Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
2.
Front Pharmacol ; 14: 1122890, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36937842

RESUMO

Background: Cancer, also known as a malignant tumor, is caused by the activation of oncogenes, which leads to the uncontrolled proliferation of cells that results in swelling. According to the World Health Organization (WHO), cancer is one of the main causes of death worldwide. The main variables limiting the efficacy of anti-tumor treatments are side effects and drug resistance. The search for natural, safe, low toxicity, and efficient chemical compounds in tumor research is essential. Berberine is a pentacyclic isoquinoline quaternary ammonium alkaloid isolated from Berberis and Coptis that has long been used in clinical settings. Studies in recent years have reported the use of berberine in cancer treatment. In this study, we performed a bibliometric analysis of berberine- and tumor-related research. Materials and methods: Relevant articles from January 1, 2002, to December 31, 2021, were identified from the Web of Science Core Collection (WOSCC) of Clarivate Analytics. Microsoft Excel, CiteSpace, VOSviewer, and an online platform were used for the literary metrology analysis. Results: A total of 1368 publications had unique characteristics. Publications from China were the most common (783 articles), and Y. B. Feng (from China) was the most productive author, with the highest total citations. China Medical University (Taiwan) and Sun Yat-sen University (China) were the two organizations with the largest numbers of publications (36 each). Frontiers in Pharmacology was the most commonly occurring journal (29 articles). The present body of research is focused on the mechanism, molecular docking, and oxidative stress of berberine in tumors. Conclusion: Research on berberine and tumors was thoroughly reviewed using knowledge map and bibliometric methods. The results of this study reveal the dynamic evolution of berberine and tumor research and provide a basis for strategic planning in cancer research.

3.
Pharmacol Res ; 172: 105814, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34450313

RESUMO

Cervical cancer is one of the common malignancies in women, which is characterized with high invasion and metastatic tendency in its advanced stage. Increasing evidence indicates that methyltransferase-like (METTL) gene family is involved in the progression of various cancers. However, the functional role of methyltransferase-like gene family in cervical cancer remains unclear. In the present study, we found that METTL11A, a member of the methyltransferase-like gene family, was significantly over-expressed in cervical carcinoma by analyzing TCGA database. This finding was further validated in clinical tissue samples. Moreover, ectopic expression of METTL11A in cervical cancer cell lines promoted cell proliferation and migration both in vitro and in vivo. Differential gene expression analysis in the transcriptomic sequencing data indicated that ELK3 was down-regulated in METTL11A-silenced cervical cancer cells, which was further verified at both protein and mRNA levels. Functional experiments identified that METTL11A promoted migration of cervical cancer cells in an ELK3-dependent manner. This study will promote understanding the mechanism of cervical cancer progression and the functional role of methyltransferase-like gene families in cancers.


Assuntos
Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-ets/genética , Neoplasias do Colo do Útero , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Metiltransferases/genética , Camundongos , Regulação para Cima , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo , Neoplasias do Colo do Útero/patologia , Cicatrização
4.
Int J Oncol ; 58(6)2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33955519

RESUMO

Kidins220 is a transmembrane scaffold protein involved in several types of cancer. The aim of the present study was to examine the role of Kidins220 in tumorigenesis and disease progression of pancreatic cancer. The relevant signalling pathways including EGFR, EMT, and MMP were also investigated. The expression of Kidins220 was examined at the transcript and protein level. The Kidins220 knockdown cell model was established and its influence on cellular functions was determined. Involvement of Kidins220 in tumorigenesis and metastasis was examined in CD1 mice, respectively. The results showed that, reduced Kidin220 expression was associated with tumorigenesis, metastasis, and overall survival of pancreatic cancer. Knockdown of Kidins220 promoted proliferation, colony formation and tumorigenic capacity of pancreatic cancer cells in vitro and in vivo, respectively. Kidins220 regulated pancreatic cancer cell migration through the EGFR/AKT/ERK signalling pathway. Furthermore, enhanced EMT was observed in the pancreatic cancer cell lines with the knockdown of Kidins220, underlying EGFR regulation. Kidins220 also affected cell invasion via MMP1. A reduced expression of Kidins220 was observed in pancreatic cancer, which is associated with disease progression, distant metastasis and poor prognosis. The loss of Kidins220 in pancreatic cancer may contribute to disease progression through the upregulation of EGFR and downstream signalling.


Assuntos
Carcinogênese/patologia , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Pancreáticas/patologia , Animais , Linhagem Celular Tumoral , Estudos de Coortes , Progressão da Doença , Receptores ErbB/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Humanos , Estimativa de Kaplan-Meier , Metaloproteinase 1 da Matriz/metabolismo , Proteínas de Membrana/genética , Camundongos , Invasividade Neoplásica/patologia , Proteínas do Tecido Nervoso/genética , Pâncreas/patologia , Pâncreas/cirurgia , Pancreatectomia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/cirurgia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Int J Oncol ; 57(3): 813-824, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32705152

RESUMO

Noggin is an antagonist of bone morphogenetic proteins (BMP), being indispensable for certain developmental events. The present study aimed to examine the role of Noggin in the development and prognosis of gastric cancer (GC) and to elucidate the underlying mechanisms. The expression of Noggin in GC was evaluated by RT­qPCR, immunohistochemistry and by the analyses of publicly available databases. The effects of Noggin on proliferation, cell cycle, adhesion, invasion, colony formation and tumour spheroid were examined following both the knockdown and overexpression of Noggin in GC cell lines. The involvement of epidermal growth factor receptor (EGFR) signalling was examined by western blot analysis and by using small molecule inhibitors. As a result, a higher expression of Noggin in GC was found to be associated with a poorer overall survival. Noggin overexpression promoted the proliferation and colony formation of GC cells by promoting cell cycle progression. The knockdown of Noggin in HGC27 cells exerted an opposite effect on proliferation, colony formation and cell cycle progression. Noggin expression positively correlated with EGFR expression in both GC cell line models and The Cancer Genome Atlas human GC cohort. Targeting EGFR and its downstream pathways diminished cell proliferation which was promoted by Noggin. Furthermore, Noggin overexpression resulted in an enhanced nuclear translocation of ß­catenin, leading to an upregulation of EGFR. Thus, the findings of the present study demonstrate that Noggin promotes the proliferation of GC cells by upregulating EGFR and enhancing a vicious circle formed by ß­catenin, EGFR, ERK and Akt.


Assuntos
Biomarcadores Tumorais/metabolismo , Proteínas de Transporte/metabolismo , Mucosa Gástrica/patologia , Neoplasias Gástricas/mortalidade , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/genética , Conjuntos de Dados como Assunto , Intervalo Livre de Doença , Receptores ErbB/genética , Feminino , Gastrectomia , Mucosa Gástrica/cirurgia , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Estimativa de Kaplan-Meier , Sistema de Sinalização das MAP Quinases/genética , Masculino , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Neoplasias Gástricas/cirurgia , Regulação para Cima , beta Catenina/metabolismo
6.
Trials ; 21(1): 77, 2020 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-31937335

RESUMO

BACKGROUND: Acupoint selection is a key factor in the treatment of diseases and has not been well studied. The aim of this trial is to explore the differences in efficacy between compatible acupoints and a single acupoint for patients with functional dyspepsia (FD). METHODS: This randomized controlled trial will be conducted in the First Affiliated Hospital of Changchun University of Chinese Medicine in China. Two hundred and sixteen FD patients will be randomly assigned to the compatible acupoints group, single acupoint group, or sham acupuncture group. This trial will include a 1-week baseline period, a 4-week treatment period, and a 4-week follow-up period. During the 4-week treatment period, patients will receive 20 sessions of acupuncture (weekly cycles of one session per day for 5 consecutive days followed by a 2-day break). The primary outcome will be a change in the Nepean Dyspepsia Life Quality Index from baseline to after the 4-week treatment period. Secondary outcome measures will include the dyspeptic symptom sum score, Overall Treatment Effect questionnaire, and 36-item Short Form survey. Adverse events also will be recorded. Ultraweak photon emission and metabolomics tests will be performed at baseline and at the end of treatment to explore the mechanisms of the differences between compatible acupoints and a single acupoint. DISCUSSION: The results of this trial will allow us to compare the difference in efficacy between compatible acupoints and a single acupoint. The findings from this trial will be published in peer-reviewed journals. TRIAL REGISTRATION: Acupuncture-Moxibustion Clinical Trial Registry, AMCTR-IPC-18000176, registered on 4 March 2019; Chinese Clinical Trial Registry, ChiCTR1900023983, registered on 23 June 2019.


Assuntos
Pontos de Acupuntura/classificação , Terapia por Acupuntura/métodos , Dispepsia/fisiopatologia , Dispepsia/terapia , Terapia por Acupuntura/efeitos adversos , Adolescente , Adulto , Estudos de Casos e Controles , China/epidemiologia , Dispepsia/psicologia , Feminino , Humanos , Masculino , Metabolômica/métodos , Pessoa de Meia-Idade , Fótons , Qualidade de Vida , Projetos de Pesquisa , Inquéritos e Questionários , Resultado do Tratamento , Adulto Jovem
7.
Oncotarget ; 8(16): 26231-26244, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-28412738

RESUMO

Aberrant expression of nephroblastoma overexpressed (NOV) has been evident in certain malignancies. In the current study, we aim to investigate the role played by NOV in colorectal cancer (CRC). NOV expression was determined in a cohort of 359 CRC tissues and 174 normal colorectal tissues. Its impact on CRC cells was investigated using in vitro NOV knockdown and overexpression models. NOV transcripts were reduced in the CRC tumours compared with the paired adjacent normal colorectal tissues (p < 0.01) and was associated with distant metastases. NOV knockdown resulted in increased cell proliferation and invasion of RKO cells, whilst an opposite effect was seen in the HT115 NOV over expressing cells. A positive association between Caspase-3/-8 and NOV was seen in NOV knockdown and overexpression cell lines which contributed to the survival of serum deprived CRC cells. Further investigation showed that NOV regulated proliferation, survival and invasion through the JNK pathway. NOV knockdown in RKO cells reduced the responsiveness to 5-Fluorouracil treatment, whilst overexpression in HT115 cells exhibited a contrasting effect. Taken together, NOV is reduced in CRC tumours and this is associated with disease progression. NOV inhibits the proliferation and invasion of CRC cells in vitro. Inhibition of proliferation is mediated by a regulation of Caspase-3/-8, via the JNK pathway, which has potential for predicting and preventing chemoresistance.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Proteína Sobre-Expressa em Nefroblastoma/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/mortalidade , Progressão da Doença , Humanos , Sistema de Sinalização das MAP Quinases , Gradação de Tumores , Metástase Neoplásica , Estadiamento de Neoplasias , Prognóstico
8.
Int J Oncol ; 50(5): 1491-1500, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28393239

RESUMO

Psoriasin (S100A7) is an 11-kDa small calcium binding protein initially isolated from psoriatic skin lesions. It belongs to the S100 family of proteins which play an important role in a range of cell functions including proliferation, differentiation, migration and apoptosis. Aberrant Psoriasin expression has been implicated in a range of cancers and is often associated with poor prognosis. This study examined the role of Psoriasin on pancreatic cancer cell functions and the implication in progression of the disease. Expression of Psoriasin was determined in a cohort of pancreatic tissues comprised of 126 pancreatic tumours and 114 adjacent non-tumour pancreatic tissues. Knockdown and overexpression of Psoriasin in pancreatic cancer cells was performed using specifically constructed plasmids, which either had anti-Psoriasin ribozyme transgene or the full length human Psoriasin coding sequence. Psoriasin knockdown and overexpression was verified using conventional RT-PCR and qPCR. The effect of manipulating Psoriasin expression on pancreatic cancer cell functions was assessed using several in vitro cell function assays. Local invasive pancreatic cancers extended beyond the pancreas expressed higher levels of Psoriasin transcripts compared with the cancers confined to the pancreas. Primary tumours with distant metastases exhibited a reduced expression of Psoriasin. Psoriasin overexpression cell lines exhibited significantly increased growth and migration compared to control cells. In addition, Psoriasin overexpression resulted in increased pancreatic cancer cell invasion which was associated with upregulation of matrix metalloproteinase-2 (MMP-2) and MMP-9. Overexpression of Psoriasin also promoted aggregation and survival of pancreatic cancer cells when they lost anchorage. Taken together, higher expression of Psoriasin was associated with local invasion in pancreatic cancers. Psoriasin expression is associated with pancreatic cancer cell growth, migration, cell-matrix adhesion, and invasion via regulation of MMPs. As such, the proposed implications of Psoriasin in invasion, disease progression and as a potential therapeutic target warrant further investigation.


Assuntos
Biomarcadores Tumorais/genética , Agregação Celular/genética , Invasividade Neoplásica/genética , Neoplasias Pancreáticas/genética , Proteínas S100/genética , Apoptose/genética , Biomarcadores Tumorais/biossíntese , Movimento Celular/genética , Sobrevivência Celular/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Invasividade Neoplásica/patologia , Metástase Neoplásica , Neoplasias Pancreáticas/patologia , Proteína A7 Ligante de Cálcio S100 , Proteínas S100/biossíntese
9.
Int J Oncol ; 50(4): 1127-1135, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28259897

RESUMO

A potential role may be played by receptor-type protein tyrosine phosphatase kappa (PTPRK) in angiogenesis due to its critical function in coordinating intracellular signal transduction from various receptors reliant on tyrosine phosphorylation. In the present study, we investigated the involvement of PTPRK in the cellular functions of vascular endothelial cells (HECV) and its role in angiogenesis using in vitro assays and a PTPRK knockdown vascular endothelial cell model. PTPRK knockdown in HECV cells (HECVPTPRKkd) resulted in a decrease of cell proliferation and cell-matrix adhesion; however, increased cell spreading and motility were seen. Reduced focal adhesion kinase (FAK) and paxillin protein levels were seen in the PTPRK knockdown cells which may contribute to the inhibitory effect on adhesion. HECVPTPRKkd cells were more responsive to the treatment of fibroblast growth factor (FGF) in their migration compared with the untreated control and cells treated with VEGF. Moreover, elevated c-Src and Akt1 were seen in the PTPRK knockdown cells. The FGF-promoted cell migration was remarkably suppressed by an addition of PLCγ inhibitor compared with other small inhibitors. Knockdown of PTPRK suppressed the ability of HECV cells to form tubules and also impaired the tubule formation that was induced by FGF and conditioned medium of cancer cells. Taken together, it suggests that PTPRK plays dual roles in coordinating angiogenesis. It plays a positive role in cell proliferation, adhesion and tubule formation, but suppresses cell migration, in particular, the FGF-promoted migration. PTPRK bears potential to be targeted for the prevention of tumour associated angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Neovascularização Patológica/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Transdução de Sinais , Proteína Tirosina Quinase CSK , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Fatores de Crescimento de Fibroblastos/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Paxilina/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Quinases da Família src/metabolismo
10.
Oncol Rep ; 37(3): 1707-1715, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28112359

RESUMO

Phospholipase C (PLC) regulates a number of cellular behaviours including cell motility, cell transformation, differentiation and cell growth. PLC plays a regulatory role in cancer cells partly by acting as signalling intermediates for cytokines such as EGF and interleukins. The current study examined the expression of the PLC isozymes in human breast cancer and corresponding clinical relevance. Transcript levels of human PLC-α, -ß1, -δ, -ε, and -γ1 in human breast cancer tissues were quantitatively determined by real-time PCR. Immunochemical staining was performed for PLC-δ. The clinical relevance was analysed with clinic pathological information. Mammary tissues widely expressed PLC-α, -ß1, -δ, -ε, and -γ1. Significantly high levels of PLC -ß1 and -ε were seen in breast cancer tissues in comparison with normal mammary gland tissues. PLC-γ1 however, showed marginally low levels in tumour tissues. No significant difference was seen in the expression of the PLC isozymes in tumours with lymph node metastases. Moderately and poorly differentiated breast tumours (grade 2 and grade 3) had significantly higher levels of PLC-γ1, compared with well differentiated tumours. High levels of PLC-δ were significantly correlated with a shorter disease-free survival. The altered expression of other isozymes had no correlation with the survival. It is concluded that mammary tissues differentially expressed PLC isozymes. These isozymes have certain implications in the disease development and progression, with PLC-δ showing a significant correlation with shorter disease-free survival.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/enzimologia , Regulação Enzimológica da Expressão Gênica , Fosfoinositídeo Fosfolipase C/metabolismo , Fosfolipase C beta/metabolismo , Fosfolipase C delta/metabolismo , Fosfolipase C gama/metabolismo , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Movimento Celular , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas Imunoenzimáticas , Isoenzimas , Gradação de Tumores , Fosfoinositídeo Fosfolipase C/genética , Fosfolipase C beta/genética , Fosfolipase C delta/genética , Fosfolipase C gama/genética , Prognóstico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Células Tumorais Cultivadas
11.
Anticancer Res ; 36(3): 1227-35, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26977019

RESUMO

BACKGROUND: Prostate apoptosis response-4 (PAR4) plays an important role in apoptosis and survival of cancer cells. The current study aimed to further elucidate its role in breast cancer. MATERIALS AND METHODS: PAR4 expression in human breast cancer tissue was examined using quantitative polymerase chain reaction (qPCR) and immunohistochemical staining (IHC). Plasmids containing full-length human PAR4 coding sequence were used to overexpress PAR4 in breast cancer cells and the effect on cellular functions was examined using both in vitro functional assays and an in vivo murine model. RESULTS: Patients with low PAR4 transcript levels had poorer overall survival. PAR4 expression may be associated with differential expression of oestrogen receptors α and ß in the tumours. Overexpression of PAR4 in MDA-MB-231 cells resulted in reduced cell growth and invasion, and also reduced in vivo tumour growth. CONCLUSION: Decreased PAR4 expression in breast cancer is associated with shorter survival. PAR4 suppresses growth and invasiveness of breast cancer cells.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Movimento Celular , Proliferação de Células , Animais , Proteínas Reguladoras de Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Estimativa de Kaplan-Meier , Células MCF-7 , Camundongos Nus , Invasividade Neoplásica , Modelos de Riscos Proporcionais , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Fatores de Tempo , Transfecção , Carga Tumoral
12.
Anticancer Res ; 36(3): 1295-303, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26977028

RESUMO

AIM: Ran binding protein M (RanBPM) is a ubiquitous, nucleocytoplasmic protein that serves as a scaffolding molecule. This study aimed to investigate the role of RanBPM in gastric cancer. MATERIALS AND METHODS: RanBPM expression in human gastric cancer tissue samples was analyzed using real-time polymerase chain reaction. The effect of RanBPM on cellular functions was examined in RanBPM-knockdown gastric cells and with in vitro cell functional assays. RESULTS: Gastric tumors with distant metastases expressed lower levels of RanBPM transcripts compared to tumours without detectable metastases (p=0.036). RanBPM knockdown in gastric cancer cells reduced adhesion and promoted survival of gastric cancer cells after exposure to methotrexate and fluorouracil. CONCLUSION: RanBPM levels were reduced in gastric tumors with distant metastases. This suggests that loss of RanBPM expression may play an important role in gastric cancer tumor development and metastasis. Reduced RanBPM expression is also associated with chemoresistance of gastric cancer cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Movimento Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fluoruracila/farmacologia , Metotrexato/farmacologia , Proteínas Nucleares/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Adenocarcinoma/genética , Adenocarcinoma/secundário , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Invasividade Neoplásica , Proteínas Nucleares/genética , RNA Catalítico/genética , RNA Catalítico/metabolismo , Transdução de Sinais , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Fatores de Tempo , Transfecção
13.
Cancer Genomics Proteomics ; 12(6): 339-45, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26543080

RESUMO

BACKGROUND: Metastasis-associated protein 1 (MTA1) plays an important role in tumourigenesis and progression of certain cancer types. In the current study, we analyzed the relationship between MTA1 expression and disease progression of colorectal cancer (CRC). MATERIALS AND METHODS: CRC tissues (n=93) and adjacent normal colorectal tissues (n=70) were analyzed by quantitative real-time polymerase chain reaction. MTA1 knockdown was established in RKO and HT115 cells using MTA1 siRNA. RESULTS: The expression of MTA1 was significantly increased in CRC tissues compared to paired normal colorectal tissues, but decreased expression of MTA1 was correlated with poor prognosis (higher lymph node involvement stage, TNM stage, local invasion and recurrence) that was associated with increased expression of VEGFC and -D and the receptor VEGFR3. CONCLUSION: MTA1 is up-regulated in CRC. MTA1 expression is inversely associated with lymphatic metastases and the expression of VEGFC, VEGFD and VEGFR3.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Metástase Linfática , Proteínas Repressoras/metabolismo , Regulação para Cima , Biomarcadores Tumorais/metabolismo , Células CACO-2 , Linhagem Celular Tumoral , Progressão da Doença , Seguimentos , Humanos , Invasividade Neoplásica , Recidiva Local de Neoplasia , Estadiamento de Neoplasias , Prognóstico , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transativadores , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Cicatrização
14.
J Transl Med ; 13: 280, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26310485

RESUMO

BACKGROUND: Hepatocyte growth factor (HGF) is a cytokine that has a profound effect on cancer cells by stimulating migration and invasion and acting as an angiogenic factor. In lung cancer, the factor also plays a pivotal role and is linked to a poor outcome in patients. In particular, HGF is known to work in combination with EGF on lung cancer cells. In the present study, we investigated the effect of a traditional Chinese medicine reported in cancer therapies, namely YangZheng XiaoJi (YZXJ) on lung cancer and on HGF mediated migration and invasion of lung cancer cells. METHODS: Human lung cancer cells, SKMES1 and A549 were used in the study. An extract from the medicine was used. Cell migration was investigated using the EVOS and by ECIS. Cell-matrix adhesion and in vitro invasion were assessed. In vivo growth of lung cancer was tested using an in vivo xenograft tumour model and activation of the HGF receptor in lung tumours by an immunofluorescence method. RESULTS: Both lung cancer cells increased their migration in response to HGF and responded to YZXJ by reducing their speed of migration. YZXJ markedly reduced the migration and in vitro invasiveness induced by HGF. It worked synergistically with PHA665752 and SU11274, HGF receptor inhibitors on the lung cancer cells both on HGF receptor activation and on cell functions. A combination of HGF and EGF resulted in a greater increase in cell migration, which was similarly inhibited by YZXJ, and in combination with the HGF receptor and EGF receptor inhibitors. In vivo, YZXJ reduced the rate of tumour growth and potentiated the effects of PHA665752 on tumour growth. It was further revealed that YZXJ significantly reduced the degree of phosphorylation of the HGF receptor in lung tumours. CONCLUSION: YZXJ has a significant role in reducing the migration, invasion and in vivo tumour growth of lung cancer and acts to inhibit the migratory and invasive effects induced by HGF and indeed by HGF/EGF. This effect is likely attributed to the inhibition of the HGF receptor activation. These results indicate that YZXJ has a therapeutic role in lung cancer and that combined strategy with methods to block HGF and EGF should be considered.


Assuntos
Antineoplásicos/química , Medicamentos de Ervas Chinesas/química , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Neoplasias Pulmonares/metabolismo , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Animais , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Citocinas/metabolismo , Impedância Elétrica , Feminino , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Concentração Inibidora 50 , Medicina Tradicional Chinesa , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Neovascularização Fisiológica , Proteínas Proto-Oncogênicas c-met/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais
15.
Cancer Cell Int ; 15: 51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26052252

RESUMO

BACKGROUND: The WASP (Wiskott-Aldrich syndrome protein) and WAVE (WASP Verpolin homologous) family of proteins are structurally related and responsible for regulation of actin polymerization through their interaction with actin related proteins 2&3 (ARP 2/3). WAVE-3 has exhibited an association with disease progression and poorer prognosis of certain malignancies. In the current study, we determined the role of WAVE-3 in hepatocyte growth factor induced cellular changes including cell matrix interaction, invasion and cellular motility, and pathways that may be responsible for the changes in prostate cancer cells. METHODS: We used hammer head ribozymes to knock down the expression of WAVE-3 in PC-3 prostate cancer cell line. In vitro cellular functional assays including growth, invasion, adhesion, motility and invasion, were performed to assess the effects of WAVE-3 knock down. Further experimentation was performed to investigate the role of different pathway through expression and phosphorylation status of various intermediate proteins. RESULTS: WAVE-3 knockdown reduced invasive potential and motility of prostate cancer cells. Following addition of HGF, control cells showed significantly increased invasion and motility (p value <0.5) and marked increase in cellular growth. However, WAVE-3 knockdown cell line failed to show any increase in these trends (p value <0.5) except increased growth compared with control cells. Further experiments revealed that HGF-induced activation of Paxillin was weakened by the knockdown of WAVE-3. Our study also indicated that reduced invasiveness following WAVE-3 knockdown, may be related to reduce activity of MMP-2. CONCLUSIONS: Our studies suggest a vital role of WAVE-3 in HGF induced invasion and migration in which Paxillin and MMP-2 are involved. Further study will shed light on its potential as therapeutic target to suppress local invasion and metastasis of prostate cancer cells.

16.
Anticancer Res ; 35(3): 1321-31, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25750281

RESUMO

Sonic Hedgehog (SHH) is a protein that is aberrantly expressed in various human tumors. SHH and its signaling molecules have been indicated as potential therapeutic targets. In the present study, we evaluated the expression of SHH transcript in human non-small cell lung cancer (NSCLC) tissues and investigated the impact of inhibiting SHH together with a traditional Chinese medicine formula, YangZheng XiaoJi (YZXJ), on the function and growth of lung cancer cells. Human NSCLC tissues had significantly higher levels of the SHH transcript compared matched normal lung tissues (n=83). TNM2 tumors and tumors with pleural invasion had higher levels than TNM1 and non-invasive tumors. High SHH levels were associated with a shorter overall survival (OS) of the patients. A SHH inhibitor, cyclopamine, and YZXJ alone or in combination had a marked inhibitory effect on cellular invasion and cellular migration of human lung cancer cells, A549 and SKMES1. YangZheng XiaoJi and its combination with cyclopamine also significantly reduced the growth of lung tumors in vivo together with a reduction of SHH and smoothened (Smo) proteins in the lung tumors. The present study provides evidence that blocking SHH by way of small inhibitor and by YangZheng XiaoJi has a profound influence on lung cancer cells as seen by in vitro invasion and cell migration and in vivo tumor growth. Together with the aberrant expression of SHH in NSCLC tumors in the patients, it is suggested that SHH is a potential target for therapies for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Proteínas Hedgehog/fisiologia , Neoplasias Pulmonares/tratamento farmacológico , Animais , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Proteínas Hedgehog/genética , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Alcaloides de Veratrum/farmacologia
17.
Int J Oncol ; 45(4): 1565-73, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24993339

RESUMO

Capillary morphogenesis gene 2 (CMG2) is a receptor of anthrax toxin and plays an important role in angiogenesis. It has been shown to be involved in the cell adhesion and motility of various cell types, including epithelia and endothelia. The present study aimed to examine the therapeutic potential of targeting CMG2 to prevent tumour­related new vasculature. The full-length coding sequence of the human CMG2 gene and different fragments of the CMG2 vWA domain were amplified and constructed into a mammalian expression plasmid vector. The effect of CMG2 and its vWA domain on endothelial cells and angiogenesis was assessed using relevant in vitro, ex vivo and in vivo models. The overexpression of CMG2 enhanced the adhesion of endothelial cells to extracellular matrix, but was negatively associated with cell migration. Overexpression of CMG2 and the vWA domain fragments inhibited the tubule formation and migration of endothelial cells. Small peptides based on the amino acid sequence of the CMG2 vWA domain fragments potently inhibited in vitro tubule formation and ex vivo angiogenesis. One of the polypeptides, LG20, showed an inhibitory effect on in vivo tumour growth of cancer cells which were co-inoculated with the vascular endothelial cells. CMG2 is a potential target for treating tumour­related angiogenesis. The polypeptides based on the CMG2 vWA domain can potently inhibit in vitro and ex vivo angiogenesis, which may contribute to the inhibitory effect on in vivo tumour growth. Further investigations are required to shed light on the machinery and may provide a novel therapeutic approach for inhibition of angiogenesis in cancer management.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/química , Neoplasias Experimentais/tratamento farmacológico , Peptídeos/administração & dosagem , Peptídeos/química , Receptores de Peptídeos/química , Receptores de Peptídeos/metabolismo , Animais , Antineoplásicos/uso terapêutico , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/patologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/patologia , Peptídeos/uso terapêutico , Receptores de Peptídeos/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Fator de von Willebrand/química
18.
Oncol Lett ; 7(6): 2149-2153, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24932305

RESUMO

Capillary morphogenesis gene 2 (CMG2), also known as anthrax toxin receptor 2, has been indicated in the formation of new vasculature and in the internalisation of the anthrax toxin. Anti-angiogenesis therapy that targets this molecule has been investigated. However, our recent studies of this molecule have indicated that this gene may also play certain roles in cancer cells. The present study aimed to examine the expression of CMG2 in prostate cancer tissues and cell lines, and also its impact on cellular functions. The expression of CMG2 was detectable in normal and prostate cancer tissues. The prostate cancer cell lines appeared to have relatively high expression compared with the prostatic epithelial cells. Knockdown of CMG2 impaired the adherence of the prostate cancer cells. CMG2 overexpression resulted in decreasing invasiveness, while the knockdown of CMG2 contrastingly enhanced this ability. The altered expression of CMG2 in the prostate cancer cells did not affect the in vitro or in vivo growth of the cells. Taken together, these results show that CMG2 is expressed in prostatic epithelia and cancer cells. In addition to its role in the angiogenesis and the internalisation of anthrax toxin, CMG2 also plays an important role in regulating the adhesion and invasion of prostate cancer cells.

19.
J Cancer Res Clin Oncol ; 140(6): 957-67, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24667935

RESUMO

BACKGROUND: Capillary morphogenesis gene 2 (CMG2) also known as anthrax toxin receptor 2 was identified as a gene being up-regulated in capillary morphogenesis. It has been shown to be involved in cell adhesion and motility which are critical functions for cancerous cells to disseminate. The present study aimed to examine the expression of CMG2 in breast cancer and its implication in the disease progression. MATERIALS AND METHODS: Breast primary tumours and background tissues were collected immediately after surgery and stored at -80 °C with approval by the local ethics committee, and written consent obtained from patients. The expression of CMG2 in 127 breast cancer tumour samples and 34 normal mammary tissues was determined using real-time PCR. Knockdown and over-expression in breast cancer cells were established using constructed plasmid vectors carrying either anti-CMG2 ribozyme or full-coding sequence of human CMG2. The effect on growth of breast cancer cells was examined using in vitro and in vivo models. RESULTS: The CMG2 transcript levels were reduced in advanced tumours compared with its expression in tumours of early stage according to their TNM staging. The reduced expression was associated with shorter overall survival, p = 0.004 compared with patients who had higher expression. The knockdown of CMG2 resulted in an increased in vitro growth of MDA-MB-231 cells which express this gene at relatively higher levels. This is consistent with the finding from MCF-7 cells which express lower levels of CMG2 and exhibited reduced growth following over-expression of CMG2. The over-expression of CMG2 also demonstrated an inhibitory effect on in vivo growth of MCF-7 cells. CONCLUSION: Reduced expression of CMG2 is associated with disease progression and poor prognosis of breast cancer. CMG2 has an inhibitory effect on growth of breast cancer cells. Further investigation is required to shed light on the prognostic and therapeutic potential of targeting this molecule.


Assuntos
Neoplasias da Mama/patologia , Mama/patologia , Proliferação de Células , Recidiva Local de Neoplasia/patologia , Receptores de Peptídeos/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Adenocarcinoma/patologia , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/mortalidade , Adenocarcinoma Mucinoso/patologia , Animais , Western Blotting , Mama/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/mortalidade , Carcinoma Ductal de Mama/patologia , Carcinoma Medular/metabolismo , Carcinoma Medular/mortalidade , Carcinoma Medular/patologia , Estudos de Casos e Controles , Adesão Celular , Estudos de Coortes , Progressão da Doença , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Camundongos , Camundongos Nus , Gradação de Tumores , Invasividade Neoplásica , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/mortalidade , Estadiamento de Neoplasias , Prognóstico , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Int J Oncol ; 43(5): 1560-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24002526

RESUMO

Receptor-like protein tyrosine phosphatase κ (PTPRK) has been indicated as a putative tumour suppressor in primary central nervous system lymphomas and colorectal cancer. The present study investigated the expression of PTPRK in prostate cancer and the biological impact of PTPRK on prostate cancer cells. The expression of the PTPRK protein and transcript in prostate cancer was examined using IHC and PCR. Knockdown of PTPRK in prostate cancer cells was performed using a specific anti-PTPRK transgene. The impact of PTPRK knockdown on prostate cancer cells was evaluated using in vitro cell models and the apoptosis was analysed using flow cytometry. PTPRK expression was increased in prostate cancer tissues and knockdown of PTPRK in PC-3 cells suppressed the in vitro cell growth in which an increased apoptotic population was seen. Accompanied with the knockdown of PTPRK, increased expression of caspase-3, caspase-8 and p53, and a decreased ID1 expression were evident in the cells. Furthermore, an increased tyrosine phosphorylated c-Jun N-terminal kinase (JNK) was seen in the PTPRK knockdown cells. The effect on apoptosis was diminished by a JNK inhibitor. In conclusion, PTPRK knockdown resulted in increased apoptosis leading to the inhibition of in vitro growth of prostate cancer cells. PTPRK is a key factor in coordinating apoptosis via the regulation of MAPK pathways, in particular the JNK pathway in prostate cancer cells.


Assuntos
Apoptose , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias da Próstata/patologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Western Blotting , Proliferação de Células , Humanos , Técnicas Imunoenzimáticas , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/antagonistas & inibidores , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...