Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Methods ; 112: 68-74, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27582124

RESUMEN

Adult humans need to make 2.5million red blood cells (RBCs) every second to maintain a steady state level of 25trillion circulating RBCs. Understanding normal erythropoiesis as well as diseases that afflict the erythron, such as genetic anemias, hyperproliferative disorders, and myelodysplastic syndromes, requires a robust method to delineate erythropoietic intermediates. In order to apply the power of flow cytometry to these studies, challenges of limited immunophenotypic markers, incorporation of significant changes in morphology, and maturational changes that occur along a continuum need to be met. Imaging flow cytometry (IFC) provides a solution to address these challenges. Integration of changes in immunophenotype, loss of RNA (ribosomes), and enucleation, with morphological characteristics of cell and nuclear size, can be used to delineate erythroblasts that correlate with classical histological classifications. A protocol is described that demonstrates the basic approaches of staining panel selection, mask generation and selection of features to best sequentially refine erythroid intermediates and remove contaminating cells with overlapping immunophenotype. Ultimately erythroid cells in the murine bone marrow are divided into seven sub-populations using IFC including four erythroblasts (pro-, basophilic, polychromatophilic and orthochromatic), the pyrenocyte, which contains the eliminated nucleus, the enucleated reticulocyte and the mature RBC.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula/genética , Eritropoyesis/genética , Citometría de Flujo/métodos , Citometría de Imagen/métodos , Animales , Biomarcadores/metabolismo , Células de la Médula Ósea/clasificación , Células de la Médula Ósea/metabolismo , Ciclo Celular/genética , Diferenciación Celular , Núcleo Celular/ultraestructura , Eritroblastos/citología , Eritroblastos/metabolismo , Eritrocitos/citología , Eritrocitos/metabolismo , Citometría de Flujo/instrumentación , Humanos , Citometría de Imagen/instrumentación , Ratones , Cultivo Primario de Células , Reticulocitos/citología , Reticulocitos/metabolismo , Ribosomas/ultraestructura , Coloración y Etiquetado/métodos
2.
J Math Biol ; 70(7): 1485-521, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24989701

RESUMEN

The erythroid lineage is a particularly sensitive target of radiation injury. We model the dynamics of immature (BFU-E) and mature (CFU-E) erythroid progenitors, which have markedly different kinetics of recovery, following sublethal total body irradiation using a two-type reducible age-dependent branching process with immigration. Properties of the expectation and variance of the frequencies of both types of progenitors are presented. Their explicit expressions are derived when the process is Markovian, and their asymptotic behavior is identified in the age-dependent (non-Markovian) case. Analysis of experimental data on the kinetics of BFU-E and CFU-E reveals that the probability of self-renewal increases transiently for both cell types following sublethal irradiation. In addition, the probability of self-renewal increased more for CFU-E than for BFU-E. The strategy adopted by the erythroid lineage ensures replenishment of the BFU-E compartment while optimizing the rate of CFU-E recovery. Finally, our analysis also indicates that radiation exposure causes a delay in BFU-E recovery consistent with injury to the hematopoietic stem/progenitor cell compartment that give rise to BFU-E. Erythroid progenitor self-renewal is thus an integral component of the recovery of the erythron in response to stress.


Asunto(s)
Eritropoyesis/fisiología , Eritropoyesis/efectos de la radiación , Modelos Biológicos , Animales , Ensayo de Unidades Formadoras de Colonias , Simulación por Computador , Células Precursoras Eritroides/patología , Células Precursoras Eritroides/fisiología , Células Precursoras Eritroides/efectos de la radiación , Humanos , Cinética , Cadenas de Markov , Conceptos Matemáticos , Ratones , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/fisiopatología , Procesos Estocásticos , Estrés Fisiológico , Irradiación Corporal Total/efectos adversos
3.
Radiat Res ; 196(3): 284-296, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34153091

RESUMEN

Thrombocytopenia is a major complication in hematopoietic-acute radiation syndrome (H-ARS) that increases the risk of mortality from uncontrolled hemorrhage. There is a great demand for new therapies to improve survival and mitigate bleeding in H-ARS. Thrombopoiesis requires interactions between megakaryocytes (MKs) and endothelial cells. 16, 16-dimethyl prostaglandin E2 (dmPGE2), a longer-acting analogue of PGE2, promotes hematopoietic recovery after total-body irradiation (TBI), and various angiotensin-converting enzyme (ACE) inhibitors mitigate endothelial injury after radiation exposure. Here, we tested a combination therapy of dmPGE2 and lisinopril to mitigate thrombocytopenia in murine models of H-ARS following TBI. After 7.75 Gy TBI, dmPGE2 and lisinopril each increased survival relative to vehicle controls. Importantly, combined dmPGE2 and lisinopril therapy enhanced survival greater than either individual agent. Studies performed after 4 Gy TBI revealed reduced numbers of marrow MKs and circulating platelets. In addition, sublethal TBI induced abnormalities both in MK maturation and in in vitro and in vivo platelet function. dmPGE2, alone and in combination with lisinopril, improved recovery of marrow MKs and peripheral platelets. Finally, sublethal TBI transiently reduced the number of marrow Lin-CD45-CD31+Sca-1- sinusoidal endothelial cells, while combined dmPGE2 and lisinopril treatment, but not single-agent treatment, accelerated their recovery. Taken together, these data support the concept that combined dmPGE2 and lisinopril therapy improves thrombocytopenia and survival by promoting recovery of the MK lineage, as well as the MK niche, in the setting of H-ARS.


Asunto(s)
16,16-Dimetilprostaglandina E2/uso terapéutico , Síndrome de Radiación Aguda/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Plaquetas/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Trastornos Hemorrágicos/tratamiento farmacológico , Lisinopril/uso terapéutico , Megacariocitos/efectos de los fármacos , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Síndrome de Radiación Aguda/complicaciones , Animales , Plaquetas/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Proteína C-Reactiva/análisis , Radioisótopos de Cesio , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de la radiación , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/efectos de la radiación , Femenino , Rayos gamma/efectos adversos , Trastornos Hemorrágicos/etiología , Megacariocitos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Selectina-P/análisis , Agregación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de la radiación , Factor Plaquetario 4/análisis , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/etiología , Trombocitopenia/etiología , Trombopoyesis/efectos de la radiación , Irradiación Corporal Total , Factor de von Willebrand/análisis
4.
Mol Cell Biol ; 19(4): 3029-38, 1999 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10082570

RESUMEN

Using PCR-coupled subtractive screening-representational difference analysis, we have cloned a novel gene from AML1-ETO knockin mice. This gene is highly expressed in the yolk sac and fetal liver of the knockin mice. Nucleotide sequence analysis indicates that its cDNA contains an 1,107-bp open reading frame encoding a 368-amino-acid polypeptide. Further protein sequence and protein translation analysis shows that it belongs to a family of ubiquitin-specific proteases (UBP), and its molecular mass is 43 kDa. Therefore, we have named this gene UBP43. Like other ubiquitin proteases, the UBP43 protein has deubiquitinating enzyme activity. Protein ubiquitination has been implicated in many important cellular events. In wild-type adult mice, UBP43 is highly expressed in the thymus and in peritoneal macrophages. Among nine different murine hematopoietic cell lines analyzed, UBP43 expression is detectable only in cell lines related to the monocytic lineage. Furthermore, its expression is regulated during cytokine-induced monocytic cell differentiation. We have investigated its function in the hematopoietic myeloid cell line M1. UBP43 was introduced into M1 cells by retroviral gene transfer, and several high-expressing UBP43 clones were obtained for further study. Morphologic and cell surface marker examination of UBP43/M1 cells reveals that overexpression of UBP43 blocks cytokine-induced terminal differentiation of monocytic cells. These data suggest that UBP43 plays an important role in hematopoiesis by modulating either the ubiquitin-dependent proteolytic pathway or the ubiquitination state of another regulatory factor(s) during myeloid cell differentiation.


Asunto(s)
Proteínas de Unión al ADN , Endopeptidasas/genética , Hematopoyesis/genética , Leucemia Experimental/etiología , Proteínas de Fusión Oncogénica , Proteínas Proto-Oncogénicas , Factores de Transcripción/genética , Ubiquitinas/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Clonación Molecular , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Endopeptidasas/metabolismo , Células Madre Hematopoyéticas/citología , Leucemia Experimental/genética , Leucemia Mieloide Aguda/etiología , Leucemia Mieloide Aguda/genética , Macrófagos/citología , Ratones , Ratones Noqueados , Ratones Mutantes , Datos de Secuencia Molecular , Monocitos/citología , Proteína 1 Compañera de Translocación de RUNX1 , Proteínas Recombinantes de Fusión/genética , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Ubiquitina Tiolesterasa
5.
Exp Hematol ; 29(8): 927-36, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11495698

RESUMEN

OBJECTIVE: To review the process of blood-cell formation in the murine and human yolk sac. DATA SOURCES: Most articles were selected from the PubMed database. DATA SYNTHESIS: The yolk sac is the first site of blood-cell production during murine and human ontogeny. Primitive erythroid cells originate in the yolk sac and complete their maturation, including enucleation, in the bloodstream. Though species differences exist, the pattern of hematopoietic progenitor cell emergence in the yolk sac is similar in mouse and man. In both species, there is a stage of development where both primitive red blood cells and definitive erythroid progenitors are produced in the yolk sac. An "embryonic" hematopoietic stem cell that engrafts in myeloablated newborn but not adult mice can be detected in the murine yolk sac and embryo. Stem-cell activity in the human yolk sac has not been reported. CONCLUSIONS: The yolk sac is the sole site of embryonic erythropoiesis. However, definitive erythroid, myeloid, and multipotential progenitors also originate in the yolk sac. The relationship between these progenitors and the "embryonic" hematopoietic stem cell has not been elucidated. Yolk sac-derived progenitor cells may seed the developing liver via the circulation and serve as the immediate source of the mature blood cells that are required to meet the metabolic needs of the rapidly growing fetus.


Asunto(s)
Células Sanguíneas/fisiología , Hematopoyesis/fisiología , Saco Vitelino/fisiología , Animales , Animales Recién Nacidos , Células Sanguíneas/citología , Eritropoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones
6.
Exp Hematol ; 25(11): 1149-57, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9328451

RESUMEN

Although erythropoietin is necessary for definitive (fetal liver and bone marrow) erythropoiesis, the role of erythropoietin signaling in primitive (yolk sac) hematopoiesis has not been well defined. In situ hybridization studies have revealed that erythropoietin-receptor (EPOR) mRNA accumulation begins in mesoderm cell masses of the developing yolk sac of the neural plate stage embryo (E7.5) before the development of morphologically recognizable erythroblasts. EPOR mRNA is also present in yolk sac blood islands at early somite stages (E8.5). These findings suggest that EPOR functions during early stages of yolk sac erythropoiesis. We have used a serum-free murine yolk sac explant system (Palis et al., Blood 86:156, 1995) to investigate the initial differentiation of primitive erythroblasts from extraembryonic mesoderm cells. Exogenous erythropoietin increased both erythroblast numbers and betaH1-globin accumulation in yolk sac explants, suggesting that primary yolk sac erythroblasts are directly responsive to erythropoietin. An antisense oligodeoxynucleotide (ODN) experimental approach was used to examine the functional role of erythropoietin signaling during the initiation of yolk sac hematopoiesis in yolk sac explants. Antisense EPOR ODN produced a >50% reduction (p < 0.005) in the number of differentiating primitive erythroblasts, >95% reduction in betaH1-globin accumulation (p < 0.001), and a >50% reduction (p < 0.01) in the number of CFU-E and BFU-E compared with missense EPOR ODN-treated and untreated control explants. Antisense EPOR ODN also blocked the increase in primitive erythroblast number induced by exogenous erythropoietin. We conclude that erythropoietin/EPOR signaling is functionally active during the initial proliferation and differentiation of primary yolk sac erythroblasts. These results also suggest that other growth factor signaling cascades are active during the onset of mammalian erythropoiesis.


Asunto(s)
Embrión de Mamíferos/fisiología , Eritropoyesis , Globinas/biosíntesis , Receptores de Eritropoyetina/biosíntesis , Saco Vitelino/fisiología , Animales , Ensayo de Unidades Formadoras de Colonias , Desarrollo Embrionario y Fetal , Eritroblastos/citología , Eritroblastos/fisiología , Eritropoyesis/efectos de los fármacos , Femenino , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/efectos de los fármacos , Hematopoyesis/fisiología , Ratones , Ratones Endogámicos ICR , Oligonucleótidos Antisentido/farmacología , Técnicas de Cultivo de Órganos , Receptores de Eritropoyetina/efectos de los fármacos , Receptores de Eritropoyetina/fisiología
7.
Blood Rev ; 12(2): 106-14, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9661799

RESUMEN

A newborn infant represents the culmination of developmental events from conception through organogenesis. Red cells are critically important for survival and growth of the embryo. During development, erythropoiesis occurs in two distinct forms. The first 'primitive' form consists of nucleated erythroblasts that differentiate within the blood vessels of the extraembryonic yolk sac. The second 'definitive' form consists of anucleate erythrocytes that differentiate within the liver and third trimester bone marrow of the fetus. While adult bone marrow and cord blood now serve as sources of stem cells for the treatment by transplantation of genetic and malignant diseases, the developmental origin of hematopoietic stem cells has not been determined. During the third trimester the fetus grows rapidly and the production of red cells is approximately 3-5 times that of adult steady state levels. Birth brings dramatic changes in oxygenation and erythropoietin production that result in a tenfold drop in red cell production and in a transient 'physiologic' anemia. Other causes of fetal and infant anemias have their origins in development processes. These include globin gene switching in alpha and beta thalassemia, the expression of red cell antigens in alloimmune hemolytic disease, and the poorly understood defects in the regulation of erythropoiesis in Diamond Blackfan anemia. Even in the adult, vestiges of fetal erythropoiesis are evident during transient states of accelerated erythroid expansion. A better understanding of the development of erythropoiesis will bring improvements in the treatment of anemia, not only in the newborn, but also in the fetus and the adult.


Asunto(s)
Anemia/fisiopatología , Eritropoyesis/fisiología , Anemia/genética , Anemia/inmunología , Desarrollo Embrionario y Fetal/fisiología , Células Madre Hematopoyéticas/fisiología , Humanos , Recién Nacido , Hígado/irrigación sanguínea , Hígado/embriología , Estrés Fisiológico/sangre , Síndrome , Saco Vitelino/fisiología
8.
Int Rev Immunol ; 20(1): 137-55, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11342302

RESUMEN

For decades, numerous investigators have reported derivation of macrophage-like cells from CD5(+) pre-B cell lymphomas. Recently, it has become clear that biphenotypic CD5(+) B/macrophage cells are not a spurious result of malignancy. Indeed, the existence of normal biphenotypic cells with CD5(+) B lymphocyte and macrophage characteristics has been demonstrated in the mouse. This review considers normal B/macrophage cell function in an evolutionary context where a primitive, flexible cell type could perform dual roles in adaptive and innate immunity.


Asunto(s)
Subgrupos de Linfocitos B/citología , Antígenos CD5/análisis , Macrófagos/citología , Animales , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Clonales , Reordenamiento Génico de Linfocito B , Genes ras , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/citología , Humanos , Inmunidad Innata , Inmunofenotipificación , Hígado/citología , Hígado/embriología , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Células Madre Neoplásicas/citología , Fagocitosis , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Tumorales Cultivadas
9.
Leuk Res ; 12(4): 339-44, 1988.
Artículo en Inglés | MEDLINE | ID: mdl-3374166

RESUMEN

The human leukemia cell line HL-60 consists predominantly of abnormal promyelocytes. When grown in RPMI-1640 and 10% FCS between 5 and 10% of these cells spontaneously differentiate into more mature myeloid cells, becoming smaller in size and developing the ability to generate superoxide. Centrifugal elutriation was used to separate these G0 cells from the bulk of the cycling G1, S and G2M cells. These isolated differentiating cells are shown to be similar in size, DNA content, RNA content and NBT positivity not only to granulocyte induced HL-60 cells but also to human peripheral blood granulocytes. This methodology allows the study of differentiative vs proliferative processes through the quick one-step generation of homogeneous subpopulations of G0, G1, S and G2M cells.


Asunto(s)
Separación Celular , Granulocitos/patología , Leucemia/patología , Ciclo Celular , Diferenciación Celular , Línea Celular , Separación Celular/métodos , Humanos
10.
Ann N Y Acad Sci ; 938: 96-107; discussion 108, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11458531

RESUMEN

The in vitro differentiation of embryonic stem (ES) cells provides a powerful approach for studying the earliest events involved in the commitment of the hematopoietic and endothelial lineages. Using this model system, we have identified a precursor with the potential to generate both primitive and definitive hematopoietic cells as well as cells with endothelial characteristics. The developmental potential of this precursor suggests that it represents the in vitro equivalent of the hemangioblast, a common stem cell for both lineages. ES cells deficient for the transcription factor scl/tal-1 are unable to generate hemangioblasts, while those deficient for Runx1 generate reduced numbers of these precursors. These findings indicate that both genes play pivotal roles at the earliest stages of hematopoietic and endothelial development. In addition, they highlight the strength of this model system in studying the function of genes in embryonic development.


Asunto(s)
Hematopoyesis/fisiología , Sistema Hematopoyético/embriología , Proteínas Proto-Oncogénicas , Células Madre/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula , Células Cultivadas/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Desarrollo Embrionario y Fetal , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/citología , Regulación del Desarrollo de la Expresión Génica , Genes Letales , Edad Gestacional , Hematopoyesis/genética , Células Madre Hematopoyéticas/citología , Sistema Hematopoyético/crecimiento & desarrollo , Humanos , Hígado/citología , Hígado/embriología , Linfocinas/farmacología , Ratones , Ratones Noqueados , Modelos Biológicos , Células Madre/efectos de los fármacos , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Saco Vitelino/citología
12.
Curr Top Microbiol Immunol ; 252: 293-300, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11187084
13.
Blood ; 89(4): 1154-64, 1997 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9028937

RESUMEN

Hematopoiesis in the mouse conceptus begins in the visceral yolk (VYS), with primitive erythroblasts first evident in blood islands at the headfold stage (E8.0). VYS erythropoiesis is decreased or abrogated by targeted disruption of the hematopoietic transcription factors tal-1, rbtn2, GATA-1, and GATA-2. To better understand the potential roles of these genes, and to trace the initial temporal and spatial development of mammalian embryonic hematopoiesis, we examined their expression patterns, and that of betaH1-globin, in normal mouse conceptuses by means of in situ hybridization. Attention was focused on the 36-hour period from mid-primitive streak to early somite stages (E7.25 to E8.5), when the conceptus undergoes rapid morphologic changes with formation of the yolk sac and blood islands. Each of these genes was expressed in extraembryonic mesoderm, from which blood islands are derived. This VYS expression occurred in a defined temporal sequence: tal-1 and rbtn2 transcripts were detected earlier than the others, followed by GATA-2 and GATA-1, and then by betaH1-globin. Transcripts for all of these genes were present in VYS mesoderm cell masses at the neural plate stage (E7.5), indicating commitment of these cells to the erythroid lineage before the appearance of morphologically recognizable erythroblasts. By early somite stages (E8.5), GATA-2 mRNA expression is downregulated in VYS blood islands as terminal primitive erythroid differentiation proceeds. We conclude that primitive mammalian erythropoiesis arises during gastrulation through the ordered temporal expression of tal-1, rbtn2, GATA2, and GATA-1 in a subset of extraembryonic mesoderm cells. During the stages analyzed, tal-1 and rbtn2 expression was also present in posterior embryonic mesoderm, while GATA-1 and GATA-2 expression was evident in extraembryonic tissues of ectodermal origin.


Asunto(s)
Eritropoyesis , Sistema Hematopoyético/embriología , Proteínas Proto-Oncogénicas , Factores de Transcripción/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Factor de Transcripción GATA2 , Gástrula/fisiología , Regulación del Desarrollo de la Expresión Génica , Globinas/biosíntesis , Globinas/genética , Hibridación in Situ , Proteínas con Dominio LIM , Mesodermo/metabolismo , Metaloproteínas/genética , Metaloproteínas/fisiología , Ratones , Ratones Endogámicos ICR , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/genética , Saco Vitelino/fisiología
14.
Mol Reprod Dev ; 48(2): 145-53, 1997 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9291463

RESUMEN

The visceral yolk sac (YS), a simple bilayer structure formed during gastrulation, supplies blood cells and intestine- and liver-like functions to support embryonic growth. To better understand gene regulation in extraembryonic tissues, we examined the early murine YS for expression of the homeobox family of developmental transcription regulators. We identified a subset of known homeobox sequences (Hox 1l, b1, a9, c9, a7, b7, b8, a10, cdx-1, and PDX-1), as well as two novel homeodomains consisting of a fourth labial class Hox genes and one that matches the Antennapedia class on the amino acid level. The two most frequently isolated YS Hox genes, a9 and c9, are initially expressed only in the YS (E.5) and subsequently expressed in both the embryo and YS (E8.5). Another of the identified genes, PDX-1, is involved in pancreatic development and insulin regulation. Whereas the4 rodent YS is known to produce insulin from mid to late gestation, YS insulin expression had not been examined earlier in development . We detected insulin mRNA in the YS at both E7.5 and E8.5, prior to expression in the embryo proper or formation of the pancreas. However, other pancreatic products, such as glucagon, somatostatin, and carboxypeptidase A, are not expressed in the YS. In situ analysis indicates insulin is produced in YS mesothelial cells and endoderm cells, but not in blood cells. We hypothesize the early expression of insulin in the YS is required for the expansion of insulin responsive cells including primitive erythroblasts.


Asunto(s)
Expresión Génica , Genes Homeobox , Hematopoyesis , Proteínas de Homeodominio/genética , Transactivadores/genética , Saco Vitelino/metabolismo , Animales , ADN Complementario/química , Hibridación in Situ , Insulina/metabolismo , Ratones , Páncreas/metabolismo , Reacción en Cadena de la Polimerasa
15.
Mol Reprod Dev ; 42(1): 19-27, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-8562047

RESUMEN

The visceral yolk sac (VYS), composed of extraembryonic mesoderm and visceral endoderm, is the initial site of blood cell development and serves important nutritive and absorptive functions. In the mouse, the visceral endoderm becomes a morphologically distinct tissue at the time of implantation (E4.5), while the extraembryonic mesoderm arises during gastrulation (E6.5-8.5). To isolate genes differentially expressed in the developing yolk sac, polymerase chain reaction (PCR) methods were used to construct cDNA from late primitive streak to neural plate stage (E7.5) murine VYS mesoderm and VYS endoderm tissues. Differential screening led to the identification of six VYS mesoderm-enriched clones: ribosomal protein L13a, the heat shock proteins hsc 70 and hsp 86, guanine-nucleotide binding protein-related gene, cellular nucleic acid binding protein, and alpha-enolase. One VYS endoderm-specific cDNA was identified as apolipoprotein C2. In situ hybridization studies confirmed the differential expression of these genes in E7.5 yolk sac tissues. These results indicate that representative cDNA populations can be obtained from small numbers of cells and that PCR methodologies permit the study of gene expression during early mammalian postimplantation development. While all of the mesoderm-enriched genes were ubiquitously expressed in the embryo proper, apolipoprotein C2 expression was confined to the visceral endoderm. These results are consistent with the hypothesis that at E7.5, the yolk sac endoderm provides differentiated liver-like functions, while the newly developing extraembryonic mesoderm is still a largely undifferentiated tissue.


Asunto(s)
Proteínas Fetales/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Saco Vitelino/metabolismo , Animales , Secuencia de Bases , Diferenciación Celular , ADN Complementario/genética , Endodermo/metabolismo , Femenino , Proteínas Fetales/genética , Hibridación in Situ , Masculino , Mesodermo/metabolismo , Ratones , Ratones Endogámicos ICR , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , ARN Mensajero/genética
16.
Am J Hematol ; 66(1): 46-8, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11426492

RESUMEN

A child with Wilm's tumor and a child with immune thrombocytopenic purpura (ITP) were each noted to have persistent elevations of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and lactate dehydrogenase (LDH). Both children underwent thorough evaluation for liver disease and, as a result, experienced delays in treatment of the Wilm's tumor and ITP. Eventually both children were found to have extremely elevated serum creatine kinase (CK). Muscle biopsy confirmed diagnoses of Duchenne's muscular dystrophy in one child, and Becker's muscular dystrophy in the second. Hematologists/oncologists should consider obtaining a serum CK to rule out muscle disease in patients with unexplained elevations of AST, ALT, and LDH.


Asunto(s)
Alanina Transaminasa/sangre , Aspartato Aminotransferasas/sangre , Isoenzimas/sangre , Neoplasias Renales/enzimología , L-Lactato Deshidrogenasa/sangre , Pruebas de Función Hepática , Proteínas Musculares/sangre , Distrofia Muscular de Duchenne/enzimología , Púrpura Trombocitopénica Idiopática/enzimología , Tumor de Wilms/enzimología , Biopsia , Niño , Preescolar , Creatina Quinasa/sangre , Diagnóstico Diferencial , Humanos , Neoplasias Renales/complicaciones , Hígado/enzimología , Hígado/patología , Hepatopatías/diagnóstico , Masculino , Músculos/patología , Distrofia Muscular de Duchenne/complicaciones , Distrofia Muscular de Duchenne/diagnóstico , Proteínas de Neoplasias/sangre , Púrpura Trombocitopénica Idiopática/complicaciones , Tumor de Wilms/complicaciones
17.
Blood ; 86(1): 156-63, 1995 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-7795222

RESUMEN

The blood islands of the visceral yolk sac (VYS) are the initial sites of hematopoiesis in mammals. We have developed a yolk sac explant culture system to study the process of blood cell and endothelial cell development from extraembryonic mesoderm cells. No benzidine-positive cells or beta H1-globin mRNA expression was detected at the primitive streak or neural plate stage of development (E7.5). However, when isolated E7.5 dissected tissues were cultured for 36 to 72 hours in serum-free medium, hundreds of hemoglobin-producing cells and embryonic globin gene expression were identified in both intact yolk sac and VYS mesoderm explants. Explanted E7.5 extraembryonic mesoderm tissues thus recapitulate in vivo primitive erythropoiesis and do not require the presence of a vascular network or the VYS endoderm. Yolk sac blood islands also contain endothelial cells that arise by vasculogenesis and express flk-1. We detected flk-1 mRNA as early as the primitive streak stage of mouse embryogenesis. Culture of embryo proper and intact VYS explants, which contain both mesoderm and endoderm cells, produced capillary networks and expressed flk-1. In contrast, vascular networks were not seen when VYS mesoderm was cultured alone, although flk-1 expression was similar to that of intact VYS explants. The addition of vascular endothelial growth factor to VYS mesoderm explants did not induce vascular network formation. These results suggest that the VYS endoderm or its extracellular matrix is necessary for the coalescence of developing endothelial cells into capillary networks.


Asunto(s)
Vasos Sanguíneos/embriología , Hematopoyesis Extramedular/fisiología , Mesodermo/fisiología , Saco Vitelino/fisiología , Animales , Biomarcadores , Embrión de Pollo , Medio de Cultivo Libre de Suero , Factores de Crecimiento Endotelial/farmacología , Endotelio Vascular/embriología , Endotelio Vascular/metabolismo , Gástrula/fisiología , Regulación del Desarrollo de la Expresión Génica , Linfocinas/farmacología , Mesodermo/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Técnicas de Cultivo de Órganos , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Saco Vitelino/irrigación sanguínea , Saco Vitelino/efectos de los fármacos
18.
Development ; 114(3): 769-86, 1992 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-1618141

RESUMEN

The cloning and characterization of cDNAs representing four genes or small gene families that are coordinately expressed in a spatially restricted pattern during the very early blastula (VEB) stage of sea urchin development are presented. The VEB genes encode multiple transcripts that are expressed transiently in embryos of Strongylocentrotus purpuratus between 16-cell stage and hatching, with peak abundance 12 to 15 hours post-fertilization (approximately 150-250 cells). The VEB transcripts share the same spatial pattern in the early blastula embryo: they are asymmetrically distributed along the animal-vegetal axis but their distribution around this axis is uniform. Thus, the VEB transcripts are the earliest messages to reveal asymmetry along the primary axis in the sea urchin embryo. The temporal and spatial patterns of VEB transcript accumulation are not consistent with involvement of these gene products in cell division or in tissue-specific functions. Furthermore, VEB messages cannot be detected in either ovary or adult tissues, suggesting that these genes function exclusively during embryogenesis. We suggest that the VEB genes function in constructing the early blastula. Two VEB genes encode metalloendoproteases: one (SpHE) is hatching enzyme and the other (SpAN) is similar to bone morphogenetic protein-1 (BMP-1; Wozney et al., Science 242: 1528-1534, 1988) and the Tolloid gene product (tld) (Shimell et al., Cell 67: 459-482, 1991). Several lines of evidence suggest that the VEB genes are regulated directly by factors or regulatory activities localized along the maternally specificed animal-vegetal axis.


Asunto(s)
Blastocisto/fisiología , Expresión Génica/genética , Genes/genética , ARN Mensajero/genética , Erizos de Mar/genética , Animales , Blastocisto/citología , Células Cultivadas , Clonación Molecular , Metaloendopeptidasas/genética , Técnicas de Sonda Molecular , Datos de Secuencia Molecular , Alineación de Secuencia
19.
Proc Natl Acad Sci U S A ; 98(8): 4528-33, 2001 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-11296291

RESUMEN

During mouse embryogenesis, two waves of hematopoietic progenitors originate in the yolk sac. The first wave consists of primitive erythroid progenitors that arise at embryonic day 7.0 (E7.0), whereas the second wave consists of definitive erythroid progenitors that arise at E8.25. To determine whether these unilineage hematopoietic progenitors arise from multipotential precursors, we investigated the kinetics of high proliferative potential colony-forming cells (HPP-CFC), multipotent precursors that give rise to macroscopic colonies when cultured in vitro. No HPP-CFC were found at presomite stages (E6.5-E7.5). Rather, HPP-CFC were detected first at early somite stages (E8.25), exclusively in the yolk sac. HPP-CFC were found subsequently in the bloodstream at higher levels than the remainder of the embryo proper. However, the yolk sac remains the predominant site of HPP-CFC expansion (>100-fold) until the liver begins to serve as the major hematopoietic organ at E11.5. On secondary replating, embryonic HPP-CFC give rise to definitive erythroid and macrophage (but not primitive erythroid) progenitors. Our findings support the hypothesis that definitive but not primitive hematopoietic progenitors originate from yolk sac-derived HPP-CFC during late gastrulation.


Asunto(s)
División Celular , Desarrollo Embrionario y Fetal , Células Madre Hematopoyéticas/citología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
20.
Development ; 126(22): 5073-84, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10529424

RESUMEN

In this study, we have mapped the onset of hematopoietic development in the mouse embryo using colony-forming progenitor assays and PCR-based gene expression analysis. With this approach, we demonstrate that commitment of embryonic cells to hematopoietic fates begins in proximal regions of the egg cylinder at the mid-primitive streak stage (E7.0) with the simultaneous appearance of primitive erythroid and macrophage progenitors. Development of these progenitors was associated with the expression of SCL/tal-1 and GATA-1, genes known to be involved in the development and maturation of the hematopoietic system. Kinetic analysis revealed the transient nature of the primitive erythroid lineage, as progenitors increased in number in the developing yolk sac until early somite-pair stages of development (E8.25) and then declined sharply to undetectable levels by 20 somite pairs (E9.0). Primitive erythroid progenitors were not detected in any other tissue at any stage of embryonic development. The early wave of primitive erythropoiesis was followed by the appearance of definitive erythroid progenitors (BFU-E) that were first detectable at 1-7 somite pairs (E8.25) exclusively within the yolk sac. The appearance of BFU-E was followed by the development of later stage definitive erythroid (CFU-E), mast cell and bipotential granulocyte/macrophage progenitors in the yolk sac. C-myb, a gene essential for definitive hematopoiesis, was expressed at low levels in the yolk sac just prior to and during the early development of these definitive erythroid progenitors. All hematopoietic activity was localized to the yolk sac until circulation was established (E8.5) at which time progenitors from all lineages were detected in the bloodstream and subsequently in the fetal liver following its development. This pattern of development suggests that definitive hematopoietic progenitors arise in the yolk sac, migrate through the bloodstream and seed the fetal liver to rapidly initiate the first phase of intraembryonic hematopoiesis. Together, these findings demonstrate that commitment to hematopoietic fates begins in early gastrulation, that the yolk sac is the only site of primitive erythropoiesis and that the yolk sac serves as the first source of definitive hematopoietic progenitors during embryonic development.


Asunto(s)
Desarrollo Embrionario y Fetal , Células Precursoras Eritroides/citología , Eritropoyesis , Células Madre Hematopoyéticas/citología , Saco Vitelino/embriología , Animales , Circulación Sanguínea , Eritropoyesis/genética , Regulación del Desarrollo de la Expresión Génica , Hígado/embriología , Macrófagos/citología , Ratones
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda