Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Exp Hematol ; 76: 38-48.e2, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31295506

RESUMEN

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.


Asunto(s)
Leucemia Mieloide Aguda/genética , Proteínas de Neoplasias/fisiología , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Animales , Médula Ósea/patología , Trastornos de Fallo de la Médula Ósea/etiología , Trastornos de Fallo de la Médula Ósea/genética , Factor de Transcripción CDX2/biosíntesis , Factor de Transcripción CDX2/genética , Transformación Celular Neoplásica/genética , Islas de CpG , Metilación de ADN , Progresión de la Enfermedad , Fluorouracilo/toxicidad , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Leucemia Experimental/genética , Leucemia Experimental/prevención & control , Leucemia Mieloide Aguda/metabolismo , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Proteínas de Fusión Oncogénica/fisiología , Regiones Promotoras Genéticas , Quimera por Radiación
2.
Nature ; 549(7673): 476-481, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28825709

RESUMEN

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.


Asunto(s)
Ácido Ascórbico/metabolismo , Carcinogénesis/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia/patología , Animales , Ácido Ascórbico/análisis , Deficiencia de Ácido Ascórbico/genética , Deficiencia de Ácido Ascórbico/metabolismo , Carcinogénesis/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia/genética , Masculino , Metabolómica , Ratones , Mielopoyesis/genética , Proteínas Proto-Oncogénicas/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo
3.
Stem Cells ; 34(1): 67-82, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26439305

RESUMEN

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.


Asunto(s)
Factor de Transcripción GATA1/metabolismo , Hematopoyesis Extramedular , Selectina-P/metabolismo , Mielofibrosis Primaria/metabolismo , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Emperipolesis , Femenino , Humanos , Masculino , Megacariocitos/patología , Megacariocitos/ultraestructura , Ratones , Neutrófilos/metabolismo , Fenotipo , Mielofibrosis Primaria/patología , Bazo/patología , Bazo/ultraestructura , Factor de Crecimiento Transformador beta/metabolismo
4.
Hum Antibodies ; 22(1-2): 21-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24284306

RESUMEN

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.


Asunto(s)
Eosinófilos/metabolismo , Monocitos/metabolismo , Neutrófilos/metabolismo , Proteínas/genética , ARN Mensajero/genética , Antígenos CD34/genética , Antígenos CD34/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Eosinófilos/citología , Eosinófilos/efectos de los fármacos , Sangre Fetal/citología , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Acetiltransferasas , Humanos , Inmunofenotipificación , Interleucina-5/farmacología , Monocitos/citología , Monocitos/efectos de los fármacos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Proteínas/metabolismo , ARN Mensajero/metabolismo
5.
PLoS One ; 8(3): e57481, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23469197

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/inmunología , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/inmunología , Receptores Nicotínicos/inmunología , Traslado Adoptivo , Animales , Biomarcadores/metabolismo , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/inmunología , Cruzamientos Genéticos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Inflamación/genética , Inflamación/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Interleucina-23/biosíntesis , Interleucina-23/inmunología , Antígenos Comunes de Leucocito/genética , Antígenos Comunes de Leucocito/inmunología , Lipopolisacáridos/farmacología , Linfocitos/citología , Linfocitos/inmunología , Masculino , Ratones , Ratones Transgénicos , Células Mieloides/citología , Células Mieloides/inmunología , Receptores Nicotínicos/genética , Irradiación Corporal Total , Receptor Nicotínico de Acetilcolina alfa 7
6.
Antioxid Redox Signal ; 19(17): 2054-67, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-23249337

RESUMEN

AIMS: Vitamin C (ascorbic acid) is thought to enhance immune function, but the mechanisms involved are obscure. We utilized an in vitro model of T-cell maturation to evaluate the role of ascorbic acid in lymphocyte development. RESULTS: Ascorbic acid was essential for the developmental progression of mouse bone marrow-derived progenitor cells to functional T-lymphocytes in vitro and also played a role in vivo. Ascorbate-mediated enhancement of T-cell development was lymphoid cell-intrinsic and independent of T-cell receptor (TCR) rearrangement. Analysis of TCR rearrangements demonstrated that ascorbic acid enhanced the selection of functional TCRαß after the stage of ß-selection. Genes encoding the coreceptor CD8 as well as the kinase ZAP70 were upregulated by ascorbic acid. Pharmacologic inhibition of methylation marks on DNA and histones enhanced ascorbate-mediated differentiation, suggesting an epigenetic mechanism of Cd8 gene regulation via active demethylation by ascorbate-dependent Fe(2+) and 2-oxoglutarate-dependent dioxygenases. INNOVATION: We speculate that one aspect of gene regulation mediated by ascorbate occurs at the level of chromatin demethylation, mediated by Jumonji C (JmjC) domain enzymes that are known to be reliant upon ascorbate as a cofactor. JmjC domain enzymes are also known to regulate transcription factor activity. These two mechanisms are likely to play key roles in the modulation of immune development and function by ascorbic acid. CONCLUSION: Our results provide strong experimental evidence supporting a role for ascorbic acid in T-cell maturation as well as insight into the mechanism of ascorbate-mediated enhancement of immune function.


Asunto(s)
Ácido Ascórbico/farmacología , Factores Inmunológicos/farmacología , Linfocitos T/efectos de los fármacos , Animales , Azepinas/farmacología , Células Cultivadas , Medios de Cultivo , Epigénesis Genética/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Reordenamiento Génico de Linfocito T/efectos de los fármacos , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Metilación , Ratones , Ratones Endogámicos C57BL , Ftalimidas/farmacología , Procesamiento Proteico-Postraduccional , Quinazolinas/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Triptófano/análogos & derivados , Triptófano/farmacología
7.
PLoS Genet ; 8(11): e1003048, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144633

RESUMEN

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.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas , Transportador 1 de Catión Orgánico , Células Madre , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Biomarcadores/metabolismo , Antígeno CD24/metabolismo , Colon/citología , Colon/metabolismo , Células HeLa , Humanos , Receptores de Hialuranos/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/metabolismo , Fenotipo , ARN Mensajero/metabolismo , Células Madre/citología , Células Madre/metabolismo
8.
Eur J Immunol ; 42(4): 1038-43, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22531927

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Linfocitos/inmunología , Células Mieloides/inmunología , Proteínas Represoras/inmunología , Factores de Transcripción/inmunología , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/fisiología , Regulación de la Expresión Génica/genética , Linfocitos/metabolismo , Ratones , Ratones Transgénicos , Células Mieloides/metabolismo , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transducción Genética
9.
Blood ; 119(24): 5621-31, 2012 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-22490804

RESUMEN

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.


Asunto(s)
Antineoplásicos/toxicidad , Hidrazonas/toxicidad , Leucemia/patología , Quinolinas/toxicidad , Pez Cebra/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Crisis Blástica/patología , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Hidrazonas/química , Hidrazonas/farmacocinética , Hidrazonas/uso terapéutico , Leucemia/tratamiento farmacológico , Ratones , Mitosis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinolinas/química , Quinolinas/farmacocinética , Quinolinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Immunol ; 188(3): 1381-93, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22180617

RESUMEN

IL-10 is a nonredundant inflammatory modulator that suppresses arthritis development in Borrelia burgdorferi-infected mice. Infected C57BL/6 (B6) IL-10(-/-) mice were previously found to have a prolonged IFN-inducible response in joint tissue. Infection of B6 IL-10 reporter mice identified macrophages and CD4(+) T cells as the primary sources of IL-10 in the infected joint tissue, suggesting that early local production of IL-10 dampened the proarthritic IFN response. Treatment of B6 IL-10(-/-) mice with anti-IFN-γ reduced the increase in arthritis severity and suppressed IFN-inducible transcripts to wild-type levels, thereby linking dysregulation of IFN-γ to disease in the B6 IL-10(-/-) mouse. Arthritis in B6 IL-10(-/-) mice was associated with elevated numbers of NK cell, NKT cell, α/ß T cell, and macrophage infiltration of the infected joint. FACS lineage sorting revealed NK cells and CD4(+) T cells as sources of IFN-γ in the joint tissue of B6 IL-10(-/-) mice. These findings suggest the presence of a positive-feedback loop in the joint tissue of infected B6 IL-10(-/-) mice, in which production of inflammatory chemokines, infiltration of IFN-γ-producing cells, and additional production of inflammatory cytokines result in arthritis. This mechanism of arthritis is in contrast to that seen in C3H/He mice, in which arthritis development is linked to transient production of type I IFN and develops independently of IFN-γ. Due to the sustained IFN response driven by NK cells and T cells, we propose the B6 IL-10(-/-) mouse as a potential model to study the persistent arthritis observed in some human Lyme disease patients.


Asunto(s)
Movimiento Celular/inmunología , Inflamación/inmunología , Interferón gamma/farmacología , Interleucina-10/biosíntesis , Enfermedad de Lyme/inmunología , Animales , Borrelia burgdorferi , Retroalimentación Fisiológica , Humanos , Interferón gamma/inmunología , Interleucina-10/deficiencia , Células Asesinas Naturales/inmunología , Ratones , Ratones Noqueados , Linfocitos T/inmunología
11.
J Immunol ; 187(10): 5203-10, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21998453

RESUMEN

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.


Asunto(s)
Diferenciación Celular/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Selectina L/inmunología , Células Madre Multipotentes/inmunología , Animales , Antígenos Ly/biosíntesis , Antígenos Ly/sangre , Biomarcadores/sangre , Linaje de la Célula/inmunología , Selectina L/biosíntesis , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Multipotentes/metabolismo , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Tirosina Quinasa 3 Similar a fms/biosíntesis
12.
Blood ; 117(20): 5494-502, 2011 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-21310927

RESUMEN

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.


Asunto(s)
Anemia/etiología , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Protoporfiria Eritropoyética/etiología , Anemia/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Pérdida del Embrión/genética , Femenino , Marcación de Gen , Hemo/biosíntesis , Hepatocitos/metabolismo , Hierro/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Embarazo , Protoporfiria Eritropoyética/genética , Protoporfirinas/metabolismo
13.
Blood Cells Mol Dis ; 44(1): 1-6, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19836979

RESUMEN

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.


Asunto(s)
Células Precursoras Eritroides/citología , Eritropoyesis , Células Progenitoras de Megacariocitos/citología , Receptores de Eritropoyetina/fisiología , Transducción de Señal/genética , Trombopoyesis , Animales , Trasplante de Médula Ósea/métodos , Células Cultivadas , Citocinas/farmacología , Eritrocitos/metabolismo , Células Precursoras Eritroides/efectos de los fármacos , Células Precursoras Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Técnicas de Sustitución del Gen , Genotipo , Supervivencia de Injerto/genética , Humanos , Células Progenitoras de Megacariocitos/efectos de los fármacos , Células Progenitoras de Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo , Isoformas de Proteínas/análisis , Isoformas de Proteínas/genética , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/genética , Factores de Tiempo , Globinas beta/análisis , Globinas beta/genética
14.
Cell Transplant ; 18(8): 887-97, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19520051

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Cobre/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Animales , Técnicas de Cultivo de Célula , Células Cultivadas , Etilenodiaminas/farmacología , Células Madre Hematopoyéticas/fisiología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
J Biomed Mater Res A ; 91(1): 209-20, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18814276

RESUMEN

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.


Asunto(s)
Antineoplásicos/administración & dosificación , Asparaginasa/administración & dosificación , Sistemas de Liberación de Medicamentos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Secuencia de Aminoácidos , Animales , Antineoplásicos/uso terapéutico , Asparaginasa/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular , Femenino , Productos del Gen tat/química , Pruebas Hematológicas , Humanos , Ganglios Linfáticos/citología , Ratones , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Péptidos/química
16.
Curr Protoc Immunol ; Chapter 4: 4.6.1-4.6.9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18491297

RESUMEN

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.


Asunto(s)
Trasplante de Médula Ósea , Hematopoyesis , Antígenos Comunes de Leucocito/metabolismo , Linfocitos/inmunología , Quimera por Radiación/inmunología , Animales , Linaje de la Célula , Proliferación Celular , Femenino , Citometría de Flujo , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Inmunofenotipificación , Antígenos Comunes de Leucocito/genética , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Pseudomonas/etiología , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/veterinaria , Pseudomonas aeruginosa/aislamiento & purificación , Dosis de Radiación , Irradiación Corporal Total/efectos adversos
17.
Blood ; 109(12): 5191-8, 2007 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-17351112

RESUMEN

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.


Asunto(s)
Proteínas en los Gránulos del Eosinófilo/genética , Proteína Mayor Básica del Eosinófilo/genética , Neurotoxina Derivada del Eosinófilo/genética , Regulación de la Expresión Génica/genética , ARN no Traducido/fisiología , Células Cultivadas , Eosinófilos/metabolismo , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , ARN no Traducido/genética , Transcripción Genética
18.
Ann N Y Acad Sci ; 1106: 82-8, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17395732

RESUMEN

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.


Asunto(s)
Antígenos/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica , Hematopoyesis , Animales , Antígenos/metabolismo , Células de la Médula Ósea/citología , Separación Celular , Eritropoyesis , Citometría de Flujo , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Células Madre/citología , Factores de Tiempo
19.
Blood ; 109(6): 2618-21, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17148589

RESUMEN

Congenital erythropoietic porphyria (CEP), an autosomal recessive disorder, is due to mutations of uroporphyrinogen III synthase (UROS). Deficiency of UROS results in excess uroporphyrin I, which causes photosensitization. We evaluated a 3-year-old boy with CEP. A hypochromic, microcytic anemia was present from birth, and platelet counts averaged 70 x 10(9)/L (70,000/microL). Erythrocyte UROS activity was 21% of controls. Red cell morphology and globin chain labeling studies were compatible with beta-thalassemia. Hb electrophoresis revealed 36.3% A, 2.4% A(2), 59.5% F, and 1.8% of an unidentified peak. No UROS or alpha- and beta-globin mutations were found in the child or the parents. The molecular basis of the phenotype proved to be a mutation of GATA1, an X-linked transcription factor common to globin genes and heme biosynthetic enzymes in erythrocytes. A mutation at codon 216 in the child and on one allele of his mother changed arginine to tryptophan (R216W). This is the first report of a human porphyria due to a mutation in a trans-acting factor and the first association of CEP with thalassemia and thrombocytopenia. The Hb F level of 59.5% suggests a role for GATA-1 in globin switching. A bone marrow allograft corrected both the porphyria and the thalassemia.


Asunto(s)
Factor de Transcripción GATA1/genética , Mutación/genética , Porfiria Eritropoyética/genética , Activación Transcripcional/genética , Secuencia de Aminoácidos , Preescolar , Femenino , Factor de Transcripción GATA1/química , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Porfiria Eritropoyética/etiología , Porfiria Eritropoyética/patología , Unión Proteica , Dedos de Zinc
20.
Exp Hematol ; 34(12): 1730-40, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17157170

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Interleucina-7/fisiología , Factor de Células Madre/fisiología , Linfocitos T/inmunología , Animales , Anticuerpos/farmacología , Linfocitos B/inmunología , Benzamidas , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células Madre Hematopoyéticas/citología , Mesilato de Imatinib , Proteínas de la Membrana/fisiología , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Piperazinas/farmacología , Pirimidinas/farmacología , Coloración y Etiquetado , Factor de Células Madre/antagonistas & inhibidores , Células del Estroma/citología , Células del Estroma/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA