Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
2.
Oncoimmunology ; 12(1): 2190671, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36950218

RESUMEN

Triple-negative breast cancer (TNBC) is the most challenging subtype of breast cancer because of its aggressive behavior and the limited therapeutic strategies available. In the last decade, immunotherapy has become a promising treatment to prolong survival in advanced solid cancers including TNBC. However, the efficacy of immunotherapy in solid cancers remains limited because solid tumors contain few tumor-infiltrating lymphocytes. Here, we show that targeting an ETS transcription factor ELK3 (ELK3) recruits immune cells including natural killer (NK) cells into tumors via the chemotactic activity of chemokine. ELK3 depletion increases CXCL16 expression level and promotes NK cell cytotoxicity through CXCL16-mediated NK cell recruitment in TNBC. In silico analysis showed that ELK3 is negatively correlated with CXCL16 expression in breast cancer patient samples. Low expression of ELK3 and high expression of CXCL16 were associated with a better prognosis. Low expression of ELK3 and high expression of CXCL16 were associated with increased expression of NK cell-related genes. Our findings demonstrate that the ELK3-CXCL16 axis modulates NK cell recruitment to increase NK cell cytotoxicity, suggesting that targeting the ELK3 gene could be an adjuvant strategy for increasing the efficacy of immunotherapy in TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Línea Celular Tumoral , Células Asesinas Naturales/metabolismo , Inmunoterapia , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Quimiocina CXCL16/genética , Quimiocina CXCL16/metabolismo
3.
J Immunother Cancer ; 10(8)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36028281

RESUMEN

BACKGROUND: Due to their powerful immune surveillance activity and ability to kill and clear cancer cells, natural killer (NK) cells are an emerging anticancer immunotherapeutic agent. Therefore, there is much interest in developing efficient technologies that further enhance the therapeutic antitumor efficacy of NK cells. METHODS: To produce chemically primed NK cells, we screened polymers with various electric charges and examined their ability to enhance the cytotoxicity of NK cells. The effect of primary amine and electric charges of 25 kDa branched polyethylenimine (25KbPEI) was investigated by fluorination of the chemical. The role of 25KbPEI in determining the major priming mechanism was investigated in terms of calcium influx into NK cells. In vivo therapeutic efficacy of chemically primed NK cells was evaluated against solid tumor mouse model of triple negative breast and ovarian cancers. RESULTS: Chem_NK that was produced by the priming activity of 25KbPEI showed potent antitumor activity to various cancer cells. Chem_NK showed an activated phenotype, which manifests as increased expression of activating/adhesion/chemokine receptors and perforin accumulation, leading to enhanced migration ability and antitumor activity. Chem_NK display potent therapeutic efficacy against in vivo mouse model of triple negative breast and ovarian cancers. Fluorination of the primary amine group reduces the activity of 25KbPEI to prime NK cells, indicating that the cationic charge on the chemical plays a critical role in NK cell activation. A major priming mechanism was 25KbPEI-mediated calcium influx into NK cells, which occurred mainly via the Ca2+-permeable non-selective cation channel transient receptor potential melastatin 2. CONCLUSIONS: NK cells can be chemically primed with 25KbPEI to express potent antitumor activity as well as enhanced migration ability. Because PEI is a biocompatible and Food and Drug Administration-approved chemical for biomedical use, these results suggest a cost-effective and simple method of producing therapeutic NK cells.


Asunto(s)
Antineoplásicos , Neoplasias Ováricas , Neoplasias de la Mama Triple Negativas , Aminas , Animales , Calcio , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia , Células Asesinas Naturales , Ratones , Polietileneimina , Estados Unidos
4.
J Immunother Cancer ; 10(7)2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35858708

RESUMEN

BACKGROUND: Triple negative breast cancer (TNBC) is the most lethal subtype of breast cancer due to its aggressive behavior and frequent development of resistance to chemotherapy. Although natural killer (NK) cell-based immunotherapy is a promising strategy for overcoming barriers to cancer treatment, the therapeutic efficacy of NK cells against TNBC is below expectations. E26 transformation-specific transcription factor ELK3 (ELK3) is highly expressed in TNBCs and functions as a master regulator of the epithelial-mesenchymal transition. METHODS: Two representative human TNBC cell lines, MDA-MB231 and Hs578T, were exposed to ELK3-targeting shRNA or an ELK3-expressing plasmid to modulate ELK3 expression. The downstream target genes of ELK3 were identified using a combined approach comprising gene expression profiling and molecular analysis. The role of ELK3 in determining the immunosensitivity of TNBC to NK cells was investigated in terms of mitochondrial fission-fusion transition and reactive oxygen species concentration both in vitro and in vivo. RESULTS: ELK3-dependent mitochondrial fission-fusion status was linked to the mitochondrial superoxide concentration in TNBCs and was a main determinant of NK cell-mediated immune responses. We identified mitochondrial dynamics proteins of 51 (Mid51), a major mediator of mitochondrial fission, as a direct downstream target of ELK3 in TNBCs. Also, we demonstrated that expression of ELK3 correlated inversely with that of Mid51, and that the ELK3-Mid51 axis is associated directly with the status of mitochondrial dynamics. METABRIC analysis revealed that the ELK3-Mid51 axis has a direct effect on the immune score and survival of patients with TNBC. CONCLUSIONS: Taken together, the data suggest that NK cell responses to TNBC are linked directly to ELK3 expression levels, shedding new light on strategies to improve the efficacy of NK cell-based immunotherapy of TNBC.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Humanos , Células Asesinas Naturales , Dinámicas Mitocondriales , Proteínas Proto-Oncogénicas c-ets , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/terapia
5.
Int J Mol Sci ; 23(7)2022 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-35409069

RESUMEN

Current therapeutic strategies for gastric cancer, including surgery and chemotherapy improve patient survival; however, the survival rate of patients with metastatic gastric cancer is very low. The molecular mechanisms underlying the dissemination of gastric cancer cells to distant organs are currently unknown. Here, we demonstrate that the E26 transformation-specific (ETS) transcription factor ELK3 (ELK3) gene is required for the migration and invasion of gastric cancer cells. The ELK3 gene modulates the expression of extracellular matrix (ECM) remodeling-related genes, such as bone morphogenetic protein (BMP1), lysyl oxidase like 2 (LOXL2), Snail family transcriptional repressor 1 (SNAI1), serpin family F member 1 (SERPINF1), decorin (DCN), and nidogen 1 (NID1) to facilitate cancer cell dissemination. Our in silico analyses indicated that ELK3 expression was positively associated with these ECM remodeling-related genes in gastric cancer cells and patient samples. The high expressions of ELK3 and other ECM remodeling-related genes were also closely associated with a worse prognosis of patients with gastric cancer. Collectively, these findings suggest that ELK3 acts as an important regulator of gastric cancer cell dissemination by regulating ECM remodeling.


Asunto(s)
Neoplasias Gástricas , Línea Celular Tumoral , Movimiento Celular/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Humanos , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Neoplasias Gástricas/genética
6.
ACS Appl Mater Interfaces ; 12(51): 56731-56740, 2020 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-33290037

RESUMEN

Natural killer (NK) cells have been recognized as a next-generation therapy for cancer as they are less likely to trigger adverse events (e.g., cytokine storm or graft-versus-host disease) than T cell-based therapeutics. Although NK cell activation strategies through genetic engineering and cytokine treatment have been actively studied for successful cancer treatment, the approaches are inefficient, expensive, and involve complex processing. Here, we developed a facile and efficient method of activating NK cells using cationic nanoparticles (cNPs). The cytotoxic activity of cNP-treated primary NK and NK-92MI cells against triple-negative breast cancer cells was over 2-fold higher than that of control NK cells in vitro. Molecular biological analyses confirmed that cNPs altered the expression of CCR4 and CXCR4 of NK cells that function as chemokine receptors. In vitro live cell imaging showed that the NK cells treated with cNPs were better than control NK cells at interacting with cancer cells. Consistent with these in vitro results, cNP-treated NK cells effectively inhibited tumor growth in an in vivo tumor animal model of triple-negative breast cancer. Additionally, NK cells treated with cNPs were tracked effectively in vivo by magnetic resonance imaging. Thus, cNP-mediated activation of NK cells has great potential as an NK cell-based cancer immunotherapy. Most of all, activating NK cells using cNPs has a great advantage over conventional methods in that immune cells can be activated by a one-step facile process with exogenously charged nanomaterials, without the need for genetic engineering or cytokine treatment.


Asunto(s)
Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Nanopartículas/química , Neoplasias/terapia , Polietileneimina/farmacología , Animales , Línea Celular Tumoral , Femenino , Humanos , Factores Inmunológicos/química , Factores Inmunológicos/farmacología , Indoles/química , Células Asesinas Naturales/efectos de los fármacos , Ratones Desnudos , Polietileneimina/química , Polímeros/química , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Transl Med ; 18(1): 307, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32762722

RESUMEN

BACKGROUND: Treatment of human lung squamous cell carcinoma (LUSC) using current targeted therapies is limited because of their diverse somatic mutations without any specific dominant driver mutations. These mutational diversities preventing the use of common targeted therapies or the combination of available therapeutic modalities would require a preclinical animal model of this tumor to acquire improved clinical responses. Patient-derived xenograft (PDX) models have been recognized as a potentially useful preclinical model for personalized precision medicine. However, whether the use of LUSC PDX models would be appropriate enough for clinical application is still controversial. METHODS: In the process of developing PDX models from Korean patients with LUSC, the authors investigated the factors influencing the successful initial engraftment of tumors in NOD scid gamma mice and the retainability of the pathological and genomic characteristics of the parental patient tumors in PDX tumors. CONCLUSIONS: The authors have developed 62 LUSC PDX models that retained the pathological and genomic features of parental patient tumors, which could be used in preclinical and co-clinical studies. Trial registration Tumor samples were obtained from 139 patients with LUSC between November 2014 and January 2019. All the patients provided signed informed consents. This study was approved by the institutional review board (IRB) of Samsung Medical Center (2018-03-110).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Animales , Carcinoma de Células Escamosas/genética , Humanos , Pulmón , Neoplasias Pulmonares/genética , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Dev Cell ; 54(3): 302-316.e7, 2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32574556

RESUMEN

Mechanical cues from the extracellular matrix (ECM) regulate various cellular processes via distinct mechanotransduction pathways. In breast cancer, increased ECM stiffness promotes epithelial-to-mesenchymal transition (EMT), cell invasion, and metastasis. Here, we identify a mechanosensitive EPHA2/LYN protein complex regulating EMT and metastasis in response to increasing ECM stiffness during tumor progression. High ECM stiffness leads to ligand-independent phosphorylation of ephrin receptor EPHA2, which recruits and activates the LYN kinase. LYN phosphorylates the EMT transcription factor TWIST1 to release TWIST1 from its cytoplasmic anchor G3BP2 to enter the nucleus, thus triggering EMT and invasion. Genetic and pharmacological inhibition of this pathway prevents breast tumor invasion and metastasis in vivo. In human breast cancer samples, activation of this pathway correlates with collagen fiber alignment, a marker of increasing ECM stiffness. Our findings reveal an EPHA2/LYN/TWIST1 mechanotransduction pathway that responds to mechanical signals from the tumor microenvironment to drive EMT, invasion, and metastasis.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Matriz Extracelular/metabolismo , Proteínas Nucleares/metabolismo , Receptor EphA2/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/genética , Humanos , Neoplasias Mamarias Animales/metabolismo , Mecanotransducción Celular/genética , Ratones , Receptor EphA2/genética , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología
9.
Cancers (Basel) ; 12(5)2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32414208

RESUMEN

Triple-negative breast cancer (TNBC) refers to breast cancer that does not have receptors for estrogen, progesterone, and HER2 protein. TNBC accounts for 10-20% of all cases of breast cancers and is characterized by its metastatic aggressiveness, poor prognosis, and limited treatment options. Here, we show that the metastatic nature of TNBC is critically regulated by a functional link between miR-200a and the transcription factor ELK3. We found that the expression levels of miR-200a and the ELK3 mRNA were negatively correlated in the luminal and TNBC subtypes of breast cancer cells. In vitro experiments revealed that miR-200a directly targets the 3' untranslated region (UTR) of the ELK3 mRNA to destabilize the transcripts. Furthermore, ectopic expression of miR-200a impaired the migration and invasion of TNBC cells by reducing the expression level of the ELK3 mRNA. In in vivo studies, transfection of MDA-MB 231 cells (a claudin-low TNBC cell type) with exogenous miR-200a reduced their extravasation into the lung during 48 h after tail vein injection, and co-transfection of the cells with an expression plasmid harboring ELK3 that lacked an intact 3'UTR recovered their extravasation ability. Overall, our findings provide evidences that miR-200a and ELK3 is functionally linked to regulate invasive characteristics of breast cancers.

10.
Nat Cell Biol ; 21(3): 359-371, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30804505

RESUMEN

Loss of apical-basal polarity and activation of epithelial-mesenchymal transition (EMT) both contribute to carcinoma progression and metastasis. Here, we report that apical-basal polarity inhibits EMT to suppress metastatic dissemination. Using mouse and human epithelial three-dimensional organoid cultures, we show that the PAR-atypical protein kinase C (aPKC) polarity complex inhibits EMT and invasion by promoting degradation of the SNAIL family protein SNAI1. Under intact apical-basal polarity, aPKC kinases phosphorylate S249 of SNAI1, which leads to protein degradation. Loss of apical-basal polarity prevents aPKC-mediated SNAI1 phosphorylation and stabilizes the SNAI1 protein to promote EMT and invasion. In human breast tumour xenografts, inhibition of the PAR-complex-mediated SNAI1 degradation mechanism promotes tumour invasion and metastasis. Analyses of human breast tissue samples reveal negative correlations between PAR3 and SNAI1 protein levels. Our results demonstrate that apical-basal polarity functions as a critical checkpoint of EMT to precisely control epithelial-mesenchymal plasticity during tumour metastasis.


Asunto(s)
Polaridad Celular , Transición Epitelial-Mesenquimal , Complejos Multiproteicos/metabolismo , Neoplasias/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Células CACO-2 , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Proteína Quinasa C/metabolismo , Proteolisis , Interferencia de ARN , Factores de Transcripción de la Familia Snail/genética , Trasplante Heterólogo
11.
Nat Cell Biol ; 20(12): 1421-1433, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30374053

RESUMEN

Vesicular acidification and trafficking are associated with various cellular processes. However, their pathologic relevance to cancer remains elusive. We identified transmembrane protein 9 (TMEM9) as a vesicular acidification regulator. TMEM9 is highly upregulated in colorectal cancer. Proteomic and biochemical analyses show that TMEM9 binds to and facilitates assembly of vacuolar-ATPase (v-ATPase), a vacuolar proton pump, resulting in enhanced vesicular acidification and trafficking. TMEM9-v-ATPase hyperactivates Wnt/ß-catenin signalling via lysosomal degradation of adenomatous polyposis coli (APC). Moreover, TMEM9 transactivated by ß-catenin functions as a positive feedback regulator of Wnt signalling in colorectal cancer. Genetic ablation of TMEM9 inhibits colorectal cancer cell proliferation in vitro, ex vivo and in vivo mouse models. Moreover, administration of v-ATPase inhibitors suppresses intestinal tumorigenesis of APC mouse models and human patient-derived xenografts. Our results reveal the unexpected roles of TMEM9-controlled vesicular acidification in hyperactivating Wnt/ß-catenin signalling through APC degradation, and propose the blockade of TMEM9-v-ATPase as a viable option for colorectal cancer treatment.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas de la Membrana/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Vía de Señalización Wnt , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células HCT116 , Células HT29 , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Intestinos/química , Intestinos/patología , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Unión Proteica , Trasplante Heterólogo
12.
Cell Stem Cell ; 16(1): 1-2, 2015 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-25575073

RESUMEN

Twist1 has a well-established role in inducing Epithelial-Mesenchymal Transition (EMT) to promote tumor invasion and metastasis and can also inhibit apoptosis and confer stemness. In this issue of Cell Stem Cell, Beck et al. (2015) now show that low levels of Twist1 are essential for tumor initiation, maintenance, and stemness independent of its EMT-inducing activity.


Asunto(s)
Carcinogénesis/metabolismo , Progresión de la Enfermedad , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Proteína 1 Relacionada con Twist/metabolismo , Animales , Humanos
13.
Clin Cancer Res ; 21(5): 962-968, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25107915

RESUMEN

During tumor development, tumor cells constantly communicate with the surrounding microenvironment through both biochemical and biophysical cues. In particular, the tumor microenvironment can instruct carcinoma cells to undergo a morphogenesis program termed epithelial-to-mesenchymal transition (EMT) to facilitate local invasion and metastatic dissemination. Growing evidence uncovered a plethora of microenvironmental factors in promoting EMT, including proinflammatory cytokines secreted by locally activated stromal cells, hypoxia conditions, extracellular matrix components, and mechanical properties. Here, we review various biochemical and biophysical factors in the tumor microenvironment that directly impinge upon the EMT program. Specifically, cytokines such as TGFß, TNFα, and IL6 and hypoxia are capable of inducing EMT in various tumors. Several extracellular matrix (ECM) proteins, including collagen-I, fibronectin, and hyaluronan, and ECM remodeling via extracellular lysyl oxidase are also implicated in regulating EMT. In preclinical studies and ongoing clinical trials, targeting these tumor microenvironmental signals has shown promises in halting tumor progression in various human cancers.


Asunto(s)
Transición Epitelial-Mesenquimal , Neoplasias/metabolismo , Neoplasias/patología , Transducción de Señal , Microambiente Tumoral , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos Clínicos como Asunto , Citocinas/genética , Citocinas/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Hipoxia/genética , Hipoxia/metabolismo , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Transducción de Señal/efectos de los fármacos , Investigación Biomédica Traslacional , Resultado del Tratamiento , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética
14.
Mol Cell ; 52(2): 193-205, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24055345

RESUMEN

Fine control of Wnt signaling is essential for various cellular and developmental decision-making processes. However, deregulation of Wnt signaling leads to pathological consequences, one of which is cancer. Here, we identify a function of PAF, a component of translesion DNA synthesis, in modulating Wnt signaling. PAF is specifically overexpressed in colon cancer cells and intestinal stem cells and is required for colon cancer cell proliferation. In Xenopus laevis, ventrovegetal expression of PAF hyperactivates Wnt signaling, developing a secondary axis with ß-catenin target gene upregulation. Upon Wnt signaling activation, PAF dissociates from PCNA and binds directly to ß-catenin. Then, PAF recruits EZH2 to the ß-catenin transcriptional complex and specifically enhances Wnt target gene transactivation, independently of EZH2's methyltransferase activity. In mice, conditional expression of PAF induces intestinal neoplasia via Wnt signaling hyperactivation. Our studies reveal an unexpected role of PAF in regulating Wnt signaling and propose a regulatory mechanism of Wnt signaling during tumorigenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Proteínas Portadoras/genética , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Proteínas de Unión al ADN , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/embriología , Embrión no Mamífero/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Regulación del Desarrollo de la Expresión Génica , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/genética , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Xenopus laevis , beta Catenina/genética
15.
Cancer Res Treat ; 45(2): 134-44, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23864847

RESUMEN

PURPOSE: The RAS association domain family protein 1 (RASSF1) has been implicated in a tumor-suppressive function through the induction of acetylated α-tubulin and modulation of cell migration. However, the mechanisms of how RASSF1A is associated with acetylation of α-tubulin for controlling cell migration have not yet been elucidated. In this study, we found that RASSF1A regulated cell migration through the regulation of histon deacetylase 6 (HDAC6), which functions as a tubulin deacetylase. MATERIALS AND METHODS: The cell migration was assessed using wound-healing and transwell assays. The role of RASSF1A on cell migration was examined by immunofluorescence staining, HDAC activity assay and western blot analysis. RESULTS: Cell migration was inhibited and cell morphology was changed in RASSF1A-transfected H1299 cells, compared with controls, whereas HDAC6 protein expression was not changed by RASSF1A transfection in these cells. However, RASSF1A inhibited deacetylating activity of HDAC6 protein and induced acetylated α-tubulin expression. Furthermore, acetylated α-tubulin and HDAC6 protein were co-localized in the cytoplasm in RASSF1A-transfected H1299 cells. Conversely, when the endogenous RASSF1A expression in HeLa cells was blocked with RASSF1A siRNA treatment, acetylated α-tubulin was co-localized with HDAC6 protein throughout the whole cells, including the nucleus, compared with scramble siRNA-treated HeLa cells. The restoration of RASSF1A by 5-Aza-dC treatment also induced acetylated α-tubulin through inhibition of HDAC6 activity that finally resulted in suppressing cell migration in H1299 cells. To further confirm the role of HDAC6 in RASSF1A-mediated cell migration, the HDAC6 expression in H1299 cells was suppressed by using HDAC6 siRNA, and cell motility was found to be decreased through enhanced acetylated α-tubulin. CONCLUSION: The results of this study suggest that the inactivation of HDAC6 by RASSF1A regulates cell migration through increased acetylated α-tubulin protein.

16.
J Biol Chem ; 288(22): 15474-80, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23612978

RESUMEN

Viral pathogens utilize host cell machinery for their benefits. Herein, we identify that HIV-1 Vpr (viral protein R) negatively modulates telomerase activity. Telomerase enables stem and cancer cells to evade cell senescence by adding telomeric sequences to the ends of chromosomes. We found that Vpr inhibited telomerase activity by down-regulating TERT protein, a catalytic subunit of telomerase. As a molecular adaptor, Vpr enhanced the interaction between TERT and the VPRBP substrate receptor of the DYRK2-associated EDD-DDB1-VPRBP E3 ligase complex, resulting in increased ubiquitination of TERT. In contrast, the Vpr mutant identified in HIV-1-infected long-term nonprogressors failed to promote TERT destabilization. Our results suggest that Vpr inhibits telomerase activity by hijacking the host E3 ligase complex, and we propose the novel molecular mechanism of telomerase deregulation in possibly HIV-1 pathogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Proteolisis , Telomerasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Proteínas Portadoras/genética , Proteínas de Unión al ADN/genética , Infecciones por VIH/genética , Infecciones por VIH/patología , VIH-1/genética , Células HeLa , Humanos , Células Jurkat , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Estabilidad Proteica , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Telomerasa/genética , Ubiquitina-Proteína Ligasas/genética , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética , Quinasas DyrK
17.
J Biol Chem ; 288(10): 7252-62, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23362280

RESUMEN

Telomerase maintains the telomere, a specialized chromosomal end structure that is essential for genomic stability and cell immortalization. Telomerase is not active in most somatic cells, but its reactivation is one of the hallmarks of cancer. In this study, we found that dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 2 (Dyrk2) negatively regulates telomerase activity. Dyrk2 phosphorylates TERT protein, a catalytic subunit of telomerase. Phosphorylated TERT is then associated with the EDD-DDB1-VprBP E3 ligase complex for subsequent ubiquitin-mediated TERT protein degradation. During the cell cycle, Dyrk2 interacts with TERT at the G2/M phase and induces degradation. In contrast, depletion of endogenous Dyrk2 disrupts the cell cycle-dependent regulation of TERT and elicits the constitutive activation of telomerase. Similarly, a Dyrk2 nonsense mutation identified in breast cancer compromises ubiquitination-mediated TERT protein degradation. Our findings suggest the novel molecular mechanism of kinase-associated telomerase regulation.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Telomerasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Portadoras/genética , División Celular , Células Cultivadas , Proteínas de Unión al ADN/genética , Fase G2 , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Células MCF-7 , Ratones , Modelos Biológicos , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Proteolisis , Interferencia de ARN , Telomerasa/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación , Quinasas DyrK
18.
Am J Clin Oncol ; 36(1): 57-63, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22237146

RESUMEN

BACKGROUND: Epidermal growth factor receptor (EGFR) mutations as prognostic or predictive marker in patients with non-small cell lung cancer (NSCLC) have been used widely. However, it may be difficult to get tumor tissue for analyzing the status of EGFR mutation status in large proportion of patients with advanced disease. PATIENTS AND METHODS: We obtained pairs of tumor and serum samples from 57 patients with advanced NSCLC, between March 2006 and January 2009. EGFR mutation status from tumor samples was analyzed by genomic polymerase chain reaction and direct sequence and EGFR mutation status from serum samples was determined by the peptide nucleic acid locked nucleic acid polymerase chain reaction clamp. RESULTS: EGFR mutations were detected in the serum samples of 11 patients and in the tumor samples of 12 patients. EGFR mutation status in the serum and tumor samples was consistent in 50 of the 57 pairs (87.7%). There was a high correlation between the mutations detected in serum sample and the mutations detected in the matched tumor sample (correlation index 0.62; P<0.001). Twenty-two of 57 patients (38.5%) received EGFR-tyrosine kinase inhibitors as any line therapy. The response for EGFR-tyrosine kinase inhibitors was significantly associated with EGFR mutations in both tumor samples and serum samples (P<0.05). There was no significant differences in overall survival according to the status of EGFR mutations in both serum and tumor samples (P>0.05). CONCLUSIONS: Serum sample might be alternatively used in the difficult time of getting tumor tissue for analyzing the status of EGFR mutation status in patients with advanced NSCLC.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/genética , Análisis Mutacional de ADN/métodos , Genes erbB-1/genética , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/genética , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Femenino , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa , Sensibilidad y Especificidad
19.
Cancer Res Treat ; 44(4): 251-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23341789

RESUMEN

PURPOSE: c-Met is an attractive potential target for novel therapeutic inhibition of human cancer, and c-Met tyrosine kinase inhibitors (TKIs) are effective growth inhibitors of various malignancies. However, their mechanisms in anticancer effects are not clear. In the present study, we investigated the possibility that blocking c-Met signaling induces p53-mediated growth inhibition in lung cancer. MATERIALS AND METHODS: The growth inhibitory effects of c-Met TKI (SU11274) on lung cancer cells and a xenograft model were assessed using the MTT assay, flow cytometry, and terminal deoxyribonucleotide transferase-mediated nick-end labeling staining. The role of p53 protein in the sensitivity of c-Met TKI (SU11274) was examined by Western blot analysis and immunohistochemistry. RESULTS: SU11274 significantly induced apoptosis in A549 cells with wild-type p53, compared with that in Calu-1 cells with null-type p53. SU11274 increased p53 protein by enhancing the stability of p53 protein. Increased p53 protein by SU11274 induced up-regulation of Bax and PUMA expression and down-regulation of Bcl-2 expression, subsequently activating caspase 3. In p53 knock-out and knock-in systems, we confirmed that SU11274 caused apoptosis through the p53-mediated apoptotic pathway. Likewise, in the A549 xenograft model, SU11274 effectively shrank tumor volume and induced apoptosis via increased p53 protein expression. Blocking c-Met signaling increased the level of p53 protein. CONCLUSION: Our finding suggested that p53 plays an important role in SU11274-induced apoptosis, and p53 status seems to be related to the sensitivity to SU11274 in lung cancer.

20.
Cancer Res Treat ; 43(2): 108-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21811427

RESUMEN

PURPOSE: The genetic alteration of the janus kinases (JAKs), non-receptor tyrosine kinase, is related to the development of human cancers. However, little is known about how the sequence variation of JAK3 contributes to the development of lung cancer. This study investigated whether polymorphisms at the promoter region of the JAK3 gene are associated with the risk of lung cancer in the Korean population. MATERIALS AND METHODS: A total of 819 subjects, including 409 lung cancer patients and 410 healthy controls were recruited. The SNaPshot assay and polymerase chain reaction-restriction fragment length polymorphism analysis were used, and logistic regression analyses were performed to characterize the association between polymorphisms of JAK3 and lung cancer risk. RESULTS: Three polymorphisms (-672 G>A, +64 A>G and +227 G>A) of JAK3 were analyzed for large-scale genotyping (n=819). Statistical analyses revealed that polymorphisms and haplotypes in the JAK3 gene were not significantly associated with lung cancer. CONCLUSION: JAK3 gene was not significantly associated with the risk of lung cancer in the Korean population.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...