Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(23)2023 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-38069015

RESUMEN

This study aimed to explore the role of Akt protein in the induction and inhibition of prostaglandin (PG) in human follicular dendritic cell (FDC)-like cells. FDC-like cells and B cells were isolated from human tonsils. PG production was assessed using enzyme immunoassay, while the upstream cyclooxygenase-2 (COX-2) protein levels were measured using immunoblotting with FDC-like cells transfected with Akt siRNA to analyze the impact of Akt knockdown. The COX-2 expression and PG production induced with IL-1ß were significantly increased by Akt knockdown. However, IL-1ß did not significantly alter either total or phosphorylated Akt protein levels. Akt knockdown resulted in the augmentation of COX-2 expression induced by B cells, although the addition of B cells did not significantly modulate both total and phosphorylated Akt proteins. In contrast, IL-4 specifically exhibited a potent inhibitory effect on COX-2 protein induction and PG production via STAT6. The inhibitory activity of IL-4 was not hampered by Akt knockdown. Interestingly, COX-2 expression levels induced with IL-1ß were markedly modulated with STAT1 and STAT3 knockdown. STAT1 silencing resulted in further augmentation of COX-2, whereas STAT3 silencing prohibited IL-1ß from stimulating COX-2 expression. The current results suggest that Akt, IL-4, and STAT1 play inhibitory roles in PG production in FDC-like cells and expand our knowledge of the immune inflammatory milieu.


Asunto(s)
Células Dendríticas Foliculares , Interleucina-4 , Humanos , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Células Dendríticas Foliculares/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Prostaglandinas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción STAT/metabolismo
2.
Prostaglandins Other Lipid Mediat ; 151: 106487, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33007445

RESUMEN

Lipid mediators play active roles in each stage of inflammation under physiological and pathologic conditions. We have investigated the cellular source and functions of several prostanoids in the immune inflammatory responses using follicular dendritic cell (FDC)-like cells. In this study, we report a novel finding on the role of 15(S)- hydroxyeicosatetraenoic acid (HETE). Our observation of 15(S)-HETE uptake by FDC-like cells prompted to hypothesize that 15(S)-HETE might have a regulatory role in the other branch of eicosanoid production. The effects of 15(S)-HETE on COX-2 expression and prostaglandin (PG) production were analyzed by immunoblotting and specific enzyme immunoassays. The addition of 15(S)-HETE resulted in elevated levels of COX-2 expression and PG production. The enhanced PG production was not due to growth stimulation of FDC-like cells since 15(S)-HETE did not modulate FDC-like cell proliferation by the culture period of PG measurement. Peroxisome proliferator-activated receptor gamma (PPARγ) seems to mediate the augmenting activity as the antagonist GW9662 dose- dependently prevented 15(S)-HETE from increasing PG production. In addition, PPARγ protein expression was readily detected in FDC-like cells. These effects of 15(S)-HETE were displayed in the combined addition with IL-1ß. Based on these results, we suggest that 15(S)-HETE is an inflammatory costimulator of FDC acting in a paracrine fashion.


Asunto(s)
Células Dendríticas Foliculares/efectos de los fármacos , Células Dendríticas Foliculares/metabolismo , Ácidos Hidroxieicosatetraenoicos/farmacología , Comunicación Paracrina/efectos de los fármacos , Prostaglandinas/biosíntesis , Línea Celular , Células Dendríticas Foliculares/citología , Humanos
3.
Exp Mol Med ; 52(8): 1298-1309, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32770080

RESUMEN

Endothelial progenitor cell (EPC) dysfunction impairs vascular function and remodeling in inflammation-associated diseases, including preeclampsia. However, the underlying mechanism of this inflammation-induced dysfunction remains unclear. In the present study, we found increases in TNF-α and miR-31/155 levels and reduced numbers of circulating EPCs in patients with preeclampsia. Patient-derived mononuclear cells (MNCs) cultured in autologous serum had decreased endothelial nitric oxide synthase (eNOS) expression, nitric oxide production, and differentiation into EPCs with angiogenic potential, and these effects were inhibited by a TNF-α-neutralizing antibody and miR-31/155 inhibitors. Moreover, TNF-α treatment of normal MNCs increased miR-31/155 biogenesis, decreased eNOS expression, reduced EPC differentiation, and impaired angiogenic potential. The TNF-α-induced impairment of EPC differentiation and function was rescued by NF-κB p65 knockdown or miR-31/155 inhibitors. In addition, treatment of MNCs with synthetic miR-31/155 or an eNOS inhibitor mimicked the inhibitory effects of TNF-α on eNOS expression and EPC functions. Moreover, transplantation of EPCs that had been differentiated from TNF-α-treated MNCs decreased neovascularization and blood perfusion in ischemic mouse hindlimbs compared with those of normally differentiated EPCs. These findings suggest that NF-κB activation is required for TNF-α-induced impairment of EPC mobilization, differentiation, and function via miR-31/155 biogenesis and eNOS downregulation. Our data provide a new role for NF-κB-dependent miR-31/155 in EPC dysfunction under the pathogenic conditions of inflammation-associated vascular diseases, including preeclampsia.


Asunto(s)
Células Progenitoras Endoteliales/metabolismo , MicroARNs/metabolismo , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Diferenciación Celular/genética , Regulación hacia Abajo/genética , Células Progenitoras Endoteliales/patología , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/patología , Masculino , Ratones Desnudos , MicroARNs/sangre , MicroARNs/genética , Neovascularización Fisiológica/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Preeclampsia/sangre , Preeclampsia/genética , Embarazo , Factor de Necrosis Tumoral alfa/sangre
4.
BMC Immunol ; 21(1): 20, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32303181

RESUMEN

BACKGROUND: Prostaglandin E2 (PGE2) is an endogenous lipid mediator of inflammation. Its production is regulated by the rate-limiting upstream enzyme cyclooxygenase-2 (COX-2). We have recently demonstrated that the major cell type expressing COX-2 in the germinal center is follicular dendritic cell (FDC). In this study, to elucidate the molecular mechanism of PGE2 in COX-2 production, we asked whether mitogen-activated protein kinases ERK and p38 might regulate COX-2 expression. RESULTS: FDC-like cells were used to analyze the phosphorylation kinetics of ERK and p38 and the impact of genetic knockdown. PGE2 stimulation gave rise to a rapid increase of p38 but not ERK phosphorylation. In contrast, IL-1ß induced phosphorylation of both MAPKs. Knockdown of p38 resulted in a marked suppression of COX-2 expression induced by either PGE2 or IL-1ß. ERK knockdown did not significantly affect the effect of PGE2 and IL-1ß on COX-2 induction. The differential results of p38 and ERK siRNA transfection were reproduced in the production of prostaglandins and in experiments performed with pharmacologic inhibitors. CONCLUSIONS: Our data indicate that p38 is essentially required for PGE2 to induce COX-2 expression in FDC-like cells. The current study helps to expand our understanding of the biological function of FDC at the molecular level and provides a potential rationale for the pharmacologic or genetic approaches to regulate p38 MAPK in the treatment of various inflammatory disorders.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Células Dendríticas Foliculares/efectos de los fármacos , Dinoprostona/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Cultivadas , Células Dendríticas Foliculares/metabolismo , Humanos , Interleucina-1beta/metabolismo , Fosforilación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
5.
J Biol Chem ; 293(38): 14812-14822, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30104414

RESUMEN

cGMP-dependent protein kinase 1 (PKG1) plays an important role in nitric oxide (NO)/cGMP-mediated maintenance of vascular smooth muscle cell (VSMC) phenotype and vasorelaxation. Inflammatory cytokines, including tumor necrosis factor-α (TNFα), have long been understood to mediate several inflammatory vascular diseases. However, the underlying mechanism of TNFα-dependent inflammatory vascular disease is unclear. Here, we found that TNFα treatment decreased PKG1 expression in cultured VSMCs, which correlated with NF-κB-dependent biogenesis of miR-155-5p that targeted the 3'-UTR of PKG1 mRNA. TNFα induced VSMC phenotypic switching from a contractile to a synthetic state through the down-regulation of VSMC marker genes, suppression of actin polymerization, alteration of cell morphology, and elevation of cell proliferation and migration. All of these events were blocked by treatment with an inhibitor of miR-155-5p or PKG1, whereas transfection with miR-155-5p mimic or PKG1 siRNA promoted phenotypic modulation, similar to the response to TNFα. In addition, TNFα-induced miR-155-5p inhibited the vasorelaxant response of de-endothelialized mouse aortic vessels to 8-Br-cGMP by suppressing phosphorylation of myosin phosphatase and myosin light chain, both of which are downstream signal modulators of PKG1. Moreover, TNFα-induced VSMC phenotypic alteration and vasodilatory dysfunction were blocked by NF-κB inhibition. These results suggest that TNFα impairs NO/cGMP-mediated maintenance of the VSMC contractile phenotype and vascular relaxation by down-regulating PKG1 through NF-κB-dependent biogenesis of miR-155-5p. Thus, the NF-κB/miR-155-5p/PKG1 axis may be crucial in the pathogenesis of inflammatory vascular diseases, such as atherosclerotic intimal hyperplasia and preeclamptic hypertension.


Asunto(s)
Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Regulación hacia Abajo/fisiología , MicroARNs/fisiología , Músculo Liso Vascular/citología , Factor de Necrosis Tumoral alfa/fisiología , Regiones no Traducidas 3' , Actinas/metabolismo , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/genética , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Masculino , Ratones Endogámicos C57BL , Músculo Liso Vascular/enzimología , Músculo Liso Vascular/metabolismo , FN-kappa B/metabolismo , Polimerizacion , ARN Mensajero/genética
6.
Front Immunol ; 9: 1742, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30100910

RESUMEN

Migration of human plasmablast to the bone marrow is essential for the final differentiation of plasma cells and maintenance of effective humoral immunity. This migration is controlled by CXCL12/CXCR4-mediated activation of the protein kinase AKT. Herein, we show that the CXCL12-induced migration of human plasmablasts is dependent on glucose oxidation. Glucose depletion markedly inhibited plasmablast migration by 67%, and the glucose analog 2-deoxyglucose (2-DG) reduced the migration by 53%; conversely, glutamine depletion did not reduce the migration. CXCL12 boosted the oxygen consumption rate (OCR), and 2-DG treatment significantly reduced the levels of all measured tricarboxylic acid (TCA) cycle intermediates. AKT inhibitors blocked the CXCL12-mediated increase of OCR. CXCL12 enhanced the pyruvate dehydrogenase (PDH) activity by 13.5-fold in an AKT-dependent manner to promote mitochondrial oxidative phosphorylation. The knockdown and inhibition of PDH confirmed its indispensable role in CXCL12-induced migration. Cellular ATP levels fell by 91% upon exposure to 2-DG, and the mitochondrial ATP synthase inhibitor oligomycin inhibited CXCL12-induced migration by 85%. Low ATP levels inhibited the CXCL12-induced activation of AKT and phosphorylation of myosin light chains by 42%, which are required for cell migration. Thus, we have identified a mechanism that controls glucose oxidation via AKT signaling and PDH activation, which supports the migration of plasmablasts. This mechanism can provide insights into the proper development of long-lived plasma cells and is, therefore, essential for optimal humoral immunity. To our knowledge, this study is the first to investigate metabolic mechanisms underlying human plasmablast migration toward CXCL12.


Asunto(s)
Quimiocina CXCL12/metabolismo , Quimiotaxis/inmunología , Glucosa/metabolismo , Oxidación-Reducción , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Complejo Piruvato Deshidrogenasa/metabolismo , Diferenciación Celular/inmunología , Movimiento Celular , Centro Germinal/inmunología , Centro Germinal/metabolismo , Humanos , Inmunidad Humoral , Mitocondrias/metabolismo , Fosforilación Oxidativa , Células Plasmáticas/citología , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , Células Precursoras de Linfocitos B/metabolismo , Transducción de Señal
7.
Mol Immunol ; 94: 1-6, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29241029

RESUMEN

In spite of the potential importance of cyclooxygenase (COX)-2 expression in the germinal center, its underlying cellular and molecular mechanisms are largely unknown. COX-2 is the key enzyme generating pleiotropic prostaglandins. Based on our previous findings, we hypothesized that lymphocytes would stimulate COX-2 expression in follicular dendritic cell (FDC) by liberating cytokines. In this study, we examined the effect of tonsillar lymphocytes on COX-2 expression in FDC-like cells by immunoblotting. B but not T cells induced COX-2 protein in a time- and dose-dependent manner. Sub-fractionation analysis of B cell subsets revealed that activated but not resting B cells were responsible for the COX-2 induction. Confocal microscopy of frozen tonsils demonstrated that FDCs indeed express COX-2 in situ, in line with the in vitro results. To identify the stimulating molecule, we added neutralizing antibodies to the coculture of FDC-like cells and B cells. COX-2 induction in FDC-like cells was markedly inhibited by TNF-α neutralizing antibody. Finally, the actual production of TNF-α by activated B cells was confirmed by an enzyme immunoassay. The current study implies an unrecognized cellular interaction between FDC and B cells leading to COX-2 expression during immune inflammatory responses.


Asunto(s)
Linfocitos B/inmunología , Ciclooxigenasa 2/metabolismo , Células Dendríticas Foliculares/metabolismo , Activación de Linfocitos/fisiología , Factor de Necrosis Tumoral alfa/metabolismo , Linfocitos B/citología , Células Cultivadas , Células Dendríticas Foliculares/citología , Humanos , Tonsila Palatina/citología , Transducción de Señal/inmunología , Regulación hacia Arriba
8.
Oncotarget ; 8(38): 63155-63176, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28968979

RESUMEN

The regulatory role of suppressor of cytokine signaling 1 (SOCS1) in inflammation has been reported. However, its role in allergic inflammation has not been previously reported. SOCS1 mediated in vitro and in vivo allergic inflammation. Histone deacetylase-3 (HDAC3), a mediator of allergic inflammation, interacted with SOCS1, and miR-384 inhibitor, a positive regulator of HDAC3, induced features of allergic inflammation in an SOCS1-dependent manner. miRNA array analysis showed that the expression of miR-122 was decreased by antigen-stimulation. TargetScan analysis predicted the binding of miR-122 to the 3'-UTR of SOCS1. miR-122 inhibitor induced in vitro and in vivo allergic features in SOCS1-dependent manner. SOCS1 was necessary for allergic inflammation-promoted enhanced tumorigenic and metastatic potential of cancer cells. SOCS1 and miR-122 regulated cellular interactions involving cancer cells, mast cells and macrophages during allergic inflammation. SOCS1 mimetic peptide, D-T-H-F-R-T-F-R-S-H-S-D-Y-R-R-I, inhibited in vitro and in vivo allergic inflammation, allergic inflammation-promoted enhanced tumorigenic and metastatic potential of cancer cells, and cellular interactions during allergic inflammation. Janus kinase 2 (JAK2) exhibited binding to SOCS1 mimetic peptide and mediated allergic inflammation. Transforming growth factor- Δ1 (TGF-Δ1) was decreased during allergic inflammation and showed an anti-allergic effect. SOCS1 and JAK2 regulated the production of anti-allergic TGF-Δ1. Taken together, our results show that miR-122-SOCS1 feedback loop can be employed as a target for the development of anti-allergic and anti-cancer drugs.

9.
Free Radic Biol Med ; 112: 567-577, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28888620

RESUMEN

Ligation of the death receptors for TNF-α, FasL, and TRAIL triggers two common pathways, caspase-dependent intrinsic apoptosis and intracellular reactive oxygen species (ROS) generation. The apoptotic pathway is well characterized; however, a signaling linker between the death receptor and ROS production has not been clearly elucidated. Here, we found that death receptor-induced ROS generation was strongly inhibited by mitochondrial complex I and II inhibitors, but not by inhibitors of NADPH oxidase, lipoxygenase, cyclooxygenase or xanthine oxidase, indicating that ROS are mostly generated by the impairment of the mitochondrial respiratory chain. ROS generation was accompanied by caspase-8 activation, Bid cleavage, and cytochrome c release; it was blocked in FADD- and caspase-8-deficient cells, as well as by caspase-8 knockdown and inhibitor. Moreover, Bid knockdown abrogated TNF-α- or TRAIL-induced ROS generation, whereas overexpression of truncated Bid (tBid) or knockdown of cytochrome c spontaneously elevated ROS production. In addition, p53-overexpressing cells accumulated intracellular ROS via cytochrome c release mediated by the BH3-only protein Noxa induction. In a cell-free reconstitution system, caspase-8-mediated Bid cleavage and recombinant tBid induced mitochondrial cytochrome c release and ROS generation, which were blocked by Bcl-xL and antioxidant enzymes. These data suggest that anti-apoptotic Bcl-2 proteins play an important role in mitochondrial ROS generation by preventing cytochrome c release. These data provide evidence that the FADD/caspase-8/Bid/cytochrome c axis is a crucial linker between death receptors and mitochondria, where they play a role in ROS generation and apoptosis.


Asunto(s)
Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Caspasa 8/genética , Citocromos c/genética , Mitocondrias Hepáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Apoptosis/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Caspasa 8/metabolismo , Línea Celular Tumoral , Citocromos c/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/genética , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Regulación de la Expresión Génica , Células HeLa , Humanos , Células Jurkat , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
10.
Clin Exp Vaccine Res ; 6(2): 135-145, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28775978

RESUMEN

PURPOSE: The goal of this study was to investigate the utility of DNA vaccines encoding Ebola virus glycoprotein (GP) as a vaccine type for the production of GP-specific hybridomas and antibodies. MATERIALS AND METHODS: DNA vaccines were constructed to express Ebola virus GP. Mice were injected with GP DNA vaccines and their splenocytes were used for hybridoma production. Enzyme-linked immunosorbent assays (ELISAs), limiting dilution subcloning, antibody purification methods, and Western blot assays were used to select GP-specific hybridomas and purify monoclonal antibodies (MAbs) from the hybridoma cells. RESULTS: Twelve hybridomas, the cell supernatants of which displayed GP-binding activity, were selected by ELISA. When purified MAbs from 12 hybridomas were tested for their reactivity to GP, 11 MAbs, except for 1 MAb (from the A6-9 hybridoma) displaying an IgG2a type, were identified as IgM isotypes. Those 11 MAbs failed to recognize GP. However, the MAb from A6-9 recognized the mucin-like region of GP and remained reactive to the antigen at the lowest tested concentration (1.95 ng/mL). This result suggests that IgM-secreting hybridomas are predominantly generated by DNA vaccination. However, boosting with GP resulted in greater production of IgG-secreting hybridomas than GP DNA vaccination alone. CONCLUSION: DNA vaccination may preferentially generate IgM-secreting hybridomas, but boosting with the protein antigen can reverse this propensity. Thus, this protein boosting approach may have implications for the production of IgG-specific hybridomas in the context of the DNA vaccination platform. In addition, the purified monoclonal IgG antibodies may be useful as therapeutic antibodies for controlling Ebola virus infection.

11.
Cell Death Dis ; 8(5): e2808, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28518140

RESUMEN

Serum deprivation or withdrawal induces apoptosis in endothelial cells, resulting in endothelial cell dysfunction that is associated with cardiovascular disease. However, there is still limited information on the role of miRNA in serum deprivation-induced apoptosis. Here we found that serum deprivation increased caspase-dependent apoptosis through miRNA-101-3p downregulation, without altering expression of its host gene RNA 3'-terminal phosphate cyclase-like 1, which was highly correlated with suppressed expression levels of Dicer and Argonaute 2 (Ago2), indicating that miR-101-3p is post-transcriptionally elevated in serum-deprived conditions. The decreased miR-101-3p caused elevated Bim expression by targeting its 3'-untranslated region (3'-UTR). This resulted in activation of the intrinsic pathway of apoptosis via interaction with Bcl-2, decreased mitochondrial membrane potential, cytochrome c release, mitochondrial reactive oxygen species (ROS) production, and caspase activation. These events were abrogated by miR-101-3p mimic and the proapoptotic Bim siRNA, which suggest a determinant role of the miR-101-3p/Bim axis in serum deprivation-induced apoptosis. The apoptosis induced by miR-101-3p-mediated Bim expression is mediated by both caspase-3 and -1, which are activated by two distinct intrinsic mechanisms, cytochrome c release and ROS-induced inflammasome activation, respectively. In other words, the antioxidant inhibited endothelial cell death mediated by caspase-1 that activated caspase-7, but not caspase-3. These findings provide mechanistic insight into a novel function of miR-101-3p in serum withdrawal-induced apoptosis triggered by activating two different intrinsic or mitochondrial apoptosis pathways, implicating miR-101-3p as a therapeutic target that limits endothelial cell death associated with vascular disorders.


Asunto(s)
Apoptosis , Proteína 11 Similar a Bcl2/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , MicroARNs/metabolismo , Transducción de Señal , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Proteínas Argonautas/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Medio de Cultivo Libre de Suero , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Ribonucleasa III/metabolismo , Transducción de Señal/efectos de los fármacos
12.
Mol Immunol ; 87: 60-66, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28407559

RESUMEN

Prostaglandins (PGs) are bioactive lipid mediators generated from the phospholipids of cell membrane in response to various inflammatory signals. To understand the potential role of PGs in PG production itself during immune inflammatory responses, we examined the effect of PGE2, PGF2α, and beraprost on COX-2 expression using follicular dendritic cell (FDC)-like HK cells isolated from human tonsils. Those three PGs specifically augmented COX-2 protein expression in a dose-dependent manner after 4 or 8h of treatment. The enhancing effect was also reflected in the actual production of PGs and the viable cell recovery of germinal center B-cells. To investigate the underlying molecular mechanism, we examined the impact of PI3K inhibitors on PG-induced COX-2 expression. Interestingly, COX-2 induction by PGE2 and beraprost, but not PGF2α, was enhanced by wortmannin and LY294002. In line with this result, Akt phosphorylation was inhibited by PGE2 and beraprost but not by PGF2α. The distinct effect of PGE2 and beraprost from PGF2α was reproduced in Akt-knockdowned HK cells. Our current findings imply that PGE2 and PGI2 stimulate COX-2 expression in FDC by inhibiting Akt phosphorylation. Additional studies are warranted to determine the potential role of Akt as a therapeutic target in patients with inflammatory disorders.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Células Dendríticas Foliculares/metabolismo , Dinoprostona/metabolismo , Fosforilación/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Linfocitos B/metabolismo , Línea Celular , Epoprostenol/análogos & derivados , Epoprostenol/metabolismo , Centro Germinal/metabolismo , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo
13.
Oncotarget ; 8(15): 24932-24948, 2017 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-28212561

RESUMEN

Toll-like receptor (TLR) ligands are strongly considered immune-adjuvants for cancer immunotherapy and have been shown to exert direct anti-cancer effects. This study was performed to evaluate the synergistic anti-cancer and anti-metastatic effects of the TLR7 agonist imiquimod (IMQ) during radiotherapy for melanoma. The pretreatment of B16F10 or B16F1 cells with IMQ combined with γ-ionizing radiation (IR) led to enhanced cell death via autophagy, as demonstrated by increased expression levels of autophagy-related genes, and an increased number of autophagosomes in both cell lines. The results also confirmed that the autophagy process was accelerated via the reactive oxygen species (ROS)-mediated MAPK and NF-κB signaling pathway in the cells pretreated with IMQ combined with IR. Mice subcutaneously injected with melanoma cells showed a reduced tumor growth rate after treatment with IMQ and IR. Treatment with 3-methyladenine (3-MA), ameliorated the anti-cancer effect of IMQ combined with IR. Additionally, the combination therapy enhanced anti-cancer immunity, as demonstrated by an increased number of CD8+ T cells and decreased numbers of regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSCs) in the tumor lesions. Moreover, the combination therapy decreased the number of metastatic nodules in the lungs of mice that were injected with B16F10 cells via the tail vein. In addition, the combination therapy enhanced systemic anti-cancer immunity by increasing the abundances of T cell populations expressing IFN-γ and TNF-α. Therefore, these findings suggest that IMQ could serve as a radiosensitizer and immune booster during radiotherapy for melanoma patients.


Asunto(s)
Aminoquinolinas/farmacología , Antineoplásicos/farmacología , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/radioterapia , Glicoproteínas de Membrana/agonistas , Receptor Toll-Like 7/agonistas , Animales , Autofagia/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Quimioradioterapia , Modelos Animales de Enfermedad , Imiquimod , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL
14.
Oncotarget ; 8(8): 13632-13651, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28099142

RESUMEN

We previously reported the role of cancer/testis antigen CAGE in the response to anti-cancer drugs. CAGE increased the expression of cyclinD1, and pGSK3ßSer9, an inactive GSK3ß, while decreasing the expression of phospho-cyclinD1Thr286. CAGE showed binding to GSK3ß and the domain of CAGE (amino acids 231-300) necessary for binding to GSK3ß and for the expression regulation of cyclinD1 was determined. 269GTGKT273 peptide, corresponding to the DEAD box helicase domain of CAGE, decreased the expression of cyclinD1 and pGSK3ßSer9 while increasing the expression of phospho-cyclinD1Thr286. GTGKT peptide showed the binding to CAGE and prevented CAGE from binding to GSK3ß. GTGKT peptide changed the localization of CAGE and inhibited the binding of CAGE to the promoter sequences of cyclin D1. GTGKT peptide enhanced the apoptotic effects of anti-cancer drugs and decreased the migration, invasion, angiogenic, tumorigenic and metastatic potential of anti-cancer drug-resistant cancer cells. We found that Lys272 of GTGKT peptide was necessary for conferring anti-cancer activity. Peptides corresponding to the DEAD box helicase domain of CAGE, such as AQTGTGKT, QTGTGKT and TGTGKT, also showed anti-cancer activity by preventing CAGE from binding to GSK3ß. GTGKT peptide showed ex vivo tumor homing potential. Thus, peptides corresponding to the DEAD box helicase domain of CAGE can be developed as anti-cancer drugs in cancer patients expressing CAGE.


Asunto(s)
Antineoplásicos/farmacología , Ciclina D1/biosíntesis , ARN Helicasas DEAD-box/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Oligopéptidos/farmacología , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Movimiento Celular/fisiología , Ciclina D1/genética , ARN Helicasas DEAD-box/metabolismo , Resistencia a Antineoplásicos , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas , Dominios Proteicos
15.
Free Radic Biol Med ; 104: 185-198, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28087411

RESUMEN

Preeclampsia is an inflammatory disease with endothelial cell dysfunction that occurs via decreased endothelial nitric oxide synthase/nitric oxide (eNOS/NO) activity. Aspirin reduces the incidence of hypertensive pregnancy complications. However, the underlying mechanism has not been clearly explained. Here, we found that tumor necrosis factor (TNF)-α, microRNA (miR)-155, and eNOS levels as well as endothelial redox phenotype were differentially regulated in preeclamptic patients, implying the involvement of TNF-α- and redox signal-mediated miR-155 biogenesis and eNOS downregulation in the pathogenesis of preeclampsia. Aspirin prevented the TNF-α-mediated increase in miR-155 biogenesis and decreases in eNOS expression and NO/cGMP production in cultured human umbilical vein endothelial cells (HUVECs). Similar effects of aspirin were also observed in HUVECs treated with H2O2. The preventive effects of aspirin was associated with the inhibition of nuclear factor-κB (NF-κB)-dependent MIR155HG (miR-155 host gene) expression. Aspirin recovered the TNF-α-mediated decrease in wild-type, but not mutant, eNOS 3'-untranslated region reporter activity, whose effect was blocked by miR-155 mimic. Moreover, aspirin prevented TNF-α-mediated endothelial cell dysfunction associated with impaired vasorelaxation, angiogenesis, and trophoblast invasion, and the preventive effects were blocked by miR-155 mimic or an eNOS inhibitor. Aspirin rescued TNF-α-mediated eNOS downregulation coupled with endothelial dysfunction by inhibiting NF-κB-dependent transcriptional miR-155 biogenesis. Thus, the redox-sensitive NF-κB/miR-155/eNOS axis may be crucial in the pathogenesis of vascular disorders including preeclampsia.


Asunto(s)
Aspirina/administración & dosificación , MicroARNs/genética , Óxido Nítrico Sintasa de Tipo III/genética , Preeclampsia/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/genética , Adulto , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , FN-kappa B/genética , Preeclampsia/metabolismo , Preeclampsia/patología , Embarazo , Transducción de Señal , Transfección , Factor de Necrosis Tumoral alfa/metabolismo
16.
Mol Immunol ; 76: 41-8, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27344616

RESUMEN

Prostaglandins (PGs) are recognized as important immune regulators. Using human follicular dendritic cell (FDC)-like HK cells, we have investigated the immunoregulatory role of PGs and their production mechanisms. The present study was aimed at determining the role of TGF-ß in IL-1ß-induced cyclooxygenase-2 (COX-2) expression by immunoblotting. COX-2 is the key enzyme responsible for PG production in HK cells. TGF-ß, when added simultaneously with IL-1ß, gave rise to an additive effect on COX-2 expression in a dose-dependent manner. However, TGF-ß inhibited IL-1ß-stimulated COX-2 expression when it was added at least 12h before IL-1ß addition. The inhibitory effect of TGF-ß was specific to IL-1ß-induced COX-2 expression in HK cells. The stimulating and inhibitory effects of TGF-ß were reproduced in IL-1ß-stimulated PG production. Based on our previous results of the essential requirement of ERK and p38 MAPKs in TGF-ß-induced COX-2 expression, we examined whether the differential activation of these MAPKs would underlie the opposing activities of TGF-ß. The phosphorylation of ERK and p38 MAPKs was indeed enhanced or suppressed by the simultaneous treatment or pre-treatment, respectively. These results suggest that TGF-ß exerts opposing effects on IL-1ß-induced COX-2 expression in HK cells by differentially regulating activation of ERK and p38 MAPKs.


Asunto(s)
Células Dendríticas Foliculares/metabolismo , Regulación de la Expresión Génica/inmunología , Prostaglandinas/biosíntesis , Transducción de Señal/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Ciclooxigenasa 2/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Humanos , Immunoblotting , Interleucina-1beta/metabolismo , Proteínas Quinasas Activadas por Mitógenos
17.
Oncotarget ; 7(9): 10297-321, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26863629

RESUMEN

MicroRNA array analysis revealed that miR-217 expression was decreased in anti-cancer drug-resistant Malme3MR cancer cells. CAGE, a cancer/testis antigen, was predicted as a target of miR-217. Luciferase activity and ChIP assays revealed a negative feedback relationship between CAGE and miR-217. miR-217 and CAGE oppositely regulated the response to anti-cancer drugs such as taxol, gefitinib and trastuzumab, an inhibitor of HER2. miR-217 negatively regulated the tumorigenic, metastatic, angiogenic, migration and invasion potential of cancer cells. The xenograft of Malme3MR cells showed an increased expression of pEGFRY845. CAGE and miR-217 inhibitor regulated the expression of pEGFRY845. CAGE showed interactions with EGFR and HER2 and regulated the in vivo sensitivity to trastuzumab. The down-regulation of EGFR or HER2 enhanced the sensitivity to anti-cancer drugs. CAGE showed direct regulation of HER2 and was necessary for the interaction between EGFR and HER2 in Malme3MR cells. miR-217 inhibitor induced interactions of CAGE with EGFR and HER2 in Malme3M cells. The inhibition of EGFR by CAGE-binding GTGKT peptide enhanced the sensitivity to gefitinib and trastuzumab and prevented interactions of EGFR with CAGE and HER2. Our results show that miR-217-CAGE feedback loop serves as a target for overcoming resistance to various anti-cancer drugs, including EGFR and HER2 inhibitors.


Asunto(s)
Antineoplásicos/farmacología , ARN Helicasas DEAD-box/metabolismo , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , MicroARNs/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , ARN Helicasas DEAD-box/genética , Femenino , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Neovascularización Patológica/tratamiento farmacológico , Paclitaxel/farmacología , Quinazolinas/farmacología , Transducción de Señal/efectos de los fármacos , Trastuzumab/farmacología
18.
Biochem Biophys Res Commun ; 463(4): 532-7, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26051280

RESUMEN

Kringle 5, derived from plasminogen, is highly capable of inhibiting angiogenesis. Here, we have designed and synthesized 10 tetrapeptides, based on the amino acid properties of the core tetrapeptide Lys-Leu-Tyr-Asp (KLYD) originating from anti-angiogenic kringle 5 of human plasminogen. Of these, Arg-Leu-Tyr-Glu (RLYE) effectively inhibited vascular endothelial growth factor (VEGF)-induced endothelial cell proliferation, migration and tube formation, with an IC50 of 0.06-0.08 nM, which was about ten-fold lower than that of the control peptide KLYD (0.79 nM), as well as suppressed developmental angiogenesis in a zebrafish model. Furthermore, this peptide effectively inhibited the cellular events that precede angiogenesis, such as ERK and eNOS phosphorylation and nitric oxide production, in endothelial cells stimulated with VEGF. Collectively, these data demonstrate that RLYE is a potent anti-angiogenic peptide that targets the VEGF signaling pathway.


Asunto(s)
Neovascularización Fisiológica/efectos de los fármacos , Oligopéptidos/farmacología , Factor A de Crecimiento Endotelial Vascular/farmacología , Secuencia de Aminoácidos , Animales , Células Endoteliales de la Vena Umbilical Humana , Humanos , Homología de Secuencia de Aminoácido , Transducción de Señal/efectos de los fármacos , Pez Cebra
19.
J Immunol ; 194(9): 4287-97, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825445

RESUMEN

PGs are emerging as important immune modulators. Since our report on the expression of PG synthases in human follicular dendritic cells, we investigated the potential immunoregulatory function of PGs and their production mechanisms. In this study, we explored the intracellular signaling molecules mediating TGF-ß-induced cyclooxygenase (COX)-2 augmentation in follicular dendritic cell-like cells. TGF-ß triggered phosphorylation of Smad3 and ERK, which were essential for the increase in COX-2 protein. Interestingly, depletion of suppressor of cytokine signaling 1 (SOCS1) resulted in an almost complete inhibition of Smad3 phosphorylation and COX-2 induction. Nuclear translocation of Smad3 was inhibited in SOCS1-depleted cells. SOCS1 knockdown also downregulated TGF-ß-stimulated Snail expression and its binding to the Cox-2 promoter. In contrast, overexpression of SOCS1 gave rise to a significant increase in Snail and COX-2 proteins. SOCS1 was reported to be a negative regulator of cytokine signaling by various investigators. However, our current data suggest that SOCS1 promotes TGF-ß-induced COX-2 expression and PG production by facilitating Smad3 phosphorylation and Snail binding to the Cox-2 promoter. The complete understanding of the biological function of SOCS1 might be obtained via extensive studies with diverse cell types.


Asunto(s)
Células Dendríticas Foliculares/inmunología , Células Dendríticas Foliculares/metabolismo , Prostaglandinas/biosíntesis , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Ciclooxigenasa 2/metabolismo , Células Dendríticas Foliculares/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Fosforilación , Transporte de Proteínas , Transducción de Señal , Proteína smad3/metabolismo , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Factor de Crecimiento Transformador beta/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
20.
J Biol Chem ; 290(22): 14245-66, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25907560

RESUMEN

Cyclooxgenase-2 (COX-2) knock-out mouse experiments showed that COX-2 was necessary for in vivo allergic inflammation, such as passive cutaneous anaphylaxis, passive systemic anaphylaxis, and triphasic cutaneous allergic reaction. TargetScan analysis predicted COX-2 as a target of miR-26a and miR-26b. miR-26a/-26b decreased luciferase activity associated with COX-2-3'-UTR. miR-26a/-26b exerted negative effects on the features of in vitro and in vivo allergic inflammation by targeting COX-2. ChIP assays showed the binding of HDAC3 and SNAIL, but not COX-2, to the promoter sequences of miR-26a and miR-26b. Cytokine array analysis showed that the induction of chemokines, such as MIP-2, in the mouse passive systemic anaphylaxis model occurred in a COX-2-dependent manner. ChIP assays showed the binding of HDAC3 and COX-2 to the promoter sequences of MIP-2. In vitro and in vivo allergic inflammation was accompanied by the increased expression of MIP-2. miR-26a/-26b negatively regulated the expression of MIP-2. Allergic inflammation enhanced the tumorigenic and metastatic potential of cancer cells and induced positive feedback involving cancer cells and stromal cells, such as mast cells, macrophages, and endothelial cells. miR-26a mimic and miR-26b mimic negatively regulated the positive feedback between cancer cells and stromal cells and the positive feedback among stromal cells. miR-26a/-26b negatively regulated the enhanced tumorigenic potential by allergic inflammation. COX-2 was necessary for the enhanced metastatic potential of cancer cells by allergic inflammation. Taken together, our results indicate that the miR26a/-26b-COX-2-MIP-2 loop regulates allergic inflammation and the feedback relationship between allergic inflammation and the enhanced tumorigenic and metastatic potential.


Asunto(s)
Quimiocina CXCL2/metabolismo , Ciclooxigenasa 2/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Neoplasias/metabolismo , Regiones no Traducidas 3' , Animales , Antineoplásicos/química , Línea Celular Tumoral , Colágeno/química , Combinación de Medicamentos , Femenino , Hipersensibilidad/metabolismo , Inmunoglobulina E/metabolismo , Laminina/química , Pulmón/metabolismo , Macrófagos/metabolismo , Masculino , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Proteoglicanos/química , Ratas , Especies Reactivas de Oxígeno/metabolismo , beta-N-Acetilhexosaminidasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...