Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 485(7400): 656-60, 2012 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-22660330

RESUMO

How environmental cues regulate adult stem cell and cancer cell activity through surface receptors is poorly understood. Angiopoietin-like proteins (ANGPTLs), a family of seven secreted glycoproteins, are known to support the activity of haematopoietic stem cells (HSCs) in vitro and in vivo. ANGPTLs also have important roles in lipid metabolism, angiogenesis and inflammation, but were considered 'orphan ligands' because no receptors were identified. Here we show that the immune-inhibitory receptor human leukocyte immunoglobulin-like receptor B2 (LILRB2) and its mouse orthologue paired immunoglobulin-like receptor (PIRB) are receptors for several ANGPTLs. LILRB2 and PIRB are expressed on human and mouse HSCs, respectively, and the binding of ANGPTLs to these receptors supported ex vivo expansion of HSCs. In mouse transplantation acute myeloid leukaemia models, a deficiency in intracellular signalling of PIRB resulted in increased differentiation of leukaemia cells, revealing that PIRB supports leukaemia development. Our study indicates an unexpected functional significance of classical immune-inhibitory receptors in maintenance of stemness of normal adult stem cells and in support of cancer development.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Leucemia/metabolismo , Leucemia/patologia , Glicoproteínas de Membrana/metabolismo , Receptores Imunológicos/metabolismo , Animais , Diferenciação Celular , Divisão Celular , Células Cultivadas , Modelos Animais de Doenças , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Proteína de Leucina Linfoide-Mieloide , Receptores Imunológicos/genética
2.
Blood ; 117(2): 470-9, 2011 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-20959605

RESUMO

The physiologic roles of angiopoietin-like proteins (Angptls) in the hematopoietic system remain unknown. Here we show that hematopoietic stem cells (HSCs) in Angptl3-null mice are decreased in number and quiescence. HSCs transplanted into Angptl3-null recipient mice exhibited impaired repopulation. Bone marrow sinusoidal endothelial cells express high levels of Angptl3 and are adjacent to HSCs. Importantly, bone marrow stromal cells or endothelium deficient in Angptl3 have a significantly decreased ability to support the expansion of repopulating HSCs. Angptl3 represses the expression of the transcription factor Ikaros, whose unregulated overexpression diminishes the repopulation activity of HSCs. Angptl3, as an extrinsic factor, thus supports the stemness of HSCs in the bone marrow niche.


Assuntos
Angiopoietinas/metabolismo , Células da Medula Óssea/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Nicho de Células-Tronco/metabolismo , Proteína 3 Semelhante a Angiopoietina , Proteínas Semelhantes a Angiopoietina , Animais , Medula Óssea , Células da Medula Óssea/citologia , Separação Celular , Citometria de Fluxo , Células-Tronco Hematopoéticas/citologia , Fator de Transcrição Ikaros/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicho de Células-Tronco/citologia
3.
Blood ; 118(12): 3236-43, 2011 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-21821709

RESUMO

The role of IGF binding protein 2 (IGFBP2) in cell growth is intriguing and largely undefined. Previously we identified IGFBP2 as an extrinsic factor that supports ex vivo expansion of hematopoietic stem cells (HSCs). Here we showed that IGFBP2-null mice have fewer HSCs than wild-type mice. While IGFBP2 has little cell-autonomous effect on HSC function, we found decreased in vivo repopulation of HSCs in primary and secondary transplanted IGFBP2-null recipients. Importantly, bone marrow stromal cells that are deficient for IGFBP2 have significantly decreased ability to support the expansion of repopulating HSCs. To investigate the mechanism by which IGFBP2 supports HSC activity, we demonstrated that HSCs in IGFBP2-null mice had decreased survival and cycling, down-regulated expression of antiapoptotic factor Bcl-2, and up-regulated expression of cell cycle inhibitors p21, p16, p19, p57, and PTEN. Moreover, we found that the C-terminus, but not the RGD domain, of extrinsic IGFBP2 was essential for support of HSC activity. Defective signaling of the IGF type I receptor did not rescue the decreased repopulation of HSCs in IGFBP2-null recipients, suggesting that the environmental effect of IGFBP2 on HSCs is independent of IGF-IR mediated signaling. Therefore, as an environmental factor, IGFBP2 supports the survival and cycling of HSCs.


Assuntos
Proteínas Inibidoras de Quinase Dependente de Ciclina/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Contagem de Células , Ciclo Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Proteínas Inibidoras de Quinase Dependente de Ciclina/genética , Regulação para Baixo/efeitos dos fármacos , Feminino , Citometria de Fluxo , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Proteína 2 de Ligação a Fator de Crescimento Semelhante à Insulina/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , PTEN Fosfo-Hidrolase/genética , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/genética , Receptor IGF Tipo 1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
4.
J Neurosci ; 27(49): 13481-90, 2007 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-18057206

RESUMO

The adult brain maintains two regions of neurogenesis from which new neurons are born, migrate to their appropriate location, and become incorporated into the circuitry of the CNS. One of these, the subgranular zone of the hippocampal dentate gyrus, is of primary interest because of the role of this region in learning and memory. We show that mice lacking EphB1, and more profoundly EphB1 and EphB2, have significantly fewer neural progenitors in the hippocampus. Furthermore, other aspects of neurogenesis, such as polarity, cell positioning, and proliferation are disrupted in animals lacking the EphB1 receptor or its cognate ephrin-B3 ligand. Our data strongly suggest that EphB1 and ephrin-B3 cooperatively regulate the proliferation and migration of neural progenitors in the hippocampus and should be added to a short list of candidate target molecules for modulating the production and integration of new neurons as a treatment for neurodegenerative diseases or brain injury.


Assuntos
Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Proliferação de Células , Hipocampo/citologia , Neurônios/citologia , Receptores da Família Eph/fisiologia , Células-Tronco/fisiologia , Animais , Hipocampo/metabolismo , Hipocampo/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Receptor EphB1/biossíntese , Receptor EphB1/genética , Receptor EphB1/fisiologia , Receptor EphB3/biossíntese , Receptor EphB3/genética , Receptor EphB3/fisiologia , Receptores da Família Eph/biossíntese , Receptores da Família Eph/genética , Células-Tronco/citologia , Células-Tronco/metabolismo
5.
Cell Stem Cell ; 9(2): 119-30, 2011 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-21816363

RESUMO

The lack of understanding of the interplay between hematopoietic stem cells (HSCs) and the immune system has severely hampered the stem cell research and practice of transplantation. Major problems for allogeneic transplantation include low levels of donor engraftment and high risks of graft-versus-host disease (GVHD). Transplantation of purified allogeneic HSCs diminishes the risk of GVHD but results in decreased engraftment. Here we show that ex vivo expanded mouse HSCs efficiently overcame the major histocompatibility complex barrier and repopulated allogeneic-recipient mice. An 8-day expansion culture led to a 40-fold increase of the allograft ability of HSCs. Both increased numbers of HSCs and culture-induced elevation of expression of the immune inhibitor CD274 (B7-H1 or PD-L1) on the surface of HSCs contributed to the enhancement. Our study indicates the great potential of utilizing ex vivo expanded HSCs for allogeneic transplantation and suggests that the immune privilege of HSCs can be modulated.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Animais , Antígeno B7-H1/metabolismo , Contagem de Células , Membrana Celular/imunologia , Proliferação de Células , Células Cultivadas , Técnicas de Introdução de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Fenótipo , Transplante Homólogo , Regulação para Cima
6.
PLoS One ; 6(3): e18054, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21464968

RESUMO

Solid tumors are composed of cancerous cells and non-cancerous stroma. A better understanding of the tumor stroma could lead to new therapeutic applications. However, the exact compositions and functions of the tumor stroma are still largely unknown. Here, using a Lewis lung carcinoma implantation mouse model, we examined the hematopoietic compartments in tumor stroma and tumor-bearing mice. Different lineages of differentiated hematopoietic cells existed in tumor stroma with the percentage of myeloid cells increasing and the percentage of lymphoid and erythroid cells decreasing over time. Using bone marrow reconstitution analysis, we showed that the tumor stroma also contained functional hematopoietic stem cells. All hematopoietic cells in the tumor stroma originated from bone marrow. In the bone marrow and peripheral blood of tumor-bearing mice, myeloid populations increased and lymphoid and erythroid populations decreased and numbers of hematopoietic stem cells markedly increased with time. To investigate the function of hematopoietic cells in tumor stroma, we co-implanted various types of hematopoietic cells with cancer cells. We found that total hematopoietic cells in the tumor stroma promoted tumor development. Furthermore, the growth of the primary implanted Lewis lung carcinomas and their metastasis were significantly decreased in mice reconstituted with IGF type I receptor-deficient hematopoietic stem cells, indicating that IGF signaling in the hematopoietic tumor stroma supports tumor outgrowth. These results reveal that hematopoietic cells in the tumor stroma regulate tumor development and that tumor progression significantly alters the host hematopoietic compartment.


Assuntos
Compartimento Celular , Células-Tronco Hematopoéticas/patologia , Neoplasias/patologia , Animais , Contagem de Células , Diferenciação Celular , Proliferação de Células , Transplante de Células-Tronco Hematopoéticas , Camundongos , Lesões Pré-Cancerosas/patologia , Células Estromais/patologia
7.
Cell ; 125(6): 1151-63, 2006 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-16777604

RESUMO

More than 10(10) cells are generated every day in the human intestine. Wnt proteins are key regulators of proliferation and are known endogenous mitogens for intestinal progenitor cells. The positioning of cells within the stem cell niche in the intestinal epithelium is controlled by B subclass ephrins through their interaction with EphB receptors. We report that EphB receptors, in addition to directing cell migration, regulate proliferation in the intestine. EphB signaling promotes cell-cycle reentry of progenitor cells and accounts for approximately 50% of the mitogenic activity in the adult mouse small intestine and colon. These data establish EphB receptors as key coordinators of migration and proliferation in the intestinal stem cell niche.


Assuntos
Movimento Celular , Proliferação de Células , Intestinos/citologia , Receptor EphB2/fisiologia , Receptor EphB3/fisiologia , Células-Tronco/citologia , Adenoma/metabolismo , Adenoma/patologia , Animais , Ciclo Celular , Diferenciação Celular , Colo/citologia , Colo/metabolismo , Humanos , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Knockout , Receptor EphB2/biossíntese , Receptor EphB2/genética , Receptor EphB3/biossíntese , Receptor EphB3/genética , Transdução de Sinais , Proteínas Wnt/fisiologia
8.
Dev Biol ; 271(2): 272-90, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15223334

RESUMO

Incomplete urethral tubularization (hypospadias) and anorectal abnormalities are two common and poorly understood birth defects that affect the extreme caudal midline of the human embryo. We now show that cell surface molecules essential for proper axon pathfinding in the developing nervous system, namely ephrin-B2 and the ephrin receptors EphB2 and EphB3, also play major roles in cell adhesion events that tubularize the urethra and partition the urinary and alimentary tracts. Mice carrying mutations which disrupt the bidirectional signals that these molecules transduce develop with variably penetrant severe hypospadias and incomplete midline fusion of the primitive cloaca. We further show that animals completely lacking ephrin-B2 reverse signaling present a fully penetrant failure in cloacal septation. This results in severe anorectal malformations characterized by an absence of the terminal-most hindgut (rectum) and formation of a fistula that aberrantly connects the intestines to the urethra at the base of the bladder. Consistent with an apparent requisite for both forward and reverse signaling in these caudal remodeling events, EphB2 and ephrin-B2 are coexpressed at the midline in the fusing urethral/cloacal endoderm and underlying lateral mesoderm of the urorectal septum that migrates toward the caudal midline as the cloaca septates. Our data thus indicate that B-subclass Eph and ephrin molecules play an important role in these clinically significant midline cell-cell adhesion and fusion events.


Assuntos
Canal Anal/embriologia , Efrina-B2/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hipospadia/embriologia , Receptor EphB2/fisiologia , Transdução de Sinais/fisiologia , Canal Anal/anormalidades , Animais , Adesão Celular/fisiologia , Primers do DNA , Efrina-B2/genética , Imunofluorescência , Componentes do Gene , Técnicas de Transferência de Genes , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Mutação/fisiologia
9.
Dev Biol ; 271(2): 263-71, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15223333

RESUMO

Vascular development begins with the formation of a primary vascular plexus that is rapidly remodeled by angiogenesis into the interconnected branched patterns characteristic of mature vasculature. Several receptor tyrosine kinases and their ligands have been implicated to control early development of the vascular system. These include the vascular endothelial growth factor receptors (VEGFR-1 and VEGFR-2) that bind VEGF, the Tie-1 and Tie-2 receptors that bind the angiopoietins, and the EphB4 receptor that binds the membrane-anchored ligand ephrin-B2. Targeted mutations in the mouse germline have revealed essential functions for these molecules in vascular development. In particular, protein-null mutations that delete either EphB4 or ephrin-B2 from the mouse have been shown to result in early embryonic lethality due to failed angiogenic remodeling. The venous expression of EphB4 and arterial expression of ephrin-B2 has lead to the speculation that the interaction of these two molecules leads to bidirectional signaling into both the receptor-expressing cell and the ligand-expressing cell, and that both forward and reverse signals are required for proper development of blood vessels in the embryo. Indeed, targeted removal of the ephrin-B2 carboxy-terminal cytoplasmic tail by another group was shown to perturb vascular development and result in the same early embryonic lethality as the null mutation, leading the authors to propose that ephrin-B2 reverse signaling directs early angiogenic remodeling of the primary vascular plexus [Cell 104 (2001) 57]. However, we show here that the carboxy-terminal cytoplasmic domain of ephrin-B2, and hence reverse signaling, is not required during early vascular development, but it is necessary for neonatal survival and functions later in cardiovascular development in the maturation of cardiac valve leaflets. We further show that ephrin-B2 reverse signaling is required for the pathfinding of axons that form the posterior tract of the anterior commissure. Our results thus indicate that ephrin-B2 functions in the early embryo as a typical instructive ligand to stimulate EphB4 receptor forward signaling during angiogenic remodeling and that later in embryonic development ephrin-B2 functions as a receptor to transduce reverse signals involved in cardiac valve maturation and axon pathfinding.


Assuntos
Axônios/fisiologia , Efrina-B2/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Valvas Cardíacas/embriologia , Transdução de Sinais/fisiologia , Animais , Movimento Celular/fisiologia , Efrina-B2/metabolismo , Citometria de Fluxo , Imunofluorescência , Técnicas Histológicas , Ligantes , Camundongos , Neovascularização Fisiológica/fisiologia , Receptor EphB4/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA