Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Oncol Rep ; 52(2)2024 Aug.
Article in English | MEDLINE | ID: mdl-38963046

ABSTRACT

Arsenic trioxide (ATO) is expected to be a chemical drug with antitumor activity against acute promyelocytic leukemia (APL), a type of acute myeloid leukemia. In Japan, its antitumor effects were confirmed in clinical trials for APL, and it has been approved in various countries around the world. However, there have been no reports on ATO's antitumor effects on radioresistant leukemia cells, which can be developed during radiotherapy and in combination with therapeutic radiation beams. The present study sought to clarify the antitumor effect of ATO on APL cells with radiation resistance and determine its efficacy when combined with ionizing radiation (IR). The radiation­resistant HL60 (Res­HL60) cell line was generated by subjecting the native cells to 4­Gy irradiation every week for 4 weeks. The half­maximal inhibitory concentration (IC50) for cell proliferation by ATO on native cell was 0.87 µM (R2=0.67), while the IC50 for cell proliferation by ATO on Res­HL60 was 2.24 µM (R2=0.91). IR exposure increased the sub­G1 and G2/M phase ratios in both cell lines. The addition of ATO resulted in a higher population of G2/M after 24 h rather than 48 h. When the rate of change in the sub­G1 phase was examined in greater detail, the sub­G1 phase in both control cells without ATO significantly increased by exposure to IR at 24 h, but only under the condition of 2 Gy irradiation, it had continued to increase at 48 h. Res­HL60 supplemented with ATO showed a higher rate of sub­G1 change at 24 h; however, 2 Gy irradiation resulted in a decrease compared with the control. There was a significant increase in the ratio of the G2/M phase in cells after incubation with ATO for 24 h, and exposure to 2 Gy irradiation caused an even greater increase. To determine whether the inhibition of cell proliferation and cell cycle disruptions is related to reactive oxygen species (ROS) activity, intracellular ROS levels were measured with a flow cytometric assay. Although the ROS levels of Res­HL60 were higher than those of native cells in the absence of irradiation, they did not change after 0.5 or 2 Gy irradiation. Furthermore, adding ATO to Res­HL60 reduced intracellular ROS levels. These findings provide important information that radioresistant leukemia cells respond differently to the antitumor effect of ATO and the combined effect of IR.


Subject(s)
Arsenic Trioxide , Arsenicals , Cell Proliferation , Leukemia, Promyelocytic, Acute , Oxides , Radiation, Ionizing , Humans , Arsenic Trioxide/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/pathology , Leukemia, Promyelocytic, Acute/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , HL-60 Cells , Arsenicals/pharmacology , Oxides/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Radiation Tolerance/drug effects , Antineoplastic Agents/pharmacology , Reactive Oxygen Species/metabolism
2.
Future Med Chem ; 16(11): 1053-1073, 2024.
Article in English | MEDLINE | ID: mdl-38708686

ABSTRACT

Aim: This study focused on designing and synthesizing novel derivatives of 3,5,8-trisubstituted coumarin. Results: The synthesized compounds, particularly compound 5, exhibited significant cytotoxic effects on MCF-7 cells, surpassing staurosporine, and reduced toxicity toward MCF-10A cells, highlighting potential pharmacological advantages. Further, compound 5 altered the cell cycle and significantly increased apoptosis in MCF-7 cells, involving both early (41.7-fold) and late stages (33-fold), while moderately affecting necrotic signaling. The antitumor activity was linked to a notable reduction (4.78-fold) in topoisomerase IIß expression. Molecular modeling indicated compound 5's strong affinity for EGFR, human EGF2 and topoisomerase II proteins. Conclusion: These findings highlight compound 5 as a multifaceted antitumor agent for breast cancer.


[Box: see text].


Subject(s)
Antineoplastic Agents , Apoptosis , Breast Neoplasms , Cell Proliferation , Coumarins , Drug Screening Assays, Antitumor , Humans , Coumarins/chemistry , Coumarins/pharmacology , Coumarins/chemical synthesis , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Female , Apoptosis/drug effects , DNA Topoisomerases, Type II/metabolism , Structure-Activity Relationship , MCF-7 Cells , Molecular Structure , Cell Line, Tumor , Cell Cycle/drug effects , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors
3.
J Oral Biol Craniofac Res ; 13(6): 796-800, 2023.
Article in English | MEDLINE | ID: mdl-38111634

ABSTRACT

Introduction: Nicotine, the main ingredient in tobacco, acts as a key alkaloid of nearly all tobacco products and has been demonstrated to facilitate tumorigenesis and accelerate metastasis. Further traditional tobacco products have shown to give systemic oral effects such as vasoconstriction, inflammation, and delayed wound healing, however; none of the reports have confirmed the significant knowledge of oral sequel of the effect of nicotine on oral epithelial cells. So, the current study aimed to investigate the effect of nicotine on epithelial transformation to a malignant state. Material & methods: Through in-vitro experiments, the effects of nicotine on epithelial cells obtained from nicotine never exposed buccal mucosa were analyzed using total count and viability test, proliferation assay, cell cycle distribution assay, and PI3K/MAPK dual pathway activation assay. Result & conclusion: MTT assay demonstrated that the proliferation of epithelial cells takes place at a 150 mM concentration of nicotine. Further, we identified the significantly increased cell count and viability in nicotine-exposed cells. Further, cell cycle distribution assay results demonstrated that nicotine forced the epithelial cells to enter the first growth phase. The same influence of nicotine was observed on the PI3K/MAPK dual pathway activation assay where a greater number of nicotine exposed cells showed dual pathway activation. In conclusion, the current study determined the potential mechanism of action of nicotine on oral epithelial cell proliferation through activating the oncogenic pathway. This may help to develop novel therapeutic strategies for the prevention of malignant transformation from smokeless tobacco-caused oral cancer.

4.
J Taibah Univ Med Sci ; 18(5): 1076-1086, 2023 Oct.
Article in English | MEDLINE | ID: mdl-36994222

ABSTRACT

Objectives: Lung cancer is a leading cause of mortality worldwide. In lung cancer treatment, nebulized solid lipid nanoparticles may be a viable drug delivery method, helping the drug reach sites of action, and improving its inhalation efficiency and pulmonary deposition. This research focused on evaluating the effectiveness of solid lipid nanoparticles of favipiravir (Fav-SLNps) in facilitating drug delivery to sites of action in lung cancer treatment. Methods: The hot-evaporation method was used to formulate Fav-SLNps. The in vitro cell viability, anti-cancer effects, and cellular uptake activity were evaluated in A549 human lung adenocarcinoma cells treated with the Fav-SLNp formulation. Results: The Fav-SLNps were formulated successfully. Importantly, Fav-SLNps at a concentration of 322.6 µg/ml were found to be safe and non-toxic toward A549 cells in vitro. The formulation had potential anti-proliferative properties via increasing the proportions of cells in G2/M and G0/G1 phases to 1.20 and 1.13 times those in untreated cells. Additionally, Fav-SLNp treatment significantly induced necrosis in A549 cells. Furthermore, the use of SLNps in the Fav formulation resulted in a macrophage drug uptake 1.23 times that of the free drug. Conclusion: Our results confirmed the internalization and anti-cancer activity of the Fav-SLNp formulation in the A549 lung cancer cell line. Our findings suggest that Fav-SLNps could potentially be used as lung cancer treatment to facilitate drug delivery to sites of action in the lungs.

5.
Curr Mol Med ; 23(3): 266-274, 2023.
Article in English | MEDLINE | ID: mdl-35040412

ABSTRACT

BACKGROUND: DNA methylation was considered as prognostic information in some hematological malignancies. Previous studies have reported the in vitro and in vivo biology role of mesenchymal stem cells (MSCs) on leukemic cells. The aim of this study was to investigate the effect of MSCs on the promoter methylation status of hTERT as a catalytic subunit of telomerase enzyme. METHODS: In the experimental study, the Molt-4 leukemic cells were co-cultured with MSCs for 7 days. At the end of the co-culture period, the Molt-4 cells were collected, DNA and protein were extracted. Then methylation specific-PCR and western blotting were done for evaluating the hTERT gene promoter methylation status and cyclin D1 and hTERT protein expression, respectively. In the following, the flow cytometry was done for cell cycle distribution assay. RESULTS: It was found that MSCs resulted in a significant decrease in the cyclin D1 and hTERT protein expression levels. Also, MSCs caused changes in the methylation status of the CpG islands in the hTERT gene promoter region. The following results showed that MSCs caused a significant increase in the number of cells at G0/G1 phase and arrest the G0/G1 phase as well as decrease in the cell proliferation of Molt-4 cells. CONCLUSION: It is concluded that co-culture of MSCs with Molt-4 cells could be involved in changing the methylation status of hTERT gene promoter, cell cycle and hTERT protein expression; it could be potentially beneficial for further investigations regarding the cell transplantation and cell-based therapy.


Subject(s)
Leukemia , Mesenchymal Stem Cells , Humans , CpG Islands/genetics , Cyclin D1/genetics , DNA Methylation , Promoter Regions, Genetic , Adipose Tissue
6.
In Vivo ; 36(6): 2722-2729, 2022.
Article in English | MEDLINE | ID: mdl-36309382

ABSTRACT

BACKGROUND/AIM: The influence of surgical interventions and anaesthesiological procedures on tumour progression was investigated as early as the 1920s. In current cancer management, the perioperative phase is increasingly being considered a vulnerable period with an increased risk of tumour cell dissemination due to medication, surgical manipulation, and immunosuppression. The extent to which narcotics administered in the perioperative setting influence the oncological outcomes of patients with pancreatic cancer is still unclear. MATERIALS AND METHODS: To investigate the effect of propofol and etomidate on the proliferation, cell-cycle distribution, apoptosis, and necrosis of pancreatic tumour cells in vitro, PaTu 8988t and Panc-1 pancreatic cancer cells were treated with 0-1,000 µM propofol or etomidate for 24 h each. Cell proliferation was measured with enzyme-linked immunosorbent-bromodeoxyuridine assay. The apoptosis rate was analysed with annexin V staining and the cell-cycle distribution with flow cytometry. RESULTS: Propofol at 1,000 µM induced apoptosis and inhibited cell proliferation. The cell cycle showed an increased S-phase and reduced cells in the G1-phase. At 100 µM, propofol significantly inhibited proliferation of the pancreatic cancer cell line PaTu 8988t and reduced cells in the G2-phase in the cell cycle. Etomidate had no effects on cell-cycle distribution, proliferation, apoptosis, and necrosis at the concentrations used. CONCLUSION: In this study, propofol was shown to have anticancer effects by induction of apoptosis and inhibition of cell proliferation, while etomidate did not affect pancreatic cancer cells. However, it is too early to make any recommendation for changes in clinical practice and further clinical studies are warranted to investigate the effect of anaesthetics on cancer progression.


Subject(s)
Etomidate , Pancreatic Neoplasms , Propofol , Humans , Etomidate/pharmacology , Etomidate/therapeutic use , Propofol/pharmacology , Propofol/therapeutic use , Apoptosis , Necrosis , Pancreatic Neoplasms/pathology , Cell Cycle , Cell Proliferation , Pancreatic Neoplasms
7.
Part Fibre Toxicol ; 19(1): 6, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35031062

ABSTRACT

BACKGROUND: Silver nanoparticles (AgNPs) are considered a double-edged sword that demonstrates beneficial and harmful effects depending on their dimensions and surface coating types. However, mechanistic understanding of the size- and coating-dependent effects of AgNPs in vitro and in vivo remains elusive. We adopted an in silico decision tree-based knowledge-discovery-in-databases process to prioritize the factors affecting the toxic potential of AgNPs, which included exposure dose, cell type and AgNP type (i.e., size and surface coating), and exposure time. This approach also contributed to effective knowledge integration between cell-based phenomenological observations and in vitro/in vivo mechanistic explorations. RESULTS: The consolidated cell viability assessment results were used to create a tree model for generalizing cytotoxic behavior of the four AgNP types: SCS, LCS, SAS, and LAS. The model ranked the toxicity-related parameters in the following order of importance: exposure dose > cell type > particle size > exposure time ≥ surface coating. Mechanistically, larger AgNPs appeared to provoke greater levels of autophagy in vitro, which occurred during the earlier phase of both subcytotoxic and cytotoxic exposures. Furthermore, apoptosis rather than necrosis majorly accounted for compromised cell survival over the above dosage range. Intriguingly, exposure to non-cytotoxic doses of AgNPs induced G2/M cell cycle arrest and senescence instead. At the organismal level, SCS following a single intraperitoneal injection was found more toxic to BALB/c mice as compared to SAS. Both particles could be deposited in various target organs (e.g., spleen, liver, and kidneys). Morphological observation, along with serum biochemical and histological analyses, indicated that AgNPs could produce pancreatic toxicity, apart from leading to hepatic inflammation. CONCLUSIONS: Our integrated in vitro, in silico, and in vivo study revealed that AgNPs exerted toxicity in dose-, cell/organ type- and particle type-dependent manners. More importantly, a single injection of lethal-dose AgNPs (i.e., SCS and SAS) could incur severe damage to pancreas and raise blood glucose levels at the early phase of exposure.


Subject(s)
Metal Nanoparticles , Silver , Animals , Cell Survival , Knowledge Discovery , Metal Nanoparticles/toxicity , Mice , Particle Size , Silver/toxicity
8.
Pharmaceuticals (Basel) ; 14(6)2021 Jun 08.
Article in English | MEDLINE | ID: mdl-34201348

ABSTRACT

Heat utility as a critical component of fever is often ignored, although the symptom is observed in many medical conditions. Mistletoe extract (ME) is an adjunctive medication prescribed to cancer patients. The increase in body temperature is frequently observed in patients following ME administration. Nevertheless, the impact of this fever on the effectiveness of therapy is unknown. Therefore, we aimed to investigate the effect of fever-range temperatures on ME-treated breast cancer cells and macrophages. The cells were simultaneously stimulated with ME and subjected to fever-range hyperthermia (FRH; 39 °C or 41 °C). After co-treatment, the cell viability, generation of reactive oxygen species (ROS), cell cycle distribution, and production of pro-inflammatory factors (interleukin (IL)-1ß, IL-6, and cyclooxygenase (COX)-2) were evaluated. The results showed that the exposure of ME-treated breast cancer cells to FRH at 39 °C resulted in a slight decrease in their viability, whereas FRH of 41 °C enhanced this effect. Only FRH of 41 °C induced minor changes in ROS level in ME-treated breast cancer cell lines. In ME-treated macrophages, FRH stimulated cell proliferation. The cell cycle distribution analysis showed a difference between cells cultured at 39 °C and 41 °C in all examined cell lines. Moreover, hyperthermia at 41 °C completely inhibited the ME-induced increase in IL-1ß and IL-6 expression in MCF-7 breast cancer cells, whereas this effect was not observed in 4T1 breast cancer cells. In contrast, in ME-treated macrophages, FRH of 41 °C strongly up-regulated expression of the pro-inflammatory factors. We conclude that fever is an important component of ME therapy that differentially affects cancer and immune cells.

9.
J Cardiothorac Surg ; 16(1): 194, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-34233689

ABSTRACT

OBJECTIVE: C-erbB-2 has been confirmed to be an oncogene that participates in cell growth, differentiation and division of tumors. We are wondered if its silenced expression can exert an anti-tumor effect. Therefore, this study is conducted to investigate the mechanism of C-erbB-2 silencing and IGF-1 pathway on esophageal carcinoma (EC) cell biological behaviors. METHODS: The objects of study were 84 EC patients from Heping Hospital Affiliated to Changzhi Medical College, with the collection of EC tissue and adjacent normal tissue (> 5 cm away from cancer tissue). C-erbB-2 protein expression in EC tissues was detected by immunohistochemistry. Human EC cell line Eca-109 was purchased from Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. Based on different transfection protocols, EC cells with logarithmic growth phase of 3-5 passages were divided into blank control group, oe-C-erbB-2 NC group, siRNA C-erbB-2 NC group, oe-C-erbB-2 group, siRNA C-erbB-2 group, OSI-906 group, Rg5 group, Rg5 + siRNA C-erbB-2 NC group and Rg5 + siRNA C-erbB-2 group. Cell proliferation was detected by MTT assay; cell cycle distribution and apoptosis by flow cytometry; C-erbB-2, IGF-1, IGF-1R and Akt mRNA and protein expressions by qRT-PCR and western blot; and cell invasion and migration by Transwell assay and scratch test. Tumor growth was observed in male BALB/c nude mice (Shanghai Experimental Animal Center) based on Eca109 cell implantation, raising, and measurement. RESULTS: C-erbB-2, IGF-1, IGF-1R and Akt expression were higher in EC tissues than those in adjacent tissues (all P < 0.05). Compared with blank control group, both si-C-erbB-2 and OSI-906 groups had decreased IGF-1, IGF-1R and Akt mRNA and protein expressions, decreased cell proliferation, migration and invasion, prolonged G0/G1 phase, shortened S phase, increased cell apoptosis, and inhibited tumor growth (all P < 0.05); while opposite trends were detected in C-erbB-2 vector and Rg5 groups (all P < 0.05), without statistical differences in siRNA C-erbB-2 + Rg5 group (all P > 0.05). CONCLUSION: Silencing C-erbB-2 expression may inhibit EC cell proliferation, promote cell apoptosis and block cell cycle progression by inhibiting IGF-1 pathway activation. The beneficial effect of silencing C-erbB-2 expression can be reversed by promoting the activation of IGF-1 pathway. Findings in our study may provide potential reference for understanding the molecular mechanism of EC and supply possible axis for preventing the development of EC from the perspective of molecular biology.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/physiology , Gene Silencing/physiology , Insulin-Like Growth Factor I/metabolism , Receptor, ErbB-2/genetics , Adult , Aged , Animals , Apoptosis/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Flow Cytometry , Humans , Immunohistochemistry , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Transplantation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, IGF Type 1 , Transfection
10.
Nat Prod Res ; 35(23): 4953-4959, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32352327

ABSTRACT

Usnic acid has anti-cancer activity, however, low solubility and toxicity limit the potential. To investigate biological activity of usnic acid derivatives, enantiopure derivatives were synthesised by reacting usnic acid with ethylenediamine, which yielded one dimer product ((+)-1), and two tetra cyclic compounds ((+)-2 and (-)-2). The products were characterised with NMR, and evaluated in vitro in human colon cancer cell line Caco-2 by cell count, phase-contrast microscopy, MTT-assay, measurement of DNA content and cell cycle distribution. All compounds tested showed cytostatic effect in Caco-2 cells, but each compound had a distinct cellular effect. Compound (+)-1 showed anti-proliferative activity by increasing the percentage of cells in S-phase with 25% compared to the control. Compounds (+)-2 and (-)-2 induced paraptosis, but only compound (+)-2 modulated cell cycle distribution by accumulating cells in G2/M-phase by 47% and reduced DNA content by 60%. All compounds express interesting cellular and potential anti-proliferative activity.


Subject(s)
Benzofurans , Cytostatic Agents , Caco-2 Cells , Cell Cycle , Cytostatic Agents/pharmacology , Humans
11.
Molecules ; 25(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422931

ABSTRACT

Bovine bone collagen hydrolysates promote bone formation through regulating bone growth. However, the peptide sequences within these isolates have not been characterized. In this study, twenty-nine peptides from bovine bone collagen hydrolysates were purified and identified using nano-HPLC-MS-MS and Peak Studio analysis. HHGDQGAPGAVGPAGPRGPAGPSGPAGKDGR (Deamidation) and GPAGANGDRGEAGPAGPAGPAGPR (Deamidation) enhanced cell viability, inhibited apoptosis, and significantly altered the cell cycle of MC3T3-E1 osteoblast cells. These peptides were selected to perform molecular docking analysis to examine the mechanism underlying these bioactivities. Molecular docking analysis showed that these two peptides formed hydrophobic interactions and hydrogen bonds with epidermal growth factor receptor (EGFR) to activate the EGFR-signaling pathway, which may explain their bioactivity. These findings indicate that these and other similar peptides might be candidates for the treatment of osteoporosis.


Subject(s)
Bone and Bones/chemistry , Collagen/chemistry , ErbB Receptors/chemistry , Peptides/chemistry , Protein Hydrolysates/chemistry , Amino Acid Sequence , Animals , Apoptosis/drug effects , Binding Sites , Cattle , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Cell Survival/drug effects , Collagen/isolation & purification , ErbB Receptors/genetics , ErbB Receptors/metabolism , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Docking Simulation , Osteoblasts/cytology , Osteoblasts/drug effects , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
12.
China Pharmacy ; (12): 614-620, 2019.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-817061

ABSTRACT

OBJECTIVE: To study the effects of aspirin on the growth and autoghagy of human gastric cancer cells SGC-7901 and BGC-823. METHODS: SGC-7901 and BGC-823 cells were selected as research objects, with phosphate buffer (PBS) as negative control treated for 48 h, MTT assay was used to detect the effects of 1, 2, 4, 6, 8, 10 mmol/L aspirin, 5 mmol/L aspirin alone or combined with 2.5 μmol/L chloroquine, 2.5 μmol/L 3-methyladenine (3-MA) on survival rate of gastric cancer cells. Flow cytometry was used to detect the effects of 2 and 5 mmol/L aspirin, 5 mmol/L aspirin alone or combined with 2.5 μmol/L chloroquine and 2.5 μmol/L 3-MA on the apoptosis rate and cell cycle distribution of gastric cancer cells. Hoechst33258 staining was used to observe the effects of 5 mmol/L aspirin on morphology of gastric cancer cell nucleus; Transwell chamber test was adopted to detect the effects of 5 mmol/L aspirin on the migration of gastric cancer cell. Laser confocal scanning microscopy was used to observe the effects of 5 mmol/L aspirin on autophagy formation in gastric cancer cells. Western blot method was used to detect the effects of 2 and 5 mmol/L aspirin on the protein expression of autophagy markers LC3-Ⅱin gastric cancer cells. RESULTS: Compared with negative control group, aspirin could inhibit the survival rates of SGC-7901 and BGC-823 cells in dose-dependent manner, but had no significant effects on apoptosis rate of SGC-7901 and BGC-823 cells; SGC-7901 and BGC-823 cells were blocked in G1 phase. Compared with aspirin alone group, the survival rates of SGC-7901 and BGC-823 were increased significantly after treated with aspirin+chloroquine and aspirin+3-MA, while the distribution rate of SGC-7901 and BGC-823 cells at G1 phase were decreased significantly, with statistical significance (P<0.05 or P<0.01). Compared with negative control group, there were no obvious DNA fragmentation fragments, apoptotic bodies and fragments of dense bright blue, while the number of migration cells were decreased significantly in SGC-7901 and BGC-823 cells after treated with aspirin (P<0.001); the number of autophagosome was increased significantly and the protein expression of LC3-Ⅱ was enhanced significantly (P<0.05). CONCLUSIONS: Aspirin can significantly inhibit the growth of SGC-7901 and BGC-823 cells, and arrest cell cycle in G1 phase, the mechanism of which may be associated with the activation of autophagy.

13.
J Biol Inorg Chem ; 24(2): 151-169, 2019 03.
Article in English | MEDLINE | ID: mdl-30564887

ABSTRACT

Three iridium(III) polypyridyl complexes [Ir(ppy)2(PYTA)](PF6) (1) (ppy = 2-phenylpyridine), [Ir(bzq)2(PYTA)](PF6) (2) (bzq = benzo[h]quinolone) and [Ir(piq)2(PYTA)](PF6) (3) (piq = 1-phenylisoquinoline, PYTA = 2,4-diamino-6-(2'-pyridyl)-1,3,5-triazine) were synthesized and characterized by elemental analysis, IR, 1H NMR and 13C NMR. The cytotoxic activity of the complexes toward cancer SGC-7901, Eca-109, A549, HeLa, HepG2, BEL-7402 and normal LO2 cell lines was investigated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method. Complex 3 shows the most effective on inhibiting the above cell growth among these complexes. The complexes locate at the lysosomes and mitochondria. AO/EB, Annex V and PI and comet assays indicate that the complexes can induce apoptosis in SGC-7901 cells. Intracellular ROS and mitochondrial membrane potential were examined under fluorescence microscopy. The results demonstrate that the complexes increase the intracellular ROS levels and induce a decrease in the mitochondrial membrane potential. The complexes can enhance intracellular Ca2+ concentration and cause a release of cytochrome c. The autophagy was studied using MDC staining and western blot. Complexes 1-3 can effectively inhibit the cell invasion with a concentration-dependent manner. Additionally, the complexes target tubules and inhibit the polymerization of tubules. The antimicrobial activity of the complexes against S. aureus, E. coli, Salmonella and L. monocytogenes was explored. The mechanism shows that the complexes induce apoptosis in SGC-7901 cells through ROS-mediated lysosomal-mitochondrial, targeting tubules and damage DNA pathways. Three iridium(III) complexes [Ir(N-C)2(PYTA)](PF6) (N-C = ppy, 1; bzq, 2; piq, 3) were synthesized and characterized. The anticancer activity of the complexes against SGC-7901 cells was studied by apoptosis, comet assay, autophagy, ROS, mitochondrial membrane potential, intracellular Ca2+ levels, release of cytochrome c, tubules and western blot analysis. The antibacterial activity in vitro was also assayed.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Iridium/pharmacology , Phenazopyridine/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Escherichia coli/drug effects , Humans , Iridium/chemistry , Listeria monocytogenes/drug effects , Microbial Sensitivity Tests , Molecular Structure , Phenazopyridine/chemistry , Salmonella/drug effects , Staphylococcus aureus/drug effects , Structure-Activity Relationship
14.
Phytomedicine ; 43: 78-85, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29747757

ABSTRACT

INTRODUCTION: Multidrug resistance of cancer cells constitutes a serious problem in chemotherapy and a challenging issue in the discovery of new cytotoxic drugs. Many saponins are known to display anti-cancer effects. In this study, the cytotoxicity and the modes of action of a naturally occuring oleanane-type tritepene saponin, ardisiacrispin B isolated from the fruit of Ardisia kivuensis Taton (Myrsinaceae) was evaluated on a panel of 9 cancer cell lines including various sensitive and drug-resistant phenotypes. METHODS: Resazurin reduction assay was used to evaluate cytotoxicity and ferroptotic cell death of samples; caspase-Glo assay was used to detect the activation of caspases in CCRF-CEM leukemia cells. Flow cytometry was used for cell cycle analysis and detection of apoptotic cells by annexin V/PI staining, analysis of mitochondrial membrane potential (MMP) and measurement of reactive oxygen species (ROS). RESULTS: Ardisiacrispin B displayed significant cytotoxic effects in the 9 tested cancer cell lines with IC50 values below 10 µM. The IC50 values ranges were 1.20 µM (towards leukemia CCRF-CEM cells) to 6.76 µM [against heptocarcinoma HepG2 cells] for ardisiacrispin B and 0.02 µM (against CCRF-CEM cells) to 122.96 µM (against resistant CEM/ADR5000 leukemia cells) for doxorubicin. Collateral sensitivity of resistant HCT116p53-/- colon adenocarcinoma cells to ardisiacripsin B was observed. Ardisiacrispin B induced apoptosis in CCRF-CEM cells via activation of inititator caspases 8 and 9 and effector caspase 3/7, alteration of MMP and increase in ROS production. Ferroptosis also contributed to the cytotoxicity of ardisiacrispin B. CONCLUSIONS: The studied oleanane-type triterpene saponin is a good cytotoxic molecule that deserve more detailed exploration in the future, to develop novel cytotoxic drugs to combat both sensitive and drug-resistant cancers.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm/drug effects , Oleanolic Acid/analogs & derivatives , Saponins/chemistry , Saponins/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , HCT116 Cells , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Reactive Oxygen Species/metabolism
15.
Environ Pollut ; 239: 300-307, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29665550

ABSTRACT

Polyfluorinated iodine alkanes (PFIs) are a kind of emerging chemicals with endocrine disrupting effects. Based on the different binding preferences of PFIs to estrogen receptor alpha and beta isoforms (ERα and ß), two representative PFIs, dodecafluoro-1,6-diiodohexane (PFHxDI) and tridecafluorohexyl iodide (PFHxI), were selected to evaluate their effects on the proliferation of two kinds of breast cancer cells with different ERα/ß expression levels, MCF-7 and T47D. The cell viability assay showed PFHxDI could cause higher cellular toxicity than did PFHxI in both MCF-7 and T47D. MCF-7 with relatively higher ERα/ß expression ratio was more vulnerable to the cytotoxic treatments of PFHxI and PFHxDI when compared with T47D cells with relatively lower ERα/ß expression ratio. EdU incorporation and cell cycle analysis revealed that, similar to 17ß-estrodiol (E2), non-cytotoxic levels of PFHxDI could significantly promote the proliferation of MCF-7 by increasing cell population at S phase (p < 0.01), while T47D proliferation was not influenced by PFHxI exposure due to cell cycle arrest at G2/M phase. The cellular responses caused by estrogenic PFIs were dominantly mediated by their preferential binding affinities for ER isoforms, which would be helpful in the accurate assessment for their potential influences on the breast cancer progression.


Subject(s)
Alkanes/chemistry , Estrogen Receptor alpha/chemistry , Fluorocarbons/chemistry , Iodine/chemistry , Alkanes/toxicity , Breast Neoplasms , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Estradiol , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Fluorocarbons/toxicity , Humans , Iodine/toxicity , Protein Isoforms
16.
Oncol Lett ; 15(1): 175-182, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29375710

ABSTRACT

S100A11, a member of S100 calcium-binding protein family, is associated with the numerous processes of tumorigenesis and metastasis. In the present study, the role of S100A11, and its possible underlying mechanisms in cell proliferation, apoptosis and cell cycle distribution in human pancreatic cancer were explored. Immunohistochemical analyses of S100A11 and phosphorylated (p)-AKT serine/threonine kinase (AKT) were performed in 30 resected specimens from patients with pancreatic cancer. PANC-1 cells were transfected with pcDNA3.1-S100A11 or treated with 50 µmol/l LY294002 for 48 h. Cell proliferation was determined using a cell counting kit-8 assay, whereas apoptosis and cell cycle distribution were determined by flow cytometry analysis. The mRNA and protein levels of S100A11, and AKT were determined using semi quantitative reverse transcription-polymerase chain reaction and western blot analyses, respectively. Pearson correlation analysis revealed that the expression levels of S100A11 and p-AKT were positively correlated (r, 0.802; P<0.05). Compared with the control group, S100A11 overexpression significantly promoted PANC-1 cell proliferation and reduced the percentage of early apoptotic cells. Flow cytometric analysis indicated that the proportion of PANC-1 cells in the S phase was significantly elevated and cell percentage in the G0/G1 phase declined in response to S100A11 overexpression (all P<0.05). S100A11 overexpression also significantly increased AKT mRNA and p-AKT protein expression levels (both P<0.05). The phosphoinositide 3-kinase (PI3K) inhibitor, LY294002, significantly inhibited PANC-1 cell proliferation, promoted apoptosis and caused G1/S phase arrest in PANC-1 cells (all P<0.05). These findings together suggest that S100A11 promotes the viability and proliferation of human pancreatic cancer PANC-1 cells through the upregulation of the PI3K/AKT signaling pathway. Thus, S100A11 may be considered as a novel drug target for targeted therapy of pancreatic cancer.

17.
Int J Mol Sci ; 17(7)2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27399677

ABSTRACT

1α,25-dihydroxyvitamin D3 (1,25D3) is a powerful differentiation inducer for acute myeloid leukemia (AML) cells. However, 1,25D3 doses required for differentiation of AML cells may cause lethal hypercalcemia in vivo. There is evidence that vitamin D2 is less toxic than vitamin D3 in animals. Here, we determined the differentiation effects of novel analogs of 1α,25-dihydroxyvitamin D2 (1,25D2), PRI-1916 and PRI-1917, in which the extended side chains of their previously reported precursors (PRI-1906 and PRI-1907, respectively) underwent further 24Z (24-cis) modification. Using four human AML cell lines representing different stages of myeloid maturation (KG-1a, HL60, U937, and MOLM-13), we found that the potency of PRI-1916 was slightly higher or equal to that of PRI-1906 while PRI-1917 was significantly less potent than PRI-1907. We also demonstrated that 1,25D2 was a less effective differentiation agent than 1,25D3 in these cell lines. Irrespective of their differentiation potency, all the vitamin D2 derivatives tested were less potent than 1,25D3 in transactivating the DR3-type vitamin D response elements. However, similar to 1,25D3, both 1,25D2 and its analogs could strongly cooperate with the plant polyphenol carnosic acid in inducing cell differentiation and inhibition of G1-S cell cycle transition. These results indicate that the 24Z modification has contrasting effects on the differentiation ability of PRI-1906 and PRI-1907 and that the addition of a plant polyphenol could result in a similar extent of cell differentiation induced by different vitamin D compounds. The enhanced antileukemic effects of the tested combinations may constitute the basis for the development of novel approaches for differentiation therapy of AML.


Subject(s)
Cell Differentiation/drug effects , Ergocalciferols/pharmacology , Plant Extracts/pharmacology , Polyphenols/pharmacology , Abietanes/pharmacology , Cell Line, Tumor , Ergocalciferols/chemistry , G1 Phase Cell Cycle Checkpoints/drug effects , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Plant Extracts/chemistry
18.
Molecules ; 20(7): 11994-2015, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26133763

ABSTRACT

To examine the effect of hydrophobicity on the anticancer activity of 1,4-naphthoquinone derivatives, a series of compounds bearing a 2-O-alkyl-, 3-C-alkyl- or 2/3-N-morpholinoalkyl group were synthesized and evaluated for their anticancer activity against five human cancer cell lines in vitro. The cytotoxicity of these derivatives was assayed against HT-29, SW480, HepG2, MCF-7 and HL-60 cells by the MTT assay. Among them, 2-hydroxy-3-farnesyl-1,4-naphthoquinone (11a) was found to be the most cytotoxic against these cell lines. Our results showed that the effectiveness of compound 11a may be attributed to its suppression of the survival of HT-29. Secondly, in the Hoechst 33258 staining test, compound 11a-treated cells exhibited nuclear condensation typical of apoptosis. Additionally, cell cycle analysis by flow cytometry indicated that compound 11a arrested HT-29 cells in the S phase. Furthermore, cell death detected by Annexin V-FITC/propidium iodide staining showed that compound 11a efficiently induced apoptosis of HT-29 in a concentration-dependent manner. Taken together, compound 11a effectively inhibits colon cancer cell proliferation and may be a potent anticancer agent.


Subject(s)
Lipids/chemistry , Naphthoquinones/chemical synthesis , Naphthoquinones/pharmacology , Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Flow Cytometry , Humans , Naphthoquinones/chemistry
19.
Int J Nanomedicine ; 9: 3119-30, 2014.
Article in English | MEDLINE | ID: mdl-25061290

ABSTRACT

Despite the promising anticancer potential of curcumin, its therapeutic application has been limited, owing to its poor solubility, bioavailability, and chemical fragility. Therefore, various formulation approaches have been attempted to address these problems. In this study, we entrapped curcumin into monoolein (MO)-based liquid crystalline nanoparticles (LCNs) and evaluated the physicochemical properties and anticancer activity of the LCN dispersion. The results revealed that particles in the curcumin-loaded LCN dispersion were discrete and monodispersed, and that the entrapment efficiency was almost 100%. The stability of curcumin in the dispersion was surprisingly enhanced (about 75% of the curcumin survived after 45 days of storage at 40°C), and the in vitro release of curcumin was sustained (10% or less over 15 days). Fluorescence-activated cell sorting (FACS) analysis using a human colon cancer cell line (HCT116) exhibited 99.1% fluorescence gating for 5 µM curcumin-loaded LCN dispersion compared to 1.36% for the same concentration of the drug in dimethyl sulfoxide (DMSO), indicating markedly enhanced cellular uptake. Consistent with the enhanced cellular uptake of curcumin-loaded LCNs, anticancer activity and cell cycle studies demonstrated apoptosis induction when the cells were treated with the LCN dispersion; however, there was neither noticeable cell death nor significant changes in the cell cycle for the same concentration of the drug in DMSO. In conclusion, entrapping curcumin into MO-based LCNs may provide, in the future, a strategy for overcoming the hurdles associated with both the stability and cellular uptake issues of the drug in the treatment of various cancers.


Subject(s)
Antineoplastic Agents/chemistry , Curcumin/chemistry , Glycerides/chemistry , Nanoparticles/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Curcumin/pharmacology , Drug Carriers/chemistry , Drug Stability , HCT116 Cells , Humans , Particle Size , Solubility
20.
Phytomedicine ; 21(3): 315-22, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24075210

ABSTRACT

BACKGROUND: Resistance of cancer to chemotherapy remains a challenging issue for scientists as well as physicians. Naturally occurring xanthones possess a variety of biological activities such as anti-inflammatory, anti-bacterial, and anti-cancer effects. The present study was aimed at investigating the cytotoxicity and the modes of action of three naturally occurring xanthones namely, morusignin I (1), 8-hydroxycudraxanthone G (2) and cudraxanthone I (3) against a panel of nine cancer cell lines, including various sensitive and drug-resistant phenotypes. METHODS: The cytotoxicity of the compounds was determined using a resazurin reduction assay, whereas the caspase-Glo assay was used to detect the activation of caspases 3/7, caspase 8 and caspase 9 in cells treated with compounds 3. Flow cytometry was used for cell cycle analysis and detection of apoptotic cells, analysis of mitochondrial membrane potential (MMP) as well as measurement of reactive oxygen species (ROS). RESULTS: Compounds 1 and 3 inhibited the proliferation of all tested cancer cell lines including sensitive and drug-resistant phenotypes. Compound 2 was active on 8/9 cell lines with the IC50 values ranging from 16.65 µM (against leukemia CCRF-CEM cells) to 70.38 µM (against hepatocarcinoma HepG2 cells). The IC50 value ranged from 7.15 µM (against CCRF-CEM cells) to 53.85 µM [against human glioblastoma U87MG.ΔEGFR cells] for compound 1, and from 2.78 µM (against breast cancer MDA-MB231 BCRP cells) to 22.49 µM (against U87MG cells) for compound 3. P-glycoprotein expressing CEM/ADR5000 cells were cross-resistant to compounds 1 and 2 (4.21- to 610-fold) while no cross-resistance or even collateral cross-sensitivity were observed in other drug-resistant cell lines to the three compounds. Normal AML12 liver cells were more resistant to the three compounds than HepG2 liver cancer cells. Compounds 3 arrested the cell cycle between G0/G1 and S phases, strongly induced apoptosis via caspases 3/7, caspase 8, caspase 9 activation and disrupted the MMP in CCRF-CEM cells. CONCLUSIONS: The cytotoxicity of the studied xanthones and especially compound 3 deserve more detailed exploration in the future to develop novel anticancer drugs against sensitive and otherwise drug-resistant phenotypes.


Subject(s)
Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Garcinia/chemistry , Neoplasms/drug therapy , Phytotherapy , Plant Extracts/therapeutic use , Xanthones/therapeutic use , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Neoplasms/metabolism , Plant Extracts/pharmacology , Xanthones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL