Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Genes Dev ; 32(5-6): 359-372, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29563184

RESUMO

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.


Assuntos
Medula Óssea/metabolismo , Células-Tronco Mesenquimais/metabolismo , Fatores de Transcrição/metabolismo , Fatores Etários , Animais , Medula Óssea/patologia , Diferenciação Celular , Linhagem da Célula , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Osteogênese/genética , Nicho de Células-Tronco , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética
2.
Int Immunol ; 36(7): 339-352, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38430523

RESUMO

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.


Assuntos
Linfócitos B , Medula Óssea , Quimiocina CXCL12 , Malária , Camundongos Endogâmicos C57BL , Animais , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/imunologia , Camundongos , Malária/imunologia , Malária/parasitologia , Linfócitos B/imunologia , Medula Óssea/imunologia , Medula Óssea/parasitologia , Nicho de Células-Tronco/imunologia , Interferon gama/metabolismo , Interferon gama/imunologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo
3.
Curr Top Microbiol Immunol ; 434: 33-54, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34850281

RESUMO

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.


Assuntos
Neoplasias Hematológicas , Neoplasias , Medula Óssea , Neoplasias Hematológicas/genética , Hematopoese , Células-Tronco Hematopoéticas , Homeostase , Humanos , Nicho de Células-Tronco , Microambiente Tumoral
4.
Int Immunol ; 33(12): 821-826, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34668936

RESUMO

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.


Assuntos
Medula Óssea/imunologia , Fibroblastos/imunologia , Células-Tronco Hematopoéticas/imunologia , Nicho de Células-Tronco/imunologia , Animais , Humanos
5.
Int Immunol ; 31(1): 5-11, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30169696

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Sistema Imunitário/citologia , Sistema Imunitário/imunologia , Nicho de Células-Tronco , Animais , Biomarcadores , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Humanos , Sistema Imunitário/metabolismo , Leucócitos/imunologia , Leucócitos/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Células de Schwann/imunologia , Células de Schwann/metabolismo , Nicho de Células-Tronco/genética , Nicho de Células-Tronco/imunologia
6.
Nature ; 508(7497): 536-40, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24590069

RESUMO

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.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Hematopoéticas/citologia , Nicho de Células-Tronco/fisiologia , Adipócitos/citologia , Adipócitos/metabolismo , Animais , Células da Medula Óssea/citologia , Contagem de Células , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Desenvolvimento Embrionário/genética , Fatores de Transcrição Forkhead/deficiência , Fatores de Transcrição Forkhead/genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Fator de Células-Tronco/metabolismo
7.
Immunity ; 33(3): 387-99, 2010 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-20850355

RESUMO

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.


Assuntos
Adipogenia , Células-Tronco Hematopoéticas/fisiologia , Osteogênese , Células-Tronco/fisiologia , Adipócitos/citologia , Animais , Diferenciação Celular , Células Cultivadas , Quimiocina CXCL12/fisiologia , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Fator de Células-Tronco/genética
8.
Clin Calcium ; 26(5): 671-6, 2016 May.
Artigo em Japonês | MEDLINE | ID: mdl-27117611

RESUMO

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.


Assuntos
Medula Óssea/imunologia , Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Osteoblastos/citologia , Nicho de Células-Tronco/imunologia , Animais , Diferenciação Celular/imunologia , Movimento Celular/imunologia , Movimento Celular/fisiologia , Células-Tronco Hematopoéticas/imunologia , Humanos
9.
Curr Opin Hematol ; 22(4): 330-6, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26049754

RESUMO

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.


Assuntos
Quimiocina CXCL12/genética , Fatores de Transcrição Forkhead/genética , Células-Tronco Hematopoéticas/metabolismo , Fator de Células-Tronco/genética , Nicho de Células-Tronco/genética , Transcrição Gênica , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula/genética , Quimiocina CXCL12/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Complexo Repressor Polycomb 1/genética , Complexo Repressor Polycomb 1/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Transdução de Sinais , Fator de Transcrição Sp7 , Fator de Células-Tronco/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Blood ; 119(23): 5429-37, 2012 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-22553313

RESUMO

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.


Assuntos
Células da Medula Óssea/citologia , Medula Óssea/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Tenascina/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/efeitos da radiação , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Proliferação de Células , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Fluoruracila/administração & dosagem , Fluoruracila/efeitos adversos , Deleção de Genes , Células-Tronco Hematopoéticas/metabolismo , Cadeias alfa de Integrinas/genética , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Células Estromais/citologia , Células Estromais/metabolismo , Tenascina/análise , Tenascina/genética , Regulação para Cima , Irradiação Corporal Total
11.
Trends Immunol ; 32(7): 315-20, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21531624

RESUMO

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.


Assuntos
Medula Óssea/imunologia , Células-Tronco Hematopoéticas/imunologia , Animais , Medula Óssea/irrigação sanguínea , Movimento Celular , Células-Tronco Hematopoéticas/citologia , Homeostase , Humanos , Fenótipo
12.
Blood ; 117(2): 451-8, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20944068

RESUMO

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.


Assuntos
Envelhecimento , Diferenciação Celular/fisiologia , Quimiocina CXCL12/metabolismo , Células Matadoras Naturais/citologia , Receptores CXCR4/metabolismo , Transdução de Sinais/fisiologia , Animais , Separação Celular , Citometria de Fluxo , Hematopoese/fisiologia , Imuno-Histoquímica , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Inflamm Regen ; 43(1): 15, 2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36805714

RESUMO

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.

14.
Nat Commun ; 14(1): 6402, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880234

RESUMO

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.


Assuntos
Quimiocinas CXC , Células-Tronco Hematopoéticas , Camundongos , Animais , Ligantes , Células-Tronco Hematopoéticas/fisiologia , Medula Óssea , Hematopoese , Quimiocina CXCL12 , Nicho de Células-Tronco , Fatores de Transcrição
15.
Nat Commun ; 13(1): 2654, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551452

RESUMO

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.


Assuntos
Células-Tronco Hematopoéticas , Nicho de Células-Tronco , Animais , Medula Óssea/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Fibrose , Hematopoese/genética , Células-Tronco Hematopoéticas/metabolismo , Camundongos
16.
Cell Rep ; 37(3): 109875, 2021 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-34686326

RESUMO

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.


Assuntos
Células-Tronco Germinativas Adultas/fisiologia , Linhagem da Célula , Espermatogênese , Testículo/fisiologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Autorrenovação Celular , Regulação da Expressão Gênica no Desenvolvimento , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Mitose , Modelos Biológicos , Fenótipo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Testículo/citologia , Testículo/metabolismo , Fatores de Tempo
17.
J Exp Med ; 195(10): 1289-302, 2002 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12021309

RESUMO

Dendritic cells (DCs) are able in tissue culture to phagocytose and present antigens derived from infected, malignant, and allogeneic cells. Here we show directly that DCs in situ take up these types of cells after fluorescent labeling with carboxyfluorescein succinimidyl ester (CFSE) and injection into mice. The injected cells include syngeneic splenocytes and tumor cell lines, induced to undergo apoptosis ex vivo by exposure to osmotic shock, and allogeneic B cells killed by NK cells in situ. The CFSE-labeled cells in each case are actively endocytosed by DCs in vivo, but only the CD8+ subset. After uptake, all of the phagocytic CD8+ DCs can form major histocompatibility complex class II-peptide complexes, as detected with a monoclonal antibody specific for these complexes. The CD8+ DCs also selectively present cell-associated antigens to both CD4+ and CD8+ T cells. Similar events take place with cultured DCs; CD8+ DCs again selectively take up and present dying cells. In contrast, both CD8+ and CD8- DCs phagocytose latex particles in culture, and both DC subsets present soluble ovalbumin captured in vivo. Therefore CD8+ DCs are specialized to capture dying cells, and this helps to explain their selective ability to cross present cellular antigens to both CD4+ and CD8+ T cells.


Assuntos
Antígenos CD8/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Fagocitose , Animais , Apoptose , Linfócitos B/citologia , Linfócitos B/metabolismo , Morte Celular , Células Dendríticas/imunologia , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Fígado/citologia , Linfonodos/citologia , Camundongos , Pressão Osmótica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Fatores de Tempo , Células Tumorais Cultivadas
18.
J Exp Med ; 208(5): 949-60, 2011 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-21536740

RESUMO

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.


Assuntos
Antígenos de Diferenciação/metabolismo , Células Endoteliais/metabolismo , Proteína P0 da Mielina/metabolismo , Neovascularização Fisiológica/fisiologia , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/genética , Linhagem Celular Tumoral , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Células Endoteliais/citologia , Camundongos , Camundongos Transgênicos , Proteína P0 da Mielina/genética , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Receptores de Fatores de Crescimento do Endotélio Vascular/genética , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Retina/citologia , Retina/metabolismo , Células-Tronco/citologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
19.
Blood ; 110(13): 4153-60, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17827391

RESUMO

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.


Assuntos
Medula Óssea , Quimiocina CXCL12/fisiologia , Células Dendríticas/citologia , Receptores CXCR4/fisiologia , Transdução de Sinais , Células Estromais/citologia , Animais , Contagem de Células , Movimento Celular , Genótipo , Células-Tronco Hematopoéticas , Camundongos , Camundongos Knockout , Camundongos Transgênicos
20.
J Immunol ; 174(11): 6657-62, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15905504

RESUMO

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.


Assuntos
Antígenos Ly/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Receptores Imunológicos/biossíntese , Animais , Antígenos Ly/sangue , Antígenos Ly/fisiologia , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Células Cultivadas , Citocinas/biossíntese , Células Dendríticas/metabolismo , Feminino , Imunofenotipagem , Células Matadoras Naturais/metabolismo , Lectinas Tipo C , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Pulmão/citologia , Pulmão/imunologia , Pulmão/metabolismo , Tecido Linfoide/citologia , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Subfamília A de Receptores Semelhantes a Lectina de Células NK , Especificidade de Órgãos/imunologia , Receptores Imunológicos/sangue , Receptores Imunológicos/fisiologia , Receptores Semelhantes a Lectina de Células NK
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA