mutations promote protein misfolding, resulting in endoplasmic reticulum stress and activation of the unfolded protein response (UPR), triggering death of neutrophil precursors and resulting in neutropenia. Here we studied the ELANE mutation p.G185R, often associated with greater clinical severity (e.g. decreased responsiveness to granulocyte colony-stimulating factor and increased leukemogenesis). Using an inducible expression system, we observed that this ELANE mutation diminishes enzymatic activity and granulocytic differentiation without significantly affecting cell proliferation, cell death, or UPR induction in murine myeloblast 32D and human promyelocytic NB4 cells. Impaired differentiation was associated with decreased expression of genes encoding critical hematopoietic transcription factors (Gfi1, Cebpd, Cebpe, and Spi1), cell surface proteins (Csf3r and Gr1), and neutrophil granule proteins (Mpo and Elane). Together, these findings challenge the currently prevailing model that SCN results from mutant ELANE, which triggers endoplasmic reticulum stress, UPR, and apoptosis.


Assuntos
Síndrome Congênita de Insuficiência da Medula Óssea , Regulação Enzimológica da Expressão Gênica , Granulócitos/enzimologia , Elastase de Leucócito , Mutação de Sentido Incorreto , Neutropenia/congênito , Resposta a Proteínas não Dobradas , Substituição de Aminoácidos , Animais , Apoptose , Linhagem Celular Tumoral , Síndrome Congênita de Insuficiência da Medula Óssea/enzimologia , Síndrome Congênita de Insuficiência da Medula Óssea/genética , Estresse do Retículo Endoplasmático , Humanos , Elastase de Leucócito/biossíntese , Elastase de Leucócito/genética , Camundongos , Neutropenia/enzimologia , Neutropenia/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
6.
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
7.
PLoS Comput Biol ; 15(1): e1006664, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30615612

RESUMO

Cancer development is driven by series of events involving mutations, which may become fixed in a tumor via genetic drift and selection. This process usually includes a limited number of driver (advantageous) mutations and a greater number of passenger (neutral or mildly deleterious) mutations. We focus on a real-world leukemia model evolving on the background of a germline mutation. Severe congenital neutropenia (SCN) evolves to secondary myelodysplastic syndrome (sMDS) and/or secondary acute myeloid leukemia (sAML) in 30-40%. The majority of SCN cases are due to a germline ELANE mutation. Acquired mutations in CSF3R occur in >70% sMDS/sAML associated with SCN. Hypotheses underlying our model are: an ELANE mutation causes SCN; CSF3R mutations occur spontaneously at a low rate; in fetal life, hematopoietic stem and progenitor cells expands quickly, resulting in a high probability of several tens to several hundreds of cells with CSF3R truncation mutations; therapeutic granulocyte colony-stimulating factor (G-CSF) administration early in life exerts a strong selective pressure, providing mutants with a growth advantage. Applying population genetics theory, we propose a novel two-phase model of disease development from SCN to sMDS. In Phase 1, hematopoietic tissues expand and produce tens to hundreds of stem cells with the CSF3R truncation mutation. Phase 2 occurs postnatally through adult stages with bone marrow production of granulocyte precursors and positive selection of mutants due to chronic G-CSF therapy to reverse the severe neutropenia. We predict the existence of the pool of cells with the mutated truncated receptor before G-CSF treatment begins. The model does not require increase in mutation rate under G-CSF treatment and agrees with age distribution of sMDS onset and clinical sequencing data.


Assuntos
Modelos Genéticos , Mutação/genética , Síndromes Mielodisplásicas , Neutropenia/congênito , Ciclo Celular/genética , Biologia Computacional , Síndrome Congênita de Insuficiência da Medula Óssea , Neoplasias Hematológicas/genética , Humanos , Elastase de Leucócito/genética , Taxa de Mutação , Síndromes Mielodisplásicas/etiologia , Síndromes Mielodisplásicas/genética , Neutropenia/complicações , Neutropenia/genética , Neutropenia/fisiopatologia , Receptores de Fator Estimulador de Colônias/genética , Seleção Genética/genética
8.
Int J Mol Sci ; 21(13)2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32630050

RESUMO

Inherited bone marrow failure syndromes (IBMFS) are a group of cancer-prone genetic diseases characterized by hypocellular bone marrow with impairment in one or more hematopoietic lineages. The pathogenesis of IBMFS involves mutations in several genes which encode for proteins involved in DNA repair, telomere biology and ribosome biogenesis. The classical IBMFS include Shwachman-Diamond syndrome (SDS), Diamond-Blackfan anemia (DBA), Fanconi anemia (FA), dyskeratosis congenita (DC), and severe congenital neutropenia (SCN). IBMFS are associated with high risk of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML), and solid tumors. Unfortunately, no specific pharmacological therapies have been highly effective for IBMFS. Hematopoietic stem cell transplantation provides a cure for aplastic or myeloid neoplastic complications. However, it does not affect the risk of solid tumors. Since approximately 28% of FA, 24% of SCN, 21% of DBA, 20% of SDS, and 17% of DC patients harbor nonsense mutations in the respective IBMFS-related genes, we discuss the use of the nonsense suppression therapy in these diseases. We recently described the beneficial effect of ataluren, a nonsense suppressor drug, in SDS bone marrow hematopoietic cells ex vivo. A similar approach could be therefore designed for treating other IBMFS. In this review we explain in detail the new generation of nonsense suppressor molecules and their mechanistic roles. Furthermore, we will discuss strengths and limitations of these molecules which are emerging from preclinical and clinical studies. Finally we discuss the state-of-the-art of preclinical and clinical therapeutic studies carried out for IBMFS.


Assuntos
Aminoglicosídeos/uso terapêutico , Códon sem Sentido/efeitos dos fármacos , Síndrome Congênita de Insuficiência da Medula Óssea/terapia , Degradação do RNAm Mediada por Códon sem Sentido/efeitos dos fármacos , Oxidiazóis/uso terapêutico , Aminoglicosídeos/farmacologia , Síndrome Congênita de Insuficiência da Medula Óssea/genética , Humanos , Oxidiazóis/farmacologia
9.
Haematologica ; 104(1): 13-24, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30573510

RESUMO

Inherited bone marrow failure syndromes are experiments of nature characterized by impaired hematopoiesis with cancer and leukemia predisposition. The mutations associated with inherited bone marrow failure syndromes affect fundamental cellular pathways, such as DNA repair, telomere maintenance, or proteostasis. How these disturbed pathways fail to produce sufficient blood cells and lead to leukemogenesis are not understood. The rarity of inherited cytopenias, the paucity of affected primary human hematopoietic cells, and the sometime inadequacy of murine or induced pluripotential stem cell models mean it is difficult to acquire a greater understanding of them. Zebrafish offer a model organism to study gene functions. As vertebrates, zebrafish share with humans many orthologous genes involved in blood disorders. As a model organism, zebrafish provide advantages that include rapid development of transparent embryos, high fecundity (providing large numbers of mutant and normal siblings), and a large collection of mutant and transgenic lines useful for investigating the blood system and other tissues during development. Importantly, recent advances in genomic editing in zebrafish can speedily validate the new genes or novel variants discovered in clinical investigation as causes for marrow failure. Here we review zebrafish as a model organism that phenocopies Fanconi anemia, Diamond-Blackfan anemia, dyskeratosis congenita, Shwachman-Diamond syndrome, congenital amegakaryocytic thrombocytopenia, and severe congenital neutropenia. Two important insights, provided by modeling inherited cytopenias in zebrafish, widen understanding of ribosome biogenesis and TP53 in mediating marrow failure and non-hematologic defects. They suggest that TP53-independent pathways contribute to marrow failure. In addition, zebrafish provide an attractive model organism for drug development.


Assuntos
Transtornos da Insuficiência da Medula Óssea , Doenças Genéticas Inatas , Proteína Supressora de Tumor p53 , Proteínas de Peixe-Zebra , Peixe-Zebra , Animais , Transtornos da Insuficiência da Medula Óssea/genética , Transtornos da Insuficiência da Medula Óssea/metabolismo , Transtornos da Insuficiência da Medula Óssea/patologia , Modelos Animais de Doenças , Doenças Genéticas Inatas/genética , Doenças Genéticas Inatas/metabolismo , Doenças Genéticas Inatas/patologia , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
10.
Pediatr Blood Cancer ; 66(5): e27597, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30604473

RESUMO

Shwachman-Diamond syndrome (SDS) is one of the more common inherited bone marrow failure syndromes, characterized by neutropenia, occasional thrombocytopenia, and anemia. Bone marrow evaluation reveals an increased number of monocytes and mature B cells along with decreased granulocytes. However, little is known about the subpopulations of peripheral blood cells, and few previous publications have been based on a small number of patients. Here, we report a comprehensive immunophenotypic analysis from a cohort of 37 SDS patients who display impairment mostly in the myeloid compartment with a deficiency also in the number of B cells and CD4/CD8 double-negative T cells.


Assuntos
Linfócitos B/imunologia , Doenças da Medula Óssea/sangue , Doenças da Medula Óssea/imunologia , Insuficiência Pancreática Exócrina/sangue , Insuficiência Pancreática Exócrina/imunologia , Imunofenotipagem/métodos , Leucócitos Mononucleares/imunologia , Lipomatose/sangue , Lipomatose/imunologia , Adolescente , Adulto , Doenças da Medula Óssea/patologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Estudos de Coortes , Insuficiência Pancreática Exócrina/patologia , Feminino , Seguimentos , Humanos , Lactente , Lipomatose/patologia , Masculino , Prognóstico , Síndrome de Shwachman-Diamond , Adulto Jovem
11.
J Cell Mol Med ; 22(9): 4274-4282, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29971909

RESUMO

Allogeneic stem cell transplantation is currently the only curative therapy for primary myelofibrosis (MF), while the JAK2 inhibitor, ruxolitinib. Has been approved only for palliation. Other therapies are desperately needed to reverse life-threatening MF. However, the cell(s) and cytokine(s) that promote MF remain unclear. Several reports have demonstrated that captopril, an inhibitor of angiotensin-converting enzyme that blocks the production of angiotensin II (Ang II), mitigates fibrosis in heart, lung, skin and kidney. Here, we show that captopril can mitigate the development of MF in the Gata1low mouse model of primary MF. Gata1low mice were treated with 79 mg/kg/d captopril in the drinking water from 10 to 12 months of age. At 13 months of age, bone marrows were examined for fibrosis, megakaryocytosis and collagen expression; spleens were examined for megakaryocytosis, splenomegaly and collagen expression. Treatment of Gata1low mice with captopril in the drinking water was associated with normalization of the bone marrow cellularity; reduced reticulin fibres, splenomegaly and megakaryocytosis; and decreased collagen expression. Our findings suggest that treating with the ACE inhibitors captopril has a significant benefit in overcoming pathological changes associated with MF.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/farmacologia , Antineoplásicos/farmacologia , Captopril/farmacologia , Fator de Transcrição GATA1/genética , Mielofibrose Primária/tratamento farmacológico , Esplenomegalia/tratamento farmacológico , Administração Oral , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Colágeno/antagonistas & inibidores , Colágeno/genética , Colágeno/metabolismo , Modelos Animais de Doenças , Água Potável/administração & dosagem , Reposicionamento de Medicamentos , Feminino , Fator de Transcrição GATA1/deficiência , Expressão Gênica , Masculino , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Camundongos Knockout , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , Mielofibrose Primária/patologia , Reticulina/antagonistas & inibidores , Reticulina/genética , Reticulina/metabolismo , Esplenomegalia/genética , Esplenomegalia/metabolismo , Esplenomegalia/patologia
14.
Pediatr Blood Cancer ; 64(11)2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28475293

RESUMO

Pathologic variants in TP53 are known risk factors for the development of cancer. We report a 17-year-old male who presented with two primary sarcomas. Germline sequencing revealed a novel TP53 c.672 G>A mutation. Sequencing revealed wild-type TP53 in the parents, and there was no history of cancer in first-degree relatives. This de novo synonymous germline mutation results in a 5' cryptic splice site that is bound by U1, resulting in a shift of the splice site by 5 base pairs. The frame shift results in a truncated protein at residue 246, which disrupts the DNA-binding domain of p53.


Assuntos
DNA de Neoplasias/metabolismo , Mutação em Linhagem Germinativa/genética , Leiomiossarcoma/genética , Neoplasias Pélvicas/genética , Splicing de RNA/genética , Proteína Supressora de Tumor p53/genética , Adolescente , Sítios de Ligação , Evolução Fatal , Humanos , Leiomiossarcoma/metabolismo , Leiomiossarcoma/patologia , Leiomiossarcoma/terapia , Masculino , Neoplasias Pélvicas/metabolismo , Neoplasias Pélvicas/patologia , Neoplasias Pélvicas/terapia , Proteína Supressora de Tumor p53/metabolismo
15.
Pediatr Blood Cancer ; 64(12)2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28708320

RESUMO

Inherited bone marrow failure syndromes (IBMFS) are rare cancer predisposition syndromes with an especially high risk of transformation to myelodysplastic syndrome (MDS) and/or acute myeloid leukemia (AML). We performed a retrospective systematic review of reported MDS/AML arising in the eight most common IBMFS to determine the frequency and outcome of chromosome 7 abnormalities. We identified 738 MDS/AML cases of 4,293 individuals. Monosomy 7 or del (7q) occurred in ∼17%. Greater understanding of the roles played by sequential acquisition of genetic and cytogenetic changes will provide insights into myeloid leukemogenesis and improve the surveillance and hopefully outcomes for individuals with IBMFS.


Assuntos
Anemia Aplástica/genética , Doenças da Medula Óssea/genética , Deleção Cromossômica , Hemoglobinúria Paroxística/genética , Adolescente , Adulto , Transtornos da Insuficiência da Medula Óssea , Criança , Pré-Escolar , Cromossomos Humanos Par 7 , Humanos , Lactente , Recém-Nascido , Estudos Retrospectivos , Adulto Jovem
16.
J Immunol ; 195(4): 1341-9, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26254266

RESUMO

G-CSF and GM-CSF are used widely to promote the production of granulocytes or APCs. The U.S. Food and Drug Administration approved G-CSF (filgrastim) for the treatment of congenital and acquired neutropenias and for mobilization of peripheral hematopoietic progenitor cells for stem cell transplantation. A polyethylene glycol-modified form of G-CSF is approved for the treatment of neutropenias. Clinically significant neutropenia, rendering an individual immunocompromised, occurs when their number is <1500/µl. Current guidelines recommend their use when the risk for febrile neutropenia is >20%. GM-CSF (sargramostim) is approved for neutropenia associated with stem cell transplantation. Because of its promotion of APC function, GM-CSF is being evaluated as an immunostimulatory adjuvant in a number of clinical trials. More than 20 million persons have benefited worldwide, and >$5 billion in sales occur annually in the United States.


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neutropenia/tratamento farmacológico , Neutropenia/etiologia , Fator Estimulador de Colônias de Granulócitos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Neutropenia/metabolismo , Transdução de Sinais
17.
Blood ; 123(4): 562-9, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24184683

RESUMO

Hereditary neutropenia is usually caused by heterozygous germline mutations in the ELANE gene encoding neutrophil elastase (NE). How mutations cause disease remains uncertain, but two hypotheses have been proposed. In one, ELANE mutations lead to mislocalization of NE. In the other, ELANE mutations disturb protein folding, inducing an unfolded protein response in the endoplasmic reticulum (ER). In this study, we describe new types of mutations that disrupt the translational start site. At first glance, they should block translation and are incompatible with either the mislocalization or misfolding hypotheses, which require mutant protein for pathogenicity. We find that start-site mutations, instead, force translation from downstream in-frame initiation codons, yielding amino-terminally truncated isoforms lacking ER-localizing (pre) and zymogen-maintaining (pro) sequences, yet retain essential catalytic residues. Patient-derived induced pluripotent stem cells recapitulate hematopoietic and molecular phenotypes. Expression of the amino-terminally deleted isoforms in vitro reduces myeloid cell clonogenic capacity. We define an internal ribosome entry site (IRES) within ELANE and demonstrate that adjacent mutations modulate IRES activity, independently of protein-coding sequence alterations. Some ELANE mutations, therefore, appear to cause neutropenia via the production of amino-terminally deleted NE isoforms rather than by altering the coding sequence of the full-length protein.


Assuntos
Elastase de Leucócito/genética , Elastase de Leucócito/metabolismo , Mutação , Neutropenia/metabolismo , Biossíntese de Proteínas , Apoptose , Códon , Análise Mutacional de DNA , Retículo Endoplasmático/metabolismo , Células HL-60 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neutrófilos/citologia , Fenótipo , Desnaturação Proteica , Dobramento de Proteína , Isoformas de Proteínas/metabolismo , Células U937
19.
Nat Rev Cancer ; 7(2): 118-29, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17251918

RESUMO

The prevalence of patients with myelodysplastic syndromes (MDS) is increasing owing to an ageing population and increased awareness of these diseases. MDS represent many different conditions, not just a single disease, that are grouped together by several clinical characteristics. A striking feature of MDS is genetic instability, and a large proportion of cases result in acute myeloid leukaemia (AML). We Review three emerging principles of MDS biology: stem-cell dysfunction and the overlap with AML, genetic instability and the deregulation of apoptosis, in the context of inherited bone marrow-failure syndromes, and treatment-related MDS and AML.


Assuntos
Síndromes Mielodisplásicas/patologia , Células-Tronco/patologia , Antineoplásicos/efeitos adversos , Apoptose , Humanos , Síndromes Mielodisplásicas/induzido quimicamente , Síndromes Mielodisplásicas/epidemiologia , Transdução de Sinais
20.
Semin Cell Dev Biol ; 24(4): 280-6, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23384583

RESUMO

The Bin-Amphiphysin-Rvs (BAR) domain family of proteins includes groups which promote positive (classical BAR, N-BAR, and F-BAR) and negative (I-BAR) membrane deformation. Of these groups, the F-BAR subfamily is the most diverse in its biochemical properties. F-BAR domain proteins dimerize to form a tight scaffold about the membrane. The F-BAR domain provides a banana-shaped, alpha-helical structure that senses membrane curvature. Different types of F-BAR domain proteins contain tyrosine kinase or GTPase activities; some interact with phosphatases and RhoGTPases. Most possess an SH3 domain that facilitates the recruitment and activation of WASP/N-WASP. Thus, F-BAR domain proteins affect remodeling of both membrane and the actin cytoskeleton. The purpose of this review is to highlight the role of F-BAR proteins in coupling WASP/N-WASP to cytoskeletal remodeling. A role for F-BAR/WASP interaction in human diseases affecting nervous, blood, and neoplastic tissues is discussed.


Assuntos
Citoesqueleto/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Humanos , Domínios e Motivos de Interação entre Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA