Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Small ; : e2401394, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709222

ABSTRACT

Transition metal silicates (TMSs) are attempted for the electrocatalyst of oxygen evolution reaction (OER) due to their special layered structure in recent years. However, defects such as low theoretical activity and conductivity limit their application. Researchers always prefer to composite TMSs with other functional materials to make up for their deficiency, but rarely focus on the effect of intrinsic structure adjustment on their catalytic activity, especially anion structure regulation. Herein, applying the method of interference hydrolysis and vacancy reserve, new silicate vacancies (anionic regulation) are introduced in cobalt silicate hydroxide (CoSi), named SV-CoSi, to enlarge the number and enhance the activity of catalytic sites. The overpotential of SV-CoSi declines to 301 mV at 10 mA cm-2 compared to 438 mV of CoSi. Source of such improvement is verified to be not only the increase of active sites, but also the positive effect on the intrinsic activity due to the enhancement of cobalt-oxygen covalence with the variation of anion structure by density functional theory (DFT) method. This work demonstrates that the feasible intrinsic anion structure regulation can improve OER performance of TMSs and provides an effective idea for the development of non-noble metal catalyst for OER.

2.
Biomed Pharmacother ; 166: 115373, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37647693

ABSTRACT

With the progression of tumor treatment, the 5-year survival rate of breast cancer is close to 90%. Cardiovascular toxicity caused by chemotherapy has become a vital factor affecting the survival of patients with breast cancer. Anthracyclines, such as doxorubicin, are still some of the most effective chemotherapeutic agents, but their resulting cardiotoxicity is generally considered to be progressive and irreversible. In addition to anthracyclines, platinum- and alkyl-based antitumor drugs also demonstrate certain cardiotoxic effects. Targeted drugs have always been considered a relatively safe option. However, in recent years, some random clinical trials have observed the occurrence of subclinical cardiotoxicity in targeted antitumor drug users, which may be related to the effects of targeted drugs on the angiotensin converting enzyme, angiotensin receptor and ß receptor. The use of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and beta-blockers may prevent clinical cardiotoxicity. This article reviews the toxicity and mechanisms of current clinical anti-breast cancer drugs and proposes strategies for preventing cardiovascular toxicity to provide recommendations for the clinical prevention and treatment of chemotherapy-related cardiomyopathy.


Subject(s)
Breast Neoplasms , Cardiomyopathies , Humans , Female , Cardiotoxicity , Breast Neoplasms/complications , Breast Neoplasms/drug therapy , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Doxorubicin/adverse effects , Anthracyclines
3.
JCO Precis Oncol ; 7: e2200463, 2023 03.
Article in English | MEDLINE | ID: mdl-36996375

ABSTRACT

PURPOSE: To investigate the efficacy of PD-1/PD-L1 inhibitors plus chemotherapy versus anti-PD-1/PD-L1 monotherapy in advanced microsatellite instability (MSI)/mismatch repair-deficient (dMMR) gastrointestinal cancers. METHODS: We retrospectively recruited patients with MSI/dMMR gastrointestinal cancer who received anti-PD-1/PD-L1 with or without chemotherapy and compared objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) of PD-1/PD-L1 inhibitor plus chemotherapy (chemo-anti-PD-1/PD-L1 group) and PD-1/PD-L1 inhibitor alone (anti-PD-1/PD-L1 group). Propensity score-based overlap weighting analysis was conducted to adjust the baseline covariable imbalance. Sensitivity analysis was performed to confirm the stability of the results by propensity score matching and multivariable Cox and logistic regression models. RESULTS: A total of 256 patients were eligible, with 68 and 188 receiving chemo-anti-PD-1/PD-L1 and anti-PD-1/PD-L1, respectively. The chemo-anti-PD-1/PD-L1 group showed significant improvements versus the anti-PD-1/PD-L1 group in ORR (61.8% v 38.8%; P = .001), DCR (92.6% v 74.5%; P = .002), PFS (median PFS [mPFS], not reached [NR] v 27.9 months; P = .004), and OS (median OS [mOS], NR v NR; P = .014). After overlap weighting, the improvements tended to be more significant with chemo-anti-PD-1/PD-L1 versus anti-PD-1/PD-L1 in ORR (62.5% v. 38.3%; P < .001), DCR (93.8% v 74.2%; P < .001), PFS (mPFS, NR v 26.0 months; P = .004), and OS (mOS, NR v NR; P = .010). These results were solidified through sensitivity analysis. CONCLUSION: Chemo-anti-PD-1/PD-L1 is superior to anti-PD-1/PD-L1 in MSI/dMMR gastrointestinal cancers with improved efficacy.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/genetics , Retrospective Studies , Microsatellite Instability , Colorectal Neoplasms/drug therapy
5.
Pharmacol Res ; 183: 106364, 2022 09.
Article in English | MEDLINE | ID: mdl-35901939

ABSTRACT

Pancreatic cancer (PC) is the most lethal malignancy worldwide due to its delayed diagnosis and limited treatment options. Despite great progress in clinical trials of immunotherapies for various cancers, their effectiveness in PC is very low, indicating that immune evasion is still a major obstacle to immunotherapy in PC. However, the mechanism of immune escape in PC is not fully understood, which substantially restricts the development of immunotherapy. As an important component of intercellular communication networks, extracellular vesicles (EVs) have attracted increasing attention in relation to immune escape. This review aims to provide a better understanding of the roles of EVs in tumor immune escape and the potential to expand their application in cancer immunotherapy. The relationship between PC and the tumor immune microenvironment is briefly introduced. Then, the mechanism by which EVs are involved in immune regulation is summarized, and the latest progress in determining the role of EVs in regulating PC immune escape is highlighted.


Subject(s)
Extracellular Vesicles , Neoplasms , Pancreatic Neoplasms , Humans , Immunotherapy , Neoplasms/pathology , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/therapy , Tumor Escape , Tumor Microenvironment , Pancreatic Neoplasms
6.
J Control Release ; 349: 679-698, 2022 09.
Article in English | MEDLINE | ID: mdl-35878728

ABSTRACT

Extracellular vesicles (EVs) are heterogeneous membranous vesicles secreted by living cells that are involved in many physiological and pathological processes as intermediaries for intercellular communication and molecular transfer. Recent studies have shown that EVs can regulate the occurrence and development of tumors by transferring proteins, lipids and nucleic acids to immune cells as signaling molecules. As a new diagnostic biomarker and drug delivery system, EVs have broad application prospects in immunotherapy. In addition, the breakthrough of nanotechnology has promoted the development and exploration of engineered EVs for immune-targeted therapy. Herein, we review the uniqueness of EVs in immune regulation and the engineering strategies used for immunotherapy and highlight the logic of their design through typical examples. The present situation and challenges of clinical transformation are discussed, and the development prospects of EVs in immunotherapy are proposed. The goal of this review is to provide new insights into the design of immune-regulatory EVs and expand their application in cancer immunotherapy.


Subject(s)
Extracellular Vesicles , Neoplasms , Nucleic Acids , Biomarkers/metabolism , Extracellular Vesicles/metabolism , Humans , Immunotherapy , Lipids , Neoplasms/metabolism , Neoplasms/therapy
7.
Front Cardiovasc Med ; 8: 758050, 2021.
Article in English | MEDLINE | ID: mdl-34957249

ABSTRACT

Cardiovascular disease is the leading cause of human death worldwide. Drug thrombolysis, percutaneous coronary intervention, coronary artery bypass grafting and other methods are used to restore blood perfusion for coronary artery stenosis and blockage. The treatments listed prolong lifespan, however, rate of mortality ultimately remains the same. This is due to the irreversible damage sustained by myocardium, in which millions of heart cells are lost during myocardial infarction. The lack of pragmatic methods of myocardial restoration remains the greatest challenge for effective treatment. Exosomes are small extracellular vesicles (EVs) actively secreted by all cell types that act as effective transmitters of biological signals which contribute to both reparative and pathological processes within the heart. Exosomes have become the focus of many researchers as a novel drug delivery system due to the advantages of low toxicity, little immunogenicity and good permeability. In this review, we discuss the progress and challenges of EVs in myocardial repair, and review the recent development of extracellular vesicle-loading systems based on their unique nanostructures and physiological functions, as well as the application of engineering modifications in the diagnosis and treatment of myocardial repair.

8.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34184089

ABSTRACT

Following the publication of the above article, an interested reader drew to the authors' attention that various of the data panels shown for the cell migration assay experiments in Figs. 2B and 3 appeared to show overlapping regions, such that they were not generated from discretely performed experiments.The authors have re-examined their original data, and realize that the figures in question were assembled incorrectly. In addition, the authors have also realized that certain of the western blotting data panels in Figs. 5 and 6 had likewise been assembled incorrectly. The corrected versions of Figs. 2, 3, 5 and 6 are shown on the next two pages. All these corrections were approved by all authors. The authors regret that these errors were included in the paper, and are grateful to the Editor of Molecular Medicine Reports for allowing them the opportunity to publish this corrigendum. They also wish to emphasize that the errors made during the compilation of the figures did not substantially alter any of the major conclusions reported in the study, and apologize to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 16: 2461-2468, 2017; DOI: 10.3892/mmr.2017.6905].

9.
Inflammation ; 44(5): 1762-1770, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33768402

ABSTRACT

Fourteen constituents were recently isolated from the roots of Dendropanax dentiger with cyclooxygenase-2 (COX-2) inhibitory effects. However, the effect of 14 constituents on rheumatoid arthritis (RA) and their action mechanism remain unclear. The study aimed to explore the anti-RA effect and potential mechanism of these constituents in tumor necrosis factor α (TNF-α)-stimulated human RA fibroblast-like synoviocytes (MH7A cells). The cell viability, nitric oxide (NO) production, inflammatory cytokine levels, and protein expressions were measured by cell counting kit-8 (CCK-8), Griess reagent, ELISA, and Western blot assays, respectively. Results showed that 14 constituents (40 µM) have no cytotoxicity for MH7A cells. Among them, two phenols including 3,4-dimethoxyphenyl-1-O-α-L-rhamnopyranosyl-(1→6)-O-ß-D-glucopyranoside (DRG) and 3,4-dimethoxyphenol-ß-D-apiofuranosyl-(1→6)-ß-D-glucopyranoside (DAG) were shown to significantly inhibit the NO production with IC50 values of 5.25±0.34 and 5.35±0.31 µM, respectively. They also remarkably decreased the release of interleukin (IL)-2, 6, 8, and interferon (IFN)-γ, as well as prominently reduced the phosphorylation protein levels of p65, IkBα, AKT, and JNK at a concentration of 10 µM. Taken together, DRG and DAG could inhibit TNF-α-induced inflammatory response through blocking NF-kB/AKT/JNK signaling pathways in MH7A cells, thus could be promising against RA and other inflammation-related agents.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Glycosides/pharmacology , MAP Kinase Signaling System/drug effects , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Tumor Necrosis Factor-alpha/toxicity , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Drugs, Chinese Herbal/isolation & purification , Glycosides/isolation & purification , Humans , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/metabolism , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Phenol/isolation & purification , Phenol/pharmacology , Proto-Oncogene Proteins c-akt/metabolism
10.
Open Med (Wars) ; 16(1): 419-427, 2021.
Article in English | MEDLINE | ID: mdl-33758783

ABSTRACT

FGF16 is implicated in the progression of some specific types of cancers, such as embryonic carcinoma, ovarian cancer, and liver cancer. Yet, the function of FGF16 in the development of lung cancer remains largely unexplored. In this study, we present the novel function of FGF16 and the regulation of miR-520b on FGF16 in lung cancer progression. In clinical lung cancer tissues, FGF16 is overexpressed and its high level is negatively associated with the low level of miR-520b. Furthermore, both the transcription and translation levels of FGF16 are restrained by miR-520b in lung cancer cells. For the regulatory mechanism investigation, miR-520b is able to directly bind to the 3'-untranslated region (3'UTR) of FGF16 mRNA, leading to its mRNA cleavage in the cells. Functionally, miR-520b reduces the growth of lung cancer and its inhibitor anti-miR520b is able to promote the growth through competing endogenous miR-520b. Moreover, FGF16 silence using RNA interference is capable of doing great damage to anti-miR-520b-accelerated growth of lung cancer. Thus, our finding indicates that FGF16 is a new target gene of miR-520b in lung cancer. For lung cancer, FGF16 may serve as a novel biomarker and miR-520b/FGF16 may be useful in clinical treatment.

11.
Front Pharmacol ; 11: 580064, 2020.
Article in English | MEDLINE | ID: mdl-33597870

ABSTRACT

Caffeoylquinic acids, coumarins and dicaffeoyl derivatives are considered to be three kinds of the most abundant bioactive components in Sarcandra glabra, an anti-inflammatory herb mainly found in Southern Asia. The combined anti-inflammatory effect of three typical constituents C + R + I (chlorogenic acid + rosmarinic acid + isofraxidin) from this plant has been investigated. The result implies that targeting the MAPK-NF-κB pathway would be one of the major mechanisms involved, using LPS stimulated RAW 264.7 cells as in vitro model and LPS-induced acute lung injury in mice as in vivo model. C + R + I can significantly suppress the levels of nitric oxide (NO), pro-inflammatory cytokines, and inhibit iNOS and COX-2 expression in LPS-treated RAW264.7 macrophage cells. Western blot analysis showed that C + R + I suppressed phosphorylation of NF-κB and MAPK, including phosphorylation of p65-NF-κB, IKB, ERK, JNK and P38. Besides, C + R + I suppressed MPO protein expression, but promoted SOD and HO-1 expression, and the related targets for C, R, and I were also predicted by molecular docking. This indicated that C + R + I could alleviate oxidative stress induced by LPS, which were further verified in the in vivo model of mice with acute lung injury through the measurement of corresponding inflammatory mediators and the analysis of immunehistochemistry.

12.
J Am Chem Soc ; 141(43): 17314-17321, 2019 10 30.
Article in English | MEDLINE | ID: mdl-31603315

ABSTRACT

Rhomboid proteases are intramembrane proteases that hydrolyze substrate peptide bonds within the lipid bilayer and are important for a wide range of biological processes. The bacterial intramembrane protease GlpG is one of the model systems for structural investigations of the rhomboid family. Two different models of substrate gating have been proposed, based on crystal structures of GlpG in detergent micelles. Here, we present a detailed investigation of enzymatically active GlpG in a native-like lipid environment using solid-state NMR spectroscopy. Proton-detected experiments confirm the presence of water molecules in the catalytic cavity. A secondary chemical shift analysis indicates a previously unobserved kink in the central part of the gating helix TM5. Dynamics measurements revealed a dynamic hotspot of GlpG at the N-terminal part of TM5 and the adjacent loop L4, indicating that this region is important for gating. In addition, relaxation dispersion experiments suggest that TM5 is in conformational exchange between an open and a closed conformation.


Subject(s)
DNA-Binding Proteins/chemistry , Endopeptidases/chemistry , Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Liposomes/chemistry , Membrane Proteins/chemistry , DNA-Binding Proteins/metabolism , Endopeptidases/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Magnetic Resonance Spectroscopy , Membrane Proteins/metabolism , Models, Molecular , Protein Conformation , Water/chemistry
13.
Mol Med Rep ; 16(3): 2461-2468, 2017 09.
Article in English | MEDLINE | ID: mdl-28677797

ABSTRACT

Lung cancer is a leading cause of cancer­associated mortality worldwide, including in developing countries such as China. Traditional Chinese medicine may provide a novel insight for the treatment of patients with lung cancer. The present study aimed to uncover the mechanism by which the Chinese herbal medicine, Fuzheng Kang­Ai (FZKA), functions on lung cancer cell metastasis. The results demonstrated that treatment with FZKA markedly inhibited cell viability, migration and invasion of lung cancer cells, as determined by cell viability and Transwell assays. Notably, the activity and expression of matrix metalloproteinase 9 (MMP9) was significantly inhibited by FZKA treatment on lung cancer cells, as determined by an MMP9 activity assay and western blot analysis. Furthermore, FZKA markedly inhibited epithelial­mesenchymal transition (EMT) of lung cancer cells by inhibiting the expression of the mesenchymal markers N­cadherin and vimentin. In addition, activation of the oncoprotein signal transducer and activator of transcription 3 (STAT3) was suppressed following treatment with FZKA. Conversely, overexpression of STAT3 was able to rescue MMP9 activity following FZKA treatment. The present study indicated that FZKA may inhibit lung cancer metastasis via the STAT3/MMP9 pathway and EMT, suggesting that FZKA may serve as a novel promising therapeutic strategy for the treatment of patients with late stage lung cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/drug therapy , Neoplasm Invasiveness/prevention & control , Signal Transduction/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness/pathology , STAT3 Transcription Factor/metabolism
14.
J Exp Clin Cancer Res ; 36(1): 56, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28420432

ABSTRACT

BACKGROUND: Grb2-associated binder 2 (Gab2) is a scaffolding protein that serves as a critical signaling amplifier downstream of tyrosine kinase receptors. Our previous study has shown that Gab2 induces epithelial-to-mesenchymal transition (EMT) and promotes metastasis in colorectal cancer (CRC). However, the role of Gab2 in CRC growth and angiogenesis remains unclear. METHODS: The expression of vascular endothelial growth factor (VEGF) in different colorectal tissues was detected by immunohistochemistry and qRT-PCR to evaluate its correlation with Gab2. Lentiviral vectors bearing Gab2 gene and its small interfering RNAs were constructed and transfected into CRC cell lines. The effects of Gab2 on the cell proliferation in vitro and tumorigenesis in vivo, were examined via CCK­8 assay, colony formation assay as well as tumorigenicity assay respectively. Moreover, to assess its potential role in tumor growth and angiogenesis, the expression of Ki67, CD34 and vascular endothelial growth factor receptor-2 (VEGFR2) were detected by immunohistochemistry in CRC cells tumors. Finally, we evaluated the impact of Gab2 on the expression of c-Myc and VEGF, and the probable effect of mechanistic targeted extracellular signal-regulated kinase (ERK) pathway in suppressing tumor growth and angiogenesis. RESULTS: Up-regulation of Gab2 expression was found to be positively correlated with VEGF in CRC tissues. Exogenous expression of Gab2 obviously promoted, whereas silencing of Gab2 inhibited, proliferation and clone formation of human CRC cells in vitro. Of note, Gab2 enhanced tumorigenesis and tumor growth in mouse xenografts with high Ki67 expression, and led to an increased vessel density with strong CD34 and VEGFR2 activity. In addition, elevated Gab2 expression obviously up-regulated the expression of VEGF, and stimulated the activation of its downstream genes, ERK1/2 and c-Myc in CRC cells. Instead, down-regulated Gab2 expression significantly reduced the levels of VEGF, and inhibited the transduction of ERK/c-Myc pathway. Finally, we revealed that mechanistic target of mitogen-activated protein kinase (MEK) could attenuate Gab2-induced tumor growth and angiogenesis via altering VEGF and c-Myc levels. CONCLUSIONS: The results from our study suggest that Gab2 promotes intestinal tumor growth and angiogenesis through upregulation of VEGF expression mediated by the MEK/ERK/c-Myc pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Ki-67 Antigen/metabolism , MAP Kinase Signaling System , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-myc/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
15.
J Ethnopharmacol ; 194: 918-929, 2016 Dec 24.
Article in English | MEDLINE | ID: mdl-27989877

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Chinese herbal medicine (CHM) Fuzheng Kang-Ai (FZKA for short) decoction has been used as adjuvant treatment strategies in lung cancer patients for decades. However, the molecular mechanism underlying the therapeutic potential especially in sensitizing the effect of EGFR-TKI gefitinib has not been well elucidated. MATERIALS AND METHODS: Cell viability was detected by MTT assay. Cell cycle distribution was detected by flow cytometry. Western blot were used to examine phosphorylation and protein levels of Akt, p65, p50 and MUC1. The mRNA level of MUC1 was measured by qRT-PCR. Transient transfection experiments were used to overexpression of Akt, p65 and MUC1. Tumor xenograft and bioluminescent imaging experiments were carried out to confirm the in vitro findings. RESULTS: Cell viability was inhibited by FZKA treatment and showed more significant when treated with FZKA and gefitinib in combine in lung cancer cells. FZKA induced the cell arrest at G0/G1 phase. Mechanistically, we showed that the phosphorylation of Akt, protein expressions of p65 and MUC1 were suppressed by FZKA and even more responses were observed in the FZKA and gefitinib combining. Overexpressed Akt overcame the effect of FZKA on p65 protein, and exogenously expressed p65 resisted the inhibitory effect of MUC1 protein expression by FZKA. On the contrary, while overexpressed MUC1 had no effect on p65 expression, it feedback increased phosphorylation of Akt, and more importantly, reversed the cell growth inhibition affected by FZKA. In line with the above, our results confirmed the synergistic effects of FZKA and gefitinib combination on tumor growth, the phosphorylation of Akt, and protein expression of p65 and MUC1 in vivo. CONCLUSION: This study shows that FZKA decoction inhibits the growth of NSCLC cells through Akt-mediated inhibition of p65, followed by reducing the expression of MUC1. More importantly, there is a synergistic effect of FZKA decoction and gefitinib combination with greater suppression. The positive feedback regulatory loop of MUC1 to Akt signaling pathway further added the important role of MUC1 in mediating the overall responses of FZKA decoction in this process. The in vitro and in vivo study provides an additional and a novel mechanism by which the FZKA decoction enhances the growth inhibition of gefitinib in gefitinib-resistant NSCLC cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Drugs, Chinese Herbal/pharmacology , Mucin-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drugs, Chinese Herbal/administration & dosage , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mucin-1/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Quinazolines/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism
16.
BMC Cancer ; 16(1): 791, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27729020

ABSTRACT

BACKGROUND: Angiogenesis is generally involved during the cancer development and hematogenous metastasis. Vascular endothelial growth factor (VEGF) and epidermal growth factor receptor (EGFR) are considered to have an important role in tumor-associated angiogenesis. However, the effects of simultaneously targeting on VEGF and EGFR on the growth and angiogenesis of colorectal cancer (CRC), and its underlying mechanisms remain unknown. METHODS: Immunohistochemical staining was used to detect the VEGF and EGFR expression in different CRC tissue specimens, and the correlation between VEGF/EGFR expression with the clinicopathologic features was analyzed. Cell counting kit­8 (CCK-8) and transwell assays were used to assess the cellular proliferation and invasion of CRC cells after treated with anti-VEGF antibody and/or anti-EGFR antibody in vitro, respectively. Moreover, in vivo tumor formation was performed on nude mice model, and the tumor microvessel density (MVD) was determined by anti-CD34 staining in different groups. Finally, we evaluated the impact of anti-VEGF antibody and/or anti-EGFR antibody on the activation of downstream signaling effectors using western blot. RESULTS: VEGF and EGFR were upregulated in CRC tissues, and their expression levels were correlated with hepatic metastasis. Blockage on VEGF or EGFR alone could inhibit the cellular proliferation and metastasis while their combination could reach a good synergism in vitro. In addition, in vivo xenograft mice model demonstrated that the tumor formation and angiogenesis were strongly suppressed by combination treatment of anti-VEGF and anti-EGFR antibodies. Besides, the combination treatment significantly reduced the activation of AKT and ERK1/2, but barely affected the activation of c-Myc, NF-κB/p65 and IκBα in CRC cells tumors. Interestingly, anti-VEGF antibody or anti-EGFR antibody alone could attenuate the phosphorylation of STAT3 as compared with negative control group, whereas the combined application not further suppressed but at least partially restored the activation of STAT3 in vivo. CONCLUSIONS: Simultaneous targeting on VEGF and EGFR does show significant inhibition on CRC tumor growth and angiogenesis in mice model, and these effects are mainly attributed to suppression of the AKT and ERK signaling pathways.


Subject(s)
Antibodies, Monoclonal/pharmacology , Colorectal Neoplasms/metabolism , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adult , Aged , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neovascularization, Pathologic/drug therapy , Tumor Burden , Xenograft Model Antitumor Assays
17.
J Exp Clin Cancer Res ; 35(1): 112, 2016 07 16.
Article in English | MEDLINE | ID: mdl-27421653

ABSTRACT

BACKGROUND: Polyphyllin I (PPI), a bioactive phytochemical extracted from the Rhizoma of Paris polyphylla, has been reported to exhibit anti-cancer activity. However, the detailed mechanism underlying this remains to be elucidated. METHODS: Cell viability and cell cycle distribution were measured using a 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) and flow cytometry assays, respectively. The expression of enhancer of zeste homolog 2 (EZH2) mRNA was measured by quantitative real time PCR (qRT-PCR). Western blot analysis was performed to examine the phosphorylation and protein expression of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), p65, DNA methyltransferase 1 (DNMT1) and EZH2. Exogenous expression of p65, DNMT1, and EZH2 were carried out by transient transfection assays. Promoter activity of EZH2 gene was determined using Secrete-Pair Dual Luminescence Assay Kit. A xenografted tumor model in nude mice and bioluminescent imaging system were used to further test the effect of PPI in vivo. RESULTS: We showed that PPI significantly inhibited growth and induced cell cycle arrest of non-small cell lung cancer (NSCLC) cells in a dose-dependent manner. Mechanistically, we found that PPI increased the phosphorylation of SAPK/JNK, reduced protein expression of p65 and DNMT1. The inhibitor of SAPK/JNK (SP600125) blocked the PPI-inhibited p65 and DNMT1 protein expression. Interestingly, exogenously expressed p65 overcame PPI-inhibited protein expression of DNMT1. Moreover, PPI reduced EZH2 protein, mRNA, and promoter activity; overexpression of EZH2 resisted the PPI-inhibited cell growth, and intriguingly, negative feedback regulation of SAPK/JNK signaling. Finally, exogenous expression of DNMT1 antagonized the PPI-suppressed EZH2 protein expression. Consistent with this, PPI inhibited tumor growth, protein expression levels of p65, DNMT1 and EZH2, and increased phosphorylation of SAPK/JNK in vivo. CONCLUSION: Our results show that PPI inhibits growth of NSCLC cells through SAPK/JNK-mediated inhibition of p65 and DNMT1 protein levels, subsequently; this results in the reduction of EZH2 gene expression. The interactions among p65, DNMT1 and EZH2, and feedback regulation of SAPK/JNK by EZH2 converge on the overall responses of PPI. This study reveals a novel mechanism for regulating EZH2 gene in response to PPI and suggests a new strategy for NSCLC associated therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Diosgenin/analogs & derivatives , Enhancer of Zeste Homolog 2 Protein/genetics , Lung Neoplasms/drug therapy , MAP Kinase Kinase 4/metabolism , Transcription Factor RelA/metabolism , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Diosgenin/administration & dosage , Diosgenin/pharmacology , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phosphorylation , Xenograft Model Antitumor Assays
18.
J Exp Clin Cancer Res ; 35: 5, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26754532

ABSTRACT

BACKGROUND: Grb2-associated binder 2 (Gab2), a scaffolding adaptor protein, has recently been implicated in cancer progression. However, the role of Gab2 in the progression and metastasis of colorectal cancer (CRC) remains unclear. METHODS: Gab2 expression was assessed in CRC patient specimens as well as in CRC cell lines. Recombinant lentivirus vector containing Gab2 gene and its small interfering RNAs were constructed and introduced into CRC cells. Cell migration and invasion ability were evaluated by transwell assays in vitro, and in vivo metastasis was performed on nude mice model. Moreover, the expression of Gab2 and epithelial-to-mesenchymal transition (EMT)-associated proteins (E-cadherin and vimentin) were assessed by western blot and qRT-PCR in CRC cells to evaluate the correlation between Gab2 and EMT. Finally, we evaluated the impact of Gab2 on the activation of its downstream signaling effectors, and furthermore the effects of these pathways on Gab2 induced-EMT were also detected. RESULTS: We confirmed that increased Gab2 expression correlated with higher tumor node metastasis stage and highly invasive CRC cell lines. Ectopic expression of Gab2 promoted metastasis of CRC cells, whereas silencing of Gab2 resulted in inhibited metastasis both in vitro and in vivo. Overexpression of Gab2 in CRC cells induced EMT, whereas knockdown of Gab2 had the opposite effect. Furthermore, upregulation of Gab2 expression obviously stimulated the activation of extracellular signal-regulated kinase-1/2 (ERK1/2), and increased the expression of matrix metalloproteinase-7 (MMP7) and matrix metalloproteinase-9 (MMP9) in CRC cells. Conversely, downregulation of Gab2 expression significantly decreased the activation of ERK1/2, and inhibited MMP7 and MMP9 expression. U0126, an inhibitor of mitogen-activated protein kinase (MEK), can reverse the effects of Gab2 on EMT. CONCLUSIONS: Our work highlights that Gab2 induces EMT through the MEK/ERK/MMP pathway, which in turn promotes intestinal tumor metastasis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Metalloproteases/metabolism , Animals , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , MAP Kinase Signaling System , Mice , Neoplasm Metastasis , Neoplasm Transplantation
SELECTION OF CITATIONS
SEARCH DETAIL
...