Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Protein Expr Purif ; 182: 105857, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33639277

RESUMEN

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.


Asunto(s)
Escherichia coli , Expresión Génica , Receptores Tipo II del Factor de Necrosis Tumoral , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/química , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/aislamiento & purificación , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Solubilidad
2.
Medicine (Baltimore) ; 100(7): e24321, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33607766

RESUMEN

ABSTRACT: Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 induces severe infection, and it is responsible for a worldwide disease outbreak starting in late 2019. Currently, there are no effective medications against coronavirus. In the present study, we utilized a holistic bioinformatics approach to study gene signatures of SARS-CoV- and SARS-CoV-2-infected Calu-3 lung adenocarcinoma cells. Through the Gene Ontology platform, we determined that several cytokine genes were up-regulated after SARS-CoV-2 infection, including TNF, IL6, CSF2, IFNL1, IL-17C, CXCL10, and CXCL11. Differentially regulated pathways were detected by the Kyoto Encyclopedia of Genes and Genomes, gene ontology, and Hallmark platform, including chemokines, cytokines, cytokine receptors, cytokine metabolism, inflammation, immune responses, and cellular responses to the virus. A Venn diagram was utilized to illustrate common overlapping genes from SARS-CoV- and SARS-CoV-2-infected datasets. An Ingenuity pathway analysis discovered an enrichment of tumor necrosis factor- (TNF-) and interleukin (IL)-17-related signaling in a gene set enrichment analysis. Downstream networks were predicted by the Database for Annotation, Visualization, and Integrated Discovery platform also revealed that TNF and TNF receptor 2 signaling elicited leukocyte recruitment, activation, and survival of host cells after coronavirus infection. Our discovery provides essential evidence for transcript regulation and downstream signaling of SARS-CoV and SARS-CoV-2 infection.


Asunto(s)
COVID-19/genética , COVID-19/inmunología , Quimiocinas/biosíntesis , Citocinas/biosíntesis , Mediadores de Inflamación/metabolismo , Línea Celular Tumoral , Quimiocinas/genética , Citocinas/genética , Perfilación de la Expresión Génica , Ontología de Genes , Interacciones Huésped-Patógeno , Humanos , Interleucina-17/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , SARS-CoV-2 , Factor de Necrosis Tumoral alfa/biosíntesis , Regulación hacia Arriba
3.
Int J Mol Sci ; 21(9)2020 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397546

RESUMEN

Around a 20-30% of inflammatory bowel disease (IBD) patients are diagnosed before they are 18 years old. Anti-TNF drugs can induce and maintain remission in IBD, however, up to 30% of patients do not respond. The aim of the work was to identify markers that would predict an early response to anti-TNF drugs in pediatric patients with IBD. The study population included 43 patients aged <18 years with IBD who started treatment with infliximab or adalimumab. Patients were classified into primary responders (n = 27) and non-responders to anti-TNF therapy (n = 6). Response to treatment could not be analyzed in 10 patients. Response was defined as a decrease in over 15 points in the disease activity indexes from week 0 to week 10 of infliximab treatment or from week 0 to week 26 of adalimumab treatment. The expression profiles of nine genes in total RNA isolated from the whole-blood of pediatric IBD patients taken before biologic administration and after 2 weeks were analyzed using qPCR and the 2-∆∆Ct method. Before initiation and after 2 weeks of treatment the expression of SMAD7 was decreased in patients who were considered as non-responders (p value < 0.05). Changes in expression were also observed for TLR2 at T0 and T2, although that did not reach the level of statistical significance. In addition, the expression of DEFA5 decreased 1.75-fold during the first 2 weeks of anti-TNF treatment in responders, whereas no changes were observed in non-responders. Expression of the SMAD7 gene is a pharmacogenomic biomarker of early response to anti-TNF agents in pediatric IBD. TLR2 and DEFA5 need to be validated in larger studies.


Asunto(s)
Adalimumab/farmacología , Antiinflamatorios/farmacología , Antirreumáticos/farmacología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Infliximab/farmacología , Transcriptoma/efectos de los fármacos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Adalimumab/uso terapéutico , Adolescente , Antiinflamatorios/uso terapéutico , Antirreumáticos/uso terapéutico , Niño , Preescolar , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lactante , Enfermedades Inflamatorias del Intestino/sangre , Enfermedades Inflamatorias del Intestino/genética , Infliximab/uso terapéutico , Masculino , ARN Mensajero/biosíntesis , ARN Mensajero/sangre , ARN Mensajero/genética , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Proteína smad7/biosíntesis , Proteína smad7/genética , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/genética , Resultado del Tratamiento , Receptor Activador Expresado en Células Mieloides 1/biosíntesis , Receptor Activador Expresado en Células Mieloides 1/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , alfa-Defensinas/biosíntesis , alfa-Defensinas/genética
4.
Environ Pollut ; 244: 486-494, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30366296

RESUMEN

Environmental estrogens are capable of interfering with the spermatogenesis and fertility of fish. However in natural waters, these chemicals are more likely to occur as a combination rather than a single stressor. Whether and how the mixture of xenoestrogens with environmental relevant concentrations may affect fish spermatogenesis remains largely unknown. In this study, male zebrafish adults were administered to 17alpha-ethinylestradiol (EE2) and a mixture of xenoestrogens (Mix (E2, EE2, DES, 4-t-OP, 4-NP and BPA)), with the estrogenic potency equivalent to EE2. After a 60-day exposures, elevated mRNA levels of vitellogenin 1 (vtg1) and estrogen receptor 1 (esr1) in the liver of fish in both treated groups were observed. Moreover, the plasma level of E2 declined significantly in the Mix group and the ratio of 11-KT/E2 was significantly elevated in both treated groups. Consistently, the mRNA level of P450 side-chain cleavage (scc) in the EE2 group and ovarian type aromatase (cyp19a1a) in the Mix group was significantly suppressed. In addition, decreased gonadosomatic index and sperm count in the fish of Mix group were present. Furthermore, increased number of the proliferating germ cells (such as spermatogonia and spermatocytes) was observed in the fish of both groups, suggesting a stimulated germ cell proliferation and meiosis. Accordingly, both exposures significantly up-regulated the mRNA levels of genes in mitosis (cyclinb1) and meiosis (cyp26a1 in EE2 group, aldh1a2, cyp26a1, sycp3 and spo11 in Mix). In addition, decreased number of spermatozoa and increased number of TUNEL-positive signals were present in the testis of fish in the Mix group, indicating an enhanced apoptosis. Further analyses demonstrated the significant elevated expressions of tnfrsf1a and the ratio of tnfrsf1a/tnfrsf1b in the Mix group, suggesting an elevated apoptosis in the testis of fish in the Mix group via extrinsic pathway. The present study greatly extends our understanding of the underlying mechanisms of the reproductive toxicity of xenoestrogens on fish.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Estrógenos/sangre , Etinilestradiol/toxicidad , Meiosis/efectos de los fármacos , Espermatogénesis/fisiología , Espermatozoides/citología , Animales , Exposición a Riesgos Ambientales , Receptor alfa de Estrógeno/metabolismo , Femenino , Hormonas Esteroides Gonadales , Masculino , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Reproducción/fisiología , Ácido Retinoico 4-Hidroxilasa/biosíntesis , Recuento de Espermatozoides , Espermatogonias/citología , Testículo/metabolismo , Vitelogeninas/metabolismo , Pez Cebra
5.
Cell Physiol Biochem ; 47(6): 2307-2318, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29975930

RESUMEN

BACKGROUND/AIMS: Plastrum testudinis extracts (PTE) show osteoprotective effects on glucocorticoid-induced osteoporosis in vivo and in vitro. However, the underlying molecular mechanism of PTE in promoting osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is unclear. METHODS: BMSC proliferation was investigated using the Cell Counting Kit-8 assay. BMSC differentiation and osteogenic mineralization were assayed using alkaline phosphatase and Alizarin red staining, respectively. The mRNA expression levels of Let-7f-5p, Tnfr2, Traf2, Pi3k, Akt, ß-catenin, Gsk3ß, Runx2, and Ocn were measured using real time quantitative polymerase chain reaction. Protein levels of TNFR2, TRAF2, p-PI3K, p-AKT, p-ß-CATENIN, and p-GSK3ß were analyzed by western blotting. The functional relationship of Let-7f-5p and Tnfr2 was determined by luciferase reporter assays. RESULTS: The optimum concentration for PTE was 30 µg/ml. PTE significantly promoted BMSC osteogenic differentiation and mineralization after 7 and 14 days in culture, respectively. The combination of PTE and osteogenic induction exhibited significant synergy. PTE upregulated Let-7f-5p, ß-catenin, Runx2, and Ocn mRNA expression, and downregulated Tnfr2, Traf2, Pi3k, Akt, and Gsk3ß mRNA expression. PTE inhibited TNFR2, TRAF2, and p-ß-CATENIN protein expression, and promoted p-PI3K, p-AKT, and p-GSK3ß protein expression. In addition, Tnfr2 was a functional target of Let-7f-5p in 293T cells. CONCLUSIONS: Our results suggested that PTE may promote BMSC proliferation and osteogenic differentiation via a mechanism associated with the regulation of Let-7f-5p and the TNFR2/PI3K/AKT signaling pathway.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , MicroARNs/biosíntesis , Osteogénesis/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/biosíntesis , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Transducción de Señal/efectos de los fármacos , Extractos de Tejidos/farmacología , Animales , Células de la Médula Ósea/citología , Femenino , Células Madre Mesenquimatosas/citología , Ratas , Ratas Sprague-Dawley
6.
Clin Exp Med ; 18(4): 547-554, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29802567

RESUMEN

The aims of the study were to compare the levels of tumor necrosis factor alpha (TNF-α) and its soluble type I (sTNF-R1) and type II (sTNF-R2) receptors detected in intracystic liquid and serum from benign and malignant ovarian neoplasms and to relate them to prognostic factors in epithelial ovarian cancer. The patients were divided into benign ovarian neoplasms (n = 46) and malignant ovarian neoplasms (n = 17). The serum and intracystic samples were collected before and during surgery for ovarian cyst, respectively. The levels of TNF-α, sTNF-R1, and sTNF-R2 were measured using ELISA. Results were compared with the Mann-Whitney test. Concentration of sTNF-R2 in the intracystic samples collected from the malignant neoplasia was significantly higher than that of the benign neoplasias (p = 0.02). Higher intracystic levels of sTNF-R2 exhibited a significant association with tumor differentiation grades 2 and 3 (p = 0.0087). There was no statistical significance in relation to serum levels. Tumor microenvironment levels of sTNF-R2 may represent a factor of poor prognosis in epithelial ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/metabolismo , Neoplasias Ováricas/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Microambiente Tumoral , Factor de Necrosis Tumoral alfa/biosíntesis , Carcinoma Epitelial de Ovario/diagnóstico , Femenino , Humanos , Neoplasias Ováricas/diagnóstico , Pronóstico
7.
Neurochem Int ; 118: 292-303, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29777731

RESUMEN

Tumor Necrosis Factor-α (TNF-α) is a proinflammatory cytokine implicated in neuronal damage in response to cerebral ischemia. Ischemic preconditioning (IPC) provides neuroprotection against a subsequent severer or longer transient ischemia by ischemic tolerance. Here, we focused on the role of TNF-α in IPC-mediated neuroprotection against neuronal death following a subsequent longer transient cerebral ischemia (TCI). Gerbils used in this study were randomly assigned to eight groups; sham group, TCI operated group, IPC plus (+) sham group, IPC + TCI operated group, sham + etanercept (an inhibitor of TNF-a) group, TCI + etanercept group, IPC + sham + etanercept group, and IPC + TCI + etanercept group. IPC was induced by a 2-min sublethal transient ischemia, which was operated 1 day prior to a longer (5-min) TCI. A significant death of neurons was found in the stratum pyramidale (SP) in the CA1 area (CA1) of the hippocampus 5 days after TCI; however, IPC protected SP neurons from TCI. We found that TNF-α immunoreactivity was significantly increased in CA1 pyramidal neurons in the TCI and IPC + TCI groups compared to the sham group. TNF-R1 expression in CA1 pyramidal neurons of the TCI group was also increased 1 and 2 days after TCI; however, in the IPC + TCI group, TNF-R1 expression was significantly lower than that in the TCI group. On the other hand, we did not detect TNF-R2 immunoreactivity in CA1 pyramidal neurons 1 and 2 days after TCI; meanwhile, in the IPC + TCI group, TNF-R2 expression was significantly increased compared to TNF-R2 expression at 1 and 2 days after TCI. In addition, in this group, TNF-R2 was newly expressed in pericytes, which are important cells in the blood brain barrier, from 1 day after TCI. When we treated etanercept to the IPC + TCI group, IPC-induced neuroprotection was significantly weakened. In brief, this study indicates that IPC confers neuroprotection against TCI by TNF-α signaling through TNF-R2 and suggests that the enhancement of TNF-R2 expression by IPC may be a legitimate strategy for a therapeutic intervention of TCI.


Asunto(s)
Hipocampo/metabolismo , Ataque Isquémico Transitorio/metabolismo , Precondicionamiento Isquémico/métodos , Neuroprotección/fisiología , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Animales , Gerbillinae , Hipocampo/patología , Ataque Isquémico Transitorio/patología , Masculino , Factores de Tiempo
8.
Neurosci Res ; 137: 36-42, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29481885

RESUMEN

1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine (MPTP) -induced neuroinflammation and its impact in hippocampus remain elusive till date. Our present study includes the time dependent changes of inflammatory molecules in mouse hippocampus during MPTP treatment. MPTP treatment increased level of TNF-α, enhanced expression of TNFR2 along with PI3 kinase (PI3K) induced phosphorylation of Akt resulting in persistent nuclear factor-κB (NF-κB) activation. The expressions gradually increased from Day1 post-MPTP treatment, maximally at Day3 post-treatment. MPTP induced translocation of p65 and p52, two subunits of NF-κB family, to nucleus where they had been found to dimerize. Therefore, MPTP induced TNF-α signaling through TNFR2 mediated pathway and recruited p65-p52 dimer in hippocampal nucleus which is reported to have protective effect on hippocampal neurons indicated by unchanged neuronal count in hippocampus in treated groups with respect to control. Our finding suggests that this unique NF-κB dimer plays some role in providing inherent protection to hippocampus during MPTP-treatment.


Asunto(s)
1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , FN-kappa B/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Proteínas de Unión al ADN , Hipocampo/patología , Ratones , FN-kappa B/biosíntesis , Subunidad p52 de NF-kappa B/metabolismo , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo
9.
J Musculoskelet Neuronal Interact ; 17(3): 226-236, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860425

RESUMEN

BACKGROUND: The peritendinous connective tissues can have importance in chronic tendon pain. Recently cytokine TNF-α has been suggested to be involved in tendinopathic processes. It is not known how TNF-α and its receptors TNFR1 and TNFR2 are expressed in peritendinous tissues. METHODS: The objective for this study was to immunohistochemically evaluate the expression patterns of these in the peritendinous tissue located between the plantaris and Achilles tendons and the one located superficially to the extensor origin at the elbow region for patients with tendinopathy/tennis elbow. RESULTS: The nerve fascicles were of two types, one type being homogenously stained for the nerve markers ßIII-tubulin and neurofilament and the other showing deficits for these suggesting features of axonal damage. Much more distinct TNFR1/TNFR2 immunoreactions were seen for the latter nerve fascicles. TNFR1 was seen in axons, TNFR2 mainly in Schwann cells. TNFR1 and particularly TNFR2 were seen in walls of parts of blood vessels. The dispersed cells showed frequently TNFR1 and TNFR2 immunoreactivity. DISCUSSION: These findings suggest that TNF-α can be related to degenerative events but also attempts for healing concerning the nerve structures. The marked expression of the TNF-α system in the peritendinous tissue suggests an impact of TNF-α in tendinopathy/tennis elbow.


Asunto(s)
Tejido Conectivo/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Codo de Tenista/metabolismo , Adulto , Axones/patología , Tejido Conectivo/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nervios Periféricos , Codo de Tenista/patología
10.
PLoS Genet ; 13(4): e1006712, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28379965

RESUMEN

Somatosensory information from the periphery is routed to the spinal cord through centrally-projecting sensory axons that cross into the central nervous system (CNS) via the dorsal root entry zone (DREZ). The glial cells that ensheath these axons ensure rapid propagation of this information. Despite the importance of this glial-axon arrangement, how this afferent nerve is assembled during development is unknown. Using in vivo, time-lapse imaging we show that as centrally-projecting pioneer axons from dorsal root ganglia (DRG) enter the spinal cord, they initiate expression of the cytokine TNFalpha. This induction coincides with ensheathment of these axons by associated glia via a TNF receptor 2 (TNFR2)-mediated process. This work identifies a signaling cascade that mediates peripheral glial-axon interactions and it functions to ensure that DRG afferent projections are ensheathed after pioneer axons complete their navigation, which promotes efficient somatosensory neural function.


Asunto(s)
Neuroglía/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Raíces Nerviosas Espinales/metabolismo , Factor de Necrosis Tumoral alfa/genética , Animales , Astrocitos/metabolismo , Axones/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Ganglios Espinales , Regulación del Desarrollo de la Expresión Génica , Ratones , Neuroglía/citología , Neuronas Aferentes/metabolismo , Sistema Nervioso Periférico/crecimiento & desarrollo , Sistema Nervioso Periférico/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Transducción de Señal , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Raíces Nerviosas Espinales/crecimiento & desarrollo , Factor de Necrosis Tumoral alfa/biosíntesis , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
11.
Eur J Pharmacol ; 771: 48-55, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26688568

RESUMEN

Ginsenoside metabolite compound K (CK), metabolite of the ginsenoside, is considered to exert numerous pharmacological efficacies of ginsenoside, including anti-inflammation and immunoregulatory effects. Rheumatoid arthritis (RA) is a multi-systemic autoimmune disease characterized by hyperplastic synovial membrane and systemic inflammation, which ultimately lead to progressive destructive inflammatory arthropathy. To evaluate the potential joint-protective effects of CK and the underlying mechanism, adjuvant arthritis (AA) was induced by complete Freund's adjuvant in rats. After the onset of arthritis, The effect of CK on AA rats was evaluated by histopathology of the joint. The proliferation of fibroblast-like synoviocyte(FLS) was assayed by the Cell Counting Kit-8.The migration of FLS was assayed by transwell migration assay. Cytokines in the supernatant from FLS were measured by ELISA kit. Expression of Tumor Necrosis Factor Receptor Type 1(TNFR1) and Tumor Necrosis Factor Receptor Type 2(TNFR2) were detected by immunostaining analysis and western blot analysis. CK (80mg/kg) significantly ameliorated the histopathological change of joint in AA rats, balanced the RANKL/OPG ratio and attenuated the proliferation and migration of AA-FLS. CK suppressed the secretion of proinflammatory cytokines TNF-α and downregulated the expression of TNFR2 on AA-FLS. In vitro CK also significantly suppressed proliferation, migration and secretion of AA-FLS mediated by TNF-α. Further studies showed that the effects of CK on AA-FLS were reversed by using glucocorticoid receptor (GR) antagonist (mifepristone). Our data suggest that CK exerts joint-protective effect by interfering with synoviocyte function mediated by TNF-α and TNFR2, and this effect may be mediated by GR.


Asunto(s)
Antiinflamatorios/farmacología , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Sapogeninas/farmacología , Membrana Sinovial/citología , Membrana Sinovial/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Antagonistas de Hormonas/farmacología , Masculino , Mifepristona/farmacología , Ligando RANK/biosíntesis , Ligando RANK/genética , Ratas , Ratas Sprague-Dawley , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Membrana Sinovial/patología
12.
J Immunol ; 195(6): 2633-47, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26268655

RESUMEN

The double-edged sword nature by which IL-2 regulates autoimmunity and the unpredictable outcomes of anti-TNF therapy in autoimmunity highlight the importance for understanding how TNF regulates IL-2. Transmembrane TNF (tmTNF) preferentially binds TNFR2, whereas soluble TNF (sTNF) binds TNFR1. We previously showed reduced IL-2 production in TNFR1(-/-) TNFR2(-/-) CD4(+) T cells. In this study, we generated TNFR1(-/-), TNFR2(-/-), or TNFR1(-/-) TNFR2(-/-) 5C.C7 TCR Il2-GFP mice and report that CD4(+) T cell-intrinsic tmTNF/TNFR2 stimulates Il2 promoter activity and Il2 mRNA stability. We further used tmTNF Foxp3 gfp reporter mice and pharmacological TNF blockade in wild-type mice to report a tmTNF/TNFR2 interaction for Il2 expression. IL-17 is critical for host defense, but its overabundance promotes autoimmunity. IL-2 represses Th17 differentiation, but the role for TNFR2 in this process is not well understood. We report elevated expression of TNFR2 under Th17-polarization conditions. Genetic loss-of-function experimental models, as well as selective TNF blockade by etanercept and XPro1595 in wild-type mice, demonstrate that impaired tmTNF/TNFR2, but not sTNF/TNFR1, promotes Th17 differentiation in vivo and in vitro. Under Th17-polarizing conditions, elevated IL-17 production by TNFR2-knockout CD4(+) T cells was associated with increased STAT3 activity and decreased STAT5 activity. Increased IL-17 production in TNFR2-knockout T cells was prevented by adding exogenous IL-2. We conclude that CD4(+) T cell-intrinsic tmTNF/TNFR2 promotes IL-2 production that inhibits the generation of Th17 cells in a Foxp3-independent manner. Moreover, under Th17-polarizing conditions, selective blockade of CD4(+) T cell-intrinsic TNFR2 appears to be sufficient to promote Th17 differentiation.


Asunto(s)
Interleucina-17/biosíntesis , Interleucina-2/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Células Th17/citología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Diferenciación Celular/inmunología , Etanercept/farmacología , Factores de Transcripción Forkhead/inmunología , Proteínas Fluorescentes Verdes/genética , Inmunosupresores/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
Tumour Biol ; 36(12): 9421-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26113409

RESUMEN

Although the expression of tumor necrosis factor receptors (TNFRs) has been associated with clinicopathologic features of some other cancers, their roles in hypopharyngeal squamous cell carcinoma (HPSCC) have not been documented. Forty-five HPSCC specimens were analyzed for the expression of TNFR1 and TNFR2 and its relationship with clinicopathologic factors. Interaction between the two receptors and its effects on TNF-α was investigated by neutralizing TNFR1 and upregulation of TNFR2. The results indicated that, in HPSCC specimens, the expression of TNFR1 but not TNFR2 is associated with clinical staging, T stage, cervical lymph node metastasis, and histologic grade in HPSCC. In Fadu cells, when conjugating with its receptors, TNF-α mediates proliferation effects, and neutralizing TNFR1 and/or upregulating TNFR2 evokes proliferation-inhibiting and apoptosis-inducing effects and potentiates cisplatin (DDP)-induced growth inhibition and apoptosis induction. In conclusion, interaction of TNFR1 with TNFR2 determines the biological characters of HPSCC, and TNFR1 may dominate this process. Moreover, interaction between the two receptors plays important roles in determining the fates of HPSCC cells and thus may serve as a therapeutic target for developing new therapeutic strategies for HPSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias Hipofaríngeas/genética , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Adulto , Anciano , Apoptosis/genética , Carcinoma de Células Escamosas/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hipofaríngeas/patología , Masculino , Persona de Mediana Edad , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética
14.
Immunology ; 144(2): 254-62, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25087772

RESUMEN

Intestinal inflammation causes tight junction changes and death of epithelial cells, and plays an important role in the development of Crohn's disease (CD). CD52 monoclonal antibody (CD52 mAb) directly targets the cell surface CD52 and is effective in depleting mature lymphocytes by cytolytic effects in vivo, leading to long-lasting changes in adaptive immunity. The aim of this study was to investigate the therapeutic effect of CD52 mAb on epithelial barrier function in animal models of IBD. Interleukin-10 knockout mice (IL-10(-/-) ) of 16 weeks with established colitis were treated with CD52 mAb once a week for 2 weeks. Severity of colitis, CD4(+) lymphocytes and cytokines in the lamina propria, epithelial expression of tight junction proteins, morphology of tight junctions, tumour necrosis factor-α (TNF-α)/TNF receptor 2 (TNFR2) mRNA expression, myosin light chain kinase (MLCK) expression and activity, as well as epithelial apoptosis in proximal colon were measured at the end of the experiment. CD52 mAb treatment effectively attenuated colitis associated with decreased lamina propria CD4(+) lymphocytes and interferon-γ/IL-17 responses in colonic mucosa in IL-10(-/-) mice. After CD52 mAb treatment, attenuation of colonic permeability, increased epithelial expression and correct localization of tight junction proteins (occludin and zona occludens protein-1), as well as ameliorated tight junction morphology were observed in IL-10(-/-) mice. CD52 mAb treatment also effectively suppressed the epithelial apoptosis, mucosa TNF-α mRNA expression, epithelial expression of long MLCK, TNFR2 and phosphorylation of MLC. Our results indicated that anti-CD52 therapy may inhibit TNF-α/TNFR2-mediated epithelial apoptosis and MLCK-dependent tight junction permeability by depleting activated T cells in the gut mucosa.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Glicoproteínas/antagonistas & inhibidores , Interleucina-10/genética , Mucosa Intestinal/fisiología , Uniones Estrechas/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Antígenos de Neoplasias/inmunología , Apoptosis/inmunología , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/inmunología , Antígeno CD52 , Colitis/tratamiento farmacológico , Colitis/inmunología , Colon/inmunología , Glicoproteínas/inmunología , Inflamación/inmunología , Interferón gamma/biosíntesis , Interleucina-17/biosíntesis , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Quinasa de Cadena Ligera de Miosina/biosíntesis , Ocludina/biosíntesis , ARN Mensajero/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Uniones Estrechas/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Proteína de la Zonula Occludens-1/biosíntesis
15.
PLoS One ; 9(10): e111229, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25340707

RESUMEN

TNFα is one of the initial and important mediators to activate downstream signaling pathways by binding to trimerized TNFα receptors (TNFR), and thus is an ideal drug target for cancer therapy. Taking advantage of intrinsic homotimerization of the globular domain of adiponectin (gAD), we have developed a novel TNFα antagonist, the trimerized fusion protein named sTNFRII-gAD. However, our previously-used CHO expression system yielded less than 10 mg/L of sTNFRII-gAD. To produce large quantities of sTNFRII-gAD efficiently, we used a modified CHO-S cell expression system, which is based on a pMH3 vector with non-coding GC-rich DNA fragments for high-level gene expression. We obtained stable clones that produced 75 mg/L of sTNFRII-gAD in the 96-well plate, adapted the clones to 40 ml suspension serum-free batch culture, then optimized the culturing conditions to scale up the fed-batch culture in a 3 L shake-flask and finally in a 5 L AP30 bioreactor. We achieved a final yield of 52 mg/L of sTNFRII-gAD. The trimerized sTNFRII-gAD exhibited the higher affinity to TNFα with a dissociation constant (Kd) of 5.63 nM than the dimerized sTNFRII-Fc with a Kd of 13.4 nM, and further displayed the higher TNFα-neutralizing activity than sTNFRII-Fc (p<0.05) in a L929 cytotoxicity assay. Therefore, the strategy employed in this study may provide an efficient avenue for large-scale production of other recombinant proteins by use of the modified CHO-S cell expression system.


Asunto(s)
Adiponectina/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Animales , Bioensayo , Células CHO , Cricetinae , Cricetulus , Medios de Cultivo/química , Enzimas de Restricción del ADN , Perfilación de la Expresión Génica , Humanos , Plásmidos , Multimerización de Proteína , Estructura Terciaria de Proteína , Transducción de Señal , Resonancia por Plasmón de Superficie , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
17.
Cell Signal ; 26(7): 1539-48, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24703938

RESUMEN

Progranulin (PGRN) was reported to be a stress-response factor in response to hypoxia and acidosis. Here we present evidences demonstrating that PGRN is also an endoplasmic reticulum (ER) stress responsive factor: PGRN expression was induced and its activation of Erk1/2 and Akt signaling enhanced in response to ER stress; Normal ER stress response was lost in PGRN deficient cells and PGRN deficient cells became hypersusceptible to ER stress-induced apoptosis; additionally, recombinant PGRN could rescue the defects in ER-stress responses seen in PGRN deficient cells. Mechanistic studies indicated that PGRN/TNFR2 was critical for PGRN mediated regulation of ER stress response: similar to PGRN, the expression of TNFR2, but not TNFR1, was also induced in the course of ER stress; in addition, the association between PGRN and TNFR2 was markedly enhanced following ER stress; More importantly, PGRN protection of ER stress induced apoptosis was abolished when TNFR2 signaling was blocked. In addition, the 2nd and 3rd cysteine-rich domains (CRD) in the extracellular portion of TNFR2 (CRD2CRD3), known to directly bind to PGRN, disturbed the interaction of PGRN with TNFR2, and in turn abolished PGRN-mediated activation of Erk1/2 and Akt signaling and protection against apoptosis in response to ER-stress. Collectively, PGRN plays an important role in ER stress and regulates ER stress response through interacting with TNFR2. This study provides new insight into PGRN regulation of stress response and may also present PGRN as a potential molecular target for treating stress-associated disorders.


Asunto(s)
Apoptosis/fisiología , Estrés del Retículo Endoplásmico/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Animales , Apoptosis/genética , Células Cultivadas , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Granulinas , Ratones , Ratones Noqueados , Progranulinas , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Receptores Tipo II del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Regulación hacia Arriba
18.
Anticancer Res ; 34(2): 745-52, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24511008

RESUMEN

BACKGROUND/AIM: It has been previously shown that epithelial ovarian carcinoma tissues express high levels of tumor necrosis alpha (TNF-α), interleukin (IL)-6, IL-1α and IL-1ß. The aim of the present study was to evaluate the localization of TNF-α and its receptors (TNFR1 and TNFR2) in different types of ovarian carcinoma tissues and the possible role of TNF in the pathogenesis of epithelial ovarian carcinoma. MATERIALS AND METHODS: Total RNA was extracted from normal and cancerous ovarian tissues and mRNA was analyzed with semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR). Immunohistochemical staining was performed with use of antibodies against human (ah)TNFR1 and TNF2. RESULTS: TNF-α mRNA and TNFR2 mRNA levels were significantly higher in ovarian carcinoma tissues than in normal ovarian tissues, whereas TNFR1 mRNA levels were similar. TNFR1 and TNFR2 were mainly localized in the epithelial neoplastic cells of the tumor. Knocking-down TNF-α activity with αhTNF-a altered ovarian carcinoma cell morphology (with more branches) in vitro. CONCLUSION: Our study indicates a possible role of TNF-α in epithelial ovarian carcinoma pathogenesis through TNFR2, which affects morphological changes, which may be involved ovarian cancer pathogenesis.


Asunto(s)
Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Receptores Tipo I de Factores de Necrosis Tumoral/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Actinas/biosíntesis , Actinas/genética , Comunicación Celular/fisiología , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Ováricas/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Factor de Necrosis Tumoral alfa/genética
19.
Cytokine ; 65(1): 56-64, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24080164

RESUMEN

CD4(+) T cells stimulate immune responses through distinct patterns of cytokine produced by Th1, Th2 or Th17 cells, or inhibit immune responses through Foxp3-expressing regulatory T cells (Tregs). Paradoxically, effector T cells were recently shown to activate Tregs, however, it remains unclear which Th subset is responsible for this effect. In this study, we found that Th17 cells expressed the highest levels of TNF among in vitro generated Th subsets, and most potently promoted expansion and stabilized Foxp3 expression by Tregs when co-transferred into Rag1(-/-) mice. Both TNF and IL-2 produced by Th17 cells contributed to this effect. The stimulatory effect of Th17 cells on Tregs was largely abolished when co-transferred with TNFR2-deficient Tregs. Furthermore, Tregs deficient in TNFR2 also supported a much lower production of IL-17A and TNF expression by co-transferred Th17 cells. Thus, our data indicate that the TNF-TNFR2 pathway plays a crucial role in the reciprocal stimulatory effect of Th17 cells and Tregs. This bidirectional interaction should be taken into account when designing therapy targeting Th17 cells, Tregs, TNF and TNFR2.


Asunto(s)
Activación de Linfocitos/inmunología , Receptores Tipo II del Factor de Necrosis Tumoral/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Traslado Adoptivo , Animales , Antígenos CD4/inmunología , Citocinas/inmunología , Factores de Transcripción Forkhead/inmunología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-2/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis , Células TH1/inmunología , Células Th2/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis
20.
Biochem Biophys Res Commun ; 440(2): 336-41, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24076392

RESUMEN

The neuroprotective role of TNF receptor 2 (TNFR2) has been shown in various studies. However, a direct role of TNFR2 in oligodendrocyte function has not yet been demonstrated. Using primary oligodendrocytes of transgenic mice expressing human TNFR2, we show here that TNFR2 is primarily expressed on oligodendrocyte progenitor cells. Interestingly, preconditioning with a TNFR2 agonist protects these cells from oxidative stress, presumably by increasing the gene expression of distinct anti-apoptotic and detoxifying proteins, thereby providing a potential mechanism for the neuroprotective role of TNFR2 in oligodendrocyte progenitor cells.


Asunto(s)
Oligodendroglía/efectos de los fármacos , Receptores Tipo II del Factor de Necrosis Tumoral/fisiología , Células Madre/efectos de los fármacos , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Humanos , Ratones , Ratones Transgénicos , Oligodendroglía/fisiología , Estrés Oxidativo , Receptores Tipo II del Factor de Necrosis Tumoral/agonistas , Receptores Tipo II del Factor de Necrosis Tumoral/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...