Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Immunology ; 172(3): 486-499, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38547355

RESUMEN

To explore the effect of K33 only mutant ubiquitin (K33O) on bone marrow-derived dendritic cells' (BMDCs') maturity, antigen uptake capability, surface molecule expressions and BMDC-mediated CTL priming, and further investigate the role of PI3K-Akt engaged in K33O-increased BMDC maturation, antigen uptake and presentation, surface molecule expressions and BMDC-based CTL priming. BMDCs were conferred K33O and other ubiquitin mutants (K33R, K48R, K63R-mutant ubiquitin) incubation or LY294002 and wortmannin pretreatment. PI3K-Akt phosphorylation, antigen uptake, antigenic presentation and CD86/MHC class I expression in BMDC were determined by western blot or flow cytometry. BMDC-based CTL proliferation and priming were determined by in vitro mixed lymphocyte reaction (MLR), ex vivo enzyme-linked immunospot assay (Elispot) and flow cytometry with intracellular staining, respectively. The treatment with K33O effectively augmented PI3K-Akt phosphorylation, BMDCs' antigen uptake, antigenic presentation, CD86/MHC class I and CD11c expressions. MLR, Elispot and flow cytometry revealed that K33O treatment obviously enhanced CTL proliferation, CTL priming and perforin/granzyme B expression. The pretreatment with PI3K-Akt inhibitors efficiently abrogated K33O's effects on BMDC. The replenishment of K33 only mutant ubiquitin augments BMDC-mediated CTL priming in bone marrow-derived dendritic cells via PI3K-Akt signalling.


Asunto(s)
Presentación de Antígeno , Células de la Médula Ósea , Células Dendríticas , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Linfocitos T Citotóxicos , Ubiquitina , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Animales , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ubiquitina/metabolismo , Linfocitos T Citotóxicos/inmunología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Presentación de Antígeno/inmunología , Ratones Endogámicos C57BL , Fosforilación , Activación de Linfocitos , Diferenciación Celular , Mutación , Morfolinas/farmacología , Prueba de Cultivo Mixto de Linfocitos , Proliferación Celular , Antígeno B7-2/metabolismo , Antígeno B7-2/genética , Antígeno B7-2/inmunología , Células Cultivadas , Cromonas/farmacología , Wortmanina/farmacología , Androstadienos/farmacología
2.
J Leukoc Biol ; 112(1): 157-172, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35352390

RESUMEN

K48-linked ubiquitination determines antigen degradation and plays vital roles in the process of cross-presentation of bone marrow precursor cell (BMPC)-derived dendritic cells (DCs). Although previous studies revealed that K48 and K27-linked ubiquitination regulates innate immunity, the exact roles of K48 and K27-linked ubiquitination in cross-presentation and BMPC-based adaptive immunity are still uncertain. In this study, we investigated the effects of K48- and K27-mutant ubiquitin (Ub) on BMPC-based adaptive immune response by observing the effects of MG132, Ub deficiency, and K48/K27-mutant Ub on cross-presentation, T cell proliferation, IFN-γ secretion, BMPC-based CTL priming, and thereby the efficiency of cytolytic capacity of BMPC-activate T cells. We demonstrated that MG132, Ub deficiency, and K48-mutant Ub impair cross-presentation, T cell proliferation, IFN-γ secretion, BMPC-based CTL priming, and the cytolytic capacity of BMPC-activated T cells. Moreover, although K27-only Ub decreases cross-presentation, the replenishment of K27-mutant Ub restores Ub deficiency impaireds the abilities of T cell proliferation, IFN-γ secretion, CTL priming, and the cytolytic capacity of BMPC-activated T cells. Thus, these data suggest that K48- and K27-linked ubiquitination contributes to BMPC-mediated adaptive immune response by affecting BMPC cross-presentation and the cytolytic capacity by up-regulating both perforin and granzyme B in BMPC-activated T cells. K48- and K27-mutant Ub might have potential clinical therapeutic function in adaptive immune response-associated diseases.


Asunto(s)
Reactividad Cruzada , Ubiquitina , Inmunidad Adaptativa/genética , Médula Ósea/metabolismo , Ubiquitina/metabolismo , Ubiquitinación
3.
Vaccines (Basel) ; 9(3)2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33808531

RESUMEN

K48-linked ubiquitination determining antigen degradation and the endosomal recruitments of p97 and Sec61 plays vital roles in dendritic cell (DC) cross-presentation. Our previous studies revealed that nicotine treatment increases bone marrow-derived dendritic cell (BM-DC) cross-presentation and promotes BM-DC-based cytotoxic T lymphocyte (CTL) priming. But the effect of nicotine on K48-linked ubiquitination and the mechanism of nicotine-increased BM-DC cross-presentation are still uncertain. In this study, we first demonstrated that ex vivo nicotine administration obviously increased K48-linked ubiquitination in BM-DC. Then, we found that K48-linked ubiquitination was essential for nicotine-augmented cross-presentation, BM-DC-based CTL priming, and thereby the superior cytolytic capacity of DC-activated CTL. Importantly, K48-linked ubiquitination was verified to be necessary for nicotine-augmented endosomal recruitments of p97 and Sec61. Importantly, mannose receptor (MR), which is an important antigenic receptor for cross-presentation, was exactly catalyzed with K48-linked ubiquitination by the treatment with nicotine. Thus, these data suggested that K48-linked ubiquitination contributes to the superior adaptive immunity of nicotine-administrated BM-DC. Regulating K48-linked ubiquitination might have therapeutic potential for DC-mediated immune therapy.

4.
Vaccines (Basel) ; 8(3)2020 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957586

RESUMEN

Cross-presentation in dendritic cells (DC) requires the endosomal relocations of internalized antigens and the endoplasmic reticulum protein Sec61. Despite the fact that endotoxin-containing pathogen and endotoxin-free antigen have different effects on protein kinase B (Akt) and I-kappa B Kinase α/ß (IKKα/ß) activation, the exact roles of Akt phosphorylation, IKKα or IKKß activation in endotoxin-containing pathogen-derived cross-presentation are poorly understood. In this study, endotoxin-free ovalbumin supplemented with endotoxin was used as a model pathogen. We investigated the effects of endotoxin-containing pathogen and endotoxin-free antigen on Akt phosphorylation, IKKα/ß activation, and explored the mechanisms that the endotoxin-containing pathogen orchestrating the endosomal recruitment of Sec61 of the cross-presentation in bone marrow precursor cells (BMPC). We demonstrated that endotoxin-containing pathogen and endotoxin-free antigen efficiently induced the phosphorylation of Akt-IKKα/ß and Akt-IKKα, respectively. Endotoxin-containing pathogen derived Akt+ IKKα/ß+ Rab5+ signalosome, together with augmented the recruitment of Sec61 toward endosome, lead to the increased cross-presentation in BMPC. Importantly, the endosomal recruitment of Sec61 was partly mediated by the formation of Akt+ IKKα/ß+ signalosome. Thus, these data suggest that Akt+ IKKα/ß+ Rab5+ signalosome contribute to endotoxin-containing pathogen-induced the endosomal recruitment of Sec61 and the superior efficacy of cross-presentation in BMPC.

5.
J Leukoc Biol ; 108(1): 177-188, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32293057

RESUMEN

This study tested the hypothesis that PI3K-Akt activity contributes to the superior immune function of IL-15-administrated bone marrow precursor cells (BMPC). Our previous studies revealed that PI3K-Akt play vital role in dendritic cells (DCs) cross-presentation and DC-based CTL priming. Despite the fact that IL-15 serves multiple functions in its therapeutic potential for the induction and maintenance of T cell response, the exact role of PI3K-Akt in IL-15 increased adaptive immunity is still poorly understood. In this study, we demonstrated that ex vivo IL-15 administration increased BMPC capability of antigen uptake and the expression of costimulatory molecules (such as CD80 and 4-1BB(CD137) ligand [4-1BBL]) and MHC class I molecule via PI3K-Akt pathway. Importantly, PI3K-Akt activity was not only necessary for IL-15 augmented BMPC cross-presentation and CTL priming, but also facilitated IL-15 increased therapeutic potential of the cytolytic capacity and maintenance of BMPC-activated T cells. Thus, these data suggested that PI3K-Akt activity contribute to the superior immune function of IL-15-administrated BMPC and thereby might be therapeutic potential for adaptive immunity.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Células de la Médula Ósea/inmunología , Interleucina-15/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Animales , Antígenos/metabolismo , Reactividad Cruzada/efectos de los fármacos , Citosol/metabolismo , Femenino , Memoria Inmunológica/efectos de los fármacos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Linfocitos T Citotóxicos/inmunología
6.
Exp Cell Res ; 381(2): 165-171, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31100307

RESUMEN

Matrix metalloproteinases (MMPs) and the epithelial-mesenchymal transition (EMT) contribute to metastasis. As shown in our previous studies, interleukin-6 (IL-6) induces ATM phosphorylation to increase MMP expression and metastasis in lung cancer. However, the exact roles of ATM activation in the IL-6-induced epithelial-mesenchymal transition and lung cancer metastasis are currently unclear. Here, ATM phosphorylation exerts its pro-metastatic effect via vimentin-mediated epithelial-mesenchymal transition, which was supported by the evidence described below. Firstly, IL-6 treatment increases vimentin expression via the ATM-NF-κB pathway. Second, ATM inactivation not only abolishes IL-6-induced increases in vimentin expression but also inhibits IL-6-induced nest formation in a xenograft lung metastasis model. Moreover, close positive correlations were observed between ATM phosphorylation and vimentin upregulation, IL-6 levels and metastasis in lung cancer specimens. Hence, ATM modulates vimentin expression to facilitate IL-6-induced epithelial-mesenchymal transition and metastasis in lung cancer, indicating that ATM and vimentin might be potential therapeutic targets for inflammation-associated lung cancer metastasis.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Interleucina-6/farmacología , Neoplasias Pulmonares/patología , Carcinoma Pulmonar de Células Pequeñas/patología , Vimentina/genética , Células A549 , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Vimentina/metabolismo
7.
J Immunol Res ; 2018: 5070573, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30069488

RESUMEN

PYR-41 and thalidomide have therapeutic effects on inflammation-associated diseases with side effects such as tumorigenesis. Cross-presentation allows dendritic cells (DC) to present endogenous antigen and induce protective immunity against microbe infection and tumors. But, up to now, the effects of PYR-41 and thalidomide on cross-presentation are still uncertain. In this study, we investigated the effect and mechanism of PYR-41 and thalidomide on DC cross-presentation by observing Myddosome formation, endosomal recruitment of p97 and Sec61, NF-κB activation, and cross-priming ability. We demonstrated that the inhibition of endosomal recruitment of p97 and Sec61, together with attenuated NF-κB activation and Myddosome formation, contributes to PYR-41- and thalidomide-impaired cross-presentation and thereby reverses cross-activation of T cells. These observations suggest that NF-κB signaling and p97 and Sec61 molecules are candidates for dealing with the side effects of PYR-41 and thalidomide.


Asunto(s)
Benzoatos/toxicidad , Reactividad Cruzada/efectos de los fármacos , Células Dendríticas/efectos de los fármacos , Furanos/toxicidad , Inmunosupresores/toxicidad , Pirazoles/toxicidad , Talidomida/toxicidad , Animales , Reactividad Cruzada/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Endosomas/efectos de los fármacos , Endosomas/inmunología , Endosomas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/efectos de los fármacos , Complejos Multiproteicos/inmunología , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Proteínas Nucleares/metabolismo , Canales de Translocación SEC/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
8.
Oncol Rep ; 39(6): 2688-2694, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29620232

RESUMEN

Tumor necrosis factor α­induced protein 8 (TIPE) is highly expressed in many types of malignancies. Apoptosis is the process of programmed cell death which maintains the balance of cell survival and death. TIPE is involved in the carcinogenesis of many tumor types, yet the exact role of TIPE in defective apoptosis­associated carcinogenesis remains uncertain. In the present study, TIPE­overexpressing Raw264.7 and EL4 cells and vector control cells were treated with 4 mJ/cm2 ultraviolet radiation or 2 µg/ml cisplatin. Following ultraviolet irradiation, TIPE overexpression decreased the percentage of apoptotic cells as detected by flow cytometric and reversed the cisplatin­mediated decrease in mitochondrial membrane potential by JC­1 assay. Western blot analyses also revealed that TIPE overexpression inhibited cisplatin­induced activation of caspase­3 and ­9 and PARP. Secondly, TIPE overexpression increased the levels of phosphorylated JNK, MEK and p38. Moreover, inhibition of JNK and p38, but not MEK, efficiently abolished the cell pro­survival effect of TIPE. Most importantly, an in vivo tumor implantation model revealed that TIPE overexpression augmented the volume and weight of the implanted tumors, indicating that TIPE facilitated tumor formation. We found that TIPE exhibited an anti­apoptotic effect via JNK and p38 activation, which ultimately promoted tumor. Hence, the present study revealed that activation of JNK and p38 kinases contribute to the TIPE­mediated anti­apoptotic effect, indicating that JNK and p38 may be potential therapeutic molecules for TIPE overexpression­associated diseases.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Cisplatino/farmacología , MAP Quinasa Quinasa 4/metabolismo , Neoplasias Experimentales/metabolismo , Transfección , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Ratones , Ratones Desnudos , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Células RAW 264.7
9.
Int J Oncol ; 51(3): 987-995, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28766689

RESUMEN

Cholinergic anti-inflammatory pathway has therapeutic effect on inflammation-associated diseases. However, the exact mechanism of nicotine-mediated anti-inflammatory effect is still unclear. TIPE2, a new member of tumor necrosis factor-α-induced protein-8 family, is a negative regulator of immune homeostasis. However, the roles of TIPE2 in cholinergic anti-inflammatory effect are still uncertain. Here, we demonstrated that nicotine exerts its anti-inflammatory effect by TIPE2 upregulation and phosphorylated stat3 mediated the inhibition of NF-κB activation, which was supported by the following evidence: firstly, both nicotine and TIPE2 inhibit pro-inflammatory cytokine release via NF-κB inactivation. Secondly, nicotine upregulates TIPE2 expression via α7 nicotinic acetylcholine receptor. Moreover, the enhancement of stat3 phosphorylation and decrease of LPS-induced p65 translocation were achieved by nicotine treatment. Importantly, nicotine treatment augments the interaction of phosphorylated stat3 and p65, indicating that the inhibitory effect of nicotine on NF-κB activation was mediated with protein-protein interactions. Hence, this study revealed that TIPE2 upregulation and stat3 phosphorylation contribute to nicotine-mediated anti-inflammation effect, indicating that TIPE2 and stat3 might be potential molecules for dealing with inflammation-associated diseases.


Asunto(s)
Inflamación/tratamiento farmacológico , Péptidos y Proteínas de Señalización Intracelular/genética , Nicotina/administración & dosificación , Factor de Transcripción STAT3/genética , Factor de Transcripción ReIA/genética , Animales , Colinérgicos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , Inflamación/patología , Ratones , FN-kappa B/genética , Fosforilación/efectos de los fármacos , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética
10.
Oncotarget ; 7(38): 62070-62083, 2016 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-27556690

RESUMEN

Despite that ataxia-telangiectasia mutated (ATM) is involved in IL-6 promoted lung cancer chemotherapeutic resistance and metastasis, the exact role of ATM in tumor necrosis factor-alpha (TNF-α) increasing tumor migration is still elusive. In the present study, we demonstrated that TNF-α promoted lung cancer cell migration by up-regulation of matrix metalloproteinase-13 (MMP-13). Notably, by gene silencing or kinase inhibition, we proposed for the first time that ATM is a key up-stream regulator of TNF-α activated ERK/p38-NF-κB pathway. The existence of TNF-α secreted in autocrine or paracrine manner by components of tumor microenvironment highlights the significance of TNF-α in inflammation-associated tumor metastasis. Importantly, in vivo lung cancer metastasis test showed that ATM depletion actually reduce the number of metastatic nodules and cancer nests in lung tissues, verifying the critical role of ATM in metastasis. In conclusion, our findings demonstrate that ATM, which could be activated by lung cancer-associated TNF-α, up-regulate MMP-13 expression and thereby augment tumor metastasis. Therefore, ATM might be a promising target for prevention of inflammation-associated lung cancer metastasis.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células A549 , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Silenciador del Gen , Humanos , Inflamación , Leucocitos Mononucleares/citología , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , FN-kappa B/metabolismo , Metástasis de la Neoplasia , ARN Interferente Pequeño/metabolismo , Bazo/citología , Regulación hacia Arriba
11.
Oncol Rep ; 36(1): 455-61, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27221310

RESUMEN

Hypoxia always exists in the processes involved in the development of lung cancer and contributes to an acidic microenvironment. Despite that hypoxia in the tumor microenvironment is associated with the formation of chemotherapeutic resistance, the exact role of an acidic microenvironment in the development of hypoxia-induced lung cancer multidrug resistance is still unknown. In the present study, we acidized the medium with 2-(N-morpholino)-ethanesulfonic acid (MES monohydrate) to mimic the acidic tumor microenvironment and observed the effects of acidification on lung cancer cell viability, the expression of ATP-binding cassette sub-family G member 2 (ABCG2) and myeloid cell leukemia­1 (Mcl-1), and activation of the PI3K-Akt pathway. Thereafter, we investigated the mechanisms involved in the acidification-induced expression of ABCG2 and Mcl-1, and the potential therapeutic strategy to overcome acidification-associated multidrug resistance formation. We demonstrated that acidification obviously increased the expression of ABCG2 and Mcl-1 via PI3K­Akt­mTOR-S6 pathway activation and contributed to multidrug resistance. Inhibition of PI3K-Akt activity efficiently abolished the effect of acidification on cell viability, indicating that the PI3K-Akt pathway may include potential therapeutic target molecules in acidized microenvironment-associated lung cancer chemotherapeutic resistance.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Regulación hacia Arriba/genética , Células A549 , Línea Celular Tumoral , Supervivencia Celular/genética , Resistencia a Múltiples Medicamentos/genética , Humanos , Hipoxia/genética , Neoplasias Pulmonares/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteínas Quinasas S6 Ribosómicas/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética , Activación Transcripcional/genética , Microambiente Tumoral/genética
12.
Oncotarget ; 7(25): 38451-38466, 2016 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-27224911

RESUMEN

Cross-presentation by dendritic cells (DCs) requires surface molecules such as lectin, CD40, langerin, heat shock protein, mannose receptor, mediated endocytosis, the endosomal translocation of internalized antigen, and the relocation of transporter associated with antigen processing (TAP). Although the activation of α7 nicotinic acetylcholine receptor (α7 nAchR) up-regulate surface molecule expression, augment endocytosis, and enhance cross-presentation, the molecular mechanism of α7 nAchR activation-increased cross-presentation is still poorly understood. In this study, we investigated the role of mannose receptor in nicotine-increased cross-presentation and the mechanism that endotoxins orchestrating the recruitment of TAP toward endosomes. We demonstrated that nicotine increase the expressiones of mannose receptor and Toll-like receptor 4 (TLR4) via PI3K-Akt-mTOR-p70S6 pathway. Both endosomal translocation of mannose receptor-internalized antigens and TLR4 sig- naling are necessary for nicotine-augmented cross-presentation and cross-priming. Importantly, the recruitment of TAP toward endosomes via TLR4-MyD88-IRAK4 signaling contributes to nicotine-increased cross-presentation and cross-activation of T cells. Thus, these data suggest that increased recruitment of TAP to Ag-containing vesicles contributes to the superior cross-presentation efficacy of α7 nAchR activated DCs.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Células Dendríticas/metabolismo , Endosomas/metabolismo , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Nicotina/farmacología , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 4/metabolismo , Transportadoras de Casetes de Unión a ATP/inmunología , Animales , Presentación de Antígeno/efectos de los fármacos , Reactividad Cruzada , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Femenino , Humanos , Activación de Linfocitos , Receptor de Manosa , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo
13.
Oncotarget ; 6(38): 40719-33, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26528698

RESUMEN

Our previous studies show that the phosphorylation of ataxia-telangiectasia mutated (ATM) induced by interleukin 6 (IL-6) treatment contributes to multidrug resistance formation in lung cancer cells, but the exact role of ATM activation in IL-6 increased metastasis is still elusive. In the present study, matrix metalloproteinase-3 (MMP-3) and MMP-13 were firstly demonstrated to be involved in IL-6 correlated cell migration. Secondly, IL-6 treatment not only increased MMP-3/MMP-13 expression but also augmented its activities. Thirdly, the inhibition of ATM phosphorylation efficiently abolished IL-6 up-regulating MMP-3/MMP-13 expression and increasing abilities of cell migration. Most importantly, the in vivo test showed that the inhibition of ATM abrogate the effect of IL-6 on lung cancer metastasis via MMP-3/MMP-13 down-regulation. Taken together, these findings demonstrate that IL-6 inducing ATM phosphorylation increases the expression of MMP-3/MMP-13, augments the abilities of cell migration, and promotes lung cancer metastasis, indicating that ATM is a potential target molecule to overcome IL-6 correlated lung cancer metastasis.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Interleucina-6/farmacología , Neoplasias Pulmonares/secundario , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/genética , Western Blotting , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Técnicas para Inmunoenzimas , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Anal Cell Pathol (Amst) ; 2015: 741487, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26351626

RESUMEN

Our previous studies showed that α7 nicotinic acetylcholine receptor (nAchR) agonist nicotine has stimulatory effects on murine bone marrow-derived semimature DCs, but the effect of nicotine on peripheral blood mononuclear cell- (PBMC-) derived human semimature dendritic cells (hu-imDCs) is still to be clarified. In the present study, hu-imDCs (cultured 4 days) were conferred with ex vivo lower dose nicotine stimulation and the effect of nicotine on surface molecules expression, the ability of cross-presentation, DCs-mediated PBMC priming, and activated signaling pathways were determined. We could demonstrate that the treatment with nicotine resulted in increased surface molecules expression, enhanced hu-imDCs-mediated PBMC proliferation, upregulated release of IL-12 in the supernatant of cocultured DCs-PBMC, and augmented phosphorylation of Akt and ribosomal protein S6. Nicotine associated with traces of LPS efficiently enhanced endosomal translocation of internalized ovalbumin (OVA) and increased TAP-OVA colocalization. Importantly, the upregulation of nicotine-increased surface molecules upregulation was significantly abrogated by the inhibition of Akt kinase. These findings demonstrate that ex vivo nicotine stimulation augments hu-imDCs surface molecules expression via Akt-S6 pathway, combined with increased Ag-presentation result in augmented efficacy of DCs-mediated PBMC proliferation and Th1 polarization.


Asunto(s)
Células Dendríticas/citología , Leucocitos Mononucleares/citología , Nicotina/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína S6 Ribosómica/metabolismo , Transducción de Señal/efectos de los fármacos , Vacunación , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Interleucina-12/metabolismo , Lipopolisacáridos/farmacología , Ovalbúmina/metabolismo , Transporte de Proteínas/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
Int J Oncol ; 46(1): 254-64, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25339267

RESUMEN

Tumor necrosis factor-α-induced protein 8-like 2 (TNFAIP8L2, TIPE2), which belongs to the TNF-α-induced protein 8 family, is a negative regulator of immune homeostasis. Although pro-inflammatory cytokines such as TNF-α have been reported to be involved in liver carcinoma metastasis, the effect of TIPE2 on hepatocellular carcinoma metastasis remains unknown. We demonstrate that TNF-α clearly augments MMP-13/MMP-3 expression and promotes cell migration in HepG2 cells through activation of the Erk1/2-NF-κB pathways. Interestingly, in addition to human PBLs, macrophages and fibroblasts, liver cancer cells specifically express TNF-α following LPS treatment. Most importantly, TIPE2 overexpression efficiently abrogates the effects of LPS on TNF-α secretion and abolishes the effects of TNF-α on MMP-13/MMP-3 upregulation, cell migration and Erk1/2-NF-κB activation. Taken together, these findings demonstrate that TIPE2 was able to suppress TNF-α-induced hepatocellular carcinoma metastasis by inhibiting Erk1/2 and NF-κB activation, indicating that both TNF-α and TIPE2 might be potential targets for the treatment of HCC metastasis.


Asunto(s)
Carcinoma Hepatocelular/patología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neoplasias Hepáticas/patología , Factor de Necrosis Tumoral alfa/farmacología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Hep G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , FN-kappa B/metabolismo , Metástasis de la Neoplasia , Transfección
16.
PLoS One ; 9(8): e105636, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25144375

RESUMEN

Dendritic cells are able to present Ag-derived peptides on MHC class I and II molecules and induce T cells priming. Lipopolysaccharides (LPS), an activator of Toll-like 4 receptor (TLR4) signaling, has been demonstrated to facilitate Ag-presentation, up-regulate surface molecules expression but impair T cells priming. In this study, we investigated the effect of LPS on nicotine-enhanced DCs-dependent T cells priming and the mechanisms of LPS orchestrating the immunosuppressive program. We could demonstrate that the treatment with LPS resulted in increased surface molecules expression, enhanced Ag-presentation, up-regulated release of TGF-beta, TNF-alpha, IL-6, and IFN-beta. Concomititantly, the upregulation of IFN-beta in DCs induces the up-regulation of coinhibitory molecules B7H1 and GITRL, which cause an impaired activation of naïve Ag-specific T cells and the induction of T cell tolerance by enhancing B7H1-PD-1 interactions and promoting GITRL-GITL facilitated Treg generation, respectively. These data provide a mechanistic basis for the immunomodulatory action of IFN-beta which might open new possibilities in the development of therapeutic approaches aimed at the control of excessive immune response and persistent infection.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Antígeno B7-H1/inmunología , Interferón beta/inmunología , Lipopolisacáridos/farmacología , Linfocitos T/inmunología , Factores de Necrosis Tumoral/inmunología , Regulación hacia Arriba/efectos de los fármacos , Animales , Presentación de Antígeno/inmunología , Células Cultivadas , Células Dendríticas/inmunología , Femenino , Tolerancia Inmunológica/efectos de los fármacos , Interleucina-6/inmunología , Ratones , Factor de Crecimiento Transformador beta/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Regulación hacia Arriba/inmunología
17.
Cancer Sci ; 105(9): 1220-7, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24988892

RESUMEN

Although it is known that ataxia-telangiectasia mutated (ATM) and interleukin 6 (IL-6) contribute to multiple drug resistance (MDR) in tumor chemotherapy, the exact role of ATM activation in MDR resulting from increased IL-6 expression is still unclear. In the present study, we demonstrate that the activation of the ATM-NF-kappaB pathway, resulting from increased IL-6 expression, plays a central role in augmented chemoresistance in lung cancer cell lines. This result was supported by the increased expressions of Bcl-2, Mcl-1, Bcl-xl, and the upregulation of MDR-associated protein ABCG2. The higher level of IL-6 reveals not only higher ATM/NF-kappaB activity but also increased expressions of ABCG2, Bcl-2, Mcl-1 and Bcl-xl. Most importantly, lung cancer cells themselves upregulated IL-6 secretion by activating the p38/NF-kappaB pathway through treatment with cisplatin and camptothecin. Taken together, these findings demonstrate that chemotherapeutic agents increase IL-6 expression, hence activating the ATM/NF-kappaB pathway, augmenting anti-apoptotic protein expression and contributing to MDR. This indicates that both IL-6 and ATM are potential targets for the treatment of chemotherapeutic resistance in lung cancer.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Resistencia a Antineoplásicos , Interleucina-6/fisiología , FN-kappa B/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Camptotecina/farmacología , Línea Celular Tumoral , Cisplatino/farmacología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Imidazoles/farmacología , Neoplasias Pulmonares , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Nitrilos/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Piridinas/farmacología , Sulfonas/farmacología , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Int J Mol Med ; 33(4): 925-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24481039

RESUMEN

Epidemiological data have indicated that smoking tobacco can decrease the risk of developing Alzheimer's disease (AD). Nicotine, a main component of tobacco, has been shown to have therapeutic effects in AD. The aim of the present study was to assess the neuroprotective effects of nicotine against toxicity induced by ß-amyloid (Aß) in relation to cell apoptosis, and to elucidate the role of the activation of the Erk1/2-p38-JNK pathway and the modulation of anti-apoptotic proteins in the nicotine-induced neuroprotective effects. We performed in vitro and in vivo experiments using SH-SY5Y cells and C57BL/6 mice, respectively. The effects of nicotine on cell apoptosis were determined by flow cytmetry and microscopic observation. The effects of nicotine on the expression of anti-apoptotic proteins were also determined by western blot analysis. Our results demonstrated that nicotine protected the SH-SY5Y cells against Aß25-35-induced toxicity by inhibiting apoptosis and upregulating the expression of anti-apoptotic proteins. As shown by our in vivo experiments, nicotine effectively ameliorated the impairment in spatial working memory induced by Aß25-35; this was confirmed by a Morris water maze navigation test and further supported by the upregulation of Bcl-2 in the hippocampus of Aß25-35-injected mice treated with nicotine. The phosphorylation of Erk1/2, p38 and JNK increased following treatment with nicotine in the SH-SY5Y cells, whereas caspase-3 activation was inhibited by treatment with nicotine prior to exposure to Aß25-35. Of note, these effects of nicotine against Aß25-35-induced damage were abolished by inhibitors of Erk1/2, p38 and JNK phosphorylation. These findings suggest that nicotine prevents Aß25-35-induced neurotoxicity through the inhibition of neuronal apoptosis, and may thus prove to be a potential preventive or therapeutic agent for AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fármacos Neuroprotectores/farmacología , Nicotina/farmacología , Fragmentos de Péptidos/toxicidad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Trastornos del Conocimiento/tratamiento farmacológico , Trastornos del Conocimiento/fisiopatología , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Masculino , Memoria a Corto Plazo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Neurotoxinas/toxicidad , Nicotina/uso terapéutico , Factores de Tiempo , Proteína bcl-X/metabolismo
19.
Oncol Rep ; 31(3): 1480-8, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24399025

RESUMEN

Although nicotine is a risk factor for carcinogenesis and atherosclerosis, epidemiological data indicate that nicotine has therapeutic benefits in treating Alzheimer's disease. Our previous studies also showed that nicotine-treated dendritic cells have potential antitumor effects. Hence, the precise effects of nicotine on the biological characterizations of cells are controversial. The aim of the present study was to assess the roles of α7 nicotinic acetylcholine receptors (nAChRs), Erk1/2-p38-JNK and PI3K-Akt pathway in nicotine-mediated proliferation and anti-apoptosis effects. The results firstly showed that nicotine treatment clearly augmented cell viability and upregulated PCNA expression in both Raw264.7 and El4 cells. Meanwhile, nicotine afforded protection against cisplatin-induced toxicity through inhibiting caspase-3 activation and upregulating anti-apoptotic protein expression. Further exploration demonstrated that nicotine efficiently abolished cisplatin-promoted mitochondria translocation of Bax and the release of cytochrome c. The pretreatment of α-bungarotoxin and tubocurarine chloride significantly attenuated nicotine-augmented cell viability, abolished caspase-3 activation and α7 nAChR upregulation. Both Erk-JNK-p38 and PI3K-Akt signaling pathways could be activated by nicotine treatment in Raw264.7 and El4 cells. Notably, when Erk-JNK and PI3K-Akt activities were inhibited, nicotine-augmented cell proliferation and anti-apoptotic effects were abolished accordingly. The results presented here indicate that nicotine could achieve α7 nAChR-mediated proliferation and anti-apoptotic effects by activating Erk-JNK and PI3K-Akt pathways respectively, providing potential therapeutic molecules to deal with smoking-associated human diseases.


Asunto(s)
Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos , Nicotina/farmacología , Agonistas Nicotínicos/farmacología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citocromos c/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Mitocondrias/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
20.
Int J Oncol ; 42(4): 1289-96, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23381786

RESUMEN

Multidrug resistance (MDR) formation is an important problem in lung cancer chemotherapy. Our study showed that both camptothecin and cisplatin could not only induce ATM and NF-κB activation but also upregulate expression of the MDR-related genes ABCG2, MRP2 in NCI-H446 cells. Moreover, camptothecin and cisplatin-induced ABCG2 and MRP2 upregulation could be impaired by ATM and NF-κB inhibitors, indicating a relationship between ATM, NF-κB activation and MDR formation in lung cancer chemo-therapy. Our study indicates that ATM may serve as a potential molecular target for MDR formation in lung cancer chemotherapy.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Antineoplásicos/farmacología , Camptotecina/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Proteínas de Neoplasias/genética , Transducción de Señal , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Proteína 2 Asociada a Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...