Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cancer Med ; 12(17): 18032-18049, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37578396

RESUMEN

BACKGROUND & AIMS: In liver cancer, leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) compartment represents an important tumor-initiating cell (TIC) population and served as a potential therapeutic target. Cancer-associated fibroblasts (CAFs) is a critical part of the tumor microenvironment, heavily influenced TIC function and fate. However, deeply investigations have been hindered by the lack of accurate preclinical models to investigate the interaction between CAFs and TIC. Organoids model have achieved major advancements as a precious research model for recapitulating the morphological aspects of organs, and thus also serving as a candidate model to investigate the mutual interaction between different cell types. Consequently, this study aimed to construct a three-dimensional (3D) co-culture organoid model of primary LGR5-expressing tumor stem cells from primary murine liver tumors with CAFs to investigate the impact of CAFs on LGR5 marked TICs in liver cancer. MATERIALS AND METHODS: First, both of the transgenic LGR5-diphtheria toxin receptor (DTR)-GFP knock-in mice and transgenic Rosa26-mT mice developed primary liver tumors by diethylnitrosamine (DEN) administration. Tumor organoids and CAFs were generated from those primary liver cancer separately. Second, LGR5-expressing TICs organoid with CAFs were established ex vivo based on cell-cell contact or trans-well co-culture system, and the mutual influence between those two types of cells was further investigated. Subsequently, immunodeficient mouse-based xenograft model was further adopted to evaluate the influence of CAFs to LGR5 tumor stem cell, tumor formation, and metastasis. RESULTS: The co-culture organoid model composed of murine liver tumor LGR5+ tumor-initiating cells and CAFs in 3D co-culture was successfully established, with the intention to investigate their mutual interaction. The existence of CAFs upon engrafting tumor organoids resulted in dramatic higher number of LGR5+ cells in the neoplasia when compared with engrafting tumor organoids alone. Furthermore, ex vivo culture of isolated LGR5+ cells from tumors of co-engrafted mice formed significantly larger size of organoids than mono-engrafted. Our results also indicated significantly larger size and number of formed organoids, when LGR5+ cells co-cultured with CAF in both cell-cell contact and paracrine signaling in vitro, comparing to LGR5+ cells alone. Furthermore, we found that specific knockout of LGR5 expressing cells suppressed CAF-mediated promotion of tumor formation, growth, and metastasis in the experimental mice model. CONCLUSIONS: Altogether, in a 3D co-culture type of murine liver LGR5+ cells and cancer-associated fibroblasts, we have demonstrated robust effects of CAFs in the promotion of LGR5 marked liver TICs. We also further revealed the influence of tumor microenvironment on stem cell-related therapy, suggesting the possibility of combing CAF-targeted and tumor stem cell targeted therapy in treating liver cancer.

3.
Front Oncol ; 12: 1029926, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36439513

RESUMEN

Introduction: An emerging public health issue is brought on by the worldwide increase of thyroid nodules (TNs). The goal of the current study is to determine the global prevalence of TNs among the general population. Methods: We screened articles published from January 2000 to May 2022. TN prevalence was calculated with the DerSimonian-Laird random effects model with arcsine transformation. Results: A total of 20,358 entries were found in our research, and 102 of them met our inclusion criteria. A total of 9,276,178 individuals have been diagnosed as TNs; the overall prevalence was 24.83% (95% CI 21.44-28.55), regardless of the diagnostic techniques. TNs have become more prevalent during 2012-2022 (29.29%) compared with 2000-2011 (21.53%, p = 0.02). In addition, we discovered that women (36.51%) were more likely to have TNs than men (23.47%, p < 0.01). Interestingly, we found that obesity was correlated with the prevalence of TNs. Additionally, age-specific-stratified TN prevalence was found in our results. Discussion: This meta-analysis shows that, regardless of country development and economic status, TNs are spreading more widely over the world. Our findings showed a strong correlation between rising TN prevalence and older age, female sex, and elevated weight. To stop the TN epidemic from spreading over the world, increased awareness, the understanding of the disease, and quick action are required.

4.
Chin Med J (Engl) ; 135(14): 1682-1691, 2022 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-36070463

RESUMEN

BACKGROUND: The increasing burden of non-alcoholic fatty liver disease (NAFLD) worldwide imposes an emerging public health issue. We perform the current study to estimate the global prevalence, incidence, disease progression, and clinical outcomes of NAFLD. METHODS: A systematic search was conducted in Medline, Embase, Web of Science, Google Scholar, and Cochrane CENTRAL that screened articles in English language published from January 2000 to December 2021. NAFLD prevalence, incidence, rate of disease progression, and outcomes were calculated with the DerSimonian-Laird random effects model with arcsine transformation. RESULTS: Our search identified 59,156 records, of which 578 studies fulfilled our inclusion criteria. The overall prevalence of NAFLD was 29.38% (95% confidence interval [CI] 28.09-30.69) regardless of the diagnostic techniques. Looking at the group in which the diagnosis was made by ultrasound exclusively, the pooled prevalence was 30.49% (95% CI 29.55-31.43). NAFLD has become more prevalent during the year 2011-2021 (31.63%, 95% CI 30.23-33.04) compared with year 2000-2010 (27.94%, 95% CI 26.23-29.69). The pooled estimation of non-alcoholic steatohepatitis prevalence was 8.26% (95% CI 1.13-21.01), 46.49% (95% CI 35.93-57.20), and 46.72% (95% CI 37.57-55.98) in general population, NAFLD patients, and severe/morbidly obese patients, respectively. Based on a total of 110,142 newly developed NAFLD patients, the pooled incident rate was estimated as 46.24 cases per 1000 person-years (95% CI 43.21-49.30). In patients with NAFLD, the incident rate of hepatocellular carcinoma was 1.46 (95% CI 0.90-2.03) cases per 1000 person-years. The overall pooled estimate of NAFLD related mortality was 23.91 (95% CI 13.55-37.18) death per 1000 person-years. CONCLUSIONS: The prevalence of NAFLD is increasing globally. It is contributing to poor clinical outcomes including hepatocellular carcinoma and death. Rising awareness and urgent actions are warranted to control the NAFLD pandemic across the globe. REGISTRATION: PROSPERO, No. CRD42020171104.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Enfermedad del Hígado Graso no Alcohólico , Obesidad Mórbida , Progresión de la Enfermedad , Humanos , Incidencia , Enfermedad del Hígado Graso no Alcohólico/diagnóstico , Prevalencia
5.
Cancer Sci ; 113(11): 3722-3734, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36087034

RESUMEN

Enhanced fatty acid synthesis provides proliferation and survival advantages for tumor cells. Apelin is an adipokine, which serves as a ligand of G protein-coupled receptors that promote tumor growth in malignant cancers. Here, we confirmed that apelin increased sterol regulatory element-binding protein 1 (SREBP1) activity and induced the expression of glutamine amidotransferase for deamidating high-mobility group A 1 (HMGA1) to promote fatty acid synthesis and proliferation of lung cancer cells. This post-translational modification stabilized the HMGA1 expression and enhanced the formation of the apelin-HMGA1-SREBP1 complex to facilitate SREBP1 activity for lipid metabolism and lung cancer cell growth. We uncovered the pivotal role of apelin-mediated deamidation of HMGA1 in lipid metabolism and tumorigenesis of lung cancer cells.


Asunto(s)
Proteína HMGA1a , Neoplasias Pulmonares , Humanos , Apelina , Carcinogénesis/genética , Línea Celular Tumoral , Transformación Celular Neoplásica , Ácidos Grasos , Proteína HMGA1a/genética , Lípidos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo
6.
Signal Transduct Target Ther ; 7(1): 289, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36008393

RESUMEN

A thorough interrogation of the immune landscape is crucial for immunotherapy strategy selection and prediction of clinical responses in non-small-cell lung cancer (NSCLC) patients. Single-cell RNA sequencing (scRNA-seq) techniques have prompted the opportunity to dissect the distinct immune signatures between lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), the two major subtypes of NSCLC. Here, we performed scRNA-seq on 72,475 immune cells from 40 samples of tumor and matched adjacent normal tissues spanning 19 NSCLC patients, and drew a systematic immune cell transcriptome atlas. Joint analyses of the distinct cellular compositions, differentially expressed genes (DEGs), cell-cell interactions, pseudotime trajectory, transcriptomic factors and prognostic factors based on The Cancer Genome Atlas (TCGA), revealed the central roles of cytotoxic and effector T and NK cells and the distinct functional macrophages (Mφ) subtypes in the immune microenvironment heterogeneity between LUAD and LUSC. The dominant subtype of Mφ was FABP4-Mφ in LUAD and SPP1-Mφ in LUSC. Importantly, we identified a novel lymphocyte-related Mφ cluster, which we named SELENOP-Mφ, and further established its antitumor role in both types, especially in LUAD. Our comprehensive depiction of the immune heterogeneity and definition of Mφ clusters could help design personalized treatment for lung cancer patients in clinical practice.


Asunto(s)
Adenocarcinoma del Pulmón , Carcinoma de Pulmón de Células no Pequeñas , Carcinoma de Células Escamosas , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Análisis de Secuencia de ARN , Microambiente Tumoral/genética
7.
Front Immunol ; 13: 854724, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35874785

RESUMEN

Understanding immune cell phenotypes in the tumor microenvironment (TME) is essential for explaining and predicting progression of non-small cell lung cancer (NSCLC) and its response to immunotherapy. Here we describe the single-cell transcriptomics of CD45+ immune cells from tumors, normal tissues and blood of NSCLC patients. We identified three clusters of immune cells exerting immunosuppressive effects: CD8+ T cells with exhausted phenotype, tumor-associated macrophages (TAMs) with a pro-inflammatory M2 phenotype, and regulatory B cells (B regs) with tumor-promoting characteristics. We identified genes that may be mediating T cell phenotypes, including the transcription factors ONECUT2 and ETV4 in exhausted CD8+ T cells, TIGIT and CTL4 high expression in regulatory T cells. Our results highlight the heterogeneity of CD45+ immune cells in the TME and provide testable hypotheses about the cell types and genes that define the TME.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Linfocitos T CD8-positivos , Proteínas de Homeodominio/genética , Humanos , Factores de Transcripción/genética , Transcriptoma , Microambiente Tumoral/genética
8.
Nat Commun ; 13(1): 4308, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35879297

RESUMEN

Cancer associated fibroblasts (CAFs) support tumors via multiple mechanisms, including maintaining the immunosuppressive tumor microenvironment and limiting infiltration of immune cells. The prolyl isomerase Pin1, whose overexpression in CAFs has not been fully profiled yet, plays critical roles in tumor initiation and progression. To decipher effects of selective Pin1 inhibition in CAFs on pancreatic cancer, here we formulate a DNA-barcoded micellular system (DMS) encapsulating the Pin1 inhibitor AG17724. DMS functionalized with CAF-targeting anti-FAP-α antibodies (antiCAFs-DMS) can selectively inhibit Pin1 in CAFs, leading to efficacious but transient tumor growth inhibition. We further integrate DNA aptamers (AptT), which can engage CD8+ T lymphocytes, to obtain a bispecific antiCAFs-DMS-AptT system. AntiCAFs-DMS-AptT inhibits tumor growth in subcutaneous and orthotopic pancreatic cancer models.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias Pancreáticas , Fibroblastos Asociados al Cáncer/patología , Línea Celular Tumoral , Fibroblastos/patología , Humanos , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Microambiente Tumoral , Neoplasias Pancreáticas
9.
Bioeng Transl Med ; 7(1): e10263, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35111955

RESUMEN

Besides its well-known benefits on human health, calcitriol, the hormonally active form of vitamin D3, has been being evaluated in clinical trials as an anticancer agent. However, currently available results are contradictory and not fundamentally deciphered. To the best of our knowledge, hypercalcemia caused by high-dose calcitriol administration and its low bioavailability limit its anticancer investigations and translations. Here, we show that the one-step self-assembly of calcitriol and amphiphilic cholesterol-based conjugates leads to the formation of a stable minimalist micellar nanosystem. When administered to mice, this nanosystem demonstrates high calcitriol doses in breast tumor cells, significant tumor growth inhibition and antimetastasis capability, as well as good biocompatibility. We further reveal that the underlying molecular antimetastatic mechanisms involve downregulation of proteins facilitating metastasis and upregulation of paxillin, the key protein of focal adhesion, in primary tumors.

10.
J Cell Mol Med ; 26(7): 1918-1931, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35122390

RESUMEN

Lymphocytes and neutrophils are involved in the immune response against cancer. This study aimed to investigate the relationship between lymphocyte percentage/neutrophil percentage and the clinical characteristics of lung cancer patients, and to explore whether they could act as valuable predictors to ameliorate lung cancer prognosis. A total of 1312 patients were eligible to be recruited. Lymphocyte percentage and neutrophil percentage were classified based on their reference ranges. Survival curves were determined using Kaplan-Meier method, and univariate and multivariate cox regression analyses were performed to identify the significant predictors. Decision curve analysis was used to evaluate the clinical benefit. The results of both training and validation cohorts indicated that lymphocyte percentage exhibited high correlation with clinical characteristics and metastasis of lung cancer patients. Both lymphocyte percentage and neutrophil percentage were closely associated with survival status (all p < 0.0001). Low lymphocyte percentage could act as an indicator of poor prognosis; it offered a higher clinical benefit when combined with the clinical characteristic model. Our findings suggested that pretreatment lymphocyte percentage served as a reliable predictor of lung cancer prognosis, and it was also an accurate response indicator in lung adenocarcinoma and advanced lung cancer. Measurement of lymphocyte percentage improved the clinical utility of patient characteristics in predicting mortality of lung cancer patients.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/patología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Recuento de Linfocitos , Linfocitos/patología , Neutrófilos/patología , Curva ROC , Estudios Retrospectivos
11.
Signal Transduct Target Ther ; 7(1): 9, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35027529

RESUMEN

Lung adenocarcinoma (LUAD) and squamous carcinoma (LUSC) are two major subtypes of non-small cell lung cancer with distinct pathologic features and treatment paradigms. The heterogeneity can be attributed to genetic, transcriptional, and epigenetic parameters. Here, we established a multi-omics atlas, integrating 52 single-cell RNA sequencing and 2342 public bulk RNA sequencing. We investigated their differences in genetic amplification, cellular compositions, and expression modules. We revealed that LUAD and LUSC contained amplifications occurring selectively in subclusters of AT2 and basal cells, and had distinct cellular composition modules associated with poor survival of lung cancer. Malignant and stage-specific gene analyses further uncovered critical transcription factors and genes in tumor progression. Moreover, we identified subclusters with proliferating and differentiating properties in AT2 and basal cells. Overexpression assays of ten genes, including sub-cluster markers AQP5 and KPNA2, further indicated their functional roles, providing potential targets for early diagnosis and treatment in lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón , Biomarcadores de Tumor , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Transcripción Genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo
12.
Front Pharmacol ; 13: 1109822, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36712673

RESUMEN

Background: Manoalide (MA), a proven natural inhibitor of PLA2 has anticancer effects, but its potential application and mechanism as an anticancer drug to promote EGFR-TKI sensitivity in lung cancer cells have not been studied. Methods: KRAS-mutated lung cancer cells and organoids, acquired osimertinib-resistant lung cancer cell lines HCC827OR, were used as EGFR-TKI-resistant models. CCK-8, clone formation, apoptosis assays, and calcein-AM staining were performed to investigate the inhibitory effects of MA in lung cancer cells and organoids. The flow cytometry or confocal microscope was used to detect lipid droplets, ROS, lipid peroxidation, mitochondria Ca2+, and iron content. The oxygen consumption rate (OCR) and fatty acid oxidation (FAO) were used to estimate the effect of MA on mitochondrial function. Results: MA inhibits the proliferation of KRAS-mutated lung cancer cells and organoids. In addition, MA induces ER stress in a ROS-dependent mechanism. The ROS induced by MA is mainly in mitochondrial and causes lipid peroxidation, thereby inhibiting mitochondrial FAO metabolism and promoting the accumulation of lipid droplets. MA also suppresses the KRAS-ERK pathway through ROS and promotes the sensitivity of KRAS-mutated lung cancer cells and organoids to osimertinib. Furthermore, MA induces ferroptosis by suppressing the NRF2-SLC7A11 axis and mitochondrial Ca2+ overload induced-FTH1 pathways to promote the sensitivity of osimertinib-resistant lung cancer cells to osimertinib. Conclusions: MA is a candidate EGFR-TKI sensitizer in KRAS-mutated and osimertinib-resistant lung cancer cells.

13.
Nat Commun ; 12(1): 6500, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764257

RESUMEN

Lung adenocarcinomas (LUAD) arise from precancerous lesions such as atypical adenomatous hyperplasia, which progress into adenocarcinoma in situ and minimally invasive adenocarcinoma, then finally into invasive adenocarcinoma. The cellular heterogeneity and molecular events underlying this stepwise progression remain unclear. In this study, we perform single-cell RNA sequencing of 268,471 cells collected from 25 patients in four histologic stages of LUAD and compare them to normal cell types. We detect a group of cells closely resembling alveolar type 2 cells (AT2) that emerged during atypical adenomatous hyperplasia and whose transcriptional profile began to diverge from that of AT2 cells as LUAD progressed, taking on feature characteristic of stem-like cells. We identify genes related to energy metabolism and ribosome synthesis that are upregulated in early stages of LUAD and may promote progression. MDK and TIMP1 could be potential biomarkers for understanding LUAD pathogenesis. Our work shed light on the underlying transcriptional signatures of distinct histologic stages of LUAD progression and our findings may facilitate early diagnosis.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Neoplasias Pulmonares/metabolismo , Ribosomas/metabolismo , Adenocarcinoma del Pulmón/genética , Linaje de la Célula , Metabolismo Energético/genética , Metabolismo Energético/fisiología , Humanos , Neoplasias Pulmonares/genética , Midkina/genética , Midkina/metabolismo , Ribosomas/genética , Inhibidor Tisular de Metaloproteinasa-1/genética , Inhibidor Tisular de Metaloproteinasa-1/metabolismo
14.
Cell Rep ; 36(10): 109660, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34496260

RESUMEN

Non-small cell lung carcinoma (NSCLC), the most common form of lung cancer, is the leading cause of cancer-related death worldwide. We perform whole-genome sequencing (WGS) on samples from 43 primary patients with NSCLC and matched normal samples and analyze their matched open chromatin data and transcriptome data. Our results indicate that next-generation sequencing (NGS) and the Bionano Genomics (BNG) platform should be viewed as complementary technologies in terms of structural variations detection. By creating a framework integrating these two platforms, we detect high-technical-confidence somatic structural variations (SVs) in NSCLC cases, which could aid in the efficient investigation of new candidate oncogenes, such as TRIO and SESTD1. Our findings highlight the impact of somatic SVs on NSCLC oncogenesis and lay a foundation for exploring associations among somatic SVs, gene expression, and regulatory networks in patients with NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Genoma Humano/genética , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mutación/genética , Oncogenes/genética , Secuenciación Completa del Genoma/métodos
15.
Carcinogenesis ; 42(2): 243-253, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32808032

RESUMEN

Apelin acts as a tumor promoter in multiple malignant tumors; however, its regulatory mechanism remains unclear. Previous studies have indicated that exosomes are pivotal to mediating tumor progression and metastasis. This study examined whether apelin enhances proliferation and invasion ability of lung cancer cells via exosomal microRNA (miRNA). Lung cancer A549 cells overexpressing apelin and control vector were generated by lentiviral transfection. Exosomes were isolated from the culture supernatant of each cell group and characterized. A-exo and V-exo were, respectively, cocultured with A549 cells, and assays of proliferation, apoptosis, colony formation and invasion were conducted. Exosomal miRNA sequencing (miRNA-seq) was performed on A-exo and V-exo to select a candidate miRNA. It was found that A549 cells absorbed more A-exo than V-exo, and A-exo could promote proliferation, colony formation, migration and invasion of A549 cells more than V-exo. Exosomal miRNA-seq data revealed that miR-15a-5p was markedly lower in A-exo compared with V-exo. Low expression of miR-15a-5p was also found in lung cancer tissues and cell lines, suggesting that miR-15a-5p may have an anti-tumor role. Overexpression of miR-15a-5p in A549 cells was associated with less cell proliferation, migration, invasion and suppressed cell cycle, and lower amounts of CDCA4 (cell division cycle-associated protein 4) indicated that it may be a potential target for miR-15a-5p. This study elucidated a novel regulatory mechanism that apelin may promote proliferation and invasion of lung cancer cells by inhibiting miR-15a-5p encapsulated in exosomes.


Asunto(s)
Apelina/metabolismo , Proteínas de Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , MicroARNs/antagonistas & inhibidores , Células A549 , Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular/genética , Técnicas de Cocultivo , Regulación hacia Abajo , Exosomas/metabolismo , Humanos , Neoplasias Pulmonares/patología , MicroARNs/agonistas , MicroARNs/metabolismo , RNA-Seq
16.
Free Radic Biol Med ; 161: 150-162, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33059020

RESUMEN

Metabolic reprogramme was a key characteristic of malignant tumors. Increased evidences indicated that besides Warburg effect (abnormal glucose metabolism), abnormal lipid metabolism played more and more important in progression and metastasis of malignant tumors. MiR-15a-5p could inhibit development of lung cancer, while its regulating mechanism, especially the role in lipid metabolism still remained unclear. In this study, we confirmed that miR-15a-5p inhibited proliferation, migration and invasion of lung cancer cells. The online analysis of Mirpath v.3 predicted that miR-15a-5p was closely associated with fatty acid synthesis and lipid metabolism. In vitro cell experiments revealed that miR-15a-5p significantly suppressed fatty acid synthesis of lung cancer cells by inhibiting acetate uptake. Extensive analysis indicated that miR-15a-5p could suppress acetyl-CoA activity and decrease histone H4 acetylation by inhibiting ACSS2 expression. In addition, we also observed that ACSS2 located in nucleus under hypoxic conditions, while miR-15a-5p could be transported into nucleus to inhibit the function of ACSS2. Our study unveiled a novel mechanism of miR-15a-5p in inhibiting metastasis of lung cancer cells by suppressing lipid metabolism via suppression of ACSS2 mediated acetyl-CoA activity and histone acetylation.


Asunto(s)
Histonas , Metabolismo de los Lípidos , Neoplasias Pulmonares , MicroARNs , Acetato CoA Ligasa/metabolismo , Acetilación , Histonas/genética , Histonas/metabolismo , Humanos , Metabolismo de los Lípidos/genética , Neoplasias Pulmonares/genética
17.
Eur J Cancer Prev ; 29(4): 315-320, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31764214

RESUMEN

This study aimed to evaluate the diagnostic efficacy of seven autoantibodies in all lung cancer, lung adenocarcinoma, lung squamous cell carcinoma and early-stage lung cancer patients. ELISA testing of a seven autoantibody panel was performed on 386 lung cancer patients and 238 normal controls. The sensitivity and specificity of each autoantibody were analyzed using the receiver operating characteristic curve analysis. The diagnostic efficacy of a combination of these seven autoantibodies was evaluated by binary logistic regression. The results indicated that six of the seven autoantibodies (p53, SOX2, GAGE7, GBU4-5, MAGEA1 and CAGE) had high specificity and low sensitivity, while PGP9.5 had high sensitivity and low specificity. Further analysis showed that all seven autoantibodies had better diagnostic value in lung squamous cell carcinoma patients when compared to lung adenocarcinoma or all lung cancer patients. Logistic regression showed that a combination of the seven autoantibodies resulted in more reliable detection of lung cancer than any individual autoantibody in early-stage lung cancer (sensitivity/specificity: 47.8%/81.4%, areas under the curve: 0.764, 95% confidence interval: 0.718-0.811). Additionally, this panel had a better sensitivity of 56.5% for detection of lung squamous cell carcinoma than for all lung cancer (50.1%) or adenocarcinoma (51.7%) (P < 0.05). Our results indicated that the seven autoantibody panel could be used for early lung cancer detection, and it had better sensitivity in diagnosis of lung squamous cell carcinoma.


Asunto(s)
Adenocarcinoma del Pulmón/diagnóstico , Autoanticuerpos/sangre , Biomarcadores de Tumor/sangre , Carcinoma de Células Escamosas/diagnóstico , Detección Precoz del Cáncer/métodos , Neoplasias Pulmonares/diagnóstico , Adenocarcinoma del Pulmón/sangre , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/patología , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Autoanticuerpos/inmunología , Biomarcadores de Tumor/inmunología , Biopsia , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Estudios de Casos y Controles , Femenino , Humanos , Pulmón/patología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Curva ROC
18.
J Cell Biochem ; 119(7): 6104-6112, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29575175

RESUMEN

Natural products are a great source of cancer chemotherapeutic agents. In the present study, the anticancer effects of cucurbitacin I on A549 cells were investigated. Cucurbitacin I decreased cell viability, inhibited colony formation, and induced apoptosis in A549 cells. Cucurbitacin I caused accumulation of autophagosome and dose-dependent expression of LC3II protein. Autophagy inhibitors 3-methyladenine (3-MA) inhibited autophagy induced by cucurbitacin I and relieved cucurbitacin I-triggered cell death and apoptosis in A549 Cells. Cucurbitacin I treatment inhibits the ERK activation and the downstream phosphorylation level of mTOR and STAT3, but not the PI3K/Akt pathway. Furthermore, treatment with the mTOR activator MHY-1485, which also suppressed cucurbitacin I-induced LC3II expression, and also reversed cucurbitacin I-induced cell death and apoptosis. Taken together, these results suggest that cucurbitacin I induced pro-death autophagy through ERK/mTOR/STAT3 signaling cascade in A549 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Pulmonares/patología , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Células A549 , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
19.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 33(1): 33-38, 2017 Jan.
Artículo en Chino | MEDLINE | ID: mdl-28031111

RESUMEN

Objective To study the mechanism underlying cucurbitacin I (JSI-124) inducing cell apoptosis in human hepatoma HepG2 cells. Methods HepG2 cells were exposed to 0.01, 0.10, 1.00 and 10.00 µmol/L JSI-124 for 24, 48 and 72 hours. Cell proliferation was detected by CCK-8 assay; the nuclear morphological changes were observed by Hoechst 33258 staining; the formation of tumor cell colonies was visualized by violet staining; and cell apoptosis was detected by annexin V-FITC/PI double staining combined with flow cytometry. Furthermore, the mRNA expression levels of p53 and its downstream Bax, Fas and MDM2 genes were measured by quantitative real-time PCR, and the protein levels of P53 and activated caspase-3 were evaluated by Western blotting. Results JSI-124 inhibited the proliferation and induced Hoechst 33258-stained chromatin condensation in HepG2 cells in a concentration- and time-dependent manner. Flow cytometry revealed that 1.00 µmol/L JSI-124 treatment increased the apoptotic rate significantly in HepG2 cells compared with the control cells. Furthermore, JSI-124 significantly enhanced the mRNA expressions of p53 and its downstream apoptotic factors, including Bax and Fas, but did not change the gene expression of the p53 tumor suppressor, MDM2. The 48-hour treatment of JSI-124 in HepG2 cells significantly increased the levels of p53 and cleaved caspase-3 proteins. Conclusion JSI-124 induces the apoptosis of HepG2 cells through the activation of p53 and its downstream pro-apoptotic factors.


Asunto(s)
Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Células Hep G2/efectos de los fármacos , Humanos
20.
Appl Biochem Biotechnol ; 172(7): 3593-605, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24557956

RESUMEN

Glutamate dehydrogenase (GDH) from Bacillus subtilis natto was purified to apparent homogeneity by ammonium sulfate precipitation, ion-exchange chromatography, size exclusion chromatography, and hydroxyapatite (HA) affinity chromatography. The GDH was purified 34-fold, with a yield of 41 % of total activity and a specific activity of 34.29 U/mg proteins. The molecular weight (Mr) of was measured at 47 kDa with sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and 264 kDa with high-performance liquid chromatography (HPLC). The optimum pH and temperature for the deammoniation reaction were measured to be 7.5 and 30 °C, respectively. The active-site amino acid residues of GDH were investigated by chemical modification. The compounds 2,4,6-trinitrobenzenesulfonic acid (TNBS), phenylglyoxal (PG), and phenylmethanesulfonyl fluoride (PMSF) were used to modify lysine, arginine, and serine active site residues, respectively. After treatment with modifying reagents at concentrations of 1 mM, GDH activity fell to 10.7 % with TNBS, 83.3 % with PG, and 12.8 % with PMSF. However, with substrate protection, there was almost no loss in GDH activity following treatment with any modifying reagent. The kinetic parameters K m and V max were determined in each case. K m values for native GDH, 50 % TNBS-inactivated GDH, and 50 % PMSF-inactivated GDH were 0.037, 0.104, and 0.017 mM, respectively. V max values were 0.048, 0.022, and 0.031 mM/s, respectively. These results suggest that the active site contains one or more lysine residues that play a role in substrate binding and one or more serine residues that may maintain the enzyme conformation. However, arginine residues played less of a role in the activity of GDH.


Asunto(s)
Bacillus subtilis/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/aislamiento & purificación , Glutamato Deshidrogenasa/química , Glutamato Deshidrogenasa/aislamiento & purificación , Bacillus subtilis/química , Proteínas Bacterianas/metabolismo , Estabilidad de Enzimas , Glutamato Deshidrogenasa/metabolismo , Cinética , Conformación Proteica , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...