Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Immunol ; 22(1): 44, 2021 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-34253166

RESUMEN

BACKGROUND: Transforming growth factor beta (TGF-ß) is a typical immuno-inhibitory cytokine and highly secreted by lung cancer cells. It was supposed that its immunosuppressive effects to NK cell might be related with the altered expression of activating and inhibitory molecules in lung cancer cells. In this study, we examined the expression of NKG2DLs, PD-L1 and PD-L2 in lung cancer cells after treatment of TGF-ß and a TGF-ß inhibitor, Galunisertib (LY2157299). RESULTS: TGF-ß reduced the level of surface proteins of five NKG2DLs without altered transcription levels in lung cancer cells. Galunisertib reversed the effect of TGF-ß on the expression of NKG2DLs. Since MMP inhibitors, MMPi III and MMP2 inhibitor I, restored the reduced expression of NKG2DLs after treatment of TGF-ß, it was thought that TGF-ß induced the expression of MMP2 which facilitated the shedding of the NKG2DLs in cancer cells. However, the expression of PD-L1, L2 were not changed by treatment with TGF-ß or Galunisertib. CONCLUSIONS: Therefore, inhibition of TGF-ß might reverse the immunosuppressive status on immune cells and restore NK cell mediated anticancer immune responses by upregulation of NKG2DLs in cancer cells.


Asunto(s)
Adenocarcinoma Bronquioloalveolar/inmunología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Adenocarcinoma Bronquioloalveolar/tratamiento farmacológico , Citotoxicidad Inmunológica , Regulación hacia Abajo , Proteínas Ligadas a GPI/metabolismo , Humanos , Tolerancia Inmunológica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Pirazoles/farmacología , Quinolinas/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Escape del Tumor
2.
Molecules ; 26(13)2021 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203519

RESUMEN

Histone acetylation is an epigenetic mechanism that regulates the expression of various genes, such as natural killer group 2, member D (NKG2D) ligands. These NKG2D ligands are the key molecules that activate immune cells expressing the NKG2D receptor. It has been observed that cancer cells overexpress histone deacetylases (HDACs) and show reduced acetylation of nuclear histones. Furthermore, HDAC inhibitors are known to upregulate the expression of NKG2D ligands. Humans have 18 known HDAC enzymes that are divided into four classes. At present, it is not clear which types of HDAC are involved in the expression of NKG2D ligands. We hypothesized that specific types of HDAC genes might be responsible for altering the expression of NKG2D ligands. In this study, we monitored the expression of NKG2D ligands and major histocompatibility complex (MHC) class I molecules in lung cancer cells which were treated with six selective HDAC inhibitors and specific small interfering RNAs (siRNAs). We observed that treatment with FK228, which is a selective HDAC1/2 inhibitor, also known as Romidepsin, induced NKG2D ligand expression at the transcriptional and proteomic levels in two different lung cancer cell lines. It also caused an increase in the susceptibility of NCI-H23 cells to NK cells. Silencing HDAC1 or HDAC2 using specific siRNAs increased NKG2D ligand expression. In conclusion, it appears that HDAC1 and HDAC2 might be the key molecules regulating the expression of NKG2D ligands. These results imply that specifically inhibiting HDAC1 and HDAC2 could induce the expression of NKG2D ligands and improve the NK cell-mediated anti-cancer immunity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/inmunología , Histona Desacetilasa 1/inmunología , Histona Desacetilasa 2/inmunología , Inmunidad Celular/inmunología , Neoplasias Pulmonares/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Proteínas de Neoplasias/inmunología , Células A549 , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Humanos , Células Asesinas Naturales , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Subfamilia K de Receptores Similares a Lectina de Células NK/genética , Proteínas de Neoplasias/genética
3.
Med Sci Monit ; 26: e926395, 2020 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-33139690

RESUMEN

BACKGROUND The purpose of this study was to investigate the effects of sevoflurane on cancer immunosurveillance and metastasis in non-small-cell lung cancer (NSCLC). MATERIAL AND METHODS NCI-H23 cells, a human NSCLC cell line, were incubated with or without sevoflurane at the concentrations of 0, 12.5, 25, 50, 100, and 200 µM for 6 h. Cell viability, the expression of natural killer group 2, member D ligands (NKG2D ligands: UL16-binding proteins 1-3 [ULBP1-3] and major histocompatibility complex class I chain-related molecules A/B [MICA/B]), the expression of matrix metalloproteinases (MMPs), NK cell-mediated cytotoxicity, and cancer cell migration were measured. RESULTS At 12.5, 25, 50, and 100 µM, sevoflurane increased the expression of NKG2D ligands (ULBP2-3 and MICA, ULBP1-3, ULBP1-3, and ULBP1, respectively). Sevoflurane decreased the expression of NKG2D ligands at 200 µM (MICA/B). NK cell-mediated lysis of NCI-H23 cells at 200 µM sevoflurane was significantly reduced compared with the control (P=0.025; target cell: effect cell=1: 10). Sevoflurane increased the expression of MMP-1, -2, and -9 and increased cell migration in NCI-H23 cells at 50, 100, and 200 µM (P=0.001, 0.035, and 0.039, respectively, compared with the control after 18 h of wound formation). CONCLUSIONS Sevoflurane could suppress NKG2D-mediated NK cell cytotoxicity and increased expression of MMPs and migration in NCI-H23 cells. Further research is needed to determine the effects of sevoflurane on cancer immunosurveillance and metastasis in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/inmunología , Inmunidad/efectos de los fármacos , Neoplasias Pulmonares/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Sevoflurano/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Citotoxicidad Inmunológica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Cicatrización de Heridas/efectos de los fármacos
4.
Korean J Parasitol ; 58(6): 689-694, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33412774

RESUMEN

Strongyloidiasis is caused by Strongyloides stercoralis and is one of the most neglected tropical diseases in tropical and subtropical regions. Although several strongyloidiasis cases have been reported in Korea, genetic analysis of Korean isolates is still incomplete. In this study, a parasite was isolated from a 61-year-old man diagnosed with strongyloidiasis during the treatment of lymphoma on his retroperitoneal lymph node. Diffuse symmetric wall thickening from the ascending to descending colon and a nematode-infected intestine was observed following microscopic examination. Genomic DNA was isolated from a patient tissue block, and S. stercoralis was identified by PCR and sequencing (18S rDNA). In order to determine phylogenetic location of a Korean isolate (named KS1), we analyzed cox1 gene (500-bp) and compared it with that from 47 previous S. stercoralis isolates (28 human isolates and 19 canid isolates) from Asian countries. Our results showed that phylogenetic tree could clearly be divided into 5 different groups according to hosts and regions. KS1 was most closely related with the Chinese isolates in terms of genetic distance.


Asunto(s)
Filogenia , Strongyloides stercoralis/genética , Estrongiloidiasis/parasitología , Animales , Asia , Pueblo Asiatico , ADN de Helmintos/genética , ADN de Helmintos/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad , Análisis de Secuencia de ADN , Strongyloides stercoralis/clasificación , Strongyloides stercoralis/aislamiento & purificación
5.
Biochem Biophys Res Commun ; 514(4): 1278-1284, 2019 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-31113615

RESUMEN

Our previous study reported that cancer upregulated gene (CUG)2, a novel oncogene, induces both faster cell migration and anti-cancer drug resistance. We thus wonder whether CUG2 also induces stemness, a characteristic of cancer stem cells (CSCs) and further examine the molecular mechanism of this phenotype. To test that CUG2 induces stemness, we examined expression of stemness-related factors. Overexpression of CUG2 enhanced expression levels of stemness-related factors in human lung carcinoma A549 and immortalized bronchial BEAS-2B cells. Consequently, CUG2 increased cellular spherical cluster forming ability. Overexpression of CUG2 also induced tumor formation in xenotransplanted nude mice whereas transplantation of control cells failed to, implying that CUG2 possesses malignant tumorigenic potential. We paid attention to nucleophosmin (NPM1) for its known interaction with CUG2. Suppression of NPM1 hindered the CUG2-mediated stemness-like phenotypes and diminished TGF-ß transcriptional activity and signaling. TGF-ß increased stemness-like phenotypes in the control cells whereas TGF-ß inhibitor blocked induction of the phenotypes, indicating that NPM1 is required for CUG2-mediated stemness-like phenotypes through TGF-ß signaling. Furthermore, the suppression of Smad- and non-Smad-dependent TGF-ß signaling pathways also prevented CUG2 from inducing stemness-like phenotypes. Altogether, we suggest that the novel CUG2 oncogene promotes cellular transformation and stemness, mediated by nuclear NPM1 protein and TGF-ß signaling.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Células A549 , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nucleofosmina , Fenotipo
6.
Biochem Biophys Res Commun ; 484(2): 298-303, 2017 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-28126340

RESUMEN

The cancer/testis (CT) antigen NY-SAR-35 gene is located on the X chromosome and is aberrantly expressed in various cancers but not in normal tissues, other than testes. Previously, we reported the expression of NY-SAR-35 enhanced cell growth, proliferation, and invasion in HEK293 and cancer cells. To extend understanding of the NY-SAR-35 gene, we used a next generation sequencing (NGS) approach. NY-SAR-35 expression induced growth, proliferation, metastasis, and stemness genes, as indicated by the up-regulations of CXCR4, EpCAM, CD133, and CD44, at the mRNA and protein levels. The expression of NY-SAR-35 in HEK293 cells significantly increased ERK phosphorylation, but not the phosphorylation of AKT. In HEK293/NY-SAR-35 cells, the expressions of pro-apoptotic proteins, including p53, Bax, and p21, were reduced and that of cyclin E was increased. Also, NY-SAR-35 increased the expressions of pluripotency genes (Nanog, Oct-4, and Sox2) and the ability of HEK293 cells to form colonies. Taken together, the present study indicates NY-SAR-35 functions as a CT antigen that triggers oncogenesis and self-renewal.


Asunto(s)
Antígeno AC133/metabolismo , Antígenos de Neoplasias/fisiología , Molécula de Adhesión Celular Epitelial/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Antígeno AC133/genética , Molécula de Adhesión Celular Epitelial/genética , Regulación de la Expresión Génica/fisiología , Células HEK293 , Humanos , Receptores de Hialuranos/genética , Regulación hacia Arriba
7.
J Arthroplasty ; 32(3): 824-829, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27667531

RESUMEN

BACKGROUND: Both rotating-platform (RP) mobile-bearing and medial-pivot (MP) fixed-bearing prostheses allow axial femorotibial rotation using a highly conforming polyethylene insert. However, limited comparative data are available between the 2 designs. This study was performed to compare the midterm clinical outcomes and patient-reported outcome measures (PROMs) of RP and MP prostheses. METHODS: We retrospectively reviewed the records of 52 total knee arthroplasties using RP mobile-bearing prosthesis and 49 total knee arthroplasties using MP fixed prosthesis with a minimum follow-up period of 5 years. Clinical and radiological outcomes, failure rates, and PROMs, including the Western Ontario and McMaster Universities Osteoarthritis Index score and satisfaction, were compared. RESULTS: There was no difference in clinical or radiographic outcomes (P > .1 for all comparisons), with the exception of the larger flexion contracture (FC) in the MP group (0.3° in RP vs 2.3° in MP, P < .01). No failure in either group was recorded during the study period. PROMs were comparable (P > .1 in all comparisons), with the exception of higher satisfactions in the RP group while performing light household duties (P < .01) and leisure or recreational activities (P = .014) in patients without FC. CONCLUSION: The midterm clinical results with both the RP mobile-bearing and MP fixed-bearing prostheses were satisfactory. Although both prostheses provided comparable PROMs, patients with an RP prosthesis were more satisfied than those with an MP prosthesis for highly demanding activities that are strongly associated with the presence of postoperative FC.


Asunto(s)
Artroplastia de Reemplazo de Rodilla/instrumentación , Prótesis de la Rodilla , Medición de Resultados Informados por el Paciente , Diseño de Prótesis , Anciano , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Polietileno , Radiografía , Rango del Movimiento Articular , Estudios Retrospectivos
8.
Exp Cell Res ; 330(2): 451-459, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25218028

RESUMEN

In the present study, we investigated whether celecoxib could induce the expression of NKG2D ligands in clonogenic colon cancer cells, and increase their susceptibility to NK cell-mediated cell death. Celecoxib and its non-coxib analog, 2,5-dimethyl celecoxib, induced ULBP-1 and DR5 in both COX-2 negative HCT-15 cells and COX-2 positive HT-29 cells. Celecoxib increased their susceptibility to NK92 cells in both DELFIA assay and soft agar colony forming assay. The inducibility of ULBP-1 and DR5 by celecoxib was not different between CD44- and CD44+ HCT-15 cells, and CD133- and CD133+ HT-29 cells. Celecoxib increased the susceptibility of highly clonogenic CD44+ HCT-15 and CD133+ HT-29 cells to NK92 cells, at least comparable to less clonogenic CD44- HCT-15 and CD133- HT-29 cells, respectively. In addition, celecoxib induced CHOP, and thapsigargin, an inducer of ER (endoplasmic reticulum) stress, induced DR5 but not ULBP1 in HCT-15. Taken together, these findings suggest that celecoxib induces the expression of ULBP-1 as well as DR5 in clonogenic colon cancer cells via COX-2 and ER stress-independent pathways, and increases their susceptibility to NK cells.


Asunto(s)
Neoplasias del Colon/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Células Asesinas Naturales/inmunología , Pirazoles/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Sulfonamidas/farmacología , Antígeno AC133 , Antígenos CD/biosíntesis , Celecoxib , Ciclooxigenasa 2/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proteínas Ligadas a GPI/biosíntesis , Glicoproteínas/biosíntesis , Células HT29 , Humanos , Receptores de Hialuranos/biosíntesis , Péptidos , Tapsigargina/farmacología , Factor de Transcripción CHOP/biosíntesis
9.
Water Sci Technol ; 74(1): 130-7, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27386990

RESUMEN

The effects on sulfur removal and membrane fouling resulting from FeCl(3) addition to an anaerobic fluidized membrane bioreactor (AFMBR) in a staged AFMBR (SAF-MBR) was investigated. Total sulfur removal in the SAF-MBR was 42-59% without FeCl(3) addition, but increased to 87-95% with FeCl(3) addition. Sulfide removal in the AFMBR increased to 90% with addition of FeCl(3) at a molar Fe(3+)/S ratio of 0.54 and to 95% when the ratio was increased to 0.95. Effluent sulfide concentration then decreased to 0.3-0.6 mg/L. Phosphate removals were only 19 and 37% with the above added FeCl(3) ratios, indicating that iron removed sulfide more readily than phosphate. Neither chemical oxygen demand nor biochemical oxygen demand removal efficiencies were affected by the addition of FeCl(3). When the AFMBR permeate became exposed to air, light brown particles were formed from effluent Fe(2+) oxidation to Fe(3+). FeCl(3) addition, while beneficial for sulfide removal, did increase the membrane fouling rate due to the deposition of inorganic precipitates in the membrane pores.


Asunto(s)
Reactores Biológicos/microbiología , Metaloporfirinas/química , Aguas Residuales/química , Purificación del Agua/métodos , Anaerobiosis , Análisis de la Demanda Biológica de Oxígeno , Membranas Artificiales , Sulfuros/química , Azufre/química , Purificación del Agua/instrumentación
10.
Water Sci Technol ; 74(2): 457-65, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27438251

RESUMEN

A bench-scale short-term test, developed to predict the long-term integrity of membranes with potential for use in anaerobic fluidized-bed membrane bioreactors, was used to evaluate several commercial hollow-fiber membranes. It was found that membrane performance varied widely, some membranes failing much more rapidly than others. Also found was that larger sizes of the fluidized media, in this case granular activated carbon (GAC), severely affected membrane structural integrity more than did smaller sizes, as did the method used for membrane attachment. Within the limits studied, the GAC packing ratio had only a minor impact. A decrease in membrane permeability that sometimes resulted during the testing and was caused by the deposition of fine GAC particles could be eliminated without membrane damage through simultaneous chemical cleaning and sonication. This new testing procedure should be useful for selecting membranes and reactor operating conditions to better ensure long-term operating performance of anaerobic fluidized-bed membrane bioreactors.


Asunto(s)
Reactores Biológicos , Carbón Orgánico/análisis , Membranas Artificiales , Eliminación de Residuos Líquidos/métodos , Eliminación de Residuos Líquidos/instrumentación
11.
Immunol Invest ; 43(5): 447-62, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24654594

RESUMEN

Various ex vivo or in vivo loading protocols have been developed or evaluated for the delivery of tumor antigens to dendritic cells (DCs). We compared the antitumor effect of mature DCs electroporation-pulsed (EP/mDC) ex vivo with tumor cell lysate and immature DCs (iDCs) injected into the tumor apoptosed by ionizing radiation (IR/iDC) in lung cancer model. DCs were generated from bone marrow of C57BL/6 mice. Ionizing radiation (IR) was applied at a dose of 10 Gy to the tumor on the right thigh. iDCs were intratumorally injected into the irradiated tumor and EP/mDC was injected subcutaneously in the right flank. DC injection induced strong tumor-specific immunity against Lewis lung carcinoma, as compared with the tumor-bearing control and IR only treated mice. The growth of a distant tumor on the right and left flank was inhibited by IR/iDC and EP/mDC. Particularly, IR/iDC resulted in a more significant inhibition of tumor growth and prolonged survival time. It was related to increase of tumor-specific interferon-gamma, cytotoxicity, and decrease of regulatory T-cells. The results indicate that DCs electroporation-pulsed with tumor cell lysate induce a potent antitumor effect, but that iDCs intratumoral injected into the irradiated tumor induce a more potent antitumor effect.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Neoplasias Pulmonares/inmunología , Animales , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Citotoxicidad Inmunológica , Inyecciones Intralesiones , Inyecciones Subcutáneas , Interferón gamma/biosíntesis , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/terapia , Masculino , Ratones , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
12.
Immunol Invest ; 42(4): 341-55, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23509890

RESUMEN

Dendritic cells (DCs) are potent antigen-presenting cells that can be matured in vitro from immature dendritic cells (iDCs) in the presence of several biological agents such as cytokine cocktail, CD40L, TNF-a and antigen loading, which are necessary and achieved using various protocols, such as lipofection, passive pulse or electroporation. However, these DCs maturation protocols may cause with a significant loss of cells because of cellular attachment and spreading during culturing. Some biomaterials that influence adhesion and development of cells have been used in cell culture techniques, and it was thought that they might be applied on the culture of DCs. In this study, we used polyHEMA, which is a hydrogel coating biomaterial that prevents DCs from adherence, and investigated whether hydrogel coating affects the maturation of iDCs. The efficiency in the generation of mDCs was improved through hydrogel coating procedure and a dendritic cell maturation marker, CD83, was significantly increased in hydrogel-coated culture condition. The antigen-loaded mDCs from electroporation were further expressed the CD83. The mDCs generated in the hydrogel-coated culture condition showed more, longer and thicker dendrites, and produced more amounts of cytokines such as IL-12 and IFN-γ. Therefore, it was suggested that the hydrogel-coated culture condition could improve function of mDCs. Cheol-Hun Son and Jae-Ho Bae contributed equally to this work.


Asunto(s)
Células Dendríticas/citología , Polihidroxietil Metacrilato/farmacología , Anticuerpos/farmacología , Antígenos/química , Adhesión Celular/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Electroporación , Humanos , Hidrogel de Polietilenoglicol-Dimetacrilato , Interferón gamma/inmunología , Interleucina-12/inmunología , Células K562
13.
Oncol Lett ; 25(6): 232, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37153058

RESUMEN

Natural killer (NK) cells play a crucial role in early immune defenses against transformed cells and are used in the therapeutic management of cancer. However, it is difficult to sufficiently obtain high purity activated NK cells for clinical application. The function of NK cells is dependent on the balance of activating and inhibitory signals. Strong and diverse stimuli are required to increase the function of NK cells. Radiotherapy modulates the expression of various immunomodulatory molecules that recruit and activate NK cells. NK cell-mediated antibody-dependent cellular cytotoxicity is one of the most potent cytotoxic effects of NK cells against target cancer cells. To generate activated and irradiated autologous peripheral blood mononuclear cells (PBMCs), cytokine and monoclonal antibody stimulation followed by ionizing radiation was performed in the present study. The expanded NK cells were cultured for 21 days using activated/irradiated autologous PBMCs. Colorectal cancer cells (SW480 and HT-29) were used to analyze the expression of NK group 2D ligands and EGFR by radiation. The cytotoxicity of radiation plus NK cell-based targeted therapy against colorectal cancer cell lines was analyzed using flow cytometry. Activated and irradiated PBMCs exhibited significantly increased expression of various activating ligands that stimulated NK cells. In total, >10,000-fold high-purity activated NK cells were obtained, with negligible T-cell contamination. To confirm the antitumor activity of the NK cells expanded by this method, the expanded NK cells were treated with cetuximab, radiotherapy, or a combination of cetuximab and radiotherapy in the presence of human colorectal cancer cells. Expanded NK cells were effective at targeting human colorectal cancer cells, particularly when combined with cetuximab and radiotherapy. Thus, in the present study, a novel method for high-purity activated NK cell expansion was developed using activated and irradiated PBMCs. In addition, combined radiotherapy and antibody-based immunotherapy with expanded NK cells may be an effective strategy to enhance the efficiency of treatment against colorectal cancer.

14.
Sci Rep ; 13(1): 7656, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37169953

RESUMEN

Pancreatic cancer is difficult to diagnose at the initial stage and is often discovered after metastasis to nearby organs. Gemcitabine is currently used as a standard treatment for pancreatic cancer. However, since chemotherapy for pancreatic cancer has not yet reached satisfactory therapeutic results, adjuvant chemotherapy methods are attempted. It can be expected that combining immune cell therapy with existing anticancer drug combination treatment will prevent cancer recurrence and increase survival rates. We isolated natural killer (NK) cells and co-cultured them with strongly activated autologous peripheral blood mononuclear cells (PBMCs) as feeder cells, activated using CD3 antibody, IFN-r, IL-2, and γ-radiation. NK cells expanded in this method showed greater cytotoxicity than resting NK cells, when co-cultured with pancreatic cancer cell lines. Tumor growth was effectively inhibited in a pancreatic cancer mouse xenograft model. Therapeutic efficacy was increased by using gemcitabine and erlotinib in combination. These findings suggest that NK cells cultured by the method proposed here have excellent anti-tumor activity. We demonstrate that activated NK cells can efficiently inhibit pancreatic tumors when used in combination with gemcitabine-based therapy.


Asunto(s)
Gemcitabina , Neoplasias Pancreáticas , Humanos , Animales , Ratones , Leucocitos Mononucleares , Recurrencia Local de Neoplasia/metabolismo , Células Asesinas Naturales , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Inmunoterapia/métodos , Línea Celular Tumoral , Neoplasias Pancreáticas
15.
Korean J Anesthesiol ; 76(6): 627-639, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37435613

RESUMEN

BACKGROUND: We investigated the effects of sevoflurane exposure on the expression of matrix metalloproteinase (MMP), expression and ablation of natural killer group 2, member D (NKG2D) ligands (UL16-binding proteins 1-3 and major histocompatibility complex class I chain-related molecules A/B), and natural killer (NK) cell-mediated cytotoxicity in breast cancer cells. METHODS: Three human breast cancer cell lines (MCF-7, MDA-MB-453, and HCC-70) were incubated with 0 (control), 600 (S6), or 1200 µM (S12) sevoflurane for 4 h. The gene expression of NKG2D ligands and their protein expression on cancer cell surfaces were measured using multiplex polymerase chain reaction (PCR) and flow cytometry, respectively. Protein expression of MMP-1 and -2 and the concentration of soluble NKG2D ligands were analyzed using western blotting and enzyme-linked immunosorbent assays, respectively. RESULTS: Sevoflurane downregulated the mRNA and protein expression of the NKG2D ligand in a dose-dependent manner in MCF-7, MDA-MB-453, and HCC-70 cells but did not affect the expression of MMP-1 or -2 or the concentration of soluble NKG2D ligands in the MCF-7, MDA-MB-453, and HCC-70 cells. Sevoflurane attenuated NK cell-mediated cancer cell lysis in a dose-dependent manner in MCF-7, MDA-MB-453, and HCC-70 cells (P = 0.040, P = 0.040, and P = 0.040, respectively). CONCLUSIONS: Our results demonstrate that sevoflurane exposure attenuates NK cell-mediated cytotoxicity in breast cancer cells in a dose-dependent manner. This could be attributed to a sevoflurane-induced decrease in the transcription of NKG2D ligands rather than sevoflurane-induced changes in MMP expression and their proteolytic activity.


Asunto(s)
Neoplasias de la Mama , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Femenino , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Metaloproteinasa 1 de la Matriz/metabolismo , Sevoflurano , Ligandos , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Células Asesinas Naturales/metabolismo
16.
Biochim Biophys Acta ; 1812(7): 796-805, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21514380

RESUMEN

In this study, we investigated the role of c-Myc in overcoming multidrug resistance (MDR) in human ovarian and breast cancer cells by TRAIL. We showed that P-gp expressing MDR variants (Hey A8-MDR and MCF7-MDR cells) with high level of c-Myc were highly susceptible to TRAIL treatment when compared to their drug-sensitive parental human ovarian cancer Hey A8 and breast MCF-7 cells, respectively. Up-regulation of DR5 TRAIL receptor and down-regulation of c-FLIP and the promotion of caspase-dependent cell death, which contribute to TRAIL sensitization of MDR cells, were regulated by the over-expressed c-Myc in the MDR cells. After targeted inhibition of c-Myc with specific siRNA, these responses to TRAIL disappeared and TRAIL-induced apoptosis was also suppressed in MCF7-MDR cells. Treatment with TRAIL significantly reduced P-glycoprotein (P-gp)-mediated efflux of rhodamine123 in both Hey A8-MDR and MCF7-MDR cells. Furthermore, TRAIL significantly potentiated the cytotoxicity of vinblastine, vincristine, doxorubicin and VP-16 that are P-gp substrate anticancer drugs in both MDR cells, which resulted in the reversal effect of TRAIL on the MDR phenotype. The present study shows for the first time that elevated c-Myc expression in the MDR cells plays a critical role in overcoming MDR by TRAIL that can act as a specific sensitizer for P-gp substrate anticancer drug.


Asunto(s)
Neoplasias de la Mama/patología , Resistencia a Múltiples Medicamentos , Neoplasias Ováricas/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Secuencia de Bases , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Cartilla de ADN , Femenino , Citometría de Flujo , Humanos , Neoplasias Ováricas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Cancer Sci ; 103(1): 7-16, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21951556

RESUMEN

We have previously shown that inhibition of intracellular signaling pathways by treatment with quercetin induced the expression of natural killer cell group 2D (NKG2D) ligands on cancer cells and made the cells sensitive to natural killer (NK)-cell mediated cytotoxicity. In the present study, we investigated whether epidermal growth factor receptor (EGFR) inhibitors could induce the expression of NKG2D ligands in colon cancer cells. Treatment with EGFR inhibitors predominantly increased the levels of mRNA transcripts and surface protein of UL16-binding protein-1 (ULBP1) in various colon cancer cells, including KM12, Caco-2, HCT-15, and HT-29, which express EGFR, and increased susceptibility of these colon cancer cells to NK-92 cells. The expression of ULBP1 was not induced by inhibitors of nuclear factor-κB, phosphatidylinositol 3 kinase, and MAPK, but was induced by inhibitors of PKC, and the induction of ULBP1 expression with EGFR inhibitors was prevented by treatment with PMA in colon cancer cells. A transcription factor, activator protein-2 alpha (AP-2α), which has a suppressive effect on ULBP1 transcription, was prevented from binding to the ULBP1 promoter by treatment with EGFR inhibitors. The present study suggests that EGFR inhibitors can enhance the susceptibility to NK cell-mediated lysis of colon cancer cells by induction of ULBP1 via inhibition of the PKC pathway.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Citotoxicidad Inmunológica/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Western Blotting , Inmunoprecipitación de Cromatina , Neoplasias del Colon/tratamiento farmacológico , Receptores ErbB/metabolismo , Clorhidrato de Erlotinib , Citometría de Flujo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Gefitinib , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Células Asesinas Naturales/metabolismo , Quinazolinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas , Regulación hacia Arriba
18.
Mol Cancer ; 10: 46, 2011 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-21513580

RESUMEN

BACKGROUND: Tumor recurrence and metastasis develop as a result of tumors' acquisition of anti-apoptotic mechanisms and therefore, it is necessary to develop novel effective therapeutics against metastatic cancers. In this study, we showed the differential TRAIL responsiveness of human prostate adenocarcinoma PC3 and human colon carcinoma KM12 cells and their respective highly metastatic PC3-MM2 and KM12L4A sublines and investigated the mechanism underlying high susceptibility of human metastatic cancer cells to TRAIL. RESULTS: PC3-MM2 and KM12L4A cells with high level of c-Myc and DNA-PKcs were more susceptible to TRAIL than their poorly metastatic primary PC3 and KM12 cells, which was associated with down-regulation of c-FLIPL/S and Mcl-1 and up-regulation of the TRAIL receptor DR5 but not DR4 in both metastatic cells. Moreover, high susceptibility of these metastatic cells to TRAIL was resulted from TRAIL-induced potent activation of caspase-8, -9, and -3 in comparison with their primary cells, which led to cleavage and down-regulation of DNA-PKcs. Knockdown of c-Myc gene in TRAIL-treated PC3-MM2 cells prevented the increase of DR5 cell surface expression, caspase activation and DNA-PKcs cleavage and attenuated the apoptotic effects of TRAIL. Moreover, the suppression of DNA-PKcs level with siRNA in the cells induced the up-regulation of DR5 and active caspase-8, -9, and -3. We also found that 4,5-dimethoxy-2-nitrobenzaldehyde (DMNB), a specific inhibitor of DNA-PK, potentiated TRAIL-induced cytotoxicity and apoptosis in relatively TRAIL-insensitive PC3 and KM12 cells and therefore functioned as a TRAIL sensitizer. CONCLUSION: This study showed the positive relationship between c-Myc expression in highly metastatic human prostate and colon cancer cells and susceptibility to TRAIL-induced apoptosis and therefore indicated that TRAIL might be used as an effective therapeutic modality for advanced metastatic cancers overexpressing c-Myc and combination of TRAIL therapy with agent that inhibits the DNA-PKcs/Akt signaling pathway might be clinically useful for the treatment of relatively TRAIL-insensitive human cancers.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzaldehídos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Apoptosis/genética , Western Blotting , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/genética , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 8/genética , Caspasa 8/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Humanos , Masculino , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Interferencia de ARN , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Células Tumorales Cultivadas
19.
Chemistry ; 17(45): 12802-8, 2011 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-21954062

RESUMEN

Magnetic particles have become very promising materials for drug delivery. However, preparation of magnetite particles with high surface area, biocompatibility, strong magnetic response, and suitable particle size still remains a major challenge. In this report, magnetite nanocrystal clusters with high surface areas were fabricated through a solvothermal process by introducing ammonium acetate as a porogen and trisodium citrate as a surface modification agent. The porosity, which was controlled by the reactant concentration, has been investigated in detail. The surface area of the nanocrystal clusters was as high as 141 m(2) g(-1). Ibuprofen, as a model drug, was entrapped into the magnetite carriers. The interfacial interaction between the carboxylic groups on the drug molecules and the carboxylate groups on the carriers enhanced the loading efficiency. Low cytotoxicity in MCF-7 cell and in vitro constant drug release behavior combined with the high drug loading efficiency and high saturation magnetization values demonstrated the potential of the as-synthesized magnetite materials in targeted drug release systems.


Asunto(s)
Sistemas de Liberación de Medicamentos , Óxido Ferrosoférrico/química , Ibuprofeno/administración & dosificación , Supervivencia Celular/efectos de los fármacos , Femenino , Óxido Ferrosoférrico/síntesis química , Humanos , Porosidad , Solubilidad , Agua
20.
Immunol Invest ; 40(4): 367-82, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21314289

RESUMEN

Natural killer (NK) cells are important innate effector cells which can irradicate tumor cells through specific interactions between activating receptors on NK cells and their cognate ligands on cancer cells. Recently, it has been known that induction of activating NKG2D ligands including MHC class I chain-related (MIC) and UL16-binding protein (ULBP) families on tumor cells by various stresses makes them more susceptible to NK cell-mediated cytotoxicity. Therefore, it was investigated whether sublethal dose of hematoporphyrin-based photodynamic therapy (PDT) could up-regulate NKG2D ligands on tumor cells and increase the susceptibility of cancer cells against NK cells. Treatment with sublethal dose of hematoporphyrin-based PDT increased mRNA transcription and surface expression of ULBP1 and ULBP2 genes in SNU-1 human gastric tumor cell line and MICA/B, ULBP1, ULBP2 and ULBP3 genes in SW-900 human lung cancer cell line. These results were followed by increased susceptibility of cancer cells to NK cell-mediated cytotoxicity after sublethal PDT, which was abolished by addition of a blocking NKG2D mAb. Therefore, it could be suggested that the effect of hematoporphyrin-based PDT might be mediated in part by the increased susceptibility to NK cells via induction of NKG2D ligands on tumor cells, which survived after treatment with PDT.


Asunto(s)
Células Asesinas Naturales/inmunología , Ligandos , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Fotoquimioterapia , Regulación hacia Arriba , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/inmunología , Carcinoma/tratamiento farmacológico , Carcinoma/inmunología , Línea Celular Tumoral/inmunología , Línea Celular Tumoral/metabolismo , Citotoxicidad Inmunológica , Regulación Neoplásica de la Expresión Génica , Hematoporfirinas/metabolismo , Humanos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/inmunología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA