Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Signal ; 96: 110351, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35523402

RESUMEN

BACKGROUND: Cervical cancer belongs to the most common gynecological malignant cancers. EZH2 has been found to be dysregulated in different kinds of tumors and acts as an oncogene to promote cancer development. However, its upstream regulators and downstream targets in cervical cancer remain unclear. PD-L1 is a surface marker of cancer cells, facilitating the immunosuppressive microenvironment for escape from immunity attack. The molecular mechanism of increased PD-L1 expression in cervical cancer is needed to be explored. METHODS: The expression levels of USP7, EZH2 and TIMP2 in cervical cancer patients' samples and cell lines were detected by qRT-PCR and histopathology staining. The functions of USP7, EZH2 and TIMP2 were evaluated by MTT, cell migration and invasion assays after knocking down or overexpression of indicated genes. The tumor microenvironment was determined by testing of PD-L1 expression and cytotoxicity when co-cultured with NK-92 cells. Xenograft model was used to test the function of USP7 in vivo. RESULTS: Our data demonstrated that USP7 and EZH2 were upregulated in cervical cancer, while TIMP2 was downregulated. Inhibition of USP7 and EZH2, or overexpression of TIMP2 suppressed proliferation, migration, invasion and immune escape ability of cervical cancer cells. USP7 could increase EZH2 level, which in turn inhibited TIMP2 expression via methylation in its promoter. TIMP2 was able to mediate PD-L1 expression via NF-κB signaling pathway. Knocking down of USP7 could inhibit tumor development in vivo of cervical cancer. CONCLUSIONS: The study discovered the function and mechanism of USP7 and highlighted its oncogenic role in cervical cancer development. Our results indicated that targeting USP7 could be a therapeutic strategy the treatment of cervical cancer.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica , Inhibidor Tisular de Metaloproteinasa-2 , Peptidasa Específica de Ubiquitina 7 , Neoplasias del Cuello Uterino , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Femenino , Humanos , FN-kappa B/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/genética , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Microambiente Tumoral , Peptidasa Específica de Ubiquitina 7/metabolismo , Neoplasias del Cuello Uterino/metabolismo
2.
BBA Adv ; 2: 100052, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-37082587

RESUMEN

Filopodia are cellular protrusions that respond to a variety of stimuli. Filopodia are formed when actin is bound to the protein Fascin, which may play a crucial role in cellular interactions and motility during cancer metastasis. Significantly, the noncanonical features of Fascin-1 are gradually being clarified, including the related molecular network contributing to metabolic reprogramming, chemotherapy resistance, stemness ac-tivity, and tumor microenvironment events. However, the relationship between biological characteristics and pathological features to identify effective therapeutic strategies needs to be studied further. The pur-pose of this review article is to provide a broad overview of the latest molecular networks and multiomics research regarding fascins and cancer. It also highlights their direct and indirect effects on available cancer treatments. With this multidisciplinary approach, researchers and clinicians can gain the most relevant in-formation on the function of fascins in cancer progression, which may facilitate clinical applications in the future.

3.
Biomedicines ; 9(9)2021 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-34572346

RESUMEN

Changes in cell growth and metabolism are affected by the surrounding environmental factors to adapt to the cell's most appropriate growth model. However, abnormal cell metabolism is correlated with the occurrence of many diseases and is accompanied by changes in galectin (Gal) performance. Gals were found to be some of the master regulators of cell-cell interactions that reconstruct the microenvironment, and disordered expression of Gals is associated with multiple human metabolic-related diseases including cancer development. Cancer cells can interact with surrounding cells through Gals to create more suitable conditions that promote cancer cell aggressiveness. In this review, we organize the current understanding of Gals in a systematic way to dissect Gals' effect on human disease, including how Gals' dysregulated expression affects the tumor microenvironment's metabolism and elucidating the mechanisms involved in Gal-mediated diseases. This information may shed light on a more precise understanding of how Gals regulate cell biology and facilitate the development of more effective therapeutic strategies for cancer treatment by targeting the Gal family.

4.
Immunol Lett ; 223: 10-16, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32333963

RESUMEN

The mechanism of recognition of the foot-and-mouth disease virus (FMDV) by host innate immune cells is not well-understood. In this study, we first found that binary ethylenimine inactivated-FMDV (BEI-FMDV) with structurally intact capsid activated TLR2, but not other TLRs, and this specific activation was blocked by anti-TLR2 Abs or knockout of TLR2. BEI-FMDV activated NF-κB to induce cytokines, notably interferon-ß and IL-6, in a TLR2 and MyD88-dependent manner. Coexpression of TLR6 and CD14 showed additive effects on BEI-FMDV/TLR2-mediated activation of NF-κB. Further studies demonstrated that recombinant capsid proteins rVP1 and rVP3 of FMDV but not rVP0 bound directly with CD14 and TLR2. The rVP1- and rVP3-mediated activation of TLR2 and NF-κB were enhanced by the coexpression of TLR1 or TLR6. Immunoprecipitation of either rVP1 or rVP3 with mouse macrophage cell extracts revealed that rVP1 or rVP3 associated with TLR2, CD14 and TLR6 suggesting that rVP1 and rVP3 interact with CD14, TLR2/TLR1, and TLR2/TLR6 heterodimer. Additional study confirmed that rVP1 and rVP3 interacted with the swine TLR2 signaling pathway to induce IL-6 in swine macrophages. Our results identify VP1 and VP3 of FMDV as novel TLR agonists whose recognition by CD14, TLR2/TLR1, and TLR2/TLR6 of host innate immune cells is critical for the induction of cytokine production.


Asunto(s)
Virus de la Fiebre Aftosa/fisiología , Fiebre Aftosa/inmunología , Receptores de Lipopolisacáridos/metabolismo , Macrófagos/inmunología , Receptor Toll-Like 2/metabolismo , Animales , Proteínas de la Cápside/metabolismo , Células Cultivadas , Células HEK293 , Humanos , Inmunidad Innata , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Transducción de Señal , Porcinos , Receptor Toll-Like 2/genética
5.
J Biomed Sci ; 27(1): 42, 2020 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-32169072

RESUMEN

BACKGROUND: The underlying mechanism involved in ovarian cancer stemness and chemoresistance remains largely unknown. Here, we explored whether the regulation of c-Kit and plasma membrane prohibitin (PHB) affects ovarian cancer stemness and chemotherapy resistance. METHODS: Mass spectrum analysis and an in vitro kinase assay were conducted to examine the phosphorylation of PHB at tyrosine 259 by c-Kit. The in vitro effects of c-Kit on membrane raft-PHB in ovarian cancer were determined using tissue microarray (TMA)-based immunofluorescence, western blotting, immunoprecipitation, colony and spheroid formation, cell migration and cell viability assays. In vivo tumor initiation and carboplatin treatment were conducted in nude mice. RESULTS: We found that c-Kit and PHB colocalized in the raft domain and were positively correlated in human ovarian serous carcinoma. c-Kit interacted with PHB and facilitated the phosphorylation of PHB at tyrosine 259 (phospho-PHBY259) in the membrane raft to enhance ovarian cancer cell motility. The generation of SKOV3GL-G4, a metastatic phenotype of SKOV3 green fluorescent protein and luciferase (GL) ovarian cancer cells, in xenograft murine ascites showed a correlation between metastatic potential and stem cell characteristics, as indicated by the expression of c-Kit, Notch3, Oct4, Nanog and SOX2. Further study revealed that after activation by c-Kit, raft-phospho-PHBY259 interacted with Notch3 to stabilize Notch3 and increase the downstream target PBX1. Downregulation of raft-phospho-PHBY259 increased the protein degradation of Notch3 through a lysosomal pathway and inhibited the ß-catenin-ABCG2 signaling pathway. Moreover, raft-phospho-PHBY259 played an important role in ovarian cancer stemness and tumorigenicity as well as resistance to platinum drug treatment in vitro and in vivo. CONCLUSIONS: These findings thus reveal a hitherto unreported interrelationship between c-Kit and PHB as well as the effects of raft-phospho-PHBY259 on ovarian cancer stemness and tumorigenicity mediated by the Notch3 and ß-catenin signaling pathways. Targeting the c-Kit/raft-phospho-PHBY259 axis may provide a new therapeutic strategy for treating patients with ovarian cancer.


Asunto(s)
Proliferación Celular , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/genética , Transducción de Señal/genética , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/fisiopatología , Prohibitinas
6.
J Invest Dermatol ; 140(9): 1771-1783.e6, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32035924

RESUMEN

The induction of immunogenic cell death (ICD) in cancer cells triggers specific immune responses against the same cancer cells. Imiquimod (IMQ) is a synthetic ligand of toll-like receptor 7 that exerts antitumor activity by stimulating cell-mediated immunity or by directly inducing apoptosis. Whether IMQ causes tumors to undergo ICD and elicits a specific antitumor immune response is unknown. We demonstrated that IMQ-induced ICD-associated features, including the surface exposure of calreticulin and the secretion of adenosine triphosphate and HMGB1, were mediated by ROS and endoplasmic reticulum stress. In a B16F10 melanoma mouse model, vaccinating mice with IMQ-induced ICD cell lysate or directly injecting IMQ in situ reduced tumor growth that was mediated by inducing tumor-specific T-cell proliferation, promoting tumor-specific cytotoxic killing by CD8+ T cells, and increasing the infiltration of various immune cells into tumor lesions. The ICD-associated features were crucial in the induction of specific antitumor immunity in vivo. The glycolytic inhibitor 2-deoxyglucose enhanced IMQ-induced ICD-associated features and strengthened the antitumor immunity mediated by IMQ-induced ICD cell lysate in p53-mutant cancer cells, which were IMQ-resistant in vitro. We conclude that IMQ is an authentic ICD inducer and provide a concept connecting IMQ-induced cancer cell death and antitumor immune responses.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxiglucosa/farmacología , Imiquimod/farmacología , Muerte Celular Inmunogénica/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral/trasplante , Desoxiglucosa/uso terapéutico , Sinergismo Farmacológico , Glucólisis/efectos de los fármacos , Humanos , Imiquimod/uso terapéutico , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología
7.
Cancers (Basel) ; 11(12)2019 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-31795213

RESUMEN

Dysregulation of forkhead box D1 (FOXD1) is known to promote tumor progression; however, its molecular mechanism of action is unclear. Based on microarray analysis, we identified galectin-3/LGALS3 (Gal-3) as a potential downstream target of FOXD1, as FOXD1 transactivated Gal-3 by interacting with the Gal-3 promoter to upregulate Gal-3 in FOXD1-overexpressing CL1-0 lung cancer cells. Ectopic expression of FOXD1 increased the expression of Gal-3 and the growth and motility of lung cancer cells, whereas depletion of Gal-3 attenuated FOXD1-mediated tumorigenesis. ERK1/2 interacted with FOXD1 in the cytosol and translocated FOXD1 into the nucleus to activate Gal-3. Gal-3 in turn upregulated FOXD1 via the transcription factor proto-oncogene 1 (ETS-1) to transactivate FOXD1. The increase in ETS-1/FOXD1 expression by Gal-3 was through Gal-3-mediated integrin-ß1 (ITGß1) signaling. The overexpression of both FOXD1 and Gal-3 form a positive regulatory loop to promote lung cancer aggressiveness. Moreover, both FOXD1 and Gal-3 were positively correlated in human lung cancer tissues. Our findings demonstrated that FOXD1 and Gal-3 form a positive feedback loop in lung cancer, and interference of this loop may serve as an effective therapeutic target for the treatment of lung cancers, particularly those related to dysregulation of Gal-3.

9.
Radiol Oncol ; 53(3): 307-315, 2019 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-31553703

RESUMEN

Background Endometrial adenocarcinoma (EAC) is one of the most commonly diagnosed gynaecological malignancies among female population of the developed countries. DUSP6 is a negative regulator of ERK signaling, which is a molecular switch involved in MAPK signaling during the progress of malignancies. DUSP6 was previously found to inhibit tumorigenesis and EMT-associated properties in several cancers, however, its exact role in EAC remains unclear Methods The level of DUSP6, (E-cad) and (N-cad) in EAC cancerous tissues and respective adjacent non-cancerous tissues were examined by western-blot or immunohistochemistry. The cell growth, invasion and migration abilities were measured in Ishikawa 3H12 endometrial cancer cell lines with overexpressed or knock down DUSP6. Protein levels of EMT-associated markers E-cadherin, N-cadherin and Vimentin were also determined. The impacts of DUSP6 on ERK signaling was assessed by detection of ERK and p-ERK. Results Down-regulation of DUSP6 was observed in EAC compared with the normal controls. The overexpression of DUSP6 significantly attenuated tumor cell growth, invasion, migration abilities and inhibited EMT-associated markers, while knock down of DUSP6 showed opposite trends. Overexpression of DUSP6 also down-regulated p-ERK and the knock down of DUSP6 inversely up-regulated p-ERK level. Conclusions DUSP6 inhibited cell growth, invasion and migration abilities in Ishikawa 3H12 cells as well as attenuating EMT-associated properties. This tumor suppressive effect of DUSP6 in EAC is achieved by inhibiting ERK signaling pathway.


Asunto(s)
Adenocarcinoma/metabolismo , Fosfatasa 6 de Especificidad Dual/metabolismo , Neoplasias Endometriales/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Sistema de Señalización de MAP Quinasas , Adenocarcinoma/fisiopatología , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Neoplasias Endometriales/fisiopatología , Quinasas MAP Reguladas por Señal Extracelular , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica
11.
Transl Cancer Res ; 8(6): 2307-2315, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35116983

RESUMEN

BACKGROUND: Increasing evidence has revealed that N 6-methyladenosine (m6A) modification is implicated in multiple biological functions in mammals. Methyltransferase-like 14 (METTL14), an important component of m6A modification, has been reported to play important roles in the pathogenesis of acute myeloid leukaemia and hepatocellular carcinoma metastasis. However, its role in cervical cancer remains unclear. METHODS: Expression of METTL14 was knocked down by shRNA-METTL14 interference in HPV-positive and HPV-negative cervical cancer cell lines SiHa and C33a. CCK8, colony formation, wound-healing, and Transwell assays were performed to evaluate the effects of METTL14 knockdown on the proliferation, migration and invasion abilities of SiHa and C33a cells. Flow cytometry analysis was utilized to detect cell cycle distribution, and the expression of related proteins was examined by western blot analysis. RESULTS: Bioinformatics analysis demonstrated that up-regulation of METTL14 acted as an adverse prognostic factor for overall survival in cervical cancer patients. We demonstrated that down-regulation of METTL14 inhibited the proliferation, migration and invasion abilities of SiHa and C33a cells. Moreover, silencing METTL14 induced cell cycle arrest in cervical cancer cells. METTL14 knockdown suppressed the PI3K/Akt/mTOR signaling pathway by decreasing the phosphorylation of Akt and mTOR, and the expression of downstream apoptosis-related proteins was also impacted. CONCLUSIONS: In conclusion, these data suggest an important oncogenic role of METTL14 in the growth and invasion of both HPV-positive and HPV-negative cervical cancer cells.

12.
Autophagy ; 14(12): 2065-2082, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30081720

RESUMEN

The relationship between macroautophagy/autophagy and miRNA in regulating cancer cell motility is not clearly delineated. Here, we found that induction of BECN1-dependent or -independent autophagy decreased ubiquitin-binding proteins SQSTM1/p62 and CALCOCO2/NDP52. Downregulation of SQSTM1 (but not CALCOCO2) led to a decrease of the miRNA-processing enzyme DICER1 and the miRNA effector AGO2. The autophagy-mediated reduction of levels of SQSTM1, DICER1 or AGO2 resulted in increased MIRLET7A-3P (but not MIRLET7A-5P or PRE-MIRLET7A miRNA) and suppressed ovarian cancer motility. The investigation of the MIRLET7A effects on cancer cell motility showed that synthetic MIRLET7A-3P (3 nM) inhibited, whereas MIRLET7A-5P (100 nM) increased cancer cell motility. Moreover, downregulation of MIRLET7A-3P with antisense of MIRLET7A-3P miRNA (MIRLET7A-3P inhibitor; 3 nM) reversed the nutrient depletion- and rVP1-mediated suppression of ovarian cancer cell motility. In addition, restoring SQSTM1, DICER1 and AGO2 with inhibition of autophagic degradation or overexpression of DICER1 and AGO2 reversed the autophagy-associated enhancement of MIRLET7A-3P and inhibition of motility. Examination of ovarian cancer tissue microarray further showed that the levels of SQSTM1, DICER1 and AGO2 in the tumor were higher than those in the non-tumor cells and negatively correlated with the levels of autophagy and MIRLET7A-3P. Our results demonstrated that induction of autophagy to decrease SQSTM1, DICER1 and AGO2 and increase MIRLET7A-3P is a potential therapeutic strategy for suppressing ovarian cancer cell motility. Abbreviations: ACTB: actin beta; AGO2: argonaute 2, RISC catalytic component; ATG: autophagy related; BCIP/NBT: 5-bromo-4-chloro-3-indolyl-phosphate/nitro blue tetrazolium; BECN1: beclin 1, autophagy related; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CQ: chloroquine; DICER1: dicer 1, ribonuclease III; EBSS: Earle balanced salt solution; FBS: fetal bovine serum; HGF: hepatocyte growth factor; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MIRLET7A: microRNA LET-7A: MIR16: microRNA 16; MIR29C: microRNA 29C; miRNA: microRNA; MMP: matrix metallopeptidase; PRE-MIRNA: precursor microRNA; PtdIns3K: class III phosphatidylinositol 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; RISC: RNA-induced silencing complex; rVP1: recombinant foot-and-mouth disease virus capsid protein VP1; siRNA: small interfering RNA; SQSTM1/p62: sequestosome 1; WIPI: WD repeat domain, phosphoinositide interacting.


Asunto(s)
Proteínas Argonautas/genética , Autofagia/fisiología , Movimiento Celular , ARN Helicasas DEAD-box/genética , MicroARNs/genética , Neoplasias Ováricas/patología , Ribonucleasa III/genética , Proteína Sequestosoma-1/metabolismo , Adenocarcinoma Papilar/genética , Adenocarcinoma Papilar/metabolismo , Adenocarcinoma Papilar/patología , Proteínas Argonautas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , ARN Helicasas DEAD-box/metabolismo , Regulación hacia Abajo/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Proteolisis , Ribonucleasa III/metabolismo , Transducción de Señal/genética
13.
Nat Commun ; 8(1): 299, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28824166

RESUMEN

The C-type lectin member 5A (CLEC5A) is a pattern recognition receptor for members of the Flavivirus family and has critical functions in response to dengue virus and Japanese encephalitis virus. Here we show that CLEC5A is involved in neutrophil extracellular trap formation and the production of reactive oxygen species and proinflammatory cytokines in response to Listeria monocytogenes. Inoculation of Clec5a -/- mice with L. monocytogenes causes rapid bacterial spreading, increased bacterial loads in the blood and liver, and severe liver necrosis. In these mice, IL-1ß, IL-17A, and TNF expression is inhibited, CCL2 is induced, and large numbers of CD11b+Ly6ChiCCR2hiCX3CR1low inflammatory monocytes infiltrate the liver. By day 5 of infection, these mice also have fewer IL-17A+ γδ T cells, severe liver necrosis and a higher chance of fatality. Thus, CLEC5A has a pivotal function in the activation of multiple aspects of innate immunity against bacterial invasion.The lectin receptor CLEC5A is a pattern recognition receptor that has been shown to detect dengue and Japanese encephalitis virus. Here the authors show that CLEC5A is needed for optimal ROS production, NET formation and other immune responses to Listeria monocytogenes in mice.


Asunto(s)
Inmunidad Innata/inmunología , Lectinas Tipo C/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores de Superficie Celular/inmunología , Animales , Citocinas/inmunología , Citocinas/metabolismo , Trampas Extracelulares/genética , Trampas Extracelulares/inmunología , Trampas Extracelulares/microbiología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Listeria monocytogenes/fisiología , Listeriosis/genética , Listeriosis/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo
14.
Oncotarget ; 7(32): 52255-52269, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27391343

RESUMEN

Activation of IKK enhances NF-κB signaling to facilitate cancer cell migration, invasion and metastasis. Here, we uncover the existence of a negative feedback loop of IKK. The transcription factor PATZ1 induces protein phosphatase-4 (PP4) regulatory subunit 2 (PP4R2) in an IKK-dependent manner. PP4R2 enhances the binding of PP4 to phosphorylated IKK to inactivate IKK/NF-κB signaling during sustained stimulation by cellular stimuli such as growth factors and inflammatory mediators. Matched pair studies reveal that primary lung cancers express more PATZ1 and PP4R2 than lymph node metastases in patients. Ectopic PATZ1 decreases invasion/colonization of lung cancers and prolongs the survival of xenograft mice. These effects of PATZ1 are reversed by downregulating PP4R2. Our results suggest that PATZ1 and PP4R2 provide negative feedback on IKK/NF-κB signaling to prevent cancer cells from over-stimulation from cellular stimuli; a decline in PATZ1 and PP4R2 is functionally associated with cancer migration/invasion and agents enhancing PATZ1 and PP4R2 are worth exploring to prevent invasion/metastasis of lung cancers.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Quinasa I-kappa B/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Ratones , Ratones SCID , FN-kappa B/metabolismo , Transducción de Señal/fisiología
16.
Int J Clin Exp Pathol ; 8(7): 7921-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26339357

RESUMEN

High dose methylprednisolone (HDMP) has been an effective salvage therapy for patients with relapsed chronic lymphocytic leukemia (CLL), while little is known about the exact mechanisms implicated in glucocorticoid-induced cell death. To explore the mechanism of glucocorticoid-induced cell death, we investigated the effect of HDMP on canonical Wnt signaling which emerged as a key pathway implicated in the pathogenesis of CLL. In this study, the human CLL cell line MEC-1 was incubated with various concentrations of methylprednisolone. Cell proliferation activity was detected by CCK8 assay, the apoptotic effect was evaluated by TUNEL assay. Western blot was used to detect active-caspase 3, and the key proteins in Wnt signaling pathway (LEF-1, ß-catenin). RT-PCR was performed to assess the mRNA levels of ß-catenin, LEF-1, c-myc and cyclin D1. We observed that high concentration of methylprednisolone could suppress the proliferation activity of MEC-1 cells, promote the relative expression of active-caspase 3, and induce apoptotic cell death. Furthermore, methylprednisolone could inhibit LEF-1 protein expression, consequently down-regulate mRNA levels of c-myc and cyclin D1, but could not affect the transcription level of ß-catenin and LEF-1 mRNA. The results of this study indicate that methylprednisolone can suppress Wnt signaling pathway by down-regulating LEF-1 protein expression, indicating a novel mechanism for HDMP therapy in CLL.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Metilprednisolona/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Ciclina D1/metabolismo , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Factor de Unión 1 al Potenciador Linfoide/genética , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Factores de Tiempo , Regulación hacia Arriba
17.
PLoS One ; 10(6): e0128926, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26042735

RESUMEN

Macrophage proliferation and migration are important for many facets of immune response. Here we showed that stimulation of macrophages with type B CpG oligodeoxynucleotides (CpG-B ODNs) such as CpG-ODN 1668 increased the production of anti-inflammatory cytokine interleukin 1 receptor antagonist (IL-1Ra) in a TLR9- and MyD88-dependent manner. The CpG-B ODNs-induced IL-1Ra increased macrophage migration and promoted macrophage proliferation by down-regulating the expression of a cell cycle negative regulator, p27 to increase cell population in the S phase. The induction of IL-1Ra by CpG-B ODNs was F-spondin dependent. Knockdown of F-spondin and IL-1Ra decreased CpG-B ODNs-induced macrophage migration whereas overexpression of IL-1Ra increased migration of those cells. These findings demonstrated novel roles for F-spondin and IL-1Ra in CpG-B ODNs-mediated cell proliferation and migration of macrophages.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Oligodesoxirribonucleótidos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Factor 88 de Diferenciación Mieloide/metabolismo , Células RAW 264.7 , Fase S/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
J Dermatol Sci ; 78(2): 108-16, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25766763

RESUMEN

BACKGROUND: AMP-activated protein kinase (AMPK), a principal intracellular energy sensor, plays a crucial role in cell growth, proliferation, apoptosis and autophagy. Imiquimod (IMQ) directly exhibits anti-tumor activity through the induction of apoptosis and autophagic cell death. OBJECTIVE: To evaluate the role of AMPK in IMQ-induced apoptosis and autophagy. METHODS: The phosphorylation of AMPK and its substrates was detected by immunoblotting. ATP contents were analyzed by an ATP bioluminescence assay. The upstream signaling for AMPK activation was dissected by examination of TLR7/8 expression, over-expression of TLR7/8, the addition of AMPK kinase inhibitors, and the genetic silencing of Myd88 and LKB1. The role of AMPK activation in IMQ-induced autophagy and apoptosis was assessed by inhibiting AMPK, genetically silencing AMPK and over-expressing AMPK dominant-negative mutants. Autophagy and apoptosis were evaluated by a DNA content assay, immunoblotting, EGFP-LC3 puncta detection and acridine orange staining. RESULTS: IMQ could activate AMPK and autophagy in cancer cells not expressing TLR7/8. IMQ caused ATP depletion and induced LKB1-mediated AMPK activation. The down-regulation of AMPK activity via pharmacological inhibition and genetic silencing resulted in reduced IMQ-induced apoptosis but did not influence autophagy, and this rescue effect was associated with the retention of translation factor activity and anti-apoptotic Bcl-2 family member Mcl-1 protein expression levels. CONCLUSION: IMQ induces AMPK activation independent of TLR7/8 expression, resulting in translation inhibition and subsequent apoptosis through ATP depletion and LKB1 signaling, in skin tumor cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Aminoquinolinas/farmacología , Antineoplásicos/farmacología , Carcinoma Basocelular/metabolismo , Carcinoma de Células Escamosas/metabolismo , Melanoma/metabolismo , Biosíntesis de Proteínas/efectos de los fármacos , Neoplasias Cutáneas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/genética , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma Basocelular/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Regulación hacia Abajo , Activación Enzimática/efectos de los fármacos , Silenciador del Gen , Humanos , Imiquimod , Melanoma/tratamiento farmacológico , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/genética , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Neoplasias Cutáneas/tratamiento farmacológico , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo
19.
Oncotarget ; 6(1): 381-93, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25575814

RESUMEN

Growth factors and COX-2/PGE2 enhance lung cancer invasion/metastasis via PI3K/Akt and RAS/Raf. Here, we explored their mechanism of action further. We found first that higher levels of migration inducting gene-7 protein (MIG-7) and PHB phosphorylated at threonine 258 (phospho-PHBT258) are positively correlated with advanced stages of human lung cancer in tissue microarray. PGE2 or growth factors such as EGF, HGF and IGF-1 increased complex formation of phospho-PHBT258 with Ras, phospho-AktS473, phospho-Raf-1S338, MEKK1 and IKKα/ßS176/180 in the raft domain transiently within 1 hour and MIG-7 in the cytosol 12-24 hours later. Association of phospho-PHBT258 with MEKK1 but not MEKK3 activates IKK/IκB/NF-κB and MEK/ERK to increase cellular COX-2/PGE2 and an E-cadherin suppressor Snail leading to enhancement of epithelial-mesenchymal transition (EMT) and lung cancer migration/invasion. MIG-7, on the other hand, was induced by growth factors and PGE2 via Akt/GSK-3ß in a phospho-PHBT258 independent manner. MIG-7 increased two E-cadherin suppressors ZEB-1 and Twist to enhance EMT and cancer migration/invasion. Downregulating phospho-PHBT258 and MIG-7 had an additive effect on attenuating lung cancer invasion/metastasis and prolonging the survival of xenograft mice. Phospho-PHBT258 and MIG-7 may thus play complementary roles in the initiation and sustainment of the effects of growth factors and COX-2/PGE2 on cancer invasion/metastasis.


Asunto(s)
Movimiento Celular/fisiología , Neoplasias Pulmonares/patología , Invasividad Neoplásica/patología , Proteínas de Neoplasias/metabolismo , Proteínas Represoras/metabolismo , Animales , Línea Celular Tumoral , Xenoinjertos , Humanos , Immunoblotting , Inmunoprecipitación , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones SCID , Fosforilación , Prohibitinas , ARN Interferente Pequeño , Transducción de Señal/fisiología , Análisis de Matrices Tisulares , Transfección
20.
Curr Protoc Protein Sci ; 78: 6.5.1-6.5.30, 2014 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-25367010

RESUMEN

Heterologous expression of recombinant proteins in E. coli often results in the formation of insoluble and inactive protein aggregates, commonly referred to as inclusion bodies. To obtain the native (i.e., correctly folded) and hence active form of the protein from such aggregates, four steps are usually followed: (1) the cells are lysed, (2) the cell wall and outer membrane components are removed, (3) the aggregates are solubilized (or extracted) with strong protein denaturants, and (4) the solubilized, denatured proteins are folded with concomitant oxidation of reduced cysteine residues into the correct disulfide bonds to obtain the native protein. This unit features three different approaches to the final step of protein folding and purification. In the first, guanidine·HCl is used as the denaturant, after which the solubilized protein is folded (before purification) in an "oxido-shuffling" buffer system to increase the rate of protein oxidation. In the second, acetic acid is used to solubilize the protein, which is then partially purified by gel filtration before folding; the protein is then folded and oxidized by simple dialysis against water. Thirdly, folding and purification of a fusion protein using metal-chelate affinity chromatography are described.


Asunto(s)
Escherichia coli/química , Guanidina/química , Cuerpos de Inclusión/química , Desnaturalización Proteica , Replegamiento Proteico , Proteínas Recombinantes , Animales , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...