Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38430523

RESUMEN

Bone marrow is a dynamic organ composed of stem cells that constantly receive signals from stromal cells and other hematopoietic cells in the niches of the bone marrow to maintain hematopoiesis and generate immune cells. Perturbation of the bone marrow microenvironment by infection and inflammation affects hematopoiesis and may affect immune cell development. Little is known about the effect of malaria on the bone marrow stromal cells that govern the hematopoietic stem cell (HSC) niche. In this study, we demonstrate that the mesenchymal stromal CXCL12-abundant reticular (CAR) cell population is reduced during acute malaria infection. The reduction of CXCL12 and interleukin-7 signals in the bone marrow impairs the lymphopoietic niche, leading to the depletion of common lymphoid progenitors, B cell progenitors, and mature B cells, including plasma cells in the bone marrow. We found that interferon-γ (IFNγ) is responsible for the upregulation of Sca1 on CAR cells, yet the decline in CAR cell and B cell populations in the bone marrow is IFNγ-independent. In contrast to the decline in B cell populations, HSCs and multipotent progenitors increased with the expansion of myelopoiesis and erythropoiesis, indicating a bias in the differentiation of multipotent progenitors during malaria infection. These findings suggest that malaria may affect host immunity by modulating the bone marrow niche.


Asunto(s)
Linfocitos B , Médula Ósea , Quimiocina CXCL12 , Malaria , Ratones Endogámicos C57BL , Animales , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/inmunología , Ratones , Malaria/inmunología , Malaria/parasitología , Linfocitos B/inmunología , Médula Ósea/inmunología , Médula Ósea/parasitología , Nicho de Células Madre/inmunología , Interferón gamma/metabolismo , Interferón gamma/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo
2.
Nat Commun ; 14(1): 6402, 2023 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880234

RESUMEN

Lympho-hematopoiesis is regulated by cytokines; however, it remains unclear how cytokines regulate hematopoietic stem cells (HSCs) to induce production of lymphoid progenitors. Here, we show that in mice whose CXC chemokine ligand 12 (CXCL12) is deleted from half HSC niche cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, HSCs migrate from CXCL12-deficient niches to CXCL12-intact niches. In mice whose CXCL12 is deleted from all Ebf3+/leptin receptor (LepR)+ CAR cells, HSCs are markedly reduced and their ability to generate B cell progenitors is reduced compared with that to generate myeloid progenitors even when transplanted into wild-type mice. Additionally, CXCL12 enables the maintenance of B lineage repopulating ability of HSCs in vitro. These results demonstrate that CAR cell-derived CXCL12 attracts HSCs to CAR cells within bone marrow and plays a critical role in the maintenance of HSCs, especially lymphoid-biased or balanced HSCs. This study suggests an additional mechanism by which cytokines act on HSCs to produce B cells.


Asunto(s)
Quimiocinas CXC , Células Madre Hematopoyéticas , Ratones , Animales , Ligandos , Células Madre Hematopoyéticas/fisiología , Médula Ósea , Hematopoyesis , Quimiocina CXCL12 , Nicho de Células Madre , Factores de Transcripción
3.
Inflamm Regen ; 43(1): 15, 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36805714

RESUMEN

Throughout adult life, most lineages of blood cells, including immune cells, are generated from hematopoietic stem cells (HSCs) in the bone marrow. HSCs are thought to require special microenvironments, termed niches, for their maintenance in the bone marrow; however, the identity of the HSC cellular niche has been a subject of long-standing debate. Although diverse candidates have been proposed so far, accumulated studies demonstrate that the bone marrow-specific population of fibroblastic reticular cells with long processes, termed CXC chemokine ligand 12-abundant reticular cells (which overlap strongly with leptin receptor-expressing cells), termed CAR/LepR+ cells, are the pivotal cellular component of niches for HSCs and lymphoid progenitors. Sinusoidal endothelial cells (ECs) are also important for hematopoietic homeostasis and regeneration. Hematopoiesis is altered dynamically by various stimuli such as inflammation, infection, and leukemia, all of which affect cellular niches and alter their function. Therefore, it is important to consider situations in which stimuli affect HSCs, either via direct interaction or indirectly via the hematopoietic niches. In this review, the dynamics of cellular niches in the steady state and disease are described, with a focus on CAR/LepR+ cells and ECs.

4.
Nat Commun ; 13(1): 2654, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35551452

RESUMEN

In bone marrow, special microenvironments, known as niches, are essential for the maintenance of hematopoietic stem cells (HSCs). A population of mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells or leptin receptor-expressing cells are the major cellular component of HSC niches. The molecular regulation of HSC niche properties is not fully understood. The role of Runx transcription factors, Runx1 and Runx2 in HSC cellular niches remains unclear. Here we show that Runx1 is predominantly expressed in CAR cells and that mice lacking both Runx1 and Runx2 in CAR cells display an increase in fibrosis and bone formation with markedly reduced hematopoietic stem and progenitor cells in bone marrow. In vitro, Runx1 is induced by the transcription factor Foxc1 and decreases fibrotic gene expression in CAR cells. Thus, HSC cellular niches require Runx1 or Runx2 to prevent their fibrotic conversion and maintain HSCs and hematopoiesis in adults.


Asunto(s)
Células Madre Hematopoyéticas , Nicho de Células Madre , Animales , Médula Ósea/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Fibrosis , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Ratones
5.
Curr Top Microbiol Immunol ; 434: 33-54, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34850281

RESUMEN

Most types of blood cells, including immune cells are generated from hematopoietic stem cells (HSCs) within bone marrow in the adult. Most HSCs are in contact with and require the special microenvironment known as a niche for their maintenance. It has been thought that HSC niches comprise various types of support cells that provide critical signals, including cytokines and extracellular matrix for HSC regulation. However, among these cells, several lines of evidence have demonstrated that the population of bone marrow-specific mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor-expressing (LepR+) cells, is the major cellular component of HSC niches. CAR/LepR+ cells give rise to most adipocytes and osteoblasts in adult bone marrow and express much higher levels of HSC niche factors, including cytokines CXCL12 and stem cell factor (SCF), which are essential for HSC maintenance, and transcription factors Foxc1 and Ebf3, which are essential for the formation and maintenance of HSC niches than other types of cells. CAR/LepR+ cells are present in human bone marrow, undergo fibrotic expansion, and have reduced expression of HSC niche factors in hematopoietic malignancies.


Asunto(s)
Neoplasias Hematológicas , Neoplasias , Médula Ósea , Neoplasias Hematológicas/genética , Hematopoyesis , Células Madre Hematopoyéticas , Homeostasis , Humanos , Nicho de Células Madre , Microambiente Tumoral
6.
Int Immunol ; 33(12): 821-826, 2021 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-34668936

RESUMEN

Most lineages of blood cells, including immune cells, are generated from hematopoietic stem cells (HSCs) in bone marrow throughout adult life. Since HSCs cannot expand on their own, they require and contact the special microenvironments, termed niches for their maintenance. HSC niches comprise supportive cells that provide adjacent HSCs with critical signals, including cytokines. Although bone marrow microenvironments have been thought to be complex, recent studies have demonstrated that the bone marrow-specific population of fibroblastic reticular cells with long processes, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor (LepR)-expressing (LepR+) cells, is the major cellular component of niches for HSCs and lymphoid progenitors. CAR cells have salient features, expressing much higher levels of critical HSC niche factors than any other cell populations and function as self-renewing mesenchymal stem cells. Human counterpart of CAR cells is present and affected in diseases, including leukemia. Foxl1+ telocytes recently identified as the niche for intestinal stem cells share some features with CAR cells, suggesting that CAR cells might serve as a prototype for fibroblastic reticular cells creating niche for long-lived cells, including tissue stem cells and memory lymphocytes. These findings provided the basis for future mechanistic studies on the cross-talk between hematopoietic cells and microenvironments in both health and disease.


Asunto(s)
Médula Ósea/inmunología , Fibroblastos/inmunología , Células Madre Hematopoyéticas/inmunología , Nicho de Células Madre/inmunología , Animales , Humanos
7.
Cell Rep ; 37(3): 109875, 2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34686326

RESUMEN

In mouse testis, a heterogeneous population of undifferentiated spermatogonia (Aundiff) harbors spermatogenic stem cell (SSC) potential. Although GFRα1+ Aundiff maintains the self-renewing pool in homeostasis, the functional basis of heterogeneity and the implications for their dynamics remain unresolved. Here, through quantitative lineage tracing of SSC subpopulations, we show that an ensemble of heterogeneous states of SSCs supports homeostatic, persistent spermatogenesis. Such heterogeneity is maintained robustly through stochastic interconversion of SSCs between a renewal-biased Plvap+/GFRα1+ state and a differentiation-primed Sox3+/GFRα1+ state. In this framework, stem cell commitment occurs not directly but gradually through entry into licensed but uncommitted states. Further, Plvap+/GFRα1+ cells divide slowly, in synchrony with the seminiferous epithelial cycle, while Sox3+/GFRα1+ cells divide much faster. Such differential cell-cycle dynamics reduces mitotic load, and thereby the potential to acquire harmful de novo mutations of the self-renewing pool, while keeping the SSC density high over the testicular open niche.


Asunto(s)
Células Madre Germinales Adultas/fisiología , Linaje de la Célula , Espermatogénesis , Testículo/fisiología , Células Madre Germinales Adultas/metabolismo , Animales , Autorrenovación de las Células , Regulación del Desarrollo de la Expresión Génica , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Mitosis , Modelos Biológicos , Fenotipo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Testículo/citología , Testículo/metabolismo , Factores de Tiempo
8.
Int Immunol ; 31(1): 5-11, 2019 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-30169696

RESUMEN

The special microenvironments, termed niches, with which hematopoietic stem cells (HSCs) are in contact, have been thought to be required for the maintenance of HSCs and the generation of immune cells in bone marrow. Although the identity of the HSC niche has been a subject of long-standing debate, recent findings demonstrate that a population of mesenchymal stem cells, termed CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells or leptin receptor-expressing (LepR+) cells, are the major cellular components of niches for HSCs and lymphoid progenitors, which express specific transcription factors, including Foxc1 and Ebf3, and cytokines, including CXCL12 and stem cell factor (SCF), essential for their niche functions. The identity and functions of other types of cells, including osteoblasts, sinusoidal endothelial cells, periarteriolar cells, megakaryocytes and a population of macrophages in HSC maintenance, have also been shown.


Asunto(s)
Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Nicho de Células Madre , Animales , Biomarcadores , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Endoteliales/citología , Células Endoteliales/metabolismo , Humanos , Sistema Inmunológico/metabolismo , Leucocitos/inmunología , Leucocitos/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Células de Schwann/inmunología , Células de Schwann/metabolismo , Nicho de Células Madre/genética , Nicho de Células Madre/inmunología
9.
Genes Dev ; 32(5-6): 359-372, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29563184

RESUMEN

Bone marrow is the tissue filling the space between bone surfaces. Hematopoietic stem cells (HSCs) are maintained by special microenvironments known as niches within bone marrow cavities. Mesenchymal cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells or leptin receptor-positive (LepR+) cells, are a major cellular component of HSC niches that gives rise to osteoblasts in bone marrow. However, it remains unclear how osteogenesis is prevented in most CAR/LepR+ cells to maintain HSC niches and marrow cavities. Here, using lineage tracing, we found that the transcription factor early B-cell factor 3 (Ebf3) is preferentially expressed in CAR/LepR+ cells and that Ebf3-expressing cells are self-renewing mesenchymal stem cells in adult marrow. When Ebf3 is deleted in CAR/LepR+ cells, HSC niche function is severely impaired, and bone marrow is osteosclerotic with increased bone in aged mice. In mice lacking Ebf1 and Ebf3, CAR/LepR+ cells exhibiting a normal morphology are abundantly present, but their niche function is markedly impaired with depleted HSCs in infant marrow. Subsequently, the mutants become progressively more osteosclerotic, leading to the complete occlusion of marrow cavities in early adulthood. CAR/LepR+ cells differentiate into bone-producing cells with reduced HSC niche factor expression in the absence of Ebf1/Ebf3 Thus, HSC cellular niches express Ebf3 that is required to create HSC niches, to inhibit their osteoblast differentiation, and to maintain spaces for HSCs.


Asunto(s)
Médula Ósea/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Médula Ósea/patología , Diferenciación Celular , Linaje de la Célula , Regulación de la Expresión Génica , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Osteogénesis/genética , Nicho de Células Madre , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética
10.
Clin Calcium ; 26(5): 671-6, 2016 May.
Artículo en Japonés | MEDLINE | ID: mdl-27117611

RESUMEN

It has been hypothesized that the special microenvironments known as niches in the bone marrow play an essential role in maintaining hematopoietic stem and progenitor cells(HSPCs), and the identification of the HSPC niche has been a subject of long-standing argument. Recent studies identified candidate cells meeting the criteria for HSPC niches and the critical transcription factor of their development and maintenance.


Asunto(s)
Médula Ósea/inmunología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Osteoblastos/citología , Nicho de Células Madre/inmunología , Animales , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Movimiento Celular/fisiología , Células Madre Hematopoyéticas/inmunología , Humanos
11.
Curr Opin Hematol ; 22(4): 330-6, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26049754

RESUMEN

PURPOSE OF REVIEW: It has been assumed that the special microenvironments known as niches in the marrow play an essential role in maintaining hematopoietic stem and progenitor cells (HSPCs), and the identity of the HSPC niche has been a subject of long-standing debate. Recent studies identified cells, which create microenvironments meeting the criteria for HSPC niches and the critical transcriptional regulators of their development and maintenance. RECENT FINDINGS: Osterix as well as Ebf2 and Bmi1 are critical but not specific transcriptional regulators of HSPC niche development. The transcription factor Foxc1 is expressed preferentially in a population of adipo-osteogenic progenitors, termed CXCL12-abundant reticular (CAR) cells, which create HSPC niches and are largely equivalent to stem cell factor and Lepr-expressing cells, in developing and adult bone marrow. Foxc1 is essential for CAR cell development and maintenance of bone marrow niches for HSPCs upregulating CXCL12 and SCF expression and inhibition of adipogenic processes in CAR cell progenitors. SUMMARY: Foxc1 is the first critical and specific transcriptional regulator that is required for development and maintenance of cells creating HSPC niches, including a specialized population of adipo-osteogenic progenitors in bone marrow.


Asunto(s)
Quimiocina CXCL12/genética , Factores de Transcripción Forkhead/genética , Células Madre Hematopoyéticas/metabolismo , Factor de Células Madre/genética , Nicho de Células Madre/genética , Transcripción Genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Quimiocina CXCL12/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Transducción de Señal , Factor de Transcripción Sp7 , Factor de Células Madre/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Nature ; 508(7497): 536-40, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24590069

RESUMEN

Haematopoietic stem and progenitor cells are maintained by special microenvironments known as niches in bone marrow. Many studies have identified diverse candidate cells that constitute niches for haematopoietic stem cells in the marrow, including osteoblasts, endothelial cells, Schwann cells, α-smooth muscle actin-expressing macrophages and mesenchymal progenitors such as CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells, stem cell factor-expressing cells, nestin-expressing cells and platelet-derived growth factor receptor-α (PDGFR-α)(+)Sca-1(+)CD45(-)Ter119(-) (PαS) cells. However, the molecular basis of the formation of the niches remains unclear. Here we find that the transcription factor Foxc1 is preferentially expressed in the adipo-osteogenic progenitor CAR cells essential for haematopoietic stem and progenitor cell maintenance in vivo in the developing and adult bone marrow. When Foxc1 was deleted in all marrow mesenchymal cells or CAR cells, from embryogenesis onwards, osteoblasts appeared normal, but haematopoietic stem and progenitor cells were markedly reduced and marrow cavities were occupied by adipocytes (yellow adipose marrow) with reduced CAR cells. Inducible deletion of Foxc1 in adult mice depleted haematopoietic stem and progenitor cells and reduced CXCL12 and stem cell factor expression in CAR cells but did not induce a change to yellow marrow. These data suggest a role for Foxc1 in inhibiting adipogenic processes in CAR progenitors. Foxc1 might also promote CAR cell development, upregulating CXCL12 and stem cell factor expression. This study identifies Foxc1 as a specific transcriptional regulator essential for development and maintenance of the mesenchymal niches for haematopoietic stem and progenitor cells.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Células Madre Hematopoyéticas/citología , Nicho de Células Madre/fisiología , Adipocitos/citología , Adipocitos/metabolismo , Animales , Células de la Médula Ósea/citología , Recuento de Células , Diferenciación Celular , Quimiocina CXCL12/metabolismo , Desarrollo Embrionario/genética , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Factor de Células Madre/metabolismo
13.
Blood ; 119(23): 5429-37, 2012 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-22553313

RESUMEN

The BM microenvironment is required for the maintenance, proliferation, and mobilization of hematopoietic stem and progenitor cells (HSPCs), both during steady-state conditions and hematopoietic recovery after myeloablation. The ECM meshwork has long been recognized as a major anatomical component of the BM microenvironment; however, the molecular signatures and functions of the ECM to support HSPCs are poorly understood. Of the many ECM proteins, the expression of tenascin-C (TN-C) was found to be dramatically up-regulated during hematopoietic recovery after myeloablation. The TN-C gene was predominantly expressed in stromal cells and endothelial cells, known as BM niche cells, supporting the function of HSPCs. Mice lacking TN-C (TN-C(-/-)) mice showed normal steady-state hematopoiesis; however, they failed to reconstitute hematopoiesis after BM ablation and showed high lethality. The capacity to support transplanted wild-type hematopoietic cells to regenerate hematopoiesis was reduced in TN-C(-/-) recipient mice. In vitro culture on a TN-C substratum promoted the proliferation of HSPCs in an integrin α9-dependent manner and up-regulated the expression of the cyclins (cyclinD1 and cyclinE1) and down-regulated the expression of the cyclin-dependent kinase inhibitors (p57(Kip2), p21(Cip1), p16(Ink4a)). These results identify TN-C as a critical component of the BM microenvironment that is required for hematopoietic regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Médula Ósea/metabolismo , Hematopoyesis , Células Madre Hematopoyéticas/citología , Tenascina/metabolismo , Animales , Médula Ósea/efectos de los fármacos , Médula Ósea/efectos de la radiación , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Proliferación Celular , Células Endoteliales/citología , Células Endoteliales/metabolismo , Fluorouracilo/administración & dosificación , Fluorouracilo/efectos adversos , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Cadenas alfa de Integrinas/genética , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Células del Estroma/citología , Células del Estroma/metabolismo , Tenascina/análisis , Tenascina/genética , Regulación hacia Arriba , Irradiación Corporal Total
14.
Trends Immunol ; 32(7): 315-20, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21531624

RESUMEN

In the bone marrow, hematopoietic stem cells (HSCs) are maintained by special microenvironments, termed niches. The nature and function of these niches, however, remains unclear. HSCs are thought be in contact with bone-lining osteoblasts, but recent studies have suggested that only a small subpopulation of HSCs reside in this endosteal niche. By contrast, many HSCs are associated with the sinusoidal endothelium, which is referred to as the vascular niche. Recent data have suggested that primitive mesenchymal cells, including CXC chemokine ligand 12-abundant reticular cells and nestin-expressing cells act as HSC niches. Here, we review HSC niches, with an emphasis on the emerging role of reticular niches for maintaining HSCs in a proliferative and undifferentiated state.


Asunto(s)
Médula Ósea/inmunología , Células Madre Hematopoyéticas/inmunología , Animales , Médula Ósea/irrigación sanguínea , Movimiento Celular , Células Madre Hematopoyéticas/citología , Homeostasis , Humanos , Fenotipo
15.
J Exp Med ; 208(5): 949-60, 2011 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-21536740

RESUMEN

Vasculogenesis describes the process of de novo vessel formation from vascular precursor cells. Although formation of the first major vessels, such as the dorsal aorta and cardinal veins, occurs during embryonic vasculogenesis, the contribution of precursor cell populations to postnatal vessel development is not well understood. Here, we identified a novel population of postnatal vascular precursor cells in mice. These cells express the Schwann cell protein myelin protein zero (Po) and exhibit a CD45(-)CD31(-)VEcad(-)c-kit(+)CXCR4(+) surface phenotype. Po(+) vascular precursors (PVPs) are recruited into the growing vasculature, and comprise a minor population of arterial endothelial cells in adult mice. Recruitment of PVPs into growing vessels is mediated by CXCL12-CXCR4 signaling, and is enhanced during vascular expansion induced by Notch inhibition. Po-specific ablation of Flk1, a receptor for VEGF, results in branching defects and insufficient arterial patterning in the retina, as well as reduced neovascularization of tumors and ischemic tissues. Thus, in postnatal mice, although growing vessels are formed primarily by angiogenesis from preexisting vessels, a minor population of arterial endothelia may be derived from tissue-resident vascular precursor cells.


Asunto(s)
Antígenos de Diferenciación/metabolismo , Células Endoteliales/metabolismo , Proteína P0 de la Mielina/metabolismo , Neovascularización Fisiológica/fisiología , Células Madre/metabolismo , Animales , Antígenos de Diferenciación/genética , Línea Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células Endoteliales/citología , Ratones , Ratones Transgénicos , Proteína P0 de la Mielina/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Retina/citología , Retina/metabolismo , Células Madre/citología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
16.
Blood ; 117(2): 451-8, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20944068

RESUMEN

Natural killer (NK) cells are granular lymphocytes that are generated from hematopoietic stem cells and play vital roles in the innate immune response against tumors and viral infection. Generation of NK cells is known to require several cytokines, including interleukin-15 (IL-15) and Fms-like tyrosine kinase 3 ligand, but not IL-2 or IL-7. Here we investigated the in vivo role of CXC chemokine ligand-12 (CXCL12) and its primary receptor CXCR4 in NK-cell development. The numbers of NK cells appeared normal in embryos lacking CXCL12 or CXCR4; however, the numbers of functional NK cells were severely reduced in the bone marrow, spleen, and peripheral blood from adult CXCR4 conditionally deficient mice compared with control animals, probably resulting from cell-intrinsic CXCR4 deficiency. In culture, CXCL12 enhanced the generation of NK cells from lymphoid-primed multipotent progenitors and immature NK cells. In the bone marrow, expression of IL-15 mRNA was considerably higher in CXCL12-abundant reticular (CAR) cells than in other marrow cells, and most NK cells were in contact with the processes of CAR cells. Thus, CXCL12-CXCR4 chemokine signaling is essential for NK-cell development in adults, and CAR cells might function as a niche for NK cells in bone marrow.


Asunto(s)
Envejecimiento , Diferenciación Celular/fisiología , Quimiocina CXCL12/metabolismo , Células Asesinas Naturales/citología , Receptores CXCR4/metabolismo , Transducción de Señal/fisiología , Animales , Separación Celular , Citometría de Flujo , Hematopoyesis/fisiología , Inmunohistoquímica , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Immunity ; 33(3): 387-99, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20850355

RESUMEN

Hematopoietic stem cells (HSCs) and their lympho-hematopoietic progeny are supported by microenvironmental niches within bone marrow; however, the identity, nature, and function of these niches remain unclear. Short-term ablation of CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells in vivo did not affect the candidate niches, bone-lining osteoblasts, or endothelial cells but severely impaired the adipogenic and osteogenic differentiation potential of marrow cells and production of the cytokines SCF and CXCL12 and led to a marked reduction in cycling lymphoid and erythroid progenitors. HSCs from CAR cell-depleted mice were reduced in number and cell size, were more quiescent, and had increased expression of early myeloid selector genes, similar to the phenotype of wild-type HSCs cultured without a niche. Thus, the niche composed of adipo-osteogenic progenitors is required for proliferation of HSCs and lymphoid and erythroid progenitors, as well as maintenance of HSCs in an undifferentiated state.


Asunto(s)
Adipogénesis , Células Madre Hematopoyéticas/fisiología , Osteogénesis , Células Madre/fisiología , Adipocitos/citología , Animales , Diferenciación Celular , Células Cultivadas , Quimiocina CXCL12/fisiología , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular/fisiología , Ratones , Ratones Endogámicos C57BL , Osteoblastos/citología , Factor de Células Madre/genética
18.
Blood ; 110(13): 4153-60, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17827391

RESUMEN

Plasmacytoid dendritic cells (pDCs), also known as type I interferon (IFN)-producingcells, are thought to play central roles in antiviral immunity and the pathogenesis of some autoimmune diseases. pDCs are produced from hematopoietic stem cells in bone marrow. However, the environmental regulation of the development of pDCs is not fully understood. Here, we show that the numbers of pDCs and their earliest progenitors are severely reduced in the absence of CXCR4, the primary physiologic receptor for CXC chemokine ligand 12 (CXCL12), also known as stromal cell-derived factor-1 (SDF-1) in vivo. In vitro, CXCL12 induces a significant increase in pDC numbers generated from primitive hematopoietic cells, and pDCs and their progenitors migrate to CXCL12. In addition, most pDCs are in contact with CXCL12-abundant reticular (CAR) cells in the intersinal space of bone marrow, although many primitive hematopoietic cells adjoin CAR cells surrounding sinusoidal endothelial cells or residing near the bone surface. Thus we identified CXCL12 as a key regulator of pDC development produced by cellular niches, providing new targets for pDC therapeutic control.


Asunto(s)
Médula Ósea , Quimiocina CXCL12/fisiología , Células Dendríticas/citología , Receptores CXCR4/fisiología , Transducción de Señal , Células del Estroma/citología , Animales , Recuento de Células , Movimiento Celular , Genotipo , Células Madre Hematopoyéticas , Ratones , Ratones Noqueados , Ratones Transgénicos
19.
J Immunol ; 174(11): 6657-62, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15905504

RESUMEN

Plasmacytoid dendritic cells (PDCs) are defined in mice by a unique combination of markers: CD11c, B220, and Ly6C/G. We have reported previously that PDCs express Ly49Q, a lectin-type killer cell inhibitory receptor. We now find that different expression levels of Ly49Q define sequential developmental stages of PDCs in bone marrow. Although PDCs in spleen and lymph nodes express high levels of Ly49Q, a significant portion of CD11c(+)B220(+) PDCs in bone marrow lack Ly49Q, as well as the CD4 and MHC II. Purified Ly49Q(-) marrow PDCs spontaneously up-regulate Ly49Q after overnight culture without cell proliferation and acquire most features of typical PDCs in spleen. When exposed to TLR ligands, such as CpG-oligodeoxynucleotide and hemagglutinating virus of Japan (Sendai virus), Ly49Q(-) PDCs increase CD86 and MHC class II expression but produce less IFN-alphabeta, IL-6, and IL-12p70 than Ly49Q(+) PDCs, although they are able to produce comparable amounts of TNF-alpha. However, interestingly, Ly49Q(-) PDCs do not produce TNF-alpha in response to the TLR2 ligand, Pam3SCK(4), whereas Ly49Q(+) PDCs did. Therefore, Ly49Q is a new marker to identify a precursor form of PDCs that participates in innate immunity.


Asunto(s)
Antígenos Ly/biosíntesis , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/biosíntesis , Animales , Antígenos Ly/sangre , Antígenos Ly/fisiología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células Cultivadas , Citocinas/biosíntesis , Células Dendríticas/metabolismo , Femenino , Inmunofenotipificación , Células Asesinas Naturales/metabolismo , Lectinas Tipo C , Hígado/citología , Hígado/inmunología , Hígado/metabolismo , Pulmón/citología , Pulmón/inmunología , Pulmón/metabolismo , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Subfamilia A de Receptores Similares a Lectina de Células NK , Especificidad de Órganos/inmunología , Receptores Inmunológicos/sangre , Receptores Inmunológicos/fisiología , Receptores Similares a Lectina de Células NK
20.
J Immunol ; 174(8): 4621-9, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15814685

RESUMEN

Ly49Q is a member of the Ly49 family that is expressed on Gr-1+ cells but not on NK and NKT cells. Ly49Q appears to be involved in regulating cytoskeletal architectures through ITIM-mediated signaling. We provide evidence that dendritic cells (DCs) of certain maturational states expressed Ly49Q, and that IFN-alpha plays an important role in its regulation. Freshly prepared murine plasmacytoid pre-DCs as well as Flt3L-induced plasmacytoid pre-DCs expressed Ly49Q, whereas freshly prepared myeloid DCs did not. However, GM-CSF-induced myeloid DCs showed low levels of Ly49Q expression, and this was significantly enhanced by IFN-alpha. In contrast, other cytokines and ligands for TLRs such as TNF-alpha, IL-6, LPS, and CpG-ODN had little or no effect on Ly49Q expression. Plasmacytoid pre-DCs in all mouse strains examined expressed Ly49Q. Constitutive expression of Ly49Q on myeloid DCs was observed in three restricted mouse strains including 129, NZB, and NZW. As can be seen in other Ly49 family members, Ly49Q expression was affected by MHC class I expression. At the same time, Ly49Q possessed polymorphisms, including at least three alleles. The polymorphic residues lay within the stalk and carbohydrate recognition domain, and two of them, in loop 3 and loop 6 of the carbohydrate recognition domain, are located in the region implicated in the interaction of Ly49A with H-2D(d). Therefore, depending on IFN-alpha, our results imply that Ly49Q serves a role for the biological functions of certain DC subsets through recognition of MHC class I or related molecules.


Asunto(s)
Antígenos Ly/genética , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Asesinas Naturales/inmunología , Receptores Inmunológicos/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Citocinas/farmacología , ADN/genética , Células Dendríticas/clasificación , Células Dendríticas/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase I/metabolismo , Técnicas In Vitro , Interferón Tipo I/farmacología , Lectinas Tipo C , Ratones , Ratones Endogámicos , Ratones Noqueados , Datos de Secuencia Molecular , Subfamilia A de Receptores Similares a Lectina de Células NK , Polimorfismo Genético , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Similares a Lectina de Células NK , Proteínas Recombinantes , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA