Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Exp Hematol ; 76: 38-48.e2, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31295506

RESUMO

A better understanding of the development and progression of acute myelogenous leukemia (AML) is necessary to improve patient outcome. Here we define roles for the transcription factor Oct1/Pou2f1 in AML and normal hematopoiesis. Inappropriate reactivation of the CDX2 gene is widely observed in leukemia patients and in leukemia mouse models. We show that Oct1 associates with the CDX2 promoter in both normal and AML primary patient samples, but recruits the histone demethylase Jmjd1a/Kdm3a to remove the repressive H3K9me2 mark only in malignant specimens. The CpG DNA immediately adjacent to the Oct1 binding site within the CDX2 promoter exhibits variable DNA methylation in healthy control blood and bone marrow samples, but complete demethylation in AML samples. In MLL-AF9-driven mouse models, partial loss of Oct1 protects from myeloid leukemia. Complete Oct1 loss completely suppresses leukemia but results in lethality from bone marrow failure. Loss of Oct1 in normal hematopoietic transplants results in superficially normal long-term reconstitution; however, animals become acutely sensitive to 5-fluorouracil, indicating that Oct1 is dispensable for normal hematopoiesis but protects blood progenitor cells against external chemotoxic stress. These findings elucidate a novel and important role for Oct1 in AML.


Assuntos
Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/fisiologia , Fator 1 de Transcrição de Octâmero/fisiologia , Animais , Medula Óssea/patologia , Transtornos da Insuficiência da Medula Óssea/etiologia , Transtornos da Insuficiência da Medula Óssea/genética , Fator de Transcrição CDX2/biossíntese , Fator de Transcrição CDX2/genética , Transformação Celular Neoplásica/genética , Ilhas de CpG , Metilação de DNA , Progressão da Doença , Fluoruracila/toxicidade , Regulação Leucêmica da Expressão Gênica , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/prevenção & controle , Leucemia Mieloide Aguda/metabolismo , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Camundongos Endogâmicos C57BL , Fator 1 de Transcrição de Octâmero/deficiência , Proteínas de Fusão Oncogênica/fisiologia , Regiões Promotoras Genéticas , Quimera por Radiação
2.
Nature ; 549(7673): 476-481, 2017 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-28825709

RESUMO

Stem-cell fate can be influenced by metabolite levels in culture, but it is not known whether physiological variations in metabolite levels in normal tissues regulate stem-cell function in vivo. Here we describe a metabolomics method for the analysis of rare cell populations isolated directly from tissues and use it to compare mouse haematopoietic stem cells (HSCs) to restricted haematopoietic progenitors. Each haematopoietic cell type had a distinct metabolic signature. Human and mouse HSCs had unusually high levels of ascorbate, which decreased with differentiation. Systemic ascorbate depletion in mice increased HSC frequency and function, in part by reducing the function of Tet2, a dioxygenase tumour suppressor. Ascorbate depletion cooperated with Flt3 internal tandem duplication (Flt3ITD) leukaemic mutations to accelerate leukaemogenesis, through cell-autonomous and possibly non-cell-autonomous mechanisms, in a manner that was reversed by dietary ascorbate. Ascorbate acted cell-autonomously to negatively regulate HSC function and myelopoiesis through Tet2-dependent and Tet2-independent mechanisms. Ascorbate therefore accumulates within HSCs to promote Tet activity in vivo, limiting HSC frequency and suppressing leukaemogenesis.


Assuntos
Ácido Ascórbico/metabolismo , Carcinogênese/metabolismo , Células-Tronco Hematopoéticas/citologia , Leucemia/patologia , Animais , Ácido Ascórbico/análise , Deficiência de Ácido Ascórbico/genética , Deficiência de Ácido Ascórbico/metabolismo , Carcinogênese/genética , Proteínas de Ligação a DNA/metabolismo , Dioxigenases , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Leucemia/genética , Masculino , Metabolômica , Camundongos , Mielopoese/genética , Proteínas Proto-Oncogênicas/metabolismo , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
3.
Stem Cells ; 34(1): 67-82, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26439305

RESUMO

Splenomegaly is a major manifestation of primary myelofibrosis (PMF) contributing to clinical symptoms and hematologic abnormalities. The spleen from PMF patients contains increased numbers of hematopoietic stem cells (HSC) and megakaryocytes (MK). These MK express high levels of P-selectin (P-sel) that, by triggering neutrophil emperipolesis, may cause TGF-ß release and disease progression. This hypothesis was tested by deleting the P-sel gene in the myelofibrosis mouse model carrying the hypomorphic Gata1(low) mutation that induces megakaryocyte abnormalities that recapitulate those observed in PMF. P-sel(null) Gata1(low) mice survived splenectomy and lived 3 months longer than P-sel(WT) Gata1(low) littermates and expressed limited fibrosis and osteosclerosis in the marrow or splenomegaly. Furthermore, deletion of P-sel disrupted megakaryocyte/neutrophil interactions in spleen, reduced TGF-ß content, and corrected the HSC distribution that in Gata1(low) mice, as in PMF patients, is abnormally expanded in spleen. Conversely, pharmacological inhibition of TGF-ß reduced P-sel expression in MK and corrected HSC distribution. Spleens, but not marrow, of Gata1(low) mice contained numerous cKIT(pos) activated fibrocytes, probably of dendritic cell origin, whose membrane protrusions interacted with MK establishing niches hosting immature cKIT(pos) hematopoietic cells. These activated fibrocytes were not detected in spleens from P-sel(null) Gata1(low) or TGF-ß-inhibited Gata1(low) littermates and were observed in spleen, but not in marrow, from PMF patients. Therefore, in Gata1(low) mice, and possibly in PMF, abnormal P-sel expression in MK may mediate the pathological cell interactions that increase TGF-ß content in MK and favor establishment of a microenvironment that supports myelofibrosis-related HSC in spleen.


Assuntos
Fator de Transcrição GATA1/metabolismo , Hematopoese Extramedular , Selectina-P/metabolismo , Mielofibrose Primária/metabolismo , Animais , Diferenciação Celular , Modelos Animais de Doenças , Emperipolese , Feminino , Humanos , Masculino , Megacariócitos/patologia , Megacariócitos/ultraestrutura , Camundongos , Neutrófilos/metabolismo , Fenótipo , Mielofibrose Primária/patologia , Baço/patologia , Baço/ultraestrutura , Fator de Crescimento Transformador beta/metabolismo
4.
Hum Antibodies ; 22(1-2): 21-9, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24284306

RESUMO

BACKGROUND: c3orf75 is a conserved open reading frame within the human genome and has recently been identified as the Elongator subunit, ELP6 [1]. The Elongator enzyme complex has diverse roles, including translational control, neuronal development, cell migration and tumorigenicity [2]. OBJECTIVE: To identify genes expressed early in human eosinophil development. METHODS: Eosinophilopoiesis was investigated by gene profiling of IL-5 stimulated CD34+ cells; ELP6 mRNA is upregulated. A monoclonal antibody was raised to the recombinant protein predicted by the open reading frame. RESULTS: ELP6 transcripts are upregulated in a human tissue culture model of eosinophil development during gene profiling experiments. Transcripts are expressed in most tissue types, as shown by reverse-transcriptase PCR. Western blot experiments show that human ELP6 is a 30 kDa protein expressed in the bone marrow, as well as in many other tissues. Flow cytometry experiments of human bone marrow mononuclear cells show that ELP6 is expressed intracellularly, in developing and mature human neutrophils, eosinophils and monocytes. CONCLUSIONS: ELP6 is expressed intracellularly in developing and mature granulocytes and monocytes but not in lymphocytes and erythrocytes.


Assuntos
Eosinófilos/metabolismo , Monócitos/metabolismo , Neutrófilos/metabolismo , Proteínas/genética , RNA Mensageiro/genética , Antígenos CD34/genética , Antígenos CD34/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Eosinófilos/citologia , Eosinófilos/efeitos dos fármacos , Sangue Fetal/citologia , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Histona Acetiltransferases , Humanos , Imunofenotipagem , Interleucina-5/farmacologia , Monócitos/citologia , Monócitos/efeitos dos fármacos , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Proteínas/metabolismo , RNA Mensageiro/metabolismo
5.
PLoS One ; 8(3): e57481, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469197

RESUMO

How inflammatory responses are mechanistically modulated by nicotinic acetylcholine receptors (nAChR), especially by receptors composed of alpha7 (α7) subunits, is poorly defined. This includes a precise definition of cells that express α7 and how these impact on innate inflammatory responses. To this aim we used mice generated through homologous recombination that express an Ires-Cre-recombinase bi-cistronic extension of the endogenous α7 gene that when crossed with a reporter mouse expressing Rosa26-LoxP (yellow fluorescent protein (YFP)) marks in the offspring those cells of the α7 cell lineage (α7(lin+)). In the adult, on average 20-25 percent of the total CD45(+) myeloid and lymphoid cells of the bone marrow (BM), blood, spleen, lymph nodes, and Peyers patches are α7(lin+), although variability between litter mates in this value is observed. This hematopoietic α7(lin+) subpopulation is also found in Sca1(+)cKit(+) BM cells suggesting the α7 lineage is established early during hematopoiesis and the ratio remains stable in the individual thereafter as measured for at least 18 months. Both α7(lin+) and α7(lin-) BM cells can reconstitute the immune system of naïve irradiated recipient mice and the α7(lin+):α7(lin-) beginning ratio is stable in the recipient after reconstitution. Functionally the α7(lin+):α7(lin-) lineages differ in response to LPS challenge. Most notable is the response to LPS as demonstrated by an enhanced production of IL-12/23(p40) by the α7(lin+) cells. These studies demonstrate that α7(lin+) identifies a novel subpopulation of bone marrow cells that include hematopoietic progenitor cells that can re-populate an animal's inflammatory/immune system. These findings suggest that α7 exhibits a pleiotropic role in the hematopoietic system that includes both the direct modulation of pro-inflammatory cell composition and later in the adult the role of modulating pro-inflammatory responses that would impact upon an individual's lifelong response to inflammation and infection.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/imunologia , Hematopoese/imunologia , Células-Tronco Hematopoéticas/imunologia , Receptores Nicotínicos/imunologia , Transferência Adotiva , Animais , Biomarcadores/metabolismo , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/imunologia , Cruzamentos Genéticos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Células-Tronco Hematopoéticas/citologia , Inflamação/genética , Inflamação/imunologia , Interleucina-12/biossíntese , Interleucina-12/imunologia , Interleucina-23/biossíntese , Interleucina-23/imunologia , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/imunologia , Lipopolissacarídeos/farmacologia , Linfócitos/citologia , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Células Mieloides/citologia , Células Mieloides/imunologia , Receptores Nicotínicos/genética , Irradiação Corporal Total , Receptor Nicotínico de Acetilcolina alfa7
6.
PLoS Genet ; 8(11): e1003048, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23144633

RESUMO

Defining master transcription factors governing somatic and cancer stem cell identity is an important goal. Here we show that the Oct4 paralog Oct1, a transcription factor implicated in stress responses, metabolic control, and poised transcription states, regulates normal and pathologic stem cell function. Oct1(HI) cells in the colon and small intestine co-express known stem cell markers. In primary malignant tissue, high Oct1 protein but not mRNA levels strongly correlate with the frequency of CD24(LO)CD44(HI) cancer-initiating cells. Reducing Oct1 expression via RNAi reduces the proportion of ALDH(HI) and dye efflux(HI) cells, and increasing Oct1 increases the proportion of ALDH(HI) cells. Normal ALDH(HI) cells harbor elevated Oct1 protein but not mRNA levels. Functionally, we show that Oct1 promotes tumor engraftment frequency and promotes hematopoietic stem cell engraftment potential in competitive and serial transplants. In addition to previously described Oct1 transcriptional targets, we identify four Oct1 targets associated with the stem cell phenotype. Cumulatively, the data indicate that Oct1 regulates normal and cancer stem cell function.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas , Transportador 1 de Cátions Orgânicos , Células-Tronco , Aldeído Desidrogenase/genética , Aldeído Desidrogenase/metabolismo , Biomarcadores/metabolismo , Antígeno CD24/metabolismo , Colo/citologia , Colo/metabolismo , Células HeLa , Humanos , Receptores de Hialuronatos/metabolismo , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Transportador 1 de Cátions Orgânicos/genética , Transportador 1 de Cátions Orgânicos/metabolismo , Fenótipo , RNA Mensageiro/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo
7.
Eur J Immunol ; 42(4): 1038-43, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22531927

RESUMO

The altered expression of transcription factors in hematopoietic stem cells and their subsequent lineages can alter the development of lymphoid and myeloid lineages. The role of the transcriptional repressor Snai3 protein in the derivation of cells of the hemato-poietic system was investigated. Snai3 is expressed in terminal T-cell and myeloid lineages, therefore, we chose to determine if expressing Snai3 in the early stages of hematopoietic development would influence cell-lineage determination. Expression of Snai3 by retroviral transduction of hematopoietic stem cells using bone marrow chimera studies demonstrated a block in lymphoid-cell development and enhanced expansion of myeloid-lineage cells. Analysis of Snai3-expressing hematopoietic precursor cells showed normal numbers of immature cells, but a block in the development of cells committed to lymphoid lineages. These data indicate that the overexpression of Snai3 does alter bone marrow cell development and that the identification of genes whose expression is altered by the presence of Snai3 would aid in our understanding of these developmental pathways.


Assuntos
Regulação da Expressão Gênica/imunologia , Linfócitos/imunologia , Células Mieloides/imunologia , Proteínas Repressoras/imunologia , Fatores de Transcrição/imunologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/genética , Linfócitos/metabolismo , Camundongos , Camundongos Transgênicos , Células Mieloides/metabolismo , Proteínas Repressoras/biossíntese , Proteínas Repressoras/genética , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Transdução Genética
8.
Blood ; 119(24): 5621-31, 2012 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-22490804

RESUMO

To detect targeted antileukemia agents we have designed a novel, high-content in vivo screen using genetically engineered, T-cell reporting zebrafish. We exploited the developmental similarities between normal and malignant T lymphoblasts to screen a small molecule library for activity against immature T cells with a simple visual readout in zebrafish larvae. After screening 26 400 molecules, we identified Lenaldekar (LDK), a compound that eliminates immature T cells in developing zebrafish without affecting the cell cycle in other cell types. LDK is well tolerated in vertebrates and induces long-term remission in adult zebrafish with cMYC-induced T-cell acute lymphoblastic leukemia (T-ALL). LDK causes dephosphorylation of members of the PI3 kinase/AKT/mTOR pathway and delays sensitive cells in late mitosis. Among human cancers, LDK selectively affects survival of hematopoietic malignancy lines and primary leukemias, including therapy-refractory B-ALL and chronic myelogenous leukemia samples, and inhibits growth of human T-ALL xenografts. This work demonstrates the utility of our method using zebrafish for antineoplastic candidate drug identification and suggests a new approach for targeted leukemia therapy. Although our efforts focused on leukemia therapy, this screening approach has broad implications as it can be translated to other cancer types involving malignant degeneration of developmentally arrested cells.


Assuntos
Antineoplásicos/toxicidade , Hidrazonas/toxicidade , Leucemia/patologia , Quinolinas/toxicidade , Peixe-Zebra/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Crise Blástica/patologia , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Humanos , Hidrazonas/química , Hidrazonas/farmacocinética , Hidrazonas/uso terapêutico , Leucemia/tratamento farmacológico , Camundongos , Mitose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolinas/química , Quinolinas/farmacocinética , Quinolinas/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Immunol ; 187(10): 5203-10, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21998453

RESUMO

The details of the bifurcation of the lymphoid and myeloid lineages following commitment by multipotent progenitor cells (MPP) remain a topic of controversy. We report that the surface glycoprotein CD62L can be characterized as a novel marker of this and other stages of early hematopoietic differentiation. Cell isolation and transplant studies demonstrated CD62L(neg/low) long-term hematopoietic stem cells and CD62L(high) MPP within the traditionally defined c-kit(pos)Lin(neg/low)Sca-1(pos) stem/progenitor cell population. Within the MPP population, previously defined as c-kit(pos)Lin(neg/low)Sca-1(pos)-Thy-1.1(neg)Flt3(pos), Sca-1 and CD62L resolved four populations and segregated Sca-1(high)CD62L(neg/low) MPP from Sca-1(high)CD62L(high) leukocyte-biased progenitors. Using a novel transplantation method that allows tracking of erythroid and platelet engraftment as an alternative to the classical method of in vitro colony formation, we characterized Sca-1(high)CD62L(neg/low) cells as MPP, based on transient engraftment of these lineages. These data establish CD62L as a useful tool in the study of early hematopoiesis and emphasize the power of trilineage-engraftment studies in establishing the lineage potential of MPP subsets.


Assuntos
Diferenciação Celular/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Selectina L/imunologia , Células-Tronco Multipotentes/imunologia , Animais , Antígenos Ly/biossíntese , Antígenos Ly/sangue , Biomarcadores/sangue , Linhagem da Célula/imunologia , Selectina L/biossíntese , Proteínas de Membrana/biossíntese , Proteínas de Membrana/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Multipotentes/metabolismo , Proteínas Proto-Oncogênicas c-kit/biossíntese , Tirosina Quinase 3 Semelhante a fms/biossíntese
10.
Blood ; 117(20): 5494-502, 2011 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-21310927

RESUMO

Mitoferrin1 is 1 of 2 homologous mitochondrial iron transporters and is required for mitochondrial iron delivery in developing erythroid cells. We show that total deletion of Mfrn1 in embryos leads to embryonic lethality. Selective deletion of Mfrn1 in adult hematopoietic tissues leads to severe anemia because of a deficit in erythroblast formation. Deletion of Mfrn1 in hepatocytes has no phenotype or biochemical effect under normal conditions. In the presence of increased porphyrin synthesis, however, deletion of Mfrn1 in hepatocytes results in a decreased ability to convert protoporphyrin IX into heme, leading to protoporphyria, cholestasis, and bridging cirrhosis. Our results show that the activity of mitoferrin1 is required to manage an increase in heme synthesis. The data also show that alterations in heme synthesis within hepatocytes can lead to protoporphyria and hepatotoxicity.


Assuntos
Anemia/etiologia , Proteínas de Membrana Transportadoras/deficiência , Proteínas de Membrana Transportadoras/genética , Protoporfiria Eritropoética/etiologia , Anemia/genética , Animais , Sequência de Bases , Primers do DNA/genética , Perda do Embrião/genética , Feminino , Marcação de Genes , Heme/biossíntese , Hepatócitos/metabolismo , Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Gravidez , Protoporfiria Eritropoética/genética , Protoporfirinas/metabolismo
11.
Blood Cells Mol Dis ; 44(1): 1-6, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19836979

RESUMO

Transgenic expression of a gain-of-function truncated mouse erythropoietin receptor gene (EpoR) leads to expansion of the HSC pool in response to human erythropoietin (Epo). We have re-examined this observation using a knock-in mouse model, wherein the mouse EpoR gene was replaced in its proper genetic locus by a single copy of either a wild-type human or a polycythemia-inducing truncated human EPOR gene. Bone marrow cells obtained from knock-in mice were transplanted together with competitor bone marrow cells in a model that allows tracking of erythroid, platelet, and leukocyte contributions by each genotype. Secondary transplants were also performed. Stem/progenitor cells were identified phenotypically and isolated for colony-forming assays to evaluate cytokine responsiveness by cells with the wild-type human or truncated human EPOR gene. Augmented Epo signaling increased erythroid repopulation post-transplant as expected, but had no effect on short-term or long-term leukocyte repopulation. However, the wild-type human EPOR knock-in mouse showed decreases in both erythroid and platelet repopulation compared to marrow cells from the mutant human EPOR knock-in mouse or normal B6 animals. These results provide evidence supporting a role for Epo signaling in megakaryopoiesis in vivo and suggest a role for Epo signaling early in hematopoietic development.


Assuntos
Células Precursoras Eritroides/citologia , Eritropoese , Células Progenitoras de Megacariócitos/citologia , Receptores da Eritropoetina/fisiologia , Transdução de Sinais/genética , Trombopoese , Animais , Transplante de Medula Óssea/métodos , Células Cultivadas , Citocinas/farmacologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Técnicas de Introdução de Genes , Genótipo , Sobrevivência de Enxerto/genética , Humanos , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/genética , Fatores de Tempo , Globinas beta/análise , Globinas beta/genética
12.
Cell Transplant ; 18(8): 887-97, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19520051

RESUMO

Copper chelation has been shown to favor the expansion of human hematopoietic stem/progenitor cells in vitro. To further understand the effects of copper modulation on defined subsets of stem cells versus progenitor cells, we extended the studies in a mouse system. We isolated mouse hematopoietic stem cells (HSCs) or hematopoietic progenitor cells (HPCs) and cultured them with or without the copper chelator tetraethylenepentamine (TEPA) or CuCl(2). Cytokine-stimulated HPC cultures treated with TEPA for 7 days generated about two to three times more total and erythroid colony-forming cells (CFCs) compared to control cultures. In contrast, CuCl(2) treatment decreased the CFC numbers. Similar results were seen with HSC after 14, but not 7, days of culture. Transplant studies showed that HPCs cultured for 7 days in TEPA had about twofold higher short-term erythroid repopulation potential compared to control cultures, while CuCl(2) decreased the erythroid potential of cultured HPCs compared to control cultures. HSCs cultured with TEPA for 7 days did not exhibit significantly higher repopulation potential in either leukocyte or erythrocyte lineages compared to control cultures in short-term or long-term assays. Based on JC-1 staining, the mitochondrial membrane potential of HPCs cultured with TEPA was lower relative to control cultures. Our data suggest that decreasing the cellular copper content with TEPA results in preferential expansion or maintenance of HPC that are biased for erythroid differentiation in vivo, but does not enhance the maintenance of HSC activity in culture.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Cobre/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Etilenodiaminas/farmacologia , Células-Tronco Hematopoéticas/fisiologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
13.
J Biomed Mater Res A ; 91(1): 209-20, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18814276

RESUMO

Asparaginase (ASNase) is an enzyme drug presently approved for the induction of remission in the treatment of patients with acute lymphoblastic leukemia (ALL). The cytotoxic effect of ASNase is derived from its ability to deplete asparagine, an essential amino acid required by certain types of leukemia cells for protein synthesis and survival. Despite its efficacy in enhancing disease remission rate and prolonging complete remission duration in ALL patients, ASNase therapy is nevertheless confounded by a number of serious toxic effects, particularly to organs associated with high protein production (e.g., liver, pancreas), due to the systemic depletion of asparagine. Presented herein is a modified version of our previously established ATTEMPTS protein delivery system that carries the potential to permit a tumor specific, intracellular delivery of ASNase, thereby allowing for a significant reduction of ASNase-induced systemic toxicity. In a previous paper, we already demonstrated the in vitro feasibility of this heparin/protamine-regulated, TAT-mediated system in delivering ASNase directly into ASNase-sensitive murine lymphoma cells. In this article, we further validated the in vivo applicability of this system in animals harboring ASNase-encapsulated L5178Y lymphoma cells. Preliminary results showed that animals inoculated with L5178Y cells containing TAT-ASNase exhibited an extended survival rate of approximately 13% over those harboring L5178Y cells without the encapsulation of ASNase. Furthermore, the TAT-ASNase-treated mice also displayed a significantly improved hematological and liver histological status than the control groups. These findings bring promise to the use of the modified ATTEMPTS delivery system in achieving enhanced ASNase therapy.


Assuntos
Antineoplásicos/administração & dosagem , Asparaginase/administração & dosagem , Sistemas de Liberação de Medicamentos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Sequência de Aminoácidos , Animais , Antineoplásicos/uso terapêutico , Asparaginase/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Produtos do Gene tat/química , Testes Hematológicos , Humanos , Linfonodos/citologia , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Peptídeos/química
14.
Curr Protoc Immunol ; Chapter 4: 4.6.1-4.6.9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18491297

RESUMO

Transplantation of marrow between mouse strains congenic for CD45 after lethal irradiation establishes hematopoiesis driven by genetically marked cells in recipient animals. After several weeks, peripheral blood or primary and secondary lymphoid organs of transplant recipients can be evaluated for the presence of donor-derived cells. Two- or three-color flow cytometry can be used to identify the progeny of transplanted cells, to document their cell-surface phenotypes, and to follow development of T, B, and myeloid lineages in vivo.


Assuntos
Transplante de Medula Óssea , Hematopoese , Antígenos Comuns de Leucócito/metabolismo , Linfócitos/imunologia , Quimera por Radiação/imunologia , Animais , Linhagem da Célula , Proliferação de Células , Feminino , Citometria de Fluxo , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde/genética , Imunofenotipagem , Antígenos Comuns de Leucócito/genética , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Infecções por Pseudomonas/etiologia , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/veterinária , Pseudomonas aeruginosa/isolamento & purificação , Doses de Radiação , Irradiação Corporal Total/efeitos adversos
15.
Blood ; 109(12): 5191-8, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17351112

RESUMO

Gene expression profiling of early eosinophil development shows increased transcript levels of proinflammatory cytokines, chemokines, transcription factors, and a novel gene, EGO (eosinophil granule ontogeny). EGO is nested within an intron of the inositol triphosphate receptor type 1 (ITPR1) gene and is conserved at the nucleotide level; however, the largest open reading frame (ORF) is 86 amino acids. Sucrose density gradients show that EGO is not associated with ribosomes and therefore is a noncoding RNA (ncRNA). EGO transcript levels rapidly increase following interleukin-5 (IL-5) stimulation of CD34(+) hematopoietic progenitors. EGO RNA also is highly expressed in human bone marrow and in mature eosinophils. RNA silencing of EGO results in decreased major basic protein (MBP) and eosinophil derived neurotoxin (EDN) mRNA expression in developing CD34(+) hematopoietic progenitors in vitro and in a CD34(+) cell line model. Therefore, EGO is a novel ncRNA gene expressed during eosinophil development and is necessary for normal MBP and EDN transcript expression.


Assuntos
Proteínas Granulares de Eosinófilos/genética , Proteína Básica Maior de Eosinófilos/genética , Neurotoxina Derivada de Eosinófilo/genética , Regulação da Expressão Gênica/genética , RNA não Traduzido/fisiologia , Células Cultivadas , Eosinófilos/metabolismo , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , RNA não Traduzido/genética , Transcrição Gênica
16.
Ann N Y Acad Sci ; 1106: 82-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17395732

RESUMO

Prospective isolation of hematopoietic stem and progenitor cell subsets depends upon the premise that expression of combinations of surface antigens reflects developmental potential. During the process of differentiation, however, the loss of antigens associated with stem cells and the concomitant gain of those associated with progenitor cells often occurs as a continuum rather than by discrete binary steps. Coupled with the fact that assay conditions can profoundly influence the developmental fates of prospectively isolated cells, gradients of antigen expression during differentiation have led to a variety of interpretations of lineage commitment in hematopoiesis.


Assuntos
Antígenos/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Regulação da Expressão Gênica , Hematopoese , Animais , Antígenos/metabolismo , Células da Medula Óssea/citologia , Separação Celular , Eritropoese , Citometria de Fluxo , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco/citologia , Fatores de Tempo
17.
Exp Hematol ; 34(12): 1730-40, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17157170

RESUMO

OBJECTIVE: The OP9-DL1 culture system is an in vitro model for T-cell development in which activation of the Notch pathway by Delta-like 1 promotes differentiation of mature T cells from progenitors. The roles of specific cytokines in this culture system have not been well defined, and controversy regarding the role of IL-7 has recently emerged. We examined the roles played by IL-7, Flt3 ligand, and stem cell factor (SCF) in differentiation of adult bone marrow cells in the OP9-DL1 culture system. METHODS: Hematopoietic progenitor cells isolated from mouse bone marrow were cultured with OP9 or OP9-DL1 stromal cells and evaluated for T and B lymphocyte differentiation using immunofluorescent staining. RESULTS: IL-7 provided both survival/proliferation and differentiation signals in a dose-dependent manner. T-cell development from the CD4/CD8 double-negative (DN) stage to the CD4/CD8 double-positive (DP) stage required IL-7 provided by the stromal cells, while differentiation from the DP to the CD8 single-positive (SP) stage required addition of exogenous IL-7. SCF favored the proliferation of DN lymphoid progenitors and inhibited differentiation to the DP stage in a dose-dependent manner. Conversely, blocking the function of SCF expressed endogenously by OP9-DL1 cells inhibited proliferation of lymphoid progenitors and accelerated T-lineage differentiation. Flt3 ligand promoted proliferation without affecting differentiation. CONCLUSION: These results validate the OP9-DL1 model for the analysis of T-cell development from bone marrow-derived progenitor cells, and demonstrate specific roles of SCF, IL-7, and Flt3L in promoting efficient T-lineage differentiation.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Interleucina-7/fisiologia , Fator de Células-Tronco/fisiologia , Linfócitos T/imunologia , Animais , Anticorpos/farmacologia , Linfócitos B/imunologia , Benzamidas , Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Técnicas de Cocultura , Citometria de Fluxo , Imunofluorescência , Células-Tronco Hematopoéticas/citologia , Mesilato de Imatinib , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Piperazinas/farmacologia , Pirimidinas/farmacologia , Coloração e Rotulagem , Fator de Células-Tronco/antagonistas & inibidores , Células Estromais/citologia , Células Estromais/fisiologia
18.
Stem Cells ; 24(9): 2045-51, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16690777

RESUMO

Transgenic mouse strains ubiquitously expressing green fluorescent protein (GFP) have enabled investigators to develop in vivo transplant models that can detect donor contributions to many different tissues. However, most GFP transgenics lack expression of the reporter in the erythroid lineage. We evaluated expression of GFP in the bone marrow of the OsbY01 transgenic mouse (B6-GFP) in the context of CD71 and TER-119 expression and found that GFP fluorescence is lost prior to the basophilic erythroblast stage of development. However, platelets in B6-GFP mice were found to be uniformly positive for GFP. We therefore used the GFP transgenic model in combination with allelic variants of CD45 and the hemoglobin beta (Hbb) chain to develop a model system that allows all blood lineages to be followed in a mouse model of bone marrow transplantation (BMT). To detect Hbb variant molecules, we developed a new protocol based on high-performance liquid chromatography that is sensitive and precise, allowing rapid and quantitative analysis of erythroid chimerism. Platelet and leukocyte engraftment were detected by flow cytometry. BMT into sublethally irradiated (4 Gy) recipients demonstrated the failure of B6-GFP-derived cells to engraft relative to B6-CD45(a)-derived cells, suggesting that an immune barrier may prevent efficient engraftment of the transgenic cells in a setting of minimal ablation. These results establish limitations in the use of transgenic GFP expression as a donor marker in transplantation models.


Assuntos
Transplante de Medula Óssea/métodos , Quimerismo , Células Eritroides/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Transgenes/genética , Alelos , Animais , Células Sanguíneas/metabolismo , Linhagem da Célula , Cromatografia Líquida de Alta Pressão , Fluorescência , Proteínas de Fluorescência Verde/análise , Hemoglobinas/genética , Cinética , Antígenos Comuns de Leucócito/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Animais , Fenótipo
19.
Ann N Y Acad Sci ; 1044: 210-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15958714

RESUMO

Bone marrow contains a heterogeneous mixture of mature and maturing precursors of blood cells, progenitor cells for myeloid and lymphoid lineages, and hematopoietic and mesenchymal stem cells. The differentiation potential of these different stem, progenitor, and precursor populations can be evaluated by using transplantation and cell culture assays. In this study, we used a stromal cell co-culture system to evaluate the B and T lineage potential of different subsets of mouse bone marrow. We enriched hematopoietic stem (Lin(-)Sca-1(+)c-kit(+)Thy1.1(low) [Thy1.1(low)]) cells and lymphoid progenitor (Lin(-)Sca-1(+)c-kit(+)Thy1.1(-) [Thy1.1(-)]) cells from mouse bone marrow and co-cultured these populations with OP9 or OP9-DL1 stromal cell lines. Development of the B and T lineages was evaluated over time. Both populations gave rise to B and T cells but with different kinetics. Thy1.1(-) lymphoid progenitors gave rise to B and T lineage cells earlier than did Thy1.1(low) stem cells; and at any given time, percentages of differentiating B and T cells were higher in Thy1.1(-) cultures than in Thy1.1(low) cultures. We also compared the lineage potential of Thy-1.1(-) lymphoid progenitors with that of the recently described common lymphoid progenitor 2 (isolated as Lin(-)Sca-1(+)c-kit(-)Thy1.1(-)B220(+) cells [B220(+)]). B220(+) cells produced B lineage progeny in OP9 cultures more rapidly than did Thy1.1(-) cells and produced higher percentages of differentiating T cells in OP9-DL1 cultures. These studies demonstrate the utility of the OP9 and OP9-DL1 co-culture systems for evaluation of lymphoid lineage potential and for determining the relative position of specific bone marrow populations within the hematopoietic hierarchy.


Assuntos
Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Linfócitos/citologia , Animais , Linhagem da Célula/imunologia , Técnicas de Cocultura , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos
20.
Stem Cells ; 23(5): 638-43, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15849171

RESUMO

Transgenic marking approaches are increasingly used to evaluate the developmental potential of stem cells. However, cell fate mapping studies using different transgenic marking systems have produced conflicting results. These disparate findings may be due in part to the infidelity of donor marker gene expression. Analysis of hematopoietic stem cells (c-Kit+, Sca-1+, lineage marker- [KSL]) from a transgenic mouse (1Osb) engineered to ubiquitously express the enhanced green fluorescent protein (EGFP) reveals two distinct populations. Forty percent of KSL cells demonstrate intermediate levels of EGFP fluorescence and differentiate into subpopulations of B cells, T cells, and myeloid cells that do not express EGFP. By contrast, progeny of the remaining 60% of KSL cells are almost exclusively EGFP bright. Long-term multilineage hematopoietic reconstitution and serial transplantation experiments show that these differences in EGFP are a property of self-renewing stem cells. Furthermore, both the transgene integration site and the activation status of a cell are important determinants of EGFP expression. These results indicate that a combination of donor cell markers is required to reliably track the full differentiation potential of transgenic stem cells.


Assuntos
Antígenos de Diferenciação/análise , Diferenciação Celular , Linhagem da Célula , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Animais , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA