Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 11(9): e0162672, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27611322

RESUMO

BACKGROUND: Asthma is a chronic inflammatory disease of airways, but an ideal biomarker that accurately reflects ongoing airway inflammation has not yet been established. The aim of this study was to examine the potential of sputum leucine-rich alpha-2 glycoprotein (LRG) as a new biomarker for airway inflammation in asthma. METHODS: We obtained induced sputum samples from patients with asthma (N = 64) and healthy volunteers (N = 22) and measured LRG concentration by sandwich enzyme-linked immunosorbent assay (ELISA). Ovalbumin (OVA)-induced asthma model mice were used to investigate the mechanism of LRG production during airway inflammation. The LRG concentrations in the bronchoalveolar lavage fluid (BALF) obtained from mice were determined by ELISA and mouse lung sections were stained with anti-LRG antibody and periodic acid-Schiff (PAS) reagent. RESULTS: Sputum LRG concentrations were significantly higher in patients with asthma than in healthy volunteers (p = 0.00686). Consistent with patients' data, BALF LRG levels in asthma model mice were significantly higher than in control mice (p = 0.00013). Immunohistochemistry of lung sections from asthma model mice revealed that LRG was intensely expressed in a subpopulation of bronchial epithelial cells, which corresponded with PAS-positive mucus producing cells. CONCLUSION: These findings suggest that sputum LRG is a promising biomarker of local inflammation in asthma.


Assuntos
Asma/complicações , Asma/metabolismo , Glicoproteínas/metabolismo , Pneumonia/complicações , Pneumonia/metabolismo , Escarro/metabolismo , Animais , Asma/sangue , Asma/patologia , Biomarcadores/sangue , Brônquios/patologia , Líquido da Lavagem Broncoalveolar , Contagem de Células , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Glicoproteínas/sangue , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Pneumonia/sangue , Pneumonia/patologia , Regulação para Cima
2.
PLoS One ; 10(8): e0135836, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26284361

RESUMO

Human dynactin-associated protein (dynAP) is a transmembrane protein that promotes AktSer473 phosphorylation. Here, we report the oncogenic properties of dynAP. In contrast to control NIH3T3 cells expressing LacZ (NIH3T3LacZ), NIH3T3dynAP cells vigorously formed foci in two-dimensional culture, colonies on soft agar, and spheroids in anchorage-deficient three-dimensional culture. NIH3T3dynAP cells injected into nude mice produced tumors with abundant blood vessels and weak cell-cell contacts. Expression of dynAP elevated the level of rictor (an essential subunit of mTORC2) and promoted phosphorylation of FOXO3aSer253. FOXO3a is a transcriptional factor that stimulates expression of pro-apoptotic genes and phosphorylation of FOXO3a abrogates its function, resulting in promoted cell survival. Knockdown of rictor in NIH3T3dynAP cells reduced AktSer473 phosphorylation and formation of foci, colony in soft agar and spheroid, indicating that dynAP-induced activation of the mTORC2/AktSer473 pathway for cell survival contributes to cell transformation. E-cadherin and its mRNA were markedly reduced upon expression of dynAP, giving rise to cells with higher motility, which may be responsible for the weak cell-cell adhesion in tumors. Thus, dynAP could be a new oncoprotein and a target for cancer therapy.


Assuntos
Comunicação Celular , Transformação Celular Neoplásica , Proteínas Associadas aos Microtúbulos/genética , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/genética , Animais , Complexo Dinactina , Feminino , Humanos , Óperon Lac/genética , Camundongos , Células NIH 3T3 , Neoplasias/genética , Regulação para Cima
3.
Exp Eye Res ; 140: 53-64, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26297802

RESUMO

Recently, a number of biologics have been used in the treatment of autoimmune diseases. However, in the treatment of severe autoimmune uveitis, only TNF-alpha inhibitors are preferably used and the effect of other biologics such as interleukin-6 (IL-6) signaling blockade or cytotoxic T-lymphocyte antigen-4-immunoglobulin fusion protein (CTLA4-Ig) has not been well studied. Previously, we reported that IL-6 blockade effectively suppresses the development of experimental autoimmune uveitis (EAU), a mouse model for uveitis, by inhibiting Th17 cell development. In this study, we investigated the effect of CTLA4-Ig on EAU development and compared it with the effect of anti-IL-6 receptor monoclonal antibody (MR16-1). C57BL/6J mice were immunized with interphotoreceptor retinoid-binding protein (IRBP) and treated once with CTLA4-Ig or MR16-1. Both CTLA4-Ig and MR16-1 administered in the induction phase (the same day as immunization) significantly reduced the clinical and histopathological scores of EAU. Fluorescence-activated cell sorting studies using draining lymph node (LN) cells from EAU mice 10 days after immunization showed that CTLA4-Ig can suppress early T-helper cell activation. CTLA4-Ig administered in the effector phase of the disease (one week after immunization), when IRBP-reactive T cells have been primed, also significantly reduced the clinical and histopathological scores of EAU. In contrast, MR16-1 administered in the effector phase did not ameliorate EAU. To investigate the differences between these biologics in the effector phase, in vitro restimulation analysis of LN cells obtained from EAU mice one week after immunization was performed and revealed that CTLA4-Ig, but not MR16-1, added to culture media could inhibit the proliferation of IRBP-specific CD4(+) T cells which possessed capacities of producing IFN-gamma and/or IL-17. Collectively, CTLA4-Ig ameliorated EAU through preventing initial T-cell activation in the induction phase and suppressing proliferation of IRBP-specific T cells in the effector phase. Blockade of IL-6 signaling did not have such inhibitory effects after T-cell priming. CTLA4-Ig may have therapeutic effects on human chronic uveitis.


Assuntos
Abatacepte/farmacologia , Doenças Autoimunes/prevenção & controle , Modelos Animais de Doenças , Imunossupressores/farmacologia , Interleucina-6/antagonistas & inibidores , Uveíte/prevenção & controle , Animais , Anticorpos Bloqueadores/farmacologia , Doenças Autoimunes/induzido quimicamente , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Proteínas do Olho , Feminino , Citometria de Fluxo , Interferon gama/metabolismo , Interleucina-17/metabolismo , Linfonodos , Ativação Linfocitária/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fragmentos de Peptídeos , Receptores de Interleucina-6/imunologia , Proteínas de Ligação ao Retinol , Uveíte/induzido quimicamente , Uveíte/imunologia
4.
Oncotarget ; 6(13): 11009-22, 2015 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-25826092

RESUMO

Leucine-rich α2-glycoprotein (LRG) is an approximately 50-kDa glycoprotein that has been found to be elevated in the sera of patients with several types of cancer. LRG directly binds to transforming growth factor beta 1 (TGFß1) and modulates TGFß1 signaling in endothelial cells; however, the precise function of LRG in cancer remains unclear. This study aimed to investigate the role of LRG in cancer. Lewis lung carcinoma (LLC) cells hardly expressed LRG. The growth of LLC tumors allografted in the LRG knockout (KO) mice was significantly increased compared with wild-type (WT) mice. Conversely, overexpression of LRG significantly inhibited the growth of LLC tumors in WT mice. In the presence of LRG, TGFß1 significantly inhibited the proliferation of LLC cells and human hepatocellular carcinoma Hep3B cells in vitro by inducing apoptosis via the potent activation of smad2 and its downstream signaling pathway. Furthermore, administration of a TGFßR1 inhibitor (SB431542) significantly enhanced the growth of LLC tumors in WT mice compared with LRG KO mice via inhibition of apoptosis. We propose that LRG potentiates the effect of TGFß1 in cancer cells whose growth is suppressed in the presence of TGFß1.


Assuntos
Apoptose , Carcinoma Hepatocelular/prevenção & controle , Carcinoma Pulmonar de Lewis/prevenção & controle , Glicoproteínas/fisiologia , Neoplasias Hepáticas/prevenção & controle , Fator de Crescimento Transformador beta1/metabolismo , Animais , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Proliferação de Células , Feminino , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta1/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Pigment Cell Melanoma Res ; 27(4): 630-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24674392

RESUMO

Given no reliable therapy for advanced malignant melanoma, it is important to elucidate the molecular mechanisms underlying the disease progression. Using a quantitative proteomics approach, the 'isobaric tags for relative and absolute quantitation (iTRAQ)' method, we identified that the extracellular matrix protein, periostin (POSTN), was highly expressed in invasive melanoma compared with normal skin. An immunohistochemical analysis showed that POSTN was expressed in all invasive melanoma (n = 20) and metastatic lymph node (n = 5) tissue samples, notably restricted in their stroma. In terms of the intercellular regulation of POSTN, we found that there was upregulation of POSTN when melanoma cells and normal human dermal fibroblasts (NHDFs) were cocultured, with restricted expression of TGF-ß1 and TGF-ß3. In a functional analyses, recombinant and NHDF-derived POSTN significantly accelerated melanoma cell proliferation via the integrin/mitogen-activated protein kinase (MAPK) signaling pathway in vitro. The size of implanted melanoma tumors was significantly suppressed in POSTN/Rag2 double knockout mice compared with Rag2 knock-out mice. These results indicate that NHDF-derived POSTN accelerates melanoma progression and might be a promising therapeutic target for malignant melanoma.


Assuntos
Moléculas de Adesão Celular/metabolismo , Sistema de Sinalização das MAP Quinases , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Microambiente Tumoral , Animais , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Masculino , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Transplante de Neoplasias
6.
Cancer Sci ; 104(11): 1483-91, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23962256

RESUMO

Lung cancer (LC) is the major cause of death by cancer and the number of LC patients is increasing worldwide. This study investigated the therapeutic potential of gene delivery using suppressor of cytokine signaling 1 (SOCS-1), an endogenous inhibitor of intracellular signaling pathways, for the treatment of LC. To examine the antitumor effect of SOCS-1 overexpression on non-small-cell lung cancer (NSCLC) cells, NSCLC cells (A549, LU65, and PC9) were infected with adenovirus-expressing SOCS-1 vector. The cell proliferation assay showed that A549 and LU65, but not PC9, were sensitive to SOCS-1 gene-mediated suppression of cell growth. Although JAK inhibitor I could also inhibit proliferation of A549 and LU65 cells, SOCS-1 gene delivery appeared to be more potent as SOCS-1 could suppress focal adhesion kinase and epidermal growth factor receptor, as well as the JAK/STAT3 signaling pathway. Enhanced phosphorylation of the p53 protein was detected by means of phospho-kinase array in SOCS-1 overexpressed A549 cells compared with control cells, whereas no phosphorylation of p53 was observed when JAK inhibitor I was used. Furthermore, treatment with adenoviral vector AdSOCS-1 in vivo significantly suppressed NSCLC proliferation in a xenograft model. These results suggest that the overexpression of SOCS-1 gene is effective for antitumor therapy by suppressing the JAK/STAT, focal adhesion kinase, and epidermal growth factor receptor signaling pathways and enhancing p53-mediated antitumor activity in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/terapia , Neoplasias Pulmonares/terapia , Proteínas Supressoras da Sinalização de Citocina/genética , Adenoviridae/genética , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Terapia Genética , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Transplante de Neoplasias , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Transdução Genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
7.
Int J Cancer ; 132(2): 459-71, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22532243

RESUMO

Malignant pleural mesothelioma (MPM) is an aggressive tumor with poor prognosis for which an effective therapy remains to be established. This study investigated the therapeutic potential of gene delivery using suppressor of cytokine signaling 1 (SOCS-1), an endogenous inhibitor of intracellular signaling pathways, for the treatment of MPM. We infected MPM cells (MESO-4, H28 and H226) with adenovirus-expressing SOCS-1 vector to examine the effect of SOCS-1 overexpression on MPM cells. We evaluated the antitumor effect of SOCS-1 gene delivery combined with cisplatin plus pemetrexed by cell proliferation, apoptosis and invasion assay. We also investigated the regulation of NF-κB and STAT3 signaling related to apoptotic pathways. Furthermore, we evaluated the inhibition of tumor growth by SOCS-1 gene delivery combined with cisplatin plus pemetrexed in vivo. SOCS-1 gene delivery cooperated with cisplatin plus pemetrexed to inhibit cell proliferation, invasiveness and induction of apoptosis in MPM cells. SOCS-1 regulated NF-κB and STAT3 signaling to induce apoptosis in MESO-4 and H226 cells. Furthermore, SOCS-1 gene delivery cooperated with cisplatin plus pemetrexed to regulate NF-κB signaling and significantly inhibit tumor growth of MPM in vivo. These results suggest that SOCS-1 gene delivery has a potent antitumor effect against MPM and a potential for clinical use in combination with cisplatin plus pemetrexed.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Mesotelioma/terapia , Neoplasias Pleurais/terapia , Proteínas Supressoras da Sinalização de Citocina/genética , Adenoviridae/genética , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Terapia Combinada , Ativação Enzimática , Feminino , Expressão Gênica , Terapia Genética , Vetores Genéticos , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Guanina/análogos & derivados , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Interferon gama/farmacologia , Interferon gama/fisiologia , Mesotelioma/patologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , NF-kappa B/metabolismo , Invasividade Neoplásica , Pemetrexede , Neoplasias Pleurais/patologia , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
PLoS One ; 7(11): e48933, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23152823

RESUMO

Embryo implantation is a highly orchestrated process that involves blastocyst-uterine interactions. This process is confined to a defined interval during gestation referred to as the "window of embryo implantation receptivity". In mice this receptive period is controlled by ovarian estrogen and involves a coordination of blastocyst adhesion competence and uterine receptivity. Mechanisms coordinating the acquisition of blastocyst adhesion competence and uterine receptivity are largely unknown. Here, we show that ovarian estrogen indirectly regulates blastocyst adhesion competence. Acquisition of blastocyst adhesion competence was attributed to integrin activation (e.g. formation of adhesion complexes) rather than de novo integrin synthesis. Osteopontin (OPN) was identified as an estrogen-dependent uterine endometrial gland secretory factor responsible for activating blastocyst adhesion competence. Increased adhesion complex assembly in OPN-treated blastocysts was mediated through focal adhesion kinase (FAK)- and phosphatidylinositol 3-kinase (PI3K)-dependent signaling pathways. These findings define for the first time specific regulatory components of an estrogen-dependent pathway coordinating blastocyst adhesion competence and uterine receptivity.


Assuntos
Blastocisto/metabolismo , Estrogênios/metabolismo , Osteopontina/metabolismo , Útero/metabolismo , Animais , Blastocisto/citologia , Adesão Celular , Decídua/fisiologia , Feminino , Fibronectinas/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Humanos , Integrinas/metabolismo , Camundongos , Osteopontina/genética , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
9.
Invest Ophthalmol Vis Sci ; 52(6): 3264-71, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21330657

RESUMO

PURPOSE. Both Th17 and Th1 cells contribute to experimental autoimmune uveoretinitis (EAU). Interleukin-6 (IL-6) blockade inhibits Th17 differentiation in EAU and potently suppresses ocular inflammation, although its effect on Th1 cells is unknown. To clarify the mechanism of IL-6 blockade, the authors investigated T helper cells with particular focus on Th1 and regulatory T cells (Treg) in EAU of IL-6 gene knockout (KO) mice. METHODS. EAU was induced in wild-type (WT) mice and in mice lacking IL-6 (IL-6KO), IL-17 (IL-17KO), and IFN-γ (GKO) on a C57BL/6 background. Clinical scores of EAU, cytokine levels in supernatants from ocular tissue homogenates, and T helper cell differentiation in lymph nodes in each mouse were examined. To study the roles of Treg cells, EAU was induced in IL-6KO mice treated with anti-CD25 monoclonal antibody (mAb) to deplete Treg cells in vivo. RESULTS. Inflammation was comparable between WT, IL-17KO, and GKO mice but was absent in IL-6KO mice. Th17 and interphotoreceptor retinoid binding protein (IRBP)-specific Th1 cells were increased in GKO and IL-17KO mice, respectively, whereas both populations were reduced in IL-6KO mice. Th1-dominant EAU in IL-17KO mice was suppressed by anti-IL-6R mAb treatment. Treg cell depletion in vivo induced EAU in IL-6KO mice. CONCLUSIONS. After the induction of EAU, IL-6 deficiency resulted in the inhibition of the IRBP-specific Th1 response and enhanced the generation of IRBP-specific Treg cells. Furthermore, Treg was needed to inhibit Th1 responses and ocular inflammation in IL-6KO mice. Protective effects of IL-6 signaling blockade in EAU involve not only Th17 cell inhibition but also IRBP-specific Treg cell promotion.


Assuntos
Doenças Autoimunes/prevenção & controle , Proteínas do Olho/imunologia , Interleucina-6/fisiologia , Retinite/prevenção & controle , Proteínas de Ligação ao Retinol/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Células Th17/imunologia , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Retinite/imunologia , Transdução de Sinais/fisiologia , Uveíte/imunologia
10.
Int J Cancer ; 129(4): 1005-17, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20949562

RESUMO

Malignant pleural mesothelioma (MPM) is an aggressive tumor with poor prognosis for which an effective therapy remains to be established. Our study investigated the therapeutic potential of the suppressor of cytokine signaling 3 (SOCS3), an endogenous inhibitor of intracellular signaling pathways, for treatment of MPM. We infected MPM cells (H226, EHMES-1, MESO-1 and MESO-4) with an adenovirus-expressing SOCS3 (AdSOCS3) to examine the effect of SOCS3 overexpression on MPM cells. SOCS3 overexpression reduced MPM proliferation and induced apoptosis and partial G0/G1 arrest. SOCS3 also inhibited the proliferation of MPM cells via multiple signaling pathways including Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated kinase (ERK), focal adhesion kinase (FAK) and p53 pathways. Notably, AdSOCS3 treatment inhibited tumor growth in an MPM pleural xenograft model. These findings demonstrate that overexpression of SOCS3 has a potent antitumor effect against MPM both in vitro and in vivo and indicate the potential for clinical use of SOCS3 for MPM treatment.


Assuntos
Mesotelioma/prevenção & controle , Neoplasias Pleurais/prevenção & controle , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Adenoviridae/genética , Animais , Apoptose , Western Blotting , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Técnicas Imunoenzimáticas , Mesotelioma/metabolismo , Mesotelioma/patologia , Camundongos , Camundongos Endogâmicos ICR , Camundongos Nus , Neoplasias Pleurais/metabolismo , Neoplasias Pleurais/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Immunol ; 186(1): 32-40, 2011 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21106853

RESUMO

IL-6 is a proinflammatory cytokine and its overproduction is implicated in a variety of inflammatory disorders. Recent in vitro analyses suggest that IL-6 is a key cytokine that determines the balance between Foxp3(+) regulatory T cells (Tregs) and Th17 cells. However, it remains unclear whether excessive IL-6 production in vivo alters the development and function of Foxp3(+) Tregs. In this study, we analyzed IL-6 transgenic (Tg) mice in which serum IL-6 levels are constitutively elevated. Interestingly, in IL-6 Tg mice, whereas peripheral lymphoid organs were enlarged, and T cells exhibited activated phenotype, Tregs were not reduced but rather increased compared with wild-type mice. In addition, Tregs from Tg mice normally suppressed proliferation of naive T cells in vitro. Furthermore, Tregs cotransferred with naive CD4 T cells into SCID-IL-6 Tg mice inhibited colitis as successfully as those transferred into control SCID mice. These results indicate that overproduction of IL-6 does not inhibit development or function of Foxp3(+) Tregs in vivo. However, when naive CD4 T cells alone were transferred, Foxp3(+) Tregs retrieved from SCID-IL-6 Tg mice were reduced compared with SCID mice. Moreover, the Helios(-) subpopulation of Foxp3(+) Tregs, recently defined as extrathymic Tregs, was significantly reduced in IL-6 Tg mice compared with wild-type mice. Collectively, these results suggest that IL-6 overproduced in vivo inhibits inducible Treg generation from naive T cells, but does not affect the development and function of natural Tregs.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/biossíntese , Interleucina-6/biossíntese , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Diferenciação Celular/genética , Células Cultivadas , Técnicas de Cocultura , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/fisiologia , Humanos , Interleucina-6/sangue , Interleucina-6/fisiologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Camundongos Transgênicos , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/transplante
12.
Inflamm Bowel Dis ; 17(2): 491-502, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20602470

RESUMO

BACKGROUND: The efficacy of anti-tumor necrosis factor monoclonal antibody (anti-TNF mAb) for Crohn's disease (CD) is well established, and anti-interleukin-6 receptor (anti-IL-6R) mAb has also been reported to be effective in CD. It is, however, unclear if the efficacy and mechanisms of both agents are different in CD therapy. METHODS: Using an adoptive transfer colitis model, we compared the efficacy of anti-IL-6R mAb, anti-TNF mAb, and TNF receptor-Fc fusion protein (TNFR-Fc), and their modes of action on CD4+ T cells. We also investigated the role of Th1 and Th17 cells in colitis using the same model. RESULTS: The histological scores for the anti-IL-6R mAb and anti-TNF mAb groups but not for TNFR-Fc group were much lower than that for the control group, and the score was the lowest for the anti-IL-6R mAb group. The frequency of proliferating CD4+ T cells was reduced in anti-IL-6R mAb and anti-TNF mAb groups, but not in the TNFR-Fc group, whereas the frequency of apoptotic CD4+ T cells was similar in all groups. Anti-IL-6R mAb suppressed the induction of Th17 cells and increased the frequency of lamina propria regulatory T cells, whereas anti-TNF mAb exerted no influence on CD4+ T-cell differentiation. A deficiency in interferon-γ and/or IL-17 in CD4+ T cells reduced the severity of colitis. CONCLUSIONS: Our findings suggest that suppression of the proliferation of pathogenic CD4+ T cells is the major mode of action of biological agents for colitis therapy. Anti-IL-6R mAb might have benefits in CD patients with Th17 dominance and impaired Treg frequency.


Assuntos
Anticorpos Anti-Idiotípicos/farmacologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/imunologia , Colite/imunologia , Receptores de Interleucina-6/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Anticorpos Anti-Idiotípicos/imunologia , Anticorpos Monoclonais/imunologia , Western Blotting , Diferenciação Celular , Proliferação de Células , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Interferon gama/fisiologia , Interleucina-17/fisiologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Células Th1/imunologia , Células Th17/imunologia
13.
Exp Eye Res ; 91(2): 162-70, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20420831

RESUMO

The aim of this study was to investigate the effect of anti-mouse IL-6 receptor monoclonal antibody (MR16-1) treatment on CD4 T cell differentiation and compared it to the effect of anti-TNF mAb treatment with using a murine model of experimental autoimmune uveoretinitis (EAU). C57BL/6 mice were immunized with interphotoreceptor retinoid-binding protein (IRBP) to induce ocular inflammation treatment with control IgG or MR16-1 or anti-TNF mAb. Helper T cells differentiation was analyzed during the development of EAU. Immunization with IRBP increased the frequency of Th17 cells rather than Th1 cells in the early stage of EAU. Treatment with MR16-1 on the same day as immunization (day 0) or one day after (day 1) suppressed ocular inflammation in EAU mice. Treatment with MR16-1 on day 0 inhibited the induction of Th17 cells in vivo, and inhibited not only IRBP-responsive Th17 cells but also their Th1 counterparts and induced IRBP-responsive regulatory T (Treg) cells in vitro. The administration of anti-TNF mAb had no significant protective effect in EAU mice. The protective effect of anti-IL-6R mAb treatment, but not anti-TNF mAb treatment on EAU correlated with the inhibition of Th17 differentiation. This finding suggests that IL-6 blockade may have a therapeutic effect on human ocular inflammation which is mediated via mechanisms distinct from those of TNF blockade. IL-6 blockade may thus represent an alternative therapy for patients with ocular inflammation who are refractory to anti-TNF mAb therapy.


Assuntos
Anticorpos Monoclonais/farmacologia , Doenças Autoimunes/prevenção & controle , Interleucina-6/metabolismo , Retinite/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/imunologia , Uveíte/prevenção & controle , Animais , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Proteínas do Olho , Feminino , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Interleucina-6/imunologia , Retinite/imunologia , Retinite/patologia , Proteínas de Ligação ao Retinol , Fator de Necrose Tumoral alfa/imunologia , Uveíte/imunologia , Uveíte/patologia
14.
FEBS J ; 275(22): 5714-24, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18959756

RESUMO

We previously developed the bio-nanocapsule, which consists of hepatitis B virus envelope L proteins. The bio-nanocapsule can be used to deliver genes and drugs specifically to the human liver-derived tissues in xenograft models, presumably by utilizing the human liver-specific mechanism of hepatitis B virus infection. The hepatitis B virus tropism is highly restricted to humans and higher primates. Thus, to evaluate the in vivo therapeutic effects of forthcoming bio-nanocapsule-based medicines, it will be crucial to develop an animal model whose liver is susceptible to both bio-nanocapsule and hepatitis B virus. In the present study, we aimed to establish a bio-nanocapsule-susceptible animal model using transgenic rats expressing squamous cell carcinoma antigen-1 (SCCA1), which has been proposed to be a receptor for hepatitis B virus, interacting with the hepatitis B virus envelope protein and enhancing the cellular uptake of hepatitis B virus. We show that the recombinant SCCA1 protein interacts directly with bio-nanocapsule and inhibits its attachment to the cultured human liver-derived cells. Furthermore, we have established a transgenic rat that specifically expresses SCCA1 in the liver and also demonstrate that the amount of bio-nanocapsule accumulated in the liver is significantly increased by the SCCA1 expression. Histological analysis suggests that bio-nanocapsule is preferentially incorporated into the SCCA1-expressing hepatocytes but not into macrophages, such as Küppfer cells, nor into endothelial cells. Therefore, this animal model is expected to be useful for the development of bio-nanocapsule-based medicines.


Assuntos
Antígenos de Neoplasias/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Fígado/metabolismo , Nanocápsulas/uso terapêutico , Serpinas/metabolismo , Proteínas do Envelope Viral/farmacocinética , Animais , Vírus da Hepatite B/química , Humanos , Nanocápsulas/virologia , Ratos , Ratos Transgênicos , Distribuição Tecidual , Proteínas do Envelope Viral/uso terapêutico
15.
Proc Natl Acad Sci U S A ; 105(26): 9041-6, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18577591

RESUMO

The development of Th17 cells is a key event in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), a murine model of human multiple sclerosis (MS). Previous studies have demonstrated that an IL-6-dependent pathway is involved in the differentiation of Th17 cells from naïve CD4-positive T cells in vitro. However, the role of IL-6 in vivo in the development of Th17 cells in EAE has remained unclear. In the present study, we found that IL-6 blockade by treatment with an anti-IL-6 receptor monoclonal antibody (anti-IL-6R mAb) inhibited the development of EAE and inhibited the induction of myelin oligodendrocyte glycoprotein (MOG) peptide-specific CD4-positive, CD8-positive, and Th17 T cells, in inguinal lymph nodes. Thus, the protective effect of IL-6 blockade in EAE is likely to be mediated via the inhibition of the development of MOG-peptide-specific Th17 cells and Th1 cells, which in turn leads to reduced infiltration of T cells into the CNS. These findings indicate that anti-IL-6R mAb treatment might represent a novel therapy for human MS.


Assuntos
Antígenos/imunologia , Encefalomielite Autoimune Experimental/imunologia , Epitopos/imunologia , Interleucina-6/imunologia , Bainha de Mielina/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Células Th1/imunologia , Animais , Anticorpos Monoclonais/imunologia , Movimento Celular , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Encefalomielite Autoimune Experimental/patologia , Ativação Linfocitária/imunologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Peptídeos/imunologia
16.
Mol Cell Biol ; 27(5): 1730-44, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17189429

RESUMO

Transcription factor ATF-2 is a nuclear target of stress-activated protein kinases, such as p38, which are activated by various extracellular stresses, including UV light. Here, we show that ATF-2 plays a critical role in hypoxia- and high-cell-density-induced apoptosis and the development of mammary tumors. Compared to wild-type cells, Atf-2(-/-) mouse embryonic fibroblasts (MEFs) were more resistant to hypoxia- and anisomycin-induced apoptosis but remained equally susceptible to other stresses, including UV. Atf-2(-/-) and Atf-2(+/-) MEFs could not express a group of genes, such as Gadd45alpha, whose overexpression can induce apoptosis, in response to hypoxia. Atf-2(-/-) MEFs also had a higher saturation density than wild-type cells and expressed lower levels of Maspin, the breast cancer tumor suppressor, which is also known to enhance cellular sensitivity to apoptotic stimuli. Atf-2(-/-) MEFs underwent a lower degree of apoptosis at high cell density than wild-type cells. Atf-2(+/-) mice were highly prone to mammary tumors that expressed reduced levels of Gadd45alpha and Maspin. The ATF-2 mRNA levels in human breast cancers were lower than those in normal breast tissue. Thus, ATF-2 acts as a tumor susceptibility gene of mammary tumors, at least partly, by activating a group of target genes, including Maspin and Gadd45alpha.


Assuntos
Fator 2 Ativador da Transcrição/genética , Predisposição Genética para Doença , Neoplasias Mamárias Animais/genética , Fator 2 Ativador da Transcrição/análise , Fator 2 Ativador da Transcrição/metabolismo , Animais , Anisomicina/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Hipóxia Celular/efeitos dos fármacos , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Genes Reporter , Genes Supressores de Tumor , Humanos , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Proteínas Nucleares/metabolismo , Inibidores da Síntese de Ácido Nucleico/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/análise , Serpinas/metabolismo , Células Tumorais Cultivadas
18.
J Cell Physiol ; 207(2): 428-36, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16419040

RESUMO

Suppressor of cytokine signaling (SOCS)-2 regulates normal postnatal growth and its deficiency in mice causes gigantism with increased bone length and proportional enlargement in skeletal muscles. Using C2C12 mesenchymal precursor cell line as a model, we investigated a possible role of SOCS-2 in the differentiation process of mesenchymal precursors. Stable transfection of SOCS-2 into C2C12 cells resulted in the acceleration of proliferation and survival, and inhibition of spontaneous myotube formation. In addition, SOCS-2 potentiated bone morphogenic protein (BMP)-induced transdifferentiation of C2C12 cells into osteoblast phenotypes. These effects of SOCS-2 on C2C12 cells differed strikingly from that of SOCS-1, another member of SOCS family, and its mechanisms were evaluated. SOCS-2 did not alter BMP-induced phosphorylation and nuclear accumulation of Smad1, nor the expression of inhibitory-Smads mRNA. However, SOCS-2 enhanced BMP-induced transcriptional activation of the Smad-responsive reporter gene, suggesting that the action of SOCS-2 is exerted at the transcriptional level. Interestingly, SOCS-2 overexpression in C2C12 cells increased the endogenous JunB protein, one of the key transcriptional factors in the control of BMP/Smad signaling responsiveness. In addition, the proteasome inhibitor enhanced JunB protein expression in C2C12 cells. Moreover, we found that SOCS-2 reduced JunB ubiquitination in COS-7 cells. Although SOCS-2 is a modulator of growth hormone (GH) signaling, the upregulation of JunB by SOCS-2 did not require GH signaling. Taken together, these results suggest that SOCS-2 positively regulates endogenous JunB protein expression in C2C12 cells through inhibition of JunB destabilization by the ubiquitin-proteasome pathway, and thereby regulates the cell fate of mesenchymal precursors.


Assuntos
Diferenciação Celular/fisiologia , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Proteína Morfogenética Óssea 6 , Proteínas Morfogenéticas Ósseas/farmacologia , Células COS , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/genética , Chlorocebus aethiops , Expressão Gênica/genética , Leupeptinas/farmacologia , Camundongos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Proteína MyoD/genética , Mioblastos/efeitos dos fármacos , Mioblastos/metabolismo , Miogenina/genética , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteocalcina/genética , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Smad/metabolismo , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Transfecção , Regulação para Cima
20.
Cancer Res ; 63(22): 7606-8, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14633675

RESUMO

Recent studies showed that SV40 is detected in >40% of non-Hodgkin's lymphoma (NHL) in United States, suggesting SV40-contaminated poliovaccines widely used during the period 1955-1963 to be a major source of SV40 in NHL. We examined the presence of SV40 sequences in 122 cases with NHL and 3 with Hodgkin's lymphoma from Japan. The detection rate of SV40 sequences in diffuse large B-cell lymphoma (19%) was higher than that in peripheral blood cells of normal healthy volunteers in Japan (4.7%; P < 0.05) reported previously as controls for comparison with the study results from cancer patients, suggesting a role for SV40 in the development of diffuse large B-cell lymphoma. In contrast, the frequency of SV40 sequences in NHL cases born between 1951 and 1963 (12%), during which SV40-contaminated poliovaccines might have been inoculated, is not significantly different from that in cases born before 1950 (11%) or after 1964 (15%). SV40 is a new candidate etiologic factor for malignant lymphoma not only in the United States but also in Japan.


Assuntos
Linfoma não Hodgkin/virologia , Vírus 40 dos Símios/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos Transformantes de Poliomavirus/genética , Criança , Pré-Escolar , Feminino , Humanos , Hibridização In Situ , Lactente , Japão , Linfoma de Células B/genética , Linfoma de Células B/virologia , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/virologia , Linfoma não Hodgkin/genética , Linfoma de Células T/genética , Linfoma de Células T/virologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA