Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Br J Haematol ; 187(2): 163-173, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31222725

RESUMO

We established mutated and non-mutated induced pluripotent stem cell (iPSC) clones from a patient with PTPN11 (c.226G>A)-mutated juvenile myelomonocytic leukaemia (JMML). Both types of iPSCs fulfilled the quality criteria. Mutated iPSC colonies generated significantly more CD34+ and CD34+ CD45+ cells compared to non-mutated iPSC colonies in a culture coated with irradiated AGM-S3 cells to which four growth factors were added sequentially or simultaneously. The haematopoietic differentiation potential of non-mutated JMML iPSC colonies was similar to or lower than that of iPSC colonies from a healthy individual. The PTPN11 mutation coexisted with the OSBP2 c.389C>T mutation. Zinc-finger nuclease-mediated homologous recombination revealed that correction of PTPN11 mutation in iPSCs with PTPN11 and OSBP2 mutations resulted in reduced CD34+ cell generation to a level similar to that obtained with JMML iPSC colonies with the wild-type of both genes, and interestingly, to that obtained with normal iPSC colonies. Transduction of the PTPN11 mutation into JMML iPSCs with the wild-type of both genes increased CD34+ cell production to a level comparable to that obtained with JMML iPSC colonies harbouring the two genetic mutations. Thus, PTPN11 mutation may be the most essential abnormality to confer an aberrant haematopoietic differentiation potential in this disorder.


Assuntos
Diferenciação Celular/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucemia Mielomonocítica Juvenil , Células-Tronco Neoplásicas/metabolismo , Mutação Puntual , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Animais , Células-Tronco Hematopoéticas/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/patologia , Masculino , Camundongos SCID , Células-Tronco Neoplásicas/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo
2.
Blood ; 129(15): 2148-2160, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28209720

RESUMO

Chronic myelomonocytic leukemia (CMML) is a hematological malignancy characterized by uncontrolled proliferation of dysplastic myelomonocytes and frequent progression to acute myeloid leukemia (AML). We identified mutations in the Cbl gene, which encodes a negative regulator of cytokine signaling, in a subset of CMML patients. To investigate the contribution of mutant Cbl in CMML pathogenesis, we generated conditional knockin mice for Cbl that express wild-type Cbl in a steady state and inducibly express CblQ367P , a CMML-associated Cbl mutant. CblQ367P mice exhibited sustained proliferation of myelomonocytes, multilineage dysplasia, and splenomegaly, which are the hallmarks of CMML. The phosphatidylinositol 3-kinase (PI3K)-AKT and JAK-STAT pathways were constitutively activated in CblQ367P hematopoietic stem cells, which promoted cell cycle progression and enhanced chemokine-chemokine receptor activity. Gem, a gene encoding a GTPase that is upregulated by CblQ367P , enhanced hematopoietic stem cell activity and induced myeloid cell proliferation. In addition, Evi1, a gene encoding a transcription factor, was found to cooperate with CblQ367P and progress CMML to AML. Furthermore, targeted inhibition for the PI3K-AKT and JAK-STAT pathways efficiently suppressed the proliferative activity of CblQ367P -bearing CMML cells. Our findings provide insights into the molecular mechanisms underlying mutant Cbl-induced CMML and propose a possible molecular targeting therapy for mutant Cbl-carrying CMML patients.


Assuntos
Ciclo Celular , Células-Tronco Hematopoéticas , Leucemia Mielogênica Crônica BCR-ABL Positiva , Mutação de Sentido Incorreto , Mielopoese , Proteínas Proto-Oncogênicas c-cbl , Regulação para Cima , Substituição de Aminoácidos , Animais , Regulação Enzimológica da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , 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 , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/patologia , Proteínas Monoméricas de Ligação ao GTP/biossíntese , Proteínas Monoméricas de Ligação ao GTP/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-cbl/biossíntese , Proteínas Proto-Oncogênicas c-cbl/genética , Transdução de Sinais
3.
Allergol Int ; 67(2): 234-242, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28919488

RESUMO

BACKGROUND: Several methods have been developed to detect allergen-specific IgE in sera. The passive IgE sensitization assay using human IgE receptor-expressing rat cell line RBL-2H3 is a powerful tool to detect biologically active allergen-specific IgE in serum samples. However, one disadvantage is that RBL-2H3 cells are vulnerable to high concentrations of human sera. Only a few human cultured cell lines are easily applicable to the passive IgE sensitization assay. However, the use of human induced pluripotent stem cells (iPSCs) to generate human mast cells (MCs) has not yet been reported. METHODS: The nuclear factor-kappa B (NF-κB)-responsive luciferase reporter gene was stably introduced into a human iPSC line 201B7, and the transfectants were induced to differentiate into MCs (iPSC-MCs). The iPSC-MCs were sensitized overnight with sera from subjects who were allergic to cedar pollen, ragweed pollen, mites, or house dust, and then stimulated with an extract of corresponding allergens. Activation of iPSC-MCs was evaluated by ß-hexosaminidase release, histamine release, or luciferase intensity. RESULTS: iPSCs-MCs stably expressed high-affinity IgE receptor and functionally responded to various allergens when sensitized with human sera from relevant allergic subjects. This passive IgE sensitization system, which we termed the induced mast cell activation test (iMAT), worked well even with undiluted human sera. CONCLUSIONS: iMAT may serve as a novel determining system for IgE/allergens in the clinical and research settings.


Assuntos
Teste de Degranulação de Basófilos/métodos , Hipersensibilidade/diagnóstico , Células-Tronco Pluripotentes Induzidas/citologia , Mastócitos/citologia , Mastócitos/imunologia , Adulto , Alérgenos , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
4.
Proc Natl Acad Sci U S A ; 110(8): 3023-8, 2013 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-23382209

RESUMO

The derivation of induced pluripotent stem (iPS) cells from individuals of genetic disorders offers new opportunities for basic research into these diseases and the development of therapeutic compounds. Severe congenital neutropenia (SCN) is a serious disorder characterized by severe neutropenia at birth. SCN is associated with heterozygous mutations in the neutrophil elastase [elastase, neutrophil-expressed (ELANE)] gene, but the mechanisms that disrupt neutrophil development have not yet been clarified because of the current lack of an appropriate disease model. Here, we generated iPS cells from an individual with SCN (SCN-iPS cells). Granulopoiesis from SCN-iPS cells revealed neutrophil maturation arrest and little sensitivity to granulocyte-colony stimulating factor, reflecting a disease status of SCN. Molecular analysis of the granulopoiesis from the SCN-iPS cells vs. control iPS cells showed reduced expression of genes related to the wingless-type mmtv integration site family, member 3a (Wnt3a)/ß-catenin pathway [e.g., lymphoid enhancer-binding factor 1], whereas Wnt3a administration induced elevation lymphoid enhancer-binding factor 1-expression and the maturation of SCN-iPS cell-derived neutrophils. These results indicate that SCN-iPS cells provide a useful disease model for SCN, and the activation of the Wnt3a/ß-catenin pathway may offer a novel therapy for SCN with ELANE mutation.


Assuntos
Neutropenia/imunologia , Neutrófilos/imunologia , Células-Tronco Pluripotentes/imunologia , Proteína Wnt3A/fisiologia , Diferenciação Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Elastase de Leucócito/genética , Mutação , Neutropenia/congênito , Neutropenia/patologia , Neutrófilos/citologia , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Dev Biol ; 345(2): 226-36, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20647008

RESUMO

Although internal ribosome entry site (IRES)-mediated translation is considered important for proper cellular function, its precise biological role is not fully understood. Runx1 gene, which encodes a transcription factor implicated in hematopoiesis, angiogenesis, and leukemogenesis, contains IRES sequences in the 5' untranslated region. To clarify the roles of the IRES element in Runx1 function, we generated knock-in mice for either wild-type Runx1 or Runx1/Evi1, a Runx1 fusion protein identified in human leukemia. In both cases, native promoter-dependent transcription was retained, whereas IRES-mediated translation was eliminated. Interestingly, homozygotes expressing wild-type Runx1 deleted for the IRES element (Runx1(Delta IRES/Delta IRES)) died in utero with prominent dilatation of peripheral blood vessels due to impaired pericyte development. In addition, hematopoietic cells in the Runx1(Delta IRES/Delta IRES) fetal liver were significantly decreased, and exhibited an altered differentiation pattern, a reduced proliferative activity, and an impaired reconstitution ability. On the other hand, heterozygotes expressing Runx1/Evi1 deleted for the IRES element (Runx1(+/RE Delta IRES)) were born normally and did not show any hematological abnormalities, in contrast that conventional Runx1/Evi1 heterozygotes die in utero with central nervous system hemorrhage and Runx1/Evi1 chimeric mice develop acute leukemia. The findings reported here demonstrate the essential roles of the IRES element in Runx1 function under physiological and pathological conditions.


Assuntos
Regiões 5' não Traduzidas , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Hematopoese/genética , Leucemia/genética , Neovascularização Fisiológica/genética , Animais , Diferenciação Celular , Regulação Leucêmica da Expressão Gênica , Técnicas de Introdução de Genes , Heterozigoto , Fígado/patologia , Camundongos , Modelos Animais , Ribossomos/metabolismo
6.
Proc Natl Acad Sci U S A ; 105(35): 13087-92, 2008 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-18755895

RESUMO

A critical issue for clinical utilization of human ES cells (hESCs) is whether they can generate terminally mature progenies with normal function. We recently developed a method for efficient production of hematopoietic progenitors from hESCs by coculture with murine fetal liver-derived stromal cells. Large numbers of hESCs-derived erythroid progenitors generated by the coculture enabled us to analyze the development of erythropoiesis at a clone level and investigate their function. The results showed that the globin expression in the erythroid cells in individual clones changed in a time-dependent manner. In particular, embryonic epsilon-globin-expressing erythroid cells from individual clones decreased, whereas adult-type beta-globin-expressing cells increased to approximately 100% in all clones we examined, indicating that the cells undergo definitive hematopoiesis. Enucleated erythrocytes also appeared among the clonal progeny. A comparison analysis showed that hESC-derived erythroid cells took a similar differentiation pathway to human cord blood CD34(+) progenitor-derived cells when examined for the expression of glycophorin A, CD71 and CD81. Furthermore, these hESC-derived erythroid cells could function as oxygen carriers and had a sufficient glucose-6-phosphate dehydrogenase activity. The present study should provide an experimental model for exploring early development of human erythropoiesis and hemoglobin switching and may help in the discovery of drugs for hereditary diseases in erythrocyte development.


Assuntos
Células-Tronco Embrionárias/citologia , Eritrócitos/citologia , Hematopoese , Animais , Antígenos CD/metabolismo , Diferenciação Celular , Linhagem Celular , Células Clonais , Técnicas de Cocultura , Eritrócitos/metabolismo , Citometria de Fluxo , Regulação da Expressão Gênica no Desenvolvimento , Globinas/genética , Globinas/metabolismo , Glicoforinas/metabolismo , Hematopoese/genética , Humanos , Fígado/citologia , Fígado/embriologia , Camundongos , Receptores da Transferrina/metabolismo , Células Estromais/citologia , Tetraspanina 28 , Fatores de Tempo
7.
J Exp Med ; 200(12): 1681-7, 2004 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-15611294

RESUMO

Dok-1 and Dok-2 are closely related rasGAP-associated docking proteins expressed preferentially in hematopoietic cells. Although they are phosphorylated upon activation of many protein tyrosine kinases (PTKs), including those coupled with cytokine receptors and oncogenic PTKs like Bcr-Abl, their physiological roles are largely unidentified. Here, we generated mice lacking Dok-1 and/or Dok-2, which included the double-deficient mice succumbed to myeloproliferative disease resembling human chronic myelogenous leukemia (CML) and chronic myelomonocytic leukemia. The double-deficient mice displayed medullary and extramedullary hyperplasia of granulocyte/macrophage progenitors with leukemic potential, and their myeloid cells showed hyperproliferation and hypo-apoptosis upon treatment and deprivation of cytokines, respectively. Consistently, the mutant myeloid cells showed enhanced Erk and Akt activation upon cytokine stimulation. Moreover, loss of Dok-1 and/or Dok-2 induced blastic transformation of chronic phase CML-like disease in mice carrying the bcr-abl gene, a cause of CML. These findings demonstrate that Dok-1 and Dok-2 are key negative regulators of cytokine responses and are essential for myeloid homeostasis and suppression of leukemia.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Fusão bcr-abl/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Fusão bcr-abl/genética , Regulação Leucêmica da Expressão Gênica/genética , Células Precursoras de Granulócitos/metabolismo , Células Precursoras de Granulócitos/patologia , Homeostase/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Ativação Linfocitária/genética , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Mielopoese/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Proteínas de Ligação a RNA/genética
9.
Stem Cells ; 26(3): 706-14, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17991916

RESUMO

Conditions that influence the selective development or recruitment of connective tissue-type and mucosal-type mast cells (MCs) are not well understood. Here, we report that cynomolgus monkey embryonic stem (ES) cells cocultured with the murine aorta-gonad-mesonephros-derived stromal cell line AGM-S1 differentiated into cobblestone (CS)-like cells by day 10-15. When replated onto fresh AGM-S1 with the addition of stem cell factor, interleukin-6, and Flt3 ligand, these CS-like cells displayed robust growth and generated almost 100% tryptase/chymase double-positive MCs within 3 weeks. At all time points, the percentage of tryptase-positive cells did not exceed that of chymase-positive cells. These ES-derived MCs were CD45+/Kit+/CD31+/CD203c+/HLA-DR- and coexpressed a high-affinity IgE receptor on their surface, which was upregulated after IgE exposure. Electron microscopy showed that they contained many electron dense granules. Moreover, ES-derived MCs responded to stimulation by via IgE and substance P by releasing histamine. These results indicate that ES-derived MCs have the phenotype of functionally mature connective tissue-type MCs. The rapid maturation of ES-derived MCs suggests a unique embryonic pathway in primates for early development of connective tissue-type MCs, which may be independent from the developmental pathway of mucosal-type MCs.


Assuntos
Quimases/metabolismo , Técnicas de Cocultura/métodos , Células-Tronco Embrionárias/citologia , Mastócitos/citologia , Mastócitos/enzimologia , Triptases/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Citocinas/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Histamina/metabolismo , Humanos , Imunoglobulina E , Mastócitos/efeitos dos fármacos , Camundongos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/efeitos dos fármacos , Fenótipo , Primatas , Substância P/metabolismo
10.
Exp Hematol ; 36(3): 255-64, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18207304

RESUMO

OBJECTIVE: Erythrocyte production is tightly regulated by cytokines, particularly erythropoietin (EPO), which affects expansion and viability of erythroid lineage cells via induction of several factors, including Bcl2-like 1 (Bcl-XL). Because type I interferon (IFN) is known to inhibit erythropoiesis, we studied mice deficient in the gene for interferon regulatory factor 2 (IRF2), which functions as a negative regulator of type I IFN signaling, in the context of erythropoiesis regulation. MATERIALS AND METHODS: We performed hematologic analyses and detected normocytic anemia in Irf2-deficient mice. RESULTS: Assessment of the maturation of erythroid progenitors in Irf2-deficient bone marrow by flow cytometry revealed a decreased number of late erythroblasts accompanied by an increased number of early erythroid progenitors. Irf2-deficient mice manifested elevated serum EPO levels, decreased Bcl-XL expression levels and enhanced apoptosis of erythroblasts, which may account for the decreased number of late erythroblasts. We further assessed the role of IRF2 in the regulation of type I IFN signaling during erythropoiesis, and found that additional homozygous mutation of IFNAR1, a subunit of type I IFN receptor complex, led to rescue of the defect of erythropoiesis in Irf2-deficient mice. CONCLUSIONS: Impaired erythropoiesis in Irf2-deficient mice results from excessive type I IFN signaling, which inhibits Bcl-XL expression in erythroid lineage cells. Our present study provides a mechanistic understanding of the potential cross-talk between type I IFN and EPO signaling pathways during erythropoiesis and may offer therapeutic insights into anemia.


Assuntos
Anemia Aplástica/genética , Eritropoese/genética , Fator Regulador 2 de Interferon/genética , Interferon Tipo I/metabolismo , Transdução de Sinais , Anemia Aplástica/sangue , Anemia Aplástica/patologia , Animais , Apoptose , Medula Óssea/patologia , Contagem de Células , Diferenciação Celular/genética , Modelos Animais de Doenças , Eritroblastos/patologia , Células Precursoras Eritroides/patologia , Eritropoese/efeitos dos fármacos , Citometria de Fluxo , Perfilação da Expressão Gênica , Testes Hematológicos , Fator Regulador 2 de Interferon/deficiência , Interferon Tipo I/farmacologia , Camundongos , Camundongos Knockout , Mutação , Receptor de Interferon alfa e beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína bcl-X/genética
11.
Int J Hematol ; 85(5): 371-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17562610

RESUMO

We propose a novel method for the efficient production of hematopoietic progenitors from human embryonic stem cells (hESC) via coculture with murine fetal liver-derived stromal cells, in which embryonic hematopoiesis dramatically expands at midgestation. We generated various hematopoietic progenitors in coculture, and this hematopoietic activity was concentrated in cobblestone-like cells derived from differentiated hESC. The cobblestone-like cells mostly expressed CD34 and retained an endothelial cell potential. They also contained hematopoietic colony-forming cells, especially erythroid and multilineage colony-forming cells at high frequency. The multipotential hematopoietic progenitors abundant among the cobblestone-like cells produced almost all types of mature blood cells, including adult-type alpha-globin-expressing erythrocytes and tryptase/chymase double-positive mast cells. These progenitors showed neither the immature properties of ESC nor the potential to differentiate into endoderm and ectoderm at a clonal level. The coculture system developed for hESC can provide a novel source of hematopoietic and blood cells for applications in cellular therapy and drug screening.


Assuntos
Técnicas de Cocultura/métodos , Hematopoese Extramedular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Multipotentes/citologia , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Feminino , Humanos , Fígado/citologia , Fígado/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Células Estromais/citologia
12.
Int J Hematol ; 86(5): 446-50, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18192114

RESUMO

We describe the clinical course of a patient who experienced refractory pure red cell aplasia (PRCA) after undergoing HLA-matched allogeneic peripheral blood stem cell transplantation (allo-PBSCT) for refractory anemia with an excess of blasts in transformation that had evolved from Kostmann syndrome. The treatment for patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) developing from Kostmann syndrome has not been standardized. We treated this patient with allo-PBSCT using a regimen combining high-dose cytosine arabinoside with granulocyte colony-stimulating factor, in addition to total body irradiation and cyclophosphamide without preceding intensive chemotherapy. The donor was ABO incompatible. Myeloid and platelet recoveries were achieved rapidly. Erythroid engraftment was not evident, however, and the patient was given a diagnosis of PRCA. Regimen-related toxicity and graft-versus-host disease (GVHD) were limited. The PRCA did not respond to various therapies, including the discontinuation of immunosuppressants for the induction of chronic GVHD, human recombinant erythropoietin, immunosuppressive treatment with steroids, cyclosporin A, and human anti-CD20 antibody (rituximab). The patient received transfusions 48 times until the resolution of his anemia by donor leukocyte infusion (DLI) at 25 months after PBSCT. He is now clinically well (performance status, 100%) with normal blood cell counts at 5 years after SCT. An in vitro study demonstrated that serum from the recipient blocked the differentiation of erythroid cells in the bone marrow. The results indicate that the conditioning regimen we describe seems safe and effective for those who have MDS/AML and that DLI might be a valuable approach for refractory PRCA after ABO-incompatible SCT.


Assuntos
Transfusão de Leucócitos , Doadores Vivos , Síndromes Mielodisplásicas/terapia , Mielopoese , Transplante de Células-Tronco de Sangue Periférico , Aplasia Pura de Série Vermelha/terapia , Criança , Ciclofosfamida/administração & dosagem , Sobrevivência de Enxerto , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Imunossupressores/administração & dosagem , Lactente , Masculino , Agonistas Mieloablativos/administração & dosagem , Síndromes Mielodisplásicas/complicações , Proteínas Recombinantes , Aplasia Pura de Série Vermelha/etiologia , Indução de Remissão , Síndrome , Transplante Homólogo , Irradiação Corporal Total
13.
Mol Cell Biol ; 23(20): 7329-38, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14517301

RESUMO

Phospholipase C (PLC) plays important roles in phosphoinositide turnover by regulating the calcium-protein kinase C signaling pathway. PLC-L2 is a novel PLC-like protein which lacks PLC activity, although it is very homologous with PLC delta. PLC-L2 is expressed in hematopoietic cells, but its physiological roles and intracellular functions in the immune system have not yet been clarified. To elucidate the physiological function of PLC-L2, we generated mice which had a genetic PLC-L2 deficiency. PLC-L2-deficient mice grew with no apparent abnormalities. However, mature B cells from PLC-L2-deficient mice were hyperproliferative in response to B-cell receptor (BCR) cross-linking, although B2 cell development appeared to be normal. Molecular biological analysis revealed that calcium influx and NFATc accumulation in nuclei were increased in PLC-L2-deficient B cells. Extracellular signal-regulated kinase activity was also enhanced in PLC-L2-deficient B cells. These mice had a stronger T-cell-independent antigen response. These results indicate that PLC-L2 is a novel negative regulator of BCR signaling and immune responses.


Assuntos
Linfócitos B/metabolismo , Lipase/metabolismo , Proteínas Nucleares , Fosfolipases/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Southern Blotting , Cálcio/metabolismo , Divisão Celular , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Genéticos , Mutagênese , Fatores de Transcrição NFATC , Fenótipo , Fosforilação , Transdução de Sinais , Fatores de Tempo , Distribuição Tecidual , Fatores de Transcrição/metabolismo
14.
Mol Cell Biol ; 23(8): 2969-80, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12665593

RESUMO

Dorsoventral patterning depends on the local concentrations of the morphogens. Twisted gastrulation (TSG) regulates the extracellular availability of a mesoderm inducer, bone morphogenetic protein 4 (BMP-4). However, TSG function in vivo is still unclear. We isolated a TSG cDNA as a secreted molecule from the mouse aorta-gonad-mesonephros region. Here we show that TSG-deficient mice were born healthy, but more than half of the neonatal pups showed severe growth retardation shortly after birth and displayed dwarfism with delayed endochondral ossification and lymphopenia, followed by death within a month. TSG-deficient thymus was atrophic, and phosphorylation of SMAD1 was augmented in the thymocytes, suggesting enhanced BMP-4 signaling in the thymus. Since BMP-4 promotes skeletogenesis and inhibits thymus development, our findings suggest that TSG acts as both a BMP-4 agonist in skeletogenesis and a BMP-4 antagonist in T-cell development. Although lymphopenia in TSG-deficient mice would partly be ascribed to systemic effects of runtiness and wasting, our findings may also provide a clue for understanding the pathogenesis of human dwarfism with combined immunodeficiency.


Assuntos
Desenvolvimento Ósseo/genética , Proteínas Morfogenéticas Ósseas/agonistas , Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Tecido Linfoide/embriologia , Proteínas/genética , Proteínas Proto-Oncogênicas , Animais , Desenvolvimento Ósseo/fisiologia , Proteína Morfogenética Óssea 4 , Diferenciação Celular , Subunidade alfa 2 de Fator de Ligação ao Core , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Desenvolvimento Embrionário e Fetal/genética , Marcação de Genes , Transtornos do Crescimento/genética , Transtornos do Crescimento/patologia , Humanos , Hibridização In Situ , Rim/anormalidades , Tecido Linfoide/crescimento & desenvolvimento , Linfopenia/genética , Camundongos , Camundongos Knockout , Fenótipo , Proteínas/fisiologia , Transdução de Sinais , Proteínas Smad , Proteína Smad1 , Linfócitos T/citologia , Transativadores/metabolismo , Fatores de Transcrição/genética
16.
Trends Cardiovasc Med ; 16(2): 45-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16473761

RESUMO

Circulation is composed of two interactive systems, the cardiovascular and the hematopoietic, which affect each other. Recently, endothelial progenitor cells/angioblasts have been identified in the circulation of the adult mouse and human. Furthermore, some hematopoietic cells (HCs) have been shown to contribute to angiogenesis, suggesting that HCs can transdifferentiate into endothelial cells (ECs). Although these concepts in adult are still controversial, understanding the mechanisms of the relationship between ECs and HCs would benefit the clinical application for cardiovascular and hematologic disorders. Both ECs and HCs are considered to be derived from a common germ layer, the mesoderm, and have more intimate relationship in embryo than in adult. Here, we describe the relationship between ECs and HCs with special attention to the hemogenic ECs in the mouse embryo.


Assuntos
Embrião de Mamíferos/citologia , Endotélio Vascular/citologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/citologia , Adulto , Animais , Humanos , Camundongos , Modelos Animais , Saco Vitelino/citologia
18.
Stem Cells Dev ; 14(6): 687-96, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16433624

RESUMO

Previously, we reported generation of erythropoiesis from acetyl low-density lipoprotein (Ac-LDL)- incorporating endothelial cells (ECs) in the mouse embryo. However, it is still unclear whether the other types of definitive hematopoietic cells (HCs) can be generated from these cells. In this study, ECs at 10 days post coitum (dpc) were tagged with Ac-LDL-DiI and were shown to release DiI+ HCs into the circulation after 12 h of whole embryo culture. The hematopoietic clusters in the main arteries were also stained with DiI. The circulating DiI+ HCs expressed c-Kit and half of these cells displayed blastic morphology. In vitro culture and in vivo reconstitution experiments demonstrated that the circulating DiI+ HCs contained definitive multipotent progenitors, including hematopoietic stem cells (HSCs). Furthermore, the sorted DiI+ HCs were able to colonize the fetal liver (FL) when introduced back into the blood stream of a secondary recipient embryo. These results suggest that Ac-LDL incorporating ECs can produce definitive HSCs and HCs colonizing FL in the mouse embryo.


Assuntos
Diferenciação Celular , Embrião de Mamíferos/citologia , Células Endoteliais/fisiologia , Hematopoese , Células-Tronco Hematopoéticas/fisiologia , Lipoproteínas LDL/metabolismo , Animais , Técnicas de Cultura Embrionária , Células Endoteliais/metabolismo , Corantes Fluorescentes , Células-Tronco Hematopoéticas/metabolismo , Fígado/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-kit/metabolismo
19.
PLoS One ; 10(3): e0120841, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25803811

RESUMO

Induced pluripotent stem (iPS) cells provide powerful tools for studying disease mechanisms and developing therapies for diseases. The 8p11 myeloproliferative syndrome (EMS) is an aggressive chronic myeloproliferative disorder (MPD) that is caused by constitutive activation of fibroblast growth factor receptor 1. EMS is rare and, consequently, effective treatment for this disease has not been established. Here, iPS cells were generated from an EMS patient (EMS-iPS cells) to assist the development of effective therapies for EMS. When iPS cells were co-cultured with murine embryonic stromal cells, EMS-iPS cells produced more hematopoietic progenitor and hematopoietic cells, and CD34+ cells derived from EMS-iPS cells exhibited 3.2-7.2-fold more macrophage and erythroid colony forming units (CFUs) than those derived from control iPS cells. These data indicate that EMS-iPS cells have an increased hematopoietic differentiation capacity, which is characteristic of MPDs. To determine whether a tyrosine kinase inhibitor (TKI) could suppress the increased number of CFUs formed by EMS-iPS-induced CD34+ cells, cells were treated with one of four TKIs (CHIR258, PKC 412, ponatinib, and imatinib). CHIR258, PKC 412, and ponatinib reduced the number of CFUs formed by EMS-iPS-induced CD34+ cells in a dose-dependent manner, whereas imatinib did not. Similar effects were observed on primary peripheral blood cells (more than 90% of which were blasts) isolated from the patient. This study provides evidence that the EMS-iPS cell line is a useful tool for the screening of drugs to treat EMS and to investigate the mechanism underlying this disease.


Assuntos
Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/genética , Proteínas de Fusão Oncogênica/genética , Inibidores de Proteínas Quinases/uso terapêutico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Translocação Genética , Adolescente , Benzimidazóis/uso terapêutico , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Hematopoese , Humanos , Mesilato de Imatinib/uso terapêutico , Imidazóis/uso terapêutico , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Masculino , Transtornos Mieloproliferativos/patologia , Piridazinas/uso terapêutico , Quinolonas/uso terapêutico , Estaurosporina/análogos & derivados , Estaurosporina/uso terapêutico
20.
Int J Hematol ; 77(3): 245-52, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12731667

RESUMO

Tunica interna endothelial cell kinase (TEK) is expressed in both hematopoietic and endothelial cells and plays a crucial role in hematopoiesis and angiogenesis in mouse development. In humans, however, little is known about the hematopoietic and angiogenic potentials of TEK-expressing cells in umbilical cord blood (CB) cells, which originate during the human fetal period. We therefore compared the hematopoietic and angiogenic abilities of CB CD34+TEK+ and CD34+TEK- cells by using a clonogenic assay and xenotransplantation into immunodeficient NOD/SCID mice. The results showed that colony-forming cells and cells capable of repopulating in NOD/SCID mice were present in both CD34+TEK+ and CD34+TEK- cells and that the hematopoietic activities of the cell types were similar. In contrast, the potential to differentiate into endothelial cells in vivo was greater in the CD34+TEK+ cells. All NOD/SCID mice engrafted with CD34+TEK+ cells had human CD31-expressing and VE-cadherin-expressing endothelial cells in the vessels of the ischemic muscles and/ or human endothelial cells expressing CD31, kinase-insert domain-containing receptor, and endothelial nitric oxide synthase in liver sinusoidal cells, whereas such endothelial cells were detected in only 3 of the 7 recipients engrafted with CD34+TEK- cells. This result has important implications in cell therapy using CB cells for treating hematopoietic disorders and vascular diseases.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/enzimologia , Neovascularização Fisiológica , Receptor TIE-2/fisiologia , Animais , Antígenos CD , Antígenos CD34 , Caderinas/análise , Diferenciação Celular , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Endotélio Vascular/química , Endotélio Vascular/citologia , Sangue Fetal , Células-Tronco Hematopoéticas/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Receptor TIE-2/análise , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA