Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
World J Surg Oncol ; 20(1): 231, 2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35820925

RESUMO

BACKGROUND: Bladder cancer is one of the most lethal malignancy in urological system, and 20-25% of bladder cancer patients are muscle invasive with unfavorable prognosis. However, the role of alternative splicing (AS) in muscle-invasive bladder cancer (MIBC) remains to be elucidated. METHODS: Percent spliced in (PSI) data obtained from the Cancer Genome Atlas (TCGA) SpliceSeq database (n = 394) were utilized to evaluate the AS events in MIBC. Prognosis-associated AS events were screened out by univariate Cox regression. LASSO Cox regression was used to identify reliable prognostic patterns in a training set and further validated in a test set. Splicing regulatory networks were constructed by correlations between PSI of AS events and RNA expression of splicing factors. RESULTS: As a result, a total of 2589 prognosis-related AS events in MIBC were identified. Pathways of spliceosomal complex (FDR = 0.017), DNA-directed RNA polymerase II, core complex (FDR = 0.032), and base excision repair (FDR = 0.038) were observed to be significantly enriched. Additionally, we noticed that most of the prognosis-related AS events were favorable factors. According to the LASSO and multivariate Cox regression analyses, 15-AS-based signature was established with the area under curve (AUC) of 0.709, 0.823, and 0.857 at 1-, 3-, and 5- years, respectively. The MIBC patients were further divided into high- and low-risk groups based on median risk sores. Interestingly, we observed that the prevalence of FGFR3 with mutations and focal amplification was significantly higher in low-risk group. Functional and immune infiltration analysis suggested potential signaling pathways and distinct immune states between these two groups. Moreover, splicing correlation network displayed a regulatory mode of prognostic splicing factors (SF) in MIBC patients. CONCLUSIONS: This study not only provided novel insights into deciphering the possible mechanism of tumorgenesis and pathogenesis but also help refine risk stratification systems and potential treatment of decision-making for MIBC.


Assuntos
Processamento Alternativo , Neoplasias da Bexiga Urinária , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Músculos , Prognóstico , Fatores de Processamento de RNA/genética , Neoplasias da Bexiga Urinária/genética
2.
Biomed Eng Online ; 20(1): 67, 2021 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-34217312

RESUMO

Hepatocellular carcinoma (HCC) is currently the sixth most common malignancy and the second major cause of tumor-related deaths in the world. This study aimed to investigate the role of cleavage and polyadenylation factor-6 (CPSF6) and B-cell translocation gene 2 (BTG2) in regulating the glycolysis and apoptosis in HCC cells. The RNA and protein expression of CPSF6 and BTG2 in normal hepatocyte and HCC were, respectively, detected by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) analysis and Western blot analysis. The viability and apoptosis of transfected Huh-7 cells were, respectively, analyzed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and terminal deoxynucleotidyl transferase (TdT) dUTP Nick-End Labeling (TUNEL) assay. The expression of apoptosis-related proteins and HK-2 in transfected Huh-7 cells was also detected by Western blot analysis. The levels of glucose and lactate in the culture supernatant of transfected Huh-7 cells were, respectively, detected with the glucose assay kit and lactate assay kit. The interaction of CPSF6 and BTG2 was confirmed by RNA binding protein immunoprecipitation (RIP) assay. As a result, CPSF6 expression was increased while BTG2 expression was decreased in Huh-7 cells. Interference with CPSF6 suppressed the viability and glycolysis, and promoted the apoptosis of Huh-7 cells. Furthermore, CPSF6 interacted with BTG2 and interference with CPSF6 upregulated the BTG2 expression and inhibited the protein kinase B (AKT)/extracellular signal-regulated kinase (ERK)/nuclear factor (NF)-κB pathway. Interference with BTG2 could partially reverse the above cell changes caused by interference with CPSF6. In conclusion, CPSF6 inhibited the BTG2 expression to promote glycolysis and suppress apoptosis in HCC cells by activating AKT/ERK/NF-κB pathway.


Assuntos
Carcinoma Hepatocelular , Proteínas Imediatamente Precoces , Neoplasias Hepáticas , Apoptose , Carcinoma Hepatocelular/genética , Proteínas de Transporte , Linhagem Celular Tumoral , Proliferação de Células , Glicólise , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Neoplasias Hepáticas/genética , RNA , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Fatores de Poliadenilação e Clivagem de mRNA
3.
BMC Complement Altern Med ; 14: 377, 2014 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-25283233

RESUMO

BACKGROUND: Lung cancers are leading causes of cancer death, and Rumex japonicus has been traditionally used in folk medicine as anti-microorganic, anti-inflammatory and anti-tumor agents. This study was designed to investigate the anti-proliferative activity of physcion 8-O-ß-glucopyranoside (PG) isolated from Rumex japonicus Houtt. on A549 cell lines. METHODS: In our present study, PG was isolated and identified from the ethanol extracts of R. japonicus. MTT method was used to evaluate the anti-proliferative activity of PG on A549 cell lines, and cell cycle distribution assay, apoptosis assay, and western blot analysis in vitro were used to explore the possible mechanisms. RESULTS: From the results of our present study, cell viability was obviously inhibited by PG, in a dose- and time-dependent manner. Our results also suggested that the anti-proliferative effect of PG was related to cell cycle arrest at the G2/M phase through repression of cdc2 and Cyclin B1 protein expression. In addition, the results of apoptosis assay and western blot analysis indicated that the anti-proliferative activity could be related to apoptosis via up-regulating the expressions of Bax, caspase-3 and caspase-7, and down-regulating the expressions of Bcl-2. CONCLUSIONS: In conclusion, the PG has significant anti-proliferative activity on A549 cell lines, and the possible mechanism was related to cell cycle arrest at the G2/M phase, and apoptosis via the regulations of Bax, Bcl-2, and caspase-3 and caspase-7.


Assuntos
Antineoplásicos/farmacologia , Emodina/análogos & derivados , Glicosídeos/farmacologia , Extratos Vegetais/farmacologia , Rumex/química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Emodina/química , Emodina/farmacologia , Glicosídeos/química , Humanos , Extratos Vegetais/química
4.
Molecules ; 19(2): 1592-602, 2014 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-24473215

RESUMO

Resveratrol-4-O-D-(2'-galloyl)-glucopyranoside (RESG) is one of the active compounds isolated from Polygonum cuspidatum. The purpose of our present study was to investigate the anti-hepatocellular carcinoma effect of RESG in vitro and in vivo, and the possible mechanisms in vitro. In vitro, our results showed that RESG could significantly inhibit the human hepatocellular carcinoma viability in the MTT assay, in a dose- and time-dependent manner. Furthermore, our results demonstrated that RESG could induce SMMC-7721 cell apoptosis and activate caspases 3 and caspases 9 by using Annexin V-FITC staining and western blot, respectively. In vivo, RESG also showed efficacy in SMMC-7721 xenograft model in nude mice, and further molecule mechanisms were investigated in vitro. The results showed that RESG up-regulated the p-JNK expressions, whereas it down-regulated the p-ERK expressions. Above results demonstrated that RESG is a potential therapeutic agent for hepatocellular carcinoma via JNK and ERK pathway to induce apoptosis. Our finding provided a basis for further development of RESG as an anticancer agent.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Glucuronídeos/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Extratos Vegetais/farmacologia , Estilbenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fallopia japonica/química , Glucuronídeos/química , Humanos , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Extratos Vegetais/química , Estilbenos/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
MedComm (2020) ; 5(2): e482, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38344399

RESUMO

Metastasis is the leading cause of death in hepatocellular carcinoma (HCC) patients, and autophagy plays a crucial role in this process by orchestrating epithelial-mesenchymal transition (EMT). Stromal interaction molecule 1 (STIM1), a central regulator of store-operated calcium entry (SOCE) in nonexcitable cells, is involved in the development and spread of HCC. However, the impact of STIM1 on autophagy regulation during HCC metastasis remains unclear. Here, we demonstrate that STIM1 is temporally regulated during autophagy-induced EMT in HCC cells, and knocking out (KO) STIM1 significantly reduces both autophagy and EMT. Interestingly, STIM1 enhances autophagy through both SOCE-dependent and independent pathways. Mechanistically, STIM1 directly interacts with microtubule-associated protein 1A/1B-light chain 3B (LC3B) to form a complex via the sterile-α motif (SAM) domain, which promotes autophagosome formation. Furthermore, deletion of the SAM domain of STIM1 abolishes its binding with LC3B, leading to a decrease in autophagy and EMT in HCC cells. These findings unveil a novel mechanism by which the STIM1/LC3B complex mediates autophagy and EMT in HCC cells, highlighting a potential target for preventing HCC metastasis.

6.
Front Oncol ; 12: 967675, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36523965

RESUMO

Introduction: Traditional therapeutic approaches for the treatment of advanced non-small-cell lung cancer (NSCLC) are based on chemotherapy. However, the discovery and understanding of oncogenic driver alterations has led to the development of targeted therapies that have substantially improved patient outcomes. Still, to date, there have been no reports of patients with advanced anaplastic lymphoma kinase (ALK)-positive lung cancer achieving clinical complete response (cCR) in the systemic lesion and pathological complete remission (pCR) in primary lung lesion after multiple lines of conversion therapy. Methods: In this case, a 55-year-old man was diagnosed with ALK-positive, stage IV lung adenocarcinoma using immunohistochemistry and next generation sequencing (NGS) tests. Results: Crizotinib and two other ATP-competitive ALK inhibitors, ceritinib and alectinib, were used respectively as first-line, second-line, and third-line therapy. The patient received treatment with crizotinib and achieved partial response (PR), but 5 months later the efficacy was evaluated as progressive disease (PD). Ceritinib was used as the second-line treatment, but the disease progressed 6 months later. Alectinib was used as the third-line treatment, but the efficacy was evaluated as PD. From April 2019 to November 2019, the patient received 4 cycles of induction chemotherapy with pemetrexed/carboplatin/bevacizumab and then switched to pemetrexed/bevacizumab as the fourth-line treatment, and received the fifth line treatment, cetuximab/paclitaxel liposome/nedaplatin, for 1 cycle, but the disease still progressed. Then the patient received the sixth line of treatment, camrelizumab/lorlatinib, for 9 antitumor cycles, resulting in PR. The patient underwent surgery followed by maintenance treatment with lorlatinib and achieved cCR. To our knowledge, this is the first documented case of cCR in a patient with ALK-positive advanced lung adenocarcinoma treated with multiple lines of therapy followed by surgical treatment. Discussion: This case reveals the possible survival benefit of immunotherapy after multiple line treatment in ALK-positive advanced lung adenocarcinoma, indicating that it is possible find new therapeutic targets based on NGS molecular detection and provide precise therapeutic strategies for clinical practice when drug resistance or progression occurs in cancer therapy.

7.
Int J Gen Med ; 14: 10423-10428, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35002298

RESUMO

BACKGROUND: The role of circRNA circ_0071662 has been studied in bladder cancer. The present study aimed to analyze its involvement in esophageal squamous cell carcinoma (ESCC). METHODS: Patients with ESCC (n = 66), esophageal ulcer (EU, n = 66), or gastroesophageal reflux disease (GERD, n = 66) and healthy controls (n = 66) were enrolled in this study. Plasma samples were collected from all patients and controls. ESCC and paired non-tumor tissue samples were collected from ESCC patients. Circ_0071662 levels in these samples were determined by RT-qPCRs. Diagnostic and prognostic values of circ_0071662 for ESCC were analyzed with ROC curve and survival curve analyses. RESULTS: Circ_0071662 level was decreased in ESCC, but not in GERN and EU compared to the controls and in ESCC tissues compared to the non-tumor tissues. Plasma circ_0071662 was closely correlated with patients' tumor size but not with other clinical features. Decreased plasma circ_0071662 levels separated ESCC patients from GERN patients, EU patients, and healthy controls. Low plasma circ_0071662 levels were closely correlated with worse survival outcomes of ESCC patients. CONCLUSION: Circ_0071662 is lowly expressed in ESCC and may serve as a potential diagnostic and prognostic biomarker for ESCC.

8.
Theranostics ; 11(10): 5045-5060, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754043

RESUMO

Background & Aims: Liver cancer stem cells (LCSCs) mediate therapeutic resistance and correlate with poor outcomes in patients with hepatocellular carcinoma (HCC). Fibroblast growth factor (FGF)-19 is a crucial oncogenic driver gene in HCC and correlates with poor prognosis. However, whether FGF19 signaling regulates the self-renewal of LCSCs is unknown. Methods: LCSCs were enriched by serum-free suspension. Self-renewal of LCSCs were characterized by sphere formation assay, clonogenicity assay, sorafenib resistance assay and tumorigenic potential assays. Ca2+ image was employed to determine the intracellular concentration of Ca2+. Gain- and loss-of function studies were applied to explore the role of FGF19 signaling in the self-renewal of LCSCs. Results: FGF19 was up-regulated in LCSCs, and positively correlated with certain self-renewal related genes in HCC. Silencing FGF19 suppressed self-renewal of LCSCs, whereas overexpressing FGF19 facilitated CSCs-like properties via activation of FGF receptor (FGFR)-4 in none-LCSCs. Mechanistically, FGF19/FGFR4 signaling stimulated store-operated Ca2+ entry (SOCE) through both the PLCγ and ERK1/2 pathways. Subsequently, SOCE-calcineurin signaling promoted the activation and translocation of nuclear factors of activated T cells (NFAT)-c2, which transcriptionally activated the expression of stemness-related genes (e.g., NANOG, OCT4 and SOX2), as well as FGF19. Furthermore, blockade of FGF19/FGFR4-NFATc2 signaling observably suppressed the self-renewal of LCSCs. Conclusions: FGF19/FGFR4 axis promotes the self-renewal of LCSCs via activating SOCE/NFATc2 pathway; in turn, NFATc2 transcriptionally activates FGF19 expression. Targeting this signaling circuit represents a potential strategy for improving the therapeutic efficacy of HCC.


Assuntos
Sinalização do Cálcio/genética , Carcinoma Hepatocelular/genética , Autorrenovação Celular/genética , Fatores de Crescimento de Fibroblastos/genética , Neoplasias Hepáticas/genética , Fatores de Transcrição NFATC/genética , Células-Tronco Neoplásicas/metabolismo , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Fatores de Crescimento de Fibroblastos/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Sistema de Sinalização das MAP Quinases , Fatores de Transcrição NFATC/metabolismo , Fosfolipase C gama , Transdução de Sinais
9.
Theranostics ; 11(10): 4699-4709, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33754022

RESUMO

Rationale: The onset of cytokine release syndrome (CRS) and in vivo persistence of anti-CD19 chimeric antigen receptor T (CAR-T) cells after infusion correlate with clinical responsiveness. However, there are no known baseline biomarkers that can predict the prognosis of patients with B-lineage non-Hodgkin lymphoma (B-NHL). The aim of this study was to identify blood cell populations associated with beneficial outcomes in B-NHL patients administered CAR-T cell immunotherapies. Methods: We enumerated peripheral blood and CAR-T cells by retrospectively analyzing three CAR-T cell trials involving 65 B-NHL patients. We used a preclinical model to elucidate the eosinophil mechanism in CAR-T cell therapy. Results: During an observation period up to 30 mo, B-NHL patients with higher baseline eosinophil counts had higher objective response rates than those with low eosinophil counts. Higher baseline eosinophil counts were also significantly associated with durable progression-free survival (PFS). The predictive significance of baseline eosinophil counts was validated in two independent cohorts. A preclinical model showed that eosinophil depletion impairs the intratumoral infiltration of transferred CAR-T cells and reduces CAR-T cell antitumor efficacy. Conclusion: The results of this study suggest that peripheral eosinophils could serve as stratification biomarkers and a recruitment machinery to facilitate anti-CD19 CAR-T cell therapy in B-NHL patients.


Assuntos
Eosinófilos , Imunoterapia Adotiva , Linfoma de Células B/terapia , Receptores de Antígenos Quiméricos , Adulto , Idoso , Animais , Antígenos CD19 , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Contagem de Leucócitos , Linfoma de Células B/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , Prognóstico , Intervalo Livre de Progressão , Adulto Jovem
10.
Cancer Sci ; 101(11): 2384-90, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20825419

RESUMO

Interleukin-17 (IL-17), a potent pro-inflammatory cytokine, plays an active role in inflammation and cancer. Recently, we found that increased IL-17-producing cells correlate with poor survival and increased lymphangiogenesis in non-small-cell lung cancer (NSCLC), but the mechanism is unknown. Here, we show that IL-17 promotes lymphangiogenesis via inducing vascular endothelial growth factor-C (VEGF-C) production by lung cancer cells. We found that IL-17 receptor (IL-17R) is expressed on the surface of Lewis lung carcinoma (LLC) cells but not on lymphatic endothelial cells (LEC). Moreover, LEC chemotaxis and tube formation (measures of net lymphangiogenic potential) were increased by conditioned medium from recombinant mouse IL-17 (rmIL-17)-stimulated LLC but not by rmIL-17. Interleukin-17 increased production of VEGF-C in lung cancer cell lines. The enhanced chemotaxis and endothelial cord formation in the presence of LLC/rmIL-17 was inhibited by addition of recombinant mouse VEGF R3/Fc chimera. Treatment of the A549 cells with rIL-17 significantly increased VEGF-C expression, which was extracellular signal-regulated protein kinase 1/2 (ERK 1/2) dependent. Importantly, we found significant correlations between IL-17 expression, VEGF-C expression and lymphatic vascular density (LVD) in NSCLC. We conclude that IL-17 is involved in lymphangiogenesis in NSCLC by enhancing production of VEGF-C, and IL-17 may be an important target for the treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Interleucina-17/metabolismo , Neoplasias Pulmonares/metabolismo , Linfangiogênese , Animais , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Quimiotaxia/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Citoplasma/metabolismo , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Imuno-Histoquímica , Interleucina-17/genética , Interleucina-17/farmacologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores de Interleucina-17/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo
11.
Front Immunol ; 11: 594212, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552051

RESUMO

Background and Purpose: To directly reveal the change in genome mutation, RNA transcript of tumor cells, and tumor microenvironment (TME) after stereotactic body radiotherapy (SBRT) in paired human lung tumor specimens. Materials and Methods: Paired tumor samples were collected from 10 patients with non-small cell lung cancer (NSCLC) or lung metastatic carcinoma within a week before and after SBRT. DNA and RNA of tumor tissues was extracted from the paired samples. Whole-exome and RNA sequencing assays were performed by next-generation sequencing. Gene mutation, genomic expression, T-cell receptor (TCR) repertoire, and profiling of tumor-infiltrating immune cells were analyzed through bioinformatics analysis in paired tumor samples. CD8+ T-cell infiltration and PD-L1 expressions were detected by immunostaining in tumor tissues. Results: The diversity of TCR repertoire and PD-L1 expression increased significantly in the TME, and the most enriched term of the gene ontology analysis was the immune response gene after receiving SBRT. SBRT induced neo-mutation of genes in tumor cells but did not increase tumor mutation burden in tumor tissues. TME displayed complex immune cell changes and infiltration and expression of immune-regulating factors such as C-X-C motif chemokine (CXCL) 10, CXCL16, interferons (IFNs), and IFN receptors. CD8+ T-cells in tumor tissues did not improve significantly after SBRT while the infiltrating TH1 and TH2 cells decreased remarkably. Conclusion: SBRT improved the TCR repertoire diversity and PD-L1 expression in the TME and induced neo-mutation of genes in tumor cells but did not increase CD8+ T-cell infiltration and IFN expression in the tumor tissue within a week.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/etiologia , Carcinoma Pulmonar de Células não Pequenas/radioterapia , Neoplasias Pulmonares/etiologia , Neoplasias Pulmonares/radioterapia , Radiocirurgia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Microambiente Tumoral/efeitos da radiação , Adulto , Idoso , Antígeno B7-H1/genética , Biomarcadores Tumorais , Biópsia , Carcinoma Pulmonar de Células não Pequenas/patologia , Feminino , Genômica/métodos , Humanos , Neoplasias Pulmonares/patologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Linfócitos do Interstício Tumoral/patologia , Masculino , Pessoa de Meia-Idade , Mutação , Radiocirurgia/métodos , Receptores de Antígenos de Linfócitos T/metabolismo , Análise de Sequência de RNA , Sequenciamento do Exoma
12.
Oncology ; 76(6): 420-9, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19420963

RESUMO

The expression levels of the RNA-binding protein Hu antigen (HuR) and vascular endothelial growth factor-C (VEGF-C) were examined immunohistochemically in 81 non-small cell lung cancers (NSCLC) and 15 benign human lung tissues. HuR showed a nuclear overexpression in 82.7% (67/81) of NSCLC specimens. Cytoplasmic immunoreactivity for HuR was observed in 45.7% (37/81) of NSCLC, while only nuclear expression of HuR was observed in 13.3% (2/15) of benign lung tissues. The expression of VEGF-C was present in a subgroup of 70.4% (57/81) of tumor cases. In the human NSCLC samples, cytoplasmic but not nuclear HuR expression was significantly associated with increased levels of VEGF-C and with clinicopathological variables, including high tumor grade, poor differentiation and lymph node metastasis. In vitro, HuR showed a predominantly nuclear staining in Lewis lung cancer cells, as seen by confocal microscopy. When lung cancer cells were treated with siRNA targeted against HuR, expression levels of the HuR and VEGF-C proteins were significantly reduced, as seen by Western blotting. Our findings indicate that there is a dysregulation of the cellular distribution of the mRNA stability factor HuR in a subset of NSCLC. Examination of cytoplasmic HuR in NSCLC tissues will allow for valuable prognostic diagnosis of lymph node metastasis, as HuR might be an important mediator regulating the expression of VEGF-C.


Assuntos
Antígenos de Superfície/biossíntese , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Metástase Linfática , Proteínas de Ligação a RNA/biossíntese , Fator C de Crescimento do Endotélio Vascular/biossíntese , Adenocarcinoma/metabolismo , Idoso , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma de Células Escamosas/metabolismo , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Feminino , Humanos , Neoplasias Pulmonares/patologia , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Proteínas de Ligação a RNA/química
13.
Cell Death Dis ; 10(3): 148, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30770798

RESUMO

Mitochondrial pyruvate carrier 1 (MPC1), a key factor that controls pyruvate transportation in the mitochondria, is known to be frequently dysregulated in tumor initiation and progression. However, the clinical relevance and potential molecular mechanisms of MPC1 in lung adenocarcinoma (LAC) progression remain to be illustrated. Herein, MPC1 was lowly expressed in LAC tissues and significantly associated with favorable survival of patients with LAC. Functionally, MPC1 markedly suppressed stemness, invasion, and migration in vitro and spreading growth of LAC cells in vivo. Further study revealed that MPC1 could interact with mitochondrial signal transducer and activator of transcription 3 (mito-STAT3), disrupting the distribution of STAT3 and reducing cytoplasmic signal transducer and activator of transcription 3 (cyto-STAT3) as well as its phosphorylation, while the activation of cyto-STAT3 by IL-6 reversed the attenuated malignant progression in MPC1-overexpression LAC cells. Collectively, we reveal that MPC1/STAT3 axis plays an important role in the progression of LAC, and our work may promote the development of new therapeutic strategies for LAC.


Assuntos
Adenocarcinoma de Pulmão/metabolismo , Progressão da Doença , Neoplasias Pulmonares/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/deficiência , Transportadores de Ácidos Monocarboxílicos/deficiência , Fator de Transcrição STAT3/metabolismo , Células A549 , Adenocarcinoma de Pulmão/mortalidade , Adenocarcinoma de Pulmão/patologia , Animais , Movimento Celular/genética , Estudos de Coortes , Feminino , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Invasividade Neoplásica/genética , Taxa de Sobrevida , Transfecção , Carga Tumoral/genética
14.
Sci Adv ; 5(11): eaax0629, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31763448

RESUMO

The mechanisms that drive programmed resolution of inflammation remain elusive. Here, we report the temporal regulation of soluble (s) and transmembrane (m) fibrinogen-like protein 2 (Fgl2) during inflammation and show that both sFgl2 and mFgl2 correlate with the outcome. The expression and ectodomain shedding of Fgl2 are respectively promoted by miR-466l and metalloproteinases (ADAM10 and ADAM17) during inflammation resolution. Deficiency of Fgl2 enhances polymorphonuclear neutrophil (PMN) infiltration but impairs macrophage (MΦ) maturation and phagocytosis and inhibits the production of n-3 docosapentaenoic acid-derived resolvin 5 (RvDp5). In contrast, administration of sFgl2 blunts PMN infiltration as well as promotes PMN apoptosis and RvDp5 biosynthesis. By activating ALX/FPR2, RvDp5 enhances sFgl2 secretion via ADAM17 and synergistically accelerates resolution of inflammation. These results uncover a previously unknown endogenous programmed mechanism by which Fgl2 regulates resolution of inflammation and shed new light on clinical sepsis treatments.


Assuntos
Proteína ADAM10/metabolismo , Proteína ADAM17/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Fibrinogênio/metabolismo , Sepse/metabolismo , Proteína ADAM10/genética , Proteína ADAM17/genética , Adulto , Animais , Ácidos Docosa-Hexaenoicos/genética , Fibrinogênio/genética , Humanos , Inflamação/genética , Inflamação/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Pessoa de Meia-Idade , Neutrófilos/metabolismo , Sepse/sangue , Sepse/genética , Adulto Jovem
15.
Pharmacology ; 82(2): 148-55, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18667841

RESUMO

We have previously reported that dihydroartemisinin is found to have a potent ability in influencing lymphatic endothelial cell migration and tube formation. In this study, we investigated the effect of artemisinin on tumor growth, lymphangiogenesis, metastasis and survival in mouse Lewis lung carcinoma (LLC) models. We found that orally administered artemisinin inhibited lymph node and lung metastasis and prolonged survival without retarding tumor growth. Consistent with the decrease in lymph node metastasis, tumor lymphangiogenesis and expression of vascular endothelial growth factor C (VEGF-C) was significantly decreased in artemisinin-treated mice, as compared to control mice. Furthermore, IL-1beta-induced p38 mitogen-activated protein kinase (MAPK) activation and upregulation of VEGF-C mRNA and protein in LLC cells was also suppressed by artemisinin or by the p38 MAPK inhibitor SB-203580, suggesting that p38 MAPK could serve as a mediator of proinflammatory cytokine-induced VEGF-C expression. These data indicate that artemisinin may be useful for the prevention of lymph node metastasis by downregulating VEGF-C and reducing tumor lymphangiogenesis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Artemisininas/farmacologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Fator C de Crescimento do Endotélio Vascular/efeitos dos fármacos , Administração Oral , Animais , Carcinoma Pulmonar de Lewis/patologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/prevenção & controle , Neoplasias Pulmonares/secundário , Linfangiogênese/efeitos dos fármacos , Metástase Linfática/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/prevenção & controle , Taxa de Sobrevida , Fator C de Crescimento do Endotélio Vascular/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
Cancer Lett ; 396: 138-144, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28342987

RESUMO

Circular RNAs (circRNAs) are a type of non-coding RNA molecules that lack a 5'-terminal cap and 3'-terminal poly A tail. A large number of circRNAs have been identified through biological experiments, computational methods and high-throughput sequencing. CircRNA sequence composition determines if a given circRNA is exonic, intronic or retained-intronic. CircRNAs are more abundant and stable than linear mRNAs, and their expression is both step- and location-specific. CircRNAs mediate transcriptional and post-transcriptional regulation of gene and protein expression. CircRNAs regulate cancer development via multiple mechanisms, including miRNA sponges, modulating Wnt signaling pathway and epithelial-mesenchymal transition. An in-depth study of circRNA will provide a better understanding of carcinogenesis and assist in developing clinical diagnostic and therapeutic strategies.


Assuntos
Neoplasias/genética , RNA/genética , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , RNA Circular
17.
Zhongguo Fei Ai Za Zhi ; 9(2): 157-61, 2006 Apr 20.
Artigo em Zh | MEDLINE | ID: mdl-21144302

RESUMO

BACKGROUND: p16INK4a and p14ARF, encoded by gene INK4a/ARF located at chromosome 9p21, are cyclin dependent kinase (CDK) inhibitors. Both p16INK4a and p14ARF are cell cycle regulatory proteins and play an important role in Rb and p53 passways respectively. In this study, wild-type INK4a/ARF gene was transfected into human lung adenocarcinoma cell line A549, in which this gene site was lost, and the effects on the cell's biological behavior were investigated. METHODS: The recombinant eukaryotic expression plasmids pcDNA3-p16INK4a and pcDNA3-p14ARF were transfected into A549 by cationic liposome method. By RT-PCR, immunocytochemistry and Western blot after G418 selection, A549 cells that could stably express p16INK4a and p14ARF were obtained. As a control, the parental cell and negative control cell with plasmid pcDNA3-LacZ were used. Inhibition of proliferation was measured by MTT assay. The cell growth curve was drawn according to cell counts. Cell cycle distribution was measured by flow cytometry (FCM), the apoptosis indexes were observed at the same time. The colony formation rate was counted by staining the cells with Coomassie brilliant blue. RESULTS: The introduction of exogenous INK4a and ARF caused significantly growth inhibition of A549. By FCM, more percentage of A549-p16INK4a-p14ARF cells couldn't pass through the checkpoint G1. The percentage of A549-p16INK4a-p14ARF cells inhibited at G0/G1 was 59.9%, 50.3% for A549-vector and 51.2% for A549. The statistical differences were significant between A549-p16INK4a-p14ARF cell and A549-vector cell (P=0.025) and between A549-p16INK4a-p14ARF cell and A549 cell (P=0.043). The apoptosis index of A549-p16INK4a-p14ARF cell was 8.0% and 2.7% for both A549-vector and A549 cell (P < 0.01). The colony formation ability of A549-p16INK4a-p14ARF was weaker than that of A549-vector and A549, they were 63%, 87% and 85% respectively. CONCLUSIONS: The wild-type INK4a/ARF gene can be co-introduced effectively into A549 cell by cationic liposome method. The reexpression of p16INK4a and p14ARF in A549 can inhibit the growth and enhance the apoptosis. This trial will be helpful in using gene therapy of lung cancer in the future.

18.
Colloids Surf B Biointerfaces ; 46(3): 188-96, 2005 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-16337111

RESUMO

Two functionally and structurally different proteins, p16(INK4a) and p14(ARF), encoded by the gene INK4a/ARF located at 9p21 are cyclin-dependent kinase (cdk) inhibitors and important cell cycle regulators. More and more evidences have been accumulated to show that the exogenous p16(INK4a) or p14(ARF) can inhibit the cell growth and/or induce the apoptosis. But it is still unclear if they can play positive role when combine with the conventional chemotherapy in cancer treatment. Here we show that cationic liposome-mediated gene transfection of INK4a/ARF into lung cancer cell line A549, in which the INK4a/ARF locus was lost, suppressed the growth and induced apoptosis. When treated with five different chemotherapy drugs with different mechanism after the transfection, A549 got an increased chemosensitivity for adriamycin and cisplatin and an unchanged result for topotecan, taxol or vinorelbine. The results indicated that cell cycle redistribution and increased apoptosis index after transfection might be the main explanation for the enhanced chemosensitivity. The combination of gene therapy with conventional chemotherapy is not always better than single chemotherapy. This trial will be of benefit to the treatment of lung cancer when combine the conventional chemotherapy and gene therapy in the future.


Assuntos
Antineoplásicos/farmacologia , Divisão Celular/genética , Inibidor p16 de Quinase Dependente de Ciclina/genética , Neoplasias Pulmonares/patologia , Transfecção , Proteína Supressora de Tumor p14ARF/genética , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Neoplasias Pulmonares/genética , Plasmídeos
19.
Int J Clin Exp Med ; 8(2): 2317-25, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25932168

RESUMO

Sorafenib, a novel orally-available multikinase inhibitor blocking several crucial oncogenic signaling pathways, presented survival benefits and became the first-line drug for treatment of patients with Hepatocellular carcinoma (HCC). However, the acquired resistance to Sorafenib resulted in limited benefits. In this study, we aimed to explore possible agents that might overcome Sorafenib resistance by bioinformatics methods. The gene expression profiles of HCC-3sp (acquired Sorafenib-resistance) and HCC-3p (Sorafenib-sensitive) cell line were downloaded from Gene Expression Omnibus (GEO) database. Then, the differentially expressed genes (DEGs) were selected using dChip software. Furthermore, Gene Ontology (GO) and pathway enrichment analyses were performed by DAVID database. Finally, the Connectivity Map was utilized to predict potential chemicals for reversing Sorafenib resistance. Consequently, a total of 541 DEGs were identified, which were associated with cell extracellular matrix, cell adhesion and binding-related items. KEGG pathway analysis indicated that 8 dysfunctional pathways were enriched. Finally, several small molecules, such as pregnenolone and lomustine, were screened out as potential therapeutic agents capable of overcoming Sorafenib resistance. The data identified some potential small molecule drugs for treatment of Sorafenib resistance and offered a novel strategy for investigation and treatments of HCC.

20.
Int J Clin Exp Med ; 8(4): 5822-30, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26131172

RESUMO

Previous reports have indicated that MDM2 T309G polymorphism might be a risk factor for various cancers. Increasing studies have been conducted on the association of MDM2 T309G polymorphism with hepatocellular carcinoma (HCC) risk. However, the results remain inconclusive. Thus, the present study aimed to address this controversy by meta-analysis. Relevant literature up to Oct 2014 was searched and screened. Necessary information was rigorously extracted for data pooling and analyzing. Separate analyses on ethnicity, source of controls, sample size and P53 polymorphism status were also performed. As a result, eleven case-control studies were selected and the overall data indicated a significant association of MDM2 T309G polymorphism with HCC risk (GG vs. TT: OR=2.31; 95% CI=1.66-3.20; dominant model: OR=1.83; 95% CI=1.36-2.47; recessive model: OR=1.73; 95% CI=1.49-2.00). Similar results could be shown in the subgroups regarding ethnicity, source of controls and sample size. Interestingly, in the subgroup analysis regarding P53 codon 72 polymorphism, increased HCC risk could be observed in the Pro/Pro+Pro/Arg subgroup under a recessive model (OR=1.78; 95% CI=1.29-2.44). In conclusion, the results of the present study suggest that MDM2 T309G polymorphism might be a low-penetrant risk factor for HCC. Homozygous GG alleles might interact with Pro of P53 and thus confer the susceptibility to HCC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA