Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Biol Blood Marrow Transplant ; 17(4): 465-75, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20457265

RESUMEN

Stable mixed hematopoietic chimerism has been consistently established in dogs who were mildly immunosuppressed by 200 cGy of total body irradiation (TBI) before undergoing dog leukocyte antigen (DLA)-identical bone marrow (BM) transplantation and who received a brief course of immunosuppression with mycophenolate mofetil (28 days) and cyclosporine (35 days) after transplantation. However, when TBI was reduced from 200 to 100 cGy, grafts were nearly uniformly rejected within 3-12 weeks. Here, we asked whether stable engraftment could be accomplished after a suboptimal dose of 100 cGy TBI with host immunosuppression enhanced by donor-derived mesenchymal stromal cells (MSCs) given after transplantation. MSCs were cultured from BM cells and evaluated in vitro for antigen expression. They showed profound immunosuppressive properties in mixed lymphocyte reactions (MLRs) in a cell dose-dependent manner not restricted by DLA. MSC and lymphocyte contact was not required, indicating that immunosuppression was mediated by soluble factors. Prostaglandin E2 was increased in culture supernatant when MSCs were cocultured in MLRs. The addition of indomethacin restored lymphocyte proliferation in cultures containing MSCs. MSCs expressed CD10, CD13, CD29, CD44, CD73/SH-3, CD90/Thy-1, and CD106/VCAM-1. For in vivo studies, MSCs were injected on the day of BM grafting and on day 35, the day of discontinuation of posttransplantation cyclosporine. MSCs derived from the respective BM donors failed to avert BM graft rejection in 4 dogs who received DLA-identical grafts after nonmyeloablative conditioning with 100 cGy TBI in a time course not significantly different from that of control dogs not given MSCs. Although the MSCs displayed in vitro characteristics similar to those reported for MSCs from other species, their immunosuppressive qualities failed to sustain stable BM engraftment in vivo in this canine model.


Asunto(s)
Trasplante de Médula Ósea , Proliferación Celular , Rechazo de Injerto/metabolismo , Supervivencia de Injerto , Linfocitos/metabolismo , Acondicionamiento Pretrasplante , Animales , Antiinflamatorios no Esteroideos/farmacología , Antígenos CD/metabolismo , Línea Celular , Ciclosporina/farmacología , Perros , Rechazo de Injerto/patología , Antígenos de Histocompatibilidad Clase I , Terapia de Inmunosupresión/métodos , Inmunosupresores/farmacología , Linfocitos/patología , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Células del Estroma/metabolismo , Células del Estroma/patología , Factores de Tiempo , Trasplante Homólogo , Irradiación Corporal Total
2.
Nat Commun ; 12(1): 6790, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815405

RESUMEN

Lineage commitment and differentiation is driven by the concerted action of master transcriptional regulators at their target chromatin sites. Multiple efforts have characterized the key transcription factors (TFs) that determine the various hematopoietic lineages. However, the temporal interactions between individual TFs and their chromatin targets during differentiation and how these interactions dictate lineage commitment remains poorly understood. Here we perform dense, daily, temporal profiling of chromatin accessibility (DNase I-seq) and gene expression changes (total RNA-seq) along ex vivo human erythropoiesis to comprehensively define developmentally regulated DNase I hypersensitive sites (DHSs) and transcripts. We link both distal DHSs to their target gene promoters and individual TFs to their target DHSs, revealing that the regulatory landscape is organized in distinct sequential regulatory modules that regulate lineage restriction and maturation. Finally, direct comparison of transcriptional dynamics (bulk and single-cell) and lineage potential between erythropoiesis and megakaryopoiesis uncovers differential fate commitment dynamics between the two lineages as they exit the stem and progenitor stage. Collectively, these data provide insights into the temporally regulated synergy of the cis- and the trans-regulatory components underlying hematopoietic lineage commitment and differentiation.


Asunto(s)
Linaje de la Célula/genética , Cromatina/genética , Regulación del Desarrollo de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/fisiología , Línea Celular , Cromatina/metabolismo , Ensayo de Unidades Formadoras de Colonias , Desoxirribonucleasa I/metabolismo , Humanos , Leucocitos Mononucleares , Cultivo Primario de Células , Regiones Promotoras Genéticas , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/metabolismo
3.
J Biol Res (Thessalon) ; 26: 4, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31360678

RESUMEN

BACKGROUND: Hematopoiesis is a model-system for studying cellular development and differentiation. Phenotypic and functional characterization of hematopoietic progenitors has significantly aided our understanding of the mechanisms that govern fate choice, lineage specification and maturity. Methods for progenitor isolation have historically relied on complex flow-cytometric strategies based on nested, arbitrary gates within defined panels of immunophenotypic markers. The resulted populations are then functionally assessed, although functional homogeneity or absolute linkage between function and phenotype is not always achieved, thus distorting our view on progenitor biology. METHOD: In this study, we present a protocol for unbiased phenotypic identification and functional characterization which combines index sorting and clonogenic assessment of individual progenitor cells. Single-cells are plated into custom media allowing multiple hematopoietic fates to emerge and are allowed to give rise to unilineage colonies or mixed. After colony identification, lineage potential is assigned to each progenitor and finally the indexed phenotype of the initial cell is recalled and a phenotype is assigned to each functional output. CONCLUSIONS: Our approach overcomes the limitations of the current protocols expanding beyond the established cell-surface marker panels and abolishing the need for nested gating. Using this method we were able to resolve the relationships of myeloid progenitors according to the revised model of hematopoiesis, as well as identify a novel marker for erythroid progenitors. Finally, this protocol can be applied to the characterization of any progenitor cell with measurable function.

4.
Stem Cell Reports ; 5(5): 753-762, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26607951

RESUMEN

Cardiomyocytes derived from human embryonic stem cells (hESC-CMs) can improve the contractility of injured hearts.We hypothesized that mesodermal cardiovascular progenitors (hESC-CVPs), capable of generating vascular cells in addition to cardiomyocytes, would provide superior repair by contributing to multiple components of myocardium. We performed a head-to-head comparison of hESC-CMs and hESC-CVPs and compared these with the most commonly used clinical cell type, human bone marrow mononuclear cells (hBMMNCs). In a nude rat model of myocardial infarction, hESC-CMs and hESC-CVPs generated comparable grafts. Both similarly improved systolic function and ventricular dilation. Furthermore, only rare human vessels formed from hESC-CVPs. hBM-MNCs attenuated ventricular dilation and enhanced host vascularization without engrafting long-term or improving contractility. Thus, hESC-CMs and CVPs show similar efficacy for cardiac repair, and both are more efficient than hBM-MNCs. However, hESC-CVPs do not form larger grafts or more significant numbers of human vessels in the infarcted heart.


Asunto(s)
Células Madre Embrionarias/citología , Células Progenitoras Endoteliales/citología , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Trasplante de Células Madre , Animales , Células Cultivadas , Humanos , Masculino , Contracción Miocárdica , Miocitos Cardíacos/fisiología , Ratas , Ratas Sprague-Dawley , Función Ventricular
5.
J Interferon Cytokine Res ; 22(7): 765-72, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12184914

RESUMEN

Previous studies have demonstrated that the intercellular spread of cytomegalovirus (CMV) is reduced in marrow stromal cells that either secrete interleukin-1 (IL-1) or are treated with exogenous IL-1. Here, we report that IL-1-treated marrow stromal cells and fibroblasts, when infected with CMV, produce decreased amounts of infectious progeny virus. CMV-infected cells treated with IL-1 contained more interferon-beta (IFN-beta) mRNA at 24 h postinfection compared with untreated, infected cells. IFN-beta protein secreted into fibroblast culture supernatants increased from 46 +/- 1 IU/ml in untreated, infected cells to 116 +/- 5 IU/ml in IL-1-treated infected cells. When IFN-beta activity was inhibited, using blocking antibodies to either the cytokine or the IFN-alpha/beta receptor, the addition of IL-1 no longer limited viral spread. Furthermore, viral spread in nonIL-1-treated cultures was inhibited by the addition of recombinant IFN-beta. These studies suggest that IL-1 functions to limit CMV spread by increasing the expression of IFN-beta, which in turn reduces production of infectious virus.


Asunto(s)
Antivirales/farmacología , Citomegalovirus/fisiología , Interferón beta/biosíntesis , Interleucina-1/farmacología , Replicación Viral/efectos de los fármacos , Médula Ósea/virología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/virología , Humanos , Interferón beta/genética , Interferón beta/fisiología , Interleucina-1/genética , Proteínas Recombinantes/farmacología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo , Células del Estroma/virología
6.
J Clin Virol ; 25 Suppl 2: S51-6, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12361756

RESUMEN

Cytomegalovirus (CMV) has long been associated with myelosuppression. Evidence for this association has been provided from in vitro studies, statistical analysis of clinical studies, informative case reports, and murine models. Reports differ as to how CMV mediates myelosuppression. Some data indicate direct infection of hematopoietic progenitors and their progeny, others indicate that the supportive microenvironment is infected and thereby its supportive function compromised. In this report we review data suggesting that the severity of myelosuppression in patients is associated with particular CMV genotypes identified by variations in the glycoprotein B gene.


Asunto(s)
Infecciones por Citomegalovirus/fisiopatología , Citomegalovirus/patogenicidad , Células Madre Hematopoyéticas/fisiología , Trasplante de Células Madre/efectos adversos , Animales , Células de la Médula Ósea/fisiología , Células de la Médula Ósea/virología , Células Cultivadas , Citomegalovirus/genética , Infecciones por Citomegalovirus/virología , Femenino , Fibroblastos , Variación Genética , Hematopoyesis , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Proteínas del Envoltorio Viral/genética
7.
Leuk Lymphoma ; 45(12): 2403-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15621752

RESUMEN

IL-7 was first isolated in 1988, when its role in early B lymphocyte development was noted [1-3]. It soon became apparent that IL-7 is a "stromal cytokine" produced by a variety of stromal tissues including those in bone marrow and thymus [4-10]. The production of IL-7 by bone marrow stromal cells is thought to be essential for early B lymphocyte development at least in the murine model, and secreted IL-7 from extra thymic sources including bone marrow is postulated to play a critical role in post-thymic T cell homeostasis [11-15]. Although IL-7 was thought to have effects mostly within the lymphoid populations, the demonstration of IL-7 receptors in non-lymphoid cells including primary marrow stromal cells has challenged this paradigm [15]. In this report, we present data showing that functional IL-7 receptors are expressed by marrow stroma and that IL-7 mediated signaling through this receptor is distinct from that mediated by IL-7 receptors expressed on lymphoid cells.


Asunto(s)
Células de la Médula Ósea/metabolismo , Receptores de Interleucina-7/metabolismo , Células del Estroma/metabolismo , Animales , Diferenciación Celular , Interleucina-7/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Células del Estroma/citología
8.
PLoS One ; 9(10): e109304, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25275584

RESUMEN

In vitro expanded bone marrow stromal cells contain at least two populations of fibroblasts, a CD146/MCAM positive population, previously reported to be critical for establishing the stem cell niche and a CD146-negative population that expresses CUB domain-containing protein 1 (CDCP1)/CD318. Immunohistochemistry of marrow biopsies shows that clusters of CDCP1+ cells are present in discrete areas distinct from areas of fibroblasts expressing CD146. Using a stromal cell line, HS5, which approximates primary CDCP1+ stromal cells, we show that binding of an activating antibody against CDCP1 results in tyrosine-phosphorylation of CDCP1, paralleled by phosphorylation of Src Family Kinases (SFKs) Protein Kinase C delta (PKC-δ). When CDCP1 expression is knocked-down by siRNA, the expression and secretion of myelopoietic cytokines is increased. These data suggest CDCP1 expression can be used to identify a subset of marrow fibroblasts functionally distinct from CD146+ fibroblasts. Furthermore the CDCP1 protein may contribute to the defining function of these cells by regulating cytokine expression.


Asunto(s)
Antígenos CD/análisis , Células de la Médula Ósea/inmunología , Antígeno CD146/análisis , Moléculas de Adhesión Celular/análisis , Fibroblastos/inmunología , Proteínas de Neoplasias/análisis , Adulto , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Neoplasias , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Antígeno CD146/inmunología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/inmunología , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Inmunofenotipificación , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Proteína Quinasa C-delta/análisis , Proteína Quinasa C-delta/inmunología , Interferencia de ARN , ARN Interferente Pequeño/genética , Células del Estroma/citología , Células del Estroma/inmunología , Células del Estroma/metabolismo , Familia-src Quinasas/análisis , Familia-src Quinasas/inmunología
9.
Stem Cells Dev ; 23(7): 729-40, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24131213

RESUMEN

Marrow stromal cells constitute a heterogeneous population of cells, typically isolated after expansion in culture. In vivo, stromal cells often exist in close proximity or in direct contact with monocyte-derived macrophages, yet their interaction with monocytes is largely unexplored. In this report, isolated CD146(+) and CD146(-) stromal cells, as well as immortalized cell lines representative of each (designated HS27a and HS5, respectively), were shown by global DNase I hypersensitive site mapping and principal coordinate analysis to have a lineage association with marrow fibroblasts. Gene expression profiles generated for the CD146(+) and CD146(-) cell lines indicate significant differences in their respective transcriptomes, which translates into differences in secreted factors. Consequently, the conditioned media (CM) from these two populations induce different fates in peripheral blood monocytes. Monocytes incubated in CD146(+) CM acquire a tissue macrophage phenotype, whereas monocytes incubated in CM from CD146(-) cells express markers associated with pre-dendritic cells. Importantly, when CD14(+) monocytes are cultured in contact with the CD146(+) cells, the combined cell populations, assayed as a unit, show increased levels of transcripts associated with organismal development and hematopoietic regulation. In contrast, the gene expression profile from cocultures of monocytes and CD146(-) cells does not differ from that obtained when monocytes are cultured with CD146(-) CM. These in vitro results show that the CD146(+) marrow stromal cells together with monocytes increase the expression of genes relevant to hematopoietic regulation. In vivo relevance of these data is suggested by immunohistochemistry of marrow biopsies showing juxtaposed CD146(+) cells and CD68(+) cells associated with these upregulated proteins.


Asunto(s)
Células Madre Adultas/citología , Células de la Médula Ósea/citología , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Monocitos/citología , Adulto , Células Madre Adultas/metabolismo , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Antígeno CD146/genética , Antígeno CD146/metabolismo , Diferenciación Celular , Línea Celular Transformada , Linaje de la Célula/fisiología , Mapeo Cromosómico , Citocinas/genética , Citocinas/metabolismo , Desoxirribonucleasa I/química , Fibroblastos/metabolismo , Citometría de Flujo , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Monocitos/metabolismo
10.
Stem Cells Dev ; 22(13): 1932-43, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23391309

RESUMEN

Platelet-derived growth factors (PDGFs) and their tyrosine kinase receptors play instrumental roles in embryonic organogenesis and diseases of adult organs. In particular, platelet-derived growth factor receptor-alpha (PDGFRα) is expressed by multipotent cardiovascular progenitors in mouse and human embryonic stem cell systems. Although cardiac PDGFRα expression has been studied in multiple species, little is known about its expression in the human heart. Using immunofluorescence, we analyzed PDGFRα expression in both human fetal and diseased adult hearts, finding strong expression in the interstitial cells of the epicardium, myocardium, and endocardium, as well as the coronary smooth muscle. Only rare endothelial cells and cardiomyocytes expressed PDGFRα. This pattern was consistent for both the fetal and adult diseased hearts, although more PDGFRα+ cardiomyocytes were noted in the latter. In vitro differentiation assays were then performed on the PDGFRα+ cell fraction isolated from the cardiomyocyte-depleted human fetal hearts. Protocols previously reported to direct differentiation to a cardiomyocyte (5-azacytidine), smooth muscle (PDGF-BB), or endothelial cell fates (vascular endothelial growth factor [VEGF]) were used. Although no significant cardiomyocyte differentiation was observed, PDGFRα+ cells generated significant numbers of smooth muscle cells (smooth muscle-α-actin+ and smooth muscle myosin+) and endothelial cells (CD31+). These data suggest that a subfraction of the cardiac PDGFRα+ populations are progenitors contributing predominantly to the vascular and mesenchymal compartments of the human heart. It may be possible to control the fate of these progenitors to promote vascularization or limit fibrosis in the injured heart.


Asunto(s)
Linfocinas/biosíntesis , Miocardio/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Células Madre/metabolismo , Animales , Azacitidina/farmacología , Diferenciación Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Lesiones Cardíacas/metabolismo , Lesiones Cardíacas/terapia , Humanos , Linfocinas/metabolismo , Ratones , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/crecimiento & desarrollo , Miocardio/patología , Miocitos Cardíacos/citología , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/citología , Células Madre/efectos de los fármacos
11.
PLoS One ; 8(3): e57179, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520463

RESUMEN

In the current study, we used a canine model of radiation-induced lung injury to test the effect of a single i.v. infusion of 10×10(6)/kg of marrow fibroblasts on the progression of damage following 15 Gy exposure to the right lung. The fibroblasts, designated DS1 cells, are a cloned population of immortalized cells isolated from a primary culture of marrow stromal cells. DS1 cells were infused at week 5 post-irradiation when lung damage was evident by imaging with high-resolution computed tomography (CT). At 13 weeks post-irradiation we found that 4 out of 5 dogs receiving DS1 cells had significantly improved pulmonary function compared to 0 out of 5 control dogs (p = 0.047, Fisher's Exact). Pulmonary function was measured as the single breath diffusion capacity-hematocrit (DLCO-Hct), the total inspiratory capacity (IC), and the total lung capacity (TLC), which differed significantly between control and DS1-treated dogs; p = 0.002, p = 0.005, and p = 0.004, respectively. The DS1-treated dogs also had less pneumonitis detected by CT imaging and an increased number of TTF-1 (thyroid transcription factor 1, NKX2-1) positive cells in the bronchioli and alveoli compared to control dogs. Endothelial-like progenitor cells (ELC) of host origin, detected by colony assays, were found in peripheral blood after DS1 cell infusion. ELC numbers peaked one day after infusion, and were not detectable by 7 days. These data suggest that infusion of marrow fibroblasts stimulates mobilization of ELC, which is associated with a reduction in otherwise progressive radiation-induced lung injury. We hypothesize that these two observations are related, specifically that circulating ELC contribute to increased angiogenesis, which facilitates endogenous lung repair.


Asunto(s)
Células de la Médula Ósea , Bronquiolos/lesiones , Trasplante de Células , Fibroblastos/trasplante , Enfermedades Pulmonares/terapia , Alveolos Pulmonares/lesiones , Traumatismos Experimentales por Radiación/terapia , Animales , Bronquiolos/patología , Línea Celular Transformada , Perros , Femenino , Enfermedades Pulmonares/etiología , Enfermedades Pulmonares/patología , Masculino , Alveolos Pulmonares/patología , Traumatismos Experimentales por Radiación/patología , Células del Estroma/trasplante , Factores de Tiempo
12.
Blood ; 109(1): 85-92, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16954500

RESUMEN

Regulatory molecules produced by stromal cells are often membrane bound until cleaved by matrix metalloproteinases (MMPs); cleavage can either activate or inactivate regulatory functions. We report here that marrow stromal cells induce the expression of MMP-9 in monocytes. Induction was contact independent and could be reproduced with recombinant MCP-1/CCL2, whereas IL-6, M-CSF, G-CSF, GM-CSF, IL-8/CXCL8, SDF-1/CXCL12, and MGSA/CXCL1 did not have this effect. Stroma-induced levels of MMP-9 in the monocyte population from healthy donors were relatively consistent, whereas induced levels varied significantly (P < .001) in the CD14+ population from 27 patients with myelodysplastic syndrome (MDS). In patients with a clonal chromosomal marker, the level of inducible MMP-9 expression in the monocyte population was inversely correlated with the percentage of marker-positive cells (n = 11, P = .01), suggesting that the ability to induce MMP-9 may be compromised in clonally derived monocytes. The inducible levels of MMP-9 were also inversely correlated with marrow cellularity observed in biopsies from MDS patients (P < .001). We conclude that monocytes can express MMP-9 in response to stromal factors and that this response may be significantly decreased in MDS-derived monocytes.


Asunto(s)
Médula Ósea/patología , Metaloproteinasa 9 de la Matriz/biosíntesis , Monocitos/enzimología , Síndromes Mielodisplásicos/enzimología , Adulto , Anciano , Células Cultivadas/metabolismo , Quimiocinas/farmacología , Medios de Cultivo Condicionados/farmacología , Citocinas/farmacología , Inducción Enzimática , Femenino , Fibroblastos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Receptores de Lipopolisacáridos/análisis , Masculino , Metaloproteinasa 9 de la Matriz/genética , Persona de Mediana Edad , Monocitos/efectos de los fármacos , Monocitos/patología , Síndromes Mielodisplásicos/patología , ARN Mensajero/biosíntesis , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo
13.
Blood ; 107(9): 3520-6, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16391012

RESUMEN

The marrow microenvironment consists of several different interacting cell types, including hematopoietic-derived monocyte/macrophages and nonhematopoietic-derived stromal cells. Gene-expression profiles of stromal cells and monocytes cultured together differ from those of each population alone. Here, we report that CXCL7 gene expression, previously described as limited to the megakaryocyte lineage, is expressed by monocytes cocultured with stromal cells. CXCL7 gene expression was confirmed by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), and secretion of protein was detected by enzyme-linked immunosorbent assay (ELISA) and Western blot. At least 2 stromal-derived activities, one yet to be identified, were required for optimal expression of CXCL7 by monocytes. NAP-2, the shortest form of CXCL7 detected in the coculture media, was confirmed to decrease the size and number of CFU-Meg colonies. The propeptide LDGF, previously reported to be mitogenic for fibroblasts, was not secreted by stimulated monocytes. The recombinant form of LDGF produced in a prokaryotic expression system did not have biologic activity in our hands. The monocytic source of CXCL7 was also detected by immunohistochemistry in normal bone marrow biopsies, indicating an in vivo function. We conclude that stromal-stimulated monocytes can serve as an additional source for CXCL7 peptides in the microenvironment and may contribute to the local regulation of megakaryocytopoiesis.


Asunto(s)
Células de la Médula Ósea/inmunología , Monocitos/inmunología , beta-Tromboglobulina/biosíntesis , beta-Tromboglobulina/genética , Animales , Secuencia de Bases , Células de la Médula Ósea/metabolismo , Línea Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados , ADN Complementario/genética , Perfilación de la Expresión Génica , Humanos , Receptores de Lipopolisacáridos/metabolismo , Ratones , Monocitos/metabolismo , Células 3T3 NIH , Células del Estroma/inmunología , Células del Estroma/metabolismo , Trombopoyesis
14.
Wilehm Roux Arch Dev Biol ; 190(2): 83-86, 1981 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28305355

RESUMEN

Fibronectin, with a subunit molecular weight of 220,000 daltons, was isolated from the ovary of the sea urchin,Pseudocentrotus depressus, using affinity chromatography on heat-denatured mammalian collagen coupled to Sepharose 4B. The distribution of fibronectin in the sea urchin ovary was examined by indirect immunofluorescence using antifibronectin serum. The basement membrane and the connective tissues exhibited strong fluorescence. The fibronectin was localized closely together with collagen bundles in the sea urchin ovary. Biochemical and immunological examinations indicate that sea urchin fibronectin has similar properties as those of mammalian fibronectin.

15.
Wilehm Roux Arch Dev Biol ; 193(2): 71-77, 1984 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28305588

RESUMEN

Some plant lectins, Concanavalin agglutinin (Con A), succinyl Con A and wheat germ agglutinin (WGA) increased the adhesion of dissociated embryonic cells of the sea urchin,Pseudocentrotus depressus, to the substratum (plastic and glass surface) in vitro. Other plant lectins,Ulex europeus agglutinin (UEA) andDolichos biflorus agglutinin (DBA) had no effect on the cell-to-substratum interaction. A specific monocarbohydrate inhibitor of lectins, α-methyl-D-mannoside, inhibited the Con A-induced cell-to-substratum adhesion of dissociated embryonic cells. This observation suggests that the Con A-induced cell-to-substratum adhesion may be attributed to the Con A-carbohydrate interaction. In Millipore-filtered sea water (MPFSW) containing Con A (0.1 mg/ml), dissociated embryonic cells adhered to the substratum for more than 6 h at 18°C, while in MPFSW as control, almost all the dissociated cells were released from the substratum after 1 h. A scanning electron microscopic study showed that dissociated embryonic cells adhered to the substratum were surrounded by an extracellular fibrous material, when the cells were cultured in MPFSW containing Con A. The induction of the extracellular fibrous material by Con A was inhibited by α-methyl-D-mannoside. The appearance of this material may be related to the cell-to-substratum adhesion of dissociated cells. Sequential extractions of Con A-treated dissociated cells with Triton X 100 and urea solubilized most of the cellular components, leaving the fibrous material on the surface. Biochemical conponents of the isolated fibrous material included sea urchin fibronectin, Con A and minor components (88 and 140 kilodalton proteins). Fibronectin preformed in the cells was excreted after the dissociation, while the 88 and 140 kilodalton proteins were synthesized and released to the extracellular space.

16.
Blood ; 100(4): 1318-25, 2002 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-12149213

RESUMEN

DNA spotted microarrays were used to compare gene expression profiles from 2 functionally distinct human marrow stromal cell lines: HS-27a, which supports cobblestone area formation by early hematopoietic progenitors, and HS-5, which secretes multiple cytokines that support the proliferation of committed progenitors. One unexpected result was the high level of interleukin-7 receptor (IL-7R) gene expression in HS-27a stromal cells. Northern blot analysis confirmed the IL-7R RNA expression, and Western blots for the IL-7R protein detected both a full-length (90-kd) IL-7R and a smaller 30-kd fragment in both HS-27a cells and primary stromal cell cultures, whereas only the 90-kd receptor protein was detected in peripheral blood mononuclear cells. Biotinylated IL-7 was shown to bind to HS-27a cells under physiologic conditions, and this binding was inhibited by blocking anti-IL-7 antibodies. Tyrosine phosphorylation of several proteins (55 kd, 30 kd, and 24 kd) in HS-27a cells was rapidly increased after incubation with recombinant IL-7. One of the phosphorylated proteins proved to be the 30-kd IL-7R fragment. Exposure of HS-27a cells to IL-7 resulted in a 10-fold increase in secretion of IL-6 into culture supernatants but no increase in the cytokines stromal cell-derived factor 1, macrophage inflammatory protein 1 alpha, or IL-1 beta. The up-regulation of IL-6 secretion is associated with a rapid but transient increase in detectable levels of IL-6 messenger RNA. These data suggest that IL-7 may function to regulate the milieu of the microenvironment by modulating IL-6 secretion by the IL-7R-expressing stromal elements.


Asunto(s)
Células de la Médula Ósea/metabolismo , Expresión Génica , Interleucina-6/metabolismo , Interleucina-7/metabolismo , Receptores de Interleucina-7/genética , Células del Estroma/metabolismo , Anticuerpos/farmacología , Northern Blotting , Western Blotting , División Celular , Línea Celular , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Humanos , Interleucina-6/genética , Interleucina-7/inmunología , Interleucina-7/farmacología , Fosforilación , Fosfotirosina/metabolismo , ARN Mensajero/análisis , Receptores de Interleucina-7/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología
17.
Biol Blood Marrow Transplant ; 10(12): 826-33, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15570251

RESUMEN

A multivariate analysis of 121 dogs conditioned with 200, 100, or 50 cGy of total body irradiation (TBI) followed by hematopoietic stem cell transplantation from matched littermates showed that TBI dose was the only factor examined that was statistically significantly associated with the percentage of donor myeloid engraftment in stable long-term chimeras ( P = .008). To understand the direct effects of low-dose irradiation on hematopoietic stem/progenitor cells, nonirradiated and irradiated human CD34 + cells were evaluated for competitive repopulating ability in nonobese diabetic/severe combined immunodeficiency beta2m -/- mice. As expected, the results showed a radiation dose-dependent loss of competitive repopulating ability. Flow cytometric analysis indicated that, within a viable cell gate, there was reduced expression of P-selectin glycoprotein ligand-1 and L selectin on irradiated compared with nonirradiated CD34 + cells; this suggests that irradiated stem/progenitor cells may be compromised in their ability to home to or interact with the marrow microenvironment. However, the CD34 + /P-selectin glycoprotein ligand-1 dim cells also showed activation of caspase-3, indicating that they were destined to die. These results suggest that the TBI dose determines the degree of myeloid engraftment by compromising the resident stem/progenitor cell compartment.


Asunto(s)
Terapia de Inmunosupresión/métodos , Trasplante de Células Madre , Trasplante Heterólogo/fisiología , Irradiación Corporal Total , Animales , Antígenos CD/sangre , Antígenos CD34/sangre , Perros , Relación Dosis-Respuesta en la Radiación , Células Madre Hematopoyéticas/inmunología , Humanos , Inmunosupresores/farmacología , Repeticiones de Minisatélite , Modelos Animales
18.
Blood ; 103(12): 4496-502, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-14996707

RESUMEN

The hematopoietic microenvironment, approximated in vitro by long-term marrow cultures (LTCs), consists of both nonhematopoietic-derived stromal elements and hematopoietic-derived monocyte/macrophages. To better understand the consequences of monocyte-stroma interactions, we compared gene expression profiles of CD14+ peripheral blood monocytes and HS-27a stromal cells cultured alone and together in cocultures. Results from 7 separate experiments revealed 22 genes were significantly up- or down-regulated in the cocultures, with osteopontin (OPN) up-regulated more than 15-fold. The microarray OPN data were confirmed by Northern blot, real-time polymerase chain reaction (PCR), and by detection of OPN protein. High levels of OPN gene expression were also detected in 2- to 3-week-old primary LTCs. Using Transwells we determined that stromal cells were secreting a factor that up-regulated OPN gene expression in CD14+ cells. When CD34+ cells were cultured in the presence of purified OPN, tyrosine phosphorylation of a 34-kDa molecule was increased 2- to 3-fold, an effect that was diminished in the presence of an OPN neutralizing monoclonal antibody. In addition, Notch1 gene expression was decreased 5-fold in OPN-treated CD34+ cells. We conclude that interactions between stroma and monocytes can result in activities that limit the role of Notch signaling in hematopoietic regulation.


Asunto(s)
Células de la Médula Ósea/fisiología , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/fisiología , Monocitos/fisiología , Receptores de Superficie Celular/genética , Sialoglicoproteínas/metabolismo , Células del Estroma/fisiología , Factores de Transcripción , Antígenos CD/análisis , Antígenos CD34/análisis , Células de la Médula Ósea/citología , Línea Celular , Células Cultivadas , Medios de Cultivo Condicionados , Cartilla de ADN , Humanos , Masculino , Monocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteopontina , Fosforilación , Fosfotirosina/metabolismo , Reacción en Cadena de la Polimerasa , Receptor Notch1 , Valores de Referencia , Sialoglicoproteínas/farmacología , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA