Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunology ; 162(2): 220-234, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33020922

RESUMO

TNF signalling through TNFRp55 and TNFRp75, and receptor shedding is important for immune activation and regulation. TNFRp75 deficiency leads to improved control of Mycobacterium tuberculosis (M. tuberculosis) infection, but the effects of early innate immune events in this process are unclear. We investigated the role of TNFRp75 on cell activation and apoptosis of alveolar macrophages and neutrophils during M. tuberculosis and M. bovis BCG infection. We found increased microbicidal activity against M. tuberculosis occurred independently of IFNy and NO generation, and displayed an inverse correlation with alveolar macrophages (AMs) apoptosis. Both M. tuberculosis and M. bovis BCG induced higher expression of MHC-II in TNFRp75-/- AMs; however, M bovis BCG infection did not alter AM apoptosis in the absence of TNFRp75. Pulmonary concentrations of CCL2, CCL3 and IL-1ß were increased in TNFRp75-/- mice during M, bovis BCG infection, but had no effect on neutrophil responses. Thus, TNFRp75-dependent regulation of mycobacterial replication is virulence dependent and occurs independently of early alveolar macrophage apoptosis and neutrophil responses.


Assuntos
Vacina BCG/imunologia , Macrófagos Alveolares/imunologia , Neutrófilos/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Tuberculose Bovina/imunologia , Tuberculose/imunologia , Animais , Apoptose/imunologia , Bovinos , Células Cultivadas , Feminino , Pulmão/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis/imunologia , Mycobacterium tuberculosis/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Virulência/imunologia
2.
J Immunol ; 204(7): 1859-1868, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32122996

RESUMO

Dendritic cells (DCs) participate in the pathogenesis of several diseases. We investigated DCs and the connection between mucosa and joints in a murine model of Yersinia enterocolitica O:3-induced reactive arthritis (ReA) in TNFRp55-/- mice. DCs of mesenteric lymph nodes (MLN) and joint regional lymph nodes (RLN) were analyzed in TNFRp55-/- and wild-type mice. On day 14 after Y. enterocolitica infection (arthritis onset), we found that under TNFRp55 deficiency, migratory (MHChighCD11c+) DCs increased significantly in RLN. Within these RLN, resident (MHCintCD11c+) DCs increased on days 14 and 21. Similar changes in both migratory and resident DCs were also detected on day 14 in MLN of TNFRp55-/- mice. In vitro, LPS-stimulated migratory TNFRp55-/- DCs of MLN increased IL-12/23p40 compared with wild-type mice. In addition, TNFRp55-/- bone marrow-derived DCs in a TNFRp55-/- MLN microenvironment exhibited higher expression of CCR7 after Y. enterocolitica infection. The major intestinal DC subsets (CD103+CD11b-, CD103-CD11b+, and CD103+CD11b+) were found in the RLN of Y. enterocolitica-infected TNFRp55-/- mice. Fingolimod (FTY720) treatment of Y. enterocolitica-infected mice reduced the CD11b- subset of migratory DCs in RLN of TNFRp55-/- mice and significantly suppressed the severity of ReA in these mice. This result was associated with decreased articular IL-12/23p40 and IFN-γ levels. In vitro FTY720 treatment downregulated CCR7 on Y. enterocolitica-infected bone marrow-derived DCs and purified MLN DCs, which may explain the mechanism underlying the impairment of DCs in RLN induced by FTY720. Taken together, data indicate the migration of intestinal DCs to RLN and the contribution of these cells in the immunopathogenesis of ReA, which may provide evidence for controlling this disease.


Assuntos
Artrite Reativa/imunologia , Células Dendríticas/imunologia , Linfonodos/imunologia , Mesentério/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Yersiniose/imunologia , Yersinia enterocolitica/imunologia , Animais , Artrite Reativa/metabolismo , Células Dendríticas/metabolismo , Linfonodos/metabolismo , Masculino , Mesentério/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proibitinas , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Yersiniose/metabolismo
3.
J Biol Chem ; 294(13): 5214-5227, 2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30723161

RESUMO

Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.


Assuntos
Vírus da Ectromelia/imunologia , Fatores Imunológicos/imunologia , Linfotoxina-alfa/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Proteínas Virais/imunologia , Sequência de Aminoácidos , Animais , Vírus da Ectromelia/química , Ectromelia Infecciosa/virologia , Humanos , Fatores Imunológicos/química , Camundongos , Modelos Moleculares , Domínios Proteicos , Fator de Necrose Tumoral alfa/imunologia , Proteínas Virais/química
4.
Blood ; 131(25): 2803-2815, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29699990

RESUMO

Large granular lymphocyte (LGL) leukemia results from clonal expansion of CD3+ cytotoxic T lymphocytes or CD3- natural killer (NK) cells. Chronic antigen stimulation is postulated to promote long-term survival of LGL leukemia cells through constitutive activation of multiple survival pathways, resulting in global dysregulation of apoptosis and resistance to activation-induced cell death. We reported previously that nuclear factor κB (NF-κB) is a central regulator of the survival network for leukemic LGL. However, the mechanisms that trigger constitutive activation of NF-κB in LGL leukemia remain undefined. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is known to induce apoptosis in tumor cells but can also activate NF-κB through interaction with TRAIL receptors 1, 2, and 4 (also known as DR4, DR5, and DcR2, respectively). The role of TRAIL has not been studied in LGL leukemia. In this study, we hypothesized that TRAIL interaction with DcR2 contributes to NF-κB activation in LGL leukemia. We observed upregulated TRAIL messenger RNA and protein expression in LGL leukemia cells with elevated levels of soluble TRAIL protein in LGL leukemia patient sera. We also found that DcR2 is the predominant TRAIL receptor in LGL leukemia cells. We demonstrated that TRAIL-induced activation of DcR2 led to increased NF-κB activation in leukemic LGL. Conversely, interruption of TRAIL-DcR2 signaling led to decreased NF-κB activation. Finally, a potential therapeutic application of proteasome inhibitors (bortezomib and ixazomib), which are known to inhibit NF-κB, was identified through their ability to decrease proliferation and increase apoptosis in LGL leukemia cell lines and primary patient cells.


Assuntos
Leucemia Linfocítica Granular Grande/imunologia , NF-kappa B/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Apoptose , Linhagem Celular Tumoral , Humanos , Leucemia Linfocítica Granular Grande/patologia , Mapas de Interação de Proteínas , Células Tumorais Cultivadas
5.
Exp Gerontol ; 95: 116-127, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28461078

RESUMO

Cell senescence plays a major role in the progression of tumors and chronic conditions such as diabetes and chronic kidney disease. Senescent cells are an important model for the study of aging-related diseases, and there is currently no efficient method for sorting out senescent cells. Decoy receptor 2 (DcR2) is a transmembrane receptor of the tumor necrosis factor superfamily, which is specifically expressed in senescent cells. In this study, we used magnetic activated cell sorting (MACS) isolation of a highly-pure populations DcR2-positive renal tubular epithelial cells (RTECs) based on three senescent cell models including the fifth passage cells, advanced glycation end-products (AGEs)- and H2O2-induced cells. The percentages of DcR2 positive RTECs in G1 and S phases increased by 20% and 4%, respectively, as compared to that in the pre-sorted cells. The positivity rates of SA-ß-gal, p16, and senescence-associated heterochromatin foci (SAHF) in DcR2-positive RTECs were about 40%, 30%, and 44% higher than that prior to cell sorting. The levels of IL-6 and TGF-ß1 in the supernatant were increased by 1.7 and 1.5 folds, respectively, as compared to that observed prior to sorting. No significant cell death was observed after 5days of continuous culture. Ki-67 positive expression rate in DcR2 negative RTECs was significantly higher than that in DcR2 positive RTECs after MACS. We demonstrated the use of DcR2 to classify live, senescent RTECs with a high specificity and stability. Our findings lay the foundation for further study of senescent RTECs in the progression of chronic kidney disease.


Assuntos
Proliferação de Células , Senescência Celular , Células Epiteliais/imunologia , Separação Imunomagnética , Túbulos Renais/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular , Produtos Finais de Glicação Avançada/farmacologia , Peróxido de Hidrogênio/farmacologia , Interleucina-6/metabolismo , Antígeno Ki-67/metabolismo , Túbulos Renais/citologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Pontos de Checagem da Fase S do Ciclo Celular , Fatores de Tempo , Fator de Crescimento Transformador beta1/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo
6.
Trends Mol Med ; 21(6): 373-84, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25851173

RESUMO

Post-transcriptional regulation of RNA is an important mechanism for activating and resolving cellular stress responses. Poly(ADP-ribose) polymerase-13 (PARP13), also known as ZC3HAV1 and zinc-finger antiviral protein (ZAP), is an RNA-binding protein that regulates the stability and translation of specific mRNAs, and modulates the miRNA silencing pathway to globally affect miRNA targets. These functions of PARP13 are important components of the cellular response to stress. In addition, the ability of PARP13 to restrict oncogenic viruses and to repress the prosurvival cytokine receptor tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor 4 (TRAILR4) suggests that it can be protective against malignant transformation and cancer development. The relevance of PARP13 to human health and disease make it a promising therapeutic target.


Assuntos
Neoplasias/genética , Neoplasias/imunologia , Proteínas de Ligação a RNA/imunologia , RNA/genética , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Imunidade Inata , Neoplasias/virologia , RNA/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
7.
Immunol Lett ; 159(1-2): 55-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24657523

RESUMO

Tumor microenvironment represents the site where the tumor tries to survive and escape from immune system-mediated recognition. Indeed, to proliferate tumor cells can divert the immune response inducing the generation of myeloid derived suppressor cells and regulatory T cells which can limit the efficiency of effector antitumor lymphocytes in eliminating neoplastic cells. Many components of the tumor microenvironment can serve as a double sword for the tumor and the host. Several types of fibroblast-like cells, which herein we define mesenchymal stromal cells (MSC), secrete extracellular matrix components and surrounding the tumor mass can limit the expansion of the tumor. On the other hand, MSC can interfere with the immune recognition of tumor cells producing immunoregulatory cytokines as transforming growth factor (TGF)ß, releasing soluble ligands of the activating receptors expressed on cytolytic effector cells as decoy molecules, affecting the correct interaction among lymphocytes and tumor cells. MSC can also serve as target for the same anti-tumor effector lymphocytes or simply impede the interaction between these lymphocytes and neoplastic cells. Thus, several evidences point out the role of MSC, both in epithelial solid tumors and hematological malignancies, in regulating tumor cell growth and immune response. Herein, we review these evidences and suggest that MSC can be a suitable target for a more efficient anti-tumor therapy.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Mesenquimais/imunologia , Neoplasias/imunologia , Evasão Tumoral/imunologia , Matriz Extracelular/imunologia , Matriz Extracelular/metabolismo , Humanos , Sistema Imunitário , Imunoterapia , Células-Tronco Mesenquimais/patologia , Células Mieloides/imunologia , Células Mieloides/patologia , Neoplasias/genética , Neoplasias/patologia , Neoplasias/terapia , Transdução de Sinais , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/imunologia , Evasão Tumoral/genética , Microambiente Tumoral/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
8.
J Clin Invest ; 124(4): 1537-51, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24569452

RESUMO

Development of host protective immunity against Mycobacterium tuberculosis infection is critically dependent on the inflammatory cytokine TNF. TNF signals through 2 receptors, TNFRp55 and TNFRp75; however, the role of TNFRp75-dependent signaling in immune regulation is poorly defined. Here we found that mice lacking TNFRp75 exhibit greater control of M. tuberculosis infection compared with WT mice. TNFRp75-/- mice developed effective bactericidal granulomas and demonstrated increased pulmonary recruitment of activated DCs. Moreover, IL-12p40-dependent migration of DCs to lung draining LNs of infected TNFRp75-/- mice was substantially higher than that observed in WT M. tuberculosis-infected animals and was associated with enhanced frequencies of activated M. tuberculosis-specific IFN-γ-expressing CD4+ T cells. In WT mice, TNFRp75 shedding correlated with markedly reduced bioactive TNF levels and IL-12p40 expression. Neutralization of TNFRp75 in M. tuberculosis-infected WT BM-derived DCs (BMDCs) increased production of bioactive TNF and IL-12p40 to a level equivalent to that produced by TNFRp75-/- BMDCs. Addition of exogenous TNFRp75 to TNFRp75-/- BMDCs infected with M. tuberculosis decreased IL-12p40 synthesis, demonstrating that TNFRp75 shedding regulates DC activation. These data indicate that TNFRp75 shedding downmodulates protective immune function and reduces host resistance and survival; therefore, targeting TNFRp75 may be beneficial for improving disease outcome.


Assuntos
Mycobacterium tuberculosis/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Células Dendríticas/imunologia , Feminino , Granuloma/patologia , Interações Hospedeiro-Patógeno/imunologia , Subunidade p40 da Interleucina-12/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mycobacterium tuberculosis/patogenicidade , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/genética , Transdução de Sinais/imunologia , Solubilidade , Tuberculose Pulmonar/imunologia , Tuberculose Pulmonar/microbiologia , Tuberculose Pulmonar/patologia , Receptores Chamariz do Fator de Necrose Tumoral/deficiência , Receptores Chamariz do Fator de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Cell Biochem Biophys ; 67(3): 1127-36, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23677859

RESUMO

Apoptosis can occur throughout the life span of osteoblasts (OBs), beginning from the early stages of differentiation and continuing throughout all stages of their working life. Here, we investigated the effects of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) on normal human OBs showing for the first time that the expression of TRAIL receptors is modulated during OB differentiation. In particular, the TRAIL receptor ratio was in favor of the deaths because of the low expression of DcR2 in undifferentiated OBs, differently it was shifted toward the decoys in differentiated ones. Undifferentiated OBs treated with TRAIL showed reduced cell viability, whereas differentiated OBs displayed TRAIL resistance. The OB sensitiveness to TRAIL was due to the up-regulation of DR5 and the down-regulation of DcR2. The main death receptor involved in TRAIL-reduced OB viability was DR5 as demonstrated by the rescue of cell viability observed in the presence of anti-DR5 neutralizing antibody. Besides the ratio of TRAIL receptors, the sensitivity of undifferentiated OBs to TRAIL-cytotoxic effect was also associated with low mRNA levels of intracellular anti-apoptotic proteins, such as cFLIP, the activation of caspase-8 and -3, as well as the DNA fragmentation. This study suggests that apoptotic effect exerted by TRAIL/TRAIL-receptor system on normal human OB is strictly dependent upon cell differentiation status.


Assuntos
Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/toxicidade , Anticorpos Neutralizantes/imunologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , DNA/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Osteoblastos/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Regulação para Cima/efeitos dos fármacos
10.
Thorax ; 67(3): 244-51, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22156959

RESUMO

BACKGROUND: Tumour necrosis factor (TNF) is upregulated in the alveolar space early in the course of ventilator-induced lung injury (VILI). Studies in genetically modified mice indicate that the two TNF receptors play opposing roles during injurious high-stretch mechanical ventilation, with p55 promoting but p75 preventing pulmonary oedema. AIM: To investigate the effects of selective inhibition of intra-alveolar p55 TNF receptor on pulmonary oedema and inflammation during ventilator-induced lung injury using a newly developed domain antibody. METHODS: Anaesthetised mice were ventilated with high tidal volume and given an intratracheal bolus of p55-specific domain antibody or anti-TNF monoclonal antibody ('pure' VILI model). As a model of enhanced inflammation, a subclinical dose of lipopolysaccharide (LPS) was included in the intratracheal antibody bolus (LPS+VILI model). Development of lung injury was assessed by respiratory mechanics and blood gases and protein levels in lavage fluid. Flow cytometry was used to determine leucocyte recruitment and alveolar macrophage activation, while lavage fluid cytokines were assessed by ELISA. RESULTS: The ventilation protocol produced deteriorations in respiratory mechanics and gas exchange with increased lavage fluid protein levels in the two models. The p55-specific domain antibody substantially attenuated all of these changes in the 'pure' VILI model, while anti-TNF antibody was ineffective. In the LPS+VILI model, p55 blockade prevented deteriorations in respiratory mechanics and oxygenation and significantly decreased neutrophil recruitment, expression of intercellular adhesion molecule 1 on alveolar macrophages, and interleukin 6 and monocyte chemotactic protein 1 levels in lavage fluid. CONCLUSIONS: Selective inhibition of intra-alveolar p55 TNF receptor signalling by domain antibodies may open new therapeutic approaches for ventilated patients with acute lung injury.


Assuntos
Anticorpos Neutralizantes/uso terapêutico , Alvéolos Pulmonares/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/antagonistas & inibidores , Receptores Chamariz do Fator de Necrose Tumoral/antagonistas & inibidores , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/imunologia , Dióxido de Carbono/sangue , Avaliação Pré-Clínica de Medicamentos/métodos , Lipopolissacarídeos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxigênio/sangue , Pressão Parcial , Edema Pulmonar/etiologia , Edema Pulmonar/prevenção & controle , Troca Gasosa Pulmonar/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Resultado do Tratamento , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Lesão Pulmonar Induzida por Ventilação Mecânica/complicações , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/fisiopatologia
11.
Clin Dev Immunol ; 2012: 135932, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21860649

RESUMO

In this paper we focus our attention on the role of two families of receptors, Toll-like receptors (TLR) and decoy receptors (DcR) involved in the generation of systemic lupus erythematosus (SLE) and lupus-like syndromes in human and mouse models. To date, these molecules were described in several autoimmune disorders such as rheumatoid arthritis, antiphospholipids syndrome, bowel inflammation, and SLE. Here, we summarize the findings of recent investigations on TLR and DcR and their role in the immunopathogenesis of the SLE.


Assuntos
Lúpus Eritematoso Sistêmico/imunologia , Receptores Toll-Like/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Animais , Autoantígenos/imunologia , Autoimunidade , Comunicação Celular , Quimiocinas/imunologia , Modelos Animais de Doenças , Humanos , Imunidade Inata , Camundongos , Transdução de Sinais
12.
Immunobiology ; 216(12): 1322-30, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21802165

RESUMO

While cytokines are major regulators of macrophage activation following host-pathogen interactions, they also act to limit inflammation to avoid tissue damage. In previous studies we reported the development of progressive Yersinia enterocolitica-induced reactive arthritis (ReA) in mice lacking the tumor necrosis factor receptor p55 (TNFRp55). In this work, we analyzed the response of TNFRp55⁻/⁻ macrophages to Y. enterocolitica antigens. We found higher concentration of nitric oxide (NO) in TNFRp55⁻/⁻ compared to wild-type macrophages in response to heat-killed Yersinia (HKY) and Yersinia outer membranes (OM). Moreover, Toll-like receptor (TLR)4 expression was increased in OM-stimulated TNFRp55⁻/⁻ versus wild-type (WT) macrophages. Accordingly, NO production was inhibited in TLR4-deficient macrophages following stimulation with OM, suggesting that LPS may function as a major OM component implicated in these responses. Thus, augmented NO production together with enhanced expression of inducible nitric oxide synthase (iNOS) and higher IL-6 production, may provide a pro-inflammatory setting in Yersinia LPS-stimulated TNFRp55⁻/⁻ macrophages. Augmented synthesis of NO and IL-6 was prevented by treatment with Polymyxin B, or by exposure to a specific NF-κB p65 oligonucleotide antisense, indicating the involvement of TLR4-mediated NF-κB activation in the unleashed pro-inflammatory response triggered by TNFRp55 deficiency. Thus, TNFRp55 modulates macrophage functions in response to Yersinia LPS stimulation through mechanisms involving NO, IL-6 and NF-κB pathways, suggesting an essential regulatory role of TNF via TNFRp55 signaling.


Assuntos
Interleucina-6/metabolismo , Macrófagos Peritoneais/metabolismo , Óxido Nítrico/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Yersinia enterocolitica/imunologia , Animais , Antígenos de Bactérias/imunologia , Antígenos de Bactérias/metabolismo , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Inflamação , Interleucina-6/genética , Interleucina-6/imunologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/patologia , Camundongos , Camundongos Knockout , NF-kappa B/genética , NF-kappa B/metabolismo , Oligodesoxirribonucleotídeos Antissenso/genética , Polimixina B/farmacologia , Proibitinas , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/genética , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
13.
PLoS One ; 6(4): e18291, 2011 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-21483669

RESUMO

BACKGROUND: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) could induce apoptosis of HIV-1-infected monocyte-derived macrophage (MDM), but the molecular mechanisms are not well understood. METHODOLOGY/PRINCIPAL FINDINGS: By using an HIV-1 Env-pseudotyped virus (HIV-1 PV)-infected MDM cell model we demonstrate that HIV-1 PV infection down-regulates the expression of TRAIL decoy receptor 1 (DcR1) and 2 (DcR2), and cellular FLICE-inhibitory protein (c-FLIP), but dose not affect the expression of death receptor 4 and 5 (DR4, DR5), and Bcl-2 family members in MDM cells. Furthermore, recombinant soluble TRAIL and an agonistic anti-DR5 antibody, AD5-10, treatment stimulates reactive oxygen species (ROS) generation and JNK phosphorylation. CONCLUSIONS/SIGNIFICANCE: HIV infection facilitates TRIAL-induced cell death in MDM by down-regulating the expression of TRAIL decoy receptors and intracellular c-FLIP. Meanwhile, the agonistic anti-DR5 antibody, AD5-10, induces apoptosis synergistically with TRAIL in HIV-1-infected cells. ROS generation and JNK phosphorylation are involved in this process. These findings potentiate clinical usage of the combination of TRAIL and AD5-10 in eradication of HIV-infected macrophage and AIDS.


Assuntos
Apoptose/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , HIV-1/fisiologia , Macrófagos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspases/metabolismo , Sinergismo Farmacológico , Células HEK293 , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Macrófagos/virologia , Monócitos/citologia , Fosforilação/efeitos dos fármacos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacologia , Solubilidade , Ligante Indutor de Apoptose Relacionado a TNF/química , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Latência Viral/efeitos dos fármacos
14.
Int Immunopharmacol ; 11(8): 1038-44, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21382533

RESUMO

The role of tumor necrosis factor-α (TNF-α) in contributing to allergen induced airway remodeling in asthma is unknown. In this study we have utilized a mouse model of chronic OVA allergen induced airway remodeling to determine whether TNF p55/p75 receptor deficient mice (abbreviated TNF-R KO) had reduced levels of airway remodeling. Chronic OVA challenged WT mice had significantly increased levels of lung eosinophilic inflammation as well as features of airway remodeling including increased peribronchial fibrosis, thickness of the peribronchial smooth muscle layer, mucus expression, and deposition of extracellular matrix proteins. In contrast, TNF-R KO mice had significantly reduced levels of major basic protein positive peribronchial eosinophils and significantly reduced peribronchial fibrosis assessed by quantitating the area of peribronchial trichrome staining and total lung collagen. In addition, TNF-R KO mice had significantly reduced thickness of the peribronchial smooth muscle layer, area of peribronchial α-smooth muscle actin immunostaining, and levels of the extracellular matrix protein fibronectin. There was a non-significant trend for reduced mucus expression in TNF-R KO mice. Levels of peribronchial cells immunostaining positive for TGF-ß1 were significantly reduced in TNF-R KO mice suggesting that reduced levels of TGF-ß1 expression in TNF-R KO mice may contribute to reduced airway remodeling. Overall, this study suggests an important role for TNF-α in contributing to many features of allergen induced airway remodeling including changes in levels of peribronchial smooth muscle, subepithelial fibrosis, and deposition of extracellular matrix.


Assuntos
Remodelação das Vias Aéreas/efeitos dos fármacos , Alérgenos/farmacologia , Ovalbumina/farmacologia , Eosinofilia Pulmonar/metabolismo , Eosinofilia Pulmonar/patologia , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Chamariz do Fator de Necrose Tumoral/deficiência , Animais , Asma/metabolismo , Asma/patologia , Brônquios/metabolismo , Brônquios/patologia , Colágeno/metabolismo , Eosinófilos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Imunoglobulina E/sangue , Interleucina-5/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Muco/metabolismo , Músculo Liso/efeitos dos fármacos , Músculo Liso/imunologia , Músculo Liso/metabolismo , Músculo Liso/patologia , Eosinofilia Pulmonar/induzido quimicamente , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/imunologia , Fibrose Pulmonar/metabolismo , Fibrose Pulmonar/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
15.
J Immunol ; 185(9): 5259-67, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20921531

RESUMO

TRAIL is known to play a pivotal role in the inhibition of autoimmune disease. We previously demonstrated that administration of dendritic cells engineered to express TRAIL and myelin-oligodendrocyte glycoprotein reduced the severity of experimental autoimmune encephalomyelitis and suggested that CD4(+)CD25(+) regulatory T cells (Tregs) were involved in mediating this preventive effect. In the current study, we investigated the effect of TRAIL on Tregs, as well as conventional T cells, using TRAIL-deficient mice. Upon induction of experimental autoimmune encephalomyelitis, TRAIL-deficient mice showed more severe clinical symptoms, a greater frequency of IFN-γ-producing CD4(+) T (Th1) cells, and a lower frequency of CD4(+)Foxp3(+) Tregs than did wild-type mice. In vitro, conventional T cells stimulated by bone marrow-derived dendritic cells (BM-DCs) from TRAIL-deficient mice showed a greater magnitude of proliferation than did those stimulated by BM-DCs from wild-type mice. In contrast, TRAIL expressed on the stimulator BM-DCs enhanced the proliferative response of CD4(+)CD25(+) Tregs in the culture. The functional TRAILR, mouse death receptor 5 (mDR5), was expressed in conventional T cells and Tregs upon stimulation. In contrast, the decoy receptor, mDc-TRAILR1, was slightly expressed only on CD4(+)CD25(+) Tregs. Therefore, the distinct effects of TRAIL may be due to differences in the mDc-TRAILR1 expression or the signaling pathways downstream of mouse death receptor 5 between the two T cell subsets. Our data suggest that TRAIL suppresses autoimmunity by two mechanisms: the inhibition of Th1 cells and the promotion of Tregs.


Assuntos
Autoimunidade/imunologia , Transdução de Sinais/imunologia , Linfócitos T Reguladores/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Células Th1/imunologia , Animais , Proliferação de Células , Separação Celular , Encefalomielite Autoimune Experimental/imunologia , Citometria de Fluxo , Imuno-Histoquímica , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/citologia , Células Th1/citologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
16.
Cancer Treat Rev ; 35(3): 280-8, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19117685

RESUMO

Tumour necrosis factor-related apoptosis-inducing ligand or Apo2 ligand (TRAIL/Apo2L) is a member of the tumour necrosis factor (TNF) superfamily of cytokines that induces apoptosis upon binding to its death domain-containing transmembrane receptors, death receptors 4 and 5 (DR4, DR5). Importantly, TRAIL preferentially induces apoptosis in cancer cells while exhibiting little or no toxicity in normal cells. To date, research has focused on the mechanism of apoptosis induced by TRAIL and the processes involved in the development of TRAIL resistance. TRAIL-resistant tumours can be re-sensitized to TRAIL by a combination of TRAIL with chemotherapeutics or irradiation. Studies suggest that in many cancer cells only one of the two death-inducing TRAIL receptors is functional. These findings as well as the aim to avoid decoy receptor-mediated neutralization of TRAIL led to the development of receptor-specific TRAIL variants and agonistic antibodies. These molecules are predicted to be more potent than native TRAIL in vivo and may be suitable for targeted treatment of particular tumours. This review focuses on the current status of TRAIL receptor-targeting for cancer therapy, the apoptotic signalling pathway induced by TRAIL receptors, the prognostic implications of TRAIL receptor expression and modulation of TRAIL sensitivity of tumour cells by combination therapies. The mechanisms of TRAIL resistance and the potential measures that can be taken to overcome them are also addressed. Finally, the status of clinical trials of recombinant TRAIL and DR4-/DR5-specific agonistic antibodies as well as the pre-clinical studies of receptor-selective TRAIL variants is discussed including the obstacles facing the use of these molecules as anti-cancer therapeutics.


Assuntos
Antineoplásicos/farmacologia , Apoptose/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias/terapia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Receptores do Fator de Necrose Tumoral/fisiologia , Ligante Indutor de Apoptose Relacionado a TNF/fisiologia , Receptores Chamariz do Fator de Necrose Tumoral/fisiologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/fisiologia , Ensaios Clínicos como Assunto/estatística & dados numéricos , Terapia Combinada , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos , Proteínas Ligadas por GPI , Humanos , Proteínas de Neoplasias/agonistas , Proteínas de Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neoplasias/radioterapia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Receptores do Fator de Necrose Tumoral/agonistas , Membro 10c de Receptores do Fator de Necrose Tumoral , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/agonistas , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Receptores Chamariz do Fator de Necrose Tumoral/agonistas , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Regulação para Cima/efeitos dos fármacos
17.
J Biol Chem ; 283(12): 7754-62, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18178561

RESUMO

TRAIL/Apo2L (tumor necrosis factor-related apoptosis-inducing ligand) is a multifunctional protein regulating homeostasis of the immune system, infection, autoimmune diseases, and apoptosis. However, its function in normal, nontransformed tissues is not clear. Here we show that TRAIL increases vascular smooth muscle cell (VSMC) proliferation in vitro, effects that can be blocked with neutralizing antibodies to TRAIL receptors DR4 and DcR1. In aortocoronary saphenous vein bypass grafts in vivo, TRAIL co-localizes with VSMC, proliferating cell nuclear antigen, and insulin-like growth factor type 1 receptor (IGF1R) expression but not active caspase-3. TRAIL is required for serum-inducible IGF1R expression, and antisense IGF1R inhibits TRAIL-induced VSMC proliferation. At 1 ng/ml, TRAIL stimulates IGF1R mRNA expression greater than insulin-like growth factor-1 and also activates the IGF1R promoter 7-fold. TRAIL-inducible IGF1R expression requires NF-kappaB activation. Consistent with this, ammonium pyrrolidine dithiocarbamate, a pharmacological inhibitor of NF-kappaB, blocks TRAIL-induced IGF1R expression, and p65 overexpression increases IGF1R protein levels. In addition, NF-kappaB binds a novel TRAIL-responsive element on the IGF1R promoter. Our findings suggest that the biological functions of TRAIL in VSMC extend beyond its role in promoting apoptosis. Thus, TRAIL may play an important role in atherosclerosis by regulating IGF1R expression in VSMC in an NF-kappaB-dependent manner.


Assuntos
Proliferação de Células/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Receptor IGF Tipo 1/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Transcrição RelA/metabolismo , Antioxidantes/farmacologia , Apoptose/imunologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/metabolismo , Doenças Autoimunes/patologia , Caspase 3/biossíntese , Caspase 3/imunologia , Células Cultivadas , Ponte de Artéria Coronária , Doença da Artéria Coronariana/imunologia , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Relação Dose-Resposta a Droga , Proteínas Ligadas por GPI , Regulação da Expressão Gênica/fisiologia , Humanos , Infecções/imunologia , Infecções/metabolismo , Infecções/patologia , Músculo Liso Vascular/imunologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Antígeno Nuclear de Célula em Proliferação/biossíntese , Antígeno Nuclear de Célula em Proliferação/imunologia , Pirrolidinas/farmacologia , Receptor IGF Tipo 1/imunologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF , Receptores do Fator de Necrose Tumoral/imunologia , Receptores do Fator de Necrose Tumoral/metabolismo , Membro 10c de Receptores do Fator de Necrose Tumoral , Ligante Indutor de Apoptose Relacionado a TNF/imunologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Tiocarbamatos/farmacologia , Fator de Transcrição RelA/antagonistas & inibidores , Fator de Transcrição RelA/imunologia , Transplantes , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo
18.
Parasite Immunol ; 29(11): 575-82, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17944747

RESUMO

Th1 and Th2 responses to the gut-dwelling nematode Trichuris muris have been well established in mouse models of infection, with Th2 responses clearly playing an important role in resistance. TNF-alpha has previously been shown to play an undefined role in resistance, although it is not a typical Th2 cytokine. However, the relative importance of the two TNF-alpha receptors, p55 and p75, has not previously been investigated. We demonstrate that p55 is the dominant TNF-alpha receptor during T. muris infection as p55-/- mice are more susceptible to infection than p75-/- mice. Moreover, p75 clearly plays a role in negatively regulating TNF-alpha. We also demonstrate that a gender difference influences the immune response of p55-/- and p75-/- mice in response to T. muris infection, with female mice fully expelling by day 35 post-infection (p.i.) and male mice harbouring chronic infections. Further, this gender difference can be reversed with recombinant IL-13 (rIL-13) in male gene-deficient mice or IL-13R2.Fc treatment in female gene-deficient mice.


Assuntos
Interleucina-13/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Tricuríase/imunologia , Trichuris/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Fatores Sexuais , Células Th1/imunologia , Células Th2/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
19.
Blood ; 109(9): 3839-48, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17202317

RESUMO

Bone homeostasis is regulated by a delicate balance between osteoblastic bone formation and osteoclastic bone resorption. Osteoclastogenesis is controlled by the ratio of receptor activator of NF-kappaB ligand (RANKL) relative to its decoy receptor, osteoprotegerin (OPG). The source of OPG has historically been attributed to osteoblasts (OBs). While activated lymphocytes play established roles in pathological bone destruction, no role for lymphocytes in basal bone homeostasis in vivo has been described. Using immunomagnetic isolation of bone marrow (BM) B cells and B-cell precursor populations and quantitation of their OPG production by enzyme-linked immunosorbent assay (ELISA) and real-time reverse transcriptase-polymerase chain reaction (RT-PCR), cells of the B lineage were found to be responsible for 64% of total BM OPG production, with 45% derived from mature B cells. Consistently B-cell knockout (KO) mice were found to be osteoporotic and deficient in BM OPG, phenomena rescued by B-cell reconstitution. Furthermore, T cells, through CD40 ligand (CD40L) to CD40 costimulation, promote OPG production by B cells in vivo. Consequently, T-cell-deficient nude mice, CD40 KO mice, and CD40L KO mice display osteoporosis and diminished BM OPG production. Our data suggest that lymphocytes are essential stabilizers of basal bone turnover and critical regulators of peak bone mass in vivo.


Assuntos
Linfócitos B/imunologia , Reabsorção Óssea/imunologia , Homeostase/imunologia , Osteogênese/imunologia , Linfócitos T/imunologia , Animais , Densidade Óssea/imunologia , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Camundongos , Camundongos Knockout , Camundongos Nus , Osteoblastos/imunologia , Osteoclastos/imunologia , Osteoporose/imunologia , Osteoprotegerina/imunologia , Ligante RANK/imunologia , Receptores Chamariz do Fator de Necrose Tumoral/imunologia
20.
Clin Exp Immunol ; 147(1): 128-38, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17177972

RESUMO

Inflammatory immune reactions in response to periodontopathogens are thought to protect the host against infection, but may trigger periodontal destruction. Thus, we examined the mechanisms by which the proinflammatory cytokine tumour necrosis factor (TNF)-alpha modulates the outcome of Actinobacillus actinomycetemcomitans-induced periodontal disease in mice. Our results showed that TNF-alpha receptor p55-deficient mice [p55TNF-knock-out (KO)] developed a less severe periodontitis in response to A. actinomycetemcomitans infection, characterized by significantly less alveolar bone loss and inflammatory reaction. Real-time polymerase chain reaction (PCR) demonstrated that levels of chemokines (CXCL1, 3 and 10; CCL3 and 5) and their receptors (CXCR2 and 3, CCR5) were lower in p55TNF-KO mice, as were matrix metalloproteinase (MMP)-1, 2 and 9 and receptor activator of nuclear factor kB ligand (RANKL) mRNA levels. However, the absence of the TNF-alpha p55 results in an impairment of protective immunity to A. actinomycetemcomitans infection, characterized by increased bacterial load and higher levels of C-reactive protein during the course of disease. Such impaired host response may be the result of the reduced chemoattraction of lymphocytes, neutrophils and macrophages, and reduced inducible nitric oxide synthase expression (iNOS) and myeloperoxidase (MPO) production in periodontal tissues of p55 TNF-KO mice. Our results demonstrate the mechanisms involved determining periodontal disease severity by TNF-alpha receptor p55, and its role in providing immune protection to A. actinomycetemcomitans periodontal infection.


Assuntos
Infecções por Actinobacillus/imunologia , Aggregatibacter actinomycetemcomitans , Periodontite/imunologia , Periodonto/imunologia , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Infecções por Actinobacillus/patologia , Aggregatibacter actinomycetemcomitans/imunologia , Perda do Osso Alveolar , Animais , Anticorpos Antibacterianos/sangue , Proteína C-Reativa/análise , Quimiocina CCL5 , Quimiocina CXCL1 , Quimiocina CXCL10 , Quimiocinas CC/análise , Quimiocinas CC/genética , Quimiocinas CXC/análise , Quimiocinas CXC/genética , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática/métodos , Citometria de Fluxo , Interferon gama/análise , Interferon gama/genética , Interleucina-10/sangue , Interleucina-10/genética , Interleucina-1beta/análise , Interleucina-1beta/genética , Metaloproteinases da Matriz/análise , Metaloproteinases da Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Periodontite/patologia , Periodonto/patologia , Peroxidase/análise , Ligante RANK/análise , Ligante RANK/genética , Receptores CCR5/análise , Receptores CCR5/genética , Receptores CXCR3 , Receptores de Quimiocinas/análise , Receptores de Quimiocinas/genética , Receptores de Interleucina-8B/análise , Receptores de Interleucina-8B/genética , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Chamariz do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...