Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 182(6): 1401-1418.e18, 2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32810439

RESUMO

Blood myeloid cells are known to be dysregulated in coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2. It is unknown whether the innate myeloid response differs with disease severity and whether markers of innate immunity discriminate high-risk patients. Thus, we performed high-dimensional flow cytometry and single-cell RNA sequencing of COVID-19 patient peripheral blood cells and detected disappearance of non-classical CD14LowCD16High monocytes, accumulation of HLA-DRLow classical monocytes (Human Leukocyte Antigen - DR isotype), and release of massive amounts of calprotectin (S100A8/S100A9) in severe cases. Immature CD10LowCD101-CXCR4+/- neutrophils with an immunosuppressive profile accumulated in the blood and lungs, suggesting emergency myelopoiesis. Finally, we show that calprotectin plasma level and a routine flow cytometry assay detecting decreased frequencies of non-classical monocytes could discriminate patients who develop a severe form of COVID-19, suggesting a predictive value that deserves prospective evaluation.


Assuntos
Infecções por Coronavirus , Coronavirus , Pandemias , Pneumonia Viral , Betacoronavirus , COVID-19 , Citometria de Fluxo , Humanos , Complexo Antígeno L1 Leucocitário , Monócitos , Células Mieloides , Estudos Prospectivos , SARS-CoV-2
2.
Blood ; 138(17): 1603-1614, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34115825

RESUMO

EZH2, the enzymatic component of PRC2, has been identified as a key factor in hematopoiesis. EZH2 loss-of-function mutations have been found in myeloproliferative neoplasms, particularly in myelofibrosis, but the precise function of EZH2 in megakaryopoiesis is not fully delineated. Here, we show that EZH2 inhibition by small molecules and short hairpin RNA induces megakaryocyte (MK) commitment by accelerating lineage marker acquisition without change in proliferation. Later in differentiation, EZH2 inhibition blocks proliferation and polyploidization and decreases proplatelet formation. EZH2 inhibitors similarly reduce MK polyploidization and proplatelet formation in vitro and platelet levels in vivo in a JAK2V617F background. In transcriptome profiling, the defect in proplatelet formation was associated with an aberrant actin cytoskeleton regulation pathway, whereas polyploidization was associated with an inhibition of expression of genes involved in DNA replication and repair and an upregulation of cyclin-dependent kinase inhibitors, particularly CDKN1A and CDKN2D. The knockdown of CDKN1A and to a lesser extent CDKN2D could partially rescue the percentage of polyploid MKs. Moreover, H3K27me3 and EZH2 chromatin immunoprecipitation assays revealed that CDKN1A is a direct EZH2 target and CDKN2D expression is not directly regulated by EZH2, suggesting that EZH2 controls MK polyploidization directly through CDKN1A and indirectly through CDKN2D.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Megacariócitos/citologia , Trombopoese , Animais , Plaquetas/citologia , Plaquetas/metabolismo , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Megacariócitos/metabolismo , Camundongos , Interferência de RNA , Transcriptoma
3.
Blood ; 138(22): 2231-2243, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34407546

RESUMO

Classical BCR-ABL-negative myeloproliferative neoplasms (MPNs) are clonal disorders of hematopoietic stem cells (HSCs) caused mainly by recurrent mutations in genes encoding JAK2 (JAK2), calreticulin (CALR), or the thrombopoietin receptor (MPL). Interferon α (IFNα) has demonstrated some efficacy in inducing molecular remission in MPNs. To determine factors that influence molecular response rate, we evaluated the long-term molecular efficacy of IFNα in patients with MPN by monitoring the fate of cells carrying driver mutations in a prospective observational and longitudinal study of 48 patients over more than 5 years. We measured the clonal architecture of early and late hematopoietic progenitors (84 845 measurements) and the global variant allele frequency in mature cells (409 measurements) several times per year. Using mathematical modeling and hierarchical Bayesian inference, we further inferred the dynamics of IFNα-targeted mutated HSCs. Our data support the hypothesis that IFNα targets JAK2V617F HSCs by inducing their exit from quiescence and differentiation into progenitors. Our observations indicate that treatment efficacy is higher in homozygous than heterozygous JAK2V617F HSCs and increases with high IFNα dose in heterozygous JAK2V617F HSCs. We also found that the molecular responses of CALRm HSCs to IFNα were heterogeneous, varying between type 1 and type 2 CALRm, and a high dose of IFNα correlates with worse outcomes. Our work indicates that the long-term molecular efficacy of IFNα implies an HSC exhaustion mechanism and depends on both the driver mutation type and IFNα dose.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Interferon-alfa/uso terapêutico , Mutação/efeitos dos fármacos , Transtornos Mieloproliferativos/tratamento farmacológico , Calreticulina/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Fatores Imunológicos/farmacologia , Interferon-alfa/farmacologia , Janus Quinase 2/genética , Estudos Longitudinais , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Estudos Prospectivos , Receptores de Trombopoetina/genética , Células Tumorais Cultivadas
4.
Haematologica ; 105(1): 112-123, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31048357

RESUMO

The functional diversity of cells that compose myeloid malignancies, i.e., the respective roles of genetic and epigenetic heterogeneity in this diversity, remains poorly understood. This question is addressed in chronic myelomonocytic leukemia, a myeloid neoplasm in which clinical diversity contrasts with limited genetic heterogeneity. To generate induced pluripotent stem cell clones, we reprogrammed CD34+ cells collected from a patient with a chronic myelomonocytic leukemia in which whole exome sequencing of peripheral blood monocyte DNA had identified 12 gene mutations, including a mutation in KDM6A and two heterozygous mutations in TET2 in the founding clone and a secondary KRAS(G12D) mutation. CD34+ cells from an age-matched healthy donor were also reprogrammed. We captured a part of the genetic heterogeneity observed in the patient, i.e. we analyzed five clones with two genetic backgrounds, without and with the KRAS(G12D) mutation. Hematopoietic differentiation of these clones recapitulated the main features of the patient's disease, including overproduction of granulomonocytes and dysmegakaryopoiesis. These analyses also disclosed significant discrepancies in the behavior of hematopoietic cells derived from induced pluripotent stem cell clones with similar genetic background, correlating with limited epigenetic changes. These analyses suggest that, beyond the coding mutations, several levels of intraclonal heterogeneity may participate in the yet unexplained clinical heterogeneity of the disease.


Assuntos
Leucemia Mielomonocítica Crônica , Leucemia Mielomonocítica Juvenil , Transtornos Mieloproliferativos , Humanos , Leucemia Mielomonocítica Crônica/genética , Leucemia Mielomonocítica Juvenil/genética , Mutação , Sequenciamento do Exoma
5.
Am J Hematol ; 93(2): 195-204, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29090484

RESUMO

Rare gain-of-function mutations within the ITGA2B or ITGB3 genes have been recognized to cause macrothrombocytopenia (MTP). Here we report three new families with autosomal dominant (AD) MTP, two harboring the same mutation of ITGA2B, αIIbR995W, and a third family with an ITGB3 mutation, ß3D723H. In silico analysis shows how the two mutated amino acids directly modify the salt bridge linking the intra-cytoplasmic part of αIIb to ß3 of the integrin αIIbß3. For all affected patients, the bleeding syndrome and MTP was mild to moderate. Platelet aggregation tended to be reduced but not absent. Electron microscopy associated with a morphometric analysis revealed large round platelets; a feature being the presence of abnormal large α-granules with some giant forms showing signs of fusion. Analysis of the maturation and development of megakaryocytes reveal no defect in their early maturation but abnormal proplatelet formation was observed with increased size of the tips. Interestingly, this study revealed that in addition to the classical phenotype of patients with αIIbß3 intracytoplasmic mutations there is an abnormal maturation of α-granules. It is now necessary to determine if this feature is a characteristic of all mutations disturbing the αIIb R995/ß3 D723 salt bridge.


Assuntos
Grânulos Citoplasmáticos/patologia , Integrina alfa2/genética , Integrina beta3/genética , Trombocitopenia/etiologia , Plaquetas/ultraestrutura , Simulação por Computador , Família , Humanos , Megacariócitos , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química
6.
Blood ; 125(23): 3618-26, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-25852055

RESUMO

Chronic myelomonocytic leukemia (CMML) is a myelodysplastic syndrome/ myeloproliferative neoplasm whose diagnosis is currently based on the elevation of peripheral blood monocytes to >1 × 10(9)/L, measured for ≥3 months. Diagnosis can be ambiguous; for example, with prefibrotic myelofibrosis or reactive monocytosis. We set up a multiparameter flow cytometry assay to distinguish CD14(+)/CD16(-) classical from CD14(+)/CD16(+) intermediate and CD14(low)/CD16(+) nonclassical monocyte subsets in peripheral blood mononucleated cells and in total blood samples. Compared with healthy donors and patients with reactive monocytosis or another hematologic malignancy, CMML patients demonstrate a characteristic increase in the fraction of CD14(+)/CD16(-) cells (cutoff value, 94.0%). The associated specificity and sensitivity values were 95.1% and 90.6% in the learning cohort (175 samples) and 94.1% and 91.9% in the validation cohort (307 samples), respectively. The accumulation of classical monocytes, which demonstrate a distinct gene expression pattern, is independent of the mutational background. Importantly, this increase disappears in patients who respond to hypomethylating agents. We conclude that an increase in the fraction of classical monocytes to >94.0% of total monocytes is a highly sensitive and specific diagnostic marker that rapidly and accurately distinguishes CMML from confounding diagnoses.


Assuntos
Citometria de Fluxo/métodos , Leucemia Mielogênica Crônica BCR-ABL Positiva/sangue , Leucemia Mielogênica Crônica BCR-ABL Positiva/diagnóstico , Receptores de Lipopolissacarídeos/sangue , Monócitos , Receptores de IgG/sangue , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/mortalidade , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Monócitos/patologia , Sensibilidade e Especificidade
7.
Hum Mol Genet ; 23(1): 171-81, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23966205

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most prevalent adult muscular dystrophies. The common clinical signs usually appear during the second decade of life but when the first molecular dysregulations occur is still unknown. Our aim was to determine whether molecular dysregulations can be identified during FSHD fetal muscle development. We compared muscle biopsies derived from FSHD1 fetuses and the cells derived from some of these biopsies with biopsies and cells derived from control fetuses. We mainly focus on DUX4 isoform expression because the expression of DUX4 has been confirmed in both FSHD cells and biopsies by several laboratories. We measured DUX4 isoform expression by using qRT-PCR in fetal FSHD1 myotubes treated or not with an shRNA directed against DUX4 mRNA. We also analyzed DUX4 downstream target gene expression in myotubes and fetal or adult FSHD1 and control quadriceps biopsies. We show that both DUX4-FL isoforms are already expressed in FSHD1 myotubes. Interestingly, DUX4-FL expression level is much lower in trapezius than in quadriceps myotubes, which is confirmed by the level of expression of DUX4 downstream genes. We observed that TRIM43 and MBD3L2 are already overexpressed in FSHD1 fetal quadriceps biopsies, at similar levels to those observed in adult FSHD1 quadriceps biopsies. These results indicate that molecular markers of the disease are already expressed during fetal life, thus opening a new field of investigation for mechanisms leading to FSHD.


Assuntos
Proteínas de Homeodomínio/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/embriologia , Distrofia Muscular Facioescapuloumeral/genética , Adulto , Células Cultivadas , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Desenvolvimento Muscular , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Facioescapuloumeral/patologia , Isoformas de Proteínas/genética , Músculo Quadríceps/embriologia , Músculo Quadríceps/metabolismo , Isoformas de RNA/genética , Isoformas de RNA/metabolismo , RNA Interferente Pequeno/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Músculos Superficiais do Dorso/embriologia , Músculos Superficiais do Dorso/metabolismo
8.
Ann Neurol ; 78(3): 387-400, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26018399

RESUMO

OBJECTIVE: Facioscapulohumeral muscular dystrophy (FSHD) is linked to either contraction of D4Z4 repeats on chromosome 4 or to mutations in the SMCHD1 gene, both of which result in the aberrant expression of the transcription factor DUX4. However, it is still difficult to correlate these genotypes with the phenotypes observed in patients. Because we have recently shown that mice with disrupted Fat1 functions exhibit FSHD-like phenotypes, we have investigated the expression of the human FAT1 gene in FSHD. METHODS: We first analyzed FAT1 expression in FSHD adult muscles and determined whether FAT1 expression was driven by DUX4. We next determined FAT1 expression levels in 64 muscles isolated from 16 control fetuses. These data were further complemented with analysis of Fat1 expression in developing mouse embryos. RESULTS: We demonstrated that FAT1 expression is independent of DUX4. Moreover, we observed that (1) in control fetal human biopsies or in developing mouse embryos, FAT1 is expressed at lower levels in muscles that are affected at early stages of FSHD progression than in muscles that are affected later or are nonaffected; and (2) in adult muscle biopsies, FAT1 expression is lower in FSHD muscles compared to control muscles. INTERPRETATION: We propose a revised model for FSHD in which FAT1 levels might play a role in determining which muscles will exhibit early and late disease onset, whereas DUX4 may worsen the muscle phenotype.


Assuntos
Caderinas/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Distrofia Muscular Facioescapuloumeral/diagnóstico , Distrofia Muscular Facioescapuloumeral/metabolismo , Músculo Quadríceps/metabolismo , Músculo Quadríceps/patologia , Adulto , Animais , Células Cultivadas , Feminino , Feto , Humanos , Masculino , Camundongos , Músculo Esquelético/embriologia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Quadríceps/embriologia
9.
Blood ; 124(26): 3967-77, 2014 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-25298036

RESUMO

Megakaryocytes are highly specialized precursor cells that produce platelets via cytoplasmic extensions called proplatelets. Proplatelet formation (PPF) requires profound changes in microtubule and actin organization. In this work, we demonstrated that DIAPH1 (mDia1), a mammalian homolog of Drosophila diaphanous that works as an effector of the small GTPase Rho, negatively regulates PPF by controlling the dynamics of the actin and microtubule cytoskeletons. Moreover, we showed that inhibition of both DIAPH1 and the Rho-associated protein kinase (Rock)/myosin pathway increased PPF via coordination of both cytoskeletons. We provide evidence that 2 major effectors of the Rho GTPase pathway (DIAPH1 and Rock/myosin II) are involved not only in Rho-mediated stress fibers assembly, but also in the regulation of microtubule stability and dynamics during PPF.


Assuntos
Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Citoesqueleto/metabolismo , Megacariócitos/citologia , Microtúbulos/metabolismo , Antígenos CD34/metabolismo , Plaquetas/citologia , Plaquetas/metabolismo , Diferenciação Celular , Clonagem Molecular , Forminas , GTP Fosfo-Hidrolases/metabolismo , Humanos , Lentivirus/genética , Miosina Tipo II/metabolismo , RNA Interferente Pequeno/metabolismo , Trombopoetina/química , Tubulina (Proteína)/química
10.
Blood ; 124(13): 2104-15, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25143485

RESUMO

Megakaryopoiesis is a 2-step differentiation process, regulated by thrombopoietin (TPO), on binding to its cognate receptor myeloproliferative leukemia (MPL). This receptor associates with intracytoplasmic tyrosine kinases, essentially janus kinase 2 (JAK2), which regulates MPL stability and cell-surface expression, and mediates TPO-induced signal transduction. We demonstrate that JAK2 and MPL mediate TPO-induced proliferation arrest and megakaryocytic differentiation of the human megakaryoblastic leukemia cell line UT7-MPL. A decrease in JAK2 or MPL protein expression, and JAK2 chemical inhibition, suppress this antiproliferative action of TPO. The expression of JAK2 and MPL, which progressively increases along normal human megakaryopoiesis, is decreased in platelets of patients diagnosed with JAK2- or MPL-mutated essential thrombocytemia and primary myelofibrosis, 2 myeloproliferative neoplasms in which megakaryocytes (MKs) proliferate excessively. Finally, low doses of JAK2 chemical inhibitors are shown to induce a paradoxical increase in MK production, both in vitro and in vivo. We propose that JAK2 and MPL expression levels regulate megakaryocytic proliferation vs differentiation in both normal and pathological conditions, and that JAK2 chemical inhibitors could promote a paradoxical thrombocytosis when used at suboptimal doses.


Assuntos
Autoantígenos/metabolismo , Diferenciação Celular , Iodeto Peroxidase/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Janus Quinase 2/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Receptores de Trombopoetina/metabolismo , Animais , Autoantígenos/genética , Plaquetas/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Expressão Gênica , Humanos , Iodeto Peroxidase/genética , Proteínas de Ligação ao Ferro/genética , Janus Quinase 2/genética , Camundongos , Fenótipo , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , RNA Interferente Pequeno/genética , Receptores de Trombopoetina/genética , Trombocitemia Essencial/genética , Trombocitemia Essencial/metabolismo
11.
Blood ; 124(16): 2554-63, 2014 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-25061177

RESUMO

Macrothrombocytopenias are the most important subgroup of inherited thrombocytopenias. This subgroup is particularly heterogeneous because the affected genes are involved in various functions such as cell signaling, cytoskeleton organization, and gene expression. Herein we describe the clinical and hematological features of a consanguineous family with a severe autosomal recessive macrothrombocytopenia associated with a thrombocytopathy inducing a bleeding tendency in the homozygous mutated patients. Platelet activation and cytoskeleton reorganization were impaired in these homozygous patients. Exome sequencing identified a c.222C>G mutation (missense p.74Ile>Met) in PRKACG, a gene encoding the γ-catalytic subunit of the cyclic adenosine monophosphate-dependent protein kinase, the mutated allele cosegregating with the macrothrombocytopenia. We demonstrate that the p.74Ile>Met PRKACG mutation is associated with a marked defect in proplatelet formation and a low level in filamin A in megakaryocytes (MKs). The defect in proplatelet formation was rescued in vitro by lentiviral vector-mediated overexpression of wild-type PRKACG in patient MKs. We thus conclude that PRKACG is a new central actor in platelet biogenesis and a new gene involved in inherited thrombocytopenia with giant platelets associated with a thrombocytopathy.


Assuntos
Plaquetas/patologia , Subunidades Catalíticas da Proteína Quinase Dependente de AMP Cíclico/genética , Mutação em Linhagem Germinativa , Megacariócitos/patologia , Trombocitopenia/genética , Adulto , Plaquetas/metabolismo , Pré-Escolar , Citoesqueleto/genética , Citoesqueleto/patologia , Humanos , Lactente , Masculino , Megacariócitos/metabolismo , Linhagem , Contagem de Plaquetas , Trombocitopenia/complicações , Trombocitopenia/patologia , Adulto Jovem
12.
Haematologica ; 101(12): 1469-1478, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27515249

RESUMO

Megakaryocytes are naturally polyploid cells that increase their ploidy by endomitosis. However, very little is known regarding the mechanism by which they escape the tetraploid checkpoint to become polyploid. Recently, it has been shown that the tetraploid checkpoint was regulated by the Hippo-p53 pathway in response to a downregulation of Rho activity. We therefore analyzed the role of Hippo-p53 pathway in the regulation of human megakaryocyte polyploidy. Our results revealed that Hippo-p53 signaling pathway proteins are present and are functional in megakaryocytes. Although this pathway responds to the genotoxic stress agent etoposide, it is not activated in tetraploid or polyploid megakaryocytes. Furthermore, Hippo pathway was observed to be uncoupled from Rho activity. Additionally, polyploid megakaryocytes showed increased expression of YAP target genes when compared to diploid and tetraploid megakaryocytes. Although p53 knockdown increased both modal ploidy and proplatelet formation in megakaryocytes, YAP knockdown caused no significant change in ploidy while moderately affecting proplatelet formation. Interestingly, YAP knockdown reduced the mitochondrial mass in polyploid megakaryocytes and decreased expression of PGC1α, an important mitochondrial biogenesis regulator. Thus, the Hippo pathway is functional in megakaryocytes, but is not induced by tetraploidy. Additionally, YAP regulates the mitochondrial mass in polyploid megakaryocytes.


Assuntos
Diferenciação Celular , Megacariócitos/citologia , Megacariócitos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Tetraploidia , Proteínas rho de Ligação ao GTP/metabolismo , Biomarcadores , Plaquetas/citologia , Plaquetas/metabolismo , Proteínas de Ciclo Celular , Diferenciação Celular/genética , Expressão Gênica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Via de Sinalização Hippo , Humanos , Modelos Biológicos , Proteínas Nucleares/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Poliploidia , Proteínas Serina-Treonina Quinases/genética , Trombopoese/genética , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas rho de Ligação ao GTP/genética
13.
Blood ; 121(12): 2186-98, 2013 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-23319568

RESUMO

Genomic studies in chronic myeloid malignancies, including myeloproliferative neoplasms (MPN), myelodysplastic syndromes (MDS), and MPN/MDS, have identified common mutations in genes encoding signaling, epigenetic, transcription, and splicing factors. In the present study, we interrogated the clonal architecture by mutation-specific discrimination analysis of single-cell-derived colonies in 28 patients with chronic myelomonocytic leukemias (CMML), the most frequent MPN/MDS. This analysis reveals a linear acquisition of the studied mutations with limited branching through loss of heterozygosity. Serial analysis of untreated and treated samples demonstrates a dynamic architecture on which most current therapeutic approaches have limited effects. The main disease characteristics are early clonal dominance, arising at the CD34(+)/CD38(-) stage of hematopoiesis, and granulomonocytic differentiation skewing of multipotent and common myeloid progenitors. Comparison of clonal expansions of TET2 mutations in MDS, MPN, and CMML, together with functional invalidation of TET2 in sorted progenitors, suggests a causative link between early clonal dominance and skewed granulomonocytic differentiation. Altogether, early clonal dominance may distinguish CMML from other chronic myeloid neoplasms with similar gene mutations.


Assuntos
Evolução Clonal/genética , Leucemia Mielomonocítica Crônica/genética , Leucemia Mielomonocítica Crônica/patologia , Idoso , Idoso de 80 Anos ou mais , Estudos de Casos e Controles , Diferenciação Celular/genética , Evolução Clonal/imunologia , Estudos de Coortes , Feminino , Hematopoese/genética , Hematopoese/imunologia , Humanos , Leucemia Mielomonocítica Crônica/imunologia , Perda de Heterozigosidade , Masculino , Pessoa de Meia-Idade , Mutação/fisiologia , Taxa de Mutação , Células Mieloides/metabolismo , Células Mieloides/fisiologia
14.
Blood ; 120(13): 2708-18, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22898599

RESUMO

FPD/AML is a familial platelet disorder characterized by platelet defects, predisposition to acute myelogenous leukemia (AML) and germ-line heterozygous RUNX1 alterations. Here we studied the in vitro megakaryopoiesis of 3 FPD/AML pedigrees. A 60% to 80% decrease in the output of megakaryocytes (MKs) from CD34(+) was observed. MK ploidy level was low and mature MKs displayed a major defect in proplatelet formation. To explain these defects, we focused on myosin II expression as RUNX1 has been shown to regulate MYL9 and MYH10 in an inverse way. In FPD/AML MKs, expression of MYL9 and MYH9 was decreased, whereas MYH10 expression was increased and the MYH10 protein was still present in the cytoplasm of mature MKs. Myosin II activity inhibition by blebbistatin rescued the ploidy defect of FPD/AML MKs. Finally, we demonstrate that MYH9 is a direct target of RUNX1 by chromatin immunoprecipitation and luciferase assays and we identified new RUNX1 binding sites in the MYL9 promoter region. Together, these results demonstrate that the defects in megakaryopoiesis observed in FPD/AML are, in part, related to a deregulation of myosin IIA and IIB expression leading to both a defect in ploidization and proplatelet formation.


Assuntos
Transtornos Plaquetários/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Leucemia Mieloide Aguda/patologia , Megacariócitos/patologia , Mutação/genética , Miosina não Muscular Tipo IIA/metabolismo , Miosina não Muscular Tipo IIB/metabolismo , Transtornos Plaquetários/genética , Transtornos Plaquetários/metabolismo , Western Blotting , Imunoprecipitação da Cromatina , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Luciferases/metabolismo , Masculino , Miosina não Muscular Tipo IIA/genética , Miosina não Muscular Tipo IIB/genética , Linhagem , Ploidias , Prognóstico , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Blood ; 117(1): 246-9, 2011 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-20944070

RESUMO

Transforming growth factor-ß1 (TGF-ß1) is the most important cytokine involved in the promotion of myelofibrosis. Mechanisms leading to its local activation in the bone marrow environment remain unclear. As a recent study has highlighted the role of thrombospondin-1 (TSP-1) in platelet-derived TGF-ß1 activation, we investigated the role of TSP-1 in the TPO(high) murine model of myelofibrosis. Two groups of engrafted mice, WT TPO(high) and Tsp-1-null TPO(high), were constituted. All mice developed a similar myeloproliferative syndrome and an increase in total TGF-ß1 levels in the plasma and in extracellular fluids of marrow and spleen. Surprisingly, we were able to detect the active form of TGF-ß1 in Tsp-1-null TPO(high) mice. Accordingly, these mice developed marrow and spleen fibrosis, with intriguingly a higher grade than in WT TPO(high) mice. Our results show that TSP-1 is not the major activator of TGF-ß1 in TPO-induced myelofibrosis, suggesting the contribution of another mechanism in the megakaryocyte/platelet compartment.


Assuntos
Mielofibrose Primária/induzido quimicamente , Mielofibrose Primária/patologia , Trombopoetina/efeitos adversos , Trombospondina 1/fisiologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Feminino , Masculino , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mielofibrose Primária/metabolismo , Baço/metabolismo , Baço/patologia
17.
Blood ; 118(5): 1264-73, 2011 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-21653327

RESUMO

The NOTCH signaling pathway is implicated in a broad range of developmental processes, including cell fate decisions. However, the molecular basis for its role at the different steps of stem cell lineage commitment is unclear. We recently identified the NOTCH signaling pathway as a positive regulator of megakaryocyte lineage specification during hematopoiesis, but the developmental pathways that allow hematopoietic stem cell differentiation into the erythro-megakaryocytic lineages remain controversial. Here, we investigated the role of downstream mediators of NOTCH during megakaryopoiesis and report crosstalk between the NOTCH and PI3K/AKT pathways. We demonstrate the inhibitory role of phosphatase with tensin homolog and Forkhead Box class O factors on megakaryopoiesis in vivo. Finally, our data annotate developmental mechanisms in the hematopoietic system that enable a decision to be made either at the hematopoietic stem cell or the committed progenitor level to commit to the megakaryocyte lineage, supporting the existence of 2 distinct developmental pathways.


Assuntos
Diferenciação Celular , Linhagem da Célula/fisiologia , Megacariócitos/fisiologia , Proteína Oncogênica v-akt/metabolismo , Receptores Notch/metabolismo , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem da Célula/genética , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Fatores de Transcrição Forkhead/fisiologia , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/fisiologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/fisiologia , Receptor Cross-Talk/fisiologia , Receptores Notch/genética , Receptores Notch/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Trombopoese/genética
18.
Blood ; 118(22): 5928-37, 2011 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-21960593

RESUMO

Filaminopathies A caused by mutations in the X-linked FLNA gene are responsible for a wide spectrum of rare diseases including 2 main phenotypes, the X-linked dominant form of periventricular nodular heterotopia (FLNA-PVNH) and the otopalatodigital syndrome spectrum of disorders. In platelets, filamin A (FLNa) tethers the principal receptors ensuring the platelet-vessel wall interaction, glycoprotein Ibα and integrin αIIbß3, to the underlying cytoskeleton. Hemorrhage, coagulopathy, and thrombocytopenia are mentioned in several reports on patients with FLNA-PVNH. Abnormal platelet morphology in 2 patients with FLNA-PVNH prompted us to examine a third patient with similar platelet morphology previously diagnosed with immunologic thrombocytopenic purpura. Her enlarged platelets showed signs of FLNa degradation in Western blotting, and a heterozygous missense mutation in FLNA was detected. An irregular distribution of FLNa within the total platelet population was shown by confocal microscopy for all 3 patients. In vitro megakaryocyte cultures showed an abnormal differentiation, including an irregular distribution of FLNa with a frayed aspect, the presence of enlarged α-granules, and an abnormal fragmentation of the cytoplasm. Mutations in FLNA may represent an unrecognized cause of macrothrombocytopenia with an altered platelet production and a modified platelet-vessel wall interaction.


Assuntos
Proteínas Contráteis/genética , Proteínas dos Microfilamentos/genética , Mutação , Trombocitopenia/classificação , Trombocitopenia/genética , Idoso , Células Cultivadas , Feminino , Filaminas , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Mutação/fisiologia , Contagem de Plaquetas , Síndrome , Trombocitopenia/sangue , Trombocitopenia/diagnóstico
19.
Leukemia ; 37(6): 1287-1297, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37100881

RESUMO

Heterozygous mutation targeting proline 95 in Serine/Arginine-rich Splicing Factor 2 (SRSF2) is associated with V617F mutation in Janus Activated Kinase 2 (JAK2) in some myeloproliferative neoplasms (MPNs), most commonly primary myelofibrosis. To explore the interaction of Srsf2P95H with Jak2V617F, we generated Cre-inducible knock-in mice expressing these mutants under control of the stem cell leukemia (Scl) gene promoter. In transplantation experiments, Srsf2P95H unexpectedly delayed myelofibrosis induced by Jak2V617F and decreased TGFß1 serum level. Srsf2P95H reduced the competitiveness of transplanted Jak2V617F hematopoietic stem cells while preventing their exhaustion. RNA sequencing of sorted megakaryocytes identified an increased number of splicing events when the two mutations were combined. Focusing on JAK/STAT pathway, Jak2 exon 14 skipping was promoted by Srsf2P95H, an event detected in patients with JAK2V617F and SRSF2P95 co-mutation. The skipping event generates a truncated inactive JAK2 protein. Accordingly, Srsf2P95H delays myelofibrosis induced by the thrombopoietin receptor agonist Romiplostim in Jak2 wild-type animals. These results unveil JAK2 exon 14 skipping promotion as a strategy to reduce JAK/STAT signaling in pathological conditions.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Transtornos Mieloproliferativos , Mielofibrose Primária , Animais , Camundongos , Janus Quinase 2/genética , Janus Quinases/genética , Mutação , Transtornos Mieloproliferativos/genética , Mielofibrose Primária/genética , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Fatores de Transcrição STAT/genética
20.
JMIR Form Res ; 6(2): e32230, 2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35225812

RESUMO

BACKGROUND: Computed tomography pulmonary angiography (CTPA) is frequently used in the emergency department (ED) for the diagnosis of pulmonary embolism (PE), while posing risk for contrast-induced nephropathy and radiation-induced malignancy. OBJECTIVE: We aimed to create an automated process to calculate the Wells score for pulmonary embolism for patients in the ED, which could potentially reduce unnecessary CTPA testing. METHODS: We designed an automated process using electronic health records data elements, including using a combinatorial keyword search method to query free-text fields, and calculated automated Wells scores for a sample of all adult ED encounters that resulted in a CTPA study for PE at 2 tertiary care hospitals in New York, over a 2-month period. To validate the automated process, the scores were compared to those derived from a 2-clinician chart review. RESULTS: A total of 202 ED encounters resulted in a completed CTPA to form the retrospective study cohort. Patients classified as "PE likely" by the automated process (126/202, 62%) had a PE prevalence of 15.9%, whereas those classified as "PE unlikely" (76/202, 38%; Wells score >4) had a PE prevalence of 7.9%. With respect to classification of the patient as "PE likely," the automated process achieved an accuracy of 92.1% when compared with the chart review, with sensitivity, specificity, positive predictive value, and negative predictive value of 93%, 90.5%, 94.4%, and 88.2%, respectively. CONCLUSIONS: This was a successful development and validation of an automated process using electronic health records data elements, including free-text fields, to classify risk for PE in ED visits.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA