Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
J Cell Sci ; 125(Pt 19): 4640-50, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22767507

RESUMEN

The mechanisms underlying the immunomodulatory effects of mesenchymal stem cells (MSCs) have been investigated under extreme conditions of strong T cell activation, which induces the rapid death of activated lymphocytes. The objective of this study was to investigate these mechanisms in the absence of additional polyclonal activation. In co-cultures of peripheral mononuclear blood cells with human MSCs (hereafter referred to as hMSCs), we observed a striking decrease in the level of CD8 expression on CD8+ cells, together with decreased expression of CD28 and CD44, and impaired production of IFN-gamma and Granzyme B. This effect was specific to hMSCs, because it was not observed with several other cell lines. Downregulation of CD8 expression required CD14+ monocytes to be in direct contact with the CD8+ cells, whereas the effects of hMSCs on the CD14+ cells were essentially mediated by soluble factors. The CD14+ monocytes exhibited a tolerogenic pattern when co-cultured with hMSCs, with a clear decrease in CD80 and CD86 co-stimulatory molecules, and an increase in the inhibitory receptors ILT-3 and ILT-4. CD8+ cells that were preconditioned by MSCs had similar effects on monocytes and were able to inhibit lymphocyte proliferation. Injection of hMSCs in humanized NSG mice showed similar trends, in particular decreased levels of CD44 and CD28 in human immune cells. Our study demonstrates a new immunomodulation mechanism of action of hMSCs through the modulation of CD8+ cells towards a non-cytotoxic and/or suppressive phenotype. This mechanism of action has to be taken into account in clinical trials, where it should be beneficial in grafts and autoimmune diseases, but potentially detrimental in malignant diseases.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica/inmunología , Células Madre Mesenquimatosas/citología , Monocitos/citología , Monocitos/inmunología , Adulto , Animales , Biomarcadores/metabolismo , Antígenos CD8/metabolismo , Moléculas de Adhesión Celular/metabolismo , Regulación hacia Abajo , Femenino , Humanos , Inmunización , Receptores de Lipopolisacáridos/metabolismo , Activación de Linfocitos/genética , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Transcripción Genética
2.
Blood ; 119(11): 2478-88, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22279055

RESUMEN

The mechanisms of hematopoietic progenitor cell egress and clinical mobilization are not fully understood. Herein, we report that in vivo desensitization of Sphingosine-1-phosphate (S1P) receptors by FTY720 as well as disruption of S1P gradient toward the blood, reduced steady state egress of immature progenitors and primitive Sca-1(+)/c-Kit(+)/Lin(-) (SKL) cells via inhibition of SDF-1 release. Administration of AMD3100 or G-CSF to mice with deficiencies in either S1P production or its receptor S1P(1), or pretreated with FTY720, also resulted in reduced stem and progenitor cell mobilization. Mice injected with AMD3100 or G-CSF demonstrated transient increased S1P levels in the blood mediated via mTOR signaling, as well as an elevated rate of immature c-Kit(+)/Lin(-) cells expressing surface S1P(1) in the bone marrow (BM). Importantly, we found that S1P induced SDF-1 secretion from BM stromal cells including Nestin(+) mesenchymal stem cells via reactive oxygen species (ROS) signaling. Moreover, elevated ROS production by hematopoietic progenitor cells is also regulated by S1P. Our findings reveal that the S1P/S1P(1) axis regulates progenitor cell egress and mobilization via activation of ROS signaling on both hematopoietic progenitors and BM stromal cells, and SDF-1 release. The dynamic cross-talk between S1P and SDF-1 integrates BM stromal cells and hematopoeitic progenitor cell motility.


Asunto(s)
Quimiocina CXCL12/metabolismo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Lisoesfingolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Bencilaminas , Médula Ósea/metabolismo , Movimiento Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Ciclamas , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Esfingosina/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo
3.
Blood ; 117(2): 419-28, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20585044

RESUMEN

Mechanisms governing stress-induced hematopoietic progenitor cell mobilization are not fully deciphered. We report that during granulocyte colony-stimulating factor-induced mobilization c-Met expression and signaling are up-regulated on immature bone marrow progenitors. Interestingly, stromal cell-derived factor 1/CXC chemokine receptor-4 signaling induced hepatocyte growth factor production and c-Met activation. We found that c-Met inhibition reduced mobilization of both immature progenitors and the more primitive Sca-1(+)/c-Kit(+)/Lin(-) cells and interfered with their enhanced chemotactic migration to stromal cell-derived factor 1. c-Met activation resulted in cellular accumulation of reactive oxygen species by mammalian target of rapamycin inhibition of Forkhead Box, subclass O3a. Blockage of mammalian target of rapamycin inhibition or reactive oxygen species signaling impaired c-Met-mediated mobilization. Our data show dynamic c-Met expression and function in the bone marrow and show that enhanced c-Met signaling is crucial to facilitate stress-induced mobilization of progenitor cells as part of host defense and repair mechanisms.


Asunto(s)
Movimiento Celular/fisiología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Animales , Quimiocina CXCL12/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Factor de Crecimiento de Hepatocito/metabolismo , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Nat Med ; 12(6): 657-64, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16715089

RESUMEN

Here we investigated the potential role of bone-resorbing osteoclasts in homeostasis and stress-induced mobilization of hematopoietic progenitors. Different stress situations induced activity of osteoclasts (OCLs) along the stem cell-rich endosteum region of bone, secretion of proteolytic enzymes and mobilization of progenitors. Specific stimulation of OCLs with RANKL recruited mainly immature progenitors to the circulation in a CXCR4- and MMP-9-dependent manner; however, RANKL did not induce mobilization in young female PTPepsilon-knockout mice with defective OCL bone adhesion and resorption. Inhibition of OCLs with calcitonin reduced progenitor egress in homeostasis, G-CSF mobilization and stress situations. RANKL-stimulated bone-resorbing OCLs also reduced the stem cell niche components SDF-1, stem cell factor (SCF) and osteopontin along the endosteum, which was associated with progenitor mobilization. Finally, the major bone-resorbing proteinase, cathepsin K, also cleaved SDF-1 and SCF. Our findings indicate involvement of OCLs in selective progenitor recruitment as part of homeostasis and host defense, linking bone remodeling with regulation of hematopoiesis.


Asunto(s)
Resorción Ósea , Huesos/anatomía & histología , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Osteoclastos/metabolismo , Animales , Proteínas Portadoras/metabolismo , Catepsina K , Catepsinas/genética , Catepsinas/metabolismo , Línea Celular , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Homeostasis , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos , Ratones Noqueados , Osteoclastos/citología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Proteínas Tirosina Fosfatasas Clase 4 Similares a Receptores , Receptores CXCR4/metabolismo , Factor de Células Madre/metabolismo
5.
Stem Cells ; 29(11): 1672-83, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21898690

RESUMEN

p63, a member of the p53 family, is essential for skin morphogenesis and epithelial stem cell maintenance. Here, we report an unexpected role of TAp63 in cardiogenesis. p63 null mice exhibit severe defects in embryonic cardiac development, including dilation of both ventricles, a defect in trabeculation and abnormal septation. This was accompanied by myofibrillar disarray, mitochondrial disorganization, and reduction in spontaneous calcium spikes. By the use of embryonic stem cells (ESCs), we show that TAp63 deficiency prevents expression of pivotal cardiac genes and production of cardiomyocytes. TAp63 is expressed by endodermal cells. Coculture of p63-knockdown ESCs with wild-type ESCs, supplementation with Activin A, or overexpression of GATA-6 rescue cardiogenesis. Therefore, TAp63 acts in a non-cell-autonomous manner by modulating expression of endodermal factors. Our findings uncover a critical role for p63 in cardiogenesis that could be related to human heart disease.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Corazón/embriología , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Células Madre Embrionarias/ultraestructura , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Corazón/crecimiento & desarrollo , Immunoblotting , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Fosfoproteínas/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo , Transactivadores/genética
6.
Biochem Biophys Res Commun ; 395(1): 131-5, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20361935

RESUMEN

Heterozygous mutations in the TP63 transcription factor underlie the molecular basis of several similar autosomal dominant ectodermal dysplasia (ED) syndromes. Here we provide a novel cellular model derived from embryonic stem (ES) cells that recapitulates in vitro the main steps of embryonic skin development. We show that ES cells carrying AEC or EEC mutations are unable to differentiate into the epidermal fate. Comparative transcriptome analysis strongly reveals an embryonic epidermal signature and suggests that mutations in the SAM domain (AEC) provide activating properties while mutations in the DBD domain (EEC) induce strong inhibitory capabilities. Our model uncovers the effect of relevant ED mutations that otherwise are difficult to evaluate on the ectodermal embryonic stage, an embryonic event critical for proper skin formation.


Asunto(s)
Displasia Ectodérmica/genética , Células Madre Embrionarias/fisiología , Modelos Biológicos , Piel/embriología , Transactivadores/genética , Proteínas Supresoras de Tumor/genética , Animales , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Ratones , Estructura Terciaria de Proteína , Factores de Transcripción
7.
Brain Behav Immun ; 23(8): 1059-65, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19341792

RESUMEN

The nervous system regulates immunity through hormonal and neuronal routes as part of host defense and repair mechanism. Here, we review the emerging evidence for regulation of human hematopoietic stem and progenitor cells (HSPC) by the nervous system both directly and indirectly via their bone marrow (BM) niche-supporting stromal cells. Functional expression of several neurotransmitter receptors was demonstrated on HSPC, mainly on the more primitive CD34(+)/CD38(-/low) fraction. The myeloid cytokines, G-CSF and GM-CSF, dynamically upregulate neuronal receptor expression on human HSPC. This is followed by an increased response to neurotransmitters, leading to enhanced proliferation and motility of human CD34(+) progenitors, repopulation of the murine BM and their egress to the circulation. Importantly, recent observations showed rapid mobilization of human HSPC to high SDF-1 expressing ischemic tissues of stroke individuals followed by neoangiogenesis, neurological and functional recovery. Along with decreased levels of circulating immature CD34(+) cells and SDF-1 blood levels found in patients with early-stage Alzheimer's disease, these findings suggest a possible involvement of human HSPC in brain homeostasis and thus their potential clinical applications in neuropathology.


Asunto(s)
Antígenos CD34/metabolismo , Células Madre Hematopoyéticas/metabolismo , Sistema Nervioso/metabolismo , Receptores de Neurotransmisores/metabolismo , Médula Ósea/metabolismo , Hematopoyesis , Humanos , Neurotransmisores/metabolismo , Células del Estroma/metabolismo
8.
J Clin Invest ; 112(2): 160-9, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12865405

RESUMEN

Hematopoietic stem cells rarely contribute to hepatic regeneration, however, the mechanisms governing their homing to the liver, which is a crucial first step, are poorly understood. The chemokine stromal cell-derived factor-1 (SDF-1), which attracts human and murine progenitors, is expressed by liver bile duct epithelium. Neutralization of the SDF-1 receptor CXCR4 abolished homing and engraftment of the murine liver by human CD34+ hematopoietic progenitors, while local injection of human SDF-1 increased their homing. Engrafted human cells were localized in clusters surrounding the bile ducts, in close proximity to SDF-1-expressing epithelial cells, and differentiated into albumin-producing cells. Irradiation or inflammation increased SDF-1 levels and hepatic injury induced MMP-9 activity, leading to both increased CXCR4 expression and SDF-1-mediated recruitment of hematopoietic progenitors to the liver. Unexpectedly, HGF, which is increased following liver injury, promoted protrusion formation, CXCR4 upregulation, and SDF-1-mediated directional migration by human CD34+ progenitors, and synergized with stem cell factor. Thus, stress-induced signals, such as increased expression of SDF-1, MMP-9, and HGF, recruit human CD34+ progenitors with hematopoietic and/or hepatic-like potential to the liver of NOD/SCID mice. Our results suggest the potential of hematopoietic CD34+/CXCR4+cells to respond to stress signals from nonhematopoietic injured organs as an important mechanism for tissue targeting and repair.


Asunto(s)
Antígenos CD34/biosíntesis , Quimiocinas CXC/fisiología , Factor de Crecimiento de Hepatocito/fisiología , Hígado/metabolismo , Metaloproteinasa 9 de la Matriz/fisiología , Células Madre/citología , Animales , Conductos Biliares/metabolismo , Movimiento Celular , Células Cultivadas , Quimiocina CXCL12 , ADN/metabolismo , Células Madre Hematopoyéticas , Humanos , Inmunohistoquímica , Hígado/patología , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones SCID , Microscopía Fluorescente , ARN Mensajero/metabolismo , Receptores CXCR4/metabolismo , Distribución Tisular , Regulación hacia Arriba
9.
J Biomed Opt ; 11(5): 050507, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17092148

RESUMEN

We develop an optical whole-body imaging technique for monitoring normal and leukemic hematopoietic cell homing in vivo. A recently developed near-infrared (NIR) lipophilic carbocyanine dye 1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide (DiR) is used to safely and directly label the membranes of human leukemic Pre-B ALL G2 cell lines as well as primary murine lymphocytes and erythrocytes. DiR has absorption and fluorescence maxima at 750 and 782 nm, respectively, which corresponds to low light absorption and autofluorescence in living tissues. This allows us to obtain a significant signal with very low background level. A charge-coupled device (CCD)-based imager is used for noninvasive whole-body imaging of DiR-labeled cell homing in intact animals. This powerful technique can potentially visualize any cell type without use of specific antibodies conjugated with NIR fluorescent tag or loading cells with transporter-delivered NIR fluorophores. Thus, in vivo imaging based on NIR lipophilic carbocyanine dyes in combination with advanced optical techniques may serve as a powerful alternative or complementation to other small animal imaging methods.


Asunto(s)
Carbocianinas , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Leucemia/patología , Lípidos de la Membrana/análisis , Espectrofotometría Infrarroja/métodos , Imagen de Cuerpo Entero/métodos , Animales , Línea Celular , Ratones , Técnicas de Sonda Molecular
10.
J Leukoc Biol ; 75(6): 1139-46, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15020655

RESUMEN

We previously reported that disaccharides (DS), generated by enzymatic degradation of heparin or heparan sulfate, inhibit T cell-mediated immune reactions in rodents and regulate cytokine [tumor necrosis factor alpha (TNF-alpha), interleukin (IL)-8, and IL-1beta] secretion by T cells, macrophages, or intestinal epithelial cells. Here, we investigated the effects of a trisulfated heparin DS (3S-DS) on two aspects of T cell function: secretion of proinflammatory cytokines and migration to an inflamed site. 3S-DS down-regulated nuclear factor-kappaB activity and reduced the secretion of TNF-alpha and interferon-gamma (IFN-gamma) by anti-CD3-activated T cells. In addition, 3S-DS inhibited CXC chemokine ligand 12 (CXCL12; stromal cell-derived factor-1alpha)-dependent migration in vitro and in vivo and decreased CXCL12-induced T cell adhesion to the extracellular matrix glycoprotein, fibronectin (FN). This inhibition was accompanied by attenuation of CXCL12-induced Pyk2 phosphorylation but did not involve internalization of the CXCL12 receptor, CXCR4, or phosphorylation of extracellular-regulated kinase. Despite inhibiting CXCL12-induced adhesion, 3S-DS, on its own, induced T cell adhesion to FN, which was accompanied by phosphorylation of Pyk2. A monosulfated DS showed no effect. Taken together, these data provide evidence that 3S-DS can regulate inflammation by inducing and modulating T cell-signaling events, desensitizing CXCR4, and modulating T cell receptor-induced responses.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Disacáridos/farmacología , Heparina/análogos & derivados , Heparina/farmacología , FN-kappa B/metabolismo , Transducción de Señal , Linfocitos T/efectos de los fármacos , Animales , Anticuerpos Monoclonales/farmacología , Médula Ósea/metabolismo , Adhesión Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiotaxis , Fibronectinas/metabolismo , Quinasa 2 de Adhesión Focal , Heparitina Sulfato/farmacología , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Fosforilación , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR4/metabolismo , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Stem Cell Res ; 12(1): 49-59, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24145187

RESUMEN

The transcription factor p63, member of the p53 gene family, encodes for two main isoforms, TAp63 and ΔNp63 with distinct functions on epithelial homeostasis and cancer. Recently, we discovered that TAp63 is essential for in vitro cardiogenesis and heart development in vivo. TAp63 is expressed by embryonic endoderm and acts on cardiac progenitors by a cell-non-autonomous manner. In the present study, we search for cardiogenic secreted factors that could be regulated by TAp63 and, by ChIP-seq analysis, identified Angiomodulin (AGM), also named IGFBP7 or IGFBP-rP1. We demonstrate that AGM is necessary for cardiac commitment of embryonic stem cells (ESCs) and its regulation depends on TAp63 isoform. TAp63 directly activates both AGM and Activin-A during ESC cardiogenesis while these secreted factors modulate TAp63 gene expression by a feedback loop mechanism. The molecular circuitry controlled by TAp63 on AGM/Activin-A signaling pathway and thus on cardiogenesis emphasizes the importance of p63 during early cardiac development.


Asunto(s)
Activinas/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Retroalimentación Fisiológica , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Transactivadores/metabolismo , Activinas/antagonistas & inhibidores , Activinas/genética , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/metabolismo , Retroalimentación Fisiológica/efectos de los fármacos , Células HeLa , Humanos , Subunidades beta de Inhibinas/genética , Subunidades beta de Inhibinas/metabolismo , Ratones , Miocardio/citología , Proteínas de Neoplasias/genética , Fosfoproteínas/antagonistas & inhibidores , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Transducción de Señal , Transactivadores/antagonistas & inhibidores
12.
Exp Hematol ; 39(12): 1161-1170.e1, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21911094

RESUMEN

Bone marrow homing and engraftment by clinically transplanted hematopoietic stem and progenitor cells is a complex process that is not fully understood. We report that the pan-leukocyte CD45 phosphatase plays an essential role in trafficking and repopulation of the bone marrow by immature human CD34(+) cells and leukemic cells in transplanted nonobese diabetic severe combined immunodeficient mice. Inhibiting CD45 function by blocking antibodies or a CD45 inhibitor impaired the motility of both normal and leukemic human cells. Blocking CD45 inhibited homing and repopulation by immature human CD34(+) cells as well as homing of primary patient leukemic cells. In addition, CD45 inhibition negatively affected development of hematopoietic progenitors in vitro and their recovery in transplanted recipients in vivo, revealing the central role of CD45 in the regulation of hematopoiesis. Moreover, CD45 blockage induced a hyperadhesive phenotype in immature human progenitor cells as well as in murine leukocytes, leading to their defective adhesion interactions with endothelial cells. This phenotype was further manifested by the ability of CD45 blockage to prevent breakdown of adhesion interactions in the BM, which inhibited murine progenitor mobilization. The substantial effects of a direct CD45 inhibition point at its essential roles in cell trafficking, including murine progenitor cell mobilization and both normal immature and leukemic human hematopoietic cells as well as regulation of hematopoiesis and engraftment potential.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Trasplante de Células Madre de Sangre del Cordón Umbilical , Leucemia Promielocítica Aguda/patología , Antígenos Comunes de Leucocito/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Animales , Anticuerpos Monoclonales/farmacología , Adhesión Celular , Ensayo de Unidades Formadoras de Colonias , Supervivencia de Injerto , Factor Estimulante de Colonias de Granulocitos/farmacología , Células HL-60/trasplante , Movilización de Célula Madre Hematopoyética , Humanos , Antígenos Comunes de Leucocito/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Quimera por Radiación , Células Tumorales Cultivadas/trasplante
13.
J Clin Invest ; 119(3): 492-503, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19197139

RESUMEN

The mechanisms governing hematopoietic progenitor cell mobilization are not fully understood. We report higher membrane type 1-MMP (MT1-MMP) and lower expression of the MT1-MMP inhibitor, reversion-inducing cysteine-rich protein with Kazal motifs (RECK), on isolated circulating human CD34+ progenitor cells compared with immature BM cells. The expression of MT1-MMP correlated with clinical mobilization of CD34+ cells in healthy donors and patients with lymphoid malignancies. Treatment with G-CSF further increased MT1-MMP and decreased RECK expression in human and murine hematopoietic cells in a PI3K/Akt-dependent manner, resulting in elevated MT1-MMP activity. Blocking MT1-MMP function by Abs or siRNAs impaired chemotaxis and homing of G-CSF-mobilized human CD34+ progenitors. The mobilization of immature and maturing human progenitors in chimeric NOD/SCID mice by G-CSF was inhibited by anti-MT1-MMP treatment, while RECK neutralization promoted motility and egress of BM CD34+ cells. BM c-kit+ cells from MT1-MMP-deficient mice also exhibited inferior chemotaxis, reduced homing and engraftment capacities, and impaired G-CSF-induced mobilization in murine chimeras. Membranal CD44 cleavage by MT1-MMP was enhanced following G-CSF treatment, reducing CD34+ cell adhesion. Accordingly, CD44-deficient mice had a higher frequency of circulating progenitors. Our results reveal that the motility, adhesion, homing, and mobilization of human hematopoietic progenitor cells are regulated in a cell-autonomous manner by dynamic and opposite changes in MT1-MMP and RECK expression.


Asunto(s)
Antígenos CD34/análisis , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/fisiología , Metaloproteinasa 14 de la Matriz/genética , Glicoproteínas de Membrana/genética , Animales , Antígenos CD/análisis , Células de la Médula Ósea/fisiología , Movimiento Celular/fisiología , Quimiotaxis , Quimera/genética , Proteínas Ligadas a GPI , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Humanos , Metaloproteinasa 14 de la Matriz/deficiencia , Metaloproteinasa 14 de la Matriz/efectos de los fármacos , Metaloproteinasa 14 de la Matriz/metabolismo , Glicoproteínas de Membrana/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , ARN Mensajero/genética , ARN Interferente Pequeño/genética
14.
Cell Stem Cell ; 3(5): 484-92, 2008 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-18983964

RESUMEN

Physiological interactions between the nervous and immune systems with components of the local microenvironment are needed to maintain homeostasis throughout the body. Dynamic regulation of bone remodeling, hematopoietic stem cells, and their evolving niches via neurotransmitter signaling are part of the host defense and repair mechanisms. This crosstalk links activated leukocytes, neuronal, and stromal cells, which combine to directly and indirectly regulate hematopoietic stem cells. Together, interactions between diverse systems create a regulatory "brain-bone-blood triad," contributing an additional dimension to the concept of the hematopoietic stem cell niche.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Homeostasis/fisiología , Neuroinmunomodulación/fisiología , Comunicación Paracrina/fisiología , Nicho de Células Madre/fisiología , Animales , Diferenciación Celular , Movimiento Celular , Ritmo Circadiano/fisiología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Humanos , Estrés Fisiológico
15.
J Exp Med ; 205(10): 2381-95, 2008 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-18779349

RESUMEN

The CD45 phosphatase is uniquely expressed by all leukocytes, but its role in regulating hematopoietic progenitors is poorly understood. We show that enhanced CD45 expression on bone marrow (BM) leukocytes correlates with increased cell motility in response to stress signals. Moreover, immature CD45 knockout (KO) cells showed defective motility, including reduced homing (both steady state and in response to stromal-derived factor 1) and reduced granulocyte colony-stimulating factor mobilization. These defects were associated with increased cell adhesion mediated by reduced matrix metalloproteinase 9 secretion and imbalanced Src kinase activity. Poor mobilization of CD45KO progenitors by the receptor activator of nuclear factor kappaB ligand, and impaired modulation of the endosteal components osteopontin and stem cell factor, suggested defective osteoclast function. Indeed, CD45KO osteoclasts exhibited impaired bone remodeling and abnormal morphology, which we attributed to defective cell fusion and Src function. This led to irregular distribution of metaphyseal bone trabecules, a region enriched with stem cell niches. Consequently, CD45KO mice had less primitive cells in the BM and increased numbers of these cells in the spleen, yet with reduced homing and repopulation potential. Uncoupling environmental and intrinsic defects in chimeric mice, we demonstrated that CD45 regulates progenitor movement and retention by influencing both the hematopoietic and nonhematopoietic compartments.


Asunto(s)
Remodelación Ósea , Huesos , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/fisiología , Antígenos Comunes de Leucocito/metabolismo , Osteoclastos/fisiología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Huesos/anatomía & histología , Huesos/fisiología , Adhesión Celular/fisiología , Quimiocina CXCL12/metabolismo , Activación Enzimática , Factor Estimulante de Colonias de Granulocitos/metabolismo , Células Madre Hematopoyéticas/citología , Antígenos Comunes de Leucocito/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Osteoclastos/citología , Ligando RANK/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo
16.
Nat Immunol ; 8(10): 1123-31, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17828268

RESUMEN

Catecholamines are important regulators of homeostasis, yet their functions in hematopoiesis are poorly understood. Here we report that immature human CD34+ cells dynamically expressed dopamine and beta2-adrenergic receptors, with higher expression in the primitive CD34+CD38(lo) population. The myeloid cytokines G-CSF and GM-CSF upregulated neuronal receptor expression on immature CD34+ cells. Treatment with neurotransmitters increased the motility, proliferation and colony formation of human progenitor cells, correlating with increased polarity, expression of the metalloproteinase MT1-MMP and activity of the metalloproteinase MMP-2. Treatment with catecholamines enhanced human CD34+ cell engraftment of NOD-SCID mice through Wnt signaling activation and increased cell mobilization and bone marrow Sca-1+c-Kit+Lin- cell numbers. Our results identify new functions for neurotransmitters and myeloid cytokines in the direct regulation of human and mouse progenitor cell migration and development.


Asunto(s)
Antígenos CD34/análisis , Catecolaminas/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Neurotransmisores/farmacología , Transducción de Señal/fisiología , Proteínas Wnt/fisiología , Animales , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dopamina/farmacología , Agonistas de Dopamina/farmacología , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Ratones Endogámicos C57BL , Norepinefrina/farmacología , Receptores Dopaminérgicos/análisis
17.
Stem Cells ; 23(4): 561-74, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15790777

RESUMEN

The mechanism of human stem cell expansion ex vivo is not fully understood. Furthermore, little is known about the mechanisms of human stem cell homing/repopulation and the role that differentiating progenitor cells may play in these processes. We report that 2- to 3-day in vitro cytokine stimulation of human cord blood CD34(+)-enriched cells induces the production of short-term repopulating, cycling G1 CD34(+)/CD38(+) cells with increased matrix metalloproteinase (MMP)-9 secretion as well as increased migration capacity to the chemokine stromal cell-derived factor-1 (SDF-1) and homing to the bone marrow of irradiated nonobese diabetic severe/combined immunodeficiency (NOD/SCID) mice. These cycling G1 cells enhance SDF-1-mediated in vitro migration and in vivo homing of quiescent G0 CD34(+) cells, which is partially abrogated after inhibition of MMP-2/-9 activity. Moreover, the engraftment potential of quiescent G0 SCID repopulating cells (SRCs) is also increased by the cycling G1 CD34(+)/CD38(+) cells. This effect is significantly abrogated after incubation of cycling G1 cells with a neutralizing anti-CXCR4 antibody. Our data suggest synergistic interactions between accessory cycling G1 CD34(+)/CD38(+) committed progenitor cells and quiescent, primitive G0 CD34(+)/CD38(-/low) SRC/stem cells, the former increasing the motility and engraftment potential of the latter, partly via secretion of MMP-9.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Antígenos CD34/metabolismo , Fase G1 , Fase de Descanso del Ciclo Celular , Células Madre/citología , Animales , Células de la Médula Ósea/fisiología , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiotaxis , Ensayo de Unidades Formadoras de Colonias , Citocinas/metabolismo , Sangre Fetal/citología , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores CXCR4/metabolismo , Trasplante de Células Madre , Células Madre/fisiología
18.
J Immunol ; 175(1): 276-85, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15972659

RESUMEN

Previously, we reported that treatment of T cells with the 60-kDa heat shock protein (HSP60) inhibits chemotaxis. We now report that treatment of purified human T cells with recombinant human HSP60 or its biologically active peptide p277 up-regulates suppressor of cytokine signaling (SOCS)3 expression via TLR2 and STAT3 activation. SOCS3, in turn, inhibits the downstream effects of stromal cell-derived-1alpha (CXCL12)-CXCR4 interaction in: 1) phosphorylation of ERK1/2, Pyk2, AKT, and myosin L chain, required for cell adhesion and migration; 2) formation of rear-front T cell polarity; and 3) migration into the bone marrow of NOD/SCID mice. HSP60 also activates SOCS3 in mouse lymphocytes and inhibits their chemotaxis toward stromal cell-derived factor-1alpha and their ability to adoptively transfer delayed-type hypersensitivity. These effects of HSP60 could not be attributed to LPS or LPS-associated lipoprotein contamination. Thus, HSP60 can regulate T cell-mediated inflammation via specific signal transduction and SOCS3 activation.


Asunto(s)
Chaperonina 60/farmacología , Proteínas Represoras/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factores de Transcripción/metabolismo , Traslado Adoptivo , Animales , Polaridad Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Quimiotaxis de Leucocito/efectos de los fármacos , Citocinas/metabolismo , Femenino , Quinasa 2 de Adhesión Focal , Silenciador del Gen , Humanos , Técnicas In Vitro , Inflamación/etiología , Inflamación/inmunología , Activación de Linfocitos/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Inmunológicos , Cadenas Ligeras de Miosina/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Receptores de Superficie Celular/metabolismo , Receptores Inmunológicos/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Transducción de Señal/efectos de los fármacos , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Linfocitos T/fisiología , Receptor Toll-Like 2 , Receptores Toll-Like , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
19.
Eur J Immunol ; 32(8): 2264-73, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12209639

RESUMEN

B cell receptor signaling threshold regulates negative selection of autoreactive B cells and determines the mechanism of B cell tolerance. Using mice carrying immunoglobulin transgene specific for MHC class I antigen K(k) (3-83 Tg mice), and IL-7-driven bone marrow (BM) culture system, we have previously shown that receptor editing is a major mechanism in B cell tolerance. To test the role of BCR signaling competence on the induction of tolerance-mediated receptor editing, we crossed the 3-83 Tg mice with mice deficient in CD45, a protein tyrosine phosphatase that functions asa positive regulator of the BCR signaling. We found that in the absence of self-antigen allelic exclusion is efficiently imposed in 3-83 Tg CD45(-/-) mice, although numbers of peripheral B cells are reduced. Using our BM culture system, we show here that immature 3-83 Tg CD45(-/-) B cells encountering self-antigen are developmentally arrested and undergo secondary light chain recombination and receptor editing, not different than CD45-sufficient cells. Thus, lack of CD45 does not abolish the receptor editing competence in immature B cells encountering high avidity membrane-bound antigen.


Asunto(s)
Linfocitos B/fisiología , Células Madre Hematopoyéticas/fisiología , Antígenos Comunes de Leucocito/fisiología , Receptores de Antígenos de Linfocitos B/fisiología , Alelos , Animales , Apoptosis , Autoantígenos/fisiología , Células Cultivadas , Ratones , Ratones Transgénicos
20.
J Immunol ; 168(11): 5596-604, 2002 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12023356

RESUMEN

Positive signaling is now thought to be important for B cell maturation, although the nature of such signals has not yet been defined. We are studying the regulatory role of B cell Ag receptor (BCR) signaling in mediating positive selection of immature B cells. To do so, we use Ig transgenic mice (3-83Tg) that are deficient in CD19, thus generating a monoclonal immature B cell population expressing signaling-incompetent BCR. Immature 3-83Tg CD19(-/-) B cells undergo developmental arrest in the bone marrow, allowing maturation only to cells that effectively compensate for the compromised receptor by elevated levels of BCR. We find that developmentally arrested 3-83Tg CD19(-/-) B cells fail to impose L chain allelic exclusion and undergo intensive V(D)J recombination to edit their BCR. Furthermore, immature 3-83Tg CD19(-/-) B cells, which were grown in vitro, failed to undergo positive selection and to survive when adoptively transferred into normal recipients. However, elevation of BCR expression levels, obtained by transgene homozygosity, effectively compensated for the compromised BCR and completely restored BCR-mediated Ca(2+) influx, allelic exclusion, and positive selection. Our results suggest that the BCR signaling threshold mediates positive selection of developing B cells, and that a receptor-editing mechanism has an important role in rescuing cells that fail positive selection because of incompetent receptors.


Asunto(s)
Alelos , Antígenos CD19/fisiología , Linfocitos B/fisiología , Cadenas Ligeras de Inmunoglobulina/genética , Animales , Calcio/metabolismo , Células Cultivadas , Reordenamiento Génico de Linfocito T , Ratones , Receptores de Antígenos de Linfocitos B/análisis , Receptores de Antígenos de Linfocitos B/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA