1
|
Zhang X, Lou X, Qiao H, Jiang Z, Sun H, Shi X, He Z, Sun J, Sun M. Supramolecular self-sensitized dual-drug nanoassemblies potentiating chemo-photodynamic therapy for effective cancer treatment. Int J Pharm 2024; 662:124496. [PMID: 39033943 DOI: 10.1016/j.ijpharm.2024.124496] [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: 02/29/2024] [Revised: 06/30/2024] [Accepted: 07/18/2024] [Indexed: 07/23/2024]
Abstract
Chemo-photodynamic synergistic therapy (CPST) holds tremendous promise for treating cancers. Unfortunately, existing CPST applications suffer from complex synthetic procedures, low drug co-loading efficiency, and carrier-related toxicity. To address these issues, we have developed a supramolecular carrier-free self-sensitized nanoassemblies by co-assembling podophyllotoxin (PTOX) and chlorin e6 (Ce6) to enhance CPST efficiency against tumors. The nanoassemblies show stable co-assembly performance in simulative vivo neural environment (∼150 nm), with high co-loading ability for PTOX (72.2 wt%) and Ce6 (27.8 wt%). In vivo, the nanoassemblies demonstrate a remarkable ability to accumulate at tumor sites by leveraging the enhanced permeability and retention (EPR) effect. The disintegration of nanoassemblies following photosensitizer bioactivation triggered by the acidic tumor environment effectively resolves the challenge of aggregation-caused quenching (ACQ) effect. Upon exposure to external light stimulation, the disintegrated nanoassemblies not only illuminate cancer cells synergistically but also exert a more potent antitumor effect when compared with PTOX and Ce6 administered alone. This self-sensitized strategy represents a significant step forward in CPST, offering a unique co-delivery paradigm for clinic cancer treatment.
Collapse
Affiliation(s)
- Xu Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xinyu Lou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Han Qiao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Zhouyu Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Hang Sun
- Hong Kong Education University, Hong Kong SAR, 999077, China
| | - Xianbao Shi
- Department of Pharmacy, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| | - Mengchi Sun
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
2
|
Hernández ÁP, Chaparro-González L, Garzo-Sánchez O, Arias-Hidalgo C, Juanes-Velasco P, García PA, Castro MÁ, Fuentes M. Podophyllic Aldehyde, a Podophyllotoxin Derivate, Elicits Different Cell Cycle Profiles Depending on the Tumor Cell Line: A Systematic Proteomic Analysis. Int J Mol Sci 2024; 25:4631. [PMID: 38731850 PMCID: PMC11083757 DOI: 10.3390/ijms25094631] [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: 03/15/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
When new antitumor therapy drugs are discovered, it is essential to address new target molecules from the point of view of chemical structure and to carry out efficient and systematic evaluation. In the case of natural products and derived compounds, it is of special importance to investigate chemomodulation to further explore antitumoral pharmacological activities. In this work, the compound podophyllic aldehyde, a cyclolignan derived from the chemomodulation of the natural product podophyllotoxin, has been evaluated for its viability, influence on the cell cycle, and effects on intracellular signaling. We used functional proteomics characterization for the evaluation. Compared with the FDA-approved drug etoposide (another podophyllotoxin derivative), we found interesting results regarding the cytotoxicity of podophyllic aldehyde. In addition, we were able to observe the effect of mitotic arrest in the treated cells. The use of podophyllic aldehyde resulted in increased cytotoxicity in solid tumor cell lines, compared to etoposide, and blocked the cycle more successfully than etoposide. High-throughput analysis of the deregulated proteins revealed a selective antimitotic mechanism of action of podophyllic aldehyde in the HT-29 cell line, in contrast with other solid and hematological tumor lines. Also, the apoptotic profile of podophyllic aldehyde was deciphered. The cell death mechanism is activated independently of the cell cycle profile. The results of these targeted analyses have also shown a significant response to the signaling of kinases, key proteins involved in signaling cascades for cell proliferation or metastasis. Thanks to this comprehensive analysis of podophyllic aldehyde, remarkable cytotoxic, antimitotic, and other antitumoral features have been discovered that will repurpose this compound for further chemical transformations and antitumoral analysis.
Collapse
Affiliation(s)
- Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
- Department of Pharmaceutical Sciences, Laboratory of Medicinal Chemistry, Faculty of Pharmacy, University of Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (P.A.G.); (M.Á.C.)
| | - Lorea Chaparro-González
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
| | - Olga Garzo-Sánchez
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
| | - Carlota Arias-Hidalgo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
| | - Pablo A. García
- Department of Pharmaceutical Sciences, Laboratory of Medicinal Chemistry, Faculty of Pharmacy, University of Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (P.A.G.); (M.Á.C.)
| | - Mª Ángeles Castro
- Department of Pharmaceutical Sciences, Laboratory of Medicinal Chemistry, Faculty of Pharmacy, University of Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (P.A.G.); (M.Á.C.)
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), IBSAL, University of Salamanca-CSIC, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain; (L.C.-G.); (O.G.-S.); (C.A.-H.); (P.J.-V.); (M.F.)
- Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
3
|
Zhao X, Chinnathambi A, Alharbi SA, Natarajan N, Raman M. Nerolidol, Bioactive Compound Suppress Growth of HCT-116 Colorectal Cancer Cells Through Cell Cycle Arrest and Induction of Apoptosis. Appl Biochem Biotechnol 2024; 196:1365-1375. [PMID: 37395945 DOI: 10.1007/s12010-023-04612-9] [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] [Accepted: 06/19/2023] [Indexed: 07/04/2023]
Abstract
Colon cancer is the most prevalent cancer and causes the highest cancer-associated mortality in both men and women globally. It has a high incidence and fatality rate, which places a significant burden on the healthcare system. The current work was performed to understand the beneficial roles of nerolidol on the viability and cytotoxic mechanisms in the colon cancer HCT-116 cells. The MTT cytotoxicity assay was done to investigate the effect of nerolidol at different doses (5-100 µM) on the HCT-116 cell viability. The impacts of nerolidol on ROS accumulation and apoptosis were investigated using DCFH-DA, DAPI, and dual staining assays, respectively. The flow cytometry analysis was performed to study the influence of nerolidol on the cell cycle arrest in the HCT-116 cells. The outcomes of the MTT assay demonstrated that nerolidol at different doses (5-100 µM) substantially inhibited the HCT-116 cell viability with an IC50 level of 25 µM. The treatment with nerolidol appreciably boosted the ROS level in the HCT-116 cells. The findings of DAPI and dual staining revealed higher apoptotic incidences in the nerolidol-exposed HCT-116 cells, which supports its ability to stimulate apoptosis. The flow cytometry analysis demonstrated the considerable inhibition in cell cycle at the G0/G1 phase in the nerolidol-exposed HCT-116 cells. Our research showed that nerolidol can inhibit the cell cycle, increase ROS accumulation, and activate apoptosis in HCT-116 cells. In light of this, it may prove to be a potent and salutary candidate to treat colon cancer.
Collapse
Affiliation(s)
- Xiaoqian Zhao
- Nuclear Medicine Department, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, PO Box -2455, Riyadh, 11451, Saudi Arabia
| | - Nandakumar Natarajan
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX, 75708, USA
| | - Muthusamy Raman
- Department of Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
4
|
Hao M, Xu H. Chemistry and Biology of Podophyllotoxins: An Update. Chemistry 2024; 30:e202302595. [PMID: 37814110 DOI: 10.1002/chem.202302595] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/11/2023]
Abstract
Podophyllotoxin is an aryltetralin lignan lactone derived from different plants of Podophyllum. It consists of five rings with four chiral centers, one trans-lactone and one aryl tetrahydronaphthalene skeleton with multiple modification sites. Moreover, podophyllotoxin and its derivatives showed lots of bioactivities, including anticancer, anti-inflammatory, antiviral, and insecticidal properties. The demand for podophyllotoxin and its derivatives is rising as a result of their high efficacy. As a continuation of our previous review (Chem. Eur. J., 2017, 23, 4467-4526), herein, total synthesis, biotransformation, structural modifications, bioactivities, and structure-activity relationships of podophyllotoxin and its derivatives from 2017 to 2022 are summarized. Meanwhile, a piece of update information on the origin of new podophyllotoxin analogues from plants from 2014 to 2022 was compiled. We hope that this review will provide a reference for future high value-added applications of podophyllotoxin and its analogues in the pharmaceutical and agricultural fields.
Collapse
Affiliation(s)
- Meng Hao
- College of Plant Protection, Northwest A&F University, Xian Yang Shi, Yangling, 712100, P.R. China
| | - Hui Xu
- College of Plant Protection, Northwest A&F University, Xian Yang Shi, Yangling, 712100, P.R. China
| |
Collapse
|
5
|
Rungrasameviriya P, Santilinon A, Atichartsintop P, Hadpech S, Thongboonkerd V. Tight junction and kidney stone disease. Tissue Barriers 2024; 12:2210051. [PMID: 37162265 PMCID: PMC10832927 DOI: 10.1080/21688370.2023.2210051] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/30/2023] [Indexed: 05/11/2023] Open
Abstract
Defects of tight junction (TJ) are involved in many diseases related to epithelial cell functions, including kidney stone disease (KSD), which is a common disease affecting humans for over a thousand years. This review provides brief overviews of KSD and TJ, and summarizes the knowledge on crystal-induced defects of TJ in renal tubular epithelial cells (RTECs) in KSD. Calcium oxalate (CaOx) crystals, particularly COM, disrupt TJ via p38 MAPK and ROS/Akt/p38 MAPK signaling pathways, filamentous actin (F-actin) reorganization and α-tubulin relocalization. Stabilizing p38 MAPK signaling, reactive oxygen species (ROS) production, F-actin and α-tubulin by using SB239063, N-acetyl-L-cysteine (NAC), phalloidin and docetaxel, respectively, successfully prevent the COM-induced TJ disruption and malfunction. Additionally, genetic disorders of renal TJ, including mutations and single nucleotide polymorphisms (SNPs) of CLDN2, CLDN10b, CLDN14, CLDN16 and CLDN19, also affect KSD. Finally, the role of TJ as a potential target for KSD therapeutics and prevention is also discussed.
Collapse
Affiliation(s)
- Papart Rungrasameviriya
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Aticha Santilinon
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Palita Atichartsintop
- Nawamethee Project, Doctor of Medicine Program, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sudarat Hadpech
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
6
|
Kumari P, Dang S. Evaluation of Enhanced Cytotoxicity Effect of Repurposed Drug Simvastatin/Thymoquinone Combination against Breast Cancer Cell Line. Cardiovasc Hematol Agents Med Chem 2024; 22:348-366. [PMID: 37907488 DOI: 10.2174/0118715257259037231012182741] [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: 05/09/2023] [Revised: 08/02/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Repurposing of drugs for their anticancer potential is gaining a lot of importance in drug discovery. AIMS The present study aims to explore the potential of Simvastatin (SIM), a drug used in the treatment of high cholesterol, and Thymoquinone (Nigella Sativa) (THY) for its anti-cancer activity on breast cancer cell lines. Thymoquinone is reported to have many potential medicinal properties exhibiting antioxidant, antiinflammatory, anti-cancer, activities like inhibition of tissue growth and division. METHODS In this analysis, we explored the inhibitory effects of the combination of Simvastatin ad Thymoquinone on two breast cancer cell lines viz MCF-7 and MDA-MB-231 cells. The combined effect of Simvastatin and Thymoquinone on Cell viability, Colony formation, Cell migration, and orientation of more programmed cell death in vitro was studied. Cell cycle arrest in the G2/M phase was concomitant with the combined effect of SIM and THY persuading apoptosis and generating reactive oxygen species (ROS). RESULTS The cell cycle arrest with combined treatment was observed that only 1.8% and 1.1% cells gated in G2/M phase in MCF-7 & MDA-MB-231 cell. An increased apoptosis was observed when cells were treated in combination which was about 76.20% and 58.15% respectively for MCF-7 and MDA-MB-231 cells. CONCLUSION It was concluded that the combined effect of simvastatin and thymoquinone stimulates apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Pallavi Kumari
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| |
Collapse
|
7
|
Singhal S, Rani V. Cardioprotective Role of Tinospora cordifolia against Trimethylamine-N-Oxide and Glucose Induced Stress in Rat Cardiomyocytes. Cardiovasc Hematol Agents Med Chem 2024; 22:475-494. [PMID: 37907489 DOI: 10.2174/0118715257270512231013064533] [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: 07/04/2023] [Revised: 08/22/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023]
Abstract
BACKGROUND Type 2 diabetes has become a concern issue that affects the quality of life and can increase the risk of cardiac insufficiency elevating the threat to the life safety of patients. A recognized cause of cardiac insufficiency is diabetic cardiomyopathy, chronic hyperglycemia, and myocardial lipotoxicity which can reduce the myocardial contractile performance, and enhance the cardiomyocyte hypertrophy and interstitial fibrosis. The cause of diabetic cardiomyopathy is multi-factorial which includes oxidative stress, insulin resistance, inflammation, apoptosis, and autophagy. Recent clinical studies have suggested the dysbiosis of gut microbiota, secretion of metabolites, and their diffusion in to the host as to have direct detrimental effects on the cardiac contractility. MATERIALS AND METHODS In the present paper, we have done in silico studies including molecular interaction of phytoconstituents of Tinospora cordifolia against reactive oxygen species producing proteins. Whereas, in vitro studies were conducted on H9C2 cardiac cells including cell morphological examination, level of reactive oxygen species, cell count-viability, apoptotic status, in the presence of high glucose, trimethylamine-n-oxide, and plant extracts which were determined through cell analyzer and microscopic assays. RESULTS The treatment of high glucose and trimethylamine-n-oxide was found to be increase the cardiac stress approximately two fold by attenuating hypertrophic conditions, oxidative stress, and apoptosis in rat cardiomyocytes, and Tinospora cordifolia was found to be a cardioprotective agent. CONCLUSION Conclusively, our study has reported that the Indian medicinal plant Tinospora cordifolia has the ability to treat diabetic cardiomyopathy. Our study can open up a new herbal therapeutic strategy against diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Shivani Singhal
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida, 201307, Uttar Pradesh, India
| | - Vibha Rani
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida, 201307, Uttar Pradesh, India
| |
Collapse
|
8
|
Guo W, Xing Y, Luo X, Li F, Ren M, Liang Y. Reactive Oxygen Species: A Crosslink between Plant and Human Eukaryotic Cell Systems. Int J Mol Sci 2023; 24:13052. [PMID: 37685857 PMCID: PMC10487619 DOI: 10.3390/ijms241713052] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Reactive oxygen species (ROS) are important regulating factors that play a dual role in plant and human cells. As the first messenger response in organisms, ROS coordinate signals in growth, development, and metabolic activity pathways. They also can act as an alarm mechanism, triggering cellular responses to harmful stimuli. However, excess ROS cause oxidative stress-related damage and oxidize organic substances, leading to cellular malfunctions. This review summarizes the current research status and mechanisms of ROS in plant and human eukaryotic cells, highlighting the differences and similarities between the two and elucidating their interactions with other reactive substances and ROS. Based on the similar regulatory and metabolic ROS pathways in the two kingdoms, this review proposes future developments that can provide opportunities to develop novel strategies for treating human diseases or creating greater agricultural value.
Collapse
Affiliation(s)
- Wei Guo
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.G.); (Y.X.); (F.L.)
- National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Yadi Xing
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.G.); (Y.X.); (F.L.)
- National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Xiumei Luo
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610000, China;
| | - Fuguang Li
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.G.); (Y.X.); (F.L.)
- National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China
- Hainan Yazhou Bay Seed Laboratory, Sanya 572000, China
| | - Maozhi Ren
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.G.); (Y.X.); (F.L.)
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610000, China;
- Hainan Yazhou Bay Seed Laboratory, Sanya 572000, China
| | - Yiming Liang
- Zhengzhou Research Base, National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China; (W.G.); (Y.X.); (F.L.)
- National Key Laboratory of Cotton Bio-Breeding and Integrated Utilization, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang 455000, China
| |
Collapse
|
9
|
Chen JF, Wu SW, Shi ZM, Hu B. Traditional Chinese medicine for colorectal cancer treatment: potential targets and mechanisms of action. Chin Med 2023; 18:14. [PMID: 36782251 PMCID: PMC9923939 DOI: 10.1186/s13020-023-00719-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Colorectal cancer (CRC) is a disease with complex pathogenesis, it is prone to metastasis, and its development involves abnormalities in multiple signaling pathways. Surgery, chemotherapy, radiotherapy, target therapy, and immunotherapy remain the main treatments for CRC, but improvement in the overall survival rate and quality of life is urgently needed. Traditional Chinese medicine (TCM) has a long history of preventing and treating CRC. It could affect CRC cell proliferation, apoptosis, cell cycle, migration, invasion, autophagy, epithelial-mesenchymal transition, angiogenesis, and chemoresistance by regulating multiple signaling pathways, such as PI3K/Akt, NF-κB, MAPK, Wnt/β-catenin, epidermal growth factor receptors, p53, TGF-β, mTOR, Hedgehog, and immunomodulatory signaling pathways. In this paper, the main signaling pathways and potential targets of TCM and its active ingredients in the treatment of CRC were systematically summarized, providing a theoretical basis for treating CRC with TCM and new ideas for further exploring the pathogenesis of CRC and developing new anti-CRC drugs.
Collapse
Affiliation(s)
- Jin-Fang Chen
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Shi-Wei Wu
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Zi-Man Shi
- grid.412540.60000 0001 2372 7462Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China ,grid.412540.60000 0001 2372 7462Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032 Shanghai, People’s Republic of China
| | - Bing Hu
- Institute of Traditional Chinese Medicine in Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, People's Republic of China. .,Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, People's Republic of China.
| |
Collapse
|
10
|
Podophyllotoxin and its derivatives: Potential anticancer agents of natural origin in cancer chemotherapy. Biomed Pharmacother 2023; 158:114145. [PMID: 36586242 DOI: 10.1016/j.biopha.2022.114145] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/16/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
The use of plant secondary metabolites has gained considerable attention among clinicians in the prevention and treatment of cancer. A secondary metabolite isolated mainly from the roots and rhizomes of Podophyllum species (Berberidaceae) is aryltetralin lignan - podophyllotoxin (PTOX). The purpose of this review is to discuss the therapeutic properties of PTOX as an important anticancer compound of natural origin. The relevant information regarding the antitumor mechanisms of podophyllotoxin and its derivatives were collected and analyzed from scientific databases. The results of the analysis showed PTOX exhibits potent cytotoxic activity; however, it cannot be used in its pure form due to its toxicity and generation of many side effects. Therefore, it practically remains clinically unusable. Currently, high effort is focused on attempts to synthesize analogs of PTOX that have better properties for therapeutic use e.g. etoposide (VP-16), teniposide, etopophos. PTOX derivatives are used as anticancer drugs which are showing additional immunosuppressive, antiviral, antioxidant, hypolipemic, and anti-inflammatory effects. In this review, attention is paid to the high potential of the usefulness of in vitro cultures of P. peltatum which can be a valuable source of lignans, including PTOX. In conclusion, the preclinical pharmacological studies in vitro and in vivo confirm the anticancer and chemotherapeutic potential of PTOX and its derivatives. In the future, clinical studies on human subjects are needed to certify the antitumor effects and the anticancer mechanisms to be certified and analyzed in more detail and to validate the experimental pharmacological preclinical studies.
Collapse
|
11
|
Nathani S, Mishra R, Katiyar P, Sircar D, Roy P. Zinc Acts Synergistically with Berberine for Enhancing Its Efficacy as an Anti-cancer Agent by Inducing Clusterin-Dependent Apoptosis in HT-29 Colorectal Cancer Cells. Biol Trace Elem Res 2022:10.1007/s12011-022-03460-8. [PMID: 36394793 DOI: 10.1007/s12011-022-03460-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 10/22/2022] [Indexed: 11/18/2022]
Abstract
It is now widely accepted that anti-cancer medications are most effective when administered in combination. Zinc is an essential micronutrient whilst berberine is a well-known natural phytochemical, both having multiple molecular mechanisms of action. The present study aimed to determine the combinatorial effect of zinc and berberine on the human adenocarcinoma HT-29 cancer cell line. The anti-proliferative activity of berberine and zinc was determined by cell viability and colony-forming assays. The combination index and drug reduction index values of zinc and berberine co-treatments were estimated by suitable software. Flow cytometry was used to analyse cell cycle distribution and Annexin V/PI staining. The expression of apoptosis and zinc signalling markers were analysed by RT-qPCR and immunoblot analysis. Berberine decreased the viability of colon cancer cells in a dose-dependent manner whilst zinc alone had no significant influence on it. However, zinc and berberine co-treatment resulted in a synergistic anti-cancer action which was demonstrated by G2/M phase arrest of cell growth at a lower dose of berberine. Annexin V assay demonstrated that the synergistic impact of zinc and berberine boosted the number of apoptotic cells. Gene expression analysis at both transcriptional and translational levels showed the upregulation of apoptotic (caspase-3 and caspase-8) and a zinc-sensing receptor (GPR39) gene with concomitant downregulation of anti-apoptotic genes like proliferating cell nuclear antigen (PCNA) and clusterin. Our findings showed that the combination of zinc and berberine has synergistic anti-cancer efficacy and thus could be used as a potential chemopreventive option for colon cancer.
Collapse
Affiliation(s)
- Sandip Nathani
- Molecular Endocrinology Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247 667, Uttarakhand, India
| | - Rutusmita Mishra
- Molecular Endocrinology Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247 667, Uttarakhand, India
| | - Parul Katiyar
- Molecular Endocrinology Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247 667, Uttarakhand, India
| | - Debabrata Sircar
- Plant Molecular Biology Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247 667, Uttarakhand, India
| | - Partha Roy
- Molecular Endocrinology Laboratory, Department of Biosciences & Bioengineering, Indian Institute of Technology Roorkee, Roorkee, 247 667, Uttarakhand, India.
| |
Collapse
|
12
|
Shen S, Tong Y, Luo Y, Huang L, Gao W. Biosynthesis, total synthesis, and pharmacological activities of aryltetralin-type lignan podophyllotoxin and its derivatives. Nat Prod Rep 2022; 39:1856-1875. [PMID: 35913409 DOI: 10.1039/d2np00028h] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Covering: up to 2022Podophyllotoxin (PTOX, 1), a kind of aryltetralin-type lignan, was first discovered in the plant Podophyllum peltatum and its structure was clarified by W. Borsche and J. Niemann in 1932. Due to its potent anti-cancer and anti-viral activities, it is considered one of the molecules most likely to be developed into modern drugs. With the increasing market demand and insufficient storage of natural resources, it is crucial to expand the sources of PTOXs. The original extraction method from plants has gradually failed to meet the requirements, and the biosynthesis and total synthesis have become the forward-looking alternatives. As key enzymes in the biosynthetic pathway of PTOXs and their catalytic mechanisms being constantly revealed, it is possible to realize the heterogeneous biosynthesis of PTOXs in the future. Chemical and chemoenzymatic synthesis also provide schemes for strictly controlling the asymmetric configuration of the tetracyclic core. Currently, the pharmacological activities of some PTOX derivatives have been extensively studied, laying the foundation for clinical candidate drugs. This review focuses primarily on the latest research progress in the biosynthesis, total synthesis, and pharmacological activities of PTOX and its derivatives, providing a more comprehensive understanding of these widely used compounds and supporting the future search for clinical applications.
Collapse
Affiliation(s)
- Siyu Shen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China. .,Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| | - Yuru Tong
- School of Pharmaceutical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yunfeng Luo
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
| | - Luqi Huang
- State Key Laboratory Breeding Base of Dao-di Herbs, National Resource Center for Chinese Materia Medica, Chinese Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Wei Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China. .,Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China
| |
Collapse
|
13
|
Ye B, Ma J, Li Z, Li Y, Han X. Ononin Shows Anticancer Activity Against Laryngeal Cancer via the Inhibition of ERK/JNK/p38 Signaling Pathway. Front Oncol 2022; 12:939646. [PMID: 35912256 PMCID: PMC9334013 DOI: 10.3389/fonc.2022.939646] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 12/24/2022] Open
Abstract
BackgroundLaryngeal cancer is a type of head and neck tumor with a poor prognosis and survival rate. The new cases of laryngeal cancer increased rapidly with a higher mortality rate around the world.ObjectiveThe current research work was focused to unveil the in vitro antitumor effects of ononin against the laryngeal cancer Hep-2 cells.MethodologyThe cytotoxic effects of ononin against the laryngeal cancer Hep-2 cells and normal HuLa-PC laryngeal cells were studied using an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The intracellular Reactive Oxygen Species (ROS) generation, apoptotic cell death, Mitochondrial Membrane Potential (MMP), and cell adhesion on the 25 and 50 µM ononin-treated Hep-2 cells were detected using respective staining assays. The levels of TBARS and antioxidants were assayed using specific kits. The expressions of c-Jun N-terminal kinase 1/2 (JNK1/2), Extracellular Signal-regulated Kinase 1/2 (ERK1/2), p38, Phosphatidylinositol-3 Kinase 1/2 (PI3K1/2), and protein kinase-B (Akt) in the ononin-treated Hep-2 cells were investigated using Reverse Transcription-Polymerase Chain Reaction (RT-PCR) assay.ResultsThe ononin treatment effectively inhibited the Hep-2 cell viability but did not affect the viability of HuLa-PC cells. Furthermore, the ononin treatment effectively improved the intracellular ROS accumulation, depleted the MMP, and triggered apoptosis in Hep-2 cells. The Thiobarbituric acid reactive substances (TBARS) were improved, and Glutathione (GSH) levels and Superoxide dismutase (SOD) were depleted in the ononin-administered Hep-2 cells. The ononin treatment substantially inhibited the JNK/ERK/p38 axis in the Hep-2 cells.ConclusionTogether, the outcomes of this exploration proved that the ononin has remarkable antitumor activity against laryngeal cancer Hep-2 cells.
Collapse
Affiliation(s)
- Ben Ye
- Department of Ear, Nose, and Throat (ENT), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji’nan, China
| | - Jianhua Ma
- Department of Cardiology, Shandong Rongjun General Hospital, Ji’nan, China
| | - Zhaoxia Li
- Department of Ear, Nose, and Throat (ENT), Shandong Provincial Hospital Affiliated to Shandong First Medical University, Ji’nan, China
| | - Yang Li
- Department of Plastic Surgery, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaopan Han
- Department of ENT, Central Hospital Affiliated to Shandong First Medical University, Ji’nan, China
- *Correspondence: Xiaopan Han,
| |
Collapse
|