Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters











Publication year range
1.
Sci Rep ; 14(1): 18107, 2024 08 05.
Article in English | MEDLINE | ID: mdl-39103425

ABSTRACT

The limitations associated with conventional cancer treatment modalities, particularly for breast cancer, underscore the imperative for developing safer and more productive drug delivery systems. A promising strategy that has emerged is the combination of chemotherapy with gas therapy. We synthesized curcumin-loaded amorphous calcium carbonate nanoparticles (Cur-CaCO3) via a gas diffusion reaction in the present study. Subsequently, a "one-step" ethanol injection method was employed to fabricate lipid-coated calcium carbonate nanoparticles (Cur-CaCO3@LA-Lip) loaded with L-arginine, aimed at harnessing the synergistic effects of chemotherapy and nitric oxide to enhance antitumor efficacy. Transmission electron microscopy analysis revealed that Cur-CaCO3@LA-Lip nanoparticles were subspherical with a distinct lipid layer encapsulating the periphery. Fourier transform infrared spectroscopy, X-ray powder diffraction, and differential scanning calorimetry results confirmed the successful synthesis of Cur-CaCO3@LA-Lip. The nanoparticles exhibited significant drug loading capacities of 8.89% for curcumin and 3.1% for L-arginine. In vitro and in vivo assessments demonstrated that Cur-CaCO3@LA-Lip nanoparticles facilitated sustained release of curcumin and exhibited high cellular uptake, substantial tumor accumulation, and excellent biocompatibility. Additionally, the nanoparticles showed robust cytotoxicity and potent antitumor efficacy, suggesting their potential as a formidable candidate for breast cancer therapy.


Subject(s)
Breast Neoplasms , Curcumin , Nanoparticles , Nitric Oxide , Curcumin/pharmacology , Curcumin/administration & dosage , Curcumin/chemistry , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Nanoparticles/chemistry , Animals , Humans , Nitric Oxide/administration & dosage , Nitric Oxide/metabolism , Nitric Oxide/chemistry , Mice , Lipids/chemistry , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Calcium Carbonate/chemistry , Cell Line, Tumor , Drug Delivery Systems/methods , Drug Carriers/chemistry , Arginine/chemistry
2.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(3): 304-312, 2024 Jun 01.
Article in English, Chinese | MEDLINE | ID: mdl-39049649

ABSTRACT

OBJECTIVES: This study aims to investigate the influence of glucose regulated protein (GRP) 78 on osteoblast differentiation in periodontal ligament fibroblasts (PDLFs) under cyclic mechanical stretch and determine the underlying mechanism. METHODS: FlexCell 5000 cell mechanical device was applied to simulate the stress environment of orthodontic teeth. GRP78High and GRP78Low subpopulation were obtained by flow sorting. Gene transfection was performed to knockdown GRP78 and c-Src expression and overexpress c-Src. Western blot analysis was used to detect the protein expression of Runt-related gene 2 (RUNX2), Osterix, osteocalcin (OCN), and osteopontin (OPN). Immunoprecipitation assay was used to determine the interaction of GRP78 with c-Src. The formation of cellular mineralized nodules was determined by alizarin red staining. RESULTS: GRP78 was heterogeneously expressed in PDLFs, and GRP78High and GRP78Low subpopulations were obtained by flow sorting. The osteogenic differentiation ability and phosphorylation level of c-Src kinase in the GRP78High subpopulation were significantly increased compared with those in GRP78Low subpopulation after cyclic mechanical stretch (P<0.05). GRP78 interacted with c-Src in PDLFs. The overexpression c-Src group showed significantly increased osteogenic differentiation ability than the vector group (P<0.05), and the sic-Src group showed significantly decreased osteogenic differentiation ability (P<0.05) after cyclic mechanical stretch. CONCLUSIONS: GRP78 upregulates c-Src expression by interacting with c-Src kinase and promotes osteogenic differentiation under cyclic mechanical stretch in PDLFs.


Subject(s)
Cell Differentiation , Heat-Shock Proteins , Osteoblasts , Periodontal Ligament , Proto-Oncogene Proteins pp60(c-src) , Signal Transduction , Stress, Mechanical , Humans , Core Binding Factor Alpha 1 Subunit/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Endoplasmic Reticulum Chaperone BiP/metabolism , Fibroblasts/metabolism , Heat-Shock Proteins/metabolism , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis , Osteopontin/metabolism , Periodontal Ligament/metabolism , Periodontal Ligament/cytology , Phosphorylation , src-Family Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism
3.
Molecules ; 29(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38893528

ABSTRACT

The existing kinase inhibitors for hepatocellular carcinoma (HCC) have conferred survival benefits but are hampered by adverse effects and drug resistance, necessitating the development of novel agents targeting distinct pathways. To discover potent new anti-HCC compounds, we leveraged scaffold hopping from Sorafenib and introduced morpholine/piperidine moieties to develop ureido-substituted 4-phenylthiazole analogs with optimized physicochemical properties and binding interactions. Notably, compound 27 exhibited potent cytotoxicity against HepG2 cells (IC50 = 0.62 ± 0.34 µM), significantly exceeding Sorafenib (IC50 = 1.62 ± 0.27 µM). Mechanistic investigations revealed that compound 27 potently inhibited HCC cell migration and colony formation, and it induced G2/M arrest and early-stage apoptosis. Kinase profiling revealed IGF1R as a key target, which compound 27 potently inhibited (76.84% at 10 µM). Molecular modeling substantiated compound 27's strong binding to IGF1R via multiple hydrogen bonds. Computational predictions indicate favorable drug-like properties for compound 27. These findings provide a promising drug candidate for the treatment of HCC patients.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Protein Kinase Inhibitors , Receptor, IGF Type 1 , Thiazoles , Humans , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/metabolism , Cell Proliferation/drug effects , Hep G2 Cells , Thiazoles/chemistry , Thiazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/chemical synthesis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Cell Movement/drug effects , Structure-Activity Relationship , Molecular Docking Simulation , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Molecular Structure , Cell Line, Tumor , Sorafenib/pharmacology , Sorafenib/chemistry , Models, Molecular
4.
ACS Omega ; 7(44): 39760-39771, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36385800

ABSTRACT

Oral cancer is one of the most common malignancies of the head and neck, and approximately 90% of oral cancers are oral squamous cell carcinomas (OSCCs). The purinergic P2Y2 receptor is upregulated in breast cancer, pancreatic cancer, colorectal cancer, and liver cancer, but its role in OSCC is still unclear. Here, we examined the effects of P2Y2 on the invasion and migration of oral cancer cells (SCC15 and CAL27). The BALB/c mouse model was used to observe the involvement of P2Y2 with tumors in vivo. P2Y2, Src, and EGFR are highly expressed in OSCC tissues and cell lines. Stimulation with ATP significantly enhanced cell invasion and migration in oral cancer cells, and enhanced the activity of Src and EGFR protein kinases, which is mediated by the PI3K/AKT signaling pathway. P2Y2 knockdown attenuated the above ATP-driven events in vitro and in vivo. The PI3K/AKT signaling pathway was blocked by Src or EGFR inhibitor. Extracellular ATP activates the PI3K/AKT pathway through the P2Y2-Src-EGFR axis to promote OSCC invasion and migration, and thus, P2Y2 may be a potential novel target for antimetastasis therapy.

5.
Int J Biochem Cell Biol ; 153: 106316, 2022 12.
Article in English | MEDLINE | ID: mdl-36280040

ABSTRACT

Notum, which belongs to the α/ß hydrolase family, is a deacylated extracellular protein that regulates the Wnt signaling pathway. Studies have found that Notum participates in the progression of colorectal cancer and hepatocellular carcinoma, but its role in oral squamous cell carcinoma (OSCC) is currently unclear. This study aimed to explore the role of Notum in regulating OSCC and further reveal the underlying mechanisms. Various approaches including bioinformatics analysis, enzyme-linked immunosorbent assay and immunohistochemical staining were used to detect the expression of Notum in OSCC cells and tissues. Cell counting kit-8 assay, clone formation assay, wound healing assay, transwell assay and in-gel zymography assay were explored to evaluate the regulation of Notum in OSCC proliferation and migration. Hoechst 33342/PI assay, cell immunofluorescence, flow cytometry and in vivo tumorigenesis experiment were applied for OSCC apoptosis. Real-time quantitative polymerase chain reaction analysis was performed for mRNA level while western blotting was conducted to detect protein expression. The results showed that Notum was highly expressed in OSCC tissues and cells, and Notum promoted the proliferation and migration of OSCC cells while it inhibited their apoptosis. Furthermore, signaling pathway analysis showed that Notum led to potential pro-survival of OSCC through crosstalk between sonic hedgehog (Shh) and Wnt/ß-catenin signaling via phosphorylation of glycogen synthase kinase-3 beta. These results will help to elucidate the mechanism and also provide new ideas for targeted treatment of OSCC.


Subject(s)
Esterases , Mouth Neoplasms , Squamous Cell Carcinoma of Head and Neck , Humans , beta Catenin/genetics , beta Catenin/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Esterases/genetics , Esterases/metabolism , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Wnt Signaling Pathway/genetics
6.
Int J Biol Sci ; 18(2): 491-506, 2022.
Article in English | MEDLINE | ID: mdl-35002505

ABSTRACT

Oral squamous cell carcinoma (OSCC) refers to the malignant tumor of the head and neck with a highest morbidity. It exhibits a poor prognosis and unsatisfactory treatment partially attributed to delayed diagnosis. As indicated from existing reports, the protein histidine phosphatase LHPP acts as a vital factor in tumorigenesis in liver, lung, bladder, breast and pancreatic tumor tissues. Thus far, the functional mechanism of LHPP in OSCC remains unclear. DGE analysis, OSCC cell lines and OSCC cases were found that LHPP was down-regulated in OSCC tissues and cells compared with that in normal oral mucosa tissues and cells, and was closely related to OSCC differentiation. Cell counting Kit 8 test, EdU proliferation test, scratches test, invasion test, monoclonal formation test, mouse xenograft tumor model, HE staining and immunohistochemistry showed that LHPP inhibited OSCC growth, proliferation and migration in vivo and in vitro. GO and KEGG enrichment analysis, LHPP transcription factor analysis and flow cytometry found that LHPP promotes the apoptosis of OSCC by decreasing the transcriptional activity of p-PI3K and p-Akt. Finally, our results suggested that LHPP inhibited the progression of OSCC through the PI3K/AKT signaling pathway, indicating that LHPP may be a new target for the treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Inorganic Pyrophosphatase/biosynthesis , Mouth Neoplasms/metabolism , Signal Transduction/physiology , Animals , Apoptosis/physiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Genes, Tumor Suppressor , Humans , Inorganic Pyrophosphatase/genetics , Inorganic Pyrophosphatase/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Mouth Neoplasms/genetics , Mouth Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xenograft Model Antitumor Assays
7.
Front Pharmacol ; 12: 615889, 2021.
Article in English | MEDLINE | ID: mdl-33716742

ABSTRACT

The multireceptor tyrosine kinase inhibitor sorafenib is a Food and Drug Administration-approved first-line drug for the treatment of advanced liver cancer that can reportedly extend overall survival in patients with advanced hepatocellular carcinoma (HCC). Primary and acquired resistance to sorafenib are gradually increasing however, leading to failure of HCC treatment with sorafenib. It is therefore crucial to study the potential mechanism of sorafenib resistance. The results of the current study indicate that neurite outgrowth inhibitor protein B receptor (NgBR) is overexpressed in cultured sorafenib-resistant cells, and that its expression is negatively correlated with the sensitivity of liver cancer cells to sorafenib. Artesunate can inhibit the expression of NgBR, and it may block sorafenib resistance. Herein we report that sorafenib treatment in combination with artesunate overcomes HCC resistance to sorafenib alone in a cell culture model.

8.
Aging (Albany NY) ; 13(3): 4242-4257, 2021 01 20.
Article in English | MEDLINE | ID: mdl-33495407

ABSTRACT

Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors worldwide, and its prognosis is still not optimistic. Oxaliplatin is a type of platinum chemotherapeutic agent, but its treatment effects on OSCC and molecular mechanisms have not been fully elucidated. Parthanatos, a unique form of cell death, plays an important role in a variety of physiological and pathological processes. This study aims to investigate whether oxaliplatin inhibits OSCC by inducing parthanatos. Our results showed that oxaliplatin inhibited the proliferation and migration of OSCC cells in vitro, and also inhibited the tumorigenesis in vivo. Further experiments proved that oxaliplatin induced parthanatos in OSCC cells, characterized by depolarization of the mitochondrial membrane potential, up-regulation of PARP1, AIF and MIF in the nucleus, as well as the nuclear translocation of AIF. Meanwhile, PARP1 inhibitor rucaparib and siRNA against PARP1 attenuated oxaliplatin-induced parthanatos in OSCC cells. In addition, we found that oxaliplatin caused oxidative stress in OSCC cells, and antioxidant NAC not only relieved oxaliplatin-induced overproduction of reactive oxygen species (ROS) but also reversed parthanatos caused by oxaliplatin. In conclusion, our results indicate that oxaliplatin inhibits OSCC by activating PARP1-mediated parthanatos through increasing the production of ROS.


Subject(s)
Antineoplastic Agents/pharmacology , Mitochondria/drug effects , Mouth Neoplasms/metabolism , Oxaliplatin/pharmacology , Parthanatos/drug effects , Poly (ADP-Ribose) Polymerase-1/drug effects , Reactive Oxygen Species/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Cell Line, Tumor , Glutathione/drug effects , Glutathione/metabolism , Humans , Mice , Mitochondria/metabolism , Mouth Neoplasms/pathology , Neoplasm Transplantation , Squamous Cell Carcinoma of Head and Neck/pathology , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Xenograft Model Antitumor Assays
9.
Shanghai Kou Qiang Yi Xue ; 30(6): 573-578, 2021 Dec.
Article in Chinese | MEDLINE | ID: mdl-35587009

ABSTRACT

PURPOSE: To investigate the effect of Met kinase inhibitor BMS-777607 on proliferation and apoptosis of tongue squamous cell carcinoma cell line CAL27. METHODS: The effect of BMS-777607 on proliferation of CAL27 was detected by MTT method, clone formation assay and EdU cell imaging. Morphological changes of apoptosis of CAL27 cells induced by BMS-777607 were observed by Heochst33342 staining. JC-1 staining was used to detect the changes of mitochondrial membrane potential of CAL27 cells treated with BMS-777607. Western blot was used to detect the effect of BMS-777607 on the expression of proliferation protein Akt, p-Akt and apoptosis-related proteins Bcl-2, Cleaved caspase-3, Bax and Parp in CAL27 cells. The data were analyzed using SPSS 22.0 software package. RESULTS: BMS-777607 inhibited proliferation and promoted apoptosis of CAL27 cells in a concentration-dependent manner(P<0.05). It also inhibited the expression of Bcl-2 and p-Akt and promoted the expression of Bax, Cleaved caspase-3 and Parp protein (P<0.05). CONCLUSIONS: BMS-777607 can inhibit proliferation and promote apoptosis of CAL27 cells.


Subject(s)
Carcinoma, Squamous Cell , Tongue Neoplasms , Aminopyridines , Apoptosis , Apoptosis Regulatory Proteins , Carcinoma, Squamous Cell/metabolism , Caspase 3/pharmacology , Caspase 3/therapeutic use , Cell Line, Tumor , Cell Proliferation , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/pharmacology , Proto-Oncogene Proteins c-bcl-2/therapeutic use , Pyridones , Tongue , Tongue Neoplasms/drug therapy , bcl-2-Associated X Protein/pharmacology
10.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 38(5): 495-501, 2020 Oct 01.
Article in Chinese | MEDLINE | ID: mdl-33085231

ABSTRACT

OBJECTIVE: This study aims to study the effect of the enhancer of zeste homolog 2 (EZH2) inhibitor GSK126 on the proliferation and apoptosis of human tongue squamous cell carcinoma cells in vitro and explore its related mechanisms in order to obtain insights into the clinical treatment of tongue squamous cell carcinoma. METHODS: Different concentrations of GSK126 were applied to CAL-27 cells of tongue squamous cell carcinoma, and the effects of drugs on cell proliferation were detected through methyl thiazolyl tetrazolium (MTT) assay, colony formation assay, and 5-ethynyl-2'-deoxyuridine (EdU) fluorescence staining. Hoechst33342 fluorescence staining and the JC-1 method were used in observing apoptosis. The expression levels of extracellular regulated protein kinases (ERK), phospho-extracellular regulated protein kinases (p-ERK), Bax, Bcl-2, and Cleaved caspase-9 in Cal-27 cells were detected through Western blot. RESULTS: GSK126 inhibited CAL-27 cell proliferation and promoted apoptosis. GSK126 down-regulated the expression of p-ERK and Bcl-2 and increased the expression of Bax and Cleaved caspase-9 (P<0.05). CONCLUSIONS: GSK126 can inhibit the proliferation of CAL-27 cells in tongue squamous cell carcinoma and promote its apoptosis, and the related mechanism may be associated with the inhibition of the MEK/ERK signaling pathway and activation of the Bax/Bcl-2 pathway.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Tongue Neoplasms , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Humans , Indoles , Pyridones , Tongue Neoplasms/drug therapy
11.
Oncotarget ; 8(5): 7502-7520, 2017 Jan 31.
Article in English | MEDLINE | ID: mdl-27447746

ABSTRACT

Here, BTG1 overexpression inhibited proliferation, induced differentiation, autophagy, and apoptosis in colorectal cancer cells (p<0.05). BTG1 overexpression reduced mitochondrial membrane potential and caused senescence in HCT-116 transfectants (p<0.05). BTG1-induced G2 arrest might be related to Cyclin B1 and Cdc25B hypoexpression in HCT-15 transfectants, while G1 arrest in HCT-116 transfectants overexpressing p21 and p27. BTG1 overexpression decreased the expression of Bcl-2, Bcl-xL, XIAP, Akt1 or survivin and increased the expression of Bax or p53 in colorectal cancer cells. BTG1-induced autophagy was dependent on Beclin-1 expression. BTG1 overexpression might weaken ß-catenin pathway in colorectal cancer cells. The chemosensitivity of BTG1 transfectants to paclitaxel, cisplatin, MG132 or SAHA was positively correlated with its apoptotic induction. There was a lower expression level of BTG1 in cancer than matched non-neoplastic mucosa by RT-PCR (p<0.05), while versa for Western blot and immunohistochemical data (p<0.05). BTG1 overexpression significantly suppressed the growth of HCT-15 and HCT-116 via inhibiting proliferation, inducing apoptosis and autophagy in nude mice. Up-regulated BTG1 expression plays an important role in colorectal carcinogenesis as a potential biomarker. BTG1 expression might reverse aggressive phenotypes, so it might be employed as a target of gene therapy for colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Genetic Therapy/methods , Neoplasm Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Signal Transduction , Time Factors , Transfection , beta Catenin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Curr Pharm Biotechnol ; 17(13): 1117-1125, 2016.
Article in English | MEDLINE | ID: mdl-27587021

ABSTRACT

Physalis pubescens L. (P. pubescens) is an edible plant used in folk medicine in China. There is traditional, but not scientific, evidence for the anti-tumour effects of P. pubescens. This study aimed to identify whether, or not, antioxidants rich in phenols and flavonoids from fruits and calyxes of P. pubescens can be the candidates for further development of an anti-hepatoma fraction, and if such biological effects coupled with reactive oxygen species (ROS) changes, can provide a direction for subsequent biological action. The effects of calyx-origin (or fruit-origin) total phenol and flavonoid (CTPF or FTPF) from P. pubescens on Malhavu cell viability were evaluated by using a counting-kit-8 (CCK-8) method. Morphological characterisation of cells was undertaken and the structures were photographed (200 × magnification) using Hoechst 3348 staining after exposure to different concentrations of CTPF or FTPF. Induced-apoptosis activity was determined using flow cytometry (FC) after Annexin VFITC/ PI staining. The corresponding ROS changes in Malhavu cells were observed and quantified by the uploading of 2', 7'-dichlorofluorescin diacetate (DCFH-DA). Anti-oxidation was evaluated by a cellular oxidation-stress model and chemical assessments for DPPH, hydroxyl radial, super-oxide radicals, and reducing power. Result shows that CTPF led to significant anti-proliferation in a time- and dosedependent manner. However, FTPF promoted cell viability at 100-1000 µg/mL with a dose-response manner in 24 h. With the extension of exposure time to 48 h, the cell viability did not increase with the growth of FTPF. Morphological characterisation and FC assay both demonstrated that CTPF, and not FTPF possessed induced-apoptotic activity. CTPF potentially induced cell apoptosis by promoting oxidative stress. FTPF indicated pro-oxidation at a concentration of 10 µg/mL and anti-oxidation capabilities at higher concentrations. ROS scavenging assay by oxidation-stress model indicated that CTPF (10 - 400 µg/mL) had ROS inhibitory capacity (R2 = 0.5156, p < 0.0001). FTPF (10 - 100 µg/mL) boosted the level of ROS (p < 0.0001) and inhibited the generation of ROS at 100-400 µg/mL (R2 = 0.5951, p < 0.0001). CTPF is a potential candidate requiring further exploration for the development of antihepatoma ingredients. The down-regulation of cell viability was related to production and reduction of cellular ROS.


Subject(s)
Antioxidants/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Physalis/chemistry , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Flavonoids/pharmacology , Humans , Liver Neoplasms/pathology , Oxidative Stress/drug effects , Phenols/pharmacology , Reactive Oxygen Species/metabolism
13.
Oncotarget ; 7(34): 54596-54615, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27409347

ABSTRACT

ING5 can interact with p53, thereby inhibiting cell growth and inducing apoptosis. We found that ING5 overexpression not only inhibited proliferation, migration, and invasion, but also induced G2 arrest, differentiation, autophagy, apoptosis, glycolysis and mitochondrial respiration in lung cancer cells. ING5 transfection up-regulated the expression of Cdc2, ATG13, ATG14, Beclin-1, LC-3B, AIF, cytochrome c, Akt1/2/3, ADFP, PFK-1 and PDPc, while down-regulated the expression of Bcl-2, XIAP, survivin,ß-catenin and HXK1. ING5 transfection desensitized cells to the chemotherapy of MG132, paclitaxel, and SAHA, which paralleled with apoptotic alteration. ING5 overexpression suppressed the xenograft tumor growth by inhibiting proliferation and inducing apoptosis. ING5 expression level was significantly higher in normal tissue than that in lung cancer at both protein and mRNA levels. Nuclear ING5 expression was positively correlated with ki-67 expression and cytoplasmic ING5 expression. Cytoplasmic ING5 expression was positively associated with lymph node metastasis, and negatively with age, lymphatic invasion or CPP32 expression. ING5 expression was different in histological classification: squamous cell carcinoma > adenocarcinoma > large cell carcinoma > small cell carcinoma. Taken together, our data suggested that ING5 downregulation might involved in carcinogenesis, growth, and invasion of lung cancer and could be considered as a promising marker to gauge the aggressiveness of lung cancer. It might be employed as a potential target for gene therapy of lung cancer.


Subject(s)
Down-Regulation , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , A549 Cells , Aged , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Humans , Hydroxamic Acids/pharmacology , Kaplan-Meier Estimate , Leupeptins/pharmacology , Lung Neoplasms/classification , Lung Neoplasms/drug therapy , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Targeted Therapy/methods , Paclitaxel/pharmacology , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Vorinostat , Xenograft Model Antitumor Assays
14.
Oncol Lett ; 10(2): 653-660, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26622549

ABSTRACT

microRNA-34a (miRNA/miR-34a) functions as a tumor suppressor gene in gastric cancer and may be involved in system-wide regulatory networks. To clarify the expression of all predicted target genes of this miRNA, a comprehensive and systematic analysis of miR-34a-target genes in gastric cancer was conducted in the present study. In the initial analysis, the potential functions, pathways and networks of gastric cancer-associated molecules and miR-34a targets were identified. In the final integrative analysis of gastric cancer-associated miR-34a targets, 30 hub genes were identified using overlap calculations, indicating that miR-34a may be significant in the development and progression of gastric cancer through the Smad signaling pathway, the cell cycle, the mitogen-activated protein kinase signaling pathway, apoptosis, the Notch signaling pathway and other pathways. The present study provides a bioinformatic analysis of miR-34a-targets in gastric cancer, describes numerous target genes and novel coregulatory networks, and may provide an opportunity to identify a critical regulatory network for predicting the molecular mechanisms of miR-34a in the development and progression of gastric cancer.

15.
Oncotarget ; 6(32): 33658-74, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26378040

ABSTRACT

5-FU is a common first-line chemotherapeutic drug for the treatment of hepatocellular carcinoma. However the development of acquired resistance to 5-FU confines its clinical usages. Although this phenomenon has been the subject of intense investigation, the exact mechanism of acquired resistance to 5-FU remains elusive. Here, we report that over-expression of GRP78 contributes to acquired resistance to 5-FU in HCC by up-regulating the c-Src/LSF/TS axis. Moreover, we found that the resistance to 5-FU conferred by GRP78 is mediated by its ATPase domain. The ATPase domain differentially increased the expression of LSF, TS and promoted the phosphorylation of ERK and Akt. We further identified that GRP78 interacts physically with c-Src through its ATPase domain and promotes the phosphorylation of c-Src, which in turn increases the expression of LSF in the nucleus. Together, GRP78 confers the resistance to 5-FU by up-regulating the c-Src/LSF/TS axis via its ATPase domain.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , DNA-Binding Proteins/metabolism , Fluorouracil/pharmacology , Heat-Shock Proteins/metabolism , Liver Neoplasms/drug therapy , Thymidylate Synthase/metabolism , Transcription Factors/metabolism , src-Family Kinases/metabolism , Animals , Antimetabolites, Antineoplastic/pharmacology , CSK Tyrosine-Protein Kinase , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Drug Resistance, Neoplasm , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Signal Transduction , Thymidylate Synthase/genetics , Xenograft Model Antitumor Assays , src-Family Kinases/genetics
16.
Oncotarget ; 6(23): 19685-705, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26050197

ABSTRACT

Here, we found that BTG1 overexpression inhibited proliferation, migration and invasion, induced G2/M arrest, differentiation, senescence and apoptosis in BGC-823 and MKN28 cells (p < 0.05). BTG1 transfectants showed a higher mRNA expression of Cyclin D1 and Bax, but a lower mRNA expression of cdc2, p21, mTOR and MMP-9 than the control and mock (p < 0.05). After treated with cisplatin, MG132, paclitaxel and SAHA, both BTG1 transfectants showed lower mRNA viability and higher apoptosis than the control in both time- and dose-dependent manners (p < 0.05) with the hypoexpression of chemoresistance-related genes (slug, CD147, GRP78, GRP94, FBXW7 TOP1, TOP2 and GST-π). BTG1 expression was restored after 5-aza-2'-deoxycytidine treatment in gastric cancer cells. BTG1 expression was statistically lower in gastric cancer than non-neoplastic mucosa and metastatic cancer in lymph node (p < 0.05). BTG1 expression was positively correlated with depth of invasion, lymphatic and venous invasion, lymph node metastasis, TNM staging and worse prognosis (p < 0.05). The diffuse-type carcinoma showed less BTG1 expression than intestinal- and mixed-type ones (p < 0.05). BTG1 overexpression suppressed tumor growth and lung metastasis of gastric cancer cells by inhibiting proliferation, enhancing autophagy and apoptosis in xenograft models. It was suggested that down-regulated BTG1 expression might promote gastric carcinogenesis partially due to its promoter methylation. BTG1 overexpression might reverse the aggressive phenotypes and be employed as a potential target for gene therapy of gastric cancer.


Subject(s)
Genetic Therapy/methods , Lung Neoplasms/metabolism , Lung Neoplasms/prevention & control , Neoplasm Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cellular Senescence , DNA Methylation , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , Female , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Lymphatic Metastasis , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Promoter Regions, Genetic , RNA, Messenger/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection , Up-Regulation , Xenograft Model Antitumor Assays , Young Adult
17.
BMC Cancer ; 15: 389, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25958313

ABSTRACT

BACKGROUND: Emerging data have suggested that cell surface GRP78 is a multifunctional receptor and has been linked to proliferative and antiapoptotic signaling cascades. Activated α2-macroglobin (α2M*) is a natural circulating ligand of cell surface GRP78. Association of cell surface GRP78 with α2M* is involved in the regulation of cell proliferation, survival and apoptosis in human cancers. METHODS: The invasion and metastasis of HCC cells were examined using transwell and wound healing assay; Cell surface expression of GRP78 was detected by in cell western assay. Translocation of GRP78 from cytosol to cell surface was observed by transfection of GRP78-EGFP plus TRIRC-WGA staining. The levels of Src, phosphor-Src, FAK, phospho-FAK, EGFR, phospho-EGFR, phospho-Cortactin, phospho-Paxillin were determined by western blot. Cell surface expression of GRP78 in HCC tissue samples was observed by immunofluorescence. The distribution of Paxillin and Cortactin in HCC cells was also observed by immunofluorescence. The interaction between GRP78 and Src were detected by far-western blot, co-immunoprecipitation and GST pulldown. GRP78 mRNA was detected by RT-PCR. RESULTS: In the current study, we showed that association of cell surface GRP78 with α2M* stimulated the invasion and metastasis of HCC. Cell surface GRP78 could interact directly with c-Src, promoted the phosphorylation of c-Src at Y416. Inhibition of the tyrosine kinase activity of c-Src with PP2 reverted the stimulatory effect caused by association of cell surface GRP78 with α2M*. Moreover, association of cell surface GRP78 with α2M* facilitates the interaction between EGFR and c-Src and consequently phosphorylated EGFR at Y1101 and Y845, promoting the invasion and metastasis of HCCs. However, inhibition of the tyrosine kinase of c-Src do not affect the interaction between EGFR and Src. CONCLUSION: c-Src plays a critical role in the invasion and metastasis of HCC induced by association of cell surface GRP78 with α2M*. Cell surface GRP78 directly binds and phosphorylates c-Src. As a consequence, c-Src phosphorylated EGFR, promoting the invasion and metastasis of HCCs.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Heat-Shock Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , alpha-Macroglobulins/metabolism , src-Family Kinases/metabolism , CSK Tyrosine-Protein Kinase , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Membrane/metabolism , Endoplasmic Reticulum Chaperone BiP , ErbB Receptors/metabolism , Gene Expression , Heat-Shock Proteins/genetics , Humans , Liver Neoplasms/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Binding , Protein Transport , Signal Transduction
18.
Oncotarget ; 6(23): 19552-79, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25980581

ABSTRACT

Here, we found that ING5 overexpression increased autophagy, differentiation, and decreased proliferation, apoptosis, migration, invasion and lamellipodia formation in gastric cancer cells, while ING5 knockdown had the opposite effects. In SGC-7901 transfectants, ING5 overexpression caused G1 arrest, which was positively associated with 14-3-3 overexpression, Cdk4 and c-jun hypoexpression. The induction of Bax hypoexpression, Bcl-2, survivin, 14-3-3, PI3K, p-Akt and p70S6K overexpression by ING5 decreased apoptosis in SGC-7901 cells. The hypoexpression of MMP-9, MAP1B and flotillin 2 contributed to the inhibitory effects of ING5 on migration and invasion of SGC-7901 cells. ING5 overexpression might activate both ß-catenin and NF-κB pathways in SGC-7901 cells, and promote the expression of down-stream genes (c-myc, VEGF, Cyclin D1, survivin, and interleukins). Compared with the control, ING5 transfectants displayed drug resistance to triciribine, paclitaxel, cisplatin, SAHA, MG132 and parthenolide, which was positively related to their apoptotic induction and the overexpression of chemoresistance-related genes (MDR1, GRP78, GRP94, IRE, CD147, FBXW7, TOP1, TOP2, MLH1, MRP1, BRCP1 and GST-π). ING5 expression was higher in gastric cancer than matched mucosa. It was inversely associated with tumor size, dedifferentiation, lymph node metastasis and clinicopathological staging of cancer. ING5 overexpression suppressed growth, blood supply and lung metastasis of SGC-7901 cells by inhibiting proliferation, enhancing autophagy and apoptosis in xenograft models. It was suggested that ING5 expression might be employed as a good marker for gastric carcinogenesis and subsequent progression by inhibiting proliferation, growth, migration, invasion and metastasis. ING5 might induce apoptotic and chemotherapeutic resistances of gastric cancer cells by activating ß-catenin, NF-κB and Akt pathways.


Subject(s)
Apoptosis , Autophagy , Biomarkers, Tumor/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Endoplasmic Reticulum Chaperone BiP , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Lymphatic Metastasis , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Protein Interaction Maps , RNA Interference , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transcription Factors/genetics , Transfection , Tumor Suppressor Proteins/genetics
19.
Oncotarget ; 6(23): 19841-67, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25904053

ABSTRACT

BTG (B-cell translocation gene) can inhibit cell proliferation, metastasis and angiogenesis, cell cycle progression, and induce differentiation in various cells. Here, we found that BTG3 overexpression inhibited proliferation, induced S/G2 arrest, differentiation, autophagy, apoptosis, suppressed migration and invasion in MKN28 and MGC803 cells (p < 0.05). BTG3 transfectants showed a higher mRNA expression of p27, Bax, 14-3-3, Caspase-3, Caspase-9, Beclin 1, NF-κB, IL-1, -2, -4, -10 and -17, but a lower mRNA expression of p21, MMP-9 and VEGF than the control and mock (p < 0.05). At protein level, BTG3 overexpression increased the expression of CDK4, AIF, LC-3B, Beclin 1 and p38 (p < 0.05), but decreased the expression of p21 and ß-catenin in both transfectants (p < 0.05). After treated with cisplatin, MG132, paclitaxel and SAHA, both BTG3 transfectants showed lower viability and higher apoptosis than the control in both time- and dose-dependent manners (p < 0.05). BTG3 expression was restored after 5-aza-2'-deoxycytidine or MG132 treatment in gastric cancer cells. BTG3 expression was decreased in gastric cancer in comparison to the adjacent mucosa (p < 0.05), and positively correlated with venous invasion and dedifferentiation of cancer (p < 0.05). It was suggested that BTG3 expression might contribute to gastric carcinogenesis. BTG3 overexpression might reverse the aggressive phenotypes and be employed as a potential target for gene therapy of gastric cancer.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/therapy , Biomarkers, Tumor/metabolism , Genetic Therapy/methods , Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Methylation , Dose-Response Relationship, Drug , Female , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phenotype , Proteins/genetics , RNA, Messenger/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Time Factors , Transfection , Young Adult
20.
Int J Nanomedicine ; 10: 245-56, 2015.
Article in English | MEDLINE | ID: mdl-25565817

ABSTRACT

Nanoparticles (NPs) which target specific agents could effectively recognize the target cells and increase the stability of chemical agents by encapsulation. As such, NPs have been widely used in cancer treatment research. Recently, over 90% of treatment failure cases in patients with metastatic cancer were attributed to resistance to chemotherapy. Surface-exposed glucose-regulated protein of 78 kDa (GRP78) is expressed highly on many tumor cell surfaces in many human cancers and is related to the regulation of invasion and metastasis. Herein, we report that NPs conjugated with antibody against GRP78 (mAb GRP78-NPs) inhibit the adhesion, invasion, and metastasis of hepatocellular carcinoma (HCC) and promote drug delivery of 5-fluorouracil into GRP78 high-expressed human hepatocellular carcinoma cells. Our new findings suggest that mAb GRP78-NPs could enhance drug accumulation by effectively transporting NPs into cell surface GRP78-overexpressed human hepatocellular carcinoma cells and then inhibit cell proliferation and viability and induce cell apoptosis by regulating caspase-3. In brief, mAb GRP78-NPs effectively inhibit cancer cell invasion and enhance antitumor efficiency by targeted drug delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Heat-Shock Proteins/metabolism , Nanoparticles/chemistry , Neoplasm Invasiveness/prevention & control , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum Chaperone BiP , Fluorouracil/chemistry , Fluorouracil/pharmacology , Heat-Shock Proteins/genetics , Humans , Liver Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL