Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Immunol ; 191(9): 4505-13, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24068671

RESUMO

We previously demonstrated that TGF-ß1 suppresses IgE-mediated signaling in human and mouse mast cells in vitro, an effect that correlated with decreased expression of the high-affinity IgE receptor, FcεRI. The in vivo effects of TGF-ß1 and the means by which it suppresses mast cells have been less clear. This study shows that TGF-ß1 suppresses FcεRI and c-Kit expression in vivo. By examining changes in cytokine production concurrent with FcεRI expression, we found that TGF-ß1 suppresses TNF production independent of FcεRI levels. Rather, IgE-mediated signaling was altered. TGF-ß1 significantly reduced expression of Fyn and Stat5, proteins critical for cytokine induction. These changes may partly explain the effects of TGF-ß1, because Stat5B overexpression blocked TGF-mediated suppression of IgE-induced cytokine production. We also found that Stat5B is required for mast cell migration toward stem cell factor, and that TGF-ß1 reduced this migration. We found evidence that genetic background may alter TGF responses. TGF-ß1 greatly reduced mast cell numbers in Th1-prone C57BL/6, but not Th2-prone 129/Sv mice. Furthermore, TGF-ß1 did not suppress IgE-induced cytokine release and did increase c-Kit-mediated migration in 129/Sv mast cells. These data correlated with high basal Fyn and Stat5 expression in 129/Sv cells, which was not reduced by TGF-ß1 treatment. Finally, primary human mast cell populations also showed variable sensitivity to TGF-ß1-mediated changes in Stat5 and IgE-mediated IL-6 secretion. We propose that TGF-ß1 regulates mast cell homeostasis, and that this feedback suppression may be dependent on genetic context, predisposing some individuals to atopic disease.


Assuntos
Imunoglobulina E/imunologia , Mastócitos/metabolismo , Receptores de IgE/imunologia , Fator de Transcrição STAT5/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Movimento Celular/imunologia , Células Cultivadas , Citocinas/metabolismo , Humanos , Imunoglobulina E/metabolismo , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Interferência de RNA , RNA Interferente Pequeno , Receptores de IgE/biossíntese , Receptores de IgE/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/imunologia , Transdução de Sinais/imunologia , Fator de Crescimento Transformador beta1/imunologia , Fatores de Necrose Tumoral/biossíntese
2.
Blood ; 115(7): 1416-24, 2010 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-20008792

RESUMO

Phosphorylated signal transducer and activator of transcription 5 (STAT5) is a biomarker and potential molecular target for hematologic malignancies. We have shown previously that lethal myeloproliferative disease (MPD) in mice mediated by persistently activated STAT5 (STAT5a(S711F)) requires the N-domain, but the mechanism was not defined. We now demonstrate by retrovirally complementing STAT5ab(null/null) primary mast cells that relative to wild-type STAT5a, STAT5a lacking the N-domain (STAT5aDeltaN) ineffectively protected against cytokine withdrawal-induced cell death. Both STAT5a and STAT5aDeltaN bound to a site in the bcl-2 gene and both bound near the microRNA 15b/16 cluster. However, only STAT5a could effectively induce bcl-2 mRNA and reciprocally suppress miR15b/16 leading to maintained bcl-2 protein levels. After retroviral complementation of STAT5ab(null/null) fetal liver cells and transplantation, persistently active STAT5a(S711F) lacking the N-domain (STAT5aDeltaN(S711F)) was insufficient to protect c-Kit(+)Lin(-)Sca-1(+) (KLS) cells from apoptosis and unable to induce bcl-2 expression, whereas STAT5a(S711F) caused robust KLS cell expansion, induction of bcl-2, and lethal MPD. Severe attenuation of MPD by STAT5aDeltaN(S711F) was reversed by H2k/bcl-2 transgenic expression. Overall, these studies define N-domain-dependent survival signaling as an Achilles heel of persistent STAT5 activation and highlight the potential therapeutic importance of targeting STAT5 N-domain-mediated regulation of bcl-2 family members.


Assuntos
MicroRNAs/metabolismo , Transtornos Mieloproliferativos/fisiopatologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Fator de Transcrição STAT5/genética , Animais , Sobrevivência Celular/fisiologia , Regulação Neoplásica da Expressão Gênica , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Íntrons/fisiologia , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Morfinanos/metabolismo , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Estrutura Terciária de Proteína , Processamento Pós-Transcricional do RNA/fisiologia , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/fisiologia
3.
Blood ; 116(9): 1548-58, 2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20508164

RESUMO

Stat5 transcription factors are essential gene regulators promoting proliferation, survival, and differentiation of all hematopoietic cell types. Mutations or fusions of oncogenic tyrosine kinases often result in constitutive Stat5 activation. We have modeled persistent Stat5 activity by using an oncogenic Stat5a variant (cS5). To analyze the hitherto unrecognized role of Stat5 serine phosphorylation in this context, we have generated cS5 constructs with mutated C-terminal serines 725 and 779, either alone or in combination. Genetic complementation assays in primary Stat5(null/null) mast cells and Stat5(DeltaN) T cells demonstrated reconstitution of proliferation with these mutants. Similarly, an in vivo reconstitution experiment of transduced Stat5(null/null) fetal liver cells transplanted into irradiated wild-type recipients revealed that these mutants exhibit biologic activity in lineage differentiation. By contrast, the leukemogenic potential of cS5 in bone marrow transplants decreased dramatically in cS5 single-serine mutants or was completely absent upon loss of both serine phosphorylation sites. Our data suggest that Stat5a serine phosphorylation is a prerequisite for cS5-mediated leukemogenesis. Hence, interference with Stat5a serine phosphorylation might provide a new therapeutic option for leukemia and myeloid dysplasias without affecting major functions of Stat5 in normal hematopoiesis.


Assuntos
Transformação Celular Neoplásica , Hematopoese/fisiologia , Leucemia/patologia , Fator de Transcrição STAT5/metabolismo , Serina/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Adulto , Idoso , Animais , Western Blotting , Transplante de Medula Óssea , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Feminino , Feto , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Leucemia/genética , Leucemia/metabolismo , Transplante de Fígado , Masculino , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Fosforilação , Células Precursoras de Linfócitos B/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/genética , Serina/genética , Linfócitos T/metabolismo , Proteínas Supressoras de Tumor/genética
4.
Blood ; 113(20): 4856-65, 2009 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-19258595

RESUMO

Currently, there is a major need in hematopoietic stem cell (HSC) transplantation to develop reduced-intensity regimens that do not cause DNA damage and associated toxicities and that allow a wider range of patients to receive therapy. Cytokine receptor signals through c-Kit and c-Mpl can modulate HSC quiescence and engraftment, but the intracellular signals and transcription factors that mediate these effects during transplantation have not been defined. Here we show that loss of one allele of signal transducer and activator of transcription 5 (STAT5) in nonablated adult mutant mice permitted engraftment with wild-type HSC. Conditional deletion of STAT5 using Mx1-Cre caused maximal reduction in STAT5 mRNA (> 97%) and rapidly decreased quiescence-associated c-Mpl downstream targets (Tie-2, p57), increased HSC cycling, and gradually reduced survival and depleted the long-term HSC pool. Host deletion of STAT5 was persistent and permitted efficient donor long-term HSC engraftment in primary and secondary hosts in the absence of ablative conditioning. Overall, these studies establish proof of principle for targeting of STAT5 as novel transplantation conditioning and demonstrate, for the first time, that STAT5, a mitogenic factor in most cell types, including hematopoietic progenitors, is a key transcriptional regulator that maintains quiescence of HSC during steady-state hematopoiesis.


Assuntos
Células-Tronco Adultas/fisiologia , Proliferação de Células , Deleção de Genes , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/fisiologia , Fator de Transcrição STAT5/genética , Proteínas Adaptadoras de Transdução de Sinal , Células-Tronco Adultas/metabolismo , Animais , Dosagem de Genes/fisiologia , Hematopoese/genética , Mobilização de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/metabolismo , Tolerância Imunológica/genética , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoproteínas/genética , Condicionamento Pré-Transplante
5.
Exp Hematol ; 35(11): 1684-94, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17976521

RESUMO

OBJECTIVE: Signal transducer and activator of transcription 5 (STAT5) is a critical regulator of hematopoietic development and its impaired activation is associated with hematopoietic and immune cell defects. However, much of this information has been learned from knockout mice that still retain the potential for expression of STAT5 proteins that are N-terminally truncated due to alternative internal translation initiation codons. The goal of these studies was to use transplantation-based assays to analyze the degree of STAT5 deltaN activity in hematopoietic stem cells (HSC) and throughout lymphomyeloid development. METHODS: We have directly compared E14.5 fetal liver cells from mice with potential to express STAT5ab deltaN (STAT5ab(deltaN/deltaN)) with mice completely lacking STAT5a and STAT5b (STAT5abnull/null). We have also utilized retroviral complementation of STAT5abnull/null fetal liver HSC to enforce expression of full-length STAT5a or STAT5a lacking the first 136 amino acids (STAT5a deltaN). RESULTS: We report that STAT5 is required for HSC, lymphocyte, and erythrocyte development. We demonstrate that restored expression of STAT5a in STAT5abnull/null HSC provides a strong selective advantage, correcting T- and B-lymphocyte and erythrocyte development. Interestingly, Gr-1(+) blood cells were inversely correlated with B lymphocytes and both were normalized by STAT5a expression. In contrast, transduction of STAT5a deltaN only provided partial B-lymphocyte development. CONCLUSIONS: These studies define the role of STAT5 in maintaining normal lymphoid vs myeloid balance during hematopoiesis and highlight a major role for the N-domain in HSC function. The platform of retroviral complementation described here will be particularly useful for future studies to subdefine the N-domain regions that are critical for hematopoiesis.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/citologia , Fator de Transcrição STAT5/fisiologia , Animais , Linhagem da Célula , Células Cultivadas , Linfócitos/citologia , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Estrutura Terciária de Proteína , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Deleção de Sequência
6.
Oncotarget ; 6(42): 44360-72, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26517238

RESUMO

RNase L is a regulated endoribonuclease that functions in the interferon antiviral response. Activation of RNase L by 2', 5'-oligoadenylates has been linked to apoptosis, autophagy and inflammation. Genetic studies have also suggested the possible involvement of the RNase L gene (RNASEL) on chromosome 1q25.3 in several types of cancer. Here we report that ablation of RNase L in human prostate cancer PC3 cells by CRISPR/Cas9 gene editing technology enhanced cell migration as determined both by transwell assays and scratch wound healing assays. In addition, RNase L knockdown by means of RNAi increased migration of PC3 and DU145 cells in response to either fibronectin or serum stimulation, as did homozygous disruption of the RNase L gene in mouse embryonic fibroblasts. Serum or fibronectin stimulation of focal adhesion kinase (FAK) autophosphorylation on tyrosine-397 was increased by either knockdown or ablation of RNase L. In contrast, a missense mutant RNase L (R667A) lacking catalytic activity failed to suppress cell migration in PC3 cells. However, a nuclease-inactive mutant mouse RNase L (W630A) was able to partially inhibit migration of mouse fibroblasts. Consistent with a role for the catalytic activity of RNase L, transfection of PC3 cells with the RNase L activator, 2', 5'-oligoadenylate, suppressed cell migration. RNase L knockdown in PC3 cells enhanced tumor growth and metastasis following implantation in the mouse prostate. Our results suggest that naturally occurring mutations in the RNase L gene might promote enhanced cell migration and metastasis.


Assuntos
Movimento Celular , Endorribonucleases/metabolismo , Fibroblastos/enzimologia , Neoplasias da Próstata/enzimologia , Animais , Proteínas Associadas a CRISPR/genética , Proteínas Associadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Proliferação de Células , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Endorribonucleases/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Mutação de Sentido Incorreto , Invasividade Neoplásica , Metástase Neoplásica , Fosforilação , Neoplasias da Próstata/patologia , Interferência de RNA , Transdução de Sinais , Fatores de Tempo , Transfecção , Carga Tumoral
7.
Am J Blood Res ; 4(1): 20-6, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25232501

RESUMO

Signal transducer and activator of transcription 5 (STAT5) is a critical regulator of normal and leukemic lympho-myeloid development through activation downstream of early-acting cytokines, their receptors, and JAKs. Truncation of STAT5 can be mediated through alternative translation initiation from an internal start codon giving rise to N-terminally deleted isoforms. To determine whether these isoforms could be detected naturally in normal murine tissues, Western blot analyses were performed on heart, lung, brain, spleen, liver, and kidney. Relative expression of full-length to truncated STAT5 was variable among tissues. Since we have previously demonstrated that STAT5abΔN lacks the ability to effectively upregulate pro-survival signals and bcl-2 expression, we used a transgenic mouse approach to next determine whether constitutive expression of human Bcl-2 in STAT5ab(ΔN/ΔN) mouse hematopoietic cells could restore normal hematopoiesis. Transgenic H2K-Bcl-2 expression in hypomorphic STAT5ab(ΔN/ΔN) mice largely rescued peripheral B and T lymphocyte numbers whereas multilineage donor contribution was only rescued to levels about 10% of normal. At the hematopoietic stem cell level, direct competitive repopulation with H2K-Bcl-2/STAT5ab(ΔN/ΔN) against STAT5ab(ΔN/ΔN) competitor showed a corrective effect of Bcl-2 expression whether the STAT5ab(ΔN/ΔN) genotype was competed as the donor or as the host versus H2K-Bcl-2/STAT5ab(ΔN/ΔN) genotype bone marrow cells. Therefore, STAT5abΔN isoforms are heterogeneously expressed and lack key functional activities that can be partially rescued by adding back Bcl-2.

8.
Front Biosci (Landmark Ed) ; 16(8): 3043-56, 2011 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622220

RESUMO

Persistent tyrosine phosphorylation of Stat3 and Stat5 is associated with oncogenic activity. Phosphorylation of the conserved tyrosine residue (pTyr) was long believed to be the only essential prerequisite to promote activation and nuclear translocation of Stat proteins. It has become evident, however, that post-translational protein modifications like serine phosphorylation, acetylation, glycosylation as well as protein splicing and processing constitute further regulatory mechanisms to modulate Stat transcriptional activity and to provide an additional layer of specificity to Jak-Stat signal transduction. Significantly, most vertebrate Stat proteins contain one conserved serine phosphorylation site within their transactivation domains. This phosphorylation motif is located within a P(M)SP sequence. Stat transcription factor activity is negatively influenced by mutation of the serine to alanine. Moreover, it was shown for both Stat3 and Stat5 that their capacity to transform cells was diminished. This review addresses recent advances in understanding the regulation and the biochemical and biological consequences of Stat serine phosphorylation. In particular, we discuss their role in persistently activated Stat proteins for cancer research.


Assuntos
Transformação Celular Neoplásica/metabolismo , Fator de Transcrição STAT5/metabolismo , Sequência de Aminoácidos , Animais , Transformação Celular Neoplásica/genética , Sequência Conservada , Neoplasias Hematológicas/etiologia , Humanos , Dados de Sequência Molecular , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT5/química , Fator de Transcrição STAT5/genética , Homologia de Sequência de Aminoácidos , Serina/química , Transdução de Sinais
9.
PLoS One ; 5(2): e9152, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-20161778

RESUMO

BACKGROUND: Grb2-associated binding (Gab) adapter proteins play major roles in coordinating signaling downstream of hematopoietic cytokine receptors. In hematopoietic cells, Gab2 can modulate phosphatidylinositol-3 kinase and mitogen associated protein kinase activities and regulate the long-term multilineage competitive repopulating activity of hematopoietic stem cells (HSCs). Gab2 may also act in a linear pathway upstream or downstream of signal transducer and activator of transcription-5 (STAT5), a major positive regulator of HSC function. Therefore, we aimed to determine whether Gab2 and STAT5 function in hematopoiesis in a redundant or non-redundant manner. METHODOLOGY/PRINCIPAL FINDINGS: To do this we generated Gab2 mutant mice with heterozygous and homozygous deletions of STAT5. In heterozygous STAT5 mutant mice, deficiencies in HSC/multipotent progenitors were reflected by decreased long-term repopulating activity. This reduction in repopulation function was mirrored in the reduced growth response to early-acting cytokines from sorted double mutant c-Kit(+)Lin(-)Sca-1(+) (KLS) cells. Importantly, in non-ablated newborn mice, the host steady-state engraftment ability was impaired by loss of Gab2 in heterozygous STAT5 mutant background. Fetal liver cells isolated from homozygous STAT5 mutant mice lacking Gab2 showed significant reduction in HSC number (KLS CD150(+)CD48(-)), reduced HSC survival, and dramatic loss of self-renewal potential as measured by serial transplantation. CONCLUSIONS/SIGNIFICANCE: These data demonstrate new functions for Gab2 in hematopoiesis in a manner that is non-redundant with STAT5. Furthermore, important synergy between STAT5 and Gab2 was observed in HSC self-renewal, which might be exploited to optimize stem cell-based therapeutics.


Assuntos
Hematopoese , Células-Tronco Hematopoéticas/metabolismo , Fosfoproteínas/deficiência , Fator de Transcrição STAT5/deficiência , Proteínas Adaptadoras de Transdução de Sinal , Animais , Animais Recém-Nascidos , Linhagem da Célula , Proliferação de Células , Sobrevivência Celular , Ensaio de Unidades Formadoras de Colônias , Feminino , Citometria de Fluxo , Genótipo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Fígado/citologia , Fígado/embriologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfoproteínas/genética , Fator de Transcrição STAT5/genética
10.
Cancer Res ; 70(9): 3526-36, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20406969

RESUMO

The antiangiogenic drug sunitinib is a receptor tyrosine kinase inhibitor with significant, yet not curative, therapeutic effects in metastatic renal cell carcinoma (RCC). Sunitinib is also an immunomodulator, potently reversing myeloid-derived suppressor cell (MDSC) accumulation and T-cell inhibition in the blood even of nonresponder RCC patients. We observed that sunitinib similarly prevented MDSC accumulation and restored normal T-cell function to the spleens of tumor-bearing mice, independent of the capacity of sunitinib to inhibit tumor progression (RENCA>CT26>4T1). Both monocytic and neutrophilic splenic MDSC were highly repressible by sunitinib. In contrast, MDSC within the microenvironment of 4T1 tumors or human RCC tumors proved highly resistant to sunitinib and ambient T-cell function remained suppressed. Proteomic analyses comparing tumor to peripheral compartments showed that granulocyte macrophage colony-stimulating factor (GM-CSF) predicted sunitinib resistance and recombinant GM-CSF conferred sunitinib resistance to MDSC in vivo and in vitro. MDSC conditioning with GM-CSF uniquely inhibited signal transducers and activators of transcription (STAT3) and promoted STAT5 activation. STAT5ab(null/null) MDSC were rendered sensitive to sunitinib in the presence of GM-CSF in vitro. We conclude that compartment-dependent GM-CSF exposure in resistant tumors may account for the regionalized effect of sunitinib upon host MDSC modulation and hypothesize that ancillary strategies to decrease such regionalized escape will enhance the potency of sunitinib as an immunomodulator and a cancer therapy.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Carcinoma de Células Renais/imunologia , Fatores Imunológicos/farmacologia , Indóis/farmacologia , Neoplasias Renais/imunologia , Células Mieloides/efeitos dos fármacos , Pirróis/farmacologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Animais , Antígeno CD11b/imunologia , Linfócitos T CD8-Positivos/imunologia , Carcinoma de Células Renais/tratamento farmacológico , Feminino , Humanos , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/imunologia , Sunitinibe , Fatores Supressores Imunológicos/imunologia , Subpopulações de Linfócitos T/imunologia
11.
Exp Hematol ; 37(2): 276-84, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19059699

RESUMO

OBJECTIVE: The aim of this study was to identify and characterize the causative mutation in the thrombocytopenic mouse strain HLB219 that was generated at the Jackson Laboratory as part of a large-scale N-ethyl-N-nitrosourea mutagenesis screen. MATERIALS AND METHODS: The HLB219 mutation was identified by interval mapping of F2 mice generated from intercross breeding of HLB219 to both BALB/cByJ (BALB) and 129/SvImJ (129/Sv). Mpl was identified as a candidate gene and sequenced. The mutation was characterized in vivo in mouse hematopoietic stem/progenitor cell assays and in cell culture by expression in Ba/F3 cells. RESULTS: A novel mutation in the thrombopoietin (TPO) receptor Mpl in HLB219 mice caused a Cys-->Arg substitution at codon 40 in the extracellular region of the receptor. Mice homozygous for the Mpl(hlb219) mutation had an 80% decrease in the number of platelets in comparison to the wild-type C57BL/6J strain, low numbers of bone marrow megakaryocytes, high TPO levels, and decreased competitive repopulating ability, consistent with a loss-of-function mutation in the receptor. Mice heterozygous for Mpl(hlb219) however, showed an overdominance effect with a significant increase in platelet number. Functional analysis in vitro demonstrated that Ba/F3 cells expressing the mutant MPL(hlb219) protein failed to activate extracellular signal-regulated kinase and signal transducers and activators of transcription 5, but proliferated in the absence of TPO and required constitutive phosphorylation of RAC-alpha serine/threonine protein kinase (AKT) for cytokine-independent growth. CONCLUSION: Thrombocytopenia in HLB219 mice is caused by a recessive mutation in Mpl that abrogates mitogen-activated protein kinase-extracellular signal regulated kinase and janus kinase-signal transducers and activators of transcription signaling.


Assuntos
Substituição de Aminoácidos , Etilnitrosoureia/toxicidade , Genes Recessivos , Mutagênicos/toxicidade , Mutação de Sentido Incorreto , Receptores de Trombopoetina/genética , Trombocitopenia/genética , Animais , Cruzamento , Linhagem Celular , Feminino , Homozigoto , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Camundongos Endogâmicos BALB C , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Receptores de Trombopoetina/metabolismo , Trombocitopenia/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/genética
12.
J Immunol ; 179(2): 1068-79, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17617599

RESUMO

The two closely related Stat5 (Stat5A and Stat5B) proteins are activated by a broad spectrum of cytokines. However, with the complication of the involvement of Stat5A/5B in stem cell function, the role of Stat5A/5B in the development and function of lymphocytes, especially B cells, is not fully understood. In this study, we demonstrated that Stat5A/5B(-/-) fetal liver cells had severe diminution of B cell progenitors but clearly had myeloid progenitors. Consistently, the mutant fetal liver cells could give rise to hemopoietic progenitors and myeloid cells but not B cells beyond pro-B cell progenitors in lethally irradiated wild-type or Jak3(-/-) mice. Deletion of Stat5A/5B in vitro directly impaired IL-7-mediated B cell expansion. Of note, reintroduction of Stat5A back into Stat5A/5B(-/-) fetal liver cells restored their abilities to develop B cells. Importantly, CD19-Cre-mediated deletion of Stat5A/5B in the B cell compartment specifically impaired early B cell development but not late B cell maturation. Moreover, the B cell-specific deletion of Stat5A/5B did not impair splenic B cell survival, proliferation, and Ig production. Taken together, these data demonstrate that Stat5A/5B directly control IL-7-mediated early B cell development but are not required for B cell maturation and Ig production.


Assuntos
Linfócitos B/citologia , Linfócitos B/imunologia , Diferenciação Celular/imunologia , Interleucina-7/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Formação de Anticorpos/imunologia , Western Blotting , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Interleucina-7/imunologia , Fígado/citologia , Fígado/embriologia , Fígado/imunologia , Camundongos , Camundongos Mutantes , Células Progenitoras Mieloides/citologia , Células Progenitoras Mieloides/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT5/imunologia
13.
Blood ; 110(1): 116-24, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17374739

RESUMO

Gab2 is an important adapter molecule for cytokine signaling. Despite its major role in signaling by receptors associated with hematopoiesis, the role of Gab2 in hematopoiesis has not been addressed. We report that despite normal numbers of peripheral blood cells, bone marrow cells, and c-Kit(+)Lin(-)Sca-1(+) (KLS) cells, Gab2-deficient hematopoietic cells are deficient in cytokine responsiveness. Significant reductions in the number of colony-forming units in culture (CFU-C) in the presence of limiting cytokine concentrations were observed, and these defects could be completely corrected by retroviral complementation. In earlier hematopoiesis, Gab2-deficient KLS cells isolated in vitro responded poorly to hematopoietic growth factors, resulting in an up to 11-fold reduction in response to a cocktail of stem cell factor, flt3 ligand, and thrombopoietin. Gab2-deficient c-Kit(+)Lin(-) cells also demonstrate impaired activation of extracellular signal-regulated kinase (ERK) and S6 in response to IL-3, which supports defects in activating the phosphatidylinositol-3 kinase (PI-3K) and mitogen-associated protein kinase (MAPK) signaling cascades. Associated with the early defects in cytokine response, competitive transplantation of Gab2(-/-) bone marrow cells resulted in defective long-term multilineage repopulation. Therefore, we demonstrate that Gab2 adapter function is intrinsically required for hematopoietic cell response to early-acting cytokines, resulting in defective hematopoiesis in Gab2-deficient mice.


Assuntos
Hematopoese , Fosfoproteínas/deficiência , Fosfoproteínas/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Transplante de Medula Óssea , Células Cultivadas , Citocinas/farmacologia , Células-Tronco Hematopoéticas/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Mutantes , Fosfoproteínas/genética , Células-Tronco/citologia
14.
J Immunol ; 177(5): 3421-6, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920984

RESUMO

The mast cell (MC) inflammatory response is now linked not only to atopy, but also to arthritis, multiple sclerosis, heart disease, and resistance to bacterial infection. In the current study, we demonstrate that the signal transducer and activator of transcription 5 (Stat5) is rapidly activated by IgE cross-linkage, and that its expression is critical to the MC response. Stat5-deficient (Stat5KO) MC demonstrated a significant decrease in IgE-mediated degranulation, leukotriene B4 production, cytokine secretion, and survival signals. The defect in cytokine production may be caused by decreased cytokine mRNA stability. Stat5KO MC-induced cytokine mRNAs normally following IgE cross-linkage, but these mRNAs were not sustained over time and were degraded at twice the rate observed in WT cells. Interestingly, the RNA destabilizing protein tristetraprolin was induced following IgE cross-linkage in Stat5KO but not wild-type cells. Moreover, reducing tristetraprolin expression via short hairpin RNA transfection significantly increased IL-13 production in Stat5KO MC. Our work demonstrates that Stat5 is a critical factor in IgE-induced MC activation, acting in part via posttranscriptional control of cytokine mRNA stability. These data have a direct impact on MC-associated inflammatory and autoimmune diseases.


Assuntos
Imunoglobulina E/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Fator de Transcrição STAT5/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Diferenciação Celular , Células Cultivadas , Citocinas/biossíntese , Citocinas/genética , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Leucotrienos/biossíntese , Mastócitos/citologia , Camundongos , Camundongos Knockout , Estabilidade de RNA/genética , Receptores de IgE/imunologia , Fator de Transcrição STAT5/deficiência , Fator de Transcrição STAT5/genética , Transdução de Sinais , Tristetraprolina/metabolismo
15.
J Biol Chem ; 279(2): 1123-31, 2004 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-14570908

RESUMO

Cellular stress responses induced during viral infections are critical to the health and survival of organisms. In higher vertebrates, interferons (IFNs) mediate the innate antiviral response in part through the action of RNase L, a uniquely regulated enzyme. RNase L is activated by 5'-phosphorylated, 2'-5' oligoadenylates (2-5A) produced from IFN-inducible and double stranded RNA-dependent synthetases. We show that viral activation of the c-Jun NH2-terminal kinases (JNK) family of MAP kinases and viral induction of apoptosis are both deficient in mouse cells lacking RNase L. Also, JNK phosphorylation in response to 2-5A was greatly reduced in RNase L-/- mouse cells. In addition, 2-5A treatment of the human ovarian carcinoma cell line, Hey1b, resulted in specific ribosomal RNA cleavage products coinciding with JNK activation. Furthermore, suppression of JNK activity with the chemical inhibitor, SP600125, prevented apoptosis induced by 2-5A. In contrast, inhibition of alternative MAP kinases, p38 and ERK, failed to prevent 2-5A-mediated apoptosis. Short interfering RNA to JNK1/JNK2 mRNAs resulted in JNK ablation while also suppressing 2-5A-mediated apoptosis. Moreover, Jnk1-/- Jnk2-/- cells were highly resistant to the apoptotic effects of IFN and 2-5A. These findings suggest that JNK and RNase L function in an integrated signaling pathway during the IFN response that leads to elimination of virus-infected cells through apoptosis.


Assuntos
Apoptose , Endorribonucleases/metabolismo , Interferons/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Antracenos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Sistema Livre de Células , Células Cultivadas , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Ativação Enzimática , Humanos , Marcação In Situ das Extremidades Cortadas , Proteínas Quinases JNK Ativadas por Mitógeno , Cinética , Sistema de Sinalização das MAP Quinases , Camundongos , Modelos Biológicos , Modelos Genéticos , Fosforilação , Ligação Proteica , RNA/metabolismo , RNA Interferente Pequeno/metabolismo , Ribossomos/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Vírus/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA