Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 12: 715508, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34899684

RESUMO

Transmembrane Channel-like (TMC) genes are critical in the carcinogenesis, proliferation, and cell cycle of human cancers. However, the multi-omics features of TMCs and their role in the prognosis and immunotherapeutic response of human cancer have not been explored. We discovered that TMCs 4-8 were commonly deregulated and correlated with patient survival in a variety of cancers. For example, TMC5 and TMC8 were correlated with the relapse and overall survival rates of breast cancer and skin melanoma, respectively. These results were validated by multiple independent cohorts. TMCs were regulated by DNA methylation and somatic alterations, such as TMC5 amplification in breast cancer (523/1062, 49.2%). Six algorithms concordantly uncovered the critical role of TMCs in the tumor microenvironment, potentially regulating immune cell toxicity and lymphocytes infiltration. Moreover, TMCs 4-8 were correlated with tumor mutation burden and expression of PD-1/PD-L1/CTLA4 in 33 cancers. Thus, we established an immunotherapy response prediction (IRP) score based on the signature of TMCs 4-8. Patients with higher IRP scores showed higher immunotherapeutic responses in five cohorts of skin melanoma (area under curve [AUC] = 0.90 in the training cohort, AUCs range from 0.70 to 0.83 in the validation cohorts). Together, our study highlights the great potential of TMCs as biomarkers for prognosis and immunotherapeutic response, which can pave the way for further investigation of the tumor-infiltrating mechanisms and therapeutic potentials of TMCs in cancer.


Assuntos
Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Canais Iônicos/genética , Proteínas de Membrana/genética , Neoplasias , Humanos , Imunoterapia , Canais Iônicos/análise , Proteínas de Membrana/análise , Prognóstico , Resultado do Tratamento
2.
Ann Transl Med ; 8(24): 1661, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33490173

RESUMO

BACKGROUND: In recent years, nanotechnology has attracted a plethora of attention due of its ability to effectively diagnose and treat various tumors. Virus-like particles (VLPs) have good biocompatibility, are safe and non-toxic, and have an internal hollow space, and as such they are often used as nano drug carriers. In recent years, it has become one of the hot spots in the field of biopharmaceutical engineering. METHODS: In this study, the tumor-targeting peptide RGD (Arg-Gly-Asp) was genetically inserted into the major immunodominant region (MIR) of the hepatitis B virus core protein (HBc). A series of characterization, including stability and optical properties, were evaluated. A visual diagnosis and analysis of the efficacy against tumor cells were conducted at the cell level and using a live animal model. RESULTS: This study demonstrated that the recombinant HBc-based VLPs could participate in self-assembly of monodispersed nanoparticles with well-defined morphology, and the near-infrared dye indocyanine green (ICG) could be packaged into the VLPs without any chemical modification. Moreover, the HBc-based VLPs could specifically target cancer cells via the interaction with overexpressed integrin αvß3. The treatment with ICG-loaded HBc-based VLPs showed significant inhibition of 4T1 breast cancer cell growth (84.87% tumor growth inhibition). The in vivo imaging experiments demonstrated that the ICG-loaded HBc-based VLPs generated excellent fluorescence in tumor sites in 4T1 breast cancer bearing mice. This provided crucial information on tumor mass location, boundaries, and shape. Moreover, compared to free ICG, the nanosystem showed significantly longer blood circulation time and superior accuracy in targeting the tumor. CONCLUSIONS: The ICG-loaded HBc-based VLPs prepared in this study were of good stability and biocompatibility. It showed strong tumor targeting specificity and tumor visualization. Thus, it is expected to provide a new experimental basis and theoretical support for the integration of VLPs in the clinical diagnosis and treatment of breast cancer.

3.
Oncol Rep ; 42(5): 1667-1676, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31485620

RESUMO

It has been demonstrated that microRNA­192 (miR­192) serves important roles in different cancer types, including breast cancer, prostate cancer and colorectal cancer. However, its biological role and function in breast cancer remains largely unknown. The present study aimed to determine the role of miR­192 in breast cancer. In the present study, one normal breast and two breast tumor cells lines were used, which included the normal mammary fibroblast cell line Hs578Bst, a more aggressive breast tumor cell line MDA­MB­231 and a less aggressive breast tumor cell line MCF­7. The effect of miR­192 on proliferation of breast cancer cells was detected with an MTT assay. Western blot analysis was performed to determine protein expression of caveolin 1 (CAV1). A lentiviral vector that overexpresses pre­miR­192 and control lentiviral packaging plasmids were used in the present study. The Student's t­test was performed to analyze the significance of differences between samples. In the present study, it was determined that the expression of miR­192 is downregulated in breast cancer, compared with the adjacent normal tissues. Overexpression of miR­192 significantly inhibited cell proliferation, and induced cell apoptosis and cell cycle arrest in MCF7 and MDA­MB­231 cells. Using a bioinformatics method, CAV1 was considered a potential target of miR­192. Furthermore, it was demonstrated that CAV1 is a direct target of miR­192 and its protein expression is negatively regulated by miR­192. Therefore, the present study demonstrated that miR­192 serves an important role as a regulator in breast cancer and the miR­192/CAV1 axis has a potential as a therapeutic target for treatment of breast cancer.


Assuntos
Neoplasias da Mama/patologia , Caveolina 1/genética , Caveolina 1/metabolismo , MicroRNAs/genética , Regiões 3' não Traduzidas , Idoso , Animais , Apoptose , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Camundongos , Pessoa de Meia-Idade , Transplante de Neoplasias
4.
Free Radic Biol Med ; 104: 280-297, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28131902

RESUMO

Autophagy plays a key role in supporting cell survival against chemotherapy-induced apoptosis. In this study, we found the chemotherapy agent SN-38 induced autophagy in colorectal cancer (CRC) cells. However, inhibition of autophagy using a small molecular inhibitor 3-methyladenine (3-MA) and ATG5 siRNA did not increase SN-38-induced cytotoxicity in CRC cells. Notably, another autophagy inhibitor chloroquine (CQ) synergistically enhanced the anti-tumor activity of SN-38 in CRC cells with wild type (WT) p53. Subsequently, we identified a potential mechanism of this cooperative interaction by showing that CQ and SN-38 acted together to trigger reactive oxygen species (ROS) burst, upregulate p53 expression, elicit the loss of lysosomal membrane potential (LMP) and mitochondrial membrane potential (∆ψm). In addition, ROS induced by CQ plus SN-38 upregulated p53 levels by activating p38, conversely, p53 stimulated ROS. These results suggested that ROS and p53 reciprocally promoted each other's production and cooperated to induce CRC cell death. Moreover, we showed induction of ROS and p53 by the two agents provoked the loss of LMP and ∆ψm. Altogether, all results suggested that CQ synergistically sensitized human CRC cells with WT p53 to SN-38 through lysosomal and mitochondrial apoptotic pathway via p53-ROS cross-talk. Lastly, we showed that CQ could enhance CRC cells response to CPT-11 (a prodrug of SN-38) in xenograft models. Thus the combined treatment might represent an attractive therapeutic strategy for the treatment of CRC.


Assuntos
Cloroquina/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Irinotecano , Lisossomos/metabolismo , Lisossomos/patologia , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Oncotarget ; 8(14): 22414-22432, 2017 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-26461472

RESUMO

Here, we showed the antibiotic salinomycin (SAL) combined with GEF exerted synergistic cytotoxicity effects in colorectal cancer cells irrespective of their EGFR and KRAS status, with a relatively low toxicity to normal cells. Additionally, combination of the two drugs overcame Ras-induced resistance and the acquired resistance to GEF. Further, we identified a new potential mechanism of this cooperative interaction by showing that GEF and SAL acted together to enhance production of reactive oxygen species (ROS), loss of mitochondrial membrane potential (MMP) and lysosomal membrane potential (LMP). And the ROS contributed the loss of MMP and LMP. We also found that GEF and SAL acted in concert to induce apoptosis via a mitochondrial-lysosomal cross-talk and caspase-independent pathway triggered by cathepsin B and D. Lastly, SAL in combination with GEF sensitized GEF-resistant cells to GEF in a nude mouse xenograft model. This novel combination treatment might provide a potential clinical application to overcome GEF resistance in colorectal cancer.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Lisossomos/efeitos dos fármacos , Piranos/uso terapêutico , Quinazolinas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Catepsina B/metabolismo , Catepsina D/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Gefitinibe , Humanos , Lisossomos/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Transdução de Sinais , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Oncotarget ; 7(18): 26480-95, 2016 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-27034162

RESUMO

In this study, to investigate whether endoplastic reticulum (ER) stress correlated with FOXM1 in colorectal cancer, we analysed the mRNA levels of FOXM1 and ER stress markers HSPA5 and spliced XBP1 by qRT-PCR. FOXM1 mRNA levels were found to positively correlate with HSPA5 in colorectal cancer. However, no significant correlation between FOXM1 and spliced XBP1 mRNA levels was found. Theses results suggested the positive correlation between FOXM1 and HSPA5 in colorectal cancer was not associated with ER stress. Next, we provided evidences that FOXM1 promoted HSPA5 transcription by directly binding to and stimulating HSPA5 promoter. Moreover, a FOXM1-binding site mapped between -1019 and -1012 bp of the proximal HSPA5 promoter was identified. In addition, we found that enhancement of cell migration and invasion by FOXM1 was significantly attenuated by depletion of HSPA5 in colorectal cancer cell. Furthermore, FOXM1 triggered colorectal cancer cell migration and invasion was involved in activities of cell-surface HSPA5. Lastly, our results suggested FOXM1 facilitated the activities and expressions of MMP2 and 9 associated with cell-surface HSPA5 in colorectal cancer cells. Moreover, statistically significant positive correlations between FOXM1 and MMP2 mRNA expression, between HSPA5 and MMP2 were found in colorectal cancer tissue specimens. Together, our results suggested that FOXM1-HSPA5 signaling might be considered as a novel molecular target for designing novel therapeutic regimen to control colorectal cancer metastasis and progression.


Assuntos
Neoplasias Colorretais/patologia , Proteína Forkhead Box M1/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteínas de Choque Térmico/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Chaperona BiP do Retículo Endoplasmático , Proteína Forkhead Box M1/genética , Proteínas de Choque Térmico/genética , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Ativação Transcricional
7.
Oncotarget ; 6(28): 25402-17, 2015 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-26320172

RESUMO

The transcription factor Krüppel-like factor (KLF)8 plays an important role in the formation of several human tumors, including colorectal cancer. We recently identified four-and-a-half LIM protein 2 (FHL2) as a critical inducer of the epithelial-to-mesenchymal transition (EMT) and invasion. However, the molecular mechanism by which KLF8 affects FHL2-mediated tumor proliferation, EMT and metastasis remains unknown. Here, we showed that KLF8 overexpression promoted EMT and metastatic phenotypes. KLF8 expression was stimulated by transforming growth factor (TGF)-ß1. Moreover, KLF8 acted as a potential EMT inducer by stimulating vimentin expression and inducing a loss of E-cadherin in stable KLF8-transfected cells. KLF8 overexpression induced a strong increase in FHL2 expression, and a positive correlation between the expression patterns of KLF8 and FHL2 was observed in CRC cells. Promoter reporter and chromatin immunoprecipitation (ChIP) assays demonstrated that KLF8 directly bound to and activated the human FHL2 gene promoter. However, siRNA-mediated repression of FHL2 in KLF8-overexpressing cells reversed the EMT and the proliferative and metastatic phenotypes. In vivo, KLF8 promoted FHL2-mediated proliferation and metastasis via orthotopic implantation. Taken together, this work identified KLF8-induced FHL2 activation as a novel and critical signaling mechanism underlying human breast/colorectal cancer invasion and metastasis.


Assuntos
Carcinogênese , Movimento Celular , Neoplasias Colorretais/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Proteínas Musculares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Sítios de Ligação , Células CACO-2 , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Feminino , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Células HT29 , Humanos , Fatores de Transcrição Kruppel-Like , Proteínas com Homeodomínio LIM/genética , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Musculares/genética , Invasividade Neoplásica , Metástase Neoplásica , Fenótipo , Regiões Promotoras Genéticas , Ligação Proteica , Interferência de RNA , Proteínas Repressoras/genética , Transdução de Sinais , Fatores de Tempo , Fatores de Transcrição/genética , Transcrição Gênica , Transfecção
8.
Oncol Rep ; 34(3): 1221-30, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26133391

RESUMO

KLF8 is a member of the KLF transcription factor family that plays an important role in oncogenesis. However, the role of KLF8 in colorectal cancer remains unknown. The aims of the present study were to examine KLF8 expression in colorectal cancers, to determine the role of KLF8 in cell differentiation and to investigate the antiproliferative effect of KLF8 silencing. The expression of KLF8 and phospho-ERK proteins was analyzed, and the effects of KLF8 suppression on cell differentiation and growth were evaluated. In addition, the biological impact of KLF8 knockdown on colorectal cancer cells was investigated in vitro and in vivo. The expression of the KLF8 protein was higher in 10/14 (71.43%) fresh cancer tissues compared with adjacent normal tissues, and the blockade of ERK signaling by U0126 decreased the expression of KLF8 in a time- and dose-dependent manner. Furthermore, KLF8-siRNA induced the expression of carcinoembryonic antigen (CEA) and E-cadherin as well as the maturation of F-actin. KLF8 suppression inhibited serum-dependent, anchorage­dependent and -independent cell growth. Moreover, KLF8 silencing induced apoptosis and sensitized cancer cells to 5-fluorouracil (5-FU). A strong antitumorigenic effect by lenti-KLF8-shRNA, which was enhanced when combined with 5-FU treatment, was exerted in nude mice. Thus, KLF8 suppression induced cell differentiation and inhibited tumorigenesis.


Assuntos
Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas Repressoras/biossíntese , Animais , Antineoplásicos/farmacologia , Apoptose , Western Blotting , Diferenciação Celular/genética , Transformação Celular Neoplásica/genética , Imunofluorescência , Fluoruracila/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Fatores de Transcrição Kruppel-Like , Camundongos , Camundongos Nus , RNA Interferente Pequeno , Proteínas Repressoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção , Ensaios Antitumorais Modelo de Xenoenxerto
9.
PLoS One ; 10(6): e0129603, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26057751

RESUMO

It is well recognized that hypoxia-inducible factor 1 alpha (HIF-1α) is involved in cancer metastasis, chemotherapy and poor prognosis. We previously found that deferoxamine, a hypoxia-mimetic agent, induces epithelial-mesenchymal transition (EMT) in colorectal cancer. Therefore, here we explored a new molecular mechanism for HIF-1α contributing to EMT and cancer metastasis through binding to ZEB1. In this study, we showed that overexpression of HIF-1α with adenovirus infection promoted EMT, cell invasion and migration in vitro and in vivo. On a molecular level, HIF-1α directly binding to the proximal promoter of ZEB1 via hypoxia response element (HRE) sites thus increasing the transactivity and expression of ZEB1. In addition, inhibition of ZEB1 was able to abrogate the HIF-1α-induced EMT and cell invasion. HIF-1α expression was highly correlated with the expression of ZEB1 in normal colorectal epithelium, primary and metastatic CRC tissues. Interestingly, both HIF-1α and ZEB1 were positively associated with Vimentin, an important mesenchymal marker of EMT, whereas negatively associated with E-cadherin expression. These findings suggest that HIF-1α enhances EMT and cancer metastasis by binding to ZEB1 promoter in CRC. HIF-1α and ZEB1 are both widely considered as tumor-initiating factors, but our results demonstrate that ZEB1 is a direct downstream of HIF-1α, suggesting a novel molecular mechanism for HIF-1α-inducing EMT and cancer metastasis.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal , Proteínas de Homeodomínio/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Hepáticas/secundário , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Caderinas/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos Nus , Dados de Sequência Molecular , Elementos de Resposta/genética , Vimentina/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
10.
Oncol Rep ; 31(1): 111-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173124

RESUMO

Deferoxamine (DFX), a metal chelator, has been previously reported to induce hypoxia and hypoxia-inducible factor-1α (HIF-1α) expression. HIF-1α is a common inducer of epithelial-mesenchymal transition (EMT) in many solid tumors. However, the effect of DFX on cancer metastasis and the related mechanisms are not well established. In the present study, we aimed to ascertain whether DFX enhances EMT and cancer metastasis in colorectal cancer. After confirmation of DFX-inducing HIF-1α expression, we examined the effect of DFX on cell adhesion, migration and invasion abilities and found a positive effect on the above functions. Consequently, cell morphology, cell growth and expression of EMT markers were assessed in cells with or without DFX treatment. We found that cells exposed to DFX were more isolated. They were spindle-shaped and looked similar to fibroblast-like cells, accompanied by increased anchorage-independent growth. DFX-treated cells expressed E-cadherin and plakoglobin at a higher level, and vimentin and N-cadherin at a lower level, when compared with these levels in control cells. Furthermore, the expression of E-cadherin in the cell membrane was markedly decreased in DFX-treated cells. These results suggest that DFX promotes cancer migration and invasion via a process consistent with EMT in colorectal cancer.


Assuntos
Neoplasias Colorretais/patologia , Desferroxamina/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Sideróforos/farmacologia , Caderinas/biossíntese , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fibronectinas/metabolismo , Células HCT116 , Células HT29 , Humanos , Invasividade Neoplásica/patologia , Metástase Neoplásica , Transdução de Sinais/efeitos dos fármacos , Vimentina/biossíntese , Cicatrização/efeitos dos fármacos , gama Catenina/biossíntese
11.
Int J Oncol ; 43(5): 1618-26, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24008552

RESUMO

FHL2 (Four and a half LIM-only protein 2) has been identified as an oncogene in colon cancer and suppression of FHL2 induces cell differentiation and tumorigenesis in colon cancer cell lines. The aim of this study was to develop a novel and effective approach to knockdown FHL2, which can serve as a promising target of colon cancer therapy. Recombinant adeno-associated virus (rAAV) was generated bearing with FHL2-shRNA and transfected into LoVo cells. Cell cycle and growth were assessed. The interaction between FHL2 and G0/G1 cell cycle and growth was evaluated by flow cytometry, western blot analysis and WST-1 assay. We showed that suppression of FHL2 by rAAV-shRNA induced G0/G1 cell cycle arrest and inhibited cell growth. Apoptosis-related proteins and their activity was investigated at the same time. rAAV-FHL2­shRNA activated intrinsic and extrinsic apoptotic pathways and increased cell susceptibility to apoptotic stimuli by 5-FU. Moreover, a xenograft model was established to explore rAAV-FHL2-shRNA with 5-FU mediated tumorigenesis in vivo. A strong anti-tumorigenic effect of rAAV-FHL2-shRNA was shown in nude mice and this antitumor effect was enhanced when combined with 5-FU treatment. These findings implicate FHL2 as a cell cycle and growth modulator and thus inhibit apoptosis in colon cancer cells. rAAV-shRNA-FHL2 may serve as a novel and potent therapeutic or 5-FU co-therapeutic agent for colon cancer.


Assuntos
Neoplasias do Colo/terapia , Dependovirus/genética , Terapia Genética , Proteínas com Homeodomínio LIM/genética , Proteínas Musculares/genética , RNA Interferente Pequeno/genética , Fatores de Transcrição/genética , Animais , Apoptose , Western Blotting , Ciclo Celular , Proliferação de Células , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Feminino , Citometria de Fluxo , Humanos , Proteínas com Homeodomínio LIM/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Musculares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...