Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Cytokine ; 177: 156542, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38364458

RESUMO

The COVID-19 patients showed hyperinflammatory response depending on the severity of the disease but little have been reported about this response in oncologic patients that also were infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Sixty-five circulating cytokines/chemokines were quantified in 15 oncologic patients, just after SARS-CoV-2 infection and fourteen days later, and their levels were compared in patients who required hospitalisation by COVID-19 versus non-hospitalised patients. A higher median age of 72 years (range 61-83) in oncologic patients after SARS-CoV-2 infection was associated with hospitalisation requirement by COVID-19 versus a median age of 49 years (20-75) observed in the non-hospitalised oncologic patients (p = 0.008). Moreover, oncologic patients at metastatic stage or with lung cancer were significantly associated with hospitalisation by COVID-19 (p = 0.044). None of these hospitalised patients required ICU treatment. Higher basal levels of tumour necrosis factor receptor II (TNF-RII), interferon-γ (IFNγ)-induced protein 10 (IP-10) and hepatocyte growth factor (HGF) in plasma were significantly observed in oncologic patients who required hospitalisation by COVID-19. Higher TNF-RII, IP-10 and HGF levels after the SARS-CoV-2 infection in oncologic patients could be used as biomarkers of COVID-19 severity associated with hospitalisation requirements.


Assuntos
COVID-19 , Neoplasias , Idoso , Idoso de 80 Anos ou mais , Humanos , Pessoa de Meia-Idade , Quimiocina CXCL10/sangue , Quimiocina CXCL10/química , COVID-19/diagnóstico , COVID-19/metabolismo , Fator de Crescimento de Hepatócito/sangue , Fator de Crescimento de Hepatócito/química , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo II do Fator de Necrose Tumoral/química , SARS-CoV-2 , Neoplasias/metabolismo
2.
Mol Pharm ; 20(4): 1884-1897, 2023 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-36897792

RESUMO

Tumor necrosis factor (TNF) is a key regulator of immune responses and plays a significant role in the initiation and maintenance of inflammation. Upregulation of TNF expression leads to several inflammatory diseases, such as Crohn's, ulcerative colitis, and rheumatoid arthritis. Despite the clinical success of anti-TNF treatments, the use of these therapies is limited because they can induce adverse side effects through inhibition of TNF biological activity, including blockade of TNF-induced immunosuppressive function of TNFR2. Using yeast display, we identified a synthetic affibody ligand (ABYTNFR1-1) with high binding affinity and specificity for TNFR1. Functional assays showed that the lead affibody potently inhibits TNF-induced NF-κB activation (IC50 of 0.23 nM) and, crucially, does not block the TNFR2 function. Additionally, ABYTNFR1-1 acts non-competitively─it does not block TNF binding or inhibit receptor-receptor interactions in pre-ligand-assembled dimers─thereby enhancing inhibitory robustness. The mechanism, monovalent potency, and affibody scaffold give this lead molecule uniquely strong potential as a therapeutic candidate for inflammatory diseases.


Assuntos
Receptores Tipo II do Fator de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Ligantes , Inibidores do Fator de Necrose Tumoral , Fator de Necrose Tumoral alfa/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-34781108

RESUMO

HS002 is the recombinant human tumor necrosis factor-α receptor Ⅱ: IgG Fc fusion protein licensed in China to treat rheumatism and psoriasis. The aim of this study was to isolate and characterize the hydrophobic freeze-dried powder injection (HS002) and ampoule injection (HS002A) variants derived from proteins of the same sequence and then to explore the structure-function relationship. Extensive physicochemical and structural testing was performed during a side-by-side comparison of the monomer peak and variant. Then the TNF-α-related binding activity, cell biological activity and affinity with FcRn were analyzed. Finally, a transformation study of the hydrophobic variant was performed under serum-like redox conditions. This research revealed that HS002A has similar physicochemical and structure-function relationship profiles to those of HS002. The hydrophobic variant exhibited the presence of new incorrect disulfide bridging. At the same time, this novel disulfide scrambled species structure-function relationship was found to be the molecular basis for reduced TNF-α binding and cell biological activities. In addition, incorrect disulfide bridging was found to be reversible under serum-like redox conditions, restoring TNF-α binding and cell biological activities to almost normal levels, all of which indicate that the variant is probably irrelevant to clinical efficacy once the drug enters the bloodstream.


Assuntos
Anti-Inflamatórios/química , Receptores Tipo II do Fator de Necrose Tumoral/química , Fator de Necrose Tumoral alfa/química , Anti-Inflamatórios/imunologia , Humanos , Interações Hidrofóbicas e Hidrofílicas , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/imunologia , Imunoglobulina G/química , Ligação Proteica , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/imunologia , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/imunologia
4.
J Clin Invest ; 131(16)2021 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-34185706

RESUMO

TNFR1 and TNFR2 have received prominent attention because of their dominance in the pathogenesis of inflammation and autoimmunity. TNFR1 has been extensively studied and primarily mediates inflammation. TNFR2 remains far less studied, although emerging evidence demonstrates that TNFR2 plays an antiinflammatory and immunoregulatory role in various conditions and diseases. Herein, we report that TNFR2 regulates macrophage polarization, a highly dynamic process controlled by largely unidentified intracellular regulators. Using biochemical copurification and mass spectrometry approaches, we isolated the signaling molecule 14-3-3ε as a component of TNFR2 complexes in response to progranulin stimulation in macrophages. In addition, 14-3-3ε was essential for TNFR2 signaling-mediated regulation of macrophage polarization and switch. Both global and myeloid-specific deletion of 14-3-3ε resulted in exacerbated inflammatory arthritis and counteracted the protective effects of progranulin-mediated TNFR2 activation against inflammation and autoimmunity. TNFR2/14-3-3ε signaled through PI3K/Akt/mTOR to restrict NF-κB activation while simultaneously stimulating C/EBPß activation, thereby instructing macrophage plasticity. Collectively, this study identifies 14-3-3ε as a previously unrecognized vital component of the TNFR2 receptor complex and provides new insights into the TNFR2 signaling, particularly its role in macrophage polarization with therapeutic implications for various inflammatory and autoimmune diseases with activation of the TNFR2/14-3-3ε antiinflammatory pathway.


Assuntos
Proteínas 14-3-3/imunologia , Macrófagos/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Proteínas 14-3-3/química , Proteínas 14-3-3/deficiência , Proteínas 14-3-3/metabolismo , Animais , Artrite Experimental/imunologia , Artrite Experimental/metabolismo , Artrite Experimental/patologia , Autoimunidade , Humanos , Inflamação/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Complexos Multiproteicos/química , Complexos Multiproteicos/imunologia , Complexos Multiproteicos/metabolismo , Progranulinas/imunologia , Progranulinas/metabolismo , Células RAW 264.7 , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Transdução de Sinais/imunologia
5.
Protein Expr Purif ; 182: 105857, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33639277

RESUMO

TNFR2 is aberrantly expressed on various cancer cells and highly immunosuppressive regulatory T cells (Tregs) accumulated in tumor microenvironment. As an oncoprotein and a stimulator of the immune checkpoint Tregs that promote cancer cell survival and tumor growth, TNFR2 is considered to be a prospective target for cancer immunotherapy with the blockers developed to simultaneously inhibit abundant TNFR2+ tumor-associated Tregs and directly kill TNFR2-expressing tumors. The soluble ectodomain of TNFR2 has also been successfully applied in clinical treatment for TNF-related autoimmune diseases. Research practices on these therapeutic strategies need recombinant protein of human soluble TNFR2 (hsTNFR2); however, mass production of such biologics using eukaryotic cells is generally high-cost in culture materials and growth conditions. This study aimed to establish an efficient methodology to prepare bioactive hsTNFR2 through a prokaryotic expression system. Recombinant vector pMCSG7-hsTNFR2 was constructed and the His-tagged fusion protein expressed in E. coli was enriched in inclusion bodies. Recombinant hsTNFR2 was denatured, refolded, and then purified by affinity chromatography, tag removal, ion-exchange chromatography and gel filtration chromatography. A protein yield of 8.4 mg per liter of bacterial culture liquid with a purity of over 97% was obtained. Purified hsTNFR2 exhibited strong affinity to human TNF-α with a KD of 10.5 nM, and inhibited TNF-α-induced cytotoxicity in L929 cells with an EC50 of 0.57 µg/ml. The biological activity assessed in vitro indicated that this soluble protein can be promisingly used in drug discovery for immunotherapy of TNFR2+ cancers and treatment of autoimmune diseases featured by TNF-α overload.


Assuntos
Escherichia coli , Expressão Gênica , Receptores Tipo II do Fator de Necrose Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Receptores Tipo II do Fator de Necrose Tumoral/biossíntese , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/isolamento & purificação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Solubilidade
6.
Methods Mol Biol ; 2248: 109-119, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33185871

RESUMO

TNFα/TNFR signaling plays a critical role in the pathogenesis of various inflammatory and autoimmune diseases, and anti-TNFα therapies have been accepted as the effective approaches for treating several autoimmune diseases. Progranulin (PGRN), a multi-faced growth factor-like molecule, directly binds to TNFR1 and TNFR2, particularly to the latter with higher affinity than TNFα. PGRN derivative Atsttrin is composed of three TNFR-binding domain of PGRN and exhibits even better therapeutic effects than PGRN in several inflammatory disease models, including collagen-induced arthritis. Herein we describe the detailed methodology of using (1) ELISA-based solid phase protein-protein interaction assay to demonstrate the direct binding of Atsttrin to TNFR2 and its inhibition of TNFα/TNFR2 interaction; and (2) tartrate-resistant acid phosphatase (TRAP) staining of in vitro osteoclastogenesis to reveal the cell-based anti-TNFα activity of Atsttrin. Using the protocol described here, the investigators should be able to reproducibly detect the physical inhibition of TNFα binding to TNFR and the functional inhibition of TNFα activity by Atsttrin and various kinds of TNF inhibitors.


Assuntos
Progranulinas/química , Receptores Tipo II do Fator de Necrose Tumoral/química , Fator de Necrose Tumoral alfa/química , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Progranulinas/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas/métodos , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/antagonistas & inibidores
7.
Artigo em Inglês | MEDLINE | ID: mdl-31965947

RESUMO

BACKGROUND: The sTNFRII-adiponectin fusion protein previously showed strong TNFα antagonistic activity. However, the fusion protein exists as mixture of different multimers. The aim of the present study was to characterize its active components. METHODS: In this study, the fusion protein was isolated and purified by Ni-NTA affinity and gel exclusion chromatography, and further identified by Coomassie staining and western blotting. The TNFα antagonistic and glucose uptake-promoting activities were determined in vitro. The glucose detection kit and 2- NBDG (2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-D-glucose) were used to measure their effects on glucose metabolism (including glucose consumption and glucose uptake in HepG2 and H9C2 cells). The effect of the fusion protein on glucose uptake was also examined in free fatty acid (FFA)- induced insulin resistance cell model. RESULTS: The sTNFRII-adiponectin fusion protein was found to exist in three forms: 250 kDa (hexamer), 130 kDa (trimer), and 60 kDa (monomer), with the final purity of 90.2%, 60.1%, and 81.6%, respectively. The fusion protein could effectively antagonize the killing effect of TNFα in L929 cells, and the multimer was found to be superior to the monomer. In addition, the fusion protein could increase glucose consumption without impacting the number of cells (HepG2, H9C2 cells) in a dosedependent manner. Mechanistically, glucose uptake was found to be enhanced by the translocation of GLUT4. However, it could not improve glucose uptake in the cell model of insulin resistance. CONCLUSION: In summary, the active components of the fusion protein are hexamers and trimers. The hexamer and trimer of sTNFRII-adiponectin fusion protein had both TNFα-antagonizing and glucose uptake-promoting activities, although neither of them could improve glucose uptake in the cell model of insulin resistance.


Assuntos
Adiponectina , Glucose/farmacocinética , Receptores Tipo II do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adiponectina/química , Adiponectina/metabolismo , Adiponectina/farmacologia , Animais , Células CHO , Metabolismo dos Carboidratos/efeitos dos fármacos , Linhagem Celular , Cricetulus , Células Hep G2 , Humanos , Resistência à Insulina , Multimerização Proteica , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/farmacologia , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos
8.
EBioMedicine ; 41: 556-570, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30826358

RESUMO

BACKGROUND: Osteoarthritis (OA) is a common degenerative disease, and tumor necrosis factor (TNF-α) is known to play a critical role in OA. Cortistatin (CST) is a neuropeptide discovered over 20  years ago, which plays a vital role in inflammatory reactions. However, it is unknown whether CST is involved in cartilage degeneration and OA development. METHODS: The interaction between CST and TNF-α receptors was investigated through Coimmunoprecipitation and Biotin-based solid-phase binding assay. Western blot, Real-time PCR, ELISA, immunofluorescence staining, nitrite production assay and DMMB assay of GAG were performed for the primary chondrocyte experiments. Surgically induced and spontaneous OA models were established and western blot, flow cytometry, Real-time PCR, ELISA, immunohistochemistry and fluorescence in vivo imaging were performed for in vivo experiments. FINDINGS: CST competitively bound to TNFR1 as well as TNFR2. CST suppressed proinflammatory function of TNF-α. Both spontaneous and surgically induced OA models indicated that deficiency of CST led to an accelerated OA-like phenotype, while exogenous CST attenuated OA development in vivo. Additionally, TNFR1- and TNFR2-knockout mice were used for analysis and indicated that TNFRs might be involved in the protective role of CST in OA. CST inhibited activation of the NF-κB signaling pathway in OA. INTERPRETATION: This study provides new insight into the pathogenesis and therapeutic strategy of cartilage degenerative diseases, including OA. FUND: The National Natural Science Foundation of China, the Natural Science Foundation of Shandong Province, Key Research and Development Projects of Shandong Province and the Cross-disciplinary Fund of Shandong University.


Assuntos
Neuropeptídeos/metabolismo , Osteoartrite/patologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Animais , Cartilagem Articular/citologia , Caspase 3/metabolismo , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Modelos Animais de Doenças , Humanos , Interleucina-1beta/sangue , Articulação do Joelho/patologia , Metaloproteinase 13 da Matriz , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Neuropeptídeos/genética , Neuropeptídeos/farmacologia , Neuropeptídeos/uso terapêutico , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Ligação Proteica , Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
9.
BMC Vet Res ; 15(1): 68, 2019 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-30819151

RESUMO

BACKGROUND: Refractory diseases, including bacterial infections, are causing huge economic losses in dairy farming. Despite efforts to prevent and treat those diseases in cattle, including the use of antimicrobials, it is not well controlled in the field. Several inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), play important roles in disease progression; thus, blocking these cytokines can attenuate the acute and sever inflammation and may be a novel strategy for treatment. However, biological drugs targeting inflammatory cytokines have not been used in cattle. Therefore, in this study, bovine sTNFR1 and sTNFR2 IgG1 Fc-fusion proteins (TNFR1-Ig and TNFR2-Ig) were produced, and their anti-inflammatory functions were analyzed in vitro, to develop decoy receptors for bovine TNF-α. RESULTS: Both TNFR1-Ig and TNFR2-Ig were shown to bind with TNF-α, and TNFR2-Ig showed higher affinity toward TNF-α than TNFR1-Ig. We next stimulated murine fibroblast-derived cells (L929 cells) with TNF-α to induce cell death and analyzed cell viability in the presence of TNFR-Ig proteins. Both TNFR1-Ig and TNFR2-Ig suppressed TNF-α-induced cell death, significantly improving cell viability. In addition, cell death induced by TNF-α was suppressed, even at low TNFR2-Ig concentrations, suggesting TNFR2-Ig has higher activity to suppress TNF-α functions than TNFR1-Ig. Finally, to examine TNFR2-Ig's anti-inflammatory, we cultured peripheral blood mononuclear cells from cattle with TNF-α in the presence of TNFR2-Ig and analyzed the gene expression and protein production of the inflammatory cytokines IL-1ß and TNF-α. TNFR2-Ig significantly reduced the gene expression and protein production of these cytokines. Our results suggest that TNFR2-Ig inhibits inflammatory cytokine kinetics by blocking TNF-α to transmembrane TNFR, thereby attenuating excessive inflammation induced by TNF-α. CONCLUSIONS: Collectively, the findings of this study demonstrated the potential of TNFR2-Ig as a novel therapeutic for inflammatory diseases, such as bovine clinical mastitis. Further investigation is required for future clinical application.


Assuntos
Morte Celular/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Receptores Chamariz do Fator de Necrose Tumoral/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Apoptose/efeitos dos fármacos , Bovinos , Linhagem Celular , Células Cultivadas , Citocinas/metabolismo , Fibroblastos , Expressão Gênica , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/farmacologia , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Leucócitos Mononucleares , Camundongos , Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo I de Fatores de Necrose Tumoral/farmacologia , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/farmacologia
10.
Fish Shellfish Immunol ; 86: 393-402, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30502465

RESUMO

Tumor necrosis factor-alpha (TNF-α) signals through two distinct cell surface receptors, TNFR1 and TNFR2 in mammals. In the present study, grass carp Tnfr2 (gcTnfr2) was isolated and characterized. Sequence alignment and phylogenetic analysis suggested that gcTnfr2 was a homolog of goldfish and zebrafish Tnfr2. Tissue distribution assay showed gctnfr2 transcripts were expressed in all examined tissues similar to gctnfr1. To functionally characterize the newly cloned molecule, gcTnfr2 was overexpressed in COS7 cell lines and it showed the ability to mediate the recombinant grass carp Tnf (rgcTnf)-α-triggered NF-κΒ activity and gcil1b promoter activity, clarifying its role in mediating Tnf-α signaling. The recombinant soluble form of gcTnfr2 (rgcsTnfr2) was prepared and it was able to interact with rgcTnf-α with higher affinity than that of rgcsTnfr1. Moreover, grass carp soluble Tnfr2 (gcsTnfr2) were detected in the culture medium of grass carp head kidney leukocytes (HKLs) and heat-inactivated A. hydrophila challenge significantly induced its production, indicating involvement of gcsTnfr2 in inflammation response. In agreement with this notion, rgcsTnfr2 effectively antagonized the effect of rgcTnf-α on il1b mRNA expression in HKLs, suggesting anti-Tnf-α property of gcsTnfr2. To strengthen the anti-inflammatory role of soluble Tnfr2, bacteria were injected intraperitoneally in grass carp followed by rgcsTnfr2. Hematoxylin-eosin (HE) staining of head kidney, spleen and intestine showed that rgcsTnfr2 could significantly improve infection-induced histopathological changes. These results functionally identified gcTnfr2 and its soluble form, particularly highlighting the role of gcsTnfr2 against Tnf-α-triggered inflammatory signaling. In this line, rgcsTnfr2 displayed anti-inflammatory potentiality during infection, thereby providing a powerful mediator of inflammation control in fish.


Assuntos
Doenças dos Peixes/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Inata/genética , Perciformes/genética , Perciformes/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Aeromonas hydrophila/fisiologia , Sequência de Aminoácidos , Animais , Proteínas de Peixes/química , Proteínas de Peixes/genética , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica/veterinária , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/veterinária , Inflamação/imunologia , Inflamação/veterinária , Filogenia , Receptores Tipo II do Fator de Necrose Tumoral/química , Alinhamento de Sequência/veterinária , Transdução de Sinais
11.
Cytokine ; 113: 340-346, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30352759

RESUMO

BACKGROUND: Inhibiting TNF-α is an effective therapy for inflammatory diseases such as rheumatoid arthritis. However, systemic, nondiscriminatory neutralization of TNF-α is associated with considerable adverse effects. METHODS: Here, we developed a trimeric chimeric TNF receptor by linking an N-terminal mouse Acrp30 trimerization domain and an MMP-2/9 substrate sequence to the mouse extracellular domain of TNF receptor 2 followed by a C-terminal mouse tetranectin coiled-coil domain (mouse Acrp-MMP-TNFR-Tn). RESULTS: Here, we show that the Acrp30 trimerization domain inhibited the binding activity of TNFR, possibly by closing the binding site of the trimeric receptor. Cleavage of the substrate sequence by MMP-9, an enzyme highly expressed in inflammatory sites, restored the binding activity of the mouse TNF receptor. We also constructed a recombinant human chimeric TNF receptor (human Acrp-MMP-TNFR-Tn) in which an MMP-13 substrate sequence was used to link the human Acrp and the human TNF receptor 2. Human Acrp-MMP-TNFR-Tn showed reduced binding activity, and MMP-13 digestion recovered its binding activity with TNF-α. CONCLUSION: Acrp-masked chimeric TNF receptors may be able to be used for inflammatory tissue-selective neutralization of TNF-α to reduce the adverse effects associated with systemic neutralization of TNF-α.


Assuntos
Adiponectina , Metaloproteinase 13 da Matriz , Metaloproteinase 2 da Matriz , Metaloproteinase 9 da Matriz , Multimerização Proteica , Receptores Tipo II do Fator de Necrose Tumoral , Proteínas Recombinantes de Fusão , Fator de Necrose Tumoral alfa , Adiponectina/química , Adiponectina/genética , Adiponectina/metabolismo , Animais , Linhagem Celular , Humanos , Metaloproteinase 13 da Matriz/química , Metaloproteinase 13 da Matriz/genética , Metaloproteinase 13 da Matriz/metabolismo , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/química , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Especificidade de Órgãos , Ligação Proteica , Domínios Proteicos , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/metabolismo
12.
Biomaterials ; 152: 24-36, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29080421

RESUMO

Gadolinium-containing fullerenol Gd@C82(OH)22 has demonstrated low-toxicity and highly therapeutic efficacy in inhibiting tumor growth and metastasis through new strategy of encaging cancer, however, little is known about the mechanisms how this nanoparticle regulates fibroblast cells to prison (instead of poison) cancer cells. Here, we report that Gd@C82(OH)22 promote the binding activity of tumor necrosis factor (TNFα) to tumor necrosis factor receptors 2 (TNFR2), activate TNFR2/p38 MAPK signaling pathway to increase cellular collagen expression in fibrosarcoma cells and human primary lung cancer associated fibroblasts isolated from patients. We also employ molecular dynamics simulations to study the atomic-scale mechanisms that dictate how Gd@C82(OH)22 mediates interactions between TNFα and TNFRs. Our data suggest that Gd@C82(OH)22 might enhance the association between TNFα and TNFR2 through a "bridge-like" mode of interaction; by contrast, the fullerenol appears to inhibit TNFα-TNFR1 association by binding to two of the receptor's cysteine-rich domains. In concert, our results uncover a sequential, systemic process by which Gd@C82(OH)22 acts to prison tumor cells, providing new insights into principles of designs of cancer therapeutics.


Assuntos
Antineoplásicos/química , Colágeno/metabolismo , Fulerenos/química , Gadolínio/química , Nanopartículas Metálicas/química , Animais , Linhagem Celular Tumoral , Fibrossarcoma/patologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Simulação de Dinâmica Molecular , Tamanho da Partícula , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Propriedades de Superfície , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Circ Res ; 121(4): 392-410, 2017 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-28637784

RESUMO

RATIONALE: Mitochondria are important cellular organelles and play essential roles in maintaining cell structure and function. Emerging evidence indicates that in addition to having proinflammatory and proapoptotic effects, TNFα (tumor necrosis factor α) can, under certain circumstances, promote improvements in mitochondrial integrity and function, phenomena that can be ascribed to the existence of TNFR2 (TNFα receptor 2). OBJECTIVE: The present study aimed to investigate whether and how TNFR2 activation mediates the effects of TNFα on mitochondria. METHODS AND RESULTS: Freshly isolated neonatal mouse cardiac myocytes treated with shRNA targeting TNFR1 were used to study the effects of TNFR2 activation on mitochondrial function. Neonatal mouse cardiac myocytes exhibited increases in mitochondrial fusion, a change that was associated with increases in mitochondrial membrane potential, intracellular ATP levels, and oxygen consumption capacity. Importantly, TNFR2 activation-induced increases in OPA1 (optic atrophy 1) protein expression were responsible for the above enhancements, and these changes could be attenuated using siRNA targeting OPA1. Moreover, both Stat3 and RelA bound to the promoter region of OPA1 and their interactions synergistically upregulated OPA1 expression at the transcriptional level. Stat3 acetylation at lysine 370 or lysine 383 played a key role in the ability of Stat3 to form a supercomplex with RelA. Meanwhile, p300 modulated Stat3 acetylation in HEK293T (human embryonic kidney 293T) cells, and p300-mediated Stat3/RelA interactions played an indispensable role in OPA1 upregulation. Finally, TNFR2 activation exerted beneficial effects on OPA1 expression in an in vivo transverse aortic constriction model, whereby TNFR1-knockout mice exhibited better outcomes than in mice with both TNFR1 and TNFR2 knocked out. CONCLUSIONS: TNFR2 activation protects cardiac myocytes against stress by upregulating OPA1 expression. This process was facilitated by p300-mediated Stat3 acetylation and Stat3/RelA interactions, leading to improvements in mitochondrial morphology and function.


Assuntos
GTP Fosfo-Hidrolases/biossíntese , Dinâmica Mitocondrial/fisiologia , NF-kappa B/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Animais Recém-Nascidos , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Células Cultivadas , GTP Fosfo-Hidrolases/química , GTP Fosfo-Hidrolases/genética , Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miócitos Cardíacos/metabolismo , NF-kappa B/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores Tipo II do Fator de Necrose Tumoral/química , Fator de Transcrição STAT3/química
14.
FEBS Lett ; 591(8): 1187-1195, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28337757

RESUMO

Exploiting regulatory T cells (Tregs) to control aberrant immune reactions is a promising therapeutic approach, but is hampered by their relative paucity. In mice, activation of death receptor 3 (DR3), a member of the TNF-receptor superfamily (TNFRSF), increases Treg frequency and efficiently controls exuberant immune activation. For human Tregs, neither DR3 expression nor potential functions have been described. Here, we show that human Tregs express DR3 and demonstrate DR3-mediated activation of p38, ERK, and NFκB. DR3 stimulation enhances Treg expansion ex vivo while retaining their suppressive capacity. In summary, our results establish a functional role for DR3 signaling in human Tregs and could potentially help to tailor Treg-based therapies.


Assuntos
Sistema de Sinalização das MAP Quinases , Subunidade p52 de NF-kappa B/agonistas , Membro 25 de Receptores de Fatores de Necrose Tumoral/agonistas , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Regulação para Cima , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Humanos , Imunossupressores/farmacologia , Ligantes , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Subunidade p52 de NF-kappa B/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Fosforilação/efeitos dos fármacos , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Membro 25 de Receptores de Fatores de Necrose Tumoral/química , Membro 25 de Receptores de Fatores de Necrose Tumoral/genética , Membro 25 de Receptores de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/química , Membro 15 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Regulação para Cima/efeitos dos fármacos
15.
Vitam Horm ; 104: 57-87, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28215307

RESUMO

Although p75 neurotrophin receptor (p75NTR) was the founding member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), it is an atypical TNFRSF protein. p75NTR like TNF-R1 and Fas-R contain an extracellular domain with four cysteine-rich domains (CRD) and a death domain (DD) in the intracellular region. While TNFRSF proteins are activated by trimeric TNFSF ligands, p75NTR forms dimers activated by dimeric neurotrophins that are structurally unrelated to TNFSF proteins. In addition, although p75NTR shares with other members the interaction with the TNF receptor-associated factors to activate the NF-κB and cell death pathways, p75NTR does not interact with the DD-containing proteins FADD, TRADD, or MyD88. By contrast, the DD of p75NTR is able to recruit several protein interactors via a full catalog of DD interactions not described before in the TNFRSF. p75-DD forms homotypic symmetrical DD-DD complexes with itself and with the related p45-DD; forms heterotypic DD-CARD interactions with the RIP2-CARD domain, and forms a new interaction between a DD and RhoGDI. All these features, in addition to its promiscuous interactions with several ligands and coreceptors, its processing by α- and γ-secretases, the dimeric nature of its transmembrane domain and its "special" juxtamembrane region, make p75NTR a truly stranger in the TNFR superfamily. In this chapter, I will summarize the known structural aspects of p75NTR and I will analyze from a structural point of view, the similitudes and differences between p75NTR and the other members of the TNFRSF.


Assuntos
Modelos Moleculares , Receptor de Fator de Crescimento Neural/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/química , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Sítios de Ligação , Dimerização , Humanos , Ligantes , Fatores de Crescimento Neural/química , Fatores de Crescimento Neural/metabolismo , Conformação Proteica , Domínios e Motivos de Interação entre Proteínas , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Receptor de Fator de Crescimento Neural/agonistas , Receptor de Fator de Crescimento Neural/química , Receptor de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/química , Receptores de Fator de Crescimento Neural/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/agonistas , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética
16.
J Diabetes Complications ; 30(5): 852-7, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27068267

RESUMO

AIMS: The tumor necrosis factor α (TNF-α) family of inflammatory molecules plays a crucial role in the pathogenesis of type 2 diabetes mellitus (DM2) complications. TNF-α soluble receptors 1 (sTNFR1) and 2 (sTNFR2) have been associated with chronic kidney disease in DM2 patients. This cross-sectional study intended to determine serum concentrations of sTNFR1 and sTNFR2 in Colombian patients and correlated them with various clinical variables, especially kidney function. METHODS: 92 Colombian patients with DM2 were recruited. Anthropometric variables, glycemic control parameters, lipid profile and renal function were assessed for each patient. Levels of sTNFR1 and sTNFR2 were determined using ELISA. Patients were stratified in two groups according to reduced estimated glomerular filtration rate (eGFR) (<60ml/min/1.73m(2)) and normal eGFR (≥60ml/min/1.73m(2)). RESULTS: Significantly elevated levels of sTNFR1 and sTNFR2 were observed in the diminished versus normal eGFR group. Also, significant differences were noticed between both groups in haemoglobin A1c (HbA1c) values, percentage of hypertensive subjects treated with angiotensin receptor blocker (ARB) and subjects treated with metformin. No differences were observed regarding body mass index (BMI), albuminuria and lipid profile. Multivariable linear regression analysis revealed that sTNFR1 alone showed a significant association with low eGFR (p=0.009). However, after adjusting for age, the association weakens. Moreover, sTNFR1 and sTNFR2 showed a linear negative correlation with eGFR (r=-0.448, p<0.001 and r=-0.376, p<0.001, respectively). A positive correlation was also seen between sTNFR1 and HbA1c, whereas a negative correlation between both sTNFRs and high-density lipoprotein (HDL) cholesterol was found. CONCLUSION: Elevated levels of sTNFRs, especially sTNFR1, are associated with loss of kidney function in Hispanic patients with DM2. Future studies should focus on social and genetic determinants of inflammation and their association with CKD in this ethnicity.


Assuntos
Diabetes Mellitus Tipo 2/complicações , Nefropatias Diabéticas/sangue , Falência Renal Crônica/complicações , Rim/fisiopatologia , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Insuficiência Renal/complicações , Idoso , Biomarcadores/sangue , Colômbia/epidemiologia , Estudos Transversais , Nefropatias Diabéticas/epidemiologia , Nefropatias Diabéticas/fisiopatologia , Feminino , Taxa de Filtração Glomerular , Hemoglobinas Glicadas/análise , Hospitais Universitários , Humanos , Falência Renal Crônica/sangue , Falência Renal Crônica/epidemiologia , Falência Renal Crônica/fisiopatologia , Masculino , Pessoa de Meia-Idade , Receptores Tipo I de Fatores de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo II do Fator de Necrose Tumoral/química , Insuficiência Renal/sangue , Insuficiência Renal/epidemiologia , Insuficiência Renal/fisiopatologia , Fatores de Risco , Índice de Gravidade de Doença , Solubilidade
17.
Sci Rep ; 6: 20150, 2016 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-26841833

RESUMO

A considerable proportion of patients with rheumatoid arthritis (RA) do not respond to monospecific agents. The purpose of our study was to generate a hybrid form of biologics, targeting tumor-necrosis factor alpha (TNFα) and interleukin-6 receptor (IL-6R), and determine its anti-arthritic properties in vitro and in vivo. A novel dual target-directed agent (DTA(A7/sTNFR2)) was generated by conjugating soluble TNF receptor 2 (sTNFR2) to the Fc region of A7, a new anti-IL-6R antibody obtained by screening the phage display human antibody library. DTA(A7/sTNFR2) inhibited the proliferation and migration of fibroblast-like synoviocytes from patients with RA (RA-FLS) more efficiently than single target-directed agents. DTA(A7/sTNFR2) also blocked osteoclastogenesis from bone marrow cells. The arthritis severity scores of the experimental arthritis mice with DTA(A7/sTNFR2) tended to be lower than those of mice with IgG, A7, or sTNFR2. Histological data suggested that DTA(A7/sTNFR2) is more efficient than single-target drugs in preventing joint destruction and bone loss. These results were confirmed in vivo using the minicircle system. Taken together, the results show that DTA(A7/sTNFR2) may be a promising therapeutic agent for the treatment of RA.


Assuntos
Anticorpos/metabolismo , Artrite Experimental/patologia , Fragmentos Fc das Imunoglobulinas/metabolismo , Receptores de Interleucina-6/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Anticorpos/genética , Artrite Experimental/tratamento farmacológico , Artrite Experimental/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Células da Medula Óssea/citologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Fragmentos Fc das Imunoglobulinas/genética , Camundongos , Camundongos Endogâmicos DBA , Osteogênese/efeitos dos fármacos , Biblioteca de Peptídeos , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/genética , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Sinoviócitos/citologia , Sinoviócitos/metabolismo
18.
J Biol Chem ; 290(26): 15973-84, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25940088

RESUMO

The blockade of tumor necrosis factor (TNF) by etanercept, a soluble version of the human TNF receptor 2 (hTNFR2), is a well established strategy to inhibit adverse TNF-mediated inflammatory responses in the clinic. A similar strategy is employed by poxviruses, encoding four viral TNF decoy receptor homologues (vTNFRs) named cytokine response modifier B (CrmB), CrmC, CrmD, and CrmE. These vTNFRs are differentially expressed by poxviral species, suggesting distinct immunomodulatory properties. Whereas the human variola virus and mouse ectromelia virus encode one vTNFR, the broad host range cowpox virus encodes all vTNFRs. We report the first comprehensive study of the functional and binding properties of these four vTNFRs, providing an explanation for their expression profile among different poxviruses. In addition, the vTNFRs activities were compared with the hTNFR2 used in the clinic. Interestingly, CrmB from variola virus, the causative agent of smallpox, is the most potent TNFR of those tested here including hTNFR2. Furthermore, we demonstrate a new immunomodulatory activity of vTNFRs, showing that CrmB and CrmD also inhibit the activity of lymphotoxin ß. Similarly, we report for the first time that the hTNFR2 blocks the biological activity of lymphotoxin ß. The characterization of vTNFRs optimized during virus-host evolution to modulate the host immune response provides relevant information about their potential role in pathogenesis and may be used to improve anti-inflammatory therapies based on soluble decoy TNFRs.


Assuntos
Vírus da Varíola Bovina/metabolismo , Poxviridae/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Chamariz do Fator de Necrose Tumoral/química , Receptores Chamariz do Fator de Necrose Tumoral/metabolismo , Proteínas Virais/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Animais , Vírus da Varíola Bovina/química , Vírus da Varíola Bovina/genética , Humanos , Linfotoxina-beta/metabolismo , Camundongos , Dados de Sequência Molecular , Poxviridae/química , Poxviridae/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Alinhamento de Sequência , Receptores Chamariz do Fator de Necrose Tumoral/genética , Fatores de Necrose Tumoral/metabolismo , Proteínas Virais/genética
19.
J Biol Chem ; 290(2): 861-71, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25378394

RESUMO

TNF receptor 2 (TNFR2) exerts diverse roles in the pathogenesis of inflammatory and autoimmune diseases. Here, we report that TNFR2 but not TNFR1 forms a heteromer with interleukin-17 receptor D (IL-17RD), also named Sef, to activate NF-κB signaling. TNFR2 associates with IL-17RD, leading to mutual receptor aggregation and TRAF2 recruitment, which further activate the downstream cascade of NF-κB signaling. Depletion of IL-17RD impaired TNFR2-mediated activation of NF-κB signaling. Importantly, IL-17RD was markedly increased in renal tubular epithelial cells in nephritis rats, and a strong interaction of TNFR2 and IL-17RD was observed in the renal epithelia. The IL-17RD·TNFR2 complex in activation of NF-κB may explain the role of TNFR2 in inflammatory diseases including nephritis.


Assuntos
Inflamação/metabolismo , NF-kappa B/metabolismo , Nefrite/metabolismo , Receptores de Interleucina-17/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Animais , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Inflamação/etiologia , Inflamação/patologia , Túbulos Renais Distais/metabolismo , Túbulos Renais Distais/patologia , NF-kappa B/genética , Nefrite/etiologia , Nefrite/patologia , Multimerização Proteica , Ratos , Receptores de Interleucina-17/química , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/química , Transdução de Sinais/genética , Ativação Transcricional/genética
20.
Arch Dermatol Res ; 307(5): 405-12, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25537528

RESUMO

Genetic factors are involved not only in the overall risk of suffering psoriasis, but also in their clinical characteristics and eventually in drug outcome. Biological therapies have dramatically improved the prognosis of Psoriasis. However, these treatments are very expensive and patients often exhibit a heterogeneous response that could be partially attributed to their genetic background. Thus, the research for genetic markers in psoriatic patients that could predict a poor response to biological therapies is an important issue. Our aim was to evaluate the effect of DNA variants at the "TNFα pathway" that could affect the risk of developing Psoriasis or the response to biological therapies among these patients. The genetic association study included a total of 518 Psoriatic patients and 480 healthy controls. Ninety of these patients received biological treatment and based on the change in the PASI score after 24 weeks were classified as good (PASI score ≥75%), intermediate (PASI 50-75), and non-responders (PASI <50). Next generation sequencing (NGS) with semiconductor-array technology was used to identify the nucleotide variants in the TNF α, TNFRSF1A and TNFRSF1B, and we only found three missense amino acid changes, all in TNFRSF1B. Interestingly, we found a significantly higher frequency of rs1061622 G carriers among CW6-positive patients (p = 0.004; OR = 1.69, 95% CI = 1.18-2.41). Allele G (p.196R) carriers were significantly more frequent in the non-responder group (56%) (p = 0.05). In conclusion, we report a significant association between the TNFRSF1B p.M196R variant and the risk for psoriasis and the response to treatment with anti-TNF or anti-Il-12/Il-23. The genotyping of this polymorphism could help to optimize the treatment by identifying patients with a likely poor response to biological drugs.


Assuntos
Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Psoríase/tratamento farmacológico , Psoríase/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Adulto , Terapia Biológica/efeitos adversos , Resistência a Medicamentos/genética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Farmacogenética/métodos , Receptores Tipo II do Fator de Necrose Tumoral/química , Análise de Sequência de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...