Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 270
Filter
Add more filters

Country/Region as subject
Publication year range
1.
BMC Oral Health ; 24(1): 715, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907185

ABSTRACT

BACKGROUND: Dental pathogens play a crucial role in oral health issues, including tooth decay, gum disease, and oral infections, and recent research suggests a link between these pathogens and oral cancer initiation and progression. Innovative therapeutic approaches are needed due to antibiotic resistance concerns and treatment limitations. METHODS: We synthesized and analyzed piperine-coated zinc oxide nanoparticles (ZnO-PIP NPs) using UV spectroscopy, SEM, XRD, FTIR, and EDAX. Antioxidant and antimicrobial effectiveness were evaluated through DPPH, ABTS, and MIC assays, while the anticancer properties were assessed on KB oral squamous carcinoma cells. RESULTS: ZnO-PIP NPs exhibited significant antioxidant activity and a MIC of 50 µg/mL against dental pathogens, indicating strong antimicrobial properties. Interaction analysis revealed high binding affinity with dental pathogens. ZnO-PIP NPs showed dose-dependent anticancer activity on KB cells, upregulating apoptotic genes BCL2, BAX, and P53. CONCLUSIONS: This approach offers a multifaceted solution to combatting both oral infections and cancer, showcasing their potential for significant advancement in oral healthcare. It is essential to acknowledge potential limitations and challenges associated with the use of ZnO NPs in clinical applications. These may include concerns regarding nanoparticle toxicity, biocompatibility, and long-term safety. Further research and rigorous testing are warranted to address these issues and ensure the safe and effective translation of ZnO-PIP NPs into clinical practice.


Subject(s)
Alkaloids , Apoptosis , Benzodioxoles , Biofilms , Mouth Neoplasms , Piperidines , Polyunsaturated Alkamides , Zinc Oxide , bcl-2-Associated X Protein , Humans , Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Apoptosis/drug effects , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/drug effects , Benzodioxoles/pharmacology , Biofilms/drug effects , Cell Line, Tumor , KB Cells , Metal Nanoparticles/therapeutic use , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Nanoparticles , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/drug effects , X-Ray Diffraction , Zinc Oxide/pharmacology
2.
Anticancer Drugs ; 33(1): 30-47, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34261915

ABSTRACT

Atractylodes is the dry root of atractylodes macrocephala koidz and has been commonly used as a traditional Chinese medicine (TCM). Atractylenolide III, a main component of atractylodes, has displayed significant effects on anti-inflammation and anticancer. However, the effects of atractylenolide III on growth inhibition and apoptosis induction in colon cancer remain unclear. The results showed that atractylenolide III significantly inhibited the cell growth and induce cellular apoptosis in HCT-116 cells in a concentration dependence manner in vitro. Mechanistic studies further showed that atractylenolide III could regulate the Bax/Bcl-2 apoptotic signaling pathway through promoting the expression of proapoptotic related gene/proteins Bax, caspase-9 and caspase-3 but inhibiting the expression of antiapoptotic related gene/protein Bcl-2 in HCT-116 cells. Furthermore, atractylenolide III also significantly inhibited the tumor growth of HCT-116 tumor xenografts bearing in nude mice through inducing apoptosis by upregulation of the expressions of Bax, cleaved caspase-3 and p53 but downregulation of the expressions of Bcl-2 in HCT-116 tumor tissues in vivo. The studies may provide the scientific rationale for the understanding of the anticancer effect of atractylenolide III. Therefore, atractylenolide III may have the potential to be developed as a promising novel anticancer agent for the treatment of colorectal cancer clinically.


Subject(s)
Colonic Neoplasms/pathology , Lactones/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Sesquiterpenes/pharmacology , bcl-2-Associated X Protein/drug effects , Animals , Apoptosis/drug effects , Caspase 3/drug effects , Caspase 9/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
J Pharm Pharm Sci ; 25: 69-76, 2022.
Article in English | MEDLINE | ID: mdl-35030074

ABSTRACT

PURPOSE: Among abused substances, methamphetamine is a psychostimulant drug widely used recreationally with public health importance. This study investigated the effect of methamphetamine on proliferation, differentiation, and apoptosis of human adipose tissue stem cells (AdSCs). METHODS: AdSCs were isolated from human abdominal adipose tissue and were characterized for mesenchymal properties and growth kinetics. MTT assay was undertaken to assess methamphetamine toxicity on proliferation and differentiation properties and apoptosis of hAdSCs. RESULTS: Isolated cells were shown to have mesenchymal properties and a population doubling time (PDT) of 40.1 h. Following methamphetamine treatment, expressions of KI-67 and TPX2 as proliferation genes and Col1A1 and PPARg as differentiation genes decreased. Methamphetamine administration increased the expression of Bax and decreased Bcl-2 genes responsible for apoptosis. CONCLUSIONS: Our data suggested when AdSCs were exposed to methamphetamine, it decreased proliferation and differentiation properties of stem cells together with an increase in apoptosis. These findings can be added to the literature, especially when methamphetamine is used recreationally for weight loss purposes.


Subject(s)
Adipose Tissue/drug effects , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Mesenchymal Stem Cells/drug effects , Methamphetamine/pharmacology , Adipose Tissue/cytology , Cell Cycle Proteins/drug effects , Genes, bcl-2/drug effects , Humans , Ki-67 Antigen/drug effects , Microtubule-Associated Proteins/drug effects , PPAR gamma/drug effects , bcl-2-Associated X Protein/blood , bcl-2-Associated X Protein/drug effects
4.
Pharm Biol ; 60(1): 543-552, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35225146

ABSTRACT

CONTEXT: Crocin has been reported to have multiple bioactivities. However, the effect of crocin administration on caecal ligation and puncture (CLP)-induced sepsis remains unknown. OBJECTIVE: We investigated the effects of crocin on CLP-induced sepsis in mice and the underlying mechanism of action. MATERIALS AND METHODS: Five experimental groups (n = 10) of BALB/c mice were used: control, CLP (normal saline) and CLP + crocin (50, 100 and 250 mg/kg, 30 min prior to CLP). Mice were sacrificed 24 h after CLP. Liver, kidney and lung histopathology, indicator levels, apoptotic status, pro-inflammatory cytokines and relative protein levels were evaluated. RESULTS: Compared to the CLP group, crocin treatment significantly increased the survival rate (70%, 80%, 90% vs. 30%). Crocin groups exhibited protection against liver, kidney and lung damage with mild-to-moderate morphological changes and lower indicator levels: liver (2.80 ± 0.45, 2.60 ± 0.55, 1.60 ± 0.55 vs. 5.60 ± 0.55), kidney (3.00 ± 0.71, 2.60 ± 0.55, 1.40 ± 0.55 vs. 6.20 ± 0.84) and lungs (8.00 ± 1.59, 6.80 ± 1.64, 2.80 ± 0.84 vs. 14.80 ± 1.79). The proinflammatory cytokines (IL-1ß, TNF-α, IL-6 and IL-10 levels in the crocin groups) were distinctly lower and the apoptotic index showed a significant decrease. Crocin administration significantly suppressed p38 MAPK phosphorylation and inhibited NF-κB/IκBα and Bcl-2/Bax activation. DISCUSSION AND CONCLUSIONS: Pre-treatment with crocin confers protective effects against CLP-induced liver, kidney and lung injury, implying it to be a potential therapeutic agent.


Subject(s)
Carotenoids/pharmacology , NF-kappa B/metabolism , Sepsis/drug therapy , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Carotenoids/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Liver Diseases/etiology , Liver Diseases/prevention & control , Lung Diseases/etiology , Lung Diseases/prevention & control , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-bcl-2/metabolism , Sepsis/complications , bcl-2-Associated X Protein/drug effects
5.
Biochem Biophys Res Commun ; 548: 60-66, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33631675

ABSTRACT

Repeated and long-term oxaliplatin therapy leads to drug resistance and severe adverse events, which limit its clinical use. These difficulties highlight the importance of identifying potent and specific drug combinations to enhance the antitumor effects of oxaliplatin. The farnesoid X receptor (FXR) deficiency in colorectal cancer (CRC) suggests that restoring FXR function might be a promising strategy for CRC treatment. A drug combination study showed that the GW4064 acted synergistically with oxaliplatin in colon cancer cells. The combination of oxaliplatin plus GW4064 inhibited cell growth and colony formation, induced apoptosis and pyroptosis in vitro, and slowed tumor growth in vivo. Mechanistically, GW4064 enhanced the chemosensitivity of cells to oxaliplatin by inducing BAX/caspase-3/GSDME-mediated pyroptosis. Furthermore, the combination of oxaliplatin and GW4064 synergistically inhibited STAT3 signaling by restoring SHP expression. Our study revealed that GW4064 could enhance the antitumor effects of oxaliplatin against CRC, which provides a novel therapeutic strategy based on a combinational approach for CRC treatment.


Subject(s)
Colorectal Neoplasms/pathology , Isoxazoles/pharmacology , Oxaliplatin/pharmacology , Pyroptosis/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/ultrastructure , Drug Synergism , Humans , Inflammasomes/metabolism , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
6.
Invest New Drugs ; 39(1): 98-110, 2021 02.
Article in English | MEDLINE | ID: mdl-32856275

ABSTRACT

Two novel chemotherapeutic chalcones were synthesized and their structures were confirmed by different spectral tools. Theoretical studies such as molecular modeling were done to detect the mechanism of action of these compounds. In vitro cytotoxicity showed a strong effect against all tested cell lines (MCF7, A459, HepG2, and HCT116), and low toxic effect against normal human melanocytes (HFB4). The lung carcinoma cell line was chosen for further molecular studies. Real-time PCR demonstrated that the two compounds upregulated gene expression of (BAX, p53, casp-3, casp-8, casp-9) genes and decreased the expression of anti-apoptotic genes bcl2, CDK4, and MMP1. Flow-cytometry indicated that cell cycle arrest of A459 was induced at the G2/M phase and the apoptotic percentage increased significantly compared to the control sample. Cytochrome c oxidase and VEGF enzyme activity were detected by ELISA assay. SEM tool was used to follow the morphological changes that occurred on the cell surface, cell granulation, and average roughness of the cell surface. The change in the number and morphology of mitochondria, cell shrinkage, increase in the number of cytoplasmic organelles, membrane blebbing, chromatin condensation, and apoptotic bodies were observed using TEM. The obtained data suggested that new chalcones exerted their pathways on lung carcinoma through induction of two pathways of apoptosis. Graphical abstract Novel chalcones were prepared and confirmed by different spectral tools. Docking simulations were done to detect the mechanism of action. In vitro cytotoxicity indicated a strong effect against different cancer cell lines and low toxic effects against normal human melanocytes (HFB4). The lung carcinoma cell line was chosen for further molecular studies that include Real-time PCR, Flow-cytometry, Cytochrome c oxidase, and ELISA assay. SEM and TEM tool were used to follow the morphological changes occurred on the cell surface.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Chalcones/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Caspases/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chalcones/chemistry , Gene Expression/drug effects , Humans , Molecular Docking Simulation , Tumor Suppressor Protein p53/drug effects , bcl-2-Associated X Protein/drug effects
7.
Endocr Regul ; 55(2): 61-71, 2021 May 21.
Article in English | MEDLINE | ID: mdl-34020532

ABSTRACT

Objectives. The present study was designed to assess whether apoptosis-related genes as parp-1 and bax could be targets for treatment of diabetes mellitus and whether vitamin D may exert beneficial effects. Methods. Vitamin D3 treatment for 4 weeks, starting after 4 weeks of the diabetes duration. The expression of parp-1 and bax genes was estimated on mRNA levels using real time quantitative polymerase chain reaction. Results. After 8 weeks, diabetic rats had weight loss, while blood glucose was increased about 4.9-fold compared to control group. Vitamin D3 administration to diabetic animals had no effect on these parameters. It was found that total serum alkaline phosphatase activity was significantly elevated in diabetic rats as compared to control animals and was restored by vitamin D3. Diabetes was accompanied by reduction of nicotinamidadenindinucleotide, a substrate of poly-ADP-ribosylation, level by 31.7% as compared to control rats, which was not reversed in response to vitamin D3 treatment. In diabetic hearts, the mRNA expression level of parp-1 gene was 2.8-fold higher compared to control rats and partially decreased by vitamin D3 treatment. Less significant alterations were observed in diabetic hearts for the mRNA expression level of bax gene that was 2.0-fold higher compared to control animals and vitamin D3 normalized it. These results indicate that cardiomyocytes have a tendency to apoptosis. Conclusions. The findings suggest that investigated genes can be targets at the transcriptional level for vitamin D action that may be contributed to the improving metabolic/signaling pathways induced by diabetes mellitus.


Subject(s)
Cholecalciferol/pharmacology , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Diabetic Cardiomyopathies , Poly (ADP-Ribose) Polymerase-1 , bcl-2-Associated X Protein , Animals , Cholecalciferol/administration & dosage , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Diabetic Cardiomyopathies/metabolism , Male , Poly (ADP-Ribose) Polymerase-1/drug effects , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
8.
Pharmacology ; 106(9-10): 551-563, 2021.
Article in English | MEDLINE | ID: mdl-34175854

ABSTRACT

BACKGROUND: Oral squamous cell carcinoma (OSCC) is among the most prevalent head and neck malignancies globally, and it is associated with high mortality rates. Actein is one of the primary active components extractable from the rhizomes of Cimicifuga foetida. This study aimed to evaluate the anti-OSCC effects of actein and evaluate the potential underlying mechanisms. METHODS AND RESULTS: CCK-8 cell proliferation experiments demonstrated significant dose- and time-dependent anti-OSCC effects of actein, while actein had weak cytotoxic effects on normal oral cell lines. Flow cytometry for cell cycle evaluation revealed that actein could induce cell cycle arrest at the G1 phase among OSCC cell lines. In our Annexin V/PI double staining apoptosis analysis, actein induced significant apoptosis among OSCC cells, with upregulation of Bax and downregulation of Bcl-2. Our mechanistic study implicated the involvement of the Akt/FoxO1 pathway in the anti-OSCC effects of actein. Akt1 and Akt2 expression significantly decreased in association with the FoxO1 upregulation. Furthermore, Bim and p21 were significantly upregulated, while survivin expression was downregulated. Finally, actein treatment was associated with significant p-Akt downregulation and p-FoxO1 upregulation in OSCC cells, demonstrating the validated roles of Akt/FoxO1 in actein-mediated OSCC cell apoptosis and cell cycle arrest. FoxO1 knockdown significantly reversed the anti-OSCC effects of actein. Additionally, a xenograft model indicated that actein could inhibit OSCC cell growth in vivo. CONCLUSIONS: Our findings demonstrated that actein could be a strong anti-OSCC candidate. Further evaluations of its safety and effectiveness are necessary before it can be considered for clinical use.


Subject(s)
Carcinoma, Squamous Cell/pathology , Drugs, Chinese Herbal/pharmacology , Forkhead Box Protein O1/drug effects , Mouth Neoplasms/pathology , Saponins/pharmacology , Triterpenes/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Female , Genes, bcl-2/drug effects , Humans , Mice , Mice, Inbred C57BL , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/drug effects
9.
BMC Anesthesiol ; 21(1): 104, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33823789

ABSTRACT

BACKGROUND: Myocardial ischemia-reperfusion injury (MIRI) is the most common cause of death worldwide. The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome plays an important role in the inflammatory response to MIRI. Dexmedetomidine (DEX), a specific agonist of α2-adrenergic receptor, is commonly used for sedation and analgesia in anesthesia and critically ill patients. Several studies have shown that dexmedetomidine has a strong anti-inflammatory effect in many diseases. Here, we investigated whether dexmedetomidine protects against MIRI by inhibiting the activation of the NLRP3 inflammasome in vitro. METHODS: We established an MIRI model in cardiomyocytes (CMs) alone and in coculture with cardiac fibroblasts (CFs) by hypoxia/reoxygenation (H/R) in vitro. The cells were treated with dexmedetomidine with or without MCC950 (a potent selective NLRP3 inhibitor). The beating rate and cell viability of cardiomyocytes, NLRP3 localization, the expression of inflammatory cytokines and NLRP3 inflammasome-related proteins, and the expression of apoptosis-related proteins, including Bcl2 and BAX, were determined. RESULTS: Dexmedetomidine treatment increased the beating rates and viability of cardiomyocytes cocultured with cardiac fibroblasts. The expression of the NLRP3 protein was significantly upregulated in cardiac fibroblasts but not in cardiomyocytes after H/R and was significantly attenuated by dexmedetomidine treatment. Expression of the inflammatory cytokines IL-1ß, IL-18 and TNF-α was significantly increased in cardiac fibroblasts after H/R and was attenuated by dexmedetomidine treatment. NLRP3 inflammasome activation induced the increased expression of cleaved caspase1, mature IL-1ß and IL-18, while dexmedetomidine suppressed H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. In addition, dexmedetomidine reduced the expression of Bcl2 and BAX in cocultured cardiomyocytes by suppressing H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts. CONCLUSION: Dexmedetomidine treatment can suppress H/R-induced NLRP3 inflammasome activation in cardiac fibroblasts, thereby alleviating MIRI by inhibiting the inflammatory response.


Subject(s)
Dexmedetomidine/pharmacology , Myocardial Reperfusion Injury/prevention & control , NLR Family, Pyrin Domain-Containing 3 Protein/drug effects , Analgesics, Non-Narcotic/pharmacology , Cell Survival/drug effects , Coculture Techniques , Cytokines/metabolism , Fibroblasts/metabolism , Furans/pharmacology , Humans , Indenes/pharmacology , Myocytes, Cardiac/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Up-Regulation , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
10.
Mar Drugs ; 19(1)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33430124

ABSTRACT

Three new and rare chromone derivatives, epiremisporine C (1), epiremisporine D (2), and epiremisporine E (3), were isolated from marine-derived Penicillium citrinum, together with four known compounds, epiremisporine B (4), penicitrinone A (5), 8-hydroxy-1-methoxycarbonyl-6-methylxanthone (6), and isoconiochaetone C (7). Among the isolated compounds, compounds 2-5 significantly decreased fMLP-induced superoxide anion generation by human neutrophils, with IC50 values of 6.39 ± 0.40, 8.28 ± 0.29, 3.62 ± 0.61, and 2.67 ± 0.10 µM, respectively. Compounds 3 and 4 exhibited cytotoxic activities with IC50 values of 43.82 ± 6.33 and 32.29 ± 4.83 µM, respectively, against non-small lung cancer cell (A549), and Western blot assay confirmed that compounds 3 and 4 markedly induced apoptosis of A549 cells, through Bcl-2, Bax, and caspase 3 signaling cascades.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Chromones/chemistry , Chromones/pharmacology , Penicillium/chemistry , A549 Cells , Adult , Anti-Bacterial Agents/pharmacology , Caspase 3/drug effects , Cell Line, Tumor , Fermentation , Humans , In Vitro Techniques , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Structure , Neutrophils/drug effects , Neutrophils/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Signal Transduction/drug effects , Superoxides/chemistry , Young Adult , bcl-2-Associated X Protein/drug effects
11.
Pak J Pharm Sci ; 34(3(Special)): 1289-1295, 2021 May.
Article in English | MEDLINE | ID: mdl-34602402

ABSTRACT

The aim of this study was to determine the radio sensitization of docetaxel in human esophageal squamous carcinoma ECA109 cell line by observing the effects of docetaxel in ECA109 cell proliferation, cell cycle distribution, apoptosis rate and related protein expression. Docetaxel inhibits the proliferation in ECA109 cell line in a dose-dependent and time-dependent manner, and 1nM was chosen for radio sensitization according to the inhibition curves. The D0 and SF2 values of ECA109 cells were 3.00Gy and 0.95, respectively, and of docetaxel (1nM) with irradiation group were 2.54Gy and 0.88. G0/G1 decreased (P<0.05), G2/M phase saw a spike (P<0.05) in the docetaxel with radiation group at 12h, 24h and 48h, while the apoptotic index witnessed a surge at 24h and 48h (P<0.05). The docetaxel with radiation group obtained a higher expression of p21 and bax protein than the docetaxel group and the radiation group (P<0.05), and a higher ratio of bcl-2/bax than the others (P<0.05). Docetaxel could inhibit the proliferation in ECA109 cell line. p21, bax, bcl-2 and other related proteins can regulate cell cycle phase distribution and induce cell apoptosis, thereby increasing the radiosensitivity effect of docetaxel in ECA109 cell line.


Subject(s)
Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Docetaxel/pharmacology , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Apoptosis/radiation effects , Cell Cycle/radiation effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/radiation effects , Humans , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/radiation effects , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/radiation effects
12.
J Cell Physiol ; 235(11): 8495-8506, 2020 11.
Article in English | MEDLINE | ID: mdl-32324265

ABSTRACT

Radiotherapy is one of the most effective modalities for treatment of neoplastic diseases. Radiation damage is to a large extent caused by overproduction of reactive oxygen species. To improve the therapeutic index, identifying effective substances for prevention or treatment of postirradiation intestinal and bone marrow injury should be prompted. This study was designed to evaluate the protective effects of cimetidine on the in rats exposed to γ-irradiation (5 Gy) and exploring the B-cell lymphoma 2 (Bcl2)/Bcl2 associated X (bax) pathway as a probable underlying mechanism. Eighteen adult male rats were randomly grouped into three: control, untreated irradiated rats, and irradiated rats pretreated with cimetidine. Seven days postirradiation the rats were culled, the bone marrow (BM) and jejunum tissue samples were collected for biochemical, histological, and immunohistological evaluation of BM cell count (BMCs), intestinal fibrosis, oxidative stress, tumor necrosis factor-α, Bcl2, and Bax. Cimetidine pretreatment significantly reversed the loss of BMCs, intestinal lining destruction, and fibrosis seen in the untreated irradiated rats and significantly decreased the underlying oxidative stress, inflammation, and Bax/Bcl2 ratio. There was a significant differential correlation between Bax/Bcl2 ratio, tissue oxidative stress level, and tissue injury. Cimetidine represents a very promising radioprotective agent with a potential differential beneficial effect on both cancer cells (inducing apoptosis) as previously proved through different studies and adjacent healthy cells (providing radioprotection via inhibiting apoptosis) as clearly demonstrated through this study, via its antioxidant effect and subsequent regulation of type 2 apoptotic pathway through modulation of Bax/Bcl2 ratio.


Subject(s)
Apoptosis/drug effects , Cimetidine/pharmacology , Oxidative Stress/drug effects , Radiation-Protective Agents/pharmacology , bcl-2-Associated X Protein/drug effects , Animals , Antioxidants/pharmacology , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Male , Rats , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism
13.
Pharmacology ; 105(3-4): 164-172, 2020.
Article in English | MEDLINE | ID: mdl-31865327

ABSTRACT

SMBA1 (small-molecule Bax agonists 1), a small molecular activator of Bax, is a potential anti-tumour agent. In the present study, we investigated the biological effects of SMBA1 on glioblastoma (GBM) cells. SMBA1 reduced the viabilities of U87MG, U251 and T98G cells in a time- and dose-dependent manner. Moreover, treatment with SMBA1 induced cell cycle arrest at the G2/M phase transition, accompanied by the downregulation of Cdc25c and cyclin B1 and the upregulation of p21. SMBA1 also induced apoptosis of GBM cells in a dose-dependent manner. Mechanistically, SMBA1 induced apoptosis via the intrinsic pathway. Silencing of Bax or ectopic expression of Bcl-2 significantly inhibited SMBA1-induced apoptosis. Moreover, SMBA1 inhibited the growth of U87MG xenograft tumours in vivo. Overall, SMBA1 shows anti-proliferative effects against GBM cells through activation of the intrinsic apoptosis pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Glioma/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Time Factors , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
14.
Dig Dis Sci ; 64(12): 3528-3538, 2019 12.
Article in English | MEDLINE | ID: mdl-31273592

ABSTRACT

BACKGROUND: α-Hederin has been shown promising anti-tumor potential against various cancer cell lines. However, reports about effects of α-hederin on esophageal squamous cell carcinoma (ESCC) are still unavailable. AIM: To investigate the inhibitory effects of α-hederin on ESCC and explore the underlying mechanism. METHODS: Human esophageal carcinoma cell line (Eca-109) was used for the experiment. Cell Counting Kit-8, flow cytometry, Hoechst 33258 staining, enhanced ATP assay kit, 2',7'-dichlorofluorescin diacetate, JC-1 kit, and Western bolt were used to assess the cell viability, cycle, apoptosis, cellular ATP content, reactive oxygen species (ROS) level, mitochondrial membrane potential (MMP), and protein expression, respectively, in vitro. Xenografted tumor model was constructed to evaluate the in vivo anti-tumor effects of α-hederin. RESULTS: Compared with control group, α-hederin significantly inhibited the proliferation, induced apoptosis of ESCC, and arrested the cell cycle in G1 phase (P < 0.05). α-Hederin induced the accumulation of ROS, decrement of ATP levels, and disruption of MMP (P < 0.05). The detection of mitochondrial and cytosol proteins showed that AIF, Apaf-1, and Cyt C were released and increased in cytoplasm, and then, caspase-3, caspase-9, and Bax were involved and increased, while Bcl-2 level was decreased (P < 0.05). Furthermore, the above changes were amplified in the group pretreated with L-buthionine sulfoximine, while N-acetyl-L-cysteine plays an opposite role (P < 0.05). Meanwhile, α-hederin significantly inhibited the growth of xenografted tumors with favorable safety. CONCLUSION: α-Hederin could inhibit the proliferation and induce apoptosis of ESCC via dissipation of the MMP with simultaneous ROS generation and activation of the mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Oleanolic Acid/analogs & derivatives , Reactive Oxygen Species/metabolism , Saponins/pharmacology , Adenosine Triphosphate/metabolism , Animals , Apoptosis Inducing Factor/drug effects , Apoptosis Inducing Factor/metabolism , Apoptotic Protease-Activating Factor 1/drug effects , Apoptotic Protease-Activating Factor 1/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D1/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytochromes c/drug effects , Cytochromes c/metabolism , Flow Cytometry , Humans , In Situ Nick-End Labeling , In Vitro Techniques , Male , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Oleanolic Acid/pharmacology , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
15.
Med Sci Monit ; 25: 436-442, 2019 Jan 15.
Article in English | MEDLINE | ID: mdl-30643111

ABSTRACT

BACKGROUND Radix Tetrastigma Hemsleyani Flavone (RTHF) has detoxification and anti- inflammation activity and is widely used. Here, we report that RTHF inhibits cell proliferation and induces apoptosis in cutaneous squamous cell carcinoma A431 cells and is a potential strategy for cancer therapy. MATERIAL AND METHODS A431 cells were cultured in different concentrations of RTHF. The inhibition of cell proliferation was assessed by MTT assay, cell apoptosis was shown through FCM, and cell invasion was assessed by Transwell methods. Enzyme proteasome assay was used to detect the activity of proteasome and DUB. Expression of apoptosis-related and ubiquitin proteasome pathway-associated proteins were assessed by PCR and Western blot. RESULTS RTHF obviously suppressed the proliferation and induced apoptosis of A431 cells in a dose-dependent manner. Transwell assay showed that RTHF inhibited the cell metastasis significantly. Enzyme proteasome assay show that the RTHF treatment of activity of proteasome and DUB was significantly lower than in control. RTHF increased the expression of Bax and inhibited Bcl-2, pro-caspase3, and pro-caspase9 activity. The expression of USP14, UCHL5, and POH1 decreased and ub-prs increased significantly in the treatment group. CONCLUSIONS Our study reveals that RTHF-mediated inhibition of DUBs and proteasome may provide a potential strategy for cancer therapy.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Flavones/metabolism , Flavones/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Caspase 3/drug effects , Caspase 9/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Humans , Medicine, Chinese Traditional/methods , Neoplasm Invasiveness , Plants, Medicinal/metabolism , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Signal Transduction , Ubiquitin Thiolesterase/metabolism , bcl-2-Associated X Protein/drug effects
16.
Exp Mol Pathol ; 105(3): 229-235, 2018 12.
Article in English | MEDLINE | ID: mdl-30184471

ABSTRACT

The aim of this study was to confirm the effects of ginsenoside Rb1 on neural cell apoptosis in the spinal cord of rats with spinal cord ischemia-reperfusion injury (SCII) and to explore its potential mechanisms. A total of 100 healthy adult Sprague-Dawley (SD) rats were randomly divided into four groups: normal control (n = 10), sham-operated (n = 10), SCII model (n = 40), and ginsenoside Rb1-treated groups (n = 40). Basso, Beattie, Bresnahan (BBB) scale was used to examine rat hindlimb locomotor function. Nissl and Tunnel staining were used to observe neural cell injury and apoptosis, respectively, in the spinal cord of rats with SCII. Immunofluorescence staining was performed to detect the expression of Bax and Bcl-2. The levels of caspase-3 and phosphorylated Ask-1 (p-Ask-1) were detected by western blotting. Ginsenoside Rb1 prevented neural cell apoptosis in the spinal cord and improved hindlimb locomotor dysfunction of rats (P < .05). Moreover, SCII-induced upregulation of caspase-3 and p-Ask-1 levels and the Bax/Bcl-2 ratio were significantly decreased by ginsenoside Rb1 (P < .05). The protective effects of ginsenoside Rb1 on neural cells in the spinal cord of rats with SCII were mediated by the ginsenoside Rb1-induced downregulation of caspase-3 and p-Ask-1 levels and the Bax/Bcl-2 ratio.


Subject(s)
Ginsenosides/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/pathology , Spinal Cord Injuries/pathology , Animals , Apoptosis/drug effects , Caspase 3/drug effects , Caspase 3/metabolism , Down-Regulation , Female , MAP Kinase Kinase Kinase 5/drug effects , MAP Kinase Kinase Kinase 5/metabolism , Male , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Spinal Cord Injuries/metabolism , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
17.
Microb Pathog ; 111: 193-197, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28867631

ABSTRACT

Nisin is a polycyclic peptide containing 34 amino acids produced by Lactococcus lactis during fermentation. Recently, researchers considered nisin as an anticancer peptide. Herein, the authors aim to evaluate the nisin effects on the apoptosis stimulation in the colon cancer cell line. The SW480 cells were exposed to discrepant concentrations of nisin and the cell viability as well as the expression of bcl-2 and bax genes and proteins were surveyed by the MTT assay, Real-Time PCR and western blotting method, respectively. Furthermore, the Ethidium bromide/Acridine orange staining was performed to visualize apoptotic cells. 4000, 3000, 2500 and 2000 µg/ml of nisin led to significant anti-proliferative impact and augmentation apoptotic index (bax/bcl-2 ratio) both at mRNA and protein levels (p < 0.05). Furthermore, the apoptotic impacts were demonstrated after Ethidium bromide/Acridine orange (EB/AO) staining to have a dose dependent manner. Our outcome suggested that nisin could induce apoptosis via intrinsic pathways and lead to cancerous cell death.


Subject(s)
Cell Line, Tumor/drug effects , Colonic Neoplasms/drug therapy , Nisin/pharmacology , Apoptosis/drug effects , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Nisin/administration & dosage , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/biosynthesis , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
18.
Childs Nerv Syst ; 33(11): 1937-1945, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28721600

ABSTRACT

OBJECTIVE: This study was aimed to investigate the mechanism of resveratrol on amelioration of hypoxia/ischemia (H/I)-induced brain injury. METHODS: The RT-PCR and western blot were used to detect the mRNA and protein expressions, respectively. The PC12 cell induced by OGD/R was as in vitro H/I brain injury model. The luciferase reporter assay was used to prove the relationship between Bax and miR-96, and the cell apoptosis was detected by MTT assay. The loss of MBP+ area in neonatal rats analyzed by immunohistochemistry was to evaluate the extent of brain injury. RESULTS: The miR-96 expression was decreased in the hippocampus and cerebral cortex of neonatal rats with H/I brain injury and the oxygenglucose deprivation/re-oxygenation (OGD/R)-induced PC12 cell, while Bax expression was opposite. And then the H/I rats and OGD/R-induced PC12 cell were treated with resveratrol (RSV); the results showed that the RSV could reverse the miR-96 and Bax expressions. Next, the luciferase reporter assay proved that Bax was a target of miR-96. We used the miR-96 inhibitor to suppress miR-96 expression in OGD/R-induced PC12 cell, and found that RSV regulated Bax expression and prevented OGD/R-induced PC12 cell apoptosis via miR-96. In addition, the immunohistochemistry was used to analyze the loss of MBP+ area in neonatal rats, and the result showed that the RSV significantly reduced the brain damage, increased miR-96 expression, and decreased Bax expression, while inhibition of miR-96 aggravated the brain damage and reversed the effect of RSV. CONCLUSION: Resveratrol ameliorates hypoxia/ischemia-induced brain injury in neonatal rat via the miR-96/ Bax axis.


Subject(s)
Hypoxia-Ischemia, Brain/pathology , MicroRNAs/metabolism , Neuroprotective Agents/pharmacology , Stilbenes/pharmacology , bcl-2-Associated X Protein/metabolism , Animals , Animals, Newborn , Brain/drug effects , Brain/metabolism , Hypoxia-Ischemia, Brain/metabolism , MicroRNAs/drug effects , PC12 Cells , Rats , Resveratrol , bcl-2-Associated X Protein/drug effects
19.
Acta Odontol Scand ; 75(1): 21-29, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27897090

ABSTRACT

OBJECTIVE: The aim of present study was to determine the effects of conjugated linoleic acid enriched milk on alveolar bone loss, hyperglycaemia, oxidative stress and apoptosis in ligature-induced periodontal disease in diabetic rat model. METHODS: Wistar rats were divided into six experimental groups: 1; non-ligated (NL, n = 6) group, 2; ligature only (LO, n = 6) group, 3; streptozotocin only (STZ, n = 8) group, 4; STZ and ligature (STZ + L, n = 8) group, 5; ligature and conjugated linoleic acid (CLA) (L + CLA, n = 8) group, 6; STZ, ligature and CLA group (STZ + L + CLA, n = 8) group. Diabetes mellitus was induced by 60 mg/kg streptozotocin. Rats were fed with CLA enriched milk for four weeks. Silk ligatures were placed at the gingival margin of lower first molars of mandibular quadrant. The study duration was four weeks after diabetes induction and the animals were sacrificed at the end of this period. Changes in alveolar bone levels were clinically measured and tissues were histopathologically examined. Inducible nitric oxide synthase (iNOS) and Bax protein expressions, serum interleukin-1ß (IL-1ß), low-density lipoprotein (LDL), high-density lipoprotein (HDL) and triglyceride levels and tartrate resistant acid phosphatase (TRAP)+ osteoclast numbers were also evaluated. RESULTS: At the end of four weeks, alveolar bone loss was significantly higher in the STZ + LO group compared to the other groups (p < .05). CLA decreased alveolar bone loss in L + CLA and STZ + L + CLA groups. CLA significantly decreased TRAP + osteoclast numbers and increased osteoblastic activity compared to the STZ + L group (p < .05). Diabetes and CLA increased Bax protein levels (p < .05) however CLA had no effect on iNOS expression (p > .05). CONCLUSION: Within the limits of this study, commercial CLA product administration in addition to diet significantly reduced alveolar bone loss, increased osteoblastic activity and decreased osteoclastic activity in the diabetic Wistar rats.


Subject(s)
Diabetes Mellitus, Experimental/complications , Linoleic Acids, Conjugated/therapeutic use , Periodontitis/prevention & control , Alveolar Bone Loss/prevention & control , Animals , Apoptosis/drug effects , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Hyperglycemia/prevention & control , Interleukin-1beta/blood , Liver/drug effects , Male , Nitric Oxide Synthase Type II/drug effects , Osteoblasts/drug effects , Osteoclasts/drug effects , Oxidative Stress/drug effects , Random Allocation , Rats , Rats, Wistar , Streptozocin , Tartrate-Resistant Acid Phosphatase/drug effects , Time Factors , Triglycerides/blood , bcl-2-Associated X Protein/drug effects
20.
Molecules ; 22(7)2017 Jul 07.
Article in English | MEDLINE | ID: mdl-28686182

ABSTRACT

Nereis active protease (NAP) is a novel fibrinolytic active serine protease from the polychaete Nereis virens. In this study, NAP was purified from Nereis virens and the effects of NAP on human lung cancer cells were investigated. Our results indicated that NAP inhibited the proliferation and induced apoptosis of H1299 cells in a time- and dose-dependent manner. The loss of mitochondrial membrane potential, the activation of Bax and cleaved-caspase 3/9, the release of cytochrome C, and the suppression of Bcl-2 and poly-ADP ribose polymerase were observed in NAP-treated H1299 cells by flow cytometry and Western blotting. Moreover, the expression levels of Bax and Bcl-2 mRNA were determined by real-time quantitative polymerase chain reaction and the Bax/Bcl-2 expression ratio was increased in the NAP-treated cell lines. The results indicated that NAP-induced apoptosis may be related to mitochondria mediated apoptosis and occurs through caspase-dependent pathways. Then, the effects of NAP on tumor growth in animal models were observed, where 5 or 10 mg/kg of NAP noticeably reduced tumor volume and weight and increased apoptosis as determined by Western blotting when compared to the negative control group. Therefore, our findings suggest that NAP could be a hopeful anticancer medicine for its propensity to inhibit growth and induce of apoptosis in human lung cancer cells.


Subject(s)
Annelida/metabolism , Lung Neoplasms/drug therapy , Serine Proteases/isolation & purification , Serine Proteases/pharmacology , Animals , Apoptosis/drug effects , Caspases/drug effects , Cell Line, Tumor , Cell Proliferation , Cytochromes c/metabolism , Enzyme Activation , Female , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Optical Imaging/methods , Poly(ADP-ribose) Polymerases/metabolism , Serine Proteases/chemistry , bcl-2-Associated X Protein/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL