Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Neurooncol ; 140(3): 497-507, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30132163

RESUMO

INTRODUCTION: Emerging evidence suggests that effective treatment of glioblastoma (GBM), the most common and deadly form of adult primary brain cancer, will likely require concurrent treatment of multiple aspects of tumor pathobiology to overcome tumor heterogeneity and the complex tumor-supporting microenvironment. Recent studies in non-central nervous system (CNS) tumor cells have demonstrated that oxaliplatin (OXA) can induce multi-faceted anti-tumor effects, in particular at drug concentrations below those required to induce apoptosis. These findings motivated re-investigation of OXA for the treatment of GBM. METHODS: The effects of OXA on murine KR158 and GL261 glioma cells including cell growth, cell death, inhibition of signal transducer and activator of transcription (STAT) activity, O-6-methylguanine-DNA methyltransferase (MGMT) expression, and immunogenic cell death (ICD) initiation, were evaluated by cytotoxicity assays, Western blot analysis, STAT3-luciferase reporter assays, qRT-PCR assays, and flow cytometry. Chemical inhibitors of endoplasmic reticulum (ER) stress were used to investigate the contribution of this cell damage response to the observed OXA effects. The effect of OXA on bone marrow-derived macrophages (BMDM) exposed to glioma conditioned media (GCM) was also analyzed by Western blot analysis. RESULTS: We identified the OXA concentration threshold for induction of apoptosis and from this determined the drug dose and treatment period for sub-cytotoxic treatments of glioma cells. Under these experimental conditions, OXA reduced STAT3 activity, reduced MGMT levels and increased temozolomide sensitivity. In addition, there was evidence of immunogenic cell death (elevated EIF2α phosphorylation and calreticulin exposure) following prolonged OXA treatment. Notably, inhibition of ER stress reversed the OXA-mediated inhibition of STAT3 activity and MGMT expression in the tumor cells. In BMDMs exposed to GCM, OXA also reduced levels of phosphorylated STAT3 and decreased expression of Arginase 1, an enzyme known to contribute to pro-tumor functions in the tumor-immune environment. CONCLUSIONS: OXA can induce notable multi-faceted biological effects in glioma cells and BMDMs at relatively low drug concentrations. These findings may have significant therapeutic relevance against GBM and warrant further investigation.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Macrófagos/metabolismo , Oxaliplatina/farmacologia , Animais , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático , Glioma/tratamento farmacológico , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Fator de Transcrição STAT3/metabolismo , Temozolomida
2.
Sci Signal ; 9(433): ra63, 2016 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-27330190

RESUMO

Insulin receptor substrate 2 (IRS2) is an adaptor protein that becomes tyrosine-phosphorylated in response to the cytokines interleukin-4 (IL-4) and IL-13, which results in activation of the phosphoinositide 3-kinase (PI3K)-Akt pathway. IL-4 and IL-13 contribute to allergic lung inflammation. To examine the role of IRS2 in allergic disease, we evaluated the responses of IRS2-deficient (IRS2(-/-)) mice. Unexpectedly, loss of IRS2 resulted in a substantial increase in the expression of a subset of genes associated with the generation of alternatively activated macrophages (AAMs) in response to IL-4 or IL-13 in vitro. AAMs secrete factors that enhance allergic responses and promote airway remodeling. Moreover, compared to IRS2(+/+) mice, IRS2(+/-) and IRS2(-/-) mice developed enhanced pulmonary inflammation, accumulated eosinophils and AAMs, and exhibited airway and vascular remodeling upon allergen stimulation, responses that partially depended on macrophage-intrinsic IRS2 signaling. Both in unstimulated and IL-4-stimulated macrophages, lack of IRS2 enhanced phosphorylation of Akt and ribosomal S6 protein. Thus, we identified a critical inhibitory loop downstream of IRS2, demonstrating an unanticipated and previously unrecognized role for IRS2 in suppressing allergic lung inflammation and remodeling.


Assuntos
Asma/imunologia , Proteínas Substratos do Receptor de Insulina/imunologia , Pulmão/imunologia , Ativação de Macrófagos , Macrófagos/imunologia , Remodelação das Vias Aéreas/efeitos dos fármacos , Remodelação das Vias Aéreas/genética , Remodelação das Vias Aéreas/imunologia , Animais , Asma/induzido quimicamente , Asma/genética , Asma/patologia , Proteínas Substratos do Receptor de Insulina/genética , Pulmão/patologia , Macrófagos/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/imunologia
3.
J Immunol ; 187(4): 1797-806, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21734075

RESUMO

NF-κB activation is essential for receptor activator for NF-κB ligand (RANKL)-induced osteoclast formation. IL-4 is known to inhibit the RANKL-induced osteoclast differentiation while at the same time promoting macrophage fusion to form multinucleated giant cells (MNG). Several groups have proposed that IL-4 inhibition of osteoclastogenesis is mediated by suppressing the RANKL-induced activation of NF-κB. However, we found that IL-4 did not block proximal, canonical NF-κB signaling. Instead, we found that IL-4 inhibited alternative NF-κB signaling and induced p105/50 expression. Interestingly, in nfκb1(-/-) bone marrow-derived macrophages (BMM), the formation of both multinucleated osteoclast and MNG induced by RANKL or IL-4, respectively, was impaired. This suggests that NF-κB signaling also plays an important role in IL-4-induced macrophage fusion. Indeed, we found that the RANKL-induced and IL-4-induced macrophage fusion were both inhibited by the NF-κB inhibitors IκB kinase 2 inhibitor and NF-κB essential modulator inhibitory peptide. Furthermore, overexpression of p50, p65, p52, and RelB individually in nfκb1(-/-) or nfκb1(+/+) BMM enhanced both giant osteoclast and MNG formation. Interestingly, knockdown of nfκb2 in wild-type BMM dramatically enhanced both osteoclast and MNG formation. In addition, both RANKL- and IL-4-induced macrophage fusion were impaired in NF-κB-inducing kinase(-/-) BMM. These results suggest IL-4 influences NF-κB pathways by increasing p105/p50 and suppressing RANKL-induced p52 translocation and that NF-κB pathways participate in both RANKL- and IL-4-induced giant cell formation.


Assuntos
Células da Medula Óssea/imunologia , Células Gigantes/imunologia , Interleucina-4/imunologia , NF-kappa B/imunologia , Osteoclastos/imunologia , Ligante RANK/imunologia , Animais , Fusão Celular , Células Cultivadas , Interleucina-4/genética , Camundongos , Camundongos Knockout , NF-kappa B/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Ligante RANK/genética , Quinase Induzida por NF-kappaB
4.
Sci Signal ; 1(51): ra17, 2008 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-19109239

RESUMO

Although interleukin-4 (IL-4) and IL-13 participate in allergic inflammation and share a receptor subunit (IL-4Ralpha), they have different functions. We compared cells expressing type I and II IL-4Rs with cells expressing only type II receptors for their responsiveness to these cytokines. IL-4 induced highly efficient, gammaC-dependent tyrosine phosphorylation of insulin receptor substrate 2 (IRS-2), whereas IL-13 was less effective, even when phosphorylation of signal transducer and activator of transcription 6 (STAT6) was maximal. Only type I receptor, gammaC-dependent signaling induced efficient association of IRS-2 with the p85 subunit of phosphoinositide 3-kinase or the adaptor protein growth factor receptor-bound protein 2. In addition, IL-4 signaling through type I IL-4Rs induced more robust expression of a subset of genes associated with alternatively activated macrophages than did IL-13. Thus, IL-4 activates signaling pathways through type I IL-4Rs qualitatively differently from IL-13, which cooperate to induce optimal gene expression.


Assuntos
Proteínas Substratos do Receptor de Insulina/metabolismo , Interleucina-13/fisiologia , Macrófagos/metabolismo , Receptores Tipo I de Interleucina-4/fisiologia , Ativação Transcricional , Linhagem Celular , Humanos , Fosforilação , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais
5.
Cell ; 132(2): 259-72, 2008 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-18243101

RESUMO

Interleukin-4 and Interleukin-13 are cytokines critical to the development of T cell-mediated humoral immune responses, which are associated with allergy and asthma, and exert their actions through three different combinations of shared receptors. Here we present the crystal structures of the complete set of type I (IL-4R alpha/gamma(c)/IL-4) and type II (IL-4R alpha/IL-13R alpha1/IL-4, IL-4R alpha/IL-13R alpha1/IL-13) ternary signaling complexes. The type I complex reveals a structural basis for gamma(c)'s ability to recognize six different gamma(c)-cytokines. The two type II complexes utilize an unusual top-mounted Ig-like domain on IL-13R alpha1 for a novel mode of cytokine engagement that contributes to a reversal in the IL-4 versus IL-13 ternary complex assembly sequences, which are mediated through substantially different recognition chemistries. We also show that the type II receptor heterodimer signals with different potencies in response to IL-4 versus IL-13 and suggest that the extracellular cytokine-receptor interactions are modulating intracellular membrane-proximal signaling events.


Assuntos
Interleucina-13/metabolismo , Interleucina-4/metabolismo , Receptores de Citocinas/metabolismo , Receptores de Interleucina-13/metabolismo , Receptores de Interleucina-4/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Cristalografia por Raios X , Dimerização , Relação Dose-Resposta a Droga , Histidina/metabolismo , Humanos , Ligação de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Interleucina-13/genética , Interleucina-13/isolamento & purificação , Interleucina-13/farmacologia , Interleucina-4/genética , Interleucina-4/isolamento & purificação , Interleucina-4/farmacologia , Cinética , Ligantes , Modelos Moleculares , Mimetismo Molecular , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de Citocinas/química , Receptores de Interleucina-13/química , Receptores de Interleucina-4/química , Proteínas Recombinantes/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT6/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Termodinâmica , Tirosina/metabolismo , Difração de Raios X
6.
Cell Res ; 17(11): 942-55, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17968425

RESUMO

Interleukin-4 (IL-4) promotes lymphocyte survival and protects primary lymphomas from apoptosis. Previous studies reported differential requirements for the signal transducer and activator of transcription 6 (STAT6) and IRS2/phosphatidylinositol 3 kinase (PI-3K) signaling pathways in mediating the IL-4-induced protection from Fas-mediated apoptosis. In this study, we characterized IL-4-activated signals that suppress anti-IgM-mediated apoptosis and growth arrest of CH31, a model B-cell lymphoma line. In CH31, anti-IgM treatment leads to the loss of mitochondrial membrane potential, phospho-Akt, phospho-CDK2, and c-myc protein. These losses are followed by massive induction of p27(Kip1) protein expression, cell cycle arrest, and apoptosis. Strikingly, IL-4 treatment prevented or reversed these changes. Furthermore, IL-4 suppressed the activation of caspases 9 and 3, and, in contrast to previous reports, induced the phosphorylation (deactivation) of BAD. IL-4 treatment also induced expression of BclxL, a STAT6-dependent gene. Pharmacologic inhibitors and dominant inhibitory forms of PI-3K and Akt abrogated the anti-apoptotic function of IL-4. These results suggest that the IL-4 receptor activates several signaling pathways, with the Akt pathway playing a major role in suppression of the apoptotic program activated by anti-IgM.


Assuntos
Apoptose/efeitos dos fármacos , Imunoglobulina M/metabolismo , Interleucina-4/farmacologia , Linfoma de Células B/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Anticorpos/farmacologia , Linfócitos B/enzimologia , Caspase 3/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27 , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc , Receptores de Interleucina-4/agonistas , Receptores de Interleucina-4/metabolismo , Fator de Transcrição STAT6/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo , Proteína bcl-X/metabolismo
7.
J Immunol ; 168(12): 6215-23, 2002 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-12055235

RESUMO

The insulin receptor substrate (IRS) family of signaling molecules is expressed in lymphocytes, although their functions in these cells is largely unknown. To investigate the role of IRS in the protection of T cells from activation-induced cell death (AICD), we transfected the T cell hybridoma A1.1, which is IL-4 responsive but lacks expression of IRS family members with cDNA encoding IRS1 or IRS2. Stimulation of these clones with immobilized anti-CD3-induced expression of CD69 to the same level as the parental A1.1 cells. However, the A1.1 IRS1-expressing cells were markedly resistant to AICD, while the A1.1 IRS2-expressing cells were not. Inhibition of phosphatidylinositol 3'-kinase in the A1.1 IRS1-expressing cells did not abrogate their resistance to AICD. Fas mRNA was induced similarly by anti-CD3 in A1.1, A1.1 IRS1-expressing, and A1.1 IRS2-expressing cells. However, induction of Fas ligand (FasL) mRNA and functional FasL protein was delayed and decreased in IRS1-expressing cells, but not in IRS2-expressing cells. The induction of transcription from a 500-bp FasL promoter and a minimal 16-mer early growth response element linked to luciferase was also impaired in the IRS1-expressing cells. These results suggest that overexpression of IRS1, but not IRS2, protects A1.1 cells from AICD by diminishing FasL transcription through a pathway that is independent of the tyrosine phosphorylation of IRS1 and phosphatidylinositol 3'-kinase activity.


Assuntos
Hibridomas/imunologia , Ativação Linfocitária , Fosfoproteínas/biossíntese , Receptor de Insulina/metabolismo , Linfócitos T/imunologia , Animais , Morte Celular/genética , Morte Celular/imunologia , Proteína Ligante Fas , Hibridomas/citologia , Hibridomas/metabolismo , Proteínas Substratos do Receptor de Insulina , Peptídeos e Proteínas de Sinalização Intracelular , Leucemia L1210 , Ativação Linfocitária/genética , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Camundongos , Fosfatidilinositol 3-Quinases/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Receptores de Antígenos de Linfócitos T/fisiologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/citologia , Linfócitos T/metabolismo , Fatores de Transcrição/metabolismo , Transfecção , Receptor fas/biossíntese , Receptor fas/genética , Receptor fas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA