1
|
Hu J, Dai S, Yuan M, Li F, Xu S, Gao L. Isoliensinine suppressed gastric cancer cell proliferation and migration by targeting TGFBR1 to regulate TGF-β-smad signaling pathways. Front Pharmacol 2024; 15:1438161. [PMID: 39364054 PMCID: PMC11446791 DOI: 10.3389/fphar.2024.1438161] [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: 06/04/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024] Open
Abstract
Background Gastric cancer (GC) ranks as the fifth most prevalent cancer globally, and its pronounced invasiveness and propensity to spread provide significant challenges for therapy. At present, there are no efficacious medications available for the treatment of patients with GC. Isoliensinine (ISO), a bisbenzylisoquinoline alkaloid, was isolated from Nelumbo nucifera Gaertn. It possesses anti-tumor, antioxidant, and other physiological effects. Nevertheless, there is currently no available study on the impact of ISO on GC, and further investigation is needed to understand its molecular mechanism. Methods ISO target points and GC-related genes were identified, and the cross-target points of ISO and GC were obtained. We then examined cross-targeting and found genes that were differentially expressed in GCs. Kaplan-Meier survival curves were used to screen target genes, and the STRING database and Cytoscape 3.9.1 were used to construct protein-protein interactions and drug-target networks. In addition, molecular docking studies confirmed the interactions between ISO screen targets. Finally, in vitro tests were used to establish the impact of ISO on GC cells. Results Through bioinformatics research, we have identified TGFBR1 as the target of ISO in GC. In addition, we noticed a substantial inhibition in GC cell proliferation, migration, and invasion activities following ISO treatment. Moreover, we noticed that ISO treatment effectively suppressed TGF-β-induced epithelial-mesenchymal transition (EMT) and activation of the TGF-β-Smad pathway. Furthermore, we discovered that siTGFBR1 nullified the impact of ISO on TGF-β-triggered migration, invasion, and activation of the TGF-β-Smad pathway. Conclusion Our research suggests that ISO specifically targets TGFBR1 and regulates the TGF-β-Smad signaling pathway to suppress the proliferation and migration of GC cells.
Collapse
Affiliation(s)
- Jinda Hu
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Shangming Dai
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Mengqin Yuan
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Fengjiao Li
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Shuoguo Xu
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Lichen Gao
- Department of Pharmacy, School of Pharmacy, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
2
|
Li Q, Chen S, Wang X, Cai J, Huang H, Tang S, He D. Cisplatin-Based Combination Therapy for Enhanced Cancer Treatment. Curr Drug Targets 2024; 25:473-491. [PMID: 38591210 DOI: 10.2174/0113894501294182240401060343] [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: 11/22/2023] [Revised: 03/04/2024] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Cisplatin, a primary chemotherapeutic drug, is of great value in the realm of tumor treatment. However, its clinical efficacy is strictly hindered by issues, such as drug resistance, relapse, poor prognosis, and toxicity to normal tissue. Cisplatin-based combination therapy has garnered increasing attention in both preclinical and clinical cancer research for its ability to overcome resistance, reduce toxicity, and enhance anticancer effects. This review examines three primary co-administration strategies of cisplatin-based drug combinations and their respective advantages and disadvantages. Additionally, seven types of combination therapies involving cisplatin are discussed, focusing on their main therapeutic effects, mechanisms in preclinical research, and clinical applications. This review also discusses future prospects and challenges, aiming to offer guidance for the development of optimal cisplatin-based combination therapy regimens for improved cancer treatment.
Collapse
Affiliation(s)
- Qi Li
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Siwei Chen
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Xiao Wang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Jia Cai
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Hongwu Huang
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| | - Shengsong Tang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, Hunan University of Medicine, Huaihua, China
| | - Dongxiu He
- Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
3
|
Chen K, Beeraka NM, Zhang X, Sinelnikov MY, Plotnikova M, Zhao C, Basavaraj V, Zhang J, Lu P. Recent Advances in Therapeutic Modalities Against Breast Cancer-Related Lymphedema: Future Epigenetic Landscape. Lymphat Res Biol 2023; 21:536-548. [PMID: 37267206 PMCID: PMC10753987 DOI: 10.1089/lrb.2022.0016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023] Open
Abstract
Background: Lymphedema is a significant postsurgical complication observed in the majority of breast cancer patients. These multifactorial etiopathogenesis have a significant role in the development of novel diagnostic/prognostic biomarkers and the development of novel therapies. This review aims to ascertain the epigenetic alterations that lead to breast cancer-related lymphedema (BCRL), multiple pathobiological events, and the underlying genetic predisposing factors, signaling cascades pertinent to the lapses in effective prognosis/diagnosis, and finally to develop a suitable therapeutic regimen. Methods and Results: We have performed a literature search in public databases such as PubMed, Medline, Google Scholar, National Library of Medicine and screened several published reports. Search words such as epigenetics to induce BCRL, prognosis/diagnosis, primary lymphedema, secondary lymphedema, genetic predisposing factors for BRCL, conventional therapies, and surgery were used in these databases. This review described several epigenetic-based predisposing factors and the pathophysiological consequences of BCRL, which affect the overall quality of life, and the interplay of these events could foster the progression of lymphedema in breast cancer survivors. Prognosis/diagnostic and therapy lapses for treating BCRL are highly challenging due to genetic and anatomical variations, alteration in the lymphatic vessel contractions, and variable expression of several factors such as vascular endothelial growth factor (VEGF)-E and vascular endothelial growth factor receptor (VEGFR) in breast cancer survivors. Conclusion: We compared the efficacy of various conventional therapies for treating BCRL as a multidisciplinary approach. Further substantial research is required to decipher underlying signaling epigenetic pathways to develop chromatin-modifying therapies pertinent to the multiple etiopathogenesis to explore the correlation between the disease pathophysiology and novel therapeutic modalities to treat BCRL.
Collapse
Affiliation(s)
- Kuo Chen
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Narasimha M. Beeraka
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Andhra Pradesh, India
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Xinliang Zhang
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Mikhail Y. Sinelnikov
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Maria Plotnikova
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Cuiping Zhao
- The 80th Army Hospital of the Chinese People's Liberation Army, Weifang, China
| | - Vijaya Basavaraj
- Department of Pathology, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Jin Zhang
- I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Pengwei Lu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Dasari S, Pathak N, Thomas A, Bitla S, Kumar R, Munirathinam G. Neferine Targets the Oncogenic Characteristics of Androgen-Dependent Prostate Cancer Cells via Inducing Reactive Oxygen Species. Int J Mol Sci 2023; 24:14242. [PMID: 37762540 PMCID: PMC10532349 DOI: 10.3390/ijms241814242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Castration resistance poses a significant challenge in the management of advanced prostate cancer (PCa), with androgen deprivation therapy (ADT) or chemotherapy being the primary treatment options. However, these approaches often lead to significant side effects and the development of therapeutic resistance. Therefore, it is crucial to explore novel treatment options that can efficiently target PCa, improve patient survival, and enhance their quality of life. Neferine (Nef), a bioactive compound derived from plants, has emerged as a promising candidate for cancer treatment due to its ability to induce apoptosis, autophagy, and cell cycle arrest. In this study, we investigated the potential anticancer effects of Nef in androgen receptor (AR)-positive LNCaP and VCaP cells, representative models of androgen-dependent PCa. Our findings demonstrate that Nef effectively inhibits cell growth, proliferation, and the tumorigenic potential of androgen-dependent PCa cells. Furthermore, Nef treatment resulted in the excessive production of reactive oxygen species (ROS), leading to the activation of key markers of autophagy and apoptosis. These results suggest that Nef has the potential to target the oncogenic characteristics of androgen-dependent PCa cells by exploiting the potency of ROS and inducing autophagy and apoptosis in AR-positive PCa cells. These findings shed light on the therapeutic potential of Nef as a novel treatment option with reduced side effects for androgen-dependent prostate cancer. Further investigations are warranted to assess its efficacy and safety in preclinical and clinical settings.
Collapse
Affiliation(s)
- Subramanyam Dasari
- School of Medicine, Indiana University Bloomington, Bloomington, IN 47405, USA;
| | - Nishtha Pathak
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL 61108, USA; (N.P.); (A.T.); (S.B.)
| | - Amy Thomas
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL 61108, USA; (N.P.); (A.T.); (S.B.)
| | - Shreeja Bitla
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL 61108, USA; (N.P.); (A.T.); (S.B.)
| | - Raj Kumar
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Waknaghat, Solan 173234, Himachal Pradesh, India;
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, IL 61108, USA; (N.P.); (A.T.); (S.B.)
| |
Collapse
|
5
|
Jiang H, Zhu S, Wu B, Su Y, Wang Q, Lei Y, Shao Q, Gao Y, Gao K, Wu G. CDK2 and CDK4 targeted liensinine inhibits the growth of bladder cancer T24 cells. Chem Biol Interact 2023; 382:110624. [PMID: 37423554 DOI: 10.1016/j.cbi.2023.110624] [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: 05/28/2023] [Revised: 07/02/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023]
Abstract
Bladder cancer (BCa) is a urinary tumor with limited treatment options and high mortality. Liensinine (LIEN), a natural bisbenzylisoquinoline alkaloid, has shown excellent anti-tumor effects in numerous preclinical studies. However, the anti-BCa effect of LIEN remains unclear. To the best of our knowledge, this is the first study to investigate the molecular mechanism of LIEN in the management of BCa. First, we identified the treatment-related targets of BCa; those that repeatedly occur in more than two databases, including GeneCards, Online Mendelian Inheritance in Man, DisGeNET, Therapeutic Target Database, and Drugbank. The SwissTarget database was used to screen LIEN-related targets, and those with a probability >0 were possible LIEN targets. The prospective targets of LIEN in the treatment of BCa were then determined using a Venn diagram. Second, we discovered that the PI3K/AKT pathway and senescence mediated the anti-BCa action of LIEN by using GO and KEGG enrichment analysis to explore the function of LIEN therapeutic targets. A protein-protein interaction network was created using the String website, and six algorithms of the CytoHubba plug-in were then used in Cytoscape to assess the core targets of LIEN for the therapy of BCa. The outcomes of molecular docking and dynamics simulation demonstrated that CDK2 and CDK4 proteins were the direct targets of LIEN in the management of BCa, among which CDK2 was more stable in binding to LIEN than CDK4. Finally, in vitro experiments showed that LIEN inhibited the activity and proliferation of T24 cells. The expression of p-/AKT, CDK2, and CDK4 proteins progressively decreased, while the expression and fluorescence intensity of the senescence-related protein, γH2AX, gradually increased with increasing LIEN concentration in T24 cells. Therefore, our data suggest that LIEN may promote senescence and inhibit proliferation by inhibiting the CDK2/4 and PI3K/AKT pathways in BCa.
Collapse
Affiliation(s)
- Hanbing Jiang
- Department of Radiation Oncology, Tangdu Hospital, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Siying Zhu
- Department of Radiation Oncology, Tangdu Hospital, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Bin Wu
- Department of Urology, Xi'an People's Hospital(Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Yinyin Su
- Department of Emergency, Tangdu Hospital, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Qiming Wang
- Department of Radiation Oncology, Tangdu Hospital, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Yonghua Lei
- Department of Urology, Xi'an People's Hospital(Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China
| | - Qiuju Shao
- Department of Radiation Oncology, Tangdu Hospital, The Second Affiliated Hospital of Air Force Military Medical University, Xi'an, 710038, China
| | - Yun Gao
- Department of Neurosurgery, 521 Hospital of Norinco Group, Xi'an, Shaanxi, 710065, China
| | - Ke Gao
- Department of Urology, Xi'an People's Hospital(Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China.
| | - Guojun Wu
- Department of Urology, Xi'an People's Hospital(Xi'an Fourth Hospital), School of Life Sciences and Medicine, Northwest University, Xi'an, 710199, China.
| |
Collapse
|
6
|
Yang J, Zhang Q, Huang G, Cong J, Wang T, Zhai X, Zhang J, Qi G, Zhou L, Jin J. Combined effects of vitamin D and neferine on the progression and metastasis of colorectal cancer. J Cancer Res Clin Oncol 2023; 149:6203-6210. [PMID: 36697773 PMCID: PMC10356635 DOI: 10.1007/s00432-022-04552-7] [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: 11/19/2022] [Accepted: 12/21/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE To investigate the synergistic effect of vitamin D and neferine on the growth and metastasis of colorectal cancer (CRC). METHODS The synergistic effect of biologically active form of vitamin D, VD3 and neferine on the treatment of CRC was investigated by bliss analysis. Colony formation and wound healing ability, migration and invasion ability, and epithelial mesenchymal transition of HCT-116 cells, as a response to the combination treatment with VD3 and neferine were evaluated. RESULTS VD3 and neferine showed a synergistic effect on CRC cell growth at a relatively low dose. The wound healing and colony formation capacity, cell migration and invasion abilities were all decreased by combination use of VD3 and neferine, compared to the VD3 or neferine treated single group. Furthermore, VD3 and neferine significantly decreased the expressions of N-cadherin, vimentin, snail, and slug in HCT-116 cells. CONCLUSION These data suggest that neferine enhances the anticancer capability of VD3 and reduces the dose dependency of VD3. The combination of vitamin D with neferine appears to be a potential therapeutic strategy for CRC.
Collapse
Affiliation(s)
- Jinfeng Yang
- School of Pharmacy, Guilin Medical University, Guilin, 541199 China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Department of Immunology, Guilin Medical University, Guilin, 541199 China
| | - Qinyu Zhang
- School of Pharmacy, Guilin Medical University, Guilin, 541199 China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guilin, 541000 China
| | - Guanlin Huang
- School of Pharmacy, Guilin Medical University, Guilin, 541199 China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
| | - Jiacheng Cong
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Department of Immunology, Guilin Medical University, Guilin, 541199 China
| | - Ting Wang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199 China
| | - Xiaoya Zhai
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199 China
| | - Juzheng Zhang
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199 China
| | - Guangying Qi
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199 China
| | - Lihua Zhou
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
| | - Jiamin Jin
- School of Pharmacy, Guilin Medical University, Guilin, 541199 China
- Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guilin Medical University, Guilin, 541199 China
- Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Guilin, 541199 China
| |
Collapse
|
7
|
Nelson VK, Nuli MV, Mastanaiah J, Saleem T. S. M, Birudala G, Jamous YF, Alshargi O, Kotha KK, Sudhan HH, Mani RR, Muthumanickam A, Niranjan D, Jain NK, Agrawal A, Jadon AS, Mayasa V, Jha NK, Kolesarova A, Slama P, Roychoudhury S. Reactive oxygen species mediated apoptotic death of colon cancer cells: therapeutic potential of plant derived alkaloids. Front Endocrinol (Lausanne) 2023; 14:1201198. [PMID: 37560308 PMCID: PMC10408138 DOI: 10.3389/fendo.2023.1201198] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/28/2023] [Indexed: 08/11/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most deaths causing diseases worldwide. Several risk factors including hormones like insulin and insulin like growth factors (e.g., IGF-1) have been considered responsible for growth and progression of colon cancer. Though there is a huge advancement in the available screening as well as treatment techniques for CRC. There is no significant decrease in the mortality of cancer patients. Moreover, the current treatment approaches for CRC are associated with serious challenges like drug resistance and cancer re-growth. Given the severity of the disease, there is an urgent need for novel therapeutic agents with ideal characteristics. Several pieces of evidence suggested that natural products, specifically medicinal plants, and derived phytochemicals may serve as potential sources for novel drug discovery for various diseases including cancer. On the other hand, cancer cells like colon cancer require a high basal level of reactive oxygen species (ROS) to maintain its own cellular functions. However, excess production of intracellular ROS leads to cancer cell death via disturbing cellular redox homeostasis. Therefore, medicinal plants and derived phytocompounds that can enhance the intracellular ROS and induce apoptotic cell death in cancer cells via modulating various molecular targets including IGF-1 could be potential therapeutic agents. Alkaloids form a major class of such phytoconstituents that can play a key role in cancer prevention. Moreover, several preclinical and clinical studies have also evidenced that these compounds show potent anti-colon cancer effects and exhibit negligible toxicity towards the normal cells. Hence, the present evidence-based study aimed to provide an update on various alkaloids that have been reported to induce ROS-mediated apoptosis in colon cancer cells via targeting various cellular components including hormones and growth factors, which play a role in metastasis, angiogenesis, proliferation, and invasion. This study also provides an individual account on each such alkaloid that underwent clinical trials either alone or in combination with other clinical drugs. In addition, various classes of phytochemicals that induce ROS-mediated cell death in different kinds of cancers including colon cancer are discussed.
Collapse
Affiliation(s)
- Vinod K. Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Juturu Mastanaiah
- Department of Pharmacology, Balaji College of Pharmacy, Anantapur, India
| | | | - Geetha Birudala
- Faculty of Pharmacy, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Yahya F. Jamous
- Vaccines and Bioprocessing Centre, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Omar Alshargi
- College of Pharmacy, Riyadh ELM University, Riyadh, Saudi Arabia
| | - Kranthi Kumar Kotha
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, India
| | - Hari Hara Sudhan
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | | | | | | | | | | | | | - Vinyas Mayasa
- GITAM School of Pharmacy, GITAM University Hyderabad Campus, Rudraram, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| | - Adriana Kolesarova
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czechia
| | | |
Collapse
|
8
|
Moldovan OL, Sandulea A, Lungu IA, Gâz ȘA, Rusu A. Identification of Some Glutamic Acid Derivatives with Biological Potential by Computational Methods. Molecules 2023; 28:molecules28104123. [PMID: 37241864 DOI: 10.3390/molecules28104123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/07/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Glutamic acid is a non-essential amino acid involved in multiple metabolic pathways. Of high importance is its relationship with glutamine, an essential fuel for cancer cell development. Compounds that can modify glutamine or glutamic acid behaviour in cancer cells have resulted in attractive anticancer therapeutic alternatives. Based on this idea, we theoretically formulated 123 glutamic acid derivatives using Biovia Draw. Suitable candidates for our research were selected among them. For this, online platforms and programs were used to describe specific properties and their behaviour in the human organism. Nine compounds proved to have suitable or easy to optimise properties. The selected compounds showed cytotoxicity against breast adenocarcinoma, lung cancer cell lines, colon carcinoma, and T cells from acute leukaemia. Compound 2Ba5 exhibited the lowest toxicity, and derivative 4Db6 exhibited the most intense bioactivity. Molecular docking studies were also performed. The binding site of the 4Db6 compound in the glutamine synthetase structure was determined, with the D subunit and cluster 1 being the most promising. In conclusion, glutamic acid is an amino acid that can be manipulated very easily. Therefore, molecules derived from its structure have great potential to become innovative drugs, and further research on these will be conducted.
Collapse
Affiliation(s)
- Octavia-Laura Moldovan
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Alexandra Sandulea
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Ioana-Andreea Lungu
- Medicine and Pharmacy Doctoral School, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Șerban Andrei Gâz
- Organic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| |
Collapse
|
9
|
Sivaselvam S, Mohankumar A, Narmadha R, Selvakumar R, Sundararaj P, Viswanathan C, Ponpandian N. Effect of gamma-ray irradiated reduced graphene oxide (rGO) on environmental health: An in-vitro and in-vivo studies. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 318:120933. [PMID: 36565492 DOI: 10.1016/j.envpol.2022.120933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/19/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
The unique properties of reduced graphene oxide (rGO) have drawn the attention of scientists worldwide since the last decade and it is explored for a wide range of applications. However, the rapid expansion of rGO use in various products will eventually lead to environenal exposure and rises a safety concern on the environment and humal health risk. Moreover, the utilization of toxic chemicals for the reduction of graphene oxide (GO) into rGO is not environmentally friendly, warranting the exploration of non-toxic approaches. In the present work, rGO was synthesized using a different dose of gamma-ray irradiation and characterized. The in-vitro and in-vivo analysis indicated that the gamma-irradiated rGO induced toxicity depending on its degree of reduction and dosage. In the L929 cells, rGO-30 KGy significantly induced cytotoxicity even at low concentration (1 mg L-1) by inducing reactive oxygen species (ROS), lactate dehydrogenase (LDH) enzyme production, nuclear fragmentation and apoptosis. The change in morphology of the cells like membrane blebbing and cell rounding was also observed via FESEM. In the in-vivo model Caenorhabditis elegans, rGO-30 KGy significantly affected the functioning of primary and secondary targeted organs and also negatively influenced the nuclear accumulation of transcription factors (DAF-16/FOXO and SKN-1/Nrf2), neuronal health, and antioxidant defense mechanism of the nematodes. The real-time PCR analysis showed significant up-regulation (ced-3, ced-4, cep-1, egl-1, and hus-1) and down-regulation (ced-9) of the gene involved in germ-line and DNA damage-induced apoptosis. The detailed toxicity mechanism of gamma irradiated rGO has been elucidated. This work highlights the toxicity of rGO prepared by gamma-ray radiation and paves way for understating the toxicity mechanism.
Collapse
Affiliation(s)
- S Sivaselvam
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, 641 046, India
| | - A Mohankumar
- Department of Zoology, Bharathiar University, Coimbatore, 641 046, India
| | - R Narmadha
- Nanobiotechnology Laboratory, Department of Nanobiotechnology, PSG Institute of Advanced Studies, Peelamedu, Coimbatore, 641 004, India
| | - R Selvakumar
- Nanobiotechnology Laboratory, Department of Nanobiotechnology, PSG Institute of Advanced Studies, Peelamedu, Coimbatore, 641 004, India
| | - P Sundararaj
- Department of Zoology, Bharathiar University, Coimbatore, 641 046, India
| | - C Viswanathan
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, 641 046, India
| | - N Ponpandian
- Department of Nanoscience and Technology, Bharathiar University, Coimbatore, 641 046, India.
| |
Collapse
|
10
|
Beeraka NM, Zhang J, Zhao D, Liu J, A U C, Vikram Pr H, Shivaprakash P, Bannimath N, Manogaran P, Sinelnikov MY, Bannimath G, Fan R. Combinatorial Implications of Nrf2 Inhibitors with FN3K Inhibitor: In vitro Breast Cancer Study. Curr Pharm Des 2023; 29:2408-2425. [PMID: 37861038 DOI: 10.2174/0113816128261466231011114600] [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: 05/18/2023] [Revised: 08/07/2023] [Accepted: 08/14/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Platinum derivatives are chemotherapeutic agents preferred for the treatment of cancers including breast cancer. Oxaliplatin is an anticancer drug that is in phase II studies to treat metastatic breast cancer. However, its usage is constrained by chemoresistance and dose-related side effects. OBJECTIVE The objective of this study is to examine the combinatorial efficacy of brusatol, an Nrf2 blocker, with oxaliplatin (a proven FN3K blocker in our study) in mitigating breast cancer growth in vitro. METHODS We performed cytotoxicity assays, combination index (CI) analysis, colony formation assays, apoptosis assays, and Western blotting. RESULTS Results of our study described the chemosensitizing efficacy of brusatol in combination with lowdose oxaliplatin against breast cancer through synergistic effects in both BT-474 and T47D cells. A significant mitigation in the migration rate of these cancer cells was observed with the combination regimen, which is equivalent to the IC-50 dose of oxaliplatin (125 μM). Furthermore, ROS-mediated and apoptotic modes of cell death were observed with a combinatorial regimen. Colony formation of breast cancer cell lines was mitigated with a combinatorial regimen of bursatol and oxaliplatin than the individual treatment regimen. FN3K expression downregulated with oxaliplatin in T47D cells. The mitigation of FN3K protein expression with a combination regimen was not observed but the Nrf2 downstream antioxidant signaling proteins were significantly downregulated with a combination regimen similar to individual drug regimens. CONCLUSION Our study concluded the combination efficacy of phytochemicals like brusatol in combination with low-dose oxaliplatin (FN3K blocker), which could enhance the chemosensitizing effect in breast cancer and minimize the overall dose requirement of oxaliplatin.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
- Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119991, Russia
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | - Di Zhao
- Department of Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Junqi Liu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
| | - Chinnappa A U
- Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Hemanth Vikram Pr
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
- Xenone Healthcare Pvt. Ltd, #318, Third Floor, US Complex, Jasola, New Delhi 110076, India
| | - Priyanka Shivaprakash
- Faculty of Life Sciences, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Namitha Bannimath
- Department of Pharmacology and Toxicology, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Prasath Manogaran
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
| | - Mikhail Y Sinelnikov
- Sechenov First Moscow State Medical University, 8-2 Trubetskaya St., Moscow 119991, Russia
- Sinelab Biomedical Research Center, Minnesota 55905, USA
| | - Gurupadayya Bannimath
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Ruitai Fan
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou 450052, China
| |
Collapse
|
11
|
Apigenin inhibits migration and induces apoptosis of human endometrial carcinoma Ishikawa cells via PI3K-AKT-GSK-3β pathway and endoplasmic reticulum stress. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
12
|
Neferine Exerts Ferroptosis-Inducing Effect and Antitumor Effect on Thyroid Cancer through Nrf2/HO-1/NQO1 Inhibition. JOURNAL OF ONCOLOGY 2022; 2022:7933775. [PMID: 35794985 PMCID: PMC9252705 DOI: 10.1155/2022/7933775] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 05/30/2022] [Accepted: 06/02/2022] [Indexed: 12/17/2022]
Abstract
Thyroid cancer is the most prevalent endocrine malignancy with an increasing incidence in the past few decades. Neferine possesses various pharmacological activities, which have been applied in diverse disease models, including various tumors. However, the detailed effect and mechanism of neferine on thyroid cancer are still unclear. In the current study, the viability of IHH-4 and CAL-62 cells was examined by the CCK-8 assay. The effect of neferine on the proliferation, apoptosis, invasion, vascular endothelial growth factor (VEGF), epithelial-mesenchymal transition (EMT), and ferroptosis was evaluated by CCK-8, flow cytometry, western blot, and spectrophotometry assays. Mechanically, the expressions levels of Nrf2/HO-1/NQO1 signaling were first determined by a western blot, which was then verified by Nrf2 overexpression. In vivo validation was also conducted on BALB/c nude mice with an inoculation dose of 2 × 106 IHH-4 cells. The results showed that neferine repressed the viability of both IHH-4 and CAL-62 cells both in a dose-dependent way and in a time-dependent fashion, in which the IC50 value of neferine on IHH-4 and CAL-62 cells was 9.47 and 8.72 μM, respectively. Besides, neferine enhanced apoptosis but suppressed invasion, angiogenesis, and EMT of IHH-4 and CAL-62 cells. Moreover, neferine induced the activation of ferroptosis in thyroid cancer cells. Notably, it was revealed that the Nrf2/HO-1/NQO1 pathway was strongly associated with the effect of neferine on the modulation of thyroid cancer. Furthermore, these outcomes were validated in xenografted mice. Therefore, neferine exerted an antitumor effect and ferroptosis-inducing effect on thyroid cancer via inhibiting the Nrf2/HO-1/NQO1 pathway.
Collapse
|
13
|
Chen K, Zhang J, Beeraka NM, Tang C, Babayeva YV, Sinelnikov MY, Zhang X, Zhang J, Liu J, Reshetov IV, Sukocheva OA, Lu P, Fan R. Advances in the Prevention and Treatment of Obesity-Driven Effects in Breast Cancers. Front Oncol 2022; 12:820968. [PMID: 35814391 PMCID: PMC9258420 DOI: 10.3389/fonc.2022.820968] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/16/2022] [Indexed: 12/11/2022] Open
Abstract
Obesity and associated chronic inflammation were shown to facilitate breast cancer (BC) growth and metastasis. Leptin, adiponectin, estrogen, and several pro-inflammatory cytokines are involved in the development of obesity-driven BC through the activation of multiple oncogenic and pro-inflammatory pathways. The aim of this study was to assess the reported mechanisms of obesity-induced breast carcinogenesis and effectiveness of conventional and complementary BC therapies. We screened published original articles, reviews, and meta-analyses that addressed the involvement of obesity-related signaling mechanisms in BC development, BC treatment/prevention approaches, and posttreatment complications. PubMed, Medline, eMedicine, National Library of Medicine (NLM), and ReleMed databases were used to retrieve relevant studies using a set of keywords, including "obesity," "oncogenic signaling pathways," "inflammation," "surgery," "radiotherapy," "conventional therapies," and "diet." Multiple studies indicated that effective BC treatment requires the involvement of diet- and exercise-based approaches in obese postmenopausal women. Furthermore, active lifestyle and diet-related interventions improved the patients' overall quality of life and minimized adverse side effects after traditional BC treatment, including postsurgical lymphedema, post-chemo nausea, vomiting, and fatigue. Further investigation of beneficial effects of diet and physical activity may help improve obesity-linked cancer therapies.
Collapse
Affiliation(s)
- Kuo Chen
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Zhang
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Narasimha M. Beeraka
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical College, Mysuru, India
| | - Chengyun Tang
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Yulia V. Babayeva
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Mikhail Y. Sinelnikov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Xinliang Zhang
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Jiacheng Zhang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junqi Liu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Igor V. Reshetov
- Department of Human Anatomy, I. M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Olga A. Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Adelaide, SA, Australia
| | - Pengwei Lu
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruitai Fan
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Retrospective analysis: 5509 cases of "totally implantable venous access port systems implantation (TIVAPS) depth" assisted by digital radiography. Langenbecks Arch Surg 2022; 407:3123-3132. [PMID: 35660962 DOI: 10.1007/s00423-022-02573-x] [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: 09/11/2021] [Accepted: 05/26/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Modern oncological treatment in breast cancer patients requires the precise delivery of chemotherapy infusion into the central venous systems without toxicity. TIVAPS is the significant method of chemotherapy delivery although certain internal or external complications associated with their placement. However, the long-term use of TIVAPS is still a concern to minimize the complications such as venous thrombosis syndrome (VTS) and cardiac defects. The aim of this study is to investigate the potential disadvantages that may be avoided by digital radiography (DR)-assisted measurement of catheter depth pertinent to TIVAPS implanted system. METHODS Retrospective analysis related to 5509 TIVAPS recipients of 99% female breast cancer patients and 1% male blood disorder patients registered from April 2013 to November 2017 were included in the study. Patients with TIVAPS catheter tip depth into superior vena cava into upper (group A), middle (group B), and lower (group C) parts were stratified for evaluation during implantation; DR-assisted measurement of TIVAPS was performed to decipher "tip depth of catheter" and determined the relevance of tip depth to complications such as VTS and cardiac defects. RESULTS Incidence of VTS complications were significantly higher in TIVAPS recipients of group A (82.7%) than group B (16%) and group C (0.12%) in which the "tip depth of TIVAPS was deeper" (P < 0.01). Defects in heart function are higher in group C (59.6%) than group A (15.8%) and group B (24.6%) in which the "tip depth of TIVAPS was deeper" (P < 0.01). CONCLUSION DR-assisted measurement can more accurately determine the depth of TIVAPS catheter implantation, and avoid the incidence of related complications, and provide a better method for surgeons.
Collapse
|
15
|
Mello ALDN, Zancan P. Isoquinolines alkaloids and cancer metabolism: Pathways and targets to novel chemotherapy. Chem Biol Drug Des 2022; 99:944-956. [PMID: 35322534 DOI: 10.1111/cbdd.14043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/15/2022] [Accepted: 03/20/2022] [Indexed: 12/24/2022]
Abstract
Cancer is one of the main causes of death in the world. This is a complex disease where the development of resistance to chemotherapy is frequent driving the search for new anticancer compounds. In this sense, isoquinolines have gained attention in the past few years. This review aims to highlight the new advances related to the use of isoquinolines compounds against cancer cells, and we point out targets for their anti-tumor action. Isoquinolines are compounds found in plants that are important for their protection. In cancer, many representatives of this class of compounds have demonstrated their efficacy against cancer by acting on cancer metabolism, such as triggering cell death, reducing pro-survival protein expression, inducing ROS production, inhibiting pro-survival cell signaling pathways, among other effects. The mechanisms triggered by isoquinolines in cancer cells represent robust anticancer strategies, which support that this class of compounds are strong candidates for cancer treatment.
Collapse
Affiliation(s)
- Angélica Lauria do Nascimento Mello
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Zancan
- Laboratório de Oncobiologia Molecular (LabOMol), Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Synergistic effects of natural compounds and conventional chemotherapeutic agents: recent insights for the development of cancer treatment strategies. Heliyon 2022; 8:e09519. [PMID: 35669542 PMCID: PMC9163513 DOI: 10.1016/j.heliyon.2022.e09519] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/06/2022] [Accepted: 05/17/2022] [Indexed: 01/02/2023] Open
Abstract
Cancer is one of the leading causes of death in the world. Chemotherapy is presented as an option for treatment of this disease, however, low specificity, high resistance rates, toxicity and hypersensitivity reactions, make it necessary to search for therapeutic alternatives that increase the selectivity of treatment, reduce the side effects and enhance its antitumor potential. Natural products are accessible, inexpensive and less toxic sources; in addition, they have multiple mechanisms of action that can potentiate the outcome of chemotherapeutics. In this review, we present evidence on the beneficial effect of the interaction of dietary phytochemicals with chemotherapeutical agents for cancer treatment. This effect is generated by different mechanisms of action such as, increased tumoricidal effect via sensitization of cancer cells, reversing chemoresistance through inhibition of several targets involved in the development of drug resistance and, decreasing chemotherapy-induced toxicity in non-tumoral cells by the promotion of repair mechanisms. Studies discussed in this review will provide a solid basis for the exploration of the potential use of natural products in combination with chemotherapeutical agents, to overcome some of the difficulties that arise in the management of cancer patients.
Collapse
|
17
|
Lim HM, Park SH. Regulation of reactive oxygen species by phytochemicals for the management of cancer and diabetes. Crit Rev Food Sci Nutr 2022; 63:5911-5936. [PMID: 34996316 DOI: 10.1080/10408398.2022.2025574] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer and diabetes mellitus are served as typical life-threatening diseases with common risk factors. Developing therapeutic measures in cancers and diabetes have aroused attention for a long time. However, the problems with conventional treatments are in challenge, including side effects, economic burdens, and patient compliance. It is essential to secure safe and efficient therapeutic methods to overcome these issues. As an alternative method, antioxidant and pro-oxidant properties of phytochemicals from edible plants have come to the fore. Phytochemicals are naturally occurring compounds, considered promising agent applicable in treatment of various diseases with beneficial effects. Either antioxidative or pro-oxidative activity of various phytochemicals were found to contribute to regulation of cell proliferation, differentiation, cell cycle arrest, and apoptosis, which can exert preventive and therapeutic effects against cancer and diabetes. In this article, the antioxidant or pro-oxidant effects and underlying mechanisms of flavonoids, alkaloids, and saponins in cancer or diabetic models demonstrated by the recent studies are summarized.
Collapse
Affiliation(s)
- Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| |
Collapse
|
18
|
Li HL, Cheng Y, Zhou ZW, Long HZ, Luo HY, Wen DD, Cheng L, Gao LC. Isoliensinine induces cervical cancer cell cycle arrest and apoptosis by inhibiting the AKT/GSK3α pathway. Oncol Lett 2021; 23:8. [PMID: 34820007 PMCID: PMC8607237 DOI: 10.3892/ol.2021.13126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/20/2021] [Indexed: 12/26/2022] Open
Abstract
Isoliensinine is a bis-benzylisoquinoline alkaloid that can be isolated from the lotus Nelumbo nucifera Gaertn. It has been reported to exert a variety of anti-cancer properties. In the present study, the potential effects of isoliensinine on cervical cancer Siha, HeLa, Caski and C33A cell lines were investigated by using Cell Counting Kit-8 (CCK-8), flow cytometry, western blotting and reverse transcription-PCR (RT-PCR) to measure cell proliferation, the cell cycle and apoptosis, in addition to elucidating the underlying molecular mechanism. Protein levels of p21, CDK2, Cyclin E, Mcl-1, cleaved Caspase-9, AKT, phosphorylated-AKT, glycogen synthase kinase (Gsk)3α, PTEN, and mRNA levels of p21, p15, p27, CDK2, CDK4, Cyclin E, Cyclin D, Gsk3α, Gsk3β and PTEN were measured. Molecular docking assays were used to calculate the strength of binding of isoliensinine to AKT using AutoDock 4.0. Isoliensinine was found to induce cell cycle arrest at the G0/G1 phase by upregulating p21 expression and downregulating CDK2 and cyclin E in cervical cancer cells. In addition, in previous research, isoliensinine promoted cell apoptosis by downregulating myeloid-cell leukemia 1 expression and activating caspase-9. Upstream, isoliensinine significantly downregulated AKT (S473) phosphorylation and GSK3α expression in a dose- and time-dependent manner. The AKT inhibitor AKTi-1/2 enhanced the function of isoliensinine on cell cycle arrest and apoptosis through the AKT/GSK3α pathway. AutoDock analysis showed that isoliensinine can bind to the AKT protein. These findings suggest that isoliensinine can induce cervical cancer cell cycle arrest and apoptosis by inhibiting the AKT/GSK3α pathway, which represents a novel strategy for the treatment of cervical cancer.
Collapse
Affiliation(s)
- Hong-Li Li
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,School of Life Science, Hunan University of Science and Technology, Xiangtan, Hunan 411201, P.R. China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China
| | - Lin Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, Hunan 410000, P.R. China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research Affiliated to School of Pharmacy, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
19
|
Bharathi Priya L, Huang CY, Hu RM, Balasubramanian B, Baskaran R. An updated review on pharmacological properties of neferine-A bisbenzylisoquinoline alkaloid from Nelumbo nucifera. J Food Biochem 2021; 45:e13986. [PMID: 34779018 DOI: 10.1111/jfbc.13986] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/19/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022]
Abstract
Phytochemicals have recently received a lot of recognition for their pharmacological activities such as anticancer, chemopreventive, and cardioprotective properties. In traditional Indian and Chinese medicine, parts of lotus (Nelumbo nucifera) such as lotus seeds, fruits, stamens, and leaves are used for treating various diseases. Neferine is a bisbenzylisoquinoline alkaloid, a major component from the seed embryos of N. nucifera. Neferine is effective in the treatment of high fevers and hyposomnia, as well as arrhythmia, platelet aggregation, occlusion, and obesity. Neferine has been found to have a variety of therapeutic effects such as anti-inflammatory, anti-oxidant, anti-hypertensive, anti-arrhythmic, anti-platelet, anti-thrombotic, anti-amnesic, and negative inotropic. Neferine also exhibited anti-anxiety effects, anti-cancerous, and chemosensitize to other anticancer drugs like doxorubicin, cisplatin, and taxol. Induction of apoptosis, autophagy, and cell cycle arrest are the key pathways that underlying the anticancer activity of neferine. Therefore, the present review summarizes the neferine biosynthesis, pharmacokinetics, and its effects in myocardium, cancer, chemosensitizing to cancer drug, central nervous system, diabetes, inflammation, and kidney diseases. PRACTICAL APPLICATIONS: Natural phytochemical is gaining medicinal importance for a variety of diseases like including cancer, neurodegenerative disorder, diabetes, and inflammation. Alkaloids and flavonoids, which are abundantly present in Nelumbo nucifera have many therapeutic applications. Neferine, a bisbenzylisoquinoline alkaloid from N. nucifera has many pharmacological properties. This present review was an attempt to compile an updated pharmacological action of neferine in different disease models in vitro and in vivo, as well as to summarize all the collective evidence on the therapeutic potential of neferine.
Collapse
Affiliation(s)
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Centre of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Rouh-Mei Hu
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Rathinasamy Baskaran
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
20
|
Zhang C, Wang X, Wang J, Qiu Y, Qi Z, Song D, Wang M. TCPP-Isoliensinine Nanoparticles for Mild-Temperature Photothermal Therapy. Int J Nanomedicine 2021; 16:6797-6806. [PMID: 34675508 PMCID: PMC8502540 DOI: 10.2147/ijn.s317462] [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: 05/05/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Photothermal therapy (PTT) is promising for the treatment of tumors due to its advantages including minimally invasive, easy implementation and selective localized treatment. However, single PTT suffers from several limitations, such as constrained light penetration and low delivery efficiency, typically leading to heterogeneous heating and incomplete elimination of cancer cells. Therefore, combination of PTT with other therapies, eg, chemotherapy is desirable in order to achieve synergistic effects in cancer treatment. Methods Here, we designed a new type of TCPP-Iso combined nanoparticle for synergetic therapy for breast cancer. Specifically, photothermal agent tetra(4-carboxyphenyl) porphine (TCPP) and anti-cancer drug isoliensinine (Iso) were encapsulated in PEG-b-PLGA polymeric nanoparticles through a precipitation process. Results The obtained NPs displayed well-controlled size and high stability over time. Tuning TCPP-Iso/polymer ratio, or total concentration of drug and polymers led to increased hydrodynamic radius of NPs from 65 to 108 nm without disturbing the narrow size distribution. Besides, the formed NPs showed a consequently cumulative release of TCPP and of Iso. The temperature elevation ability of both TCPP NPs and TCPP-Iso NPs was TCPP-concentration dependent. Solutions of TCPP NPs that contained equivalent amount of TCPP with respect to TCPP-Iso NPs, presented the same trend and exhibited non-obvious difference in temperature elevation under certain laser power. The viability of MDA-MB-231 cells treated with TCPP-Iso NPs could be inhibited effectively at a relatively mild temperature (42–43°C) compared to the other groups, which may minimize heat damage to the surrounding healthy tissues. Conclusion The results indicate that the TCPP-Iso combined NPs showed hardly any toxicity to normal tissue cell line, but displayed an efficient synergistic effect for killing cancer cells under laser irradiation. Our study demonstrates that the successful combination of TCPP and Iso realized a synergistic therapy effect at a relatively mild temperature, and the insights obtained here shall be helpful for designing new combined PTT agents for cancer treatment.
Collapse
Affiliation(s)
- Chenglin Zhang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, People's Republic of China
| | - Xinming Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Junyou Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Yuening Qiu
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Zhiyao Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | - Dianwen Song
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, People's Republic of China
| | - Mingwei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| |
Collapse
|
21
|
Cheng Y, Li HL, Zhou ZW, Long HZ, Luo HY, Wen DD, Cheng L, Gao LC. Isoliensinine: A Natural Compound with "Drug-Like" Potential. Front Pharmacol 2021; 12:630385. [PMID: 33967765 PMCID: PMC8100667 DOI: 10.3389/fphar.2021.630385] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/31/2021] [Indexed: 12/22/2022] Open
Abstract
Isoliensinine, a bisbenzylisoquinoline alkaloid isolated from Nelumbo nucifera Gaertn, exerts a variety of beneficial effects, such as antitumor, cardioprotective, antioxidant, antidepressant, and anti-HIV effects, and ameliorates T2DM with hyperlipidemia and Alzheimer’s disease. In this article, the recent literature on isoliensinine, including its pharmacology, pharmacokinetics, and synthesis and extraction, is summarized. Moreover, possible future prospects and research directions are also discussed. Studies on isoliensinine were found by searching a combination of keywords including “pharmacology,” “pharmacokinetics,” and “synthesis and extraction” in the main databases, including PubMed, Google Scholar, Web of Science, NCBI, and Wan Fang. Many studies have pointed out that a major limitation of isoliensinine is its poor solubility in aqueous media. Considering its advantages and limitations, isoliensinine can be used as a lead compound to develop novel efficient and low-toxicity derivatives. The available literature indicates that isoliensinine displays “drug-like” potential. Additionally, there are many related issues and novel mechanisms that need to be explored.
Collapse
Affiliation(s)
- Yan Cheng
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Li Li
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China
| | - Zi-Wei Zhou
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hui-Zhi Long
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China
| | - Lin Cheng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Li-Chen Gao
- Department of Pharmacy, Phase I Clinical, Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
22
|
Wang J, Hu Y, Yuan J, Zhang Y, Wang Y, Yang Y, Alahmadi TA, Ali Alharbi S, Zhuang Z, Wu F. Chemomodulatory effect of neferine on DMBA-induced squamous cell carcinogenesis: Biochemical and molecular approach. ENVIRONMENTAL TOXICOLOGY 2021; 36:460-471. [PMID: 33156559 DOI: 10.1002/tox.23051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/25/2020] [Accepted: 10/09/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Neferine (NEF) is nontoxic, bisbenzylisoquinoline alkaloid is derived from the seed embryo of lotus, a familiar medicinal plant. Although several mechanisms have been planned, an evident antitumor action pathway of NEF on the oral tumor is still not known. In the current study, we aimed at investigating the protecting effect of NEF against experimental oral carcinoma and clarify its possible mechanism through the induction of apoptosis, proliferation, and inflammatory signaling pathways. METHODS The experimental hamsters were divided into four groups (I-IV) containing six hamsters each. The group I was control group, group II and III hamsters treated with 7,12-dimethylbenz(a)anthracene (DMBA) (0.5%) alone, thrice in a week for 10 weeks, and group III and IV hamsters received oral supplementation of NEF at a concentration of 15 mg/kg bw. All the hamsters were sacrificed after 16 weeks. RESULTS Our results revealed that DMBA treated hamsters exhibited 100% oral tumor cell formation with high-tumor incidence (TI), tumor number (TN), tumor volume (TV), decreased levels of antioxidants, increased status of lipid peroxidation (LPO), and modulated the activities of liver marker agents as well as NF-kB, cell proliferation (PCNA), and p53 proteins. NEF supplementation in DMBA treated hamsters, resulted in delayed lesion synthesis, and brought back the levels of the biochemical parameters. In addition, immunostaining of NF-kB, PCNA, and p53 showed that they were inhibited by NEF. CONCLUSION Thus, NEF might be considered a better chemopreventive drug in an experimental model of home-based primary care (HBPC). More research is necessary to study other pathways implicated in oral carcinomas and their modulation by NEF.
Collapse
Affiliation(s)
- Jingxuan Wang
- The Second Department of Stomatology, Baoding No.1 Central Hospital, Baoding, China
| | - Yan Hu
- Department of Stomatology, The Affiliated Hospital of Hebei University, Hebei, China
| | - Jieying Yuan
- The Second Department of Stomatology, Baoding No.1 Central Hospital, Baoding, China
| | - Yan Zhang
- The Second Department of Stomatology, Baoding No.1 Central Hospital, Baoding, China
| | - Yifan Wang
- Department of Stomatology, The Affiliated Hospital of Hebei University, Hebei, China
| | - Yingshun Yang
- Department of Stomatology, The Affiliated Hospital of Hebei University, Hebei, China
| | - Tahani Awad Alahmadi
- Department of Pediatrics, College of Medicine, King Saud University, [Medical City], King Khalid University Hospital, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Zhizheng Zhuang
- Department of Stomatology, The Affiliated Hospital of Hebei University, Hebei, China
| | - Fan Wu
- Department of Stomatology, The Affiliated Hospital of Hebei University, Hebei, China
| |
Collapse
|
23
|
Manogaran P, Beeraka NM, Padma VV. The Cytoprotective and Anti-cancer Potential of Bisbenzylisoquinoline Alkaloids from Nelumbo nucifera. Curr Top Med Chem 2020; 19:2940-2957. [DOI: 10.2174/1568026619666191116160908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
:
Natural product therapy has been gaining therapeutic importance against various diseases,
including cancer. The failure of chemotherapy due to its associated adverse effects promoted adjunct
therapy with natural products. Phytochemicals exert anti-carcinogenic activities through the regulation
of various cell signaling pathways such as cell survival, inflammation, apoptosis, autophagy and metastasis.
The ‘small molecule-chemosensitizing agents’ from plants induce apoptosis in drug-resistant and
host-immune resistant cancer cells in in vitro as well as in vivo models. For example, alkaloids from Nelumbo
nucifera, liensinine, isoliensinine and neferine exert the anticancer activity through enhanced
ROS generation, activation of MAP kinases, followed by induction of autophagy and apoptotic cell
death. Likewise, these alkaloids also exert their cytoprotective action against cerebrovascular
stroke/ischemic stroke, diabetes, and chemotherapy-induced cytotoxicity. Therefore, the present review
elucidates the pharmacological activities of these bisbenzylisoquinoline alkaloids which include the cytoprotective,
anticancer and chemosensitizing abilities against various diseases such as cardiovascular
diseases, neurological diseases and cancer.
Collapse
Affiliation(s)
- Prasath Manogaran
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Narasimha Murthy Beeraka
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Viswanadha Vijaya Padma
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|