Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 143
Filtrar
1.
Leukemia ; 32(2): 462-469, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28744010

RESUMO

The acquired kinase mutation JAK2V617F plays a central role in myeloproliferative neoplasms (MPNs). However, the mechanisms responsible for the malignant hematopoietic stem/progenitor cell (HSPC) expansion seen in patients with MPNs are not fully understood, limiting the effectiveness of current treatment. Endothelial cells (ECs) are an essential component of the hematopoietic niche, and they have been shown to express the JAK2V617F mutation in patients with MPNs. We show that the JAK2V617F-bearing vascular niche promotes the expansion of the JAK2V617F HSPCs in preference to JAK2WT HSPCs, potentially contributing to poor donor cell engraftment and disease relapse following stem cell transplantation. The expression of Chemokine (C-X-C motif) ligand 12 (CXCL12) and stem cell factor (SCF) were upregulated in JAK2V617F-bearing ECs compared to wild-type ECs, potentially accounting for this observation. We further identify that the thrombopoietin (TPO)/MPL signaling pathway is critical for the altered vascular niche function. A better understanding of how the vascular niche contributes to HSPC expansion and MPN development is essential for the design of more effective therapeutic strategies for patients with MPNs.


Assuntos
Janus Quinase 2/genética , Transtornos Mieloproliferativos/genética , Neoplasias/genética , Animais , Quimiocina CXCL12/genética , Células Endoteliais/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Receptores de Trombopoetina/genética , Regulação para Cima/genética
2.
Leukemia ; 30(12): 2332-2341, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27133820

RESUMO

The myeloproliferative neoplasms (MPNs) are characterized by hematopoietic stem/progenitor cell (HSPC) expansion and overproduction of mature blood cells. The JAK2V617F mutation is present in hematopoietic cells in a majority of patients with MPNs, but the mechanism(s) responsible for MPN stem cell expansion remain incomplete. One hallmark feature of the marrow in patients with MPNs is megakaryocyte (MK) hyperplasia. We report here that mice bearing a human JAK2V617F gene restricted exclusively to the MK lineage develop many of the features of a MPN. Specifically, these mice exhibit thrombocytosis, splenomegaly, increased numbers of marrow and splenic hematopoietic progenitors and a substantial expansion of HSPCs. In addition, wild-type mice transplanted with cells from JAK2V617F-bearing MK marrow develop a myeloproliferative syndrome with thrombocytosis and erythrocytosis as well as pan-hematopoietic progenitor and stem cell expansion. As marrow histology in this murine model of myeloproliferation reveals a preferentially perivascular localization of JAK2V617F-mutant MKs and an increased marrow sinusoid vascular density, it adds to accumulating data that MKs are an important component of the marrow HSPC niche, and that MK expansion might indirectly contribute to the critical role of the thrombopoietin/c-Mpl signaling pathway in HSPC maintenance and expansion.


Assuntos
Proliferação de Células/genética , Células-Tronco Hematopoéticas/patologia , Janus Quinase 2/genética , Megacariócitos/metabolismo , Mutação de Sentido Incorreto , Animais , Células da Medula Óssea/patologia , Xenoenxertos , Humanos , Camundongos , Transtornos Mieloproliferativos/patologia , Nicho de Células-Tronco
3.
J Thromb Haemost ; 7 Suppl 1: 235-8, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19630807

RESUMO

The molecular pathways that regulate thrombopoiesis are becoming increasingly understood. Upon binding to its receptor, the product of the c-Mpl proto-oncogene, thrombopoietin activates a number of secondary messengers that promote cell survival, proliferation and differentiation. Amongst the best studied are the signal transducers and activators of transcription, phosphoinositol-3-kinase, and the mitogen-activated protein kinases. Additional signals activated by these secondary mediators include mammalian target of rapamycin, beta-catenin, hypoxia-inducible factor 1alpha and the homeobox proteins HOXB4 and HOXA9, and a number that are reduced, including glycogen synthase kinase 3alpha and the FOXO3 family of forkhead proteins. More recently, a number of signaling pathways have been identified that turn the thrombopoietin signal off, a step necessary to avoid uncontrolled myeloproliferation, and include the phosphatases PTEN, SHP1 and SHIP1, the suppressors of cytokine signaling, and down-modulation of surface expression of c-Mpl. This review will focus on these pathways in normal and neoplastic hematopoiesis.


Assuntos
Hematopoese , Transdução de Sinais , Trombopoetina/metabolismo , Humanos , Neoplasias , Proto-Oncogene Mas
4.
J Thromb Haemost ; 1(7): 1587-92, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12871295

RESUMO

Although first proposed to be the primary regulator of platelet production 45 years ago, the gene for thrombopoietin was cloned only within the last decade. Since then, our understanding of megakaryocyte and platelet production has increased substantially, and it is now appreciated that in addition to its critical role in regulating thrombopoiesis, the hormone affects multiple aspects of hematopoiesis, including playing a non-redundant role in stem cell survival, self-renewal and expansion. In addition to this greater physiological understanding of thrombopoietin biology, the molecular mechanisms by which the hormone affects cell survival and proliferation are coming under increased scrutiny. At least four signaling pathways have been identified that play important and non-overlapping roles in stem cell and megakaryocyte growth and development, potentially providing new strategies to therapeutically intervene in hematopoiesis. This review will focus on our current understanding of these processes.


Assuntos
Trombopoese/fisiologia , Trombopoetina/fisiologia , Animais , Plaquetas/metabolismo , Hematopoese , Humanos , Megacariócitos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Trombopoetina , Transdução de Sinais , Trombopoetina/química
7.
J Biol Chem ; 276(44): 41014-22, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11535599

RESUMO

Thrombopoietin (TPO) stimulates a network of intracellular signaling pathways that displays extensive cross-talk. We have demonstrated previously that the ERK/mitogen-activated protein kinase pathway is important for TPO-induced endomitosis in primary megakaryocytes (MKs). One known pathway by which TPO induces ERK activation is through the association of Shc with the penultimate phosphotyrosine within the TPO receptor, Mpl. However, several investigators found that the membrane-proximal half of the cytoplasmic domain of Mpl is sufficient to activate ERK in vitro and support base-line megakaryopoiesis in vivo. Using BaF3 cells expressing a truncated Mpl (T69Mpl) as a tool to identify non-Shc/Ras-dependent signaling pathways, we describe here novel mechanisms of TPO-induced ERK activation mediated, in part, by phosphoinositide 3-kinase (PI3K). Similar to cells expressing full-length receptor, PI3K was activated by its incorporation into a complex with IRS2 or Gab2. Furthermore, the MEK-phosphorylating activity of protein kinase Czeta (PKCzeta) was also enhanced after TPO stimulation of T69Mpl, contributing to ERK activity. PKCzeta and PI3K also contribute to TPO-induced ERK activation in MKs, confirming their physiological relevance. Like in BaF3 cells, a TPO-induced signaling complex containing p85PI3K is detectable in MKs expressing T61Mpl and is probably responsible for PI3K activation. These data demonstrate a novel role of PI3K and PKCzeta in steady-state megakaryopoiesis.


Assuntos
Megacariócitos/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/metabolismo , Trombopoetina/fisiologia , Animais , Divisão Celular , Linhagem Celular , Ativação Enzimática , Camundongos , Fosforilação
9.
J Cell Physiol ; 188(3): 291-303, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11473355

RESUMO

Megakaryocytes (MKs) develop from diploid progenitor cells via successive rounds of DNA synthesis in the absence of cell division, a process termed endomitosis (EnM). While the mechanism underlying EnM is not known, studies in yeast and leukemic cell lines have suggested that it may be due to reduced levels of cyclin B1 or cdc2, leading to a decrease in mitotic kinase activity. Using flow cytometry to study EnM highly purified marrow-derived MK precursors, we found that: (1) on average, 36% of 8N-32N MKs expressed abundant cyclin B during G2/M. The percentage of cells in G2/M decreased in >64N MKs, suggesting the limit of EnM, (2) the level of cyclin B per G2/M MK increased linearly with ploidy, (3) cyclin B expression oscillated normally in polyploid MKs, (4) MPM-2, a phosphoepitope created by the action of mitotic kinases and specific to M-phase cells, was expressed in a significant fraction of polyploid MKs, and (5) there was an apparent increase of cyclin B in G1-phase in polyploid MKs. This study provides the first qualitative kinetic data regarding the cell cycle status of MKs within individual ploidy classes. It also demonstrates the feasibility of using anti-cyclin B antibody and flow cytometry to resolve G1 from G2/M populations in polyploid MKs. Finally, these findings establish that neither a relative nor absolute deficiency of mitotic kinase components is responsible for EnM, suggesting that the departure from normal cell division kinetics seen in polyploid MKs is likely due to alterations in other cell cycle regulators.


Assuntos
Células-Tronco Hematopoéticas/citologia , Mitose/fisiologia , Poliploidia , Animais , Proteína Quinase CDC2/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Separação Celular , Células Cultivadas , Ciclina B/metabolismo , Citoplasma/metabolismo , DNA/análise , DNA/biossíntese , Epitopos/biossíntese , Citometria de Fluxo , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Cinética , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos , Mitose/efeitos dos fármacos , Fuso Acromático/metabolismo , Trombopoetina/farmacologia
10.
Ann N Y Acad Sci ; 938: 131-8, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11458500

RESUMO

Hematopoietic growth factors are glycoproteins of 15-70 kDa. Although much clinical success has been obtained using recombinant proteins produced in mammalian cell lines and in microbial fermentation processes, the full-length polypeptides necessarily are expensive to produce, require parenteral administration, and in some cases have provoked detrimental immune responses. With the availability of high throughput biological function and receptor binding assays it has become possible to screen millions, if not billions, of randomly produced organic compounds and relatively short peptides to identify lead compounds for the development of small molecular mimetics of hematopoietic growth factors. Herein the strategies used to screen libraries of small molecules and peptides and the successes in finding mimetics and antagonists for/to erythropoietin, granulocyte colony-stimulating factor, and thrombopoietin are reviewed. Finally, the structural study of mimetic-receptor complexes has provided us with many molecular details of growth factor-induced receptor activation and is likely to yield new insights into the molecular basis of hematopoietic signal transduction.


Assuntos
Desenho de Fármacos , Hematopoese/efeitos dos fármacos , Fatores de Crescimento de Células Hematopoéticas/química , Proteínas de Neoplasias , Biblioteca de Peptídeos , Receptores de Citocinas , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Bacteriófagos , Ligação Competitiva , Dimerização , Avaliação Pré-Clínica de Medicamentos/métodos , Eritropoetina/química , Eritropoetina/isolamento & purificação , Eritropoetina/farmacologia , Fator Estimulador de Colônias de Granulócitos/química , Fator Estimulador de Colônias de Granulócitos/farmacologia , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Humanos , Camundongos , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/farmacologia , Peptídeos Cíclicos/química , Peptídeos Cíclicos/isolamento & purificação , Peptídeos Cíclicos/farmacologia , Ligação Proteica , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Coelhos , Receptores da Eritropoetina/efeitos dos fármacos , Receptores de Fator Estimulador de Colônias de Granulócitos/efeitos dos fármacos , Receptores de Trombopoetina , Contagem de Reticulócitos , Relação Estrutura-Atividade , Trombopoetina/química , Trombopoetina/farmacologia
11.
J Biol Chem ; 276(37): 34473-9, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11418622

RESUMO

Thrombopoietin and its receptor (Mpl) support survival and proliferation in megakaryocyte progenitors and in BaF3 cells engineered to stably express Mpl (BaF3/Mpl). The binding of thrombopoietin to Mpl activates multiple kinase pathways, including the Jak/STAT, Ras/Raf/MAPK, and phosphatidylinositol 3-kinase pathways, but it is not clear how these kinases promote cell cycling. Here, we show that thrombopoietin induces phosphatidylinositol 3-kinase and that phosphatidylinositol 3-kinase is required for thrombopoietin-induced cell cycling in BaF3/Mpl cells and in primary megakaryocyte progenitors. Treatment of BaF3/Mpl cells and megakaryocytes with the phosphatidylinositol 3-kinase inhibitor LY294002 inhibited mitotic and endomitotic cell cycl-ing. BaF3/Mpl cells treated with thrombopoietin and LY294002 were blocked in G(1), whereas megakaryocyte progenitors treated with thrombopoietin and LY294002 showed both a G(1) and a G(2) cell cycle block. Expression of constitutively active Akt in BaF3/Mpl cells restored the ability of thrombopoietin to promote cell cycling in the presence of LY294002. Constitutively active Akt was not sufficient to drive proliferation of BaF3/Mpl cells in the absence of thrombopoietin. We conclude that in BaF3/Mpl cells and megakaryocyte progenitors, thrombopoietin-induced phosphatidylinositol 3-kinase activity is necessary but not sufficient for thrombopoietin-induced cell cycle progression. Phosphatidylinositol 3-kinase activity is likely to be involved in regulating the G(1)/S transition.


Assuntos
Megacariócitos/fisiologia , Proteínas de Neoplasias , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/fisiologia , Receptores de Citocinas , Trombopoetina/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Cromonas/farmacologia , Feminino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Morfolinas/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-akt , Receptores de Trombopoetina
14.
Blood ; 97(1): 1-2, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11133735
15.
Blood ; 97(1): 3, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11133736
16.
Blood ; 97(1): 154-61, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11133755

RESUMO

With the recent cloning and characterization of thrombopoietin, appreciation of the molecular events surrounding megakaryocyte (MK) development is growing. However, the final stages of platelet formation are less well understood. Platelet production occurs after the formation of MK proplatelet processes. In a study to explore the molecular mechanisms underlying this process, mature MKs isolated from suspension murine bone marrow cell cultures were induced to form proplatelets by exposure to plasma, and the role of various cell-signaling pathways was assessed. The results showed that (1) bis-indolylmaleimide I, which blocks protein kinase C (PKC) activation; (2) down-modulation of conventional or novel classes of PKC by phorbol myristate acetate; and (3) ribozymes specific for PKCalpha each inhibited proplatelet formation. Inhibition of several MAP kinases, PI3 kinase, or protein kinase A failed to affect MK proplatelet formation. To gain further insights into the function of PKCalpha in proplatelet formation, its subcellular localization was investigated. In cultures containing active proplatelet formation, cytoplasmic polymerized actin was highly aggregated, its subcellular distribution was reorganized, and PKCalpha colocalized with the cellular actin aggregates. A number of MK manipulations, including blockade of integrin signaling with a disintegrin or inhibition of actin polymerization with cytochalasin D, interrupted actin reorganization, PKC relocalization, and proplatelet formation. These findings suggest an important role for PKCalpha in proplatelet development and suggest that it acts by altering actin dynamics in proplatelet-forming MKs. Identification of the upstream and downstream pathways involved in proplatelet formation should provide greater insights into thrombopoiesis, potentially allowing pharmacologic manipulation of the process.


Assuntos
Actinas/metabolismo , Plaquetas/citologia , Megacariócitos/citologia , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Citoesqueleto de Actina/fisiologia , Actinas/antagonistas & inibidores , Actinas/ultraestrutura , Animais , Plaquetas/efeitos dos fármacos , Citocalasina D/farmacologia , Citoplasma , Desintegrinas/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/farmacologia , Isoenzimas/fisiologia , Megacariócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Microscopia de Fluorescência , Plasma , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/farmacologia , Proteína Quinase C/fisiologia , Proteína Quinase C-alfa , RNA Catalítico/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
17.
J Biol Chem ; 276(4): 2494-502, 2001 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-11054408

RESUMO

Thrombopoietin (TPO) is a recently characterized member of the hematopoietic growth factor family that serves as the primary regulator of megakaryocyte (MK) and platelet production. The hormone acts by binding to the Mpl receptor, the product of the cellular proto-oncogene c-mpl. Although many downstream signaling targets of TPO have been identified in cell lines, primary MKs, and platelets, the molecular mechanism(s) by which many of these molecules are activated remains uncertain. In this report we demonstrate that the TPO-induced activation of phosphoinositol 3-kinase (PI3K), a signaling intermediate vital for cellular survival and proliferation, occurs through its association with inducible signaling complexes in both BaF3 cells engineered to express Mpl (BaF3/Mpl) and in primary murine MKs. Although a direct association between PI3K and Mpl could not be demonstrated, we found that several proteins, including SHP2, Gab2, and IRS2, undergo phosphorylation and association in BaF3/Mpl cells in response to TPO stimulation, complexes that recruit and enhance the enzymatic activity of PI3K. To verify the physiological relevance of the complex, SHP2-Gab2 association was disrupted by overexpressing a dominant negative SHP2 construct. TPO-induced Akt phosphorylation was significantly decreased in transfected cells suggesting an important role of SHP2 in the complex to enhance PI3K activity. In primary murine MKs, TPO also induced phosphorylation of SHP2, its association with p85 and enhanced PI3K activity, but in contrast to the results in cell lines, neither Gab2 nor IRS2 are phosphorylated in MKs. Instead, a 100-kDa tyrosine-phosphorylated protein (pp100) co-immunoprecipitated with the regulatory subunit of PI3K. These findings support a model where PI3K activity is dependent on its recruitment into TPO-induced multiphosphoprotein complexes, implicate the existence of a scaffolding protein in primary MKs distinct from the known Gab and IRS proteins, and suggest that, in contrast to erythroid progenitor cells that employ Gab1 in PI3K signaling complexes, utilization of an alternate member of the Gab/IRS family could be responsible for specificity in TPO signaling.


Assuntos
Megacariócitos/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Trombopoetina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Linhagem Celular , Ativação Enzimática , Hematopoese , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Megacariócitos/metabolismo , Camundongos , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6
18.
Zhonghua Xue Ye Xue Za Zhi ; 22(6): 296-9, 2001 Jun.
Artigo em Chinês | MEDLINE | ID: mdl-11877087

RESUMO

OBJECTIVE: To characterize the inhibitory effect of interferon-alpha (IFN-alpha) on megakaryocyte proliferation and differentiation stimulated by thrombopoietin (Tpo). METHODS: Normal murine megakaryocytes were prepared by three different cell purification procedures. Megakaryocyte development was studied by semi-solid and suspension cultures and assessed with megakaryocyte colony forming unit (CFU-MK), acetylcholinesterase activity (AChE) as well as megakaryocyte count, diameter and DNA content. RESULTS: IFN-alpha prohibited CFU-MK formation and decreased AChE of megakaryocytes. The inhibitory effects were positively related to doses of IFN-alpha and could not be overcome by escalating exogenous Tpo or depriving accessory cells in bone marrow culture. IFN-alpha affected all aspects of megakaryocyte development, including decreases in megakaryocyte count, diameter and geometric mean polyploidy. CONCLUSION: IFN-alpha directly inhibits Tpo-induced megakaryocyte proliferation and differentiation in a dose-dependent manner.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Interferon-alfa/farmacologia , Megacariócitos/citologia , Trombopoetina/farmacologia , Animais , Células Cultivadas , Feminino , Megacariócitos/efeitos dos fármacos , Camundongos
19.
J Invest Dermatol ; 115(6): 1134-43, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11121152

RESUMO

Keratinocytes of patients with atopic dermatitis produce high amounts of granulocyte/macrophage colony-stimulating factor, a factor essential for dendritic cell function and thus for the development of skin immune responses. In contrast to keratinocytes cultured from nonatopic, healthy individuals, granulocyte/macrophage colony-stimulating factor mRNA could be detected in unstimulated cultures of atopic dermatitis keratinocytes, and phorbol myristate acetate induced much greater granulocyte/macrophage colony-stimulating factor mRNA levels in these cells, although the decay kinetics were not altered. Using reporter gene (chloramphenicol acetyl transferase) analysis, a minimal granulocyte/macrophage colony-stimulating factor promoter was shown to confer constitutive and phorbol-myristate-acetate-induced regulation of transcriptional activity in keratinocytes, and significantly higher levels of chloramphenicol acetyl transferase activity were measured in lysates of unstimulated and phorbol-myristate-acetate-treated atopic dermatitis keratinocytes than in control keratinocyte cultures. Electrophoretic mobility shift assays showed that low levels of NF-kappa B binding activity could be induced by phorbol myristate acetate in both normal and atopic dermatitis keratinocytes. By contrast, activator protein 1 complexes were efficiently induced, and they were invariably present at higher levels in nuclear lysates of atopic dermatitis keratinocytes. Atopic dermatitis keratinocyte nuclear lysates had higher constitutive levels of c-Jun, and phorbol myristate acetate promoted an earlier and stronger expression of c-Jun, JunB, and of the phosphorylated forms of c-Fos. A dysregulated activation of activator protein 1 may be implicated in the molecular mechanisms leading to increased granulocyte/macrophage colony-stimulating factor expression in atopic dermatitis keratinocytes. J Invest Dermatol 115:1134-1143 2000


Assuntos
Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/biossíntese , Queratinócitos/química , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/fisiologia , Adolescente , Adulto , Eletroforese em Gel de Poliacrilamida/métodos , Feminino , Corantes Fluorescentes , Expressão Gênica , Genes Reporter , Genes fos , Genes jun/efeitos dos fármacos , Humanos , Queratinócitos/metabolismo , Masculino , Acetato de Tetradecanoilforbol/farmacologia
20.
Nature ; 408(6808): 57-63, 2000 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11081504

RESUMO

Cytokines are important in the regulation of haematopoiesis and immune responses, and can influence lymphocyte development. Here we have identified a class I cytokine receptor that is selectively expressed in lymphoid tissues and is capable of signal transduction. The full-length receptor was expressed in BaF3 cells, which created a functional assay for ligand detection and cloning. Conditioned media from activated human CD3+ T cells supported proliferation of the assay cell line. We constructed a complementary DNA expression library from activated human CD3+ T cells, and identified a cytokine with a four-helix-bundle structure using functional cloning. This cytokine is most closely related to IL2 and IL15, and has been designated IL21 with the receptor designated IL21 R. In vitro assays suggest that IL21 has a role in the proliferation and maturation of natural killer (NK) cell populations from bone marrow, in the proliferation of mature B-cell populations co-stimulated with anti-CD40, and in the proliferation of T cells co-stimulated with anti-CD3.


Assuntos
Linfócitos B/imunologia , Interleucinas/fisiologia , Células Matadoras Naturais/imunologia , Receptores de Interleucina/fisiologia , Linfócitos T/imunologia , Sequência de Aminoácidos , Animais , Células da Medula Óssea , Antígenos CD40/metabolismo , Linhagem Celular , Clonagem Molecular , Etiquetas de Sequências Expressas , Humanos , Subunidade alfa de Receptor de Interleucina-21 , Interleucinas/genética , Interleucinas/isolamento & purificação , Leucopoese , Ligantes , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Conformação Proteica , Receptores de Interleucina/genética , Receptores de Interleucina/isolamento & purificação , Receptores de Interleucina-21 , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...