Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Rev Rep ; 16(4): 684-692, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32418119

RESUMEN

Aging impairs the regenerative potential of hematopoietic stem cells (HSC) and skews differentiation towards the myeloid lineage. The bone marrow (BM) microenvironment has recently been suggested to influence HSC aging, however the mechanisms whereby BM stromal cells mediate this effect is unknown. Here we show that aging-associated decreased expression of CXCR4 expression on BM mesenchymal stem cells (MSC) plays a crucial role in the development of the hematopoietic stem and progenitor cells (HSPC) aging phenotype. The BM MSC from old mice was sufficient to drive a premature aging phenotype of young HSPC when cultured together ex vivo. The impaired ability of old MSC to support HSPC function is associated with reduced expression of CXCR4 on BM MSC of old mice. Deletion of the CXCR4 gene in young MSC accelerates an aging phenotype in these cells characterized by increased production of reactive oxygen species (ROS), DNA damage, senescence, and reduced proliferation. Culture of HSPC from young mice with CXCR4 deficient MSC also from young mice led to a premature aging phenotype in the young HSPC, as evidenced by reduced hematopoietic regeneration and enhanced myeloid differentiation. Mechanistically, CXCR4 signaling prevents BM MSC dysfunction by suppressing oxidative stress, as treatment of old or CXCR4 deficient MSC with N-acetyl-L-cysteine (NAC), improved their niche supporting activity, and attenuated the HSPC aging phenotype. Our studies suggest that age-associated reduction in CXCR4 expression on BM MSC impairs hematopoietic niche activity with increased ROS production, driving an HSC aging phenotype. Thus, modulation of the SDF-1/CXCR4 axis in MSC may lead to novel interventions to alleviate the age-associated decline in immune/hematopoietic function.


Asunto(s)
Envejecimiento/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores CXCR4/metabolismo , Acetilcisteína/farmacología , Animales , Recuento de Células , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Células Clonales , Ensayo de Unidades Formadoras de Colonias , Depuradores de Radicales Libres/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones Endogámicos C57BL , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Receptores CXCR4/deficiencia
2.
Radiat Res ; 192(4): 410-421, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31390312

RESUMEN

Radiation-induced acute skin injury and consequent fibrosis are common complications of cancer radiotherapy and radiation accidents. Stromal cell-derived factor-1α (SDF-1α) and its receptor, CXC chemokine receptor 4 (CXCR4) have been shown to be involved in multiple cellular events. However, the role of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin has not been reported. In this study, we found that the expression of SDF-1α and CXCR4 was significantly increased in irradiated skin tissues of humans, monkeys and rats, compared to their nonirradiated counterparts. Mice with keratinocyte-specific ablation of CXCR4 showed less severe skin damage than wild-type mice after receiving a 35 Gy dose of radiation. Consistently, subcutaneous injection of AMD3100, an FDA approved SDF-1α/CXCR4 inhibitor, attenuated skin injury and fibrosis induced by exposure to radiation in a rat model. Mechanically, the SDF-1α/CXCR4 axis promotes pro-fibrotic TGF-b/Smad signaling through the PI3K-MAPK signaling cascade in human keratinocyte HaCaT cells and skin fibroblast WS1 cells. AMD3100 inhibited Smad2 nuclear translocation and transcriptional activity of Smad2/3 induced by radiation, which suppressed the pro-fibrotic TGF-b/Smad signaling pathway activated by exposure. Taken together, these findings demonstrate the involvement of SDF-1α/CXCR4 axis in radiation-induced acute injury and fibrosis of skin, and indicate that AMD3100 would be an effective countermeasure against these diseases.


Asunto(s)
Quimiocina CXCL12/metabolismo , Traumatismos por Radiación/metabolismo , Receptores CXCR4/metabolismo , Piel/patología , Piel/efectos de la radiación , Animales , Bencilaminas , Ciclamas , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Técnicas de Inactivación de Genes , Compuestos Heterocíclicos/farmacología , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Traumatismos por Radiación/patología , Ratas , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Piel/lesiones , Piel/metabolismo , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
3.
J Cell Physiol ; 234(8): 13894-13905, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30656691

RESUMEN

Vulvovaginal candidiasis (VVC) is a common observed infection, affecting approximately 75% of women of reproductive age. Drug resistance represents a troublesome stumbling block associated with VVC therapy. Thus the aim of the present study was to provide information regarding the selection of potential drug targets for VVC. CXCR3-, CXCR4-, or CXCR/CXCR4 double-deficient mouse models of VVC were subsequently established, with changes to the load of Candida Albicans evaluated accordingly. The biological behaviors of the vaginal epithelial cells were characterized in response to the CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout in vivo. Our initial observations revealed that in mice with VVC, CXCR3-, CXCR4-, or CXCR3 - CXCR4 double-knockout resulted in a decreased load of C. Albicans as well as reduced levels and proportion of Th17 cells. Proinflammatory cytokine production was found to be inhibited by CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout whereby the mRNA and protein expressions CXCR3, CXCR4, IL-17, IL-6, and TNF-α exhibited decreased levels. CXCR3-, CXCR4-, or CXCR3/CXCR4 double-knockout appeared to function as positive proliferation factors, while playing a negative role in the processes of apoptosis and the cell cycle of vaginal epithelial cells. Taken together, the key findings of the study suggested that CXCR3/CXCR4 double-knockout could act to hinder the progression of VVC, highlighting its promise as a novel therapeutic target in the treatment of VVC. CXCR3 and CXCR4 genes may regulate Th17/IL-17 immune inflammatory pathways to participate in antifungal immunity.


Asunto(s)
Candidiasis Vulvovaginal/inmunología , Candidiasis Vulvovaginal/metabolismo , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Receptores CXCR3/deficiencia , Receptores CXCR4/deficiencia , Células Th17/patología , Animales , Apoptosis , Candida albicans/fisiología , Candidiasis Vulvovaginal/sangre , Candidiasis Vulvovaginal/microbiología , Ciclo Celular , Proliferación Celular , Citocinas/sangre , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células Epiteliales/patología , Femenino , Ratones Noqueados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR3/sangre , Receptores CXCR3/metabolismo , Receptores CXCR4/sangre , Receptores CXCR4/metabolismo , Vagina/microbiología , Vagina/patología
4.
PLoS One ; 13(11): e0207251, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30408103

RESUMEN

The CXCL12-CXCR4 pathway has crucial roles in stem cell homing and maintenance, neuronal guidance, cancer progression, inflammation, remote-conditioning, cell migration and development. Recently, work in chick suggested that signalling via CXCR4 in neural crest cells (NCCs) has a role in the 22q11.2 deletion syndrome (22q11.2DS), a disorder where haploinsufficiency of the transcription factor TBX1 is responsible for the major structural defects. We tested this idea in mouse models. Our analysis of genes with altered expression in Tbx1 mutant mouse models showed down-regulation of Cxcl12 in pharyngeal surface ectoderm and rostral mesoderm, both tissues with the potential to signal to migrating NCCs. Conditional mutagenesis of Tbx1 in the pharyngeal surface ectoderm is associated with hypo/aplasia of the 4th pharyngeal arch artery (PAA) and interruption of the aortic arch type B (IAA-B), the cardiovascular defect most typical of 22q11.2DS. We therefore analysed constitutive mouse mutants of the ligand (CXCL12) and receptor (CXCR4) components of the pathway, in addition to ectodermal conditionals of Cxcl12 and NCC conditionals of Cxcr4. However, none of these typical 22q11.2DS features were detected in constitutively or conditionally mutant embryos. Instead, duplicated carotid arteries were observed, a phenotype recapitulated in Tie-2Cre (endothelial) conditional knock outs of Cxcr4. Previous studies have demonstrated genetic interaction between signalling pathways and Tbx1 haploinsufficiency e.g. FGF, WNT, SMAD-dependent. We therefore tested for possible epistasis between Tbx1 and the CXCL12 signalling axis by examining Tbx1 and Cxcl12 double heterozygotes as well as Tbx1/Cxcl12/Cxcr4 triple heterozygotes, but failed to identify any exacerbation of the Tbx1 haploinsufficient arch artery phenotype. We conclude that CXCL12 signalling via NCC/CXCR4 has no major role in the genesis of the Tbx1 loss of function phenotype. Instead, the pathway has a distinct effect on remodelling of head vessels and interventricular septation mediated via CXCL12 signalling from the pharyngeal surface ectoderm and second heart field to endothelial cells.


Asunto(s)
Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Quimiocina CXCL12/deficiencia , Receptores CXCR4/deficiencia , Proteínas de Dominio T Box/deficiencia , Animales , Aorta Torácica/anomalías , Aorta Torácica/embriología , Aorta Torácica/metabolismo , Anomalías Cardiovasculares/embriología , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/metabolismo , Sistema Cardiovascular/embriología , Quimiocina CXCL12/genética , Síndrome de DiGeorge/enzimología , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/metabolismo , Modelos Animales de Enfermedad , Epistasis Genética , Femenino , Haploinsuficiencia , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Cresta Neural/metabolismo , Embarazo , Receptores CXCR4/genética , Transducción de Señal/genética , Proteínas de Dominio T Box/genética
5.
Cardiovasc Res ; 113(13): 1677-1687, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29016745

RESUMEN

AIMS: Chemokine CXCL12 (stromal derived factor 1: SDF1) has been shown to play important roles in various processes of cardiovascular development. In recent avian studies, CXCL12 signalling has been implicated in guidance of cardiac neural crest cells for their participation in the development of outflow tract and cardiac septum. The goal of this study is to investigate the extent to which CXCL12 signalling contribute to the development of aortic arch and pulmonary arteries in mammals. METHODS AND RESULTS: Novel Cxcl12-LacZ reporter and conditional alleles were generated. Using whole mount X-gal staining with the reporter allele and vascular casting techniques, we show that the domain branching pattern of pulmonary arteries in Cxcl12-null mice is completely disrupted and discordant with that of pulmonary veins and airways. Cxcl12-null mice also displayed abnormal and superfluous arterial branches from the aortic arch. The early steps of pharyngeal arch remodelling in Cxcl12-null mice appeared to be unaffected, but vertebral arteries were often missing and prominent aberrant arteries were present parallel to carotid arteries or trachea, similar to aberrant vertebral artery or thyroid ima artery, respectively. Analysis with computed tomography not only confirmed the results from vascular casting studies but also identified abnormal systemic arterial supply to lungs in the Cxcl12-null mice. Tie2-Cre mediated Cxcr4 deletion phenocopied the Cxcl12-null phenotypes, indicating that CXCR4 is the primary receptor for arterial patterning, whereas Cxcl12 or Cxcr4 deletion by Wnt1-Cre did not affect aortic arch patterning. CONCLUSION: CXCL12-CXCR4 signalling is essential for the correct patterning of aortic arches and pulmonary arteries during development. Superfluous arteries in Cxcl12-null lungs and the aortic arch infer a role of CXCL12 in protecting arteries from uncontrolled sprouting during development of the arterial system.


Asunto(s)
Aorta Torácica/metabolismo , Tipificación del Cuerpo , Quimiocina CXCL12/metabolismo , Arteria Pulmonar/metabolismo , Receptores CXCR4/metabolismo , Malformaciones Vasculares/metabolismo , Animales , Aorta Torácica/anomalías , Aorta Torácica/diagnóstico por imagen , Aortografía/métodos , Quimiocina CXCL12/deficiencia , Quimiocina CXCL12/genética , Angiografía por Tomografía Computarizada , Regulación del Desarrollo de la Expresión Génica , Genotipo , Edad Gestacional , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica , Fenotipo , Arteria Pulmonar/anomalías , Arteria Pulmonar/diagnóstico por imagen , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Transducción de Señal , Malformaciones Vasculares/diagnóstico por imagen , Malformaciones Vasculares/genética
6.
J Leukoc Biol ; 101(5): 1143-1153, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28148720

RESUMEN

Precursors of dendritic cells (pre-DCs) arise in the bone marrow (BM), egress to the blood, and finally migrate to peripheral tissue, where they differentiate to conventional dendritic cells (cDCs). Upon their activation, antigen-bearing cDCs migrate from peripheral tissue to regional lymph nodes (LNs) in a manner dependent on the chemokine receptor, CCR7. To maintain immune homeostasis, these departing cDCs must be replenished by new cDCs that develop from pre-DCs, but the molecular signals that direct pre-DC trafficking from the BM to the blood and peripheral tissues remain poorly understood. In the present study, we found that pre-DCs express the chemokine receptors CXCR4, CCR2, and CX3CR1, and that each of these receptors has a distinct role in pre-DC trafficking. Flow cytometric analysis of pre-DCs lacking CXCR4 revealed that this receptor is required for the retention of pre-DCs in the BM. Analyses of mice lacking CCR2 or CX3CR1, or both, revealed that they promote pre-DC migration to the lung at steady state. CCR2, but not CX3CR1, was required for pre-DC migration to the inflamed lung. Thus, these multiple chemokine receptors cooperate in a step-wise fashion to coordinate the trafficking of pre-DCs from the BM to the circulation and peripheral tissues.


Asunto(s)
Células de la Médula Ósea/inmunología , Células Dendríticas/inmunología , Pulmón/inmunología , Neumonía/inmunología , Receptores CCR2/inmunología , Receptores CXCR4/inmunología , Receptores de Quimiocina/inmunología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Receptor 1 de Quimiocinas CX3C , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Regulación de la Expresión Génica , Lipopolisacáridos , Pulmón/efectos de los fármacos , Pulmón/patología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Receptores CCR2/deficiencia , Receptores CCR2/genética , Receptores CCR7/genética , Receptores CCR7/inmunología , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Receptores de Quimiocina/deficiencia , Receptores de Quimiocina/genética , Transducción de Señal
7.
Stem Cells ; 34(11): 2744-2757, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27340942

RESUMEN

CXCR4 is a stem/progenitor cell surface receptor specific for the cytokine stromal cell-derived factor-1 (SDF-1α). There is evidence that bone marrow-derived CXCR4-expressing cells contribute to intimal hyperplasia (IH) by homing to the arterial subintima which is enriched with SDF-1α. We have previously found that transforming growth factor-ß (TGFß) and its signaling protein Smad3 are both upregulated following arterial injury and that TGFß/Smad3 enhances the expression of CXCR4 in vascular smooth muscle cells (SMCs). It remains unknown, however, whether locally induced CXCR4 expression in SM22 expressing vascular SMCs plays a role in neointima formation. Here, we investigated whether elevated TGFß/Smad3 signaling leads to the induction of CXCR4 expression locally in the injured arterial wall, thereby contributing to IH. We found prominent CXCR4 upregulation (mRNA, 60-fold; protein, 4-fold) in TGFß-treated, Smad3-expressing SMCs. Chromatin immunoprecipitation assays revealed a specific association of the transcription factor Smad3 with the CXCR4 promoter. TGFß/Smad3 treatment also markedly enhanced SDF-1α-induced ERK1/2 phosphorylation as well as SMC migration in a CXCR4-dependent manner. Adenoviral expression of Smad3 in balloon-injured rat carotid arteries increased local CXCR4 levels and enhanced IH, whereas SMC-specific depletion of CXCR4 in the wire-injured mouse femoral arterial wall produced a 60% reduction in IH. Our results provide the first evidence that upregulation of TGFß/Smad3 in injured arteries induces local SMC CXCR4 expression and cell migration, and consequently IH. The Smad3/CXCR4 pathway may provide a potential target for therapeutic interventions to prevent restenosis. Stem Cells 2016;34:2744-2757.


Asunto(s)
Traumatismos de las Arterias Carótidas/genética , Neointima/genética , Receptores CXCR4/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/metabolismo , Túnica Íntima/metabolismo , Animales , Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/metabolismo , Traumatismos de las Arterias Carótidas/patología , Movimiento Celular , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Neointima/patología , Fosforilación , Cultivo Primario de Células , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/deficiencia , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Túnica Íntima/lesiones
8.
Am J Physiol Heart Circ Physiol ; 310(1): H20-8, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26519029

RESUMEN

The stromal cell-derived factor-1 (SDF-1):CXCR4 is important in myocardial repair. In this study we tested the hypothesis that early upregulation of cardiomyocyte CXCR4 (CM-CXCR4) at a time of high myocardial SDF-1 expression could be a strategy to engage the SDF-1:CXCR4 axis and improve cardiac repair. The effects of the hypoxia inducible factor (HIF) hydroxylase inhibitor dimethyloxalylglycine (DMOG) on CXCR4 expression was tested on H9c2 cells. In mice a myocardial infarction (MI) was produced in CM-CXCR4 null and wild-type controls. Mice were randomized to receive injection of DMOG (DMOG group) or saline (Saline group) into the border zone after MI. Protein and mRNA expression of CM-CXCR4 were quantified. Echocardiography was used to assess cardiac function. During hypoxia, DMOG treatment increased CXCR4 expression of H9c2 cells by 29 and 42% at 15 and 24 h, respectively. In vivo DMOG treatment increased CM-CXCR4 expression at 15 h post-MI in control mice but not in CM-CXCR4 null mice. DMOG resulted in increased ejection fraction in control mice but not in CM-CXCR4 null mice 21 days after MI. Consistent with greater cardiomyocyte survival with DMOG treatment, we observed a significant increase in cardiac myosin-positive area within the infarct zone after DMOG treatment in control mice, but no increase in CM-CXCR4 null mice. Inhibition of cardiomyocyte death in MI through the stabilization of HIF-1α requires downstream CM-CXCR4 expression. These data suggest that engagement of the SDF-1:CXCR4 axis through the early upregulation of CM-CXCR4 is a strategy for improving cardiac repair after MI.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Cardiotónicos/farmacología , Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , Receptores CXCR4/metabolismo , Función Ventricular Izquierda/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Hipoxia de la Célula , Línea Celular , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Prolina Dioxigenasas del Factor Inducible por Hipoxia/antagonistas & inhibidores , Prolina Dioxigenasas del Factor Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , Ratas , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Recuperación de la Función , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Volumen Sistólico/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba
9.
Cell Tissue Res ; 362(3): 633-42, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26246398

RESUMEN

Dental stem cells are located at the proximal ends of rodent incisors. These stem cells reside in the dental epithelial stem cell niche, termed the apical bud. We focused on identifying critical features of a chemotactic signal in the niche. Here, we report that CXCR4/CXCL12 signaling impacts enamel progenitor cell proliferation and motility in dental stem cell niche cells. We report cells in the apical bud express CXCR4 mRNA at high levels while expression is restricted in the basal epithelium (BE) and transit-amplifying (TA) cell regions. Furthermore, the CXCL12 ligand is present in mesenchymal cells adjacent to the apical bud. We then performed gain- and loss-of-function analyses to better elucidate the role of CXCR4 and CXCL12. CXCR4-deficient mice contain epithelial cell aggregates, while cell proliferation in mutant incisors was also significantly reduced. We demonstrate in vitro that dental epithelial cells migrate toward sources of CXCL12, whereas knocking down CXCR4 impaired motility and resulted in formation of dense cell colonies. These results suggest that CXCR4 expression may be critical for activation of enamel progenitor cell division and that CXCR4/CXCL12 signaling may control movement of epithelial progenitors from the dental stem cell niche.


Asunto(s)
Movimiento Celular , Quimiocina CXCL12/metabolismo , Esmalte Dental/citología , Receptores CXCR4/metabolismo , Transducción de Señal , Nicho de Células Madre , Células Madre/citología , Animales , Agregación Celular , Línea Celular , Proliferación Celular , Forma de la Célula , Quimiocina CXCL12/deficiencia , Quimiocina CXCL12/genética , Células Epiteliales , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Incisivo/citología , Incisivo/embriología , Ratones Noqueados , Mutación , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Células Madre/metabolismo
10.
J Neurosci ; 35(24): 9211-24, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26085643

RESUMEN

Radial glial cells are the neural progenitors of the developing CNS and have long radial processes that guide radially migrating neurons. The integrity of the radial glial scaffold, in particular proper adhesion between the endfeet of radial processes and the pial basement membrane (BM), is important for the cellular organization of the CNS, as indicated by evidence emerging from the developing cortex. However, the mechanisms underlying the maintenance of radial glial scaffold integrity during development, when the neuroepithelium rapidly expands, are still poorly understood. Here, we addressed this issue in the developing mouse spinal cord. We show that CXCR4, a receptor of chemokine CXCL12, is expressed in spinal cord radial glia. Conditional knock-out of Cxcr4 in radial glia caused disrupted radial glial scaffold with gaps at the pial endfeet layer and consequentially led to an invasion of boundary cap (BC) cells into the spinal cord. Because BC cells are PNS cells normally positioned at the incoming and outgoing axonal roots, their invasion into the spinal cord suggests a compromised CNS/PNS boundary in the absence of CXCL12/CXCR4 signaling. Both disrupted radial glial scaffold and invasion of BC cells into the CNS were also present in mice deficient in CXCR7, a second receptor of CXCL12. We further show that CXCL12 signaling promotes the radial glia adhesion to BM components and activates integrin ß1 avidity. Our study unravels a novel molecular mechanism that deploys CXCL12/CXCR4/CXCR7 for the maintenance of radial glial scaffold integrity, which in turn safeguards the CNS/PNS boundary during spinal cord development.


Asunto(s)
Células Ependimogliales/metabolismo , Organogénesis/fisiología , Receptores CXCR4/deficiencia , Médula Espinal/embriología , Médula Espinal/metabolismo , Animales , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Ratones , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Neuroglía/metabolismo , Transducción de Señal/fisiología , Médula Espinal/citología
11.
J Mol Cell Cardiol ; 81: 49-53, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25655934

RESUMEN

The G protein-coupled receptor CXCR4 and its ligand stromal-cell derived factor 1 (SDF-1) play a crucial role in directing progenitor cell (PC) homing to ischemic tissue. The Src family protein kinases (SFK) can be activated by, and serve as effectors of, G proteins. In this study we sought to determine whether SFK play a role in SDF-1/CXCR4-mediated PC homing. First, we investigated whether SDF-1/CXCR4 signaling activates SFK. Bone-marrow mononuclear cells (BM MNCs) were isolated from WT and BM-specific CXCR4-KO mice and treated with SDF-1 and/or CXCR4 antagonist AMD3100. SDF-1 treatment rapidly induced phosphorylation (activation) of hematopoietic Src (i.e., Lyn, Fgr, and Hck) in WT cells but not in AMD3100-treated cells or CXCR4-KO cells. Then, we investigated whether SFK are involved in SDF-1/CXCR4-mediated PC chemotaxis. In a combined chemotaxis and endothelial-progenitor-cell (EPC) colony assay, Src inhibitor SU6656 dose-dependently inhibited the SDF-1-induced migration of colony-forming EPCs. Next, we investigated whether SFK play a role in SDF-1/CXCR4-mediated BM PC homing to the ischemic heart. BM MNCs from CXCR4BAC:eGFP reporter mice were i.v. injected into WT and SDF-1BAC:SDF1-RFP transgenic mice following surgically-induced myocardial infarction (MI). eGFP(+) MNCs and eGFP(+)c-kit(+) PCs that were recruited in the infarct border zone in SDF-1BAC:SDF1-RFP recipients were significantly more than that in WT recipients. Treatments of mice with SU6656 significantly reduced eGFP(+) and eGFP(+)c-kit(+) cell recruitment in both WT and SDF-1BAC:RFP recipients and abrogated the difference between the two groups. Remarkably, PCs isolated from BM-specific C-terminal Src kinase (CSK)-KO (Src activated) mice were recruited more efficiently than PCs from WT PCs in the WT recipients. In conclusion, SFK are activated by SDF-1/CXCR4 signaling and play an essential role in SDF-1/CXCR4-mediated BM PC chemotactic response and ischemic cardiac recruitment.


Asunto(s)
Células de la Médula Ósea/metabolismo , Quimiocina CXCL12/genética , Células Madre Mesenquimatosas/metabolismo , Isquemia Miocárdica/genética , Receptores CXCR4/genética , Familia-src Quinasas/genética , Animales , Bencilaminas , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Quimiocina CXCL12/antagonistas & inhibidores , Quimiocina CXCL12/metabolismo , Quimiotaxis/genética , Ciclamas , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Compuestos Heterocíclicos/farmacología , Indoles/farmacología , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Miocardio/metabolismo , Miocardio/patología , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores CXCR4/deficiencia , Transducción de Señal , Sulfonamidas/farmacología , Familia-src Quinasas/metabolismo
12.
Exp Oncol ; 36(3): 138-43, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25265345

RESUMEN

Philadelphia chromosome is a result of chromosomal rearrangement that leads to the appearing of the hybrid gene bcr/abl. A hybrid mRNA transcribes from bcr-promoter and many copies of hybrid molecules of Bcr/Abl protein are formed as a result of bcr/abl gene expression. It is supposed that a hybrid Abl molecule, replacing the normal one, in majority of cases functions abnormally or does not function at all. Also it is possible that Abl moiety of Bcr/Abl protein which is possibly recognized by some hypothetical cell control system interpreted by cell as an overproduction of c-abl. This, probably, leads to blocking the normal C-Abl molecules production from the normal c-abl gene transcribed from the second non-aberrant chromosome 9. Based on C-Abl physiological functions in conjunction with the most important proteins of which functions directly depend on its activity we tried to outline the research directions that might explain disruptions of the processes at chronic myeloleukosis such as cell migration due to CXCL12/CXCR4 axis activation, reparation, apoptosis, control for mitochondria state, and to propose new perspective therapeutic approaches based on all this knowledge.


Asunto(s)
Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Proteínas Proto-Oncogénicas c-abl/deficiencia , Receptores CXCR4/deficiencia , Proteínas de Fusión bcr-abl/genética , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo
13.
Exp Dermatol ; 23(10): 748-50, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25066162

RESUMEN

Hair follicle (HF) morphogenesis relies on the coordinated exchange of signals between mesenchymal and epithelial compartments of embryonic skin. Chemokine receptor Cxcr4 expression was recently identified in dermal condensates (DCs) of nascent HFs, but its role in promoting HF morphogenesis remains unknown. Our analyses confirmed Cxcr4 expression in condensate cells, and additionally revealed transient Cxcr4 expression in incipient epithelial hair placodes. Placodal Cxcr4 appeared prior to detection in DCs, representing a switch of expression between epithelial and mesenchymal compartments. To explore the functional role of this receptor in both compartments for early HF formation, we conditionally ablated Cxcr4 with condensate-targeting Tbx18(cre) knock-in and epidermis-targeting Krt14-cre transgenic mice. Conditional knockouts for both crosses were viable throughout embryogenesis and into adulthood. Morphological and biochemical marker analyses revealed comparable numbers of HFs forming in knockout embryos compared to wild-type littermate controls in both cases, suggesting that neither dermal nor epithelial Cxcr4 expression is required for early HF morphogenesis. We conclude that Cxcr4 expression and chemokine signaling through this receptor in embryonic mouse skin is dispensable for HF formation.


Asunto(s)
Folículo Piloso/embriología , Folículo Piloso/metabolismo , Receptores CXCR4/metabolismo , Animales , Epitelio/embriología , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Morfogénesis , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Transducción de Señal
14.
J Immunol ; 193(3): 1013-6, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973450

RESUMEN

Central memory (CM) CD8(+) T cells "remember" prior encounters because they maintain themselves through cell division in the absence of ongoing challenge (homeostatic self-renewal), as well as reproduce the CM fate while manufacturing effector cells during secondary Ag encounters (rechallenge self-renewal). We tested the consequence of conditional deletion of the bone marrow homing receptor CXCR4 on antiviral T cell responses. CXCR4-deficient CD8(+) T cells have impaired memory cell maintenance due to defective homeostatic proliferation. Upon rechallenge, however, CXCR4-deficient T cells can re-expand and renew the CM pool while producing secondary effector cells. The critical bone marrow-derived signals essential for CD8(+) T cell homeostatic self-renewal appear to be dispensable to yield self-renewing, functionally asymmetric cell fates during rechallenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Homeostasis/inmunología , Memoria Inmunológica , Receptores CXCR4/deficiencia , Receptores CXCR4/fisiología , Traslado Adoptivo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/citología , Células Clonales , Homeostasis/genética , Humanos , Memoria Inmunológica/genética , Inmunofenotipificación , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores CXCR4/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo
15.
PLoS One ; 9(4): e95626, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24752354

RESUMEN

T cell recirculation through extralymphoid tissues is essential to immune surveillance, host defense and inflammation. In this process, T cells enter the tissue from the blood and subsequently leave via the afferent lymph. In the absence of inflammation, T cells require CCR7 expression to egress from the skin or lung, which is consistent with the constitutive expression of the CCR7 ligand CCL21 on lymphatic endothelium. However, during chronic inflammation alternative chemoattractants come into play, allowing Ccr7-deficient (Ccr7-/-) T cells to egress efficiently from affected skin. As T cell egress from inflamed sites is a potential control point of the inflammatory response, we aimed to determine alternative T cell exit receptors using a mouse and a sheep model. We show that CCR7+ and CCR7- T cells exiting from the chronically inflamed skin were highly responsive to the CXCR4 ligand CXCL12, which was induced in the lymphatics in the inflamed site. Based on these findings, we hypothesized that CXCR4 mediates T cell egress from inflamed skin. However, pharmacological inhibition of CXCR4 did not affect the tissue egress of wildtype or Ccr7-/- CD4 and CD8 T cells after adoptive transfer into chronically inflamed skin. Similarly, adoptively transferred Cxcr4-/- Ccr7-/- and Ccr7-/- T cells egressed from the inflamed skin equally well. Based on these data, we conclude that, while CXCR4 might play an essential role for other cell types that enter the afferent lymphatics, it is dispensable for T cell egress from the chronically inflamed skin.


Asunto(s)
Inflamación/inmunología , Linfa/inmunología , Receptores CXCR4/metabolismo , Piel/patología , Linfocitos T/inmunología , Animales , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Enfermedad Crónica , Humanos , Inflamación/patología , Linfa/efectos de los fármacos , Ratones Endogámicos C57BL , Receptores CCR7/deficiencia , Receptores CCR7/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/deficiencia , Ovinos , Linfocitos T/efectos de los fármacos
16.
Immunity ; 39(5): 912-24, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24184055

RESUMEN

Germinal center (GC) B cells cycle between the dark zone (DZ) and light zone (LZ) during antibody affinity maturation. Whether this movement is necessary for GC function has not been tested. Here we show that CXCR4-deficient GC B cells, which are restricted to the LZ, are gradually outcompeted by WT cells indicating an essential role for DZ access. Remarkably, the transition between DZ centroblast and LZ centrocyte phenotypes occurred independently of positioning. However, CXCR4-deficient cells carried fewer mutations and were overrepresented in the CD73(+) memory compartment. These findings are consistent with a model where GC B cells change from DZ to LZ phenotype according to a timed cellular program but suggest that spatial separation of DZ cells facilitates more effective rounds of mutation and selection. Finally, we identify a network of DZ CXCL12-expressing reticular cells that likely support DZ functions.


Asunto(s)
Linfocitos B/citología , Centro Germinal/citología , Linfopoyesis/fisiología , Animales , Afinidad de Anticuerpos , Antígenos de Diferenciación de Linfocitos B/metabolismo , Ciclo Celular , Movimiento Celular , Quimiocina CXCL12/análisis , Selección Clonal Mediada por Antígenos , Centro Germinal/ultraestructura , Memoria Inmunológica , Ganglios Linfáticos/ultraestructura , Mediastino , Ratones , Infecciones por Orthomyxoviridae/inmunología , Ganglios Linfáticos Agregados/citología , Fenotipo , Células Plasmáticas/citología , Quimera por Radiación , Receptores CXCR4/análisis , Receptores CXCR4/deficiencia , Organismos Libres de Patógenos Específicos , Factores de Tiempo
17.
J Neurosci ; 33(44): 17527-37, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174685

RESUMEN

Gonadotropin-releasing hormone (GnRH) neurons are neuroendocrine cells, located in the hypothalamus, that play an essential role in mammalian reproduction. These neurons originate in the nasal placode and migrate during embryonic development, in association with olfactory/vomeronasal nerves, first in the nose, then through the cribriform plate to enter the forebrain, before settling in the hypothalamus. One of the molecules required for their early migration in the nose is the chemokine CXCL12, which is expressed in the embryonic nasal mesenchyme in an increasing ventral to dorsal gradient, presumably guiding GnRH neurons toward the forebrain. Mice lacking CXCR4, the receptor for CXCL12, exhibit defective GnRH cell movement and a significant reduction in their number, suggesting that CXCL12/CXCR4 signaling is important in the migration and survival of these neurons. Here, we investigated the role of the more recently identified second CXCL12 receptor, CXCR7, in GnRH neuron development. We demonstrate that CXCR7 is expressed along the migratory path of GnRH neurons in the nasal cavity and, although not expressed by GnRH neurons, it affects their migration as indicated by the ectopic accumulation of these cells in the nasal compartment in CXCR7(-/-) mice. Absence of CXCR7 caused abnormal accumulation of CXCL12-RFP at CXCR4-positive sites in the nasal area of CXCL12-RFP-transgenic mice and excessive CXCL12-dependent intracellular clustering of CXCR4 in GnRH neurons, suggesting internalization. These findings imply that CXCR7 regulates CXCL12 availability by acting as a scavenger along the migratory path of GnRH neurons and, thus, influences the migration of these cells in a noncell-autonomous manner.


Asunto(s)
Movimiento Celular/fisiología , Quimiocina CXCL12/genética , Hormona Liberadora de Gonadotropina/fisiología , Neuronas/citología , Neuronas/fisiología , Receptores CXCR/genética , Receptores CXCR/fisiología , Animales , Quimiocina CXCL12/biosíntesis , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Receptores CXCR/deficiencia , Receptores CXCR4/deficiencia , Receptores CXCR4/genética
18.
Glia ; 61(8): 1288-305, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23828719

RESUMEN

Embryonic meninges secrete the chemokine SDF-1/CXCL12 as a chemotactic guide for migrating neural stem cells, but SDF-1 is not known to directly regulate the functions of radial glia. Recently, the developing meninges have been shown to regulate radial glial function, yet the mechanisms and signals responsible for this phenomenon remain unclear. Moreover, as a nonmigratory cell type, radial glia do not conform to traditional models associated with chemokine signaling in the central nervous system. Using fluorescent transgenes, in vivo genetic manipulations and pharmacological techniques, we demonstrate that SDF-1 derived from the meninges exerts a CXCR4-dependent effect on radial glia. Deletion of CXCR4 expression by radial glia influences their morphology, mitosis, and progression through both oligodendroglial and astroglial lineages. Additionally, disruption of CXCR4 signaling in radial glia has a transient effect on the migration of oligodendrocyte progenitors. These data indicate that a specific chemokine signal derived from the meninges has multiple regulatory effects on radial glia.


Asunto(s)
Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Células-Madre Neurales/fisiología , Receptores CXCR4/deficiencia , Receptores CXCR4/fisiología , Transducción de Señal/fisiología , Médula Espinal/embriología , Médula Espinal/fisiología , Animales , Linaje de la Célula/fisiología , Movimiento Celular/genética , Células Ependimogliales/fisiología , Femenino , Ratones Noqueados , Mitosis/genética , Técnicas de Cultivo de Órganos , Embarazo , Transducción de Señal/genética , Médula Espinal/citología , Transgenes
19.
Circ Res ; 113(5): 505-16, 2013 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-23838132

RESUMEN

RATIONALE: Cardiac neural crest cells (NCs) contribute to heart morphogenesis by giving rise to a variety of cell types from mesenchyme of the outflow tract, ventricular septum, and semilunar valves to neurons of the cardiac ganglia and smooth muscles of the great arteries. Failure in cardiac NC development results in outflow and ventricular septation defects commonly observed in congenital heart diseases. Cardiac NCs derive from the vagal neural tube, which also gives rise to enteric NCs that colonize the gut; however, so far, molecular mechanisms segregating these 2 populations and driving cardiac NC migration toward the heart have remained elusive. OBJECTIVE: Stromal-derived factor-1 (SDF1) is a chemokine that mediates oriented migration of multiple embryonic cells and mice deficient for Sdf1 or its receptors, Cxcr4 and Cxcr7, exhibit ventricular septum defects, raising the possibility that SDF1 might selectively drive cardiac NC migration toward the heart via a chemotactic mechanism. METHODS AND RESULTS: We show in the chick embryo that Sdf1 expression is tightly coordinated with the progression of cardiac NCs expressing Cxcr4. Cxcr4 loss-of-function causes delayed migration and enhanced death of cardiac NCs, whereas Sdf1 misexpression results in their diversion from their normal pathway, indicating that SDF1 acts as a chemoattractant for cardiac NCs. These alterations of SDF1 signaling result in severe cardiovascular defects. CONCLUSIONS: These data identify Sdf1 and its receptor Cxcr4 as candidate genes responsible for cardiac congenital pathologies in human.


Asunto(s)
Quimiocina CXCL12/fisiología , Defectos del Tabique Interventricular/fisiopatología , Cresta Neural/patología , Receptores CXCR4/fisiología , Animales , Animales Modificados Genéticamente , Movimiento Celular , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/deficiencia , Quimiocina CXCL12/genética , Quimiotaxis , Embrión de Pollo , Quimera , Coturnix/embriología , Ectodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Defectos del Tabique Interventricular/genética , MicroARNs/genética , Tubo Neural/citología , Tubo Neural/trasplante , Especificidad de Órganos , Organogénesis , Receptores CXCR/biosíntesis , Receptores CXCR/genética , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/biosíntesis , Receptores CXCR4/deficiencia , Receptores CXCR4/genética , Transducción de Señal , Especificidad de la Especie , Transfección
20.
Int J Biochem Cell Biol ; 45(8): 1813-20, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23742988

RESUMEN

Cysteine (C)-X-C chemokine receptor-4 (CXCR4) is the primary transmembrane receptor for stromal cell-derived factor-1 (SDF-1). We previously reported in mouse or human bone marrow-derived mesenchymal stromal stem cells (BMSCs) that deleting or antagonizing CXCR4 inhibits bone morphogenetic protein-2 (BMP2)-induced osteogenic differentiation. The goal of this study was to determine whether CXCR4-deficiency in BMSCs is an age-related effect in association with impaired osteogenic differentiation potency of aged BMSCs. Using BMSCs derived from C57BL/6J wild type mice at ages ranging from 3 to 23 months old, we detected decreased CXCR4 mRNA and protein expression as well as SDF-1 secretion with advancing aging. Moreover, CXCR4-deficient BMSCs from elderly vs. young mice exhibited impaired osteogenic differentiation in response to BMP2 stimulation or when cultured in dexamethasone (Dex)-containing osteogenic medium, evidenced by decreased alkaline phosphatase activity, osteocalcin synthesis, and calcium deposition (markers for immature and mature osteoblasts). Mechanistically, impaired BMP2- or Dex-osteoinduction in BMSCs of elderly mice was mediated by inhibited phosphorylation of intracellular R-Smads and Erk1/2 or Erk1/2 and p38 proteins, and decreased Runx2 and Osx expression (osteogenesis "master" regulators) were also detected. Furthermore, adenovirus-mediated repair of CXCR4 expression in BMSCs of elderly mice restored their osteogenic differentiation potentials to both BMP2 treatment and osteogenic medium. Collectively, our results demonstrate for the first time that CXCR4 expression in mouse BMSCs declines with aging, and this CXCR4-deficiency impairs osteogenic differentiation potency of aged BMSCs. These findings provide novel insights into mechanisms underlying age-related changes in BMSC-osteogenesis, and will potentiate CXCR4 as a therapeutic target to improve BMSC-based bone repair and regeneration in broad orthopedic situations.


Asunto(s)
Envejecimiento/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular , Células Madre Mesenquimatosas/citología , Osteogénesis , Receptores CXCR4/deficiencia , Adenoviridae/metabolismo , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/enzimología , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular/efectos de los fármacos , Medios de Cultivo/farmacología , Dexametasona/farmacología , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/enzimología , Ratones , Ratones Endogámicos C57BL , Osteogénesis/efectos de los fármacos , Receptores CXCR4/metabolismo , Proteínas Smad/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...