Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096780

RESUMEN

ADAM9 (A disintegrin and a metalloprotease 9) is a membrane-anchored protein that participates in a variety of physiological functions, primarily through the disintegrin domain for adhesion and the metalloprotease domain for ectodomain shedding of a wide variety of cell surface proteins. ADAM9 influences the developmental process, inflammation, and degenerative diseases. Recently, increasing evidence has shown that ADAM9 plays an important role in tumor biology. Overexpression of ADAM9 has been found in several cancer types and is correlated with tumor aggressiveness and poor prognosis. In addition, through either proteolytic or non-proteolytic pathways, ADAM9 promotes tumor progression, therapeutic resistance, and metastasis of cancers. Therefore, comprehensively understanding the mechanism of ADAM9 is crucial for the development of therapeutic anti-cancer strategies. In this review, we summarize the current understanding of ADAM9 in biological function, pathophysiological diseases, and various cancers. Recent advances in therapeutic strategies using ADAM9-related pathways are presented as well.


Asunto(s)
Proteínas ADAM/química , Proteínas ADAM/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Neoplasias/patología , Enfermedades Neurodegenerativas/patología , Enfermedades de la Retina/patología , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/terapia , Enfermedades Neurodegenerativas/metabolismo , Compuestos de Fenilurea/farmacología , Piridinas/farmacología , Enfermedades de la Retina/metabolismo , Sorafenib/farmacología , Microambiente Tumoral
2.
Oncologist ; 20(5): 539-45, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25876992

RESUMEN

BACKGROUND: The role of adjuvant chemotherapy for the treatment of nasopharyngeal carcinoma (NPC) is controversial, and the identification of adequate predictive factors is warranted. Therefore, we aimed to investigate whether the mean standardized uptake value (SUV) measured on [(18)F]-fluorodeoxyglucose (FDG) positron emission tomography (PET) could predict the survival benefits for NPC patients that receive adjuvant chemotherapy. MATERIALS AND METHODS: The data for 174 NPC patients who underwent PET/computed tomography before chemoradiation between January 2004 and January 2012 were reviewed. The SUV75% was recorded for primary tumors. All patients received intensity-modulated radiotherapy and cisplatin-based chemotherapy. Adjuvant chemotherapy consisted of 3 cycles of 75 mg/m(2) cisplatin and 1,000 mg/m(2) fluorouracil for 4 days. RESULTS: The optimal cutoff value was 8.35 for SUV75%, with 112 (64.4%) patients having lower SUV75% and 62 (35.6%) having higher SUV75%. Patients with lower SUV75% had significantly better 5-year overall survival (OS) and distant metastasis-free survival. Multivariate analysis revealed that tumor stage, SUV75%, and adjuvant chemotherapy were significant prognostic factors for OS. Patients with higher SUV75% had significantly higher 5-year OS rates with adjuvant chemotherapy than without adjuvant chemotherapy (84.3% vs. 32.4%, respectively; p < .001). However, in the lower SUV75% group, no differences in 5-year OS were observed between patients who received and those who did not receive adjuvant chemotherapy (92.4% vs. 93.3%, respectively; p = .682). CONCLUSION: The SUV75% on FDG PET for primary tumors could successfully identify NPC patients who may benefit from adjuvant chemotherapy.


Asunto(s)
Quimioterapia Adyuvante , Fluorodesoxiglucosa F18/administración & dosificación , Neoplasias Nasofaríngeas/tratamiento farmacológico , Neoplasias Nasofaríngeas/radioterapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma , Cisplatino/administración & dosificación , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico por imagen , Neoplasias Nasofaríngeas/patología , Estadificación de Neoplasias , Tomografía de Emisión de Positrones , Pronóstico , Radiografía , Tasa de Supervivencia
3.
Blood ; 121(8): 1357-66, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-23243276

RESUMEN

Adult patients with disseminated nontuberculous mycobacterial (dNTM) infections usually have severe immune system defects. Recently, several studies have shown that anti-IFN-γ autoantibodies may play an important role in the pathogenicity of dNTM infections. A considerable proportion of reported cases of anti-IFN-γ autoantibodies show either clinical or laboratory evidence of autoimmune disease. In the present study, we identified 19 formerly healthy adults who later developed dNTM infections, of whom 17 were further investigated immunologically. High-titer anti-IFN-γ autoantibodies capable of inhibiting IL-12 production in vitro were found in the plasma of all of these patients. In addition to dNTM infection, 35% and 71% of our patients also suffered from salmonellosis and herpes zoster, respectively. This observation suggests that IFN-γ may be crucial in controlling salmonella infection and reactivating latent varicella-zoster virus infection in humans. 2 HLA alleles, DRB1*16:02 DQB1*05:02 (odds ratio 8.68; 95% confidence interval, 3.47-21.90; P = 1.1 × 10(-6); Pc = 3.08 × 10(-5) and odds ratio 7.16; 95% confidence interval, 3.02-17.05; P = 1 × 10(-7); Pc = 1.4 × 10(-6), respectively), were found in 82% (14 of 17) of our patients. In conclusion, our data suggest that anti-IFN-γ autoantibodies may play a critical role in the pathogenesis of dNTM infections and reactivation of latent varicella-zoster virus infection and are associated with HLA-DRB1*16:02 and HLA-DQB1*05:02.


Asunto(s)
Autoanticuerpos/inmunología , Cadenas beta de HLA-DQ/inmunología , Cadenas HLA-DRB1/inmunología , Herpes Zóster/inmunología , Interferón gamma/inmunología , Infecciones por Mycobacterium no Tuberculosas/inmunología , Anciano , Anciano de 80 o más Años , Autoanticuerpos/sangre , Autoanticuerpos/genética , Coinfección/genética , Coinfección/inmunología , Coinfección/mortalidad , Femenino , Frecuencia de los Genes , Cadenas beta de HLA-DQ/genética , Cadenas HLA-DRB1/genética , Herpes Zóster/genética , Herpes Zóster/mortalidad , Herpesvirus Humano 3/inmunología , Prueba de Histocompatibilidad , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Interferón gamma/sangre , Subunidad p40 de la Interleucina-12/sangre , Subunidad p40 de la Interleucina-12/inmunología , Masculino , Persona de Mediana Edad , Infecciones por Mycobacterium no Tuberculosas/genética , Infecciones por Mycobacterium no Tuberculosas/mortalidad , Estudios Seroepidemiológicos , Latencia del Virus/inmunología
4.
Kidney Int ; 86(5): 1023-30, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24694991

RESUMEN

Pneumococcal disease leads to renal complications ranging from persistent proteinuria to end-stage renal disease (ESRD) in pediatric patients. However, long-term renal effects after pneumococcal pneumonia infection in adult patients remains largely unknown. To evaluate this we conducted a population-based retrospective cohort study consisting of 18,733 adult patients at the time of pneumococcal pneumonia diagnosis, using claims data from Taiwan's National Health Insurance Research Database (NHIRD) with a comparison cohort of 73,409 age- and gender-matched patients without pneumococcal pneumonia. The overall incidence rate ratio of ESRD was 23% higher in those with pneumococcal pneumonia than in those without pneumococcal pneumonia (5.26 vs. 3.10 per 1000 person-years), with an adjusted hazard ratio of 1.14 (95% confidence interval 1.01-1.29). In addition, the risk of developing ESRD was associated with covariates including age, gender, chronic kidney disease, diabetes mellitus, hypertension, hyperlipidemia, chronic obstructive pulmonary disease, and heart failure. The ESRD cumulative incidence curve showed a considerably higher risk of ESRD in those with pneumococcal pneumonia than in those without pneumococcal pneumonia (significant by log-rank test). Thus, pneumococcal pneumonia may be associated with an increased risk of ESRD in adult patients. A long-term follow-up of renal function is recommended for adult hospitalized patients with pneumococcal pneumonia infection.


Asunto(s)
Hospitalización , Fallo Renal Crónico/epidemiología , Neumonía Neumocócica/epidemiología , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Comorbilidad , Bases de Datos Factuales , Femenino , Humanos , Incidencia , Fallo Renal Crónico/diagnóstico , Masculino , Persona de Mediana Edad , Neumonía Neumocócica/diagnóstico , Neumonía Neumocócica/microbiología , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo , Factores Sexuales , Taiwán/epidemiología , Factores de Tiempo , Adulto Joven
5.
Toxicol Appl Pharmacol ; 279(3): 351-363, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25018059

RESUMEN

Oxidized LDL (oxLDL) induces a pro-oxidative environment and promotes apoptosis, causing the progression of renal diseases in humans. Taurine is a semi-essential amino acid in mammals and has been shown to be a potent endogenous antioxidant. The kidney plays a pivotal role in maintaining the balance of taurine. However, the mechanisms underlying the protective effects of taurine against oxLDL-induced injury in renal epithelial cells have not been clarified. In the present study, we investigated the anti-apoptotic effects of taurine on human proximal tubular epithelial (HK-2) cells exposed to oxLDL and explored the related mechanisms. We observed that oxLDL increased the contents of ROS and of malondialdehyde (MDA), which is a lipid peroxidation by-product that acts as an indicator of the cellular oxidation status. In addition, oxLDL induced cell death and apoptosis in HK-2 cells. Pretreatment with taurine at 100 µM significantly attenuated the oxLDL-induced cytotoxicity. We determined that oxLDL triggered the phosphorylation of ERK and, in turn, the activation of p53 and other apoptosis-related events, including calcium accumulation, destabilization of the mitochondrial permeability and disruption of the balance between pro-apoptotic Bax and anti-apoptotic Bcl-2 proteins. The malfunctions induced by oxLDL were effectively blocked by taurine. Thus, our results suggested that taurine exhibits potential therapeutic activity by preventing oxLDL-induced nephrotoxicity. The inhibition of oxLDL-induced epithelial apoptosis by taurine was at least partially due to its anti-oxidant activity and its ability to modulate the ERK and p53 apoptotic pathways.


Asunto(s)
Antioxidantes , Apoptosis/fisiología , Lipoproteínas LDL/antagonistas & inhibidores , Lipoproteínas LDL/toxicidad , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología , Taurina/farmacología , Proteína p53 Supresora de Tumor/fisiología , Western Blotting , Señalización del Calcio/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN/biosíntesis , ADN/genética , Células Epiteliales , Humanos , Etiquetado Corte-Fin in Situ , Riñón/citología , Riñón/efectos de los fármacos , Peroxidación de Lípido/efectos de los fármacos , Lipoproteínas/aislamiento & purificación , Lipoproteínas/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Confocal , Superóxido Dismutasa/metabolismo
6.
Am J Cancer Res ; 14(4): 1850-1865, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38726266

RESUMEN

Chronic inflammation associated with lung cancers contributes to immunosuppressive tumor microenvironments, reducing CD8+ T-cell function and leading to poor patient outcomes. A disintegrin and metalloprotease domain 9 (ADAM9) promotes cancer progression. Here, we aim to elucidate the role of ADAM9 in the immunosuppressive tumor microenvironment. A bioinformatic analysis of TIMER2.0 was used to investigate the correlation of ADAM9 and to infiltrate immune cells in the human lung cancer database and mouse lung tumor samples. Flow cytometry, immunohistochemistry, and RNA sequencing (RNA-seq) were performed to investigate the ADAM9-mediated immunosuppressive microenvironment. The coculture system of lung cancer cells with immune cells, cytokine array assays, and proteomic approach was used to investigate the mechanism. By analyzing the human LUAD database and the mouse lung cancer models, we showed that ADAM9 was associated with the immunosuppressive microenvironment. Additionally, ADAM9 released IL6 protein from cancer cells to inhibit IL12p40 secretion from dendritic cells, therefore leading to dendritic cell dysfunction and further affecting T-cell functions. Proteomic analysis indicated that ADAM9 promoted cholesterol biosynthesis and increased IL6-STAT3 signaling. Mechanistically, ADAM9 reduced the protein stability of LDLR, resulting in reduced cholesterol uptake and induced cholesterol biosynthesis. Moreover, LDLR reduction enhanced IL6-STAT3 activation. We reveal that ADAM9 has a novel biological function that drives the immunosuppressive tumor microenvironment by linking lung cancer's metabolic and signaling axes. Thus, by targeting ADAM9 an innovative and promising therapeutic opportunity was indicated for regulating the immunosuppression of lung cancer.

7.
Nat Cancer ; 5(3): 400-419, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38267627

RESUMEN

Kirsten rat sarcoma virus (KRAS) signaling drives pancreatic ductal adenocarcinoma (PDAC) malignancy, which is an unmet clinical need. Here, we identify a disintegrin and metalloproteinase domain (ADAM)9 as a modulator of PDAC progression via stabilization of wild-type and mutant KRAS proteins. Mechanistically, ADAM9 loss increases the interaction of KRAS with plasminogen activator inhibitor 1 (PAI-1), which functions as a selective autophagy receptor in conjunction with light chain 3 (LC3), triggering lysosomal degradation of KRAS. Suppression of ADAM9 by a small-molecule inhibitor restricts disease progression in spontaneous models, and combination with gemcitabine elicits dramatic regression of patient-derived tumors. Our findings provide a promising strategy to target the KRAS signaling cascade and demonstrate a potential modality to enhance sensitivity to chemotherapy in PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Proteínas Proto-Oncogénicas p21(ras) , Proliferación Celular , Neoplasias Pancreáticas/tratamiento farmacológico , Carcinoma Ductal Pancreático/tratamiento farmacológico , Gemcitabina , Proteínas de la Membrana/metabolismo , Proteínas ADAM/metabolismo , Proteínas ADAM/uso terapéutico
8.
Microbiol Spectr ; : e0385422, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37713503

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of the Coronavirus disease-19 (COVID-19) pandemic, utilizes angiotensin-converting enzyme 2 (ACE2) as a receptor for virus infection. However, the expression pattern of ACE2 does not coincide with the tissue tropism of SARS-CoV-2, hinting that other host proteins might be involved in facilitating SARS-CoV-2 entry. To explore potential host factors for SARS-CoV-2 entry, we performed an arrayed shRNA screen in H1650 and HEK293T cells. Here, we identified a disintegrin and a metalloproteinase domain 9 (ADAM9) protein as an important host factor for SARS-CoV-2 entry. Our data showed that silencing ADAM9 reduced virus entry, while its overexpression promoted infection. The knockdown of ADAM9 decreased the infectivity of the variants of concern tested-B.1.1.7 (alpha), B.1.617.2 (delta), and B.1.1.529 (omicron). Furthermore, mechanistic studies indicated that ADAM9 is involved in the binding and endocytosis stages of SARS-CoV-2 entry. Through immunoprecipitation experiments, we demonstrated that ADAM9 binds to the S1 subunit of the SARS-CoV-2 Spike. Additionally, ADAM9 can interact with ACE2, and co-expression of both proteins markedly enhances virus infection. Moreover, the enzymatic activity of ADAM9 facilitates virus entry. Our study reveals an insight into the mechanism of SARS-CoV-2 virus entry and elucidates the role of ADAM9 in virus infection. IMPORTANCE COVID-19, an infectious respiratory disease caused by SARS-CoV-2, has greatly impacted global public health and the economy. Extensive vaccination efforts have been launched worldwide over the last couple of years. However, several variants of concern that reduce the efficacy of vaccines have kept emerging. Thereby, further understanding of the mechanism of SARS-CoV-2 entry is indispensable, which will allow the development of an effective antiviral strategy. Here, we identify a disintegrin and metalloproteinase domain 9 (ADAM9) protein as a co-factor of ACE2 important for SARS-CoV-2 entry, even for the variants of concern, and show that ADAM9 interacts with Spike to aid virus entry. This virus-host interaction could be exploited to develop novel therapeutics against COVID-19.

9.
Mol Oncol ; 17(8): 1648-1665, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37013960

RESUMEN

CUB domain-containing protein 1 (CDCP1) contributes to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) resistance by regulating EGFR signaling pathways and is a potential target in lung cancer treatment. This study aims to identify a CDCP1 reducer that synergistically improves TKI treatment. Utilizing a high-throughput drug screening system, a phytoestrogen 8-isopentenylnaringenin (8PN) was identified. Upon 8PN treatment, CDCP1 protein levels and malignant features were reduced. 8PN exposure caused the accumulation of lung cancer cells in G0/G1 phase and increased the proportion of senescent cells. In EGFR TKI-resistant lung cancer cells, the combination of 8PN and TKI synergistically reduced cell malignance, inhibited downstream EGFR pathway signaling, and exerted additive effects on cell death. Moreover, combination therapy effectively reduced tumor growth and enhanced tumor necrosis in tumor xenograft mice models. Mechanistically, 8PN increased interleukin (IL)6 and IL8 expression, induced neutrophil infiltration, and enhanced neutrophil-mediated cytotoxicity to attenuate lung cancer cell growth. In conclusion, 8PN enhances the anticancer efficacy of EGFR TKI on lung cancer and triggers neutrophil-dependent necrosis, highlighting the potential to overcome TKI resistance in lung cancer patients who have EGFR mutation.


Asunto(s)
Receptores ErbB , Neoplasias Pulmonares , Humanos , Animales , Ratones , Receptores ErbB/genética , Resistencia a Antineoplásicos , Neoplasias Pulmonares/genética , Necrosis , Inhibidores de Proteínas Quinasas/farmacología , Línea Celular Tumoral , Mutación , Antígenos de Neoplasias , Moléculas de Adhesión Celular/genética
10.
Life Sci ; 302: 120635, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35569571

RESUMEN

AIMS: Articulatin (AT), purified from the Chinese mistletoe (Viscum articulatum), belongs to the family of type II ribosome-inactivating proteins (RIPs) that contain two subunits, the A and B chains. The B chain of AT is believed to function by means of interacting with the galactose moiety of glycoproteins or glycolipids on the cell membrane and is internalized into cells through endocytosis. In the study, we aim to investigate the immunomodulatory effects of recombinant articulatin B chain (rATB) on mouse bone marrow-derived dendritic cells (BM-DCs). MAIN METHODS: Detection of surface markers expression on BM-DCs by flow cytometry. Analysis of RNA and protein expression by RNAseq and Western blotting assays. Assessment of the adaptive immune responses using an in vivo mouse model. KEY FINDING: Our study presents novel results showing the activation of mouse BM-DCs by rATB, which leads to the induction of CD80, CD86, and MHC II expression as well as primed type I CD4+ T cell differentiation and CD8+ T cell activation. RNAseq and Western blotting assays revealed rATB-induced BM-DC activation to be dependent on the MAPK and NF-κB signaling pathways. In a mouse model, rATB was observed to have adjuvant effects that induced an antigen-specific Th1 immune response. SIGNIFICANCE: Based on in vitro and in vivo assays, this study shows rATB acting as a potential adjuvant that induces BM-DC activation and antigen-specific Th1 related immune response. rATB might have potential applicability in the development of vaccines against pathogens and tumors.


Asunto(s)
Antineoplásicos , Linfocitos T Citotóxicos , Ratones , Animales , Células Dendríticas , Diferenciación Celular , Activación de Linfocitos , Antineoplásicos/farmacología , Linfocitos T Colaboradores-Inductores , Células TH1 , Ratones Endogámicos C57BL , Citocinas/metabolismo
11.
Comput Struct Biotechnol J ; 20: 4626-4635, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36090818

RESUMEN

Lung cancer is a major cause of cancer-associated deaths worldwide, and lung adenocarcinoma (LUAD) is the most common lung cancer subtype. Micro RNAs (miRNAs) regulate the pattern of gene expression in multiple cancer types and have been explored as potential drug development targets. To develop an oncomiR-based panel, we identified miRNA candidates that show differential expression patterns and are relevant to the worse 5-year overall survival outcomes in LUAD patient samples. We further evaluated various combinations of miRNA candidates for association with 5-year overall survival and identified a four-miRNA panel: miR-9-5p, miR-1246, miR-31-3p, and miR-3136-5p. The combination of these four miRNAs outperformed any single miRNA for predicting 5-year overall survival (hazard ratio [HR]: 3.47, log-rank p-value = 0.000271). Experiments were performed on lung cancer cell lines and animal models to validate the effects of these miRNAs. The results showed that singly transfected antagomiRs largely inhibited cell growth, migration, and invasion, and the combination of all four antagomiRs considerably reduced cell numbers, which is twice as effective as any single miRNA-targeted transfected. The in vivo studies revealed that antagomiR-mediated knockdown of all four miRNAs significantly reduced tumor growth and metastatic ability of lung cancer cells compared to the negative control group. The success of these in vivo and in vitro experiments suggested that these four identified oncomiRs may have therapeutic potential.

12.
Int J Biol Sci ; 17(14): 3898-3910, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34671207

RESUMEN

Hypoxia and angiogenesis play key roles in the pathogenesis of esophageal squamous cell carcinoma (ESCC), but regulators linking these two pathways to drive tumor progression remain elusive. Here we provide evidence of ADAM9's novel function in ESCC progression. Increasing expression of ADAM9 was correlated with poor clinical outcomes in ESCC patients. Suppression of ADAM9 function diminished ESCC cell migration and in vivo metastasis in ESCC xenograft mouse models. Using cellular fractionation and imaging, we found a fraction of ADAM9 was present in the nucleus and was uniquely associated with gene loci known to be linked to the angiogenesis pathway demonstrated by genome-wide ChIP-seq. Mechanistically, nuclear ADAM9, triggered by hypoxia-induced translocation, functions as a transcriptional repressor by binding to promoters of genes involved in the negative regulation of angiogenesis, and thereby promotes tumor angiogenesis in plasminogen/plasmin pathway. Moreover, ADAM9 suppresses plasminogen activator inhibitor-1 gene transcription by interacting with its transcription factors at the promoter. Our findings uncover a novel regulatory mechanism of ADAM9 as a transcriptional regulator in angiogenesis and highlight ADAM9 as a promising therapeutic target for ESCC treatment.


Asunto(s)
Proteínas ADAM/fisiología , Neoplasias Esofágicas/irrigación sanguínea , Carcinoma de Células Escamosas de Esófago/irrigación sanguínea , Proteínas de la Membrana/fisiología , Neovascularización Patológica/fisiopatología , Factores de Transcripción/fisiología , Animales , Movimiento Celular , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/patología , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Ratones , Ratones SCID , Neovascularización Patológica/genética , Inhibidor 1 de Activador Plasminogénico/genética , Pronóstico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Mol Ther Nucleic Acids ; 25: 1-10, 2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34141460

RESUMEN

Lung adenocarcinoma (LUAD), the most common histological type of non-small cell lung cancer, is one of the most malignant and deadly diseases. Current treatments for advanced LUAD patients are far from ideal and require further improvements. Here, we utilized a systematic integrative analysis of LUAD microRNA sequencing (miRNA-seq) and RNA-seq data from The Cancer Genome Atlas (TCGA) to identify clinically relevant tumor suppressor miRNAs. Three miRNA candidates (miR-195-5p, miR-101-3p, and miR-338-5p) were identified based on their differential expressions, survival significance levels, correlations with targets, and an additive effect on survival among them. We further evaluated mimics of the three miRNAs to determine their therapeutic potential in inhibiting cancer progression. The results showed not only that each of the miRNA mimics alone but also the three miRNA mimics in combination were efficient at inhibiting tumor growth and progression with equal final concentrations, meaning that the three miRNA mimics in combination were more effective than the single miRNA mimics. Moreover, the combined miRNA mimics provided significant therapeutic effects in terms of reduced tumor volume and metastasis nodules in lung tumor animal models. Hence, our findings show the potential of using the three miRNAs in combination to treat LUAD patients with poor survival outcomes.

14.
Theranostics ; 11(8): 3661-3675, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33664854

RESUMEN

Rationale: Lung adenocarcinoma (LUAD) is an aggressive disease with high propensity of metastasis. Among patients with early-stage disease, more than 30% of them may relapse or develop metastasis. There is an unmet medical need to stratify patients with early-stage LUAD according to their risk of relapse/metastasis to guide preventive or therapeutic approaches. In this study, we identified 4 genes that can serve both therapeutic and diagnostic (theranostic) purposes. Methods: Three independent datasets (GEO, TCGA, and KMPlotter) were used to evaluate gene expression profile of patients with LUAD by unbiased screening approach. Upon significant genes uncovered, functional enrichment analysis was carried out. The predictive power of their expression on patient prognosis were evaluated. Once confirmed their theranostic roles by integrated bioinformatics, we further conducted in vitro and in vivo validation. Results: We found that four genes (ADAM9, MTHFD2, RRM2, and SLC2A1) were associated with poor patient outcomes with an increased hazard ratio in LUAD. Knockdown of them, both separately and simultaneously, suppressed lung cancer cell proliferation and migration ability in vitro and prolonged survival time in metastatic tumor mouse models. Moreover, these four biomarkers were found to be overexpressed in tumor tissues from LUAD patients, and the total immunohistochemical staining scores correlated with poor prognosis. Conclusions: These results suggest that these four identified genes could be theranostic biomarkers for stratifying high-risk patients who develop relapse/metastasis in early-stage LUAD. Developing therapeutic approaches for the four biomarkers may benefit early-stage LUAD patients after surgery.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/secundario , Biomarcadores de Tumor/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células A549 , Proteínas ADAM/antagonistas & inhibidores , Proteínas ADAM/genética , Adenocarcinoma del Pulmón/cirugía , Aminohidrolasas/antagonistas & inhibidores , Aminohidrolasas/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Técnicas de Silenciamiento del Gen , Transportador de Glucosa de Tipo 1/antagonistas & inhibidores , Transportador de Glucosa de Tipo 1/genética , Células HEK293 , Humanos , Neoplasias Pulmonares/cirugía , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/antagonistas & inhibidores , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Ratones , Ratones SCID , Enzimas Multifuncionales/antagonistas & inhibidores , Enzimas Multifuncionales/genética , Medicina de Precisión , Pronóstico , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/genética , Factores de Riesgo , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Am J Cancer Res ; 10(11): 3828-3837, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33294270

RESUMEN

Metastasis is a crucial hallmark of cancer progression and remains the primary cause of patient deaths. Metastasis-associated proteases contribute to cancer progression by disrupting the extracellular matrix interaction to facilitate the spreading of cancer cells to other organs. ADAM9, a type of metalloprotease, has been reported to promote tumor biology and is associated with clinicopathological features such as poor outcome, therapy resistance, and metastasis formation. Targeting ADAM9 might serve as a putative therapeutic application; however, this option is currently unavailable. Resveratrol, a polyphenol from plants, has been shown to be promising for cancer treatment due to its wide variety of biological effects with few side effects. In this study, we demonstrated that resveratrol inhibits cancer cell migration and viability in lung and esophageal cancer cells through the regulation of ADAM9. Mechanistically, resveratrol inhibits ADAM9 protein expression in cancer cells through the ubiquitin-proteasome pathway. Moreover, resveratrol provides synergistic anticancer effects when combined with clinical chemotherapeutics. Our data suggests that resveratrol may inhibit human lung cancer and ESCC progression by inhibiting ADAM9 expression, thus providing a potential mechanism for the anticancer action of resveratrol.

16.
Int J Oncol ; 56(1): 165-177, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31789397

RESUMEN

Semaphorin 5A (SEMA5A), a member of the semaphorin family, plays an important role in axonal guidance. Previously, the authors identified another possible role of SEMA5A as a prognostic biomarker for non­smoking women with lung adenocarcinoma in Taiwan, and this phenomenon has been validated in other ethnic groups. However, the functional significance of SEMA5A in lung adenocarcinoma remains unclear. Therefore, we assessed the function of SEMA5A in three lung adenocarcinoma cell lines in this study. Kaplan­Meier Plotter for lung cancer was conducted for survival analyses. Reverse transcription­quantitative PCR (RT­qPCR) and western blot analysis were performed to investigate the expression and post­translational regulation of SEMA5A in lung adenocarcinoma cell lines. A pre­designed PyroMark CpG assay and 5­aza­2'­deoxycytidine treatment were used to measure the methylation levels of SEMA5A. The biological functions of lung adenocarcinoma cells overexpressing SEMA5A were investigated by microarrays, and validated both in vitro (proliferation, colony formation and migration assays) and in vivo (tumor xenografts) experiments. The results revealed that the hypermethylation of SEMA5A and the cleavage of the extracellular domain of SEMA5A were responsible for the downregulation of the SEMA5A levels in lung adenocarcinoma cells (A549 and H1299) as compared to the normal controls. Functional analysis of SEMA5A­regulated genes revealed that they were involved in cellular growth and proliferation. The overexpression of SEMA5A in A549 and H1299 cells significantly decreased the proliferation (P<0.01), colony formation (P<0.001) and migratory ability (P<0.01) of the cells. The suppressive effects of SEMA5A on the proliferative and migratory ability of the cells were also observed in both in vitro and in vivo experiments using brain metastatic Bm7 lung adenocarcinoma cells. On the whole, the findings of this study suggest a suppressive role for SEMA5A in lung adenocarcinoma involving the inhibition of the proliferation and migration of lung transformed cells.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Biomarcadores de Tumor/metabolismo , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Semaforinas/metabolismo , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Metilación de ADN , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Pronóstico , Semaforinas/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Vaccines (Basel) ; 8(4)2020 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-33096846

RESUMEN

Peptide vaccines are safe, and aim to elicit and expand tumor-specific immunity so as to eradicate tumors. However, achieving strong and long-lasting anti-tumor immunity with peptide vaccines for the antigen-specific treatment of cancer is challenging, in part because their efficacy depends on strong adjuvants or immunomodulators. We approached this problem by conjugating an epitope-based cancer vaccine with a lipidated sequence (an immunomodulator) to elicit a strong immune response. Lipidated and non-lipidated polyepitope proteins were generated that contained the universal T helper cell epitope (pan-DR), B cell epitopes, and the extended loop sequence of extracellular domain 2 of tumor-associated antigen L6 (TAL6). We show that the lipidated polyepitope cancer vaccine can activate bone marrow-derived dendritic cells, and trigger effective antigen-specific antibody and T helper cell responses, more effectively than the non-lipidated vaccine. Moreover, potent T cell immune responses were elicited in mice inoculated with the lipidated polyepitope cancer vaccine, providing protective antitumor immunity in mice bearing TAL6 tumors. Our study demonstrates that a lipidated polyepitope cancer vaccine could be employed to generate potent anti-tumor immune responses, including humoral and cellular immunity, which could be beneficial in the treatment of TAL6+ cancer.

19.
Redox Biol ; 34: 101571, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32446175

RESUMEN

Recurrence and metastasis remain the major cause of cancer mortality. Even for early-stage lung cancer, adjuvant chemotherapy yields merely slight increase to patient survival. EF-hand domain-containing protein D2 (EFHD2) has recently been implicated in recurrence of patients with stage I lung adenocarcinoma. In this study, we investigated the correlation between EFHD2 and chemoresistance in non-small cell lung cancer (NSCLC). High expression of EFHD2 was significantly associated with poor overall survival of NSCLC patients with chemotherapy in in silica analysis. Ectopic EFHD2 overexpression increased cisplatin resistance, whereas EFHD2 knockdown improved chemoresponse. Mechanistically, EFHD2 induced the production of NADPH oxidase 4 (NOX4) and in turn the increase of intracellular reactive oxygen species (ROS), consequently activating membrane expression of the ATP-binding cassette subfamily C member 1 (ABCC1) for drug efflux. Non-steroidal anti-inflammatory drug (NSAID) ibuprofen suppressed EFHD2 expression by leading to the proteasomal and lysosomal degradation of EFHD2 through a cyclooxygenase (COX)-independent mechanism. Combining ibuprofen with cisplatin enhanced antitumor responsiveness in a murine xenograft model in comparison with the individual treatment. In conclusion, we demonstrate that EFHD2 promotes chemoresistance through the NOX4-ROS-ABCC1 axis and therefore developing EFHD2-targeting strategies may offer a new avenue to improve adjuvant chemotherapy of lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Animales , Proteínas de Unión al Calcio , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
20.
Am J Cancer Res ; 10(8): 2621-2634, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32905506

RESUMEN

The immune checkpoint blockade therapy has emerged as encouraging treatment strategies in various cancer types. Anti-PD-L1 (programmed death-ligand 1) antibodies have been approved for triple-negative breast cancer, however the response rate yet to be optimized. It would be imperative to further understand and investigate the molecular mechanisms of PD-L1 regulation. Here, we identified glucose regulatory protein 78 (GRP78), a major endoplasmic reticulum (ER) stress responding protein, as a novel binding partner of PD-L1. GRP78 interacts with PD-L1 at the ER region and increases PD-L1 levels via regulating its stability. ER stress, triggered by different stimuli such as conventional chemotherapy, leads to the induction of PD-L1 in a GRP78-dependent manner. We showed that GRP78 modulates the response to chemotherapy, and dual-high levels of GRP78 and PD-L1 correlates with poor relapse-free survival in triple-negative breast cancer. Altogether, our study provides novel molecular insights into the regulatory mechanism of PD-L1 by revealing its interaction with GRP78, and offers a rationale to target GRP78 as a potential therapeutic strategy to enhance anti-tumor immunity.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA