Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
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
2.
Blood Adv ; 7(14): 3382-3394, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37042948

RESUMO

Cell polarity, the asymmetric distribution of proteins and organelles, is permanently or transiently established in various cell types and plays an important role in many physiological events. epidermal growth factor receptor substrate 15 homology domain-binding protein 1-like 1 (EHBP1L1) is an adapter protein that is localized on recycling endosomes and regulates apical-directed transport in polarized epithelial cells. However, the role of EHBP1L1 in nonepithelial cells, remains unknown. Here, Ehbp1l1-/- mice showed impaired erythroblast enucleation. Further analyses showed that nuclear polarization before enucleation was impaired in Ehbp1l1-/- erythroblasts. It was also revealed that EHBP1L1 interactors Rab10, Bin1, and dynamin were involved in erythroblast enucleation. In addition, Ehbp1l1-/- erythrocytes exhibited stomatocytic morphology and dehydration. These defects in erythroid cells culminated in early postnatal anemic lethality in Ehbp1l1-/- mice. Moreover, we found the mislocalization of nuclei and mitochondria in the skeletal muscle cells of Ehbp1l1-/- mice, as observed in patients with centronuclear myopathy with genetic mutations in Bin1 or dynamin 2. Taken together, our findings indicate that the Rab8/10-EHBP1L1-Bin1-dynamin axis plays an important role in multiple cell polarity systems in epithelial and nonepithelial cells.


Assuntos
Núcleo Celular , Eritroblastos , Animais , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Núcleo Celular/metabolismo , Dinaminas/metabolismo , Eritroblastos/metabolismo , Eritrócitos/metabolismo
3.
Cell Rep ; 39(6): 110805, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35545056

RESUMO

Myelodysplastic syndrome (MDS) is a clonal disorder of hematopoietic stem cells (HSCs), characterized by ineffective hematopoiesis and frequent progression to leukemia. It has long remained unresolved how MDS cells, which are less proliferative, inhibit normal hematopoiesis and eventually dominate the bone marrow space. Despite several studies implicating mesenchymal stromal or stem cells (MSCs), a principal component of the HSC niche, in the inhibition of normal hematopoiesis, the molecular mechanisms underlying this process remain unclear. Here, we demonstrate that both human and mouse MDS cells perturb bone metabolism by suppressing the osteolineage differentiation of MSCs, which impairs the ability of MSCs to support normal HSCs. Enforced MSC differentiation rescues the suppressed normal hematopoiesis in both in vivo and in vitro MDS models. Intriguingly, the suppression effect is reversible and mediated by extracellular vesicles (EVs) derived from MDS cells. These findings shed light on the novel MDS EV-MSC axis in ineffective hematopoiesis.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Síndromes Mielodisplásicas , Animais , Vesículas Extracelulares/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Síndromes Mielodisplásicas/metabolismo
4.
Br J Haematol ; 193(3): 659-668, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33837967

RESUMO

A population of mesenchymal stem cells, termed CXC chemokine ligand (CXCL)12-abundant reticular (CAR) cells or leptin receptor-expressing cells, are the major cellular component of niches for haematopoietic stem cells (HSCs) in murine bone marrow. CAR cells are characterized by several salient features, including much higher expression of CXCL12, stem cell factor (SCF), forkhead box C1 (FOXC1) and early B-cell factor 3 (EBF3), which are essential for HSC maintenance, than other cells. However, the human counterpart of CAR cells has not been fully described. Here, we show the presence of cells expressing much higher CXCL12 than other cells in human adult bone marrow using a flow cytometry-based in situ technique that enables high-throughput detection of mRNA at single-cell resolution. Most CXCL12hi cells expressed high levels of SCF, FOXC1 and EBF3 and had the potential to differentiate into adipocytes and osteoblasts. Histologically, the nuclei of CXCL12hi cells were identified and quantified by EBF3 expression in fixed marrow sections. CXCL12hi cells sorted from residual bone marrow aspirates of chronic myeloid leukaemia patients expressed reduced levels of CXCL12, SCF, FOXC1 and EBF3 in correlation with increased leukaemic burden. Together, we identified the human counterpart of CAR cells, enabling the evaluation of their alterations in various haematological disorders by flow cytometric and histological analyses.


Assuntos
Quimiocina CXCL12/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas de Neoplasias/metabolismo , Nicho de Células-Tronco , Adulto , Feminino , Fatores de Transcrição Forkhead/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Células-Tronco Mesenquimais/patologia , Fator de Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
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.
Blood ; 129(15): 2124-2131, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28130213

RESUMO

Hematopoietic stem cells (HSCs) reside in and are maintained by special microenvironments, termed niches. It is assumed that the HSC niche space remains occupied by endogenous cells and that myelosuppressive conditioning is required to achieve high levels of HSC engraftment. We herein demonstrate that upon the transplantation of very large numbers of purified HSCs into normal mice not exposed to myeloablation, donor HSCs engrafted in niches distant from filled HSC niches without replacing host HSCs and subsequently proliferated and generated hematopoietic progenitors, leading to marked increases in the overall HSC numbers in bone marrow. Additionally, stem cell factor that is produced by CXC chemokine ligand 12-abundant reticular cells is involved in HSC engraftment. In contrast, host granulocyte/macrophage progenitors (GMPs) were replaced by the progeny of transplanted donor HSCs, and overall GMP numbers remained unchanged. Thus, inconsistent with the classical concept, numerous empty HSC niches are available for engraftment and proliferation in bone marrow.


Assuntos
Sobrevivência de Enxerto , Células Progenitoras de Granulócitos e Macrófagos/metabolismo , Transplante de Células-Tronco Hematopoéticas , Homeostase , Nicho de Células-Tronco , Condicionamento Pré-Transplante , Aloenxertos , Animais , Medula Óssea/metabolismo , Quimiocina CXCL12/metabolismo , Camundongos , Camundongos Transgênicos
7.
Immunity ; 45(6): 1219-1231, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27913094

RESUMO

Hematopoietic stem cells (HSCs) self-renew in bone marrow niches formed by mesenchymal progenitors and endothelial cells expressing the chemokine CXCL12, but whether a separate niche instructs multipotent progenitor (MPP) differentiation remains unclear. We show that MPPs resided in HSC niches, where they encountered lineage-instructive differentiation signals. Conditional deletion of the chemokine receptor CXCR4 in MPPs reduced differentiation into common lymphoid progenitors (CLPs), which decreased lymphopoiesis. CXCR4 was required for CLP positioning near Interleukin-7+ (IL-7) cells and for optimal IL-7 receptor signaling. IL-7+ cells expressed CXCL12 and the cytokine SCF, were mesenchymal progenitors capable of differentiation into osteoblasts and adipocytes, and comprised a minor subset of sinusoidal endothelial cells. Conditional Il7 deletion in mesenchymal progenitors reduced B-lineage committed CLPs, while conditional Cxcl12 or Scf deletion from IL-7+ cells reduced HSC and MPP numbers. Thus, HSC maintenance and multilineage differentiation are distinct cell lineage decisions that are both controlled by HSC niches.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Multipotentes/citologia , Nicho de Células-Tronco/fisiologia , Animais , Linhagem da Célula/fisiologia , Separação Celular , Quimiocina CXCL2/metabolismo , Citometria de Fluxo , Interleucina-7/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Trends Immunol ; 36(9): 504-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26282886

RESUMO

A fraction of patients with T-cell acute lymphoblastic leukemias (T-ALLs) relapse and have a dismal prognosis. Two recent papers in Cancer Cell reveal that endothelial cell-derived CXCL12 is essential for bone marrow involvement and tumor progression in T-ALL patients, suggesting that this chemokine axis presents a potential therapeutic target for the treatment of T-ALL.


Assuntos
Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/biossíntese , Endotélio Vascular/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Piridinas/farmacologia , Animais , Feminino , Humanos
9.
Am J Pathol ; 185(7): 1859-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25934614

RESUMO

Hepatic expression levels of CXCL12, a chemokine important in inflammatory and stem cell recruitment, and its receptor, C-X-C chemokine receptor 4, are increased during all forms of liver injury. CXCL12 is expressed by both parenchymal and nonparenchymal hepatic cells, and on the basis of immunohistochemistry, biliary epithelial cells (BECs) are thought to be a predominant source of hepatic CXCL12, thereby promoting periportal recruitment of C-X-C chemokine receptor 4-expressing lymphocytes. Our study aims to show that BECs may, in fact, not be the predominant source of hepatic CXCL12. We measured CXCL12 secretion and expression from human and murine BECs using enzyme-linked immunosorbent assay and Western blot analysis from cell culture supernatants and whole cell lysates, respectively, whereas CXCL12 expression in murine livers was analyzed in a Cxcl12-Gfp reporter mouse. Cell culture supernatants and whole cell lysates from BECs failed to demonstrate their expression of CXCL12. Furthermore, we confirmed these results with a Cxcl12-Gfp reporter mouse in which green fluorescent protein expression is notably absent from BECs. Interestingly, on the basis of green fluorescent protein expression, we demonstrate a population of CXCL12-expressing cells within the portal tract that are distinct, yet intimately associated with BECs. These findings indicate that BECs are not a predominant source of CXCL12.


Assuntos
Quimiocina CXCL12/metabolismo , Células Epiteliais/metabolismo , Fígado/metabolismo , Animais , Ductos Biliares Intra-Hepáticos/citologia , Ductos Biliares Intra-Hepáticos/metabolismo , Linhagem Celular , Quimiocina CXCL12/genética , Ensaio de Imunoadsorção Enzimática , Expressão Gênica , Genes Reporter , Humanos , Fígado/citologia , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes de Fusão
10.
Immunity ; 42(1): 13-4, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25607454

RESUMO

The nature and functions of cells creating hematopoietic niches during inflammation remain incompletely understood. In this issue of Immunity, Kwak et al. (2015) reveal that myeloid cell-produced reactive oxygen species stimulate proliferation of myeloid progenitors establishing an additional mechanism to regulate hematopoiesis.


Assuntos
Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Granulócitos/fisiologia , Hematopoese , Células Mieloides/fisiologia , Células Progenitoras Mieloides/fisiologia , Animais
11.
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
12.
Dig Dis Sci ; 57(11): 2892-900, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22674400

RESUMO

BACKGROUND: The chemokine SDF-1 and its receptor CXCR4 are essential for the proper functioning of multiple organs. In the liver, cholangiocytes and hepatic progenitor cells (HPCs) are the main cells that produce SDF-1, and SDF-1 is thought to be essential for HPC-stimulated liver regeneration. AIMS: In this study, CXCR4 conditionally targeted mice were used to analyze the role of SDF-1 in chronically damaged liver. METHODS: Chronic liver damage was induced in MxCre CXCR4(f/null) mice and the control MxCre CXCR4(f/wt) mice by CCl(4). Serum markers were analyzed to assess liver function and damage, the number of cytokeratin-positive cells as a measure of HPCs, and the extent of liver fibrosis. Additional parameters relating to liver damage, such as markers of HPCs, liver function, MMPs, and TIMPs were measured by real-time PCR. RESULTS: Serum ALT was significantly higher in MxCre CXCR4(f/null) mice than MxCre CXCR4(f/wt) mice. The number of cytokeratin-positive cells and the area of fibrosis were also increased in the MxCre CXCR4(f/null) mice. The expression of mRNAs for several markers related to hepatic damage and regeneration was also increased in the liver of MxCre CXCR4(f/null) mice, including primitive HPC marker prominin-1, MMP9, TNF-α, and α-SMA. CONCLUSIONS: MxCre CXCR4(f/null) mice were susceptible to severe chronic liver damage, suggesting that SDF-1-CXCR4 signals are important for liver regeneration and preventing the progression of liver disease. Modulation of SDF-1 may therefore be a promising treatment strategy for patients with chronic liver disease.


Assuntos
Quimiocina CXCL12/metabolismo , Hepatopatias/metabolismo , Receptores CXCR4/metabolismo , Animais , Doença Crônica , Suscetibilidade a Doenças , Citometria de Fluxo , Testes de Função Hepática , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Estatísticas não Paramétricas , Células-Tronco/metabolismo
13.
J Immunol ; 189(1): 200-10, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22649198

RESUMO

Whereas most hematopoietic stem cells (HSC) are quiescent in homeostasis, they actively proliferate in response to bone marrow (BM) injury. Signals from the BM microenvironment are thought to promote entry of HSC into the cell cycle. However, it has been cumbersome to assess cycle status of viable HSC and thus explore unique features associated with division. In this study, we show that expression of endothelial cell-selective adhesion molecule (ESAM) can be a powerful indicator of HSC activation. ESAM levels clearly mirrored the shift of HSC between quiescence and activation, and it was prominent in comparison with other HSC-related Ags. ESAM(hi) HSC were actively dividing, but had surprisingly high long-term reconstituting capacity. Immunohistochemical analyses showed that most ESAM(hi) HSC were located near vascular endothelium in the BM after 5-fluorouracil treatment. To determine the importance of ESAM in the process of BM recovery, ESAM knockout mice were treated with 5-fluorouracil and their hematopoietic reconstruction was examined. The ESAM deficiency caused severe and prolonged BM suppression, suggesting that ESAM is functionally indispensable for HSC to re-establish homeostatic hematopoiesis. With respect to intracellular regulators, NF-κB and topoisomerase II levels correlated with the ESAM upregulation. Thus, our data demonstrate that the intensity of ESAM expression is useful to trace activated HSC and to understand molecular events involved in stem cell states.


Assuntos
Antígenos/fisiologia , Moléculas de Adesão Celular/fisiologia , Diferenciação Celular/imunologia , Endotélio Vascular/imunologia , Células-Tronco Hematopoéticas/imunologia , Fase de Repouso do Ciclo Celular/imunologia , Animais , Antígenos/biossíntese , Antígenos/genética , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/metabolismo , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/deficiência , Diferenciação Celular/genética , Linhagem Celular , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Fase de Repouso do Ciclo Celular/genética
14.
Inflamm Allergy Drug Targets ; 11(3): 201-6, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22452607

RESUMO

In mammals, hematopoietic stem cells (HSCs), which give rise to all blood cells and their progenies, including immune cells are controlled by special microenvironments, termed niches in the bone marrow during homeostasis and infection. However, the identity, nature and function of these niches remain unclear. It has been reported that HSCs are in contact with osteoblasts lining the bone surface and osteoblasts act as niches for HSCs (termed endosteal niche). However, recent studies suggest that only a small number of HSCs reside in the endosteal niche. In contrast, many HSCs are shown to be in contact with endothelial cells in the marrow. In addition, recent studies suggest that primitive mesenchymal cells, including CXCL12-abundant reticular (CAR) cells and Nestin-expressing cells, which have the ability to differentiate into adipocytes as well as osteoblasts act as niches for HSCs. Here we review candidate niches for HSCs in the bone marrow controlling hematopoiesis and chronic inflammation.


Assuntos
Células da Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Osteoblastos/metabolismo , Adipócitos/metabolismo , Animais , Diferenciação Celular/fisiologia , Microambiente Celular , Doença Crônica , Hematopoese/fisiologia , Homeostase , Humanos , Inflamação/fisiopatologia
15.
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
16.
Immunity ; 34(4): 463-5, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21511181

RESUMO

The roles of hematopoietic stem and/or progenitor cell niches during infection remain unclear. In this issue of Immunity, Shi et al. (2011) reveal that these niches upregulate MCP1 chemokine expression, inducing emigration of bone marrow monocytes into the circulation via the endothelium.

17.
Proc Natl Acad Sci U S A ; 108(1): 302-7, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173223

RESUMO

Increasing evidence suggests that myeloid bone marrow-derived cells (BMDCs) play a critical role in lung metastasis. Blockade of VEGF receptor 1 (VEGFR1) has been proposed as a potential strategy to limit myeloid BMDC recruitment to tumors. However, preclinical evidence indicates that this strategy may not be effective in all tumors. Thus, establishing which molecular mechanisms are responsible for the "escape" of these BMDCs from VEGFR1 inhibition would facilitate development of strategies to control metastasis. Here, we report the complementary role of the chemokine (C-X-C motif) ligand 12/C-X-C chemokine receptor 4 (CXCR4) and VEGF/VEGFR1 pathways in promoting lung metastasis in mice via BMDC recruitment using chimeric mice with deficiency in CXCR4 and VEGFR1-tyrosine kinase in the BMDCs. We first demonstrate that CXCR4 activity is essential for recruitment of myeloid differentiation antigen (Gr-1)-positive BMDCs, whereas VEGFR1 activity is responsible for macrophage recruitment in established tumors. Inhibition of both VEGFR1 and CXCR4 signaling in myeloid BMDCs exerted greater effects on tumor vascular density, growth, and lung metastasis than inhibition of VEGFR1 alone. These effects were reproduced after pharmacologic inhibition of CXCR4 with AMD3100. VEGFR1 and CXCR4 independently exerted a promigratory effect in myeloid BMDCs by activating p38 mitogen-activating protein kinase. Thus, combining CXCR4 blockade with inhibition of VEGFR1 may induce greater tumor growth delay and prevent or inhibit metastasis.


Assuntos
Neoplasias Pulmonares/fisiopatologia , Células Mieloides/metabolismo , Metástase Neoplásica/fisiopatologia , Receptores CXCR4/metabolismo , Receptores de Quimiocinas/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Transplante de Medula Óssea , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/genética
18.
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
19.
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
20.
Blood ; 115(26): 5401-11, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-20354171

RESUMO

Disrupted hematopoiesis and delayed immune reconstitution are life-threatening complications of allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although graft-versus-host disease (GVHD) is a major risk factor for the bone marrow (BM) insufficiency, how GVHD impairs BM hematopoiesis has been largely unknown. We hypothesized that BM stromal niche could be a target of GVHD. In major histocompatibility complex (MHC)-mismatched murine models of GVHD, we have demonstrated the early destruction of osteoblasts that especially affected B-cell lineages. The defective B lymphopoiesis was due to the impaired ability of BM stroma and osteoblasts to support the hematopoiesis, as evidenced by the failure of GVHD-affected BM to reconstitute the hematopoietic cells. The administration of anti-CD4 monoclonal antibody (mAb) ameliorated these effects and improved B lymphopoiesis while preserving graft-versus-tumor effects. Genetic ablation of Fas-Fas ligand signaling also partially restored B lymphopoiesis. Our present study provided evidence of BM GVHD, with the identification of osteoblasts as the main target for GVHD in BM. Moreover, our data showed the potential for mAb therapies to enhance immune reconstitution in vivo for patients undergoing allo-HSCT.


Assuntos
Medula Óssea/patologia , Doença Enxerto-Hospedeiro/imunologia , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Complexo Principal de Histocompatibilidade , Animais , Anticorpos Monoclonais/uso terapêutico , Medula Óssea/imunologia , Antígenos CD4/imunologia , Linhagem Celular , Células Endoteliais/patologia , Feminino , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/patologia , Humanos , Linfopoese , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA