1
|
Turgut GÇ, Pepe NA, Ekiz YC, Şenol H, Şen A. Therapeutic Potential of Nitrogen-Substituted Oleanolic Acid Derivatives in Neuroinflammatory and Cytokine Pathways: Insights From Cell-Based and Computational Models. Chem Biodivers 2025:e202500269. [PMID: 40262123 DOI: 10.1002/cbdv.202500269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/06/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
This study was conducted to investigate the mechanism of the potential and anti-inflammatory properties of nitrogen-substituted oleanolic acid derivatives that can be used to treat neuroinflammatory diseases. Nitrogen-containing oleanolic acid derivatives have been evaluated for their anti-neuroinflammatory effects in vitro in neuronal and monocytic cell lines at nontoxic doses, and the production of cytokines (TNF-α, IL-6 and IL-17), the inflammatory enzyme induced nitric oxide synthase (iNOS) and NF-κB signalling under LPS-stimulated conditions, and the expression of genes associated with Alzheimer's disease have been assessed. In addition, molecular docking and molecular dynamics simulation assessments are conducted in silico. Key protein markers of neurodegenerative diseases, especially Alzheimer's disease and neuroinflammation, TAU protein levels, and microglial activation, as well as ionised calcium-binding adaptor protein-1 (IBA1) levels, were significantly reduced with the addition of oleanolic acid derivatives. LPS-induced NF-κB luciferase reporter activity and iNOS activity were significantly inhibited, approaching the levels in uninduced controls. The mRNA expression of proinflammatory cytokines critical for neuroinflammation, such as TNF-α, NF-κB, IL-6 and IL-17, was reduced twofold to sevenfold. Furthermore, the molecular docking and MD simulation analyses revealed potential interactions with the TNF-α and NF-κB proteins. These findings underscore the potential of oleanolic acid derivatives, particularly compound 16, as candidates for further development as therapeutic agents for neurodegenerative diseases associated with chronic inflammation.
Collapse
Affiliation(s)
- Gurbet Çelik Turgut
- Department of Organic Agriculture Management, Pamukkale University, Denizli, Türkiye
| | - Nihan Aktaş Pepe
- Department of Molecular Biology & Genetics, Faculty of Life & Natural Sciences, Abdullah Gül University, Kayseri, Türkiye
| | - Yağmur Ceylan Ekiz
- Department of Organic Agriculture Management, Pamukkale University, Denizli, Türkiye
| | - Halil Şenol
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Türkiye
| | - Alaattin Şen
- Department of Molecular Biology & Genetics, Faculty of Life & Natural Sciences, Abdullah Gül University, Kayseri, Türkiye
- Department of Biology, Faculty of Science, Pamukkale University, Denizli, Türkiye
| |
Collapse
|
2
|
Subramanian S, Pajaniradje S, Bhat SA, Chandramohan S, Anaikutti P, Rajagopalan R. Cytotoxic potential of an indole-conjugated Oleanolic acid analogue: suppression of NSCLC proliferation through modulation of mitochondrial apoptotic dynamics. Toxicol Mech Methods 2025:1-14. [PMID: 40103544 DOI: 10.1080/15376516.2025.2481915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
Pre-clinical toxicological investigations are pivotal in the development of safer and more efficacious chemotherapeutic agents. Oleanolic acid (OA), a naturally occurring pentacyclic triterpenoid, has demonstrated anticancer potential but is often limited by the toxic side effects of its derivatives. In the current study, we carried out the facile synthesis of a modified OA analogue, OD2, and studied its cytotoxicity and efficacy analysis across several cell lines. Mechanistic toxicology was explored through fluorescence-based assays. Annexin-V/Propidium Iodide (A-V/PI) staining and TUNEL assays were used to confirm apoptosis. OD2 exhibited dose-dependent cytotoxicity, with a pronounced effect on A549 lung cancer cells compared to other cancerous and non-cancerous cell lines. Apoptosis was found to be the predominant mode of cell death, evidenced by Fluorescence imaging analysis of chromatin condensation and mitochondrial dysfunction. This was further validated by an increase in Annexin-V-positive and TUNEL-positive cells in treated groups. OD2 activated the intrinsic mitochondrial apoptotic pathway as evidenced by increased Bax and decreased Bcl-2 protein abundance levels. While the current study showcases the therapeutic potential of the selective toxicological activity of OD2, future studies will focus on the deconvolution of its potential polypharmacological mode of action and decoding the basis of its selective action, so as to glean important lessons that can be applied in the development of chemotherapeutic agents with favorable toxicological profiles.
Collapse
Affiliation(s)
- Srividya Subramanian
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Sankar Pajaniradje
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | - Suhail Ahmad Bhat
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | | - Parthiban Anaikutti
- General Pathology, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Rukkumani Rajagopalan
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| |
Collapse
|
3
|
Zhai Q, Shang S, Zhang Z, Sun L, Huang Y, Feng S, Wu Q, Cui H, Shi X. Mechanism of salvianolic phenolic acids and hawthorn triterpenic acids combination in intervening atherosclerosis: network pharmacology, molecular docking, and experimental validation. Front Pharmacol 2025; 16:1501846. [PMID: 39950115 PMCID: PMC11821658 DOI: 10.3389/fphar.2025.1501846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
Background This study employs network pharmacology and molecular docking methods in conjunction with animal experimentation to elucidate the underlying mechanism by which the combination of salvianolic phenolic acids and hawthorn triterpenic acids (SHC) exerts its therapeutic effect on carotid atherosclerosis (AS) in ApoE-/- mice. Methods A network pharmacology research approach was used to predict potential core targets for SHC intervention in atherosclerosis. The predictions were subsequently validated through the implementation of animal in vivo experiments. ApoE-/- mice were randomly assigned to three experimental groups, namely, a model group, an atorvastatin group, and an SHC group. After the administration period, the plaque area in the carotid artery and aortic arch, blood lipid levels, malondialdehyde (MDA), superoxide dismutase (SOD), glutathione (GSH), and nitric oxide (NO) content were measured. Additionally, the expression of PI3K, Akt, NF-κB, JNK1, ERK1/2, and p38-MAPK in the aortic arteries was analyzed. Based on the protein expression results, molecular docking was used to predict the binding activity between the core compounds and core targets. Results A total of 23 core compounds were identified in SHC, and 55 core targets of SHC were screened as potential targets for intervention in AS. The results of the enrichment analysis indicated that the principal mechanisms through which SHC exerts its effects in AS are associated with lipid metabolism and the PI3K-Akt and MAPK pathways. The results from animal experiments demonstrated that atorvastatin and SHC markedly reduced the area of carotid plaque and downregulated the levels of TC and LDL-C in ApoE-/- mice. The administration of SHC was associated with an increase in SOD activity and a reduction in NO levels in the livers of mice. Furthermore, SHC was observed to downregulate the expression of NF-κB and p38-MAPK in the carotid region. The results of molecular docking demonstrated that the core compounds of SHC, including salvianolic acid A, B, and C, maslinic acid, ursolic acid, and oleic acid, were capable of stably binding to the core targets NF-κB and MAPK14. Conclusion It is hypothesized that SHC may reduce lipid deposition and plaque formation in AS by regulating blood lipids, a process that may be closely linked to the inhibition of inflammatory regulator expression, including NF-κB and p38-MAPK.
Collapse
Affiliation(s)
- Qu Zhai
- Institute of Executive Development, China National Medical Products Administration, Beijing, China
| | - Shixi Shang
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zihan Zhang
- Beijing University of Chinese Medical, Beijing, China
| | - Lihua Sun
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ying Huang
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuyi Feng
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Wu
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haifeng Cui
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Shi
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Ko HH, Chou HYE, Hou HH, Kuo WT, Liu WW, Yen-Ping Kuo M, Cheng SJ. Oleanolic acid inhibits aldo-keto reductase family 1 member B10-induced cancer stemness and avoids cisplatin-based chemotherapy resistance via the Snail signaling pathway in oral squamous cell carcinoma cell lines. J Dent Sci 2025; 20:100-108. [PMID: 39873100 PMCID: PMC11762581 DOI: 10.1016/j.jds.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/22/2024] [Indexed: 01/30/2025] Open
Abstract
Background/purpose Oral squamous cell carcinoma (OSCC) is a common malignancy often associated with poor prognosis due to chemoresistance. In this study, we investigated whether arecoline, a major alkaloid in betel nuts, can stimulate aldo-keto reductase family 1 member B10 (AKR1B10) levels in OSCC, promoting cancer stemness and leading to resistance to cisplatin (CDDP)-based chemotherapy. Materials and methods Gain- and Loss- of AKR1B10 functions were analyzed using WB and q-PCR of OSCC cells. Stemness, epithelial mesenchymal transition (EMT) markers, and CDDP drug resistance in overexpressed AKR1B10 were also identified. Results Upregulated AKR1B10 in OSCC significantly increased cell motility and aggregation. The results also showed that the canonical TGF-β1-Smad3 pathway was involved in arecoline-induced AKR1B10 expression, further increasing cancer stemness with CDDP resistance via the Snail-dependent EMT pathway. Moreover, oleanolic acid (OA) and ROS/RNS (reactive oxygen/nitrogen species) inhibitors effectively reversed AKR1B10-induced CDDP-resistance. Conclusion Arecoline-induced ROS/RNS to hyper-activate AKR1B10 in tumor sphere cells via the TGF-β1-Smad3 pathway. Furthermore, AKR1B10 enhanced CDDP resistance in OSCC cells via EMT-inducing markers. Finally, Finally, OA may efficiently target CDDP resistance, reverse stemness in OSCC cells, and have the potential as a novel anticancer drug.
Collapse
Affiliation(s)
- Hui-Hsin Ko
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital Hsin-Chu Branch, Hsin-Chu, Taiwan
| | - Han-Yi E. Chou
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Hsin-Han Hou
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Ting Kuo
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Wei-Wen Liu
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| | - Mark Yen-Ping Kuo
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
| | - Shih-Jung Cheng
- Graduate Institute of Clinical Dentistry, School of Dentistry, National Taiwan University, Taipei, Taiwan
- School of Dentistry, National Taiwan University, Taipei, Taiwan
- Department of Dentistry, National Taiwan University Hospital, College of Medicine, Taipei, Taiwan
- Graduate Institute of Oral Biology, School of Dentistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Zeng Y, Wang Z, Zhang J, Jian W, Fu Q. Antitumour activity of oleanolic acid: A systematic review and meta‑analysis. Oncol Lett 2024; 28:582. [PMID: 39421313 PMCID: PMC11484195 DOI: 10.3892/ol.2024.14715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
Oleanolic acid (OA), a compound known for its potent antitumour properties, has been the subject of investigations in both cell and animal models. Although OA has good biological activity, its low water solubility and bioavailability limit its therapeutic use, and therefore translating the potential of OA into the clinical oncology setting remains challenging. The present systematic review and meta-analysis utilized evidence from animal model studies to gain insights into the antitumour mechanisms of OA to address the gap in understanding, and to provide guidance for future research directions and potential clinical applications. The guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses were applied in the present study and a comprehensive search was conducted across the PubMed/MEDLINE, Web of Science, Cochrane Library and Embase databases, with a cut-off date of June 30, 2023. The primary focus was on randomized controlled trials that used animal models to assess the antitumour effects of OA. The methodological quality appraisal was conducted using the Systematic Review Centre for Laboratory Animal Experimentation risk of bias tool, and tumour volume and weight served as the principal outcome measures. Data were analysed using the RevMan (version 5.3) and Stata SE11 software packages, with an assessment of heterogeneity conducted using the I2 statistical test, sensitivity analysis conducted using the leave-one-out approach, and evaluation of publication bias performed using Egger's test and funnel plot analysis. The present study demonstrated a significant inhibitory effect of OA intervention on tumour growth and a decrease in tumour weight in animal models. Despite the broad spectrum of antitumour effects exhibited by OA, further investigations are warranted to optimize the dosage and administration routes of OA to maximize its efficacy in clinical cancer treatment.
Collapse
Affiliation(s)
- Ying Zeng
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zhonglian Wang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Jing Zhang
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Wei Jian
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Qiaofen Fu
- Department of Radiation Oncology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
6
|
Mattingly A, Vickery Z, Ivankovic D, Farrell CL, Hakonarson H, Nguyen K, Boccuto L. Exploring the Therapeutic Potential for Breast Cancer of Phytochemicals and Secondary Metabolites in Marjoram, Thyme, and Persimmon. Metabolites 2024; 14:652. [PMID: 39728433 DOI: 10.3390/metabo14120652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Background/Objectives: Breast cancer is the most common cause of death in women worldwide and the most commonly diagnosed cancer. Although several therapeutic approaches are widely used against breast cancer, their adverse effects often lead to symptoms severely affecting the quality of life. Alternative methods have been explored to reduce these adverse effects, and nutraceuticals have yielded promising results. This review will discuss mechanisms of action and potential applications against breast cancer of some nutraceuticals, specifically marjoram, thyme, and persimmon leaves. Methods: A systematic search was conducted across the public databases of PubMed, PubChem, and Google Scholar, with a specific focus on the plant extracts and phytochemicals of interest, as well as the anticarcinogenic mechanisms. Results: Ethnopharmacological and biochemical evidence support the anticarcinogenic role of marjoram, thyme, and persimmon. Numerous phytochemicals contained in these herbs' extracts, like terpenes and flavonoids, possess remarkable potential to effectively treat breast cancer. Discussion: The phytochemicals contained in the reviewed nutraceuticals target the main cellular pathways involved in cell growth and disrupted in carcinogenesis, such as Nf-κB, MAPK/p38, TNF-α/IL-1β, and PI3K/Akt. The mechanisms of action of these compounds can successfully limit the abnormal growth and proliferation of cancerous breast cells. Conclusions: The potential use of the phytochemicals discussed in this review, either alone or in combination, may offer a valid alternative to chemotherapy against breast cancer with virtually no adverse effects, and further research on these molecules may lead to the identification of additional chemo-preventative and chemotherapeutic candidates.
Collapse
Affiliation(s)
- Aubrey Mattingly
- Healthcare Genetics Laboratory, School of Nursing, Clemson University, Clemson, SC 29634, USA
| | - Zoe Vickery
- Healthcare Genetics Laboratory, School of Nursing, Clemson University, Clemson, SC 29634, USA
| | - Diana Ivankovic
- Center for Cancer Research, Anderson University, Anderson, SC 29621, USA
| | - Christopher L Farrell
- Healthcare Genetics Laboratory, School of Nursing, Clemson University, Clemson, SC 29634, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Katie Nguyen
- Healthcare Genetics Laboratory, School of Nursing, Clemson University, Clemson, SC 29634, USA
| | - Luigi Boccuto
- Healthcare Genetics Laboratory, School of Nursing, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
7
|
Park SY, Mun JG, Lee YS, Lee SB, Kim SJ, Jang JH, Kim HY, Hong SH, Kee JY. Inhibitory Effect of Alnustone on Survival and Lung Metastasis of Colorectal Cancer Cells. Nutrients 2024; 16:3737. [PMID: 39519569 PMCID: PMC11547205 DOI: 10.3390/nu16213737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Alnustone (Aln) is an effective compound of Alpinia katsumadae Hayata. Aln possesses various pharmacological activities such as antibacterial, anti-inflammatory, and anti-cancer effects. However, the inhibitory effect of Aln on colorectal cancer (CRC) has not yet been identified. Thus, research was conducted to clarify whether Aln can suppress the proliferative and metastatic ability of CRC cells. METHODS A cell viability assay was performed to confirm the decrease in CRC cell viability following Aln treatment. Flow cytometry was carried out to evaluate the effects of Aln on cell cycle arrest, autophagy, and apoptosis in CRC cells. In addition, a lung metastasis animal model was used to check the inhibitory effect of Aln on the metastasis of CRC cells. RESULTS Aln remarkably diminished the viability and colony-forming ability of several CRC cell lines. In addition, Aln led to a halt at the G0/G1 phase through downregulating cyclin D1-CDK4 in CRC cells. The upregulation of LC3B and p62 expression by Aln triggered autophagy of CRC cells. Moreover, Aln promoted mitochondrial depolarization, resulting in apoptosis of CRC cells. Oral administration of Aln significantly restrained the metastasized lung tumor nodules. CONCLUSIONS This study demonstrated that Aln can suppress the survival and lung metastasis of CRC cells by promoting cell cycle arrest, autophagy, and apoptosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Seung-Heon Hong
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea; (S.-Y.P.); (J.-G.M.); (Y.-S.L.); (S.-B.L.); (S.-J.K.); (J.-H.J.); (H.-Y.K.)
| | - Ji-Ye Kee
- Department of Oriental Pharmacy, College of Pharmacy, Wonkwang-Oriental Medicines Research Institute, Wonkwang University, 460 Iksandae-ro, Iksan 54538, Jeonbuk, Republic of Korea; (S.-Y.P.); (J.-G.M.); (Y.-S.L.); (S.-B.L.); (S.-J.K.); (J.-H.J.); (H.-Y.K.)
| |
Collapse
|
8
|
Sarg NH, Zaher DM, Abu Jayab NN, Mostafa SH, Ismail HH, Omar HA. The interplay of p38 MAPK signaling and mitochondrial metabolism, a dynamic target in cancer and pathological contexts. Biochem Pharmacol 2024; 225:116307. [PMID: 38797269 DOI: 10.1016/j.bcp.2024.116307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Mitochondria play a crucial role in cellular metabolism and bioenergetics, orchestrating various cellular processes, including energy production, metabolism, adaptation to stress, and redox balance. Besides, mitochondria regulate cellular metabolic homeostasis through coordination with multiple signaling pathways. Importantly, the p38 mitogen-activated protein kinase (MAPK) signaling pathway is a key player in the intricate communication with mitochondria, influencing various functions. This review explores the multifaced interaction between the mitochondria and p38 MAPK signaling and the consequent impact on metabolic alterations. Overall, the p38 MAPK pathway governs the activities of key mitochondrial proteins, which are involved in mitochondrial biogenesis, oxidative phosphorylation, thermogenesis, and iron homeostasis. Additionally, p38 MAPK contributes to the regulation of mitochondrial responses to oxidative stress and apoptosis induced by cancer therapies or natural substances by coordinating with other pathways responsible for energy homeostasis. Therefore, dysregulation of these interconnected pathways can lead to various pathologies characterized by aberrant metabolism. Consequently, gaining a deeper understanding of the interaction between mitochondria and the p38 MAPK pathway and their implications presents exciting forecasts for novel therapeutic interventions in cancer and other disorders characterized by metabolic dysregulation.
Collapse
Affiliation(s)
- Nadin H Sarg
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Dana M Zaher
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nour N Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Salma H Mostafa
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hussein H Ismail
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A Omar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
9
|
Oh SH, Yook JM, Jung HY, Choi JY, Cho JH, Park SH, Kim CD, Kim YL, Lim JH. Autophagy caused by oxidative stress promotes TGF-β1-induced epithelial-to-mesenchymal transition in human peritoneal mesothelial cells. Cell Death Dis 2024; 15:365. [PMID: 38806451 PMCID: PMC11133371 DOI: 10.1038/s41419-024-06753-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 05/15/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Epithelial-to-mesenchymal transition (EMT) is one of the main causes of peritoneal fibrosis. However, the pathophysiological mechanisms of EMT, specifically its relationship with autophagy, are still unknown. This study aimed to evaluate the role of autophagy in transforming growth factor-beta 1 (TGF-β1)-induced EMT in human peritoneal mesothelial cells (HPMCs). Primary cultured HPMCs were treated with TGF-β1 (2 and 5 ng/mL) and changes in autophagy markers and the relationship between autophagy and EMT were evaluated. We also identified changes in EMT- and autophagy-related signaling pathways after autophagy and NADPH oxidase 4 (NOX4) inhibition. TGF-β1 increased the generation of NOX4 and reactive oxygen species (ROS) in HPMCs, resulting in mitochondrial damage. Treatment with GKT137831 (20 μM), a NOX1/4 inhibitor, reduced ROS in the mitochondria of HPMC cells and reduced TGF-β1-induced mitochondrial damage. Additionally, the indirect inhibition of autophagy by GKT137831 (20 μM) downregulated TGF-β1-induced EMT, whereas direct inhibition of autophagy using 3-methyladenine (3-MA) (2 mM) or autophagy-related gene 5 (ATG5) gene silencing decreased the TGF-β1-induced EMT in HPMCs. The suppressor of mothers against decapentaplegic 2/3 (Smad2/3), autophagy-related phosphoinositide 3-kinase (PI3K) class III, and protein kinase B (Akt) pathways, and mitogen-activated protein kinase (MAPK) signaling pathways, such as extracellular signal-regulated kinase (ERK) and P38, were involved in TGF-β1-induced EMT. Autophagy and NOX4 inhibition suppressed the activation of these signaling pathways. Direct inhibition of autophagy and its indirect inhibition through the reduction of mitochondrial damage by upstream NOX4 inhibition reduced EMT in HPMCs. These results suggest that autophagy could serve as a therapeutic target for the prevention of peritoneal fibrosis in patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
- Se-Hyun Oh
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Ju-Min Yook
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hee-Yeon Jung
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Ji-Young Choi
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Jang-Hee Cho
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Sun-Hee Park
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Chan-Duck Kim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Yong-Lim Kim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Jeong-Hoon Lim
- Division of Nephrology, Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea.
- Cell and Matrix Research Institute, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
10
|
Han EJ, Choi EY, Jeon SJ, Moon JM, Lee SW, Lee JH, Jung GH, Han SH, Jung SH, Yang MS, Jung JY. Anticancer Effects of α-Pinene in AGS Gastric Cancer Cells. J Med Food 2024; 27:330-338. [PMID: 38387002 DOI: 10.1089/jmf.2023.k.0267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024] Open
Abstract
Gastric cancer is the fifth most common cancer globally and the third leading cause of cancer-related mortality. Existing treatment strategies for gastric cancer often present numerous side effects. Consequently, recent studies have shifted toward devising new treatments grounded in safer natural substances. α-Pinene, a natural terpene found in the essential oils of various plants, such as Lavender angustifolia and Satureja myrtifolia, displays antioxidant, antibiotic, and anticancer properties. Yet, its impact on gastric cancer remains unexplored. This research assessed the effects of α-pinene in vitro using a human gastric adenocarcinoma cell-line (AGS) human gastric cancer cells and in vivo via a xenograft mouse model. The survival rate of AGS cells treated with α-pinene was notably lower than that of the control group, as revealed by the 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide assay. This decline in cell viability was linked to apoptosis, as verified by 4',6-diamidino-2-phenylindole and annexin V/propidium iodide staining. The α-pinene-treated group exhibited elevated cleaved-poly (ADP-ribose) polymerase and B cell lymphoma 2 (Bcl-2)-associated X (Bax) levels and reduced Bcl-2 levels compared with the control levels. Moreover, α-pinene triggered the activation of extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38 within the mitogen-activated protein kinase (MAPK) pathway. In the xenograft mouse model, α-pinene induced apoptosis through the MAPK pathway, devoid of toxicity. These findings position α-pinene as a promising natural therapeutic for gastric cancer.
Collapse
Affiliation(s)
- Eun-Ji Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Eun-Young Choi
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Su-Ji Jeon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Jun-Mo Moon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Sang-Woo Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Gi-Hwan Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Myeon-Sik Yang
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Korea
- Research Institute for Natural Products, Kongju National University, Yesan, Korea
- Research Center of Crop Breeding for Omics and Artificial Intelligence, Yesan, Korea
| |
Collapse
|
11
|
Hassan S, Ahmad B, Khan MW, Shah ZA, Ullah A, Ullah S, Khan D, Rizwan M, Ahmad A, Ali Q, Kaushik P, Yilmaz S. Cytotoxic Activity of Phytoconstituents Isolated from Monotheca buxifolia against Hepatocellular Carcinoma Cell Line HepG2: In Vitro and Molecular Docking Studies. ACS OMEGA 2023; 8:33572-33579. [PMID: 37744864 PMCID: PMC10515345 DOI: 10.1021/acsomega.3c03705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023]
Abstract
Natural products and conventional chemotherapeutic drugs are believed to enhance anticancer treatment efficacy while lowering toxicity. The current study investigates the cytotoxic and apoptogenic effects of Monotheca buxifolia bioactive compounds on HepG2 cell lines. MTT assay was used to assess the effect on the viability of HepG2 cells. Morphological changes were investigated. Annexin-V-FITC/PI was used to demonstrate apoptotic activity. A molecular dynamics simulation study was carried out to investigate the compound binding pattern in the active site of the PPRAδ protein. MTT and annexin V-FITC/PI assays revealed that the isolated compounds lauric acid, oleanolic acid, and bis(2-ethylhexyl) phthalate inhibited the growth of hepatocellular cancer cells. The IC50 value for lauric acid was 56.46 ± 1.20 μg/mL, 31.94 ± 1.03 μg/mL for oleanolic acid, and 83.80 ± 2.18 μg/mL for bis(2-ethylhexyl) phthalate. Apoptosis was observed in 29.5, 52.1 and 22.4% of HepG2 cells treated with lauric acid, oleanolic acid, and bis(2-ethylhexyl) phthalate, respectively, after 24 h of treatment. Morphological assays and Hoechst staining microscopy revealed that the treatment caused morphological changes in the cell membrane and nuclear condensation. The high fluctuation indicates that various interactions were highly potent and widely adopted, and vice versa. Oleanolic acid displayed high residue fluctuation, remaining stable in the active site of the PPRAδ protein and involved in various interactions while remaining locally fluctuating in the binding sites of the other two compounds. These findings concluded that lauric acid, oleanolic acid, and bis(2-ethylhexyl) phthalate have a significant apoptogenic effect against HepG2 cells in inducing apoptosis. Our findings suggest that these bioactive compounds could be used as adjuvant therapies.
Collapse
Affiliation(s)
- Said Hassan
- Institute
of Biotechnology and Microbiology, Bacha
Khan University Charsadda, Charsadda 24420, Pakistan
| | - Bashir Ahmad
- Center
of Biotechnology and Microbiology, University
of Peshawar, Peshawar 25120, Pakistan
| | - Muhammad Waseem Khan
- Institute
of Pharmaceutical Sciences, Khyber Medical
University, Peshawar 25001, Pakistan
| | - Zafar Abbass Shah
- Department
of Bioinformatics, Hazara University Mansehra, Mansehra 21300, Pakistan
| | - Amin Ullah
- Department
of Health & Biological Sciences, Abasyn
University, Peshawar 25120, Pakistan
| | - Sana Ullah
- Department
of Zoology, Division of Science and Technology, University of Education, Lahore 54000, Pakistan
| | - Dilfaraz Khan
- Institute
of Chemical Sciences, Gomal University Dera Ismail Khan 29050, Pakistan
| | - Muhammad Rizwan
- Center
for Biotechnology and Microbiology, Swat
University, Swat 19130, Pakistan
| | - Ajaz Ahmad
- Department
of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Qurban Ali
- Department
of Plant Breeding and Genetics, Faculty of Agricultural Sciences, University of the Punjab Lahore 54590, Pakistan
| | - Prashant Kaushik
- Instituto de Conservación y Mejora
de la Agrodiversidad Valenciana, Universitat
Politècnica de València, Valencia 46022, Spain
| | - Semih Yilmaz
- Department of Agriculture Biotechnology,
Division of Enzyme and Microbial
Biotechnology, Erciyes University, Talas 38280, Turkey
| |
Collapse
|
12
|
Senobar Tahaei SA, Kulmány Á, Minorics R, Kiss A, Szabó Z, Germán P, Szebeni GJ, Gémes N, Mernyák E, Zupkó I. Antiproliferative and Antimetastatic Properties of 16-Azidomethyl Substituted 3- O-Benzyl Estrone Analogs. Int J Mol Sci 2023; 24:13749. [PMID: 37762056 PMCID: PMC10531082 DOI: 10.3390/ijms241813749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Four diastereomers of 16-azidomethyl substituted 3-O-benzyl estradiol (1-4) and their two estrone analogs (16AABE and 16BABE) were tested for their antiproliferative properties against human gynecological cancer cell lines. The estrones were selected for additional experiments based on their outstanding cell growth-inhibiting activities. Both compounds increased hypodiploid populations of breast cancer cells, and 16AABE elicited cell cycle disturbance as evidenced by flow cytometry. The two analogs substantially increased the rate of tubulin polymerization in vitro. 16AABE and 16BABE inhibited breast cancer cells' migration and invasive ability, as evidenced by wound healing and Boyden chamber assays. Since both estrone analogs exerted remarkable estrogenic activities, as documented by a luciferase reporter gene assay, they can be considered as promising drug candidates for hormone-independent malignancies.
Collapse
Affiliation(s)
| | - Ágnes Kulmány
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary
| | - Renáta Minorics
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary
| | - Anita Kiss
- Department of Inorganic, Organic and Analytical Chemistry, University of Szeged, H-6720 Szeged, Hungary
| | - Zoltán Szabó
- Department of Medicinal Chemistry, University of Szeged, H-6720 Szeged, Hungary
| | - Péter Germán
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary
| | - Gábor J. Szebeni
- Laboratory of Functional Genomics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Nikolett Gémes
- Laboratory of Functional Genomics, Biological Research Centre, H-6726 Szeged, Hungary
| | - Erzsébet Mernyák
- Department of Inorganic, Organic and Analytical Chemistry, University of Szeged, H-6720 Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, H-6720 Szeged, Hungary
- Interdisciplinary Centre of Natural Products, University of Szeged, H-6720 Szeged, Hungary
| |
Collapse
|
13
|
Nizami ZN, Aburawi HE, Semlali A, Muhammad K, Iratni R. Oxidative Stress Inducers in Cancer Therapy: Preclinical and Clinical Evidence. Antioxidants (Basel) 2023; 12:1159. [PMID: 37371889 DOI: 10.3390/antiox12061159] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/18/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Reactive oxygen species (ROS) are metabolic byproducts that regulate various cellular processes. However, at high levels, ROS induce oxidative stress, which in turn can trigger cell death. Cancer cells alter the redox homeostasis to facilitate protumorigenic processes; however, this leaves them vulnerable to further increases in ROS levels. This paradox has been exploited as a cancer therapeutic strategy with the use of pro-oxidative drugs. Many chemotherapeutic drugs presently in clinical use, such as cisplatin and doxorubicin, induce ROS as one of their mechanisms of action. Further, various drugs, including phytochemicals and small molecules, that are presently being investigated in preclinical and clinical studies attribute their anticancer activity to ROS induction. Consistently, this review aims to highlight selected pro-oxidative drugs whose anticancer potential has been characterized with specific focus on phytochemicals, mechanisms of ROS induction, and anticancer effects downstream of ROS induction.
Collapse
Affiliation(s)
- Zohra Nausheen Nizami
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Hanan E Aburawi
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire-Université Laval, Quebec, QC G1V 0A6, Canada
| | - Khalid Muhammad
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, Al Ain PO Box 15551, United Arab Emirates
| |
Collapse
|
14
|
Martins-Gomes C, Nunes FM, Silva AM. Modulation of Cell Death Pathways for Cellular Protection and Anti-Tumoral Activity: The Role of Thymus spp. Extracts and Their Bioactive Molecules. Int J Mol Sci 2023; 24:ijms24021691. [PMID: 36675206 PMCID: PMC9864824 DOI: 10.3390/ijms24021691] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/18/2023] Open
Abstract
Natural products used for their health-promoting properties have accompanied the evolution of humanity. Nowadays, as an effort to scientifically validate the health-promoting effects described by traditional medicine, an ever-growing number of bioactivities are being described for natural products and the phytochemicals that constitute them. Among them, medicinal plants and more specifically the Thymus genus spp., arise as products already present in the diet and with high acceptance, that are a source of phytochemicals with high pharmacological value. Phenolic acids, flavonoid glycoside derivatives, and terpenoids from Thymus spp. have been described for their ability to modulate cell death and survival pathways, much-valued bioactivities in the pharmaceutical industry, that continually sought-after new formulations to prevent undesired cell death or to control cell proliferation. Among these, wound treatment, protection from endogenous/exogenous toxic molecules, or the induction of selective cell death, such as the search for new anti-tumoral agents, arise as main objectives. This review summarizes and discusses studies on Thymus spp., as well as on compounds present in their extracts, with regard to their health-promoting effects involving the modulation of cell death or survival signaling pathways. In addition, studies regarding the main bioactive molecules and their cellular molecular targets were also reviewed. Concerning cell survival and proliferation, Thymus spp. present themselves as an option for new formulations designed for wound healing and protection against chemicals-induced toxicity. However, Thymus spp. extracts and some of their compounds regulate cell death, presenting anti-tumoral activity. Therefore Thymus spp. is a rich source of compounds with nutraceutical and pharmaceutical value.
Collapse
Affiliation(s)
- Carlos Martins-Gomes
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Lab, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Lab, UTAD Quinta de Prados, 5001-801 Vila Real, Portugal
| | - Fernando M. Nunes
- Chemistry Research Centre-Vila Real (CQ-VR), Food and Wine Chemistry Lab, UTAD Quinta de Prados, 5001-801 Vila Real, Portugal
- Department of Chemistry, School of Life Sciences and Environment, UTAD, 5001-801 Vila Real, Portugal
| | - Amélia M. Silva
- Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Cell Biology and Biochemistry Lab, University of Trás-os-Montes and Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
- Department of Biology and Environment, School of Life Sciences and Environment, UTAD, 5001-801 Vila Real, Portugal
- Correspondence: ; Tel.: +351-259-350-921
| |
Collapse
|
15
|
Leenutaphong P, Tancharoen S, Nararatwanchai T, Phruksaniyom C, Sarikaphuti A, Palungwachira P, Chaichalotornkul S. Induction of Human Oral Squamous Carcinoma Apoptosis by Derris scandens Benth and Elephantopus scaber Linn Extracts. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221107970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
D scandens ( Derris scandens Benth.) and E scaber ( Elephantopus scaber Linn.) contain flavonoids and phenolic acids, which have antitumor activity in various cancer cell lines. Oral cancer is among the most common cancers in Southeast Asia, and the survival rate remains low. Thus, this study screened 2 ethanolic plant extracts for cytotoxicity on the oral human squamous carcinoma cell line (HSC-2), and compared the mechanisms of action. Extracts of D scandens and E scaber showed cytotoxicity against HSC-2 cells in a dose-dependent manner. Observation of nuclear morphology by Hoechst 33342 staining revealed chromatin condensation. Apoptosis was confirmed by Annexin V-FITC staining and cell sorting (fluorescence-activated cell sorting) analysis. We demonstrated that cancer apoptosis was accompanied by changes in the expression of procaspase 3 and that D scandens-mediated apoptosis in HSC-2 cells was potentiated by protein kinase B (Akt) and B-cell lymphoma-2 (Bcl-2), while E scaber apoptosis was mediated by mitogen-activated protein kinase (MAPK) pathways, involving stress-activated protein kinases/jun amino-terminal kinase (SAPK/JNK) and p38-MAPK. Further investigation into targets for apoptosis induction by these plant extracts may have potential in oral cancer therapy.
Collapse
Affiliation(s)
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | | | | | - Ariya Sarikaphuti
- School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Chiang Rai, Thailand
| | - Pakhawadee Palungwachira
- Department of Emergency Medicine, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | | |
Collapse
|
16
|
Identification of a novel anticancer mechanism of Paeoniae Radix extracts based on systematic transcriptome analysis. Biomed Pharmacother 2022; 148:112748. [PMID: 35219117 DOI: 10.1016/j.biopha.2022.112748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/21/2022] Open
Abstract
Paeoniae Radix (PR) has a great therapeutic value in many clinical applications; however, the presence of various bioactive compounds and its complicated effects on human health makes its precise mechanisms of action unclear. This study investigated the effects of PR at the molecular pathway level by profiling genome-wide gene expression changes following dose-dependent treatment of human lung cancer cells (A549) with PR water extract (WPR), PR ethanol extracts (EPR), as well as their individual components. We found that PR exerts anticancer effects in A549 cells by regulating numerous pathways. Specifically, EPR and two compounds, namely, hederagenin (HG) and oleanolic acid (OA), significantly downregulate the Aurora B pathway. Furthermore, we generated an integrated PR extracts-compounds-target genes network in the Aurora B pathway to understand their interactions. Our findings reinforce that inhibiting Aurora kinase activity is a therapeutic target for treating cancers, providing the potential for novel mechanisms of action for PR and its components against lung cancer.
Collapse
|
17
|
Amet R, Previtali V, Mihigo HB, Sheridan E, Brophy S, Hante NK, Santos-Martinez MJ, Hayden PJ, Browne PV, Rozas I, McElligott AM, Zisterer DM. A novel aryl-guanidinium derivative, VP79s, targets the signal transducer and activator of transcription 3 signaling pathway, downregulates myeloid cell leukaemia-1 and exhibits preclinical activity against multiple myeloma. Life Sci 2021; 290:120236. [PMID: 34953891 DOI: 10.1016/j.lfs.2021.120236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
AIMS We have recently described a novel guanidinium-based compound, VP79s, which induces cytotoxicity in various cancer cell lines. Here, we aim to investigate the activity of VP79s and associated mechanisms of action in multiple myeloma (MM) cells in vitro and ex vivo. MAIN METHODS The effects of VP79s on cell viability and induction of apoptosis was examined in a panel of drug-sensitive and drug-resistant MM cell lines, as well as ex vivo patient samples and normal donor lymphocytes and platelets. Cell signaling pathways associated with the biological effects of VP79s were analysed by immunoblotting and flow cytometry. Gene expression changes were assessed by quantitative real-time PCR analysis. KEY FINDINGS VP79s was found to rapidly inhibit both constitutively active and IL-6-induced STAT3 signaling with concurrent downregulation of the IL-6 receptors, CD130 and CD126. VP79s induced a rapid and dose-dependent downregulation of anti-apoptotic Bcl-2 family member, myeloid cell leukaemia-1 (MCL-1). VP79s enhanced bortezomib induced cell death and was also found to overcome bone marrow stromal cell induced drug resistance. VP79s exhibited activity in ex vivo patient samples at concentrations which had no effect on peripheral blood mononuclear cells, lymphocytes and platelets isolated from healthy donors. SIGNIFICANCE As VP79s resulted in rapid inhibition of the key IL-6/STAT3 signaling pathway and downregulation of MCL-1 expression with subsequent selective anti-myeloma activity, VP79s may be a potential therapeutic agent with a novel mechanism of action in MM cells.
Collapse
Affiliation(s)
- Rebecca Amet
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland; John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Viola Previtali
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Helene B Mihigo
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Emily Sheridan
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | - Sarah Brophy
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland
| | - Nadhim Kamil Hante
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland
| | - Maria Jose Santos-Martinez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland; School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Patrick J Hayden
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Paul V Browne
- Department of Haematology, St. James's Hospital, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland
| | - Isabel Rozas
- School of Chemistry, Trinity College Dublin, Dublin 2, Ireland
| | - Anthony M McElligott
- John Durkan Leukaemia Laboratories, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin 8, Ireland; Trinity St. James's Cancer Institute, Trinity College and St James's Hospital, Dublin 8, Ireland.
| | - Daniela M Zisterer
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
18
|
Woo JS, Yoo ES, Kim SH, Lee JH, Han SH, Jung SH, Jung GH, Jung JY. Anticancer effects of oleanolic acid on human melanoma cells. Chem Biol Interact 2021; 347:109619. [PMID: 34364837 DOI: 10.1016/j.cbi.2021.109619] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/27/2021] [Accepted: 08/04/2021] [Indexed: 11/23/2022]
Abstract
Owing to the ineffectiveness of the currently used therapies against melanoma, there has been a shift in focus toward alternative therapies involving the use of natural compounds. This study assessed the anticancer effects of oleanolic acid (OA) and its ability to induce apoptosis in A375SM and A375P melanoma cells in vivo. Compared to the control group, viability of A375P and A375SM cells decreased following OA treatment. In OA-treated A375SM and A375P cells, 4',6-diamidino-2-phenylindole staining showed an increase in the apoptotic body, and flow cytometry revealed increased number of apoptotic cells compared to that in the control group. OA-treated A375SM cells exhibited an increased expression of the apoptotic proteins, cleaved poly (ADP-ribose) polymerase (PARP) and B-cell lymphoma (Bcl)-2-associated X protein (Bax) as well as decreased expression of the antiapoptotic protein Bcl-2 compared to that in the control group. In OA-treated A375P cells, expression patterns of cleaved PARP and Bcl-2 were similar to those in OA-treated A375SM cells; however, no difference was reported in the expression of Bax compared to that in the control group. Additionally, OA-treated melanoma cells showed decreased expression of phospho-nuclear factor-κB (p-NF-κB), phospho-inhibitor of nuclear factor-κBα (p-IκBα), and phospho-IκB kinase αβ than that in the control group. Moreover, immunohistochemistry showed a comparatively decreased level of p-NF-κB in the OA-treated group than that in the control group. Xenograft analysis confirmed the in vivo anticancer effects of OA against A375SM cells. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay revealed an increased number of TUNEL-positive cells in the OA-treated group compared to that in the control group. In conclusion, the study results suggest that OA induces apoptosis of A375SM and A375P cells in vitro and apoptosis of A375SM cells in vivo. Furthermore, the in vitro and in vivo anticancer effects were mediated by the NF-κB pathway.
Collapse
Affiliation(s)
- Joong-Seok Woo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Eun-Seon Yoo
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Sung-Hyun Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Jae-Han Lee
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - So-Hee Han
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Soo-Hyun Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Gi-Hwan Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea
| | - Ji-Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungcheongnam, 32439, Republic of Korea.
| |
Collapse
|
19
|
Sampaio LA, Pina LTS, Serafini MR, Tavares DDS, Guimarães AG. Antitumor Effects of Carvacrol and Thymol: A Systematic Review. Front Pharmacol 2021; 12:702487. [PMID: 34305611 PMCID: PMC8293693 DOI: 10.3389/fphar.2021.702487] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/22/2021] [Indexed: 12/12/2022] Open
Abstract
Background: It is estimated that one in five people worldwide faces a diagnosis of a malignant neoplasm during their lifetime. Carvacrol and its isomer, thymol, are natural compounds that act against several diseases, including cancer. Thus, this systematic review aimed to examine and synthesize the knowledge on the antitumor effects of carvacrol and thymol. Methods: A systematic literature search was carried out in the PubMed, Web of Science, Scopus and Lilacs databases in April 2020 (updated in March 2021) based on the PRISMA 2020 guidelines. The following combination of health descriptors, MeSH terms and their synonyms were used: carvacrol, thymol, antitumor, antineoplastic, anticancer, cytotoxicity, apoptosis, cell proliferation, in vitro and in vivo. To assess the risk of bias in in vivo studies, the SYRCLE Risk of Bias tool was used, and for in vitro studies, a modified version was used. Results: A total of 1,170 records were identified, with 77 meeting the established criteria. The studies were published between 2003 and 2021, with 69 being in vitro and 10 in vivo. Forty-three used carvacrol, 19 thymol, and 15 studies tested both monoterpenes. It was attested that carvacrol and thymol induced apoptosis, cytotoxicity, cell cycle arrest, antimetastatic activity, and also displayed different antiproliferative effects and inhibition of signaling pathways (MAPKs and PI3K/AKT/mTOR). Conclusions: Carvacrol and thymol exhibited antitumor and antiproliferative activity through several signaling pathways. In vitro, carvacrol appears to be more potent than thymol. However, further in vivo studies with robust methodology are required to define a standard and safe dose, determine their toxic or side effects, and clarify its exact mechanisms of action. This systematic review was registered in the PROSPERO database (CRD42020176736) and the protocol is available at https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=176736.
Collapse
Affiliation(s)
- Laeza Alves Sampaio
- Graduate Program of Applied Sciences to Health, Federal University of Sergipe, Lagarto, Brazil
| | | | | | | | | |
Collapse
|
20
|
Hosseinzadeh E, Hassanzadeh A, Marofi F, Alivand MR, Solali S. Flavonoid-Based Cancer Therapy: An Updated Review. Anticancer Agents Med Chem 2021; 20:1398-1414. [PMID: 32324520 DOI: 10.2174/1871520620666200423071759] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 10/27/2019] [Accepted: 11/06/2019] [Indexed: 12/24/2022]
Abstract
As cancers are one of the most important causes of human morbidity and mortality worldwide, researchers try to discover novel compounds and therapeutic approaches to decrease survival of cancer cells, angiogenesis, proliferation and metastasis. In the last decade, use of special phytochemical compounds and flavonoids was reported to be an interesting and hopeful tactic in the field of cancer therapy. Flavonoids are natural polyphenols found in plant, fruits, vegetables, teas and medicinal herbs. Based on reports, over 10,000 flavonoids have been detected and categorized into several subclasses, including flavonols, anthocyanins, flavanones, flavones, isoflavones and chalcones. It seems that the anticancer effect of flavonoids is mainly due to their antioxidant and anti inflammatory activities and their potential to modulate molecular targets and signaling pathways involved in cell survival, proliferation, differentiation, migration, angiogenesis and hormone activities. The main aim of this review is to evaluate the relationship between flavonoids consumption and cancer risk, and discuss the anti-cancer effects of these natural compounds in human cancer cells. Hence, we tried to collect and revise important recent in vivo and in vitro researches about the most effective flavonoids and their main mechanisms of action in various types of cancer cells.
Collapse
Affiliation(s)
- Elham Hosseinzadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
21
|
Zhao X, Li X, Wang S, Yang Z, Liu H, Xu S. Cadmium exposure induces mitochondrial pathway apoptosis in swine myocardium through xenobiotic receptors-mediated CYP450s activation. J Inorg Biochem 2021; 217:111361. [PMID: 33581611 DOI: 10.1016/j.jinorgbio.2021.111361] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 01/12/2021] [Accepted: 01/16/2021] [Indexed: 12/20/2022]
Abstract
Cadmium (Cd) pollution has become an important public and environmental health issue. Xenobiotic receptors (XRs, aryl hydrocarbon receptor, AHR; constitutive androstane receptor, CAR; pregnane X receptor, PXR) modulate downstream cytochrome P450 enzymes (CYP450s) expression to metabolize xenobiotics and environmental contaminants. However, the underlying mechanisms of cardiotoxicity induced by Cd(II) in swine and the roles of XRs and CYP450s remain poorly understood. In this study, the cardiotoxicity of Cd(II) was investigated by establishing a Cd(II)-exposed swine model (CdCl2, 20 mg Cd/Kg diet). Terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) assay and transmission electron microscope were used to observe the apoptosis. Antioxidant capacity was evaluated by free radicals contents and antioxidant enzymes activities. RT-PCR and western blot were used to measure the expression of XRs, CYP450s and apoptosis-related genes. Our results revealed that Cd(II) exposure activated the XRs and increased the CYP450s expression, contributing to the production of reactive oxygen species (ROS). Cd(II) exposure restrained the antioxidant capacity, causing oxidative stress. Moreover, mitogen-activated protein kinase (MAPK) pathway including c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK) and P38 mitogen-activated protein kinase (P38) was activated, triggering the mitochondrial apoptotic pathway. In brief, we concluded that Cd(II) caused mitochondrial pathway apoptosis in swine myocardium via the oxidative stress-MAPK pathway, and XRs-mediated CYP450s expression might participate in this process through promoting the ROS.
Collapse
Affiliation(s)
- Xia Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaojing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Shengchen Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zijiang Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Honggui Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
22
|
Chang WT, Bow YD, Fu PJ, Li CY, Wu CY, Chang YH, Teng YN, Li RN, Lu MC, Liu YC, Chiu CC. A Marine Terpenoid, Heteronemin, Induces Both the Apoptosis and Ferroptosis of Hepatocellular Carcinoma Cells and Involves the ROS and MAPK Pathways. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7689045. [PMID: 33488943 PMCID: PMC7803406 DOI: 10.1155/2021/7689045] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/27/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of death, resulting in over 700 thousand deaths annually worldwide. Chemotherapy is the primary therapeutic strategy for patients with late-stage HCC. Heteronemin is a marine natural product isolated from Hippospongia sp. that has been found to protect against carcinogenesis in cholangiocarcinoma, prostate cancer, and acute myeloid leukemia. In this study, heteronemin was found to inhibit the proliferation of the HCC cell lines HA22T and HA59T and induce apoptosis via the caspase pathway. Heteronemin treatment also induced the formation of reactive oxygen species (ROS), which are associated with heteronemin-induced cell death, and to trigger ROS removal by mitochondrial SOD2 rather than cytosolic SOD1. The mitogen-activated protein kinase (MAPK) signaling pathway was associated with ROS-induced cell death, and heteronemin downregulated the expression of ERK, a MAPK that is associated with cell proliferation. Inhibitors of JNK and p38, which are MAPKs associated with apoptosis, restored heteronemin-induced cell death. In addition, heteronemin treatment reduced the expression of GPX4, a protein that inhibits ferroptosis, which is a novel form of nonapoptotic programmed cell death. Ferroptosis inhibitor treatment also restored heteronemin-induced cell death. Thus, with appropriate structural modification, heteronemin can act as a potent therapeutic against HCC.
Collapse
Affiliation(s)
- Wen-Tsan Chang
- Division of General and Digestive Surgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Digestive Disease Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Ding Bow
- Ph.D. Program in Life Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Pei-Jung Fu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chang-Yi Wu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Hua Chang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yen-Ni Teng
- Department of Biological Sciences and Technology, National University of Tainan, Tainan 700, Taiwan
| | - Ruei-Nian Li
- Department of Biomedical Science and Environment Biology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Mei-Chin Lu
- Graduate Institute of Marine Biotechnology, National Dong Hwa University, Pingtung 944, Taiwan
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chien-Chih Chiu
- Center for Cancer Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
- The Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
23
|
Hosny S, Sahyon H, Youssef M, Negm A. Oleanolic Acid Suppressed DMBA-Induced Liver Carcinogenesis through Induction of Mitochondrial-Mediated Apoptosis and Autophagy. Nutr Cancer 2020; 73:968-982. [PMID: 32519911 DOI: 10.1080/01635581.2020.1776887] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Phytochemicals appeared as a rich source of efficient and safe agents against many diseases like cancer. Various herbal sources are rich in oleanolic acid (OA). The scope of this study was to assess the biochemical and molecular mechanisms implicated in the ameliorative potency of OA against DMBA-induced liver carcinogenesis. Forty-eight male albino mice were assigned randomly to five groups (eight mice each) as follows: control healthy group, olive oil group, OA group, DMBA group, and DMBA with OA. Apoptosis, autophagy, inflammation, proliferation, and angiogenesis were investigated in the tissue samples. Histopathological examination was carried out as well as liver enzymes activity and other hepatic antioxidant and inflammatory biomarkers. The treatment with OA effectively suppressed the DMBA-initiated liver carcinogenesis via modulation of antioxidant status, induction of apoptosis and autophagy through modulating the expression of Caspase-3, Bcl-2 and Beclin-1, inhibiting angiogenesis (VEGF), proliferation (PCNA), and improved liver function and histological picture with a reduction in AFP level. Additionally, OA applies its antitumor effects by inhibition of proinflammatory transcription factor NF-κB and inflammatory markers (TNF-α and Cox-2) associated with DMBA administration. The present study shows that OA treatment efficiently suppressed the DMBA-initiated liver carcinogenesis through induction of mitochondrial-mediated apoptosis and autophagy and modulating inflammation.
Collapse
Affiliation(s)
- Samar Hosny
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Heba Sahyon
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Magdy Youssef
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Amr Negm
- Chemistry Department, Biochemistry Division, Faculty of Science, Mansoura University, Mansoura, Egypt.,Department of Chemistry, College of Science, King Faisal University, Al-Ahasa, Saudi Arabia
| |
Collapse
|
24
|
Rahman SMT, Zhou W, Deiters A, Haugh JM. Optical control of MAP kinase kinase 6 (MKK6) reveals that it has divergent roles in pro-apoptotic and anti-proliferative signaling. J Biol Chem 2020; 295:8494-8504. [PMID: 32371393 DOI: 10.1074/jbc.ra119.012079] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/21/2020] [Indexed: 12/24/2022] Open
Abstract
The selective pressure imposed by extrinsic death signals and stressors adds to the challenge of isolating and interpreting the roles of proteins in stress-activated signaling networks. By expressing a kinase with activating mutations and a caged lysine blocking the active site, we can rapidly switch on catalytic activity with light and monitor the ensuing dynamics. Applying this approach to MAP kinase 6 (MKK6), which activates the p38 subfamily of MAPKs, we found that decaging active MKK6 in fibroblasts is sufficient to trigger apoptosis in a p38-dependent manner. Both in fibroblasts and in a murine melanoma cell line expressing mutant B-Raf, MKK6 activation rapidly and potently inhibited the pro-proliferative extracellular signal-regulated kinase (ERK) pathway; to our surprise, this negative cross-regulation was equally robust when all p38 isoforms were inhibited. These results position MKK6 as a new pleiotropic signal transducer that promotes both pro-apoptotic and anti-proliferative signaling, and they highlight the utility of caged, light-activated kinases for dissecting stress-activated signaling networks.
Collapse
Affiliation(s)
- Shah Md Toufiqur Rahman
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Wenyuan Zhou
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexander Deiters
- Department of Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason M Haugh
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
25
|
Ryu M, Sung CK, Im YJ, Chun C. Activation of JNK and p38 in MCF-7 Cells and the In Vitro Anticancer Activity of Alnus hirsuta Extract. Molecules 2020; 25:E1073. [PMID: 32121012 PMCID: PMC7179116 DOI: 10.3390/molecules25051073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 01/02/2023] Open
Abstract
JNK and p38 are important mitogen-activated protein kinases (MAPKs) that respond to stress stimuli. The stress-activated MAPKs associated with apoptotic cell death play vital roles in mammalian cells. Alnus hirsuta, which contains abundant diarylheptanoids derivatives, is a valuable medicinal plant. The CHCl3 extract (AHC) containing platyphyllenone (1) and platyphyllone (3) as main compounds showed in vitro anticancer effects. We report the biological activities of A. hirsuta extract associated with the regulation of apoptosis and JNK and p38 in MCF-7 breast cancer cells. Levels of phospho-JNK and phospho-p38 by AHC treatment were evaluated by enzyme-linked immunosorbent assay (ELISA). ROS production, apoptotic effect, and DNA contents of the cells were measured by flow cytometry. The two diarylheptanoids 1 and 3 and the AHC extract exhibited cytotoxic effects on MCF-7 cells in MTT assay, with IC50 values of 18.1, 46.9, 260.0 μg/mL, respectively. AHC induced ROS generation and elevated the endogenous levels of phospho-JNK and phospho-p38. AHC resulted in apoptosis and cell cycle arrest. We suggest that the antitumor effect of A. hirsuta extract is achieved by apoptosis promotion and cell cycle arrest mediated by the activation of JNK and p38 signaling pathway via ROS generation.
Collapse
Affiliation(s)
| | | | - Young Jun Im
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (M.R.); (C.K.S.)
| | - ChangJu Chun
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea; (M.R.); (C.K.S.)
| |
Collapse
|
26
|
Wang K, Gong Q, Zhan Y, Chen B, Yin T, Lu Y, Zhang Y, Wang H, Ke J, Du B, Liu X, Xiao J. Blockage of Autophagic Flux and Induction of Mitochondria Fragmentation by Paroxetine Hydrochloride in Lung Cancer Cells Promotes Apoptosis via the ROS-MAPK Pathway. Front Cell Dev Biol 2020; 7:397. [PMID: 32039209 PMCID: PMC6987457 DOI: 10.3389/fcell.2019.00397] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/31/2019] [Indexed: 12/30/2022] Open
Abstract
Cancer cells are characterized by malignant proliferation and aberrant metabolism and are thereby liable to the depletion of nutrients and accumulation of metabolic waste. To maintain cellular homeostasis, cancer cells are prone to upregulating the canonical autophagy pathway. Here, we identified paroxetine hydrochloride (Paxil) as a late autophagy inhibitor and investigated its killing effect on lung cancer cells and with a xenograft mouse model in vivo. Upregulated LC3-II and p62 expression indicated that Paxil inhibited autophagy. Acid-sensitive dyes (e.g., LysoTracker and AO staining) indicated reduced lysosomal acidity following Paxil treatment; consequently, the maturation of the pH-dependent hydroxylases (e.g., cathepsin B and D) substantially declined. Paxil also induced the fragmentation of mitochondria and further intensified ROS overproduction. Since the autophagy pathway was blocked, ROS rapidly accumulated, which activated JNK and p38 kinase. Such activity promoted the localization of Bax, which led to increased mitochondrial outer membrane permeability. The release of Cytochrome c with the loss of the membrane potential triggered a caspase cascade, ultimately leading to apoptosis. In contrast, the clearance of ROS by its scavenger, NAC, rescued Paxil-induced apoptosis accompanied by reduced p38 and JNK activation. Thus, Paxil blocked the autophagic flux and induced the mitochondria-dependent apoptosis via the ROS-MAPK pathway.
Collapse
Affiliation(s)
- Kun Wang
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qing Gong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yujuan Zhan
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bonan Chen
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ting Yin
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuhua Lu
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yilin Zhang
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huiqi Wang
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junzi Ke
- Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Biaoyan Du
- Department of Pathology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jianyong Xiao
- Research Center of Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Biochemistry, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
27
|
Jeong MS, Jung JH, Lee H, Kim CG, Kim SH. Methyloleanolate Induces Apoptotic And Autophagic Cell Death Via Reactive Oxygen Species Generation And c-Jun N-terminal Kinase Phosphorylation. Onco Targets Ther 2019; 12:8621-8635. [PMID: 31695422 PMCID: PMC6815788 DOI: 10.2147/ott.s211904] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/01/2019] [Indexed: 01/16/2023] Open
Abstract
Background To develop a potent anticancer agent similar to oleanolate, the underlying mechanisms of its derivative, methyloleanolate, in the apoptosis and autophagy of A549 and H1299 cells were elucidated. Purpose The aim of the present study was to investigate the effect of methyloleanolate in inducing apoptotic and autophagic cell death in cancer cells. Materials and methods Flow cytometric analysis with Annexin V/PI staining, Western blot analysis, and immunofluorescence analysis were conducted in A549 and H1299 cells. Results Methyloleanolate increased the fraction of Annexin V/PI apoptotic cells and activated caspase-8, caspase-3, and death receptor 5 (DR5) more than oleanolate in A549 and H1299 cells pretreated with pancaspase inhibitor z-VAD-fmk and DR5 depletion. Also, methyloleanolate induced autophagic features of microtubule-associated protein light chain 3 3BII (LC3BII) conversion and puncta in A549 and H1299 cells, along with autophagosomes and vacuoles. Methyloleanolate blocked autophagy flux for impaired autophagy and chloroquine (CQ)-enhanced microtubule-associated protein LC3BII accumulation and cytotoxicity in A549 and H1299 cells, although 3-methyladenine (3-MA) did not. Interestingly, LC3BII accumulation was detected only in methyloleanolate-treated autophagy-related gene 5 (ATG5)+/+ mouse embryonic fibroblast (MEF) cells but not in ATG5 -/- MEF cells. Methyloleanolate reduced p-mTOR but activated p-c-Jun N-terminal kinases and reactive oxygen species production in A549 and H1299 cells. Conversely, n-acetyl-l-cysteine and SP600125 blocked apoptotic and autophagic cascades caused by methyloleanolate in A549 and H1299 cells. Conclusion Overall, the findings suggest that methyloleanolate induces apoptotic and autophagic cell death in non-small cell lung cancers via reactive oxygen species generation and c-Jun N-terminal kinase phosphorylation.
Collapse
Affiliation(s)
- Myoung Seok Jeong
- College of Korean Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| | - Ji Hoon Jung
- College of Korean Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| | - Hyemin Lee
- College of Korean Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| | - Chang Geun Kim
- College of Korean Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Dongdaemun-Gu, Seoul 02447, Republic of Korea
| |
Collapse
|
28
|
Abstract
Chemokines are small secreted proteins with chemoattractant properties that play a key role in inflammation. One such chemokine, Stromal cell-derived factor-1 (SDF-1) also known as CXCL12, and its receptor, CXCR4, are expressed and functional in cardiac myocytes. SDF-1 both stimulates and enhances the cellular signal which attracts potentially beneficial stem cells for tissue repair within the ischemic heart. Paradoxically however, this chemokine is known to act in concert with the inflammatory cytokines of the innate immune response which contributes to cellular injury through the recruitment of inflammatory cells during ischemia. In the present study, we have demonstrated that SDF-1 has dose dependent effects on freshly isolated cardiomyocytes. Using Tunnel and caspase 3-activation assays, we have demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations (pathological concentrations) induced apoptosis. Furthermore, ELISA data demonstrated that the treatment of isolated adult rat cardiac myocyte with SDF-1 at higher concentrations upregulated TNF-α protein expression which directly correlated with subsequent apoptosis. There was a significant reduction in SDF-1 mediated apoptosis when TNF-α expression was neutralized which suggests that SDF-1 mediated apoptosis is TNF-α-dependent. The fact that certain stimuli are capable of driving cardiomyocytes into apoptosis indicates that these cells are susceptible to clinically relevant apoptotic triggers. Our findings suggest that the elevated SDF-1 levels seen in a variety of clinical conditions, including ischemic myocardial infarction, may either directly or indirectly contribute to cardiac cell death via a TNF-α mediated pathway. This highlights the importance of this receptor/ligand in regulating the cardiomyocyte response to stress conditions.
Collapse
|
29
|
Fakai MI, Abd Malek SN, Karsani SA. Induction of apoptosis by chalepin through phosphatidylserine externalisations and DNA fragmentation in breast cancer cells (MCF7). Life Sci 2019; 220:186-193. [PMID: 30682342 DOI: 10.1016/j.lfs.2019.01.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/18/2019] [Accepted: 01/20/2019] [Indexed: 12/30/2022]
Abstract
AIMS Chalepin, a naturally occurring compound isolated from Ruta angustifolia have been shown to exert a promising anticancer activity through various mechanisms. Hence, the need to investigate the apoptotic inducing ability of chalepin in MCF7 cells by various detection assays. MATERIALS AND METHODS Cytotoxicity screening of chalepin against MCF7 cells was conducted using SRB assay. Apoptosis induction was examined by established morphological and biochemical assays including phase contrast and Hoechst/PI staining fluorescence microscope. Similarly, Annexin-V/FITC and TUNEL assays were conducted using flow cytometry whereas caspase-3 activity was evaluated using microplate reader. KEY FINDINGS The result indicates remarkable cytotoxic activity against MCF7 cells, whereas it shows moderate cytotoxic activity against MDA-MB231 cells. Interestingly, chalepin did not present any toxicity against MRC5 normal cell line. Morphological examination using both phase contrast and fluorescence microscope displays typical apoptotic features such as membrane blebbing, DNA fragmentation, chromatin condensation and apoptotic bodies' formation following chalepin treatment against MCF7 cells at different concentration for 48 h. Apoptosis induction is significantly associated with externalisation of phosphatidylserine, and DNA fragmentation in MCF7 cells chalepin treated cells when compared with control. The protein expressions of caspase-8, 9 and cleaved PARP1 were upregulated which correlated well with increased caspase-3 activity. SIGNIFICANCE From our recent findings, chalepin was able to induced apoptosis in MCF7 cells and therefore, could be evaluated further as a potential source of anticancer agent for cancer treatment such as breast cancer.
Collapse
Affiliation(s)
- Musa Isah Fakai
- Division of Biochemistry, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Department of Biochemistry, Kebbi State University of Science and Technology, P.M.B. 1144, Aliero, Kebbi State, Nigeria
| | - Sri Nurestri Abd Malek
- Division of Biochemistry, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Saiful Anuar Karsani
- Division of Biochemistry, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
30
|
Lim W, Ham J, Bazer FW, Song G. Carvacrol induces mitochondria-mediated apoptosis via disruption of calcium homeostasis in human choriocarcinoma cells. J Cell Physiol 2018; 234:1803-1815. [PMID: 30070691 DOI: 10.1002/jcp.27054] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Abstract
Carvacrol is a monoterpenoid phenol present in the oils of various plants including Origanum vulgare (oregano) or Origanum majorana (marjoram). For a long time, it has been used as spice in foods because of its antimicrobial properties. Additionally, it appears to have anticancer effects against some cancer but this has not been well studied. Therefore, we conducted various assays to confirm the effects of carvacrol on choriocarcinoma cell lines (JAR and JEG3). Our results indicate that carvacrol has antiproliferative properties and induces apoptosis, resulting in increased expression of proapoptotic proteins. Additionally, carvacrol disrupted the mitochondrial membrane potential and induced calcium ion overload in the mitochondrial matrix in both JAR and JEG3 cells. Furthermore, carvacrol generated oxidative stress and lipid peroxidation in both JAR and JEG3 cells. Moreover, carvacrol-suppressed phosphoinositide 3-kinase-protein kinase B and extracellular signal-regulated kinase 1/2 mitogen-activated protein kinase (MAPK) signal transduction whereas expression of phosphor-P38 and c-Jun N-terminal kinase MAPK was increased. Together, our results indicate that carvacrol may be a possible new therapeutic agent or supplement for the control of human choriocarcinomas.
Collapse
Affiliation(s)
- Whasun Lim
- Department of Biomedical Sciences, Catholic Kwandong University, Gangneung, Republic of Korea
| | - Jiyeon Ham
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Fuller W Bazer
- Department of Animal Science, Center for Animal Biotechnology and Genomics, Texas A&M University, College Station, Texas
| | - Gwonhwa Song
- Department of Biotechnology, Institute of Animal Molecular Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Wang L, Li Q, Ye Z, Qiao B. ZBTB7/miR-137 Autoregulatory Circuit Promotes the Progression of Renal Carcinoma. Oncol Res 2018; 27:1007-1014. [PMID: 29673422 PMCID: PMC7848413 DOI: 10.3727/096504018x15231148037228] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Renal carcinoma greatly threatens human health, but the involved molecular mechanisms are far from complete understanding. As a master oncogene driving the initiation of many other cancers, ZBTB7 has not been established to be associated with renal cancer. Our data revealed that ZBTB7 is highly expressed in renal carcinoma specimens and cell lines, compared with normal cells. The silencing of ZBTB7 suppressed the proliferation and invasion of renal cancer cells. ZBTB7 overexpression rendered normal cells with higher proliferation rates and invasiveness. An animal study further confirmed the role of ZBTB7 in the growth of renal carcinoma. Moreover, miR-137 was identified to negatively regulate the expression of ZBTB7, and its abundance is inversely correlated with that of ZBTB7 in renal carcinoma specimens and cell lines. ZBTB7 overexpression may be induced by miR-137 downregulation. Interestingly, ZBTB7 can also suppress miR-137 expression by binding to its recognition site within the miR-137 promoter region. Taken together, we identified an autoregulatory loop consisting of ZBTB7 and miR-137 in gastric cancers, and targeting this pathway may be an effective strategy for renal carcinoma cancer therapy.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Qi Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Zhuo Ye
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| | - Baoping Qiao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P.R. China
| |
Collapse
|
32
|
Zhao HG, Zhou SL, Lin YY, Dai HF, Huang FY. Toxicarioside N induces apoptosis in human gastric cancer SGC-7901 cell by activating the p38MAPK pathway. Arch Pharm Res 2018; 41:71-78. [PMID: 28940036 DOI: 10.1007/s12272-017-0956-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/11/2017] [Indexed: 12/15/2022]
Abstract
Natural plant compounds with potent proliferation inhibition and apoptosis induction properties have been screened as novel anticancer drugs. Toxicarioside N (Tox N) was isolated from the seeds of the tropical plant Antiaris toxicaria in Hainan province, China. To our knowledge, the effects that Tox N has on the apoptosis of SGC-7901 cells and its potential mechanism have never been investigated. In this study, we detected the anticancer activities of Tox N and explored the potential mechanism in the human gastrointestinal cancer cell line SGC-7901. Here, we found that Tox N inhibited SGC-7901 cell growth in a dose- and time-dependent manner and induced apoptosis in cells based on cell morphology and flow cytometry analyses. Additionally, the SGC-7901 cell treated with Tox N up-regulated the expression level of cleaved caspase-3/9 and PARP, increased the Bax/Bcl-2 ratio, and led to the release of cytochrome c into the cytoplasm. In addition, Tox N treatment led to the phosphorylation of p38MAPK. SB203580, a p38MAPK inhibitor, partially attenuated Tox N induced apoptosis by preventing the activation of caspase-3/9 and PARP. Our results indicated for the first time that Tox N can induce SGC-7901 cells apoptosis by activating the p38MAPK pathway.
Collapse
Affiliation(s)
- Huan-Ge Zhao
- Key Laboratory of Translation Medicine for Tropical Diseases, Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Song-Lin Zhou
- Key Laboratory of Translation Medicine for Tropical Diseases, Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Ying-Ying Lin
- Key Laboratory of Translation Medicine for Tropical Diseases, Ministry of Education, Hainan Medical College, Haikou, 571199, China
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China
| | - Hao-Fu Dai
- Institutes of Tropical Bioscience and Biotechnology, Chinese Academy of Tropical Agricultural Sciences, Haikou, 571199, China.
| | - Feng-Ying Huang
- Key Laboratory of Translation Medicine for Tropical Diseases, Ministry of Education, Hainan Medical College, Haikou, 571199, China.
- Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, 571199, China.
| |
Collapse
|
33
|
Yamagata K, Izawa Y, Onodera D, Tagami M. Chlorogenic acid regulates apoptosis and stem cell marker-related gene expression in A549 human lung cancer cells. Mol Cell Biochem 2017; 441:9-19. [DOI: 10.1007/s11010-017-3171-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 09/01/2017] [Indexed: 01/04/2023]
|
34
|
Tsepaeva OV, Nemtarev AV, Abdullin TI, Grigor'eva LR, Kuznetsova EV, Akhmadishina RA, Ziganshina LE, Cong HH, Mironov VF. Design, Synthesis, and Cancer Cell Growth Inhibitory Activity of Triphenylphosphonium Derivatives of the Triterpenoid Betulin. JOURNAL OF NATURAL PRODUCTS 2017; 80:2232-2239. [PMID: 28782948 DOI: 10.1021/acs.jnatprod.7b00105] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
A series of new triphenylphosphonium (TPP) derivatives of the triterpenoid betulin (1, 3-lup-20(29)-ene-3β,28-diol) have been synthesized and evaluated for cytotoxic effects against human breast cancer (MCF-7), prostate adenocarcinoma (PC-3), vinblastine-resistant human breast cancer (MCF-7/Vinb), and human skin fibroblast (HSF) cells. The TPP moiety was applied as a carrier group through the acyl linker at the 28- or 3- and 28-positions of betulin to promote cellular and mitochondrial accumulation of the resultant compounds. A structure-activity relationship study has revealed the essential role of the TPP group in the biological properties of the betulin derivatives produced. The present results showed that a conjugate of betulin with TPP (3) enhanced antiproliferative activity toward vinblastine-resistant MCF-7 cells, with an IC50 value as low as 0.045 μM.
Collapse
Affiliation(s)
- Olga V Tsepaeva
- A. E. Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences , Arbuzov Street 8, 420088, Kazan, Russian Federation
| | - Andrey V Nemtarev
- A. E. Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences , Arbuzov Street 8, 420088, Kazan, Russian Federation
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Timur I Abdullin
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Leysan R Grigor'eva
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Elena V Kuznetsova
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Rezeda A Akhmadishina
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Liliya E Ziganshina
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Hanh H Cong
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| | - Vladimir F Mironov
- A. E. Arbuzov Institute of Organic and Physical Chemistry, Kazan Scientific Center, Russian Academy of Sciences , Arbuzov Street 8, 420088, Kazan, Russian Federation
- Kazan (Volga Region) Federal University , Kremlevskaya Street 18, 420008, Kazan, Russian Federation
| |
Collapse
|
35
|
Mitsuhashi Y, Furusawa Y, Aradate T, Zhao QL, Moniruzzaman R, Kanamori M, Noguchi K, Kondo T. 3-O-trans-p-coumaroyl-alphitolic acid, a triterpenoid from Zizyphus jujuba, leads to apoptotic cell death in human leukemia cells through reactive oxygen species production and activation of the unfolded protein response. PLoS One 2017; 12:e0183712. [PMID: 28832644 PMCID: PMC5568338 DOI: 10.1371/journal.pone.0183712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/09/2017] [Indexed: 11/23/2022] Open
Abstract
3-O-trans-p-coumaroyl-alphitolic acid (3OTPCA), a triterpenoid isolated from the plant Zizyphus jujuba (ZJ), is known to be cytotoxic to cancer cells; however, the molecular mechanism underlying 3OTPCA-induced cell death remains unknown. Here, we provide novel evidence that 3OTPCA induces apoptotic cell death in human leukemia cells. We found that 3OPTCA induces DNA fragmentation within 24 h after treatment in U937 cells, which was also observed in other leukemia cell lines, including Molt-4 and Jurkat cells. We then investigated other parameters involved in apoptosis, including phosphatidylserine externalization and caspase-3 cleavage in U937 cells treated with 3OTPCA. 3OTPCA caused significant DNA fragmentation, annexin-V binding, and caspase-3 cleavage, indicating that 3OTPCA exerts cytotoxicity through apoptosis induction. RNA-seq analysis revealed that the expression of transcripts associated with the unfolded protein response (UPR), such as spliced XBP-1 and CHOP, were up-regulated by 3OTPCA treatment. 3OTPCA-induced UPR activation may be due to endoplasmic reticulum (ER) stress because both 3OTPCA and thapsigargin, an endoplasmic Ca2+ transport ATPase inhibitor, increased intracellular calcium levels. 3OTPCA down-regulated the expression of Bcl-2, a target of CHOP, and led to the loss of the mitochondrial membrane, indicating that the intrinsic (mitochondrial) apoptotic pathway was triggered by 3OTPCA, likely through UPR activation. Furthermore, we found that 3OTPCA induced superoxide anion generation and, following p38 MAPK phosphorylation, caspase-8 cleavage without affecting Fas expression. It also induced subsequent Bid cleavage, which may enhance the apoptosis triggered by the intrinsic pathway. These findings reveal for the first time that 3OTPCA induces apoptotic cell death through the generation of reactive oxygen species and activation of UPR.
Collapse
Affiliation(s)
- Yohei Mitsuhashi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Yukihiro Furusawa
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa, Toyama, Japan
- * E-mail:
| | - Tadashi Aradate
- Department of Medical Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Rohan Moniruzzaman
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Masahiko Kanamori
- Department of Human Science 1, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Kyo Noguchi
- Department of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| |
Collapse
|
36
|
Twardziok M, Meierhofer D, Börno S, Timmermann B, Jäger S, Boral S, Eggert A, Delebinski CI, Seifert G. Transcriptomic and proteomic insight into the effects of a defined European mistletoe extract in Ewing sarcoma cells reveals cellular stress responses. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:237. [PMID: 28454538 PMCID: PMC5410041 DOI: 10.1186/s12906-017-1715-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 04/01/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND The hydrophobic triterpenes, oleanolic and betulinic acid as well as the hydrophilic mistletoe lectins and viscotoxins possess anticancer properties. They do all occur in combination in European mistletoe (Viscum album L.). Commercial Viscum album L. extracts are aqueous, excluding the insoluble triterpenes. We have previously shown that mistletoe lectins and triterpene acids are effective against Ewing sarcoma in vitro, ex vivo and in vivo. METHODS We recreated a total mistletoe effect (viscumTT) by combining an aqueous extract (viscum) and a triterpene extract (TT) solubilised with cyclodextrins and analysed the effects of viscumTT and the single extracts on TC-71 Ewing sarcoma cells in vitro by transcriptomic and proteomic profiling. RESULTS Treatment with the extracts strongly impacted Ewing sarcoma cell gene and protein expression. Apoptosis-associated and stress-activated genes were upregulated, proteasomal protein abundance enhanced and ribosomal and spliceosomal proteins downregulated. The mechanism of action of viscum, TT and viscumTT in TC-71 and MHH-ES-1 cells suggests the involvement of the unfolded protein response. While viscum and viscumTT extract treatment indicate response to oxidative stress and activation of stress-mediated MAPK signalling, TT extract treatment suggests the involvement of TLR signalling and autophagy. CONCLUSIONS Since the combinatory extract viscumTT exerts highly effective pro-apoptotic effects on Ewing sarcoma cells in vitro, this phytopolychemotherapy could be a promising adjuvant therapeutic option for paediatric patients with Ewing sarcoma.
Collapse
Affiliation(s)
- M Twardziok
- Department of Paediatric Oncology/Hematology, Otto Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Institute of Pharmacy, Department of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
- Department of Paediatrics, Dr. von Haunersches Kinderspital, Klinikum der Universität München, Munich, Germany
| | - D Meierhofer
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - S Börno
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - B Timmermann
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - S Jäger
- Birken AG, Niefern-Oeschelbronn, Germany
| | - Sengül Boral
- Department of Pathology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - A Eggert
- Department of Paediatric Oncology/Hematology, Otto Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - C I Delebinski
- Department of Paediatric Oncology/Hematology, Otto Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - G Seifert
- Department of Paediatric Oncology/Hematology, Otto Heubner Centre for Paediatric and Adolescent Medicine (OHC), Charité, Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
37
|
Wang Y, Luo Y, Li C, Zhang X, Pi C, Yu L, Wang S, Zhong Z. Optimized formulation of multivesicular liposomes loaded with oleanolic acid enhanced anticancer effect in vitro. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:955-968. [PMID: 28392677 PMCID: PMC5376187 DOI: 10.2147/dddt.s128795] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Invasion and metastasis are the main causes leading to the death of patients with hepatocellular carcinoma (HCC). Multivesicular liposomes loaded with oleanolic acid (OA-MVLs) have been well demonstrated to suppress survival, growth and angiogenesis of HCC cells. Emerging evidence demonstrates that OA was able to suppress the invasion of HCC cells by down-regulating myocyte enhancer factor-2. We hypothesized that the optimized OA-MVLs could inhibit the migration and invasion of HCC cells. In this study, we utilized central composite design and response surface methodology to assess the influence of some parameters on particle size and encapsulation efficiency and obtain the optimized formulation of OA-MVLs. Subsequently, the human HCC cell lines SMMC-7721 and HepG2 were treated with different doses of OA-MVLs and OA, respectively. Cellular survival, adhesion, migration and invasion in vitro were evaluated. We found that the optimized OA-MVLs significantly decreased the ability of HCC cells to adhere, migrate and invade in vitro. Furthermore, OA-MVLs significantly inhibited the survival of HCC cells at 160 µmol/L but showed no obvious inhibition effect on the cell vitality of normal liver cells. Our findings indicate that OA-MVLs did inhibit the cell survival, adhesion, invasion and metastasis of HCC cells in vitro. Although the involved mechanisms are still unclear, our findings can contribute to a better development of a preventive and therapeutic strategy for human HCC.
Collapse
Affiliation(s)
- Yunlong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Yuling Luo
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Xiaoqin Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Chao Pi
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Lu Yu
- Department of Chemistry, The Institute of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Shurong Wang
- Department of Pharmaceutical Sciences, School of Pharmacy
| | - Zhirong Zhong
- Department of Pharmaceutical Sciences, School of Pharmacy
| |
Collapse
|
38
|
Oleanolic Acid Alters Multiple Cell Signaling Pathways: Implication in Cancer Prevention and Therapy. Int J Mol Sci 2017; 18:ijms18030643. [PMID: 28300756 PMCID: PMC5372655 DOI: 10.3390/ijms18030643] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 12/13/2022] Open
Abstract
Nowadays, much attention has been paid to diet and dietary supplements as a cost-effective therapeutic strategy for prevention and treatment of a myriad of chronic and degenerative diseases. Rapidly accumulating scientific evidence achieved through high-throughput technologies has greatly expanded the understanding about the multifaceted nature of cancer. Increasingly, it is being realized that deregulation of spatio-temporally controlled intracellular signaling cascades plays a contributory role in the onset and progression of cancer. Therefore, targeting regulators of oncogenic signaling cascades is essential to prevent and treat cancer. A plethora of preclinical and epidemiological evidences showed promising role of phytochemicals against several types of cancer. Oleanolic acid, a common pentacyclic triterpenoid, is mainly found in olive oil, as well as several plant species. It is a potent inhibitor of cellular inflammatory process and a well-known inducer of phase 2 xenobiotic biotransformation enzymes. Main molecular mechanisms underlying anticancer effects of oleanolic acid are mediated by caspases, 5' adenosine monophosphate-activated protein kinase, extracellular signal-regulated kinase 1/2, matrix metalloproteinases, pro-apoptotic Bax and bid, phosphatidylinositide 3-kinase/Akt1/mechanistic target of rapamycin, reactive oxygen species/apoptosis signal-regulating kinase 1/p38 mitogen-activated protein kinase, nuclear factor-κB, cluster of differentiation 1, CKD4, s6k, signal transducer and activator of transcription 3, as well as aforementioned signaling pathways . In this work, we critically review the scientific literature on the molecular targets of oleanolic acid implicated in the prevention and treatment of several types of cancer. We also discuss chemical aspects, natural sources, bioavailability, and safety of this bioactive phytochemical.
Collapse
|
39
|
Di Gangi IM, Mazza T, Fontana A, Copetti M, Fusilli C, Ippolito A, Mattivi F, Latiano A, Andriulli A, Vrhovsek U, Pazienza V. Metabolomic profile in pancreatic cancer patients: a consensus-based approach to identify highly discriminating metabolites. Oncotarget 2016; 7:5815-29. [PMID: 26735340 PMCID: PMC4868723 DOI: 10.18632/oncotarget.6808] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/26/2015] [Indexed: 12/21/2022] Open
Abstract
Purpose pancreatic adenocarcinoma is the fourth leading cause of cancer related deaths due to its aggressive behavior and poor clinical outcome. There is a considerable variability in the frequency of serum tumor markers in cancer' patients. We performed a metabolomics screening in patients diagnosed with pancreatic cancer. Experimental Design Two targeted metabolomic assays were conducted on 40 serum samples of patients diagnosed with pancreatic cancer and 40 healthy controls. Multivariate methods and classification trees were performed. Materials and Methods Sparse partial least squares discriminant analysis (SPLS-DA) was used to reduce the high dimensionality of a pancreatic cancer metabolomic dataset, differentiating between pancreatic cancer (PC) patients and healthy subjects. Using Random Forest analysis palmitic acid, 1,2-dioleoyl-sn-glycero-3-phospho-rac-glycerol, lanosterol, lignoceric acid, 1-monooleoyl-rac-glycerol, cholesterol 5α,6α epoxide, erucic acid and taurolithocholic acid (T-LCA), oleoyl-L-carnitine, oleanolic acid were identified among 206 metabolites as highly discriminating between disease states. Comparison between Receiver Operating Characteristic (ROC) curves for palmitic acid and CA 19-9 showed that the area under the ROC curve (AUC) of palmitic acid (AUC=1.000; 95% confidence interval) is significantly higher than CA 19-9 (AUC=0.963; 95% confidence interval: 0.896-1.000). Conclusion Mass spectrometry-based metabolomic profiling of sera from pancreatic cancer patients and normal subjects showed significant alterations in the profiles of the metabolome of PC patients as compared to controls. These findings offer an information-rich matrix for discovering novel candidate biomarkers with diagnostic or prognostic potentials.
Collapse
Affiliation(s)
- Iole Maria Di Gangi
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), San Michele all'Adige, TN, Italy
| | - Tommaso Mazza
- Unit of Bioinformatics, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Andrea Fontana
- Unit of Biostatistics I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Caterina Fusilli
- Unit of Bioinformatics, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Antonio Ippolito
- Gastroenterology Unit, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Fulvio Mattivi
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), San Michele all'Adige, TN, Italy
| | - Anna Latiano
- Gastroenterology Unit, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Angelo Andriulli
- Gastroenterology Unit, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| | - Urska Vrhovsek
- Department of Food Quality and Nutrition, Research and Innovation Centre, Fondazione Edmund Mach (FEM), San Michele all'Adige, TN, Italy
| | - Valerio Pazienza
- Gastroenterology Unit, I.R.C.C.S. "Casa Sollievo della Sofferenza" Hospital, San Giovanni Rotondo, FG, Italy
| |
Collapse
|
40
|
Wu H, Zhong Q, Zhong R, Huang H, Xia Z, Ke Z, Zhang Z, Song J, Jia X. Preparation and antitumor evaluation of self-assembling oleanolic acid-loaded Pluronic P105/d-α-tocopheryl polyethylene glycol succinate mixed micelles for non-small-cell lung cancer treatment. Int J Nanomedicine 2016; 11:6337-6352. [PMID: 27932881 PMCID: PMC5135287 DOI: 10.2147/ijn.s119839] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Oleanolic acid (OA) is a triterpenoid found in various fruits and vegetables and used in traditional Chinese medicine. OA plays a crucial role in the treatment of several cancers, but poor water solubility, low permeability, and significant efflux have limited its widespread clinical use. Vitamin E-d-α-tocopheryl polyethylene glycol succinate (vitamin E-TPGS) and Pluronic P105 were used to improve the solubility and permeability and to decrease the efflux of OA. OA-loaded mixed micelles were prepared by ethanol thin-film hydration. The physicochemical properties of the micelles, including zeta potential, morphology, particle size, solubility, drug loading, and drug entrapment efficiency were characterized. OA release from micelles was slower than that from the free drug system. OA uptake by A549 non-small-cell lung cancer (NSCLC) cells was enhanced by the micelles. A tumor model was established by injecting A549 cells into nude mice. In vivo imaging showed that OA-micelles could accumulate in the tumors of nude mice. Additionally, smaller tumor size and increased expression of pro-apoptotic proteins were observed in OA-micelle-treated mice, indicating that OA-micelles are more effective than free OA in treating cancer. In vitro experiments were performed using two NSCLC cell lines (A549 and PC-9). Cytotoxicity evaluations showed that the half-maximal inhibitory concentrations of free OA and OA-micelles were 36.8±4.8 and 20.9±3.7 μM, respectively, in A549 cells and 82.7±7.8 and 56.7±4.7 μM, respectively, in PC-9 cells. Apoptosis assays revealed that the apoptotic rate of OA-micelle-treated A549 and PC-9 cells was higher than that of cells treated with the same concentration of free OA. Wound healing and transwell assays showed that migration and invasion were significantly suppressed in OA-micelle-treated cells. Immunofluorescence and Western blot analyses confirmed that the epithelial–mesenchymal transition was reversed in OA-micelle-treated cells. Mixed micelles are a promising nano-drug delivery system for lung cancer treatment.
Collapse
Affiliation(s)
- Hao Wu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui
| | - Qingxiang Zhong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu
| | - Rongling Zhong
- Laboratory Animal Center, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu
| | - Houcai Huang
- Laboratory Animal Center, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu
| | - Zhi Xia
- Laboratory Animal Center, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu
| | - Zhongcheng Ke
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; College of Chemistry and Chemical Engineering, Huangshan University, Huangshan, Anhui, People's Republic of China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine
| | - Jie Song
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu
| | - Xiaobin Jia
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine; Key Laboratory of New Drug Delivery System of Chinese Materia Medica, Jiangsu Province Academy of Chinese Medicine, Nanjing, Jiangsu; College of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui
| |
Collapse
|
41
|
Li W, Wu J, Li Z, Zhou Z, Zheng C, Lin L, Tan B, Huang M, Fan M. Melatonin induces cell apoptosis in Mia PaCa-2 cells via the suppression of nuclear factor-κB and activation of ERK and JNK: A novel therapeutic implication for pancreatic cancer. Oncol Rep 2016; 36:2861-2867. [DOI: 10.3892/or.2016.5100] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/30/2016] [Indexed: 11/05/2022] Open
|
42
|
Cao J, Li G, Wang M, Li H, Han Z. Protective effect of oleanolic acid on oxidized-low density lipoprotein induced endothelial cell apoptosis. Biosci Trends 2016; 9:315-24. [PMID: 26559024 DOI: 10.5582/bst.2015.01094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oleanolic acid (3β-hydroxyolean-12-en-28-oic acid, OA) is a naturally-occurring triterpenoid with various promising pharmacological properties. The present study was conducted to determine the protective effects of OA against oxidized low-density lipoprotein (ox-LDL) induced endothelial cell apoptosis and the possible underlying mechanisms. Our results showed that ox-LDL significantly decreased cell viability and induced apoptosis in human umbilical vein endothelial cells (HUVECs). OA in the co-treatment showed a protective effect against ox-LDL induced loss in cell viability and an increase in apoptosis, which was associated with the modulating effect of OA on ox-LDL induced hypoxia-inducible factor 1α(HIF-1α) expression. Moreover, our results showed that the modulating effect of OA against ox-LDL induced HIF-1α expression was obtained via inhibition of lipoprotein receptor 1 (LOX-1)/reactive oxygen species (ROS) signaling. Collectively, we suggested that the protective effect of OA against ox-LDL induced HUVEC apoptosis might, at least in part, be obtained via inhibition of the LOX-1/ROS/HIF-1α signaling pathway.
Collapse
Affiliation(s)
- Jianhua Cao
- Department of Pharmacy, the Third People's Hospital of Qingdao
| | | | | | | | | |
Collapse
|
43
|
Han D, Zhang X, Zhang J, Guo X, Zheng Y, Sui S, Zheng J. Oleanolic acid suppresses vascular smooth muscle cell proliferation by increasing lincRNA-p21 expression. Oncol Lett 2016; 12:3519-3522. [PMID: 27900030 DOI: 10.3892/ol.2016.5096] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/07/2016] [Indexed: 11/06/2022] Open
Abstract
Arteriosclerosis poses a significant risk to human health and involves the thickening and hardening of the walls of arteries. Accumulated evidence demonstrates that aberrant proliferation of vascular smooth muscle cells (VSMCs) accounts for the onset and progression of arteriosclerosis. Suppression of their proliferation has been demonstrated to be an effective anti-arteriosclerosis strategy. Long non-coding RNAs (lncRNAs) have recently been observed to be implicated in the proliferation of VSMCs and arteriosclerosis. In this study, we observed that oleanolic acid (OA), a natural compound from plants, inhibited the proliferation of VSMCs. The expression of lincRNA-p21, an arteriosclerosis-associated lncRNA, was demonstrated to be elevated by OA treatment. Suppression of lincRNA-p21 rescued the effect of OA on the proliferation of VSMCs. Collectively, targeting lncRNA is a promising strategy for arteriosclerosis prevention and treatment, and OA ameliorates arteriosclerosis by increasing lncRNA levels.
Collapse
Affiliation(s)
- Di Han
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Xuejuan Zhang
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Jietao Zhang
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Xiaozi Guo
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Yu Zheng
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266031, P.R. China
| | - Shihua Sui
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| | - Jiaping Zheng
- Department of Neurology, People's Hospital of Rizhao, Rizhao, Shandong 276800, P.R. China
| |
Collapse
|
44
|
Identification of a novel oxidative stress induced cell death by Sorafenib and oleanolic acid in human hepatocellular carcinoma cells. Biochem Pharmacol 2016; 118:9-17. [PMID: 27544320 DOI: 10.1016/j.bcp.2016.08.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/03/2016] [Indexed: 12/12/2022]
Abstract
The lack of effective chemotherapies in hepatocellular carcinoma (HCC) is still an unsolved problem and underlines the need for new strategies in liver cancer treatment. In this study, we present a novel approach to improve the efficacy of Sorafenib, today's only routinely used chemotherapeutic drug for HCC, in combination with triterpenoid oleanolic acid (OA). Our data show that cotreatment with subtoxic concentrations of Sorafenib and OA leads to highly synergistic induction of cell death. Importantly, Sorafenib/OA cotreatment triggers cell damage in a sustained manner and suppresses long-term clonogenic survival. Sorafenib/OA cotreatment induces DNA fragmentation and caspase-3/7 cleavage and the addition of the pan-caspase inhibitor zVAD.fmk shows the requirement of caspase activation for Sorafenib/OA-triggered cell death. Furthermore, Sorafenib/OA co-treatment stimulates a significant increase in reactive oxygen species (ROS) levels. Most importantly, the accumulation of intracellular ROS is required for cell death induction, since the addition of ROS scavengers (i.e. α-tocopherol, MnTBAP) that prevent the increase of intracellular ROS levels completely rescues cells from Sorafenib/OA-triggered cell death. In conclusion, OA represents a novel approach to increase the sensitivity of HCC cells to Sorafenib via oxidative stress.
Collapse
|
45
|
Liu J, Ma L, Chen X, Wang J, Yu T, Gong Y, Ma A, Zheng L, Liang H. ERK inhibition sensitizes cancer cells to oleanolic acid-induced apoptosis through ERK/Nrf2/ROS pathway. Tumour Biol 2015; 37:8181-7. [DOI: 10.1007/s13277-015-4668-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/16/2015] [Indexed: 11/28/2022] Open
|
46
|
Wang Y, Wang R, Wang Y, Peng R, Wu Y, Yuan Y. Ginkgo biloba extract mitigates liver fibrosis and apoptosis by regulating p38 MAPK, NF-κB/IκBα, and Bcl-2/Bax signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:6303-17. [PMID: 26664050 PMCID: PMC4671772 DOI: 10.2147/dddt.s93732] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Liver fibrosis is the consequence of diverse liver injuries and can eventually develop into liver cirrhosis. Ginkgo biloba extract (GBE) is an extract from dried ginkgo leaves that has many pharmacological effects because of its various ingredients and has been shown to be hepatoprotective. Purpose and methods Aimed to investigate the underlying protective mechanisms of GBE on carbon tetrachloride (CCl4)-induced liver fibrosis in rats. Male Sprague Dawley rats were randomly divided into four groups: control group (C), model group (M), low-dose group (L), and high-dose group (H). Liver fibrosis was induced by CCl4 groups M, L, and H: group C was administered saline. In addition, GBE at different doses was used to treat groups L and H. Results The results of hematoxylin and eosin staining, Masson’s trichrome staining, a liver function index, and a liver fibrosis index showed that GBE application noticeably mitigated fibrosis and improved the function of the liver. The western blotting and immunohistochemistry analyses indicated that GBE reduced liver fibrosis not only by inhibiting p38 MAPK and NF-κBp65 via inhibition of IκBα degradation but also by inhibiting hepatocyte apoptosis via downregulation of Bax, upregulation of Bcl-2, and subsequent inhibition of caspase-3 activation. Inflammation-associated factors and hepatic stellate cell (HSC)-activation markers further demonstrated that GBE could effectively inhibit HSC activation and inflammation as a result of its regulation of p38 MAPK and nuclear factor-kappa B/IκBα signaling. Conclusion Our findings indicated a novel role for GBE in the treatment of liver fibrosis. The potential mechanisms may be associated with the following signaling pathways: 1) the p38 MAPK and nuclear factor-kappa B/IκBα signaling pathways (inhibiting inflammation and HSCs activation) and 2) the Bcl-2/Bax signaling pathway (inhibiting the apoptosis of hepatocytes).
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Rong Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yujie Wang
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Ruqin Peng
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yan Wu
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yongfang Yuan
- Department of Pharmacy, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
47
|
Zu C, Zhang M, Xue H, Cai X, Zhao L, He A, Qin G, Yang C, Zheng X. Emodin induces apoptosis of human breast cancer cells by modulating the expression of apoptosis-related genes. Oncol Lett 2015; 10:2919-2924. [PMID: 26722264 DOI: 10.3892/ol.2015.3646] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 07/28/2015] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the effects of emodin on the proliferation of human breast cancer cells Bcap-37 and ZR-75-30. Cell viability following emodin treatment was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effects of emodin on apoptosis were determined by flow cytometry using Annexin V-fluorescein isothiocyanate and propidium iodide staining. Quantitative polymerase chain reaction and western blot analysis were used to determine changes in the expression of apoptotic genes and protein, respectively. The effect of emodin on the invasiveness of breast cancer cells was evaluated by Matrigel invasion assay. Treatment of breast cancer cells Bcap-37 and ZR-75-30 with emodin was observed to inhibit the growth and induced apoptosis in a time- and dose-dependent manner. Emodin reduced the level of Bcl-2 and increased levels of cleaved caspase-3, PARP, p53 and Bax. These findings indicate that emodin induces growth inhibition and apoptosis in human breast cancer cells. Emodin may be a potential therapeutic agent for the treatment of breast cancer.
Collapse
Affiliation(s)
- Cong Zu
- Lab 1, Cancer Institute, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Mingdi Zhang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai 200092, P.R. China
| | - Hui Xue
- Department of Gynecology, First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaopeng Cai
- Department of Surgical Oncology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Lei Zhao
- Center of Experiment Technology and Medical Research, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Anning He
- Lab 1, Cancer Institute, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Guangyuan Qin
- Lab 1, Cancer Institute, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chunshu Yang
- Lab 1, Cancer Institute, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinyu Zheng
- Lab 1, Cancer Institute, China Medical University, Shenyang, Liaoning 110001, P.R. China ; Department of Breast Surgery, First Affiliated Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
48
|
Curcumol Inhibits Growth and Induces Apoptosis of Colorectal Cancer LoVo Cell Line via IGF-1R and p38 MAPK Pathway. Int J Mol Sci 2015; 16:19851-67. [PMID: 26307972 PMCID: PMC4581329 DOI: 10.3390/ijms160819851] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 07/27/2015] [Accepted: 07/30/2015] [Indexed: 01/07/2023] Open
Abstract
Curcumol, isolated from the traditional medical plant Rhizoma Curcumae, is the bioactive component of Zedoary oil, whose potential anti-tumor effect has attracted considerable attention in recent years. Though many researchers have reported curcumol and its bioactivity, the potential molecular mechanism for its anti-cancer effect in colorectal cancer LoVo cells still remains unclear. In the present study, we found that curcumol showed growth inhibition and induced apoptosis of LoVo cells in a dose- and time-dependent manner. The occurrence of its proliferation inhibition and apoptosis came with suppression of IGF-1R expression, and then increased the phosphorylation of p38 mitogen activated protein kinase (MAPK), which might result in a cascade response by inhibiting the CREB survival pathway and finally triggered Bax/Bcl-2 and poly(ADP-ribose) polymerase 1 (PARP-1) apoptosis signals. Moreover, curcumol inhibited colorectal cancer in xenograft models of nude mice. Immunohistochemical and Western blot analysis revealed that curcumol could decrease the expression of ki-67, Bcl-2 as well as CREB1, and increase the expression of Bax and the phosphorylation of p38, which were consistent with our in vitro study. Overall, our in vitro and in vivo data confirmed the anti-cancer activity of curcumol, which was related to a significant inhibition of IGF-1R and activation of p38 MAPKs, indicating that curcumol may be a potential anti-tumor agent for colorectal carcinoma therapy.
Collapse
|
49
|
Li HF, Wang XA, Xiang SS, Hu YP, Jiang L, Shu YJ, Li ML, Wu XS, Zhang F, Ye YY, Weng H, Bao RF, Cao Y, Lu W, Dong Q, Liu YB. Oleanolic acid induces mitochondrial-dependent apoptosis and G0/G1 phase arrest in gallbladder cancer cells. Drug Des Devel Ther 2015; 9:3017-3030. [PMID: 26109845 PMCID: PMC4472077 DOI: 10.2147/dddt.s84448] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Oleanolic acid (OA), a naturally occurring triterpenoid, exhibits potential antitumor activity in many tumor cell lines. Gallbladder carcinoma is the most common malignancy of the biliary tract, and is a highly aggressive tumor with an extremely poor prognosis. Unfortunately, the effects of OA on gallbladder carcinoma are unknown. In this study, we investigated the effects of OA on gallbladder cancer cells and the underlying mechanism. The results showed that OA inhibits proliferation of gallbladder cancer cells in a dose-dependent and time-dependent manner on MTT and colony formation assay. A flow cytometry assay revealed apoptosis and G0/G1 phase arrest in GBC-SD and NOZ cells. Western blot analysis and a mitochondrial membrane potential assay demonstrated that OA functions through the mitochondrial apoptosis pathway. Moreover, this drug inhibited tumor growth in nude mice carrying subcutaneous NOZ tumor xenografts. These data suggest that OA inhibits proliferation of gallbladder cancer cells by regulating apoptosis and the cell cycle process. Thus, OA may be a promising drug for adjuvant chemotherapy in gallbladder carcinoma.
Collapse
Affiliation(s)
- Huai-Feng Li
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Xu-An Wang
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Shan-Shan Xiang
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Yun-Ping Hu
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Lin Jiang
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Yi-Jun Shu
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Mao-Lan Li
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Xiang-Song Wu
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Fei Zhang
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Yuan-Yuan Ye
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Hao Weng
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Run-Fa Bao
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Yang Cao
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Wei Lu
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Qian Dong
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| | - Ying-Bin Liu
- Department of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Laboratory of General Surgery, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
- Institute of Biliary Tract Disease, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
50
|
Liu QQ, Zhang FF, Wang F, Qiu JH, Luo CH, Zhu GY, Liu YF. TIPE2 Inhibits Lung Cancer Growth Attributing to Promotion of Apoptosis by Regulating Some Apoptotic Molecules Expression. PLoS One 2015; 10:e0126176. [PMID: 25946186 PMCID: PMC4422750 DOI: 10.1371/journal.pone.0126176] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/30/2015] [Indexed: 12/25/2022] Open
Abstract
Recent studies found that TIPE2 was involved in cancer development. However, little is known about TIPE2 in lung cancer. Our study aims to clarify the role of TIPE2 in lung carcinogenesis. We examined the expression of TIPE2 in lung squamous cancer (LSC), small cell lung cancer and lung adenocarcinoma (AdC) tissues and found that TIPE2 expression was lost in small cell lung cancer, compared with adjacent non-tumor tissues. Overexpression of TIPE2 significantly inhibited the growth of lung cancer cell H446 in vitro and even suppressed tumor formation in vivo. Flow cytometry analysis found TIPE2 overexpression promoted apoptosis of H446. In TIPE2 over-expression cells, caspase-3, caspase-9, and Bax were significantly up-regulated while Bcl-2 was down-regulated. Moreover, coincident results were shown by immunohistochemistry in tumors from nude mice. TIPE2 inhibited the phosphorylation of Akt, while promoting the phosphorylation of P38, but had no effect on IκBα and ERK pathway. Taken together, TIPE2 promoted lung cancer cell apoptosis through affecting apoptosis-related molecules caspase-3, caspase-9, Bcl-2 and Bax, possibly via regulating P38 and Akt pathways, indicating that TIPE2 might be a novel marker for lung cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Qing-Qing Liu
- Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, Fujian, PR China
| | - Feng-Feng Zhang
- Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, Fujian, PR China
| | - Fang Wang
- Department of Basic Medicine Science, NanYang Medical College, Nanyang, China
| | - Jing-Hua Qiu
- Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, Fujian, PR China
| | - Chun-Hua Luo
- The Department of Pathology, The Traditional Chinese Medical Hospital of Xiamen, Xiamen, Fujian, People’s Republic of China
| | - Guo-Yong Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, PR China
| | - Ying-Fu Liu
- Department of Basic Medical Sciences, Medical College, Xiamen University, Xiamen, Fujian, PR China
- * E-mail:
| |
Collapse
|