1
|
Wu S, Guo Y, Wang R. Combining network pharmacology and experimental verification to explore the inhibitory effects of Deoxyelephantopin (DET) Against Non-Small Cell Lung Cancer (NSCLC). BMC Cancer 2025; 25:738. [PMID: 40259252 PMCID: PMC12010628 DOI: 10.1186/s12885-025-14066-3] [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: 10/04/2024] [Accepted: 04/01/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND DET has a significant inhibitory activity against a range of cancer cells; however, its specific effects and underlying mechanisms in Non-Small Cell Lung Cancer (NSCLC) remain to be fully elucidated. This study aimed to investigate the potential mechanisms through which DET exerts its anti-neoplastic effects on NSCLC. METHOD Targets of DET were predicted using the SwissTargetPrediction database. Disease targets for NSCLC were obtained from the GeneCards database, and the intersection between drug targets and disease targets was determined. The STRING database was then employed to construct a protein-protein interaction (PPI) network and analyze target interactions. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG)enrichment analyses were conducted to investigate their biological functions. Molecular docking simulations were conducted using AutoDock software to analyze the binding interactions between DET and key target proteins. Subsequently, both in vitro and in vivo experiments were carried out to evaluate the anticancer effects of DET, with alterations in key gene expression levels assessed through RT-qPCR and Western blot analyses. RESULTS A total of 52 potential targets were discovered for DET and NSCLC. The PPI analysis revealed 5 hub targets, including CASP3, PTGS2, TNFα, ICAM1 and JUN. GO analysis identified 164 biological processes, 44 molecular functions and 40 cellular components. KEGG analysis revealed that DET anticancer effects were mediated through multiple pathways, primarily the AGE-RAGE and TNF signaling pathways. Experimental results demonstrated that DET inhibited the proliferation and migration of H460 cells and induced apoptosis in vitro. RT-qPCR and WB indicated that DET up regulated Bax and CASP3 while down regulating Bcl2, JUN, TNFα, NF-κB, ICAM1 and PTGS2. CONCLUSION This study aims to investigate the inhibitory effect of DET on NSCLC by combining network pharmacology and experimental methods. The results demonstrate that DET effectively inhibited the proliferation of H460 cells and induced apoptosis, with significant involvement of the AGE-RAGE and TNF signaling pathways, suggesting its potential as a therapeutic intervention for NSCLC.
Collapse
Affiliation(s)
- Shenjia Wu
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang Anhui, 236041, China
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Fuyang Normal University, 100 Qinghe West Road, Fuyang Anhui, 236041, China
| | - Ying Guo
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang Anhui, 236041, China
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Fuyang Normal University, 100 Qinghe West Road, Fuyang Anhui, 236041, China
| | - Rong Wang
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang Anhui, 236041, China.
- Anhui Province Key Laboratory of Embryo Development and Reproductive Regulation, Fuyang Normal University, 100 Qinghe West Road, Fuyang Anhui, 236041, China.
| |
Collapse
|
2
|
Wang Y, Zhao Q, Ding L, Liu X, Li Z, Zhou X, Wang D, Du M, Yang G, Yin M, Guo X. Transcriptome analysis of key genes and pathways associated with cisplatin resistance in oral squamous cell carcinoma Cal27 cells. Clin Transl Oncol 2025:10.1007/s12094-025-03924-y. [PMID: 40227535 DOI: 10.1007/s12094-025-03924-y] [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: 12/09/2024] [Accepted: 04/01/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) has a poor postoperative recovery and is prone to drug resistance during long-term chemotherapy, but the molecular mechanism of its resistance has not been fully elucidated. METHODS In the present study, a cisplatin-resistant cell line Cal27R was established and the key genes and pathways associated with drug resistance were explored using bioinformatics analysis and molecular biology experimental techniques. RESULTS Transcriptome analysis reveals a total of 1927 differentially expressed genes (DEGs). GO and further KEGG analysis revealed the DEGs were primarily concentrated in the tumor necrosis factor (TNF) and the mitogen-activated protein kinase (MAPK) signaling pathway. PPI network analysis identified six genes exhibiting significant interactions. Among these, interrogation of the TCGA database revealed elevated expression levels of TNF, TGFB1, and IL1B in tumors from drug-resistant patients, whereas EGF and FOS expression was significantly downregulated. The level of immune infiltration was positive correlated with the expression of TNF, TGFB1, IL6 and EGF, conversely, negative correlated with that of IL1B. Furthermore, low expression of TNF and FOS, as well as high expression of TGFB1, IL6 and EGF, was associated with poor overall prognosis. Based on the comprehensive analysis above, TNF, TGFB1, and EGF were ultimately selected as target genes to positively regulate the cisplatin resistance of Cal27R cells. Furthermore, we validated the expression of target genes in human tongue carcinoma tissues and paired adjacent normal tissues. Knockout of these genes significantly reduced drug resistance, consistent with our initial hypothesis. Whole-exome sequencing (WES) analysis confirmed the absence of underlying mutations, thereby corroborating the bioinformatics predictions. CONCLUSION TNF, TGFB1 and EGF were regarded as the key genes associated with cisplatin resistance and poor prognosis in OSCC. Meanwhile, their related TNF and MAPK pathways were considered as the pivotal signaling pathways. Our results provide a theoretical and experimental basis for potential diagnostic and therapeutic targets to address drug resistance in clinical settings.
Collapse
Affiliation(s)
- Yu Wang
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Qiwei Zhao
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Long Ding
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Xiayang Liu
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Zhuang Li
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Xinyue Zhou
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China
- Hubei Shizhen Laboratory, Wuhan, 430065, China
| | - Danru Wang
- Wuhan Bioeagle Biological Science and Technology Co. Ltd, Wuhan, 430073, China
| | - Mengtian Du
- Wuhan Bioeagle Biological Science and Technology Co. Ltd, Wuhan, 430073, China
| | - Guohua Yang
- Wuhan Bioeagle Biological Science and Technology Co. Ltd, Wuhan, 430073, China
- Department of Medical Genetics, School of Basic Medical Science, Demonstration Center for Experimental Basic Medicine Education, Wuhan University, Wuhan, 430071, China
| | - Mingzhu Yin
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China.
- Hubei Shizhen Laboratory, Wuhan, 430065, China.
| | - Xiaohong Guo
- Department of Basic Medicine, Hubei University of Chinese Medicine, Hongshan District, 16 Huangjiahu West Road, Wuhan, 430065, Hubei, China.
- Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
3
|
Tong Z, Wang Z, Jiang J, Fu W, Hu S. Glycyrrhizin enhances the antitumor activity of cisplatin in non‑small cell lung cancer cells by influencing DNA damage and apoptosis. Oncol Lett 2025; 29:207. [PMID: 40070780 PMCID: PMC11894513 DOI: 10.3892/ol.2025.14954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/07/2025] [Indexed: 03/14/2025] Open
Abstract
The objective of the present study was to elucidate the mechanism by which glycyrrhizin enhances the antitumor activity of cisplatin in non-small cell lung cancer. Initially, A549 cells were treated with different concentrations of glycyrrhizin (0.25-8 mM) or cisplatin (10-160 µM) for 48 h to investigate the effect of glycyrrhizin combined with cisplatin on A549 cells in vitro. Subsequently, A549 cells were divided into control (untreated), CP (20 µM cisplatin), GL (2 mM glycyrrhizin) and CP + GL (20 µM cisplatin + 2 mM glycyrrhizin) groups to elucidate the underlying mechanism of glycyrrhizin. After 48 h incubation, the viability and colony-forming ability of the cells were assessed using MTT and colony formation assays. Apoptosis levels and cell cycle progression were analyzed using flow cytometry and western blotting was used to evaluate apoptosis- and cell cycle-related proteins. Additionally, comet assays and western blotting were used to evaluate DNA damage and relevant proteins. The results demonstrated both glycyrrhizin and cisplatin individually reduced A549 cell viability in a concentration-dependent manner. Cisplatin demonstrated a lower half-maximal inhibitory concentration (IC50) at higher glycyrrhizin concentrations, with an IC50 value of ~35 µM with 2 mM glycyrrhizin. Furthermore, the combined treatment of glycyrrhizin and cisplatin synergistically reduced cell colony-forming ability, induced apoptosis and arrested the cell cycle at the G2 phase, showing greater efficacy when compared with either treatment individually. In addition, western blotting analysis demonstrated that, in comparison with treatment with cisplatin or glycyrrhizin alone, the combined treatment markedly increased the protein expression levels of B-cell lymphoma 2-associated X protein, cleaved-caspase-3/caspase-3, γH2AX, phosphorylated-checkpoint kinase 1 and phosphorylated-p53/p53, while notably reducing the protein levels of B-cell lymphoma 2, cyclin D1, cyclin-dependent kinase 2 and cyclin-dependent kinase 4. The findings of the present study indicate that glycyrrhizin enhances the antitumor efficacy of cisplatin in non-small cell lung cancer cells by modulating DNA damage and apoptosis.
Collapse
Affiliation(s)
- Zhufeng Tong
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Zhen Wang
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Jinghan Jiang
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Wenqi Fu
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Siying Hu
- Department of General Practice, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| |
Collapse
|
4
|
WANG R, GAO F, HAO L, ZHANG J, CHEN Z, SONG D, GAO F, HE W, ZHAO K, GUAN J. Oxypeucedanin alleviates cisplatin-induced intestinal toxicity through impairing caspase-3/gasdermin E signaling. J Vet Med Sci 2025; 87:431-441. [PMID: 40024724 PMCID: PMC11964853 DOI: 10.1292/jvms.24-0488] [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: 12/05/2024] [Accepted: 02/14/2025] [Indexed: 03/04/2025] Open
Abstract
Oxypeucedanin (OPD) is a compound naturally present in plants such as Peucedanum praeruptorum, a kind of medicinal herb which displays anti-inflammatory activities. However, its protective role on chemotherapy-induced inflammatory injuries has not been well studied. In the present study, we found that OPD could alleviate cisplatin (CDDP)-induced intestinal inflammatory injury in mice, which was accompanied by the decrease of immune cell infiltration and reverse of mitochondrial dysfunction in intestinal tissue. Then we noticed that OPD could inhibit cisplatin-induced intestinal cell pyroptosis, which was indicated by the reduction of LDH release and PI positive signal in the cells and tissues. The reason is that OPD could suppress caspase-3/gasdermin E (GSDME) signaling, which is a critical mediator of chemotherapy-induced tissue injury. We also detected the upstream factors of caspase-3/GSDME signaling and have found that cisplatin-induced Reactive oxygen species (ROS) production and DNA damages could also be inhibited by OPD. Importantly, OPD administration did not reverse cisplatin-suppressed tumor burden in animals. Our present study provides a potential application of OPD to reduce cisplatin-induced side effects, without interfering the anti-tumor capacity, indicating a way to extend the duration of cisplatin administration.
Collapse
Affiliation(s)
- Renling WANG
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Fei GAO
- Department of Laboratory Animals, College of Animal Science,
Jilin University, Changchun, China
| | - Lingrui HAO
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Jing ZHANG
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Zhexu CHEN
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Deguang SONG
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Feng GAO
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Wenqi HE
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Kui ZHAO
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| | - Jiyu GUAN
- State Key Laboratory for Diagnosis and Treatment of Severe
Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of
Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University,
Changchun, China
| |
Collapse
|
5
|
Mazzilli R, Zamponi V, Mancini C, Giorgini B, Golisano B, Mikovic N, Pecora G, Russo F, Martiradonna M, Paravani P, Prosperi D, Faggiano A. Neuroendocrine tumors and diabetes mellitus: which treatment and which effect. Endocrine 2025; 88:36-50. [PMID: 39752043 DOI: 10.1007/s12020-024-04149-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Diabetes mellitus (DM) and neuroendocrine tumors (NET) can exert unfavorable effects on each other prognosis. In this narrative review, we evaluated the effects of NET therapies on glycemic control and DM management and the effects of anti-diabetic therapies on NET outcome and management. For this purpose, we searched the PubMed, Science Direct, and Google Scholar databases for studies reporting the effects of NET therapy on DM as well as the effect of DM therapy on NET. The majority of NET treatments appear to impair glycaemic control, both inducing hypoglycemic or, more commonly, hyperglycemia and even new-onset DM. However, glucose metabolism imbalance can be effectively managed by modulating anti-diabetic therapy and adopting an appropriate nutritional approach. On the other hand, the effects of anti-diabetic treatment, like insulin, sulfonylureas, thiazolidinediones, ipeptidyl-peptidase-4 inhibitors, Glucagon-like peptide-1 receptor agonists, and Sodium-glucose cotransporter-2 inhibitors on NET are unclear. Recently, metformin has been investigated in patients with gastroenteropancreatic NET resulting in improved progression free survival suggesting a potential antineoplastic role. Finally, the management of DM in patients with NET is of great clinical relevance to correctly perform radiological procedures and even more functional imaging procedures, as well as to optimize the therapy and avoid treatment withdrawal or discontinuation. In conclusion, understanding the mechanisms underlying therapy-induced DM and implementing appropriate monitoring and management strategies of DM are essential for optimizing NET patient outcome and quality of life.
Collapse
Affiliation(s)
- Rossella Mazzilli
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy.
| | - Virginia Zamponi
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Camilla Mancini
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Beatrice Giorgini
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Bianca Golisano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Nevena Mikovic
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Giulia Pecora
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Flaminia Russo
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Maurizio Martiradonna
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Piero Paravani
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Daniela Prosperi
- Unit of Nuclear Medicine, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
| | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Qin Z, Ye F, Wang J, Jiang J, Zhang X, Li H, Feng L. BUB1B promotes cisplatin resistance in gastric cancer via Rad51-mediated DNA damage repair. Transl Oncol 2025; 54:102334. [PMID: 40056529 PMCID: PMC11930193 DOI: 10.1016/j.tranon.2025.102334] [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: 11/28/2024] [Revised: 02/26/2025] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Cisplatin resistance significantly impedes the treatment of gastric cancer (GC). This work examined the possible therapeutic target status and function of BUB1B in controlling cisplatin resistance. METHODS Following the identification of differentially expressed genes (DEGs), protein-protein interaction (PPI) network analysis was conducted using datasets from the Cancer Genome Atlas-stomach adenocarcinoma (TCGA-STAD), GSE51575, and GSE79973. Functional tests assessed the effect of BUB1B overexpression and knockdown on the GC cells. Enrichment analysis and RNA-seq identified pathways linked to BUB1B. Additionally, the function of BUB1B in GC cells resistant to cisplatin in regulating DNA repair was examined, as its relationship with Rad51 inhibitor (B02) in regulating cell cycle, proliferation, and apoptosis. The combined effects of Rad51 suppression and BUB1B overexpression on tumor development in cisplatin-resistant GC cells were further validated in vivo xenograft models. RESULTS Significant overexpression of six critical overlapping genes was seen in GC tissues. The GC cell invasion, migration, and proliferation processes were improved by BUB1B overexpression, whereas BUB1B knockdown prevented these outcomes. Genes involved in DNA repair were downregulated by BUB1B knockdown, according to an RNA-seq study. BUB1B overexpression boosted cell survival via modulating cell cycle proteins, but BUB1B knockdown hampered DNA repair and increased death in cisplatin-resistant GC cells. Overexpression of BUB1B enhanced tumor development in vivo and counteracted the inhibitory effects of B02 on cell growth. CONCLUSION BUB1B enhances cisplatin resistance in gastric cancer by regulating DNA repair and cell cycle progression, suggesting that targeting BUB1B may be a feasible therapeutic strategy.
Collapse
Affiliation(s)
- Zhe Qin
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Fangzhou Ye
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jiayi Wang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Jun Jiang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Huanqing Li
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital Affiliated to Fudan University, Shanghai 201100, China.
| |
Collapse
|
7
|
Zhang J, Yu Q, Zhu W, Sun X. Recent advances in the role of circRNA in cisplatin resistance in tumors. Cancer Gene Ther 2025:10.1038/s41417-025-00899-4. [PMID: 40148680 DOI: 10.1038/s41417-025-00899-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/08/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major threat to human health, with chemotherapy serving as one of the main treatment strategies to alleviate patient suffering. However, prolonged chemotherapy often leads to the development of drug resistance, complicating treatment outcomes. Cisplatin, a commonly utilized chemotherapeutic agent, demonstrates efficacy against a range of cancers but frequently encounters resistance, posing a significant challenge in tumor management and prognosis. Drug resistance not only facilitates tumor progression but also reduces survival rates, highlighting the urgent need for innovative strategies to overcome this issue. In recent years, non-coding RNAs, particularly circular RNAs (circRNAs), have gained attention in cancer therapy due to their stability and specificity. Moreover, an increasing number of studies have reported that circRNAs are involved in cisplatin resistance across various types of cancer. This paper primarily reviews the mechanisms and roles of circRNA in mediating cisplatin resistance over the past 3 years. These findings highlight circRNAs as promising therapeutic targets for overcoming cancer drug resistance.
Collapse
Affiliation(s)
- Jiawen Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiwen Yu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Weijin Zhu
- Department of Clinical Laboratory Medicine, Hospital of Traditional Chinese Medicine, Changzhou, Jiangsu, China
| | - Xiaochun Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
8
|
BERNAL GIULIANO, AQUEA GISELA, RAMÍREZ-RIVERA SEBASTIÁN. Metal-based molecules in the treatment of cancer: From bench to bedside. Oncol Res 2025; 33:759-779. [PMID: 40191719 PMCID: PMC11964877 DOI: 10.32604/or.2024.057019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/15/2024] [Indexed: 04/09/2025] Open
Abstract
Cancer remains one of the leading causes of death in the world, with more than 9 million deaths in 2022, a number that continues to rise. This highlights the urgent need for the development of new drugs, with enhanced antitumor capabilities and fewer side effects. Metal-based drugs have been used in clinical practice since the late 1970s, beginning with the introduction of cisplatin. Later, two additional platinum-based molecules, carboplatin, and oxaliplatin, were introduced, and all three continue to be widely used in the treatment of various cancers. However, despite their significant anticancer activity, the undesirable side effects of these drugs have motivated the scientific community to explore other metal-based complexes with greater anticancer potential and fewer adverse effects. In this context, metals such as ruthenium, copper, gold, zinc, palladium, or iridium, present promising alternatives for the development of new anticancer agents. Unfortunately, although thousands of metal-based drugs have been synthesized and tested both in vitro and in animal models, only a few ruthenium-based drugs have entered clinical trials in recent years. Meanwhile, many other molecules with comparable or even greater anticancer potential have not advanced beyond the laboratory stage. In this review, we will revisit the mechanisms of action and anticancer activities of established platinum-based drugs and explore their use in recent clinical trials. Additionally, we will examine the development of potential new metal-based drugs that could one day contribute to cancer treatment worldwide.
Collapse
Affiliation(s)
- GIULIANO BERNAL
- />Laboratory of Molecular and Cellular Biology of Cancer, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, 1781421, Chile
| | - GISELA AQUEA
- />Laboratory of Molecular and Cellular Biology of Cancer, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, 1781421, Chile
| | - SEBASTIÁN RAMÍREZ-RIVERA
- />Laboratory of Molecular and Cellular Biology of Cancer, Department of Biomedical Sciences, Faculty of Medicine, Universidad Católica del Norte, Coquimbo, 1781421, Chile
| |
Collapse
|
9
|
Liang H, Li S, Peng X, Xiao H. Overview of the epigenetic/cytotoxic dual-target inhibitors for cancer therapy. Eur J Med Chem 2025; 285:117235. [PMID: 39788061 DOI: 10.1016/j.ejmech.2024.117235] [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: 11/07/2024] [Revised: 12/24/2024] [Accepted: 12/31/2024] [Indexed: 01/12/2025]
Abstract
Epigenetic dysregulation plays a pivotal role in the initiation and progression of various cancers, influencing critical processes such as tumor growth, invasion, migration, survival, apoptosis, and angiogenesis. Consequently, targeting epigenetic pathways has emerged as a promising strategy for anticancer drug discovery in recent years. However, the clinical efficacy of epigenetic inhibitors, such as HDAC inhibitors, has been limited, often accompanied by resistance. To overcome these challenges, innovative therapeutic approaches are required, including the combination of epigenetic inhibitors with cytotoxic agents or the design of dual-acting inhibitors that target both epigenetic and cytotoxic pathways. In this review, we provide a comprehensive overview of the structures, biological functions and inhibitors of epigenetic regulators (such as HDAC, LSD1, PARP, and BET) and cytotoxic targets (including tubulin and topoisomerase). Furthermore, we discuss recent advancement of combination therapies and dual-target inhibitors that target both epigenetic and cytotoxic pathways, with a particular focus on recent advances, including rational drug design, pharmacodynamics, pharmacokinetics, and clinical applications.
Collapse
Affiliation(s)
- Hailiu Liang
- School of Medical and Information Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Shuqing Li
- School of Medical and Information Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China
| | - Xiaopeng Peng
- School of Medical and Information Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, PR China.
| | - Hao Xiao
- School of Medical and Information Engineering, School of Pharmacy, Gannan Medical University, Ganzhou, 341000, PR China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Gannan Medical University, Ganzhou, 341000, PR China.
| |
Collapse
|
10
|
Wang H, Lu C, Zhou H, Zhao X, Huang C, Cheng Z, Liu G, You X. Synergistic effects of dihydroartemisinin and cisplatin on inducing ferroptosis in gastric cancer through GPX4 inhibition. Gastric Cancer 2025; 28:187-210. [PMID: 39733394 DOI: 10.1007/s10120-024-01574-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/30/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND In the past several decades, cisplatin (DDP), in combination with other drugs, has been used as the mainstay chemotherapy drug for the treatment of gastric cancer (GC). However, the clinical application of DDP is restricted because of its toxic side effects, it is imperative to explore less toxic and more effective treatment strategies. Dihydroartemisinin (DHA) has been shown to exert potent anticancer effects through ferroptosis in multiple malignancies and has shown high efficacy and safety. METHODS Cell viability assay, live/dead staining assay, EDU proliferation assay, MitoTracker assay, BODIPY C11 assay and other cell assays in vitro were employed to observe DHA in combination with DDP inducing ferroptosis in GC. Subsequently, proteomic analysis integrated with database analysis and clinical sample detection were utilized to elucidate the mechanism of DHA inducing ferroptosis in GC both in vitro and in vivo. RESULTS In this study, we found that DHA combined with DDP can synergistically inhibit the proliferation, invasion and migration of GC cells and induce ferroptosis. Further studies have shown that DHA acts in combination with DDP to induce ferroptosis in GC cells by inhibiting GPX4 in vivo and in vitro. CONCLUSION In summary, this study is the first to report that DHA and DDP synergically promote ferroptosis in GC cells, the combination of DDP and DHA is a promising strategy from the perspective of toxicity of DDP, which may be a promising therapeutic approach.
Collapse
Affiliation(s)
- Huina Wang
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Chanchan Lu
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Haihua Zhou
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Xiaojun Zhao
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Chuanjiang Huang
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Zhiyi Cheng
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Guiyuan Liu
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Xiaolan You
- Department of Gastrointestinal Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China.
| |
Collapse
|
11
|
Wang C, Xia W. Proanthocyanidin Regulates NETosis and Inhibits the Growth and Proliferation of Liver Cancer Cells - In Vivo, In Vitro and In Silico Investigation. Cell Biochem Biophys 2025; 83:1223-1235. [PMID: 39382828 DOI: 10.1007/s12013-024-01557-6] [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: 09/17/2024] [Indexed: 10/10/2024]
Abstract
Liver cancer ranks third in global cancer-related mortality, with about 700,000 deaths recorded yearly, making it one of the most common cancers worldwide. Even though prognoses differ according to the severity of the diseases, many patients now exhibit an increased life cycle since the implementation of chemotherapy. In the current study, we investigated the effect of proanthocyanidin ‒a polyphenol molecule found in many plants‒ on the proliferation and invasion of liver cancer cells. In particular, we determined the effect of proanthocyanidin on the serum levels of four strategic liver cancer target, TNFα, IL-6, cfDNA, and IL-1β. Further molecular insight on the inhibitory mechanism of proanthocyanidin against TNFα, IL-6, and IL-1β was obtained via molecular docking, molecular dynamics simulations and binding free energy calculations. Results showed that proanthocyanidin inhibited the growth of HepG2 and HEP3B cells, and effectively reduced clonogenic survival and invasion potential when compared to control cells. Proanthocyanidin was also found to suppress the expression of Bcl-2 (26 kDa) protein in HepG2 cells, while increasing the expression of Bax (21 kDa). Molecular dynamics (MD) and thermodynamic binding free energy calculations showed that proanthocyanidin maintained stable binding within the active site of target proteins across the entire 100 ns MD simulation period, and its binding affinity outscored respective control molecules.In conclusion, the multifaceted analysis showcased in this study demonstrated promising anti-cancer effect of proanthocyanidin on HepG2 and HEP3B cancer cells, highlighting its potential as a viable liver cancer therapeutic alternative.
Collapse
Affiliation(s)
- Chenhui Wang
- Department of Pharmaceutical, Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, No. 427, Section 3, Furong Middle Road, Changsha, 410007, China
| | - Wu Xia
- Department of Pharmaceutical, Brain Hospital of Hunan Province, The Second People's Hospital of Hunan Province, No. 427, Section 3, Furong Middle Road, Changsha, 410007, China.
| |
Collapse
|
12
|
Cabrera-Serrano AJ, Sánchez-Maldonado JM, González-Olmedo C, Carretero-Fernández M, Díaz-Beltrán L, Gutiérrez-Bautista JF, García-Verdejo FJ, Gálvez-Montosa F, López-López JA, García-Martín P, Pérez EM, Sánchez-Rovira P, Reyes-Zurita FJ, Sainz J. Crosstalk Between Autophagy and Oxidative Stress in Hematological Malignancies: Mechanisms, Implications, and Therapeutic Potential. Antioxidants (Basel) 2025; 14:264. [PMID: 40227235 PMCID: PMC11939785 DOI: 10.3390/antiox14030264] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 04/15/2025] Open
Abstract
Autophagy is a fundamental cellular process that maintains homeostasis by degrading damaged components and regulating stress responses. It plays a crucial role in cancer biology, including tumor progression, metastasis, and therapeutic resistance. Oxidative stress, similarly, is key to maintaining cellular balance by regulating oxidants and antioxidants, with its disruption leading to molecular damage. The interplay between autophagy and oxidative stress is particularly significant, as reactive oxygen species (ROS) act as both inducers and by-products of autophagy. While autophagy can function as a tumor suppressor in early cancer stages, it often shifts to a pro-tumorigenic role in advanced disease, aiding cancer cell survival under adverse conditions such as hypoxia and nutrient deprivation. This dual role is mediated by several signaling pathways, including PI3K/AKT/mTOR, AMPK, and HIF-1α, which coordinate the balance between autophagic activity and ROS production. In this review, we explore the mechanisms by which autophagy and oxidative stress interact across different hematological malignancies. We discuss how oxidative stress triggers autophagy, creating a feedback loop that promotes tumor survival, and how autophagic dysregulation leads to increased ROS accumulation, exacerbating tumorigenesis. We also examine the therapeutic implications of targeting the autophagy-oxidative stress axis in cancer. Current strategies involve modulating autophagy through specific inhibitors, enhancing ROS levels with pro-oxidant compounds, and combining these approaches with conventional therapies to overcome drug resistance. Understanding the complex relationship between autophagy and oxidative stress provides critical insights into novel therapeutic strategies aimed at improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Antonio José Cabrera-Serrano
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - José Manuel Sánchez-Maldonado
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Carmen González-Olmedo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - María Carretero-Fernández
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
| | - Leticia Díaz-Beltrán
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Juan Francisco Gutiérrez-Bautista
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Servicio de Análisis Clínicos e Inmunología, University Hospital Virgen de las Nieves, 18014 Granada, Spain
- Department of Biochemistry, Molecular Biology and Immunology III, University of Granada, 18016 Granada, Spain
| | - Francisco José García-Verdejo
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Gálvez-Montosa
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - José Antonio López-López
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Paloma García-Martín
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Eva María Pérez
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Campus de la Salud Hospital, PTS, 18016 Granada, Spain
| | - Pedro Sánchez-Rovira
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Medical Oncology Unit, University Hospital of Jaén, 23007 Jaén, Spain
| | - Fernando Jesús Reyes-Zurita
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
| | - Juan Sainz
- Genomic Oncology Area, GENYO, Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS, 18016 Granada, Spain; (A.J.C.-S.); (J.M.S.-M.); (C.G.-O.); (M.C.-F.); (L.D.-B.); (J.F.G.-B.); (F.J.G.-V.); (F.G.-M.); (J.A.L.-L.); (E.M.P.); (P.S.-R.)
- Instituto de Investigación Biosanitaria IBs.Granada, 18012 Granada, Spain;
- Department of Biochemistry and Molecular Biology I, Faculty of Sciences, University of Granada, 18012 Granada, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
| |
Collapse
|
13
|
Verma M, Fatima S, Syed A, Elgorban AM, Abid I, Wong LS, Khan MS, Ansari IA. 3-Acetyl-11-keto-β-boswellic acid (AKBA) induced antiproliferative effect by suppressing Notch signaling pathway and synergistic interaction with cisplatin against prostate cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03899-1. [PMID: 39985578 DOI: 10.1007/s00210-025-03899-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Studies on the assessment of anticancer efficacy of plant-derived phytochemicals by targeting signaling pathways have drawn a lot of attention recently for human health. Multiple investigations have proposed an involvement of Notch pathway in the processes of cancer angiogenesis and metastasis, and drug resistance. Moreover, overexpression of Notch signaling is associated with increased prostate cancer (PrCa) cell growth and development. A number of chemotherapeutic agents are reported to become resistant over a period of time and have severe side effects. To increase efficacy and lessen drug-induced toxicity, a variety of bioactive compounds have been utilized alone or as adjuncts to traditional chemotherapy. Therefore, in the present study, the potential of AKBA in inhibiting the proliferation of PrCa cells by modulating Notch signaling components and its efficacy in combination with cisplatin was investigated. The results exhibited a substantial reduction in cell survival (IC50 = 25.28 µM at 24 h and 16.50 µM at 48 h) and cellular alterations in AKBA-treated PrCa cells. Additionally, AKBA caused nuclear condensation, increased reactive oxygen species (ROS) generation, mitochondrial membrane depolarization, and caspase activation, ultimately leading to apoptosis in PrCa cells. Moreover, AKBA-elicited apoptosis was evidenced by an augmentation in the Bax to Bcl2 ratio. AKBA was also found to induce G0/G1 arrest which was substantiated by reduced cyclin D1 and CDK4 expression levels concomitantly with increased expression of p21 and p27 genes. Intriguingly, AKBA demonstrated significant downregulation of Notch signaling mediators. Furthermore, the isobolograms of the combination treatment indicated that AKBA has the potential to synergistically enhance the cytotoxic efficacy of cisplatin in DU145 cells, as evidenced by CI < 1 across all tested combinations. Overall, the results of this study suggest strong antiproliferative, apoptotic, and chemo-sensitizing potential of AKBA. Thus, AKBA holds a promising drug candidature warranting further investigation as a probable therapeutic option for both the prevention and treatment of PrCa and other solid tumors.
Collapse
Affiliation(s)
- Mahima Verma
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India
- Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Shireen Fatima
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India
- Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Centre of Excellence in Biotechnology Research (CEBR), King Saud University, Riyadh, Saudi Arabia
| | - Islem Abid
- Centre of Excellence in Biotechnology Research (CEBR), King Saud University, Riyadh, Saudi Arabia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Putra Nilai, 71800, Nilai, Negeri Sembilan, Malaysia
| | - Mohd Sajid Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Irfan Ahmad Ansari
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India.
- Department of Biosciences, Integral University, Lucknow, 226026, India.
| |
Collapse
|
14
|
El-Deeb NM, Ibrahim OM, Kamel AM, Gomaa AI, Kenawy AM. Computational development of mushroom-6-glucan/paclitaxel as a synergistic complementary medicine for breast cancer therapy. BMC Complement Med Ther 2025; 25:58. [PMID: 39955575 PMCID: PMC11830196 DOI: 10.1186/s12906-025-04772-7] [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: 07/22/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Breast cancer is chemo-resistant and highly metastatic, often resulting in patient mortality. One of the primary factors contributing to the metastasis and chemotherapy resistance is the presence of cancer stem-like cells. We posited that the natural polysaccharide known as 6-glucans, derived from Pleurotus ostreatus, could effectively counteract the chemotherapy resistance associated with cancer stem-like cells in breast cancer. METHODS We computationally developed a specific dual combinatorial therapy involving 6-glucans and Paclitaxel (PTX) and tested on preclinical 3D mammosphere human tumor models representing receptor-positive and receptor-negative breast cancer. Using this preclinical 3D spheroid technology, we tested the anti-cancer properties of these predicted treatment combinations on mammospheres containing human breast cancer stem cells. RESULTS Among the 40 distinct combinations examined, computational prediction revealed that the addition of 2.0 mg/mL of 6-glucans to a low dose of 3.0 µg/mL PTX was the sole combination demonstrating a synergistic effect. This optimized synergistic combination therapy displayed a significant inhibitory impact on human cancer epithelial and stem cell migration, evasion, and colony formation. The inclusion of 6-glucans also augmented apoptosis in both breast cancer cells and stem cells, leading to a six-fold reduction in BrdU labeled cells and an increased arrest of cells in the sub-G0 phase. These effects were mediated through mitochondrial dysfunction and the downregulation of associated oncogenes. CONCLUSION Our study revealed that the computationally predicted 6-glucans-based binary complementary medicine exhibited sequence- and concentration-dependent anticancer synergistic effects.
Collapse
Affiliation(s)
- Nehal M El-Deeb
- Pharmaceutical Bioproducts Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El- Arab City, Alexandria, Egypt.
| | - Omar M Ibrahim
- Department of Medicine and McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| | - Ayman M Kamel
- Pharmaceutical Sciences Division, Medicinal and Aromatic Plants Department, National Research Centre, Cairo, Egypt
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Ahmed I Gomaa
- National Research Center, Nutrition and Food Science Department, Doki, Giza, Egypt
| | - Ahmed M Kenawy
- Nucleic Acids Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Alexandria, Egypt
| |
Collapse
|
15
|
Shirbhate E, Singh V, Kore R, Koch B, Veerasamy R, Tiwari AK, Rajak H. Synergistic strategies: histone deacetylase inhibitors and platinum-based drugs in cancer therapy. Expert Rev Anticancer Ther 2025; 25:121-141. [PMID: 39873641 DOI: 10.1080/14737140.2025.2458156] [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: 09/20/2024] [Revised: 12/23/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
INTRODUCTION The synergistic combination of histone deacetylase inhibitors and platinum-based medicines represents a promising therapeutic strategy to efficacy and overcome drug resistance in cancer therapy, necessitating a comprehensive understanding on their molecular interactions and clinical potential. AREAS COVERED The objective of presented review is to investigate the molecular pathways of platinum medicines and HDAC inhibitors. A comprehensive literature review from 2011 to 2024 was conducted across multiple databases like MEDLINE, PubMed, Google Scholar, Science Direct, Scopus and official websites of ClinicalTrial.gov to explore publications on HDAC inhibitors, platinum drugs, and combination cancer therapies, revealing preliminary evidence of innovative treatment strategies involving HDAC inhibitors and platinum chemotherapeutics. Several new platinum (IV) complexes, with HDAC inhibitory moieties and better cytotoxicity profiles than conventional platinum drugs, are also reviewed here. EXPERT OPINION The above combination has great potential in cancer treatment, however managing toxicity, dosage regimens, and patient selection biomarkers are problematic. More selective HDAC inhibitors and innovative delivery techniques are potential areas for future research. An adaptation toward changing cancer therapeutic landscapes, highlights combining HDAC inhibitors with platinum-based medicines serves as a new concept for personalized medicine, however, a deeper research is still needed at this time.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Rakesh Kore
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| | - Biplab Koch
- Department of Zoology, Banaras Hindu University, Varanasi, India
| | | | - Amit Kumar Tiwari
- Cancer & System Therapeutics, UAMS College of Pharmacy, UAMS - University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur, India
| |
Collapse
|
16
|
Karunakar KK, Edwin ER, Gopalakrishnan M, Cheriyan BV, Ramaiyan V, Karthikha VS, Justin JP. Advances in nephroprotection: the therapeutic role of selenium, silver, and gold nanoparticles in renal health. Int Urol Nephrol 2025; 57:479-510. [PMID: 39312019 DOI: 10.1007/s11255-024-04212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
Renal toxicity is a disorder that causes considerable issues in healthcare systems world, highlighting the critical importance of creating alternative treatments. Metallic nanoparticles have recently emerged as promising therapeutic agents for nephroprotection because of their remarkable properties. Numerous disciplines, including medicine, biotechnology, and the food industry, are currently investigating and exploring metallic nanoparticles, such as selenium, silver, and gold, with promising outcomes. In this overview, we provide the most current findings on cutting-edge nephroprotection through metallic nanoparticles, especially selenium, silver, and gold nanoparticles. While outlining the benefits, we outline possible methods for developing metallic nanoparticles, characterization techniques, and nephroprotection therapies. Selenium nanoparticles (SeNPs) minimize oxidative stress, a primary cause of nephrotoxicity through cell regeneration which protects kidneys. Silver nanoparticles (AgNPs) have anti-inflammatory capabilities that help alleviate kidney damage and nephrotoxicity. Gold nanoparticles (AuNPs), which are biocompatible and immune-modifying, reduce inflammation and promote renal cell regeneration, indicating nephroprotective advantages. Renal protection via the use of metallic nanoparticles represents a promising new frontier in the fight against kidney disease and other renal disorders. Metallic nanoparticles of selenium, silver, and gold can protect the kidneys by lowering oxidative stress, reducing inflammation, and improving cell repair. Through their mechanisms, these nanoparticles effectively safeguard and repair kidney function, making them suitable for treating renal diseases. The potential applications of selenium, silver, and gold nanoparticles, as well as their complex modes of action and renal penetration, provide fresh hope for improving renal health and quality of life in patients with kidney disease. The current study highlights therapeutic ability, stability, nephroprotection, and toxicity profiles, as well as the importance of continuous research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Elizabeth Rani Edwin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Meenaloshini Gopalakrishnan
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India.
| | - Velmurugan Ramaiyan
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - V S Karthikha
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Jerry Peliks Justin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| |
Collapse
|
17
|
Zhang L, Yang Y, Tan J. Applications and emerging challenges of single-cell RNA sequencing technology in tumor drug discovery. Drug Discov Today 2025; 30:104290. [PMID: 39828052 DOI: 10.1016/j.drudis.2025.104290] [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: 03/14/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
Current therapeutic drugs are inadequate for curing tumors, highlighting the need for novel tumor drugs. The advancement of single-cell RNA sequencing (scRNA-seq) technology offers new opportunities for tumor drug discovery. This technology allows us to explore tumor heterogeneity and developmental mechanisms at the single-cell level. In this review, we outline the application of scRNA-seq in tumor drug discovery stages, including elucidating tumor mechanisms, identifying targets, screening drugs, and understanding drug action and resistance. We also discuss the challenges and future prospects of using scRNA-seq in drug development, providing a scientific foundation for advancing tumor therapies.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Yueying Yang
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China; Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China.
| |
Collapse
|
18
|
Dai WC, Chen TH, Peng TC, He YC, Hsu CY, Chang CC. Blockade of the STAT3/BCL-xL Axis Leads to the Cytotoxic and Cisplatin-Sensitizing Effects of Fucoxanthin, a Marine-Derived Carotenoid, on Human Bladder Urothelial Carcinoma Cells. Mar Drugs 2025; 23:54. [PMID: 39997178 PMCID: PMC11857094 DOI: 10.3390/md23020054] [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: 12/30/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Bladder cancer is a globally prevalent urological malignancy, with transitional carcinoma (TCC) representing the majority of cases. Cisplatin is the primary drug for metastatic bladder cancer chemotherapy; however, its application is limited by nephrotoxicity and resistance. Signal Transducer and Activator of Transcription 3 (STAT3) is an oncogenic transcription factor often overactivated in various cancers, making it an appealing drug target. Fucoxanthin, a marine carotenoid, has significant anticancer properties. This study explored Fucoxanthin's cytotoxic effects and its potential to potentiate the efficacy of Cisplatin, along with the mechanisms underlying these effects, on human bladder TCC cells. We demonstrated that Fucoxanthin is cytotoxic to bladder TCC cells by inducing apoptosis, evidenced by z-VAD-fmk-mediated annulment of Fucoxanthin's cytotoxicity. Furthermore, Fucoxanthin reduced the levels of inherent or interleukin-6-induced tyrosine 705-phosphorylated STAT3 accompanied by downregulating BCL-xL, a well-established STAT3 target. Notably, ectopic expression of STAT3-C, a dominant-active STAT3 mutant, or BCL-xL thwarted Fucoxanthin's proapoptotic and cytotoxic actions. Moreover, Fucoxanthin at subtoxic dosages enhanced the susceptibility to Cisplatin-induced apoptosis of bladder TCC cells initially resistant to Cisplatin. Remarkably, this Cisplatin-sensitizing effect of Fucoxanthin was abrogated when cells ectopically expressed STAT3-C or BCL-xL. Overall, for the first time, we proved that the proapoptotic, cytotoxic, and Cisplatin-sensitizing effects of Fucoxanthin on human bladder TCC cells are attributed to the blockade of the STAT3/BCL-xL axis. Our findings highlight that targeting the STAT3/BCL-xL axis is a promising strategy to eliminate bladder TCC cells and facilitate Cisplatin sensitization, and further support the potential of incorporating Fucoxanthin into Cisplatin-based chemotherapy for treating bladder cancer.
Collapse
Affiliation(s)
- Wen-Chyi Dai
- Doctoral Program in Biotechnology Industrial Innovation and Management, National Chung Hsing University, Taichung 402202, Taiwan;
| | - Tzu-Hsuan Chen
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan;
| | - Tzu-Ching Peng
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan; (T.-C.P.); (Y.-C.H.)
| | - Yung-Ching He
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan; (T.-C.P.); (Y.-C.H.)
| | - Chao-Yu Hsu
- Division of Urology, Department of Surgery, Tungs’ Taichung MetroHarbor Hospital, Taichung 435403, Taiwan
- Department of Rehabilitation, Jenteh Junior College of Medicine, Nursing and Management, Miaoli 356006, Taiwan
| | - Chia-Che Chang
- Doctoral Program in Biotechnology Industrial Innovation and Management, National Chung Hsing University, Taichung 402202, Taiwan;
- Department of Life Sciences, National Chung Hsing University, Taichung 402202, Taiwan;
- Graduate Institute of Biomedical Sciences, National Chung Hsing University, Taichung 402202, Taiwan; (T.-C.P.); (Y.-C.H.)
- Master Program in Precision Health, Doctoral Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, The iEGG and Animal Biotechnology Research Center, National Chung Hsing University, Taichung 402202, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung 413305, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung 404327, Taiwan
| |
Collapse
|
19
|
Hasan AA, Kalinina E, Zhdanov D, Volodina Y, Tatarskiy V. Re-Sensitization of Resistant Ovarian Cancer SKOV3/CDDP Cells to Cisplatin by Curcumin Pre-Treatment. Int J Mol Sci 2025; 26:799. [PMID: 39859517 PMCID: PMC11765683 DOI: 10.3390/ijms26020799] [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: 11/28/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
A major challenging problem facing effective ovarian cancer therapy is cisplatin resistance. Re-sensitization of cisplatin-resistant ovarian cancer cells to cisplatin (CDDP) has become a critical issue. Curcumin (CUR), the most abundant dietary polyphenolic curcuminoids derived from turmeric (Curcuma longa), has achieved previously significant anti-cancer effects against human ovarian adenocarcinoma SKOV-3/CDDP cisplatin-resistant cells by inhibition the gene expression of the antioxidant enzymes (SOD1, SOD2, GPX1, CAT and HO1), transcription factor NFE2L2 and signaling pathway (PIK3CA/AKT1/MTOR). However, the detailed mechanisms of curcumin-mediated re-sensitization to cisplatin in SKOV-3/CDDP cells still need further exploration. Here, a suggested curcumin pre-treatment therapeutic strategy has been evaluated to effectively overcome cisplatin-resistant ovarian cancer SKOV-3/CDDP and to improve our understanding of the mechanisms behind cisplatin resistance. The findings of the present study suggest that the curcumin pre-treatment significantly exhibited cytotoxic effects and inhibited the proliferation of the SKOV-3/CDDP cell line compared to the simultaneous addition of drugs. Precisely, apoptosis induced by curcumin pre-treatment in SKOV-3/CDDP cells is mediated by mitochondrial apoptotic pathway (cleaved caspases 9, 3 and cleaved PARP) activation as well as by inhibition of thioredoxin reductase (TRXR1) and mTOR/STAT3 signaling pathway. This current study could deepen our understanding of the anticancer mechanism of CUR pre-treatment, which not only facilitates the re-sensitization of ovarian cancer cells to cisplatin but may lead to the development of targeted and effective therapeutics to eradicate SKOV-3/CDDP cancer cells.
Collapse
Affiliation(s)
- Aseel Ali Hasan
- T.T. Berezov Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (E.K.); (D.Z.)
| | - Elena Kalinina
- T.T. Berezov Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (E.K.); (D.Z.)
| | - Dmitry Zhdanov
- T.T. Berezov Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia; (E.K.); (D.Z.)
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia
| | - Yulia Volodina
- Laboratory of Tumor Cell Death, Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia;
| | - Victor Tatarskiy
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Science, 34/5 Vavilov Street, 119334 Moscow, Russia
| |
Collapse
|
20
|
Sipping M, Kumar TS, Kamdem N. Scientific investigation on antibacterial, antioxidant, cytotoxic effects and TLC bioautography of Terminalia schimperiania stem bark extracts. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2025:jcim-2024-0251. [PMID: 39789714 DOI: 10.1515/jcim-2024-0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 11/08/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES Terminalia schimperiana Hochst, belonging to the Combretaceae family, is known for its ethnomedicinal values, particularly in treating various diseases in Africa. This study aimed to investigate the antibacterial, antioxidant, and cytotoxic properties of T. schimperiana stem bark extracts, with a specific focus on assessing their bioactive potential and identifying active compounds via TLC bioautography. METHODS The in vitro antimicrobial activity was assessed using the agar well diffusion method against selected clinical strains. Antioxidant activity was evaluated using several methods including free radical scavenging, ferrous ion chelation assays and total phenolic content analysis. The cytotoxicity of the extracts was assessed using MTT assay towards Raw 264.7 and Vero cell lines. RESULTS All extracts demonstrated significant antibacterial activity against the bacteria tested, with inhibition zones (IZ) ranging from 6.50 ± 0.71 to 15.50 ± 0.71 mm and minimum inhibitory concentrations (MIC) ranging from 1.95 to 1,250 μg/ml. The hydroethanolic extract exhibited strongest antioxidant activities with EC50 values of 188.50; 245.30, and 281.50 μg/mL for DPPH; ABTS, ferrous ion chelation assays respectively, and a high content of phenolic compounds (101.67 ± 2.97 µgEFA/mg DW). Importantly, no cytotoxic effects were observed on Raw 264.7 and Vero cell lines. HPTLC analysis identified alkaloids and phenolic compounds in both aqueous and hydroethanolic extracts. CONCLUSIONS These findings indicate T. schimperiana provides a wealth of bio-compounds that can be utilised in the pharmaceutical industry as antibacterial and antioxidant agents to combat antibiotic resistance.
Collapse
Affiliation(s)
- Marius Sipping
- PKFokam Institute of Excellence, Yaoundé, Cameroon
- Laboratory of Phytoprotection and Valorization of Genetic Resources, Biotechnology Centre, Nkolbisson, 201336 University of Yaoundé 1 , Yaoundé, Cameroon
| | | | | |
Collapse
|
21
|
Wang Y, Du M, Wang J, Bai Z, Cui C, Tong J, Liu Y, Guo S, Zhang W, Wu X, Li B. Carbon dots-cisplatin nano drug delivery system induces the death of oral tongue squamous cell under self-targeting chemical/photodynamic combined therapy. Colloids Surf A Physicochem Eng Asp 2025; 704:135511. [DOI: 10.1016/j.colsurfa.2024.135511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
22
|
Ma M, Hu R, Huang Q, Li J, Lv M, Sun J, Zhong X, Yi J, Peng L, Feng W, Ma W, Han Z, Zhang W, Sun X, Zhan B, Liu X, Zhou X. Digoxigenin activates autophagy in hepatocellular carcinoma cells by regulating the PI3K/AKT/mTOR pathway. Cancer Cell Int 2024; 24:405. [PMID: 39696358 DOI: 10.1186/s12935-024-03602-z] [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: 07/08/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is recognized as a highly malignant tumor. Targeted combination immunotherapy, the initially approved regimen, is compromised by adverse side effects and low response rates during clinical treatment. Traditional Chinese medicine and its derived natural compounds, known for their anticancer effects, offer advantages of low toxicity and cost. In this study, we performed high-throughput phenotypic screening in vitro to identify promising anti-HCC drugs. Among 1,444 bioactive compounds, digoxigenin (DIG) was found to significantly impede HCC cell progression. We validated DIG's therapeutic effects through assays such as cell counting by CCK8, lactate dehydrogenase, and colony formation. Analyses including transmission electron microscopy, western blotting, and immunofluorescence demonstrated that DIG inhibits HCC cell proliferation via autophagy. Network pharmacology and molecular docking studies suggest that DIG targets the PI3K/AKT/mTOR signaling pathway. Comparative treatments of Hep3B and Huh7 cells with DIG or mTOR inhibitors revealed similar inhibitory impacts, indicating that DIG induces autophagy by inhibiting the PI3K/AKT/mTOR pathway. In vivo studies confirmed that DIG halts the growth of subcutaneous xenograft tumors. In conclusion, DIG represents a potential HCC treatment by modulating the PI3K/AKT/mTOR pathway to induce autophagy. This research, via phenotypic screening, accelerates drug discovery and the development of novel therapies targeting the underlying mechanisms of liver cancer.
Collapse
Affiliation(s)
- Mengqing Ma
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Rui Hu
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Qi Huang
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Jing Li
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Minling Lv
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Jialing Sun
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xin Zhong
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Jinyu Yi
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China
| | - Lanfen Peng
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wenxing Feng
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wenfeng Ma
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Zhiyi Han
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Wei Zhang
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xinfeng Sun
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Bolin Zhan
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xingning Liu
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China
| | - Xiaozhou Zhou
- Department of Liver Disease, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China.
- Department of Liver Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, 518033, China.
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, 999078, China.
| |
Collapse
|
23
|
El-Tanani M, Rabbani SA, Ali AA, Alfaouri IGA, Al Nsairat H, Al-Ani IH, Aljabali AA, Rizzo M, Patoulias D, Khan MA, Parvez S, El-Tanani Y. Circadian rhythms and cancer: implications for timing in therapy. Discov Oncol 2024; 15:767. [PMID: 39692981 PMCID: PMC11655929 DOI: 10.1007/s12672-024-01643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 11/27/2024] [Indexed: 12/19/2024] Open
Abstract
Circadian rhythms, intrinsic cycles spanning approximately 24 h, regulate numerous physiological processes, including sleep-wake cycles, hormone release, and metabolism. These rhythms are orchestrated by the circadian clock, primarily located in the suprachiasmatic nucleus (SCN) of the hypothalamus. Disruptions in circadian rhythms, whether due to genetic mutations, environmental factors, or lifestyle choices, can significantly impact health, contributing to disorders such as sleep disturbances, metabolic syndrome, and cardiovascular diseases. Additionally, there is a profound link between the disruption of circadian rhythms and development of various cancer, the influence on disease incidence and progression. This incurred regulation by circadian clock on pathways has its implication in tumorigenesis, such as cell cycle control, DNA damage response, apoptosis, and metabolism. Furthermore, the circadian timing system modulates the efficacy and toxicity of cancer treatments. In cancer treatment, the use of chronotherapy to optimize the timing of medical treatments, involves administering chemotherapy, radiation, or other therapeutic interventions at specific intervals to enhance efficacy and minimize side effects. This approach capitalizes on the circadian variations in cellular processes, including DNA repair, cell cycle progression, and drug metabolism. Preclinical and clinical studies have demonstrated that chronotherapy can significantly improve the therapeutic index of chemotherapeutic agents like cisplatin and 5-fluorouracil by enhancing anticancer activity and reducing toxicity. Further research is needed to elucidate the mechanisms underlying circadian regulation of cancer and to develop robust chronotherapeutic protocols tailored to individual patients' circadian profiles, potentially transforming cancer care into more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates.
| | - Syed Arman Rabbani
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Areeg Anwer Ali
- RAK College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Ibrahim Ghaleb Ali Alfaouri
- Translational and Medical Research Centre (TMRC), Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
- RAK College of Nursing, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Hamdi Al Nsairat
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Israa Hamid Al-Ani
- Pharmacological and Diagnostic Research Center, Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Alaa A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Pharmacy, Yarmouk University, Irbid, Jordan
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| | - Dimitrios Patoulias
- Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
- Outpatient Department of Cardiometabolic Medicine, Second Department of Cardiology, Aristotle University of Thessaloniki, Hippokration General Hospital, Athens, Greece
| | - Mohammad Ahmed Khan
- School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Suhel Parvez
- School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | | |
Collapse
|
24
|
Ravichandiran P, Martyna A, Kochanowicz E, Maroli N, Kubiński K, Masłyk M, Boguszewska-Czubara A, Ramesh T. In Vitro and In Vivo Biological Evaluation of Novel 1,4-Naphthoquinone Derivatives as Potential Anticancer Agents. ChemMedChem 2024; 19:e202400495. [PMID: 39136593 DOI: 10.1002/cmdc.202400495] [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: 06/29/2024] [Revised: 08/13/2024] [Indexed: 10/16/2024]
Abstract
A novel library of naphthoquinone derivatives (3-5 aa) was synthesized and evaluated for their anticancer properties. Specifically, compounds 5 i, 5 l, 5 o, 5 q, 5 r, 5 s, 5 t, and 5 v demonstrated superior cytotoxic activity against the cancer cell lines that were studied. All the studied compounds exhibited a higher selectivity index (SI) and a favourable safety profile than the standard drug doxorubicin. Notably, compound 5 v displayed a greater cytotoxic effect on MCF-7 cells (IC50=1.2 μM, and 0.9 μM at 24 h and 48 h, respectively) compared to the standard drug doxorubicin (IC50=2.4 μM, and 2.1 μM at 24 h and 48 h, respectively). To further investigate the mechanism of cytotoxic effect, additional anticancer studies were conducted with 5 v in MCF-7 cells. The studies are including morphological changes, AO/EB (acridine orange/ethidium bromide) double staining, apoptosis analysis, cell colony assay, SDS-PAGE and Western blotting, cell cycle analysis, and detecting reactive oxygen species (ROS) assay. The findings showed that 5 v triggered cytotoxic effects in MCF-7 cells through the initiation of cell cycle arrest at the G1/S phase and necrosis. In vivo ecotoxicity studies indicated that 5 v had lower toxicity towards zebrafish larvae (LC50=50.15 μM) and had an insignificant impact on cardiac functions. In vivo xenotransplantation of MCF-7 cells in zebrafish larvae demonstrated a significant reduction in tumour volume in the xenograft. Approximately 95 % of the zebrafish larvae with 5 v xenografts survived after 10 days of the treatment. Finally, a computational modelling study was conducted on four protein receptors, namely ER, EFGR, BRCA1, and VEFGR2. The findings highlight the importance of the aminonaphthoquinone moiety, amide linkage, and propyl thio moiety in enhancing the anticancer properties. 5 v exhibited superior drug-likeness features and docking scores (-9.1, -7.1, -8.9, and -10.9 kcal/mol) compared to doxorubicin (-7.2, -6.1, -6.9, and -7.3 kcal/mol) against ER, EFGR, BRCA1, and VEGFR2 receptors, respectively. Therefore, the notable antitumor effects of naphthoquinone derivatives (3-5 aa) suggest that these molecular frameworks may play a role in the development of promising anticancer agents for cancer treatment.
Collapse
Affiliation(s)
- Palanisamy Ravichandiran
- R&D Education Center for Whole Life Cycle R&D of Fuel Cell Systems, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Department of Life Science, Department of Energy Storage/Conversion Engineering of Graduate School, Hydrogen and Fuel Cell Research Center, Jeonbuk National University, Jeonju, Jeollabuk-do, 54896, Republic of Korea
- Present Address: Analytical, HP Green R & D Centre, Hindustan Petroleum Corporation Limited, KIADB Industrial Area, Devangundi, Hoskote, Bengaluru, Karnataka, 562114, India
| | - Aleksandra Martyna
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Elżbieta Kochanowicz
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Nikhil Maroli
- Department of Physics and Astronomy, University of Delaware, Newark, DE, 19716, USA
| | - Konrad Kubiński
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Maciej Masłyk
- Department of Molecular Biology, Institute of Biological Sciences, The John Paul II Catholic University of Lublin, ul. Konstantynów 1i, 20-708, Lublin, Poland
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, Ul. Chodźki 4 A, 20-093, Lublin, Poland
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| |
Collapse
|
25
|
Liu Y, Guo Q, Shi Y, Cui M, Jing F. Research progress of novel anti-tumor drug formulations. Front Oncol 2024; 14:1507958. [PMID: 39737395 PMCID: PMC11683012 DOI: 10.3389/fonc.2024.1507958] [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: 10/08/2024] [Accepted: 12/02/2024] [Indexed: 01/01/2025] Open
Abstract
Cancers have become the second leading cause of death worldwide, following cardiovascular diseases.Traditional anti- cancer strategies, including radiotherapy chemotherapy, surgery, and targeted therapies, have been widely used but are often reassessed due to their significant side effects and relatively low cure rate. Recently, the development of novel formulations for anti-tumor drugs has gained considerable attention, marking a pivotal step forward in cancer treatment advancements. These innovative formulations aim to enhance the therapeutic efficacy of anti-tumor drugs by employing advanced drug formulation technologies and delivery systems. In particular, nano-drug delivery systems (NDDS) have emerged as a promising approach to improve drug targeting, reduce side effects, and overcome drug resistance. This review highlights recent progress in NDDS for anti-tumor drug development and explores the future prospects of these advanced formulations in improving cancer treatment outcomes.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pharmacology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - YunYan Shi
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - MengNa Cui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - FanBo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
26
|
Xiong L, Wei Y, Si H, Li Z, Wen J, Liu F, Wang X, Yang H, Chen L, Pi C, Han Y, Zhao L. Development of the Curcumin Analog CA7 Liposome and Its Evaluation for Efficacy Against Cervical Cancer in vitro and in vivo. Int J Nanomedicine 2024; 19:13411-13428. [PMID: 39691454 PMCID: PMC11651071 DOI: 10.2147/ijn.s493074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Objective The objective of this study was to develop liposomes (LP) containing a curcumin (CU) analog CA7 to enhance its pharmacokinetic profile and anti-cervical cancer (CC) effects. Methods Single-factor and Box-Behnken experiments were conducted to optimize the formulation of CA7-loaded liposomes (CA7-LP). The in vitro release, stability, biocompatibility, and pharmacokinetic of CA7-LP were evaluated. The biological effects of CA7-LP on Hela cells were assessed using MTT assays, colony formation assays, wound healing assays, and flow cytometry. Additionally, the anti-CC efficacy of CA7-LP was tested in mouse models of transplanted tumors. Results The optimal formulation of CA7-LP exhibited a particle size of 92.43 ± 1.52 nm, a polydispersity index of 0.27 ± 0.01, an encapsulation efficiency of 97.79 ± 1.49%, a drug loading of 3.23 ± 0.20%, and a zeta potential of -6.69 ± 0.77 mV. Transmission electron microscopy confirmed that a spherical morphology was exhibited by CA7-LP. The cumulative in vitro release of CA7-LP was found to be 2.84 times greater than that of CA7, and stability at room temperature was maintained for at least 90 d. Furthermore, a significantly higher uptake of CA7-LP by Hela cells was observed compared to curcumin and CA7, leading to enhanced inhibition of cell proliferation, migration and cell cycle, as well as increased apoptosis (p < 0.05). In vivo studies revealed that CA7-LP exhibited superior pharmacokinetic properties compared to CA7 (AUC: 3.58-fold, Cmax: 5.65-fold, t1/2z: 1.2-fold). The anti-CC effects of CA7-LP were found to be comparable to those of Cisplatin injection, with a better safety profile. Conclusion The newly developed CA7-LP is considered a promising candidate for the treatment of CC, demonstrating high potential for clinical application.
Collapse
Affiliation(s)
- Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Hui Si
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Zheng Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Jie Wen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Furong Liu
- Department of Oncology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Xiaodong Wang
- Department of Hepatobiliary Diseases, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Hongru Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| | - Ling Zhao
- Department of Psychiatry, Fundamental and Clinical Research on Mental Disorders Key Laboratory of Luzhou, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, People’s Republic of China
| |
Collapse
|
27
|
Basheer I, Wang H, Li G, Jehan S, Raza A, Du C, Ullah N, Li D, Sui G. β-caryophyllene sensitizes hepatocellular carcinoma cells to chemotherapeutics and inhibits cell malignancy through targeting MAPK signaling pathway. Front Pharmacol 2024; 15:1492670. [PMID: 39734415 PMCID: PMC11671526 DOI: 10.3389/fphar.2024.1492670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Background β-caryophyllene (BCP) is a naturally occurring bicyclic sesquiterpene extracted from various plants, and widely used as a medicinal agent for various diseases. During hepatocellular carcinoma (HCC) development, cancer cells generally exhibit increased cell proliferation due to mutations or aberrant expression of key regulatory genes. The current study determines the cytotoxic effects of BCP alone or in combination with doxorubicin (DOX) and cisplatin (DDP) on HCC cells, and elucidates the underlying mechanism of BCP to exert its anticancer activities. Materials and methods HepG2, SMMC-7721 HCC cells, and HL-7702 normal liver cells were treated with BCP, DOX, and DDP individually or combinatorially. Cell proliferation assay, flow cytometric assay, and Western blot were employed to evaluate the cytotoxic effects of these treatments. Transwell assays were used to examine BCP's effects on HCC cell migration and invasion. RNA-seq analysis was used to determine BCP's primary target genes in HepG2 cells. Integrative analysis of differentially expressed genes (DEGs) of RNA-seq data with an HCC TCGA dataset identified BCP-targeted genes that were verified by RT-qPCR analysis. Ectopic gene expression, cell viability, and colony formation assay were performed to validate the primary targets of BCP. Results BCP selectively inhibited HCC cell proliferation while exhibited relatively low toxicity in normal liver cells; however, DOX and DDP showed higher toxicity in normal cells than that in HCC cells. In combinatorial treatments, BCP synergistically enhanced cytotoxicity of DOX and DDP in HCC cells but this effect was markedly reduced in HL-7702 cells. BCP treatment reduced migration and invasion of HCC cells. Furthermore, RNA-seq analyses of BCP-treated HepG2 cells identified 433 protein-coding DEGs. Integrative analyses revealed five BCP-targeted DEGs regulating the MAPK signaling pathway. Among these five genes, three displayed a significantly positive correlation of their expression with the overall survival of HCC patients. As a primary target, PGF was significantly downregulated by BCP treatment, and its exogenous expression desensitized HCC cells to BCP-mediated inhibition. Discussion BCP inhibits malignant properties of HCC and synergistically sensitizes the anticancer activity of DOX and DDP. In HCC cells, BCP primarily targets the PGF gene and MAPK signaling pathway.
Collapse
Affiliation(s)
- Irum Basheer
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Hai Wang
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Guangyue Li
- Intelligent Biomedical Labs, Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shah Jehan
- Department of Vascular Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ali Raza
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Chentao Du
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Najeeb Ullah
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, China
| | - Dangdang Li
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| | - Guangchao Sui
- Key Laboratory of Saline-Alkali Vegetation Ecology Restoration, Ministry of Education, College of Life Science, Northeast Forestry University, Harbin, China
| |
Collapse
|
28
|
Shiri Aghbash P, Sadri Nahand J, Rahbar Farzam O, Hosseini SMR, Bayat M, Entezari Maleki T, Bannazadeh Baghi H. Combination of Vitex pseudo-negundo methanolic-extract with cisplatin can induce antioxidant activity and apoptosis in HeLa and Caski cells. Front Pharmacol 2024; 15:1476152. [PMID: 39697540 PMCID: PMC11653208 DOI: 10.3389/fphar.2024.1476152] [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: 08/05/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Background Cisplatin-based chemotherapy as a common therapeutic regimen for cervical cancer patients, is becoming more and more ineffective due to high resistance. This urges the need for introducing novel metabolics such as botanical drugs with the capacity to increase the cisplatin effectiveness. In that regard, here we investigated the anticancer effects of the Cisplatin-Vitex pseudo-Negundo combination in cervical cancer cell lines. Method and Material V. pseudo-Negundo fruits were dried and extracted methanolic fraction. The MTT assay was performed to evaluate cytotoxicity of both drugs in CaSki and HeLa cells. Then, apoptosis, ROS production, and cell cycling were assessed by flow cytometry assay in cells treated with V. pseudo-Negundo and Cisplatin and their combination. Also, the rate of cell migration and colony formation were measured, using wound healing and colony formation assay, respectively. Also, the expression level of related genes (CD133, BAX, BCL2, Casp-3/8/9, MMP-3) was evaluated using the RT-PCR method. Results The obtained results established that the V. pseudo-Negundo plant has medicinal properties to induce apoptotic and antioxidant signals. The combination treatment of methanol extraction and Cisplatin had a cytotoxic effect on cervical cancer cell lines (HeLa and CaSki) compared to monotherapy. Also, combination therapy resulted in an increased apoptosis rate and diminished ROS production in both CaSki and HeLa cell lines. Furthermore, V. pseudo-Negundo and Cisplatin combination therapy leads to cell cycle arrest in the G2-M and G0-G1 phase in HeLa and CaSki cell lines, respectively. Moreover, combination therapy decreased the colony formation and cell motility in both cell lines and upregulated caspases gene expression. Conclusion The combination of V. pseudo-Negundo with Cisplatin therapy results in a significant anti-cancer and antioxidant effect compared to cisplatin, representing a promising candidate for future clinical investigations.
Collapse
Affiliation(s)
- Parisa Shiri Aghbash
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javid Sadri Nahand
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Rahbar Farzam
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mobina Bayat
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari Maleki
- Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
29
|
Famurewa AC, George MY, Ukwubile CA, Kumar S, Kamal MV, Belle VS, Othman EM, Pai SRK. Trace elements and metal nanoparticles: mechanistic approaches to mitigating chemotherapy-induced toxicity-a review of literature evidence. Biometals 2024; 37:1325-1378. [PMID: 39347848 DOI: 10.1007/s10534-024-00637-7] [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: 04/15/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Anticancer chemotherapy (ACT) remains a cornerstone in cancer treatment, despite significant advances in pharmacology over recent decades. However, its associated side effect toxicity continues to pose a major concern for both oncology clinicians and patients, significantly impacting treatment protocols and patient quality of life. Current clinical strategies to mitigate ACT-induced toxicity have proven largely unsatisfactory, leaving a critical unmet need to block toxicity mechanisms without diminishing ACT's therapeutic efficacy. This review aims to document the molecular mechanisms underlying ACT toxicity and highlight research efforts exploring the protective effects of trace elements (TEs) and their nanoparticles (NPs) against these mechanisms. Our literature review reveals that the primary driver of ACT toxicity is redox imbalance, which triggers oxidative inflammation, apoptosis, endoplasmic reticulum stress, mitochondrial dysfunction, autophagy, and dysregulation of signaling pathways such as PI3K/mTOR/Akt. Studies suggest that TEs, including zinc, selenium, boron, manganese, and molybdenum, and their NPs, can potentially counteract ACT-induced toxicity by inhibiting oxidative stress-mediated pathways, including NF-κB/TLR4/MAPK/NLRP3, STAT-3/NLRP3, Bcl-2/Bid/p53/caspases, and LC3/Beclin-1/CHOP/ATG6, while also upregulating protective signaling pathways like Sirt1/PPAR-γ/PGC-1α/FOXO-3 and Nrf2/HO-1/ARE. However, evidence regarding the roles of lncRNA and the Wnt/β-catenin pathway in ACT toxicity remains inconsistent, and the impact of TEs and NPs on ACT efficacy is not fully understood. Further research is needed to confirm the protective effects of TEs and their NPs against ACT toxicity in cancer patients. In summary, TEs and their NPs present a promising avenue as adjuvant agents for preventing non-target organ toxicity induced by ACT.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike Ikwo, Abakaliki, Ebonyi, Nigeria.
- Centre for Natural Products Discovery, School of P harmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Cletus A Ukwubile
- Department of Pharmacognosy, Faculty of Pharmacy, University of Maiduguri, Bama Road, Maiduguri, Borno, Nigeria
| | - Sachindra Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Mehta V Kamal
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Vijetha S Belle
- Department of Biochemistry, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Eman M Othman
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
- Cancer Therapy Research Center, Department of Biochemistry-I, Biocenter, University of Würzburg, Am Hubland, 97074, Würzburg, Germany
- Department of Bioinformatics, University of Würzburg, Am Hubland, 97074, BiocenterWürzburg, Germany
| | - Sreedhara Ranganath K Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Science, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| |
Collapse
|
30
|
Chen X, Song Y, Tian Y, Dong X, Chang Y, Wang W. miR-149-3p Enhances Drug Sensitivity of AML Cells by Inhibiting Warburg Effect Through PI3K/AKT Pathway. Cell Biochem Biophys 2024; 82:3287-3296. [PMID: 39154128 DOI: 10.1007/s12013-024-01412-8] [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: 07/04/2024] [Indexed: 08/19/2024]
Abstract
Acute myeloid leukemia (AML) is a kind of heterogeneous hematologic malignancy with high incidence, which is usually treated by intensive and maintenance treatment with large dose of conventional chemotherapy drugs. However, cell resistance is still an unsolved problem. The abnormal expression of miRNAs is closely related to the pathogenesis and progression of AML, and affects the drug resistance of cancer cells. miR-149-3p plays an important role in the resistance of cancer cells to cisplatin, and plays an excellent anti-tumor activity. By studying the function of miR-149-3p, it is expected to find new therapeutic methods to reverse chemotherapy resistance. In order to explore the mechanism of action of miR-149-3p on AML chemotherapeutic drug sensitivity, we explored the relationship between the Warburg effect and AML chemotherapeutic drug resistance. Based on AML cells, transfection of miR-149-3p inhibitor/NC and Warburg effect inhibitor (2DG) and PI3K/AKT pathway inhibitor (LY294002) were used to investigate the mechanism of IFN-γ regulating chemotherapy resistance of AML cells through Warburg effect. Down-regulation of miR-149-3p significantly inhibited drug sensitivity of AML cells. Down-regulation of miR-149-3p significantly promoted proliferation and invasion of AML cells while inhibiting apoptosis by up-regulating the expression of Bcl-2 and down-regulating the expression of Bax. Down-regulation of miR-149-3p significantly promoted the expression of Warburg effect-related proteins hexokinase 2 (HK2), lactate dehydrogenase A (LDHA), and Glucose transporter 1 (GLUT1), glucose consumption, lactic acid, and intracellular ATP production. After inhibiting the Warburg effect with 2DG, the effect of miR-149-3p was inhibited, suggesting that upregulation of miR-149-3p reversed AML cell resistance by inhibiting the Warburg effect. In addition, miR-149-3p interacted with AKT1. Down-regulation of miR-149-3p increased the expression of inosine phosphate 3 kinase (PI3K), protein kinase B (AKT), and multi-drug resistance protein (MDR1). LY294002 inhibited the expression of these proteins, and down-regulation of miR-149-3p reversed the effect of LY294002 and improved the drug resistance of cells. Upregulation of miR-149-3p expression may potentially be a therapeutic target for AML resistance. It has been shown to inhibit PI3K/AKT pathway activation, thereby inhibiting the Warburg effect, and affecting cell proliferation, apoptosis, and drug resistance.
Collapse
MESH Headings
- MicroRNAs/metabolism
- MicroRNAs/genetics
- Humans
- Proto-Oncogene Proteins c-akt/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Signal Transduction/drug effects
- Drug Resistance, Neoplasm/drug effects
- Cell Line, Tumor
- Apoptosis/drug effects
- Antineoplastic Agents/pharmacology
- Cisplatin/pharmacology
- Warburg Effect, Oncologic/drug effects
- Morpholines/pharmacology
- Hexokinase/metabolism
- Hexokinase/genetics
- Chromones/pharmacology
- HL-60 Cells
Collapse
Affiliation(s)
- Xi Chen
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Song
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yaoyao Tian
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiushuai Dong
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuying Chang
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Wang
- Department of Hemotology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
31
|
Zhang M, Zhang J, Ma Y, Jin Y, Li Y, Wu X. Nephropathy induced by cisplatin results from mitochondrial disruption, impaired energy metabolism, altered expression of renal transporters, and accumulation of urinary toxins. J Trace Elem Med Biol 2024; 86:127553. [PMID: 39427559 DOI: 10.1016/j.jtemb.2024.127553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 09/09/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND The administration of platinum-based drugs such as cisplatin and its derivatives, which are frequently used during clinical chemotherapy, is highly restricted due to the incidence of nephrotoxicity. The present study focused on investigating cisplatin-induced nephrotoxicity from the perspective of energy metabolism, renal transporter expression and urinary toxin accumulation. METHODS This study investigated cisplatin's toxic effects, including nephrotoxicity, cardiotoxicity, hepatotoxicity, pulmonary toxicity, and splenotoxicity. We used transmission electron microscopy (TEM) and scanning electron microscopy (SEM) to characterize the accumulation of cisplatin in the kidney and the structure of renal mitochondria. The production of reactive oxygen species (ROS) induced by cisplatin in renal tubular epithelial cells was evaluated by in vitro experiments, and apoptosis of renal tubular epithelial cells and alterations to the renal microvasculature were assessed. Metabolites associated with the glycolytic and tricarboxylic acid pathways were measured, and renal transporters expression, autophagy, and urinary toxins (UTs) accumulation were also assessed. RESULTS Our results reveal that cisplatin-induced varying degrees of damage to the heart, liver, spleen, lungs, and kidneys, including inflammatory and fibrotic damage. Accumulation of cisplatin in renal mitochondria disrupted mitochondrial structure and mitochondrial function, as evidenced by decreased levels of glucose 6-phosphate and ribose 5-phosphate and elevated levels of isocitric acid. Cisplatin-induced accumulation of ROS in renal tubular epithelial cells led to apoptosis and, ultimately, constriction or loss of renal microvasculature. Furthermore, dysregulation of renal transporter expression, activation of autophagy and increased accumulation of UTs was observed. CONCLUSION Accumulation of cisplatin in the kidney led to damage to mitochondrial structure and function, apoptosis of renal tubular epithelial cells, constriction or loss of renal microvasculature, dysfunction of renal transporters, activation of autophagy, and accumulation of UTs.
Collapse
Affiliation(s)
- Mingkang Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Jianping Zhang
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yanrong Ma
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yongwen Jin
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Yile Li
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China
| | - Xin'an Wu
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, Gansu 730000, China; Engineering Research Centre of Prevention and Control for Clinical Medication Risk, Lanzhou, Gansu 730000, China.
| |
Collapse
|
32
|
Thapa R, Gupta S, Gupta G, Bhat AA, Smriti, Singla M, Ali H, Singh SK, Dua K, Kashyap MK. Epithelial-mesenchymal transition to mitigate age-related progression in lung cancer. Ageing Res Rev 2024; 102:102576. [PMID: 39515620 DOI: 10.1016/j.arr.2024.102576] [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: 09/05/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Epithelial-Mesenchymal Transition (EMT) is a fundamental biological process involved in embryonic development, wound healing, and cancer progression. In lung cancer, EMT is a key regulator of invasion and metastasis, significantly contributing to the fatal progression of the disease. Age-related factors such as cellular senescence, chronic inflammation, and epigenetic alterations exacerbate EMT, accelerating lung cancer development in the elderly. This review describes the complex mechanism among EMT and age-related pathways, highlighting key regulators such as TGF-β, WNT/β-catenin, NOTCH, and Hedgehog signalling. We also discuss the mechanisms by which oxidative stress, mediated through pathways involving NRF2 and ROS, telomere attrition, regulated by telomerase activity and shelterin complex, and immune system dysregulation, driven by alterations in cytokine profiles and immune cell senescence, upregulate or downregulate EMT induction. Additionally, we highlighted pathways of transcription such as SNAIL, TWIST, ZEB, SIRT1, TP53, NF-κB, and miRNAs regulating these processes. Understanding these mechanisms, we highlight potential therapeutic interventions targeting these critical molecules and pathways.
Collapse
Affiliation(s)
- Riya Thapa
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gupta
- Chameli Devi Institute of Pharmacy, Department of Pharmacology, Indore, Madhya Pradesh, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome-Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| | - Asif Ahmad Bhat
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Smriti
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Madhav Singla
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| | - Manoj Kumar Kashyap
- Molecular Oncology Laboratory, Amity Stem Cell Institute, Amity Medical School, Amity University Haryana, Panchgaon (Manesar), Gurugram, Haryana, India.
| |
Collapse
|
33
|
Ortiz N, Díaz C. Preclinical evaluation of fenretinide against primary and metastatic intestinal type‑gastric cancer. Oncol Lett 2024; 28:561. [PMID: 39372665 PMCID: PMC11450695 DOI: 10.3892/ol.2024.14694] [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: 05/15/2024] [Accepted: 08/20/2024] [Indexed: 10/08/2024] Open
Abstract
In recent years there has been a decline in the incidence of gastric cancer, however the high mortality rate has remained constant. The present study evaluated the potential effects of the retinoid fenretinide on the viability and migration of two cell lines, AGS and NCI-N87, that represented primary and metastatic intestinal gastric cancer subtypes, respectively. It was determined that a similar2 dose of fenretinide reduced the viability of both the primary and metastatic cell lines. In addition, it was demonstrated that combined treatment with fenretinide and cisplatin may affect the viability of both primary and metastatic gastric cancer cells. Furthermore, a wound healing assay demonstrated an inhibitory effect for fenretinide on cell migration. As part of the characterization of the mechanism of action, the effect of fenretinide on reactive oxygen species production and lipid droplet content was evaluated, with the latter as an indirect means of assessing autophagy. These results support the hypothesis of combining using fenretinide with conventional therapies to improve survival rates in advanced or metastatic gastric cancer.
Collapse
Affiliation(s)
- Natalia Ortiz
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica
| | - Cecilia Díaz
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica
- Institute Clodomiro Picado, Faculty of Microbiology, University of Costa Rica, San Pedro de Montes de Oca, San José 11501-2060, Costa Rica
| |
Collapse
|
34
|
Chen Y, Li H, Liu J, Ni J, Deng Q, He H, Wu P, Wan Y, Seeram NP, Liu C, Ma H, Zhu W. Cytotoxicity of natural and synthetic cannabinoids and their synergistic antiproliferative effects with cisplatin in human ovarian cancer cells. Front Pharmacol 2024; 15:1496131. [PMID: 39660007 PMCID: PMC11629013 DOI: 10.3389/fphar.2024.1496131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Cannabinoids are reported to suppress the growth of ovarian cancer cells, but it is unclear whether structural modifications can improve their cytotoxic effects. Methods Herein, an investigation into the antiproliferative effects of natural cannabinoids on human ovarian cancer Caov-3 cells identified cannabidiol (CBD) as the most promising cannabinoid. Furthermore, chemical modifications of CBD yielded a group of derivatives with enhanced cytotoxicity in Caov-3 cells. Results Two CBD piperazinyl derivatives (19 and 21) showed augmented antiproliferative effects with an IC50 of 5.5 and 4.1 µM, respectively, compared to CBD's IC50 of 22.9 µM. Further studies suggest that modulation of apoptosis and ferroptosis may contribute to the cytotoxic effects of CBD and its derivatives. In addition, CBD and its derivatives (19 and 21) were explored for their potential synergistic antiproliferative effects in combination with chemotherapeutic agent cisplatin. Compounds 19 or 21 (5 µM) combined with cisplatin (1 µM) showed a synergistic effect with a combination index of 0.23 and 0.72, respectively. This effect was supported by elevated levels of reactive oxygen species in Caov-3 cells treated with cisplatin combined with 19 or 21. Discussion Findings from this study suggest that CBD derivatives with enhanced antiproliferative effects may exert synergistic effects with chemotherapeutic drugs, providing insight into the development of cannabinoid-based adjuvant agents for the management of ovarian cancer.
Collapse
Affiliation(s)
- Ying Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Huifang Li
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Jia Liu
- Department of Operation Room, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jie Ni
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qicheng Deng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Haotian He
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Panpan Wu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Yinsheng Wan
- Department of Biology, Providence College, Providence, RI, United States
| | - Navindra P. Seeram
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Chang Liu
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Hang Ma
- Bioactive Botanical Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Weipei Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
35
|
Li N, Zhu X, Zhang H, Yang X, Shao M, Cui S, Lin C. Exploring the Target Genes of Fucosylated Chondroitin Sulfate in Treating Lung Adenocarcinoma Based on the Integration of Bioinformatics Analysis, Molecular Docking, and Experimental Verification. ACS OMEGA 2024; 9:46312-46322. [PMID: 39583738 PMCID: PMC11579779 DOI: 10.1021/acsomega.4c07295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
Fucosylated chondroitin sulfate (FCS), extracted from sea cucumbers' body walls, has been found to inhibit the proliferation of lung adenocarcinoma (LUAD) cells. However, there have been few studies of the associated drug targets. This study combined bioinformatics analysis and molecular docking to screen the main targets of FCS intervention in LUAD. Moreover, an experimental validation was performed. First, we downloaded the LUAD gene data set from The Cancer Genome Atlas (TCGA) database and the cisplatin (DDP) resistance gene data set of LUAD A549 cells from the Gene Expression Omnibus (GEO) database. Nine significant genes (PLK1, BUB1, CDK1, CDC20, CCNB1, BUB1B, KIF11, CCNB2, and DLAGP5) were identified by bioinformatics analysis, and these nine genes overlapped in both data sets. Then, molecular docking results showed that FCS had a better affinity with target proteins BUB1 and PLK1. Further experimental verification revealed that FCS inhibited the growth of A549 cells and increased the sensitivity of A549 cells to DDP. Quantitative real-time polymerase chain reaction (qRT-PCR) revealed that A549 cells treated with FCS exhibited down-regulated BUB1 and PLK1 mRNA expression. At the same time, FCS+DDP treatment resulted in a more significant reduction in BUB1 and PLK1 mRNA expression than DDP or FCS treatment alone. These findings reveal potential targets of FCS for LUAD and provide clues for the development of FCS as a potential anticancer agent.
Collapse
Affiliation(s)
- Nana Li
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xinhong Zhu
- Department
of International Medicine, Qingdao Municipal
Hospital Group, Qingdao 266071, China
| | - Hua Zhang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaohui Yang
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Mingju Shao
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Shichao Cui
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Cunzhi Lin
- Department
of Respiratory and Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
36
|
Moar K, Brahma M, Pant A, Maruthi M, Maurya PK. Synergistic anticancer activity of resveratrol with cisplatin and carboplatin in A549 lung adenocarcinoma cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2024; 17:411-420. [PMID: 39660333 PMCID: PMC11626289 DOI: 10.62347/rysq1416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/13/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND This study looked at the efficacy of combining the phytochemical resveratrol with the anticancer drugs cisplatin and carboplatin on lung adenocarcinoma cell lines. MATERIALS AND METHODS We used MTT assay and generation of Reactive Oxygen Species levels using molecular fluorogenic probe 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) to investigate the effects of resveratrol in combination with cisplatin and carboplatin on the proliferation and viability of cells and levels of reactive oxygen species (ROS). RESULTS Resveratrol has an anti-proliferative effect on A549 lung cancer cells, inhibiting cell proliferation in a dose and time-dependent manner. Resveratrol in conjunction with cisplatin and carboplatin inhibited cell proliferation synergistically. The combination therapy of cisplatin and carboplatin with Resveratrol showed enhanced growth inhibition of lung cancer cells in in-vitro with IC50 values of 15.09 ± 0.71 µM and IC50 values of 21.72 ± 1.9 µM, respectively. The present investigation also revealed the significant dose-dependent ROS generation in A549 cells by cisplatin, carboplatin, and their combination with resveratrol. Carboplatin treatment in combination with Resveratrol induced a higher generation of ROS (3.4-fold) when compared to carboplatin treatment (2.4-fold) at the highest concentration. CONCLUSIONS Our findings offered a basis for further research for assessing the potential of Resveratrol as a therapeutic agent to treat lung adenocarcinoma and whether it can be used as an adjuvant with drugs like cisplatin and carboplatin for improving their efficacies. However, the underlying processes of cell inhibition and cell death should be thoroughly investigated.
Collapse
Affiliation(s)
| | | | - Anuja Pant
- Department of Biochemistry, Central University of HaryanaMahendergarh 123031, India
| | | | | |
Collapse
|
37
|
Li B, Tan S, Yu X, Wang Y. Bufalin: A promising therapeutic drug against the cisplatin-resistance of ovarian cancer by targeting the USP36/c-Myc axis. Biochem Biophys Res Commun 2024; 733:150440. [PMID: 39067250 DOI: 10.1016/j.bbrc.2024.150440] [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: 06/11/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
Cisplatin (DPP) resistance is a severe obstacle to ovarian cancer (OC) treatment. Our research aims to uncover the therapeutic effect and the underlying mechanism of Bufalin against DDP resistance. The cell viability, proliferation capacity, γH2AX expression, and apoptosis ratio were quantified via CCK8 assay, colony formation assay, immunofluorescence, and flow cytometry analysis respectively. Xenografting experiment was performed to detect the tumor growth. Molecular docking was applied to mimic the combination of Bufalin and USP36 protein, and Western blotting was conducted to measure the Bax, Bcl-2, γH2AX, USP36, and c-Myc expression. The c-Myc ubiquitination and half-life were detected via ubiquitination assay and cycloheximide chasing assay. Bufalin treatment notably suppressed the cell viability and colony numbers, and increased the apoptosis ratio and γH2AX level in the DDP treatment group. Bufalin therapy also notably inhibited tumor growth, Bax, Bcl-2, and γH2AX expression in vivo. Moreover, the Bufalin application remarkedly reduced the c-Myc expression and half-life and increased the c-Myc ubiquitination via interaction and subsequent down-regulation of USP36. Knockdown of USP36 reversed the antiproliferative effect and proapoptotic capacity of Bufalin therapy in the DDP treatment group. In conclusion, Bufalin can overcome the DDP resistance in vitro and in vivo via the USP36/c-Myc axis, which innovatively suggests the therapeutic potential of Bufalin against DDP resistance ovarian cancer.
Collapse
Affiliation(s)
- Bing Li
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Shu Tan
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Xi Yu
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Yan Wang
- Department of Gynaecology, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| |
Collapse
|
38
|
Colliva C, Rivi V, Sarti P, Cobelli I, Blom JMC. Exploring Sex-Based Neuropsychological Outcomes in Pediatric Brain Cancer Survivors: A Pilot Study. Diseases 2024; 12:289. [PMID: 39589963 PMCID: PMC11592787 DOI: 10.3390/diseases12110289] [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: 09/06/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The increasing survival rates among pediatric cancer patients underscore the critical need to understand the long-term psychosocial impacts of cancer treatments, such as cisplatin and carboplatin. While these treatments are lifesaving, they may pose risks to neurodevelopmental processes. Despite the substantial body of research highlighting cognitive impairments associated with cancer treatments, there remains a gap in understanding how these effects differ by sex. As sex differences could inform tailored interventions and support mechanisms for affected individuals, this pilot study aimed to examine the sex differences in neuropsychological outcomes in patients treated for brain cancer with cisplatin and/or carboplatin. METHODS Our study employed rigorous/structured neuropsychological assessments to evaluate executive functions in pediatric cancer survivors treated with cisplatin and/or carboplatin. We utilized the BRIEF and TOL tests to assess the key domains of executive function, including inhibitory control, cognitive flexibility, and problem-solving abilities. Additionally, psychosocial factors were evaluated using the Resiliency Scale to measure resilience and the PAT test to assess family psychosocial risk. RESULTS In our cohort of 17 patients, significant sex differences emerged, where males outperformed females in areas such as inhibitory control, impulse regulation, and strategic planning. CONCLUSIONS These findings highlight the complexity of cognitive outcomes in pediatric cancer survivors. Understanding sex-specific differences is essential for developing tailored interventions that optimize cognitive and psychosocial outcomes. Future research should focus on larger cohorts and longitudinal studies to validate these findings and guide targeted interventions to improve survivorship outcomes.
Collapse
Affiliation(s)
- Chiara Colliva
- Local Health Unit of Modena, District of Carpi, 41012 Carpi, Italy;
| | - Veronica Rivi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (P.S.); (I.C.)
| | - Pierfrancesco Sarti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (P.S.); (I.C.)
- Department of Adult Psychiatry and Psychotherapy, Psychiatric Hospital, University of Zurich, 8008 Zurich, Switzerland
| | - Isabel Cobelli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (P.S.); (I.C.)
| | - Johanna M. C. Blom
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (V.R.); (P.S.); (I.C.)
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
39
|
Huang T, He Y, Cheng R, Zhang Q, Zhong X, Hashimoto K, Liu Y, Pu Y. Ketamine attenuates kidney damage and depression-like behaviors in mice with cisplatin-induced acute kidney injury. Transl Psychiatry 2024; 14:468. [PMID: 39521765 PMCID: PMC11550419 DOI: 10.1038/s41398-024-03176-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Acute kidney injury (AKI) is a serious condition characterized by decreased urine output, often accompanied by psychiatric symptoms like depression. However, there are limited pharmacological treatments available for AKI and its associated depressive symptoms. In this study, we investigated whether cisplatin-induced AKI in mice leads to depression-like behaviors and whether ketamine could alleviate both the kidney injury and these behaviors. Mice with cisplatin-induced AKI exhibited elevated levels of creatinine and urea, kidney damage, increased kidney injury molecule-1 protein, and pathological changes in the liver, colon, and spleen. They also showed depression-like behaviors and reduced expression of synaptic proteins in the prefrontal cortex. Remarkably, a single dose of ketamine significantly reduced these symptoms and pathological changes. Interestingly, the beneficial effects of ketamine on the kidneys, other organs, and depression-like behaviors, were reversed by the tropomyosin receptor kinase B (TrkB) inhibitor ANA-12. Western blot analysis revealed the involvement of the TrkB and ERK (extracellular signal-regulated kinase)-CREB (cAMP response element binding protein) signaling pathway. Additionally, metabolomics analysis indicated that blood metabolites, such as C16-ceramide, may contribute to the effects of ketamine in this model. These findings suggest that cisplatin-induced nephrotoxicity in AKI mice contributes to depression-like behaviors, and ketamine can alleviate both kidney damage and depression-like symptoms by modulating the TrkB and ERK-CREB signaling pathways, as well as altering blood metabolites. However, the role of the kidney-brain axis in these depression-like behaviors remains unclear. Furthermore, ketamine may have therapeutic potential for treating kidney diseases such as AKI, along with associated depressive symptoms.
Collapse
Affiliation(s)
- Tianwen Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yangyang He
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Ruijuan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiang Zhong
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba, 260-8670, Japan.
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Yaoyu Pu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
40
|
Mitra A, Roy R, Paul S. Modulating the Self-Assembly of a Camptothecin Prodrug with Paclitaxel for Anticancer Combination Therapy: A Molecular Dynamics Approach. J Phys Chem B 2024; 128:10799-10812. [PMID: 39230512 DOI: 10.1021/acs.jpcb.4c04798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Camptothecin (CPT) and paclitaxel (PTX), derived from natural products, are recognized for their significant efficacy in clinical cancer treatments. Despite its therapeutic advantages, CPT is challenged by issues of toxicity and solubility, necessitating its use in conjugation with other compounds for enhanced compatibility. This study delves into the coassembly mechanism of Evans blue-conjugated camptothecin (EB-CPT) with PTX, aiming to elucidate their synergistic potential in combination therapy applications, employing all-atom molecular dynamics simulations. The EB-CPT prodrug is reported to form a self-aggregated cluster. Our findings suggest that increasing the PTX concentration induces a dispersion of EB-CPT clusters, thereby disrupting their inherent self-assembly. This disruption is explained to be facilitated by the coassembly of EB-CPT and PTX. With increasing concentration of PTX, a lengthening of the coassembled structures is observed, supporting the experimental findings of tube-like coassembled structures at higher weight ratios of PTX. Hydrophobic interactions and π-π stacking are the primary forces responsible for the formation of both self- and coassembled structures. Interestingly, the structural analysis reveals that the CPT moiety of EB-CPT is less involved in assemblies due to steric hindrances. Instead, the interaction and coassembly processes are predominantly mediated by the EB derivative component of the prodrug. This research underscores the critical role of the solubilizing agent, EB derivative, in mediating the flexibility and interaction of CPT in combination therapy strategies, particularly with PTX, thus emphasizing the importance of conjugates for therapeutic developments. Furthermore, the molecular insights into the interaction sites and mechanisms facilitating coassembly between EB-CPT and PTX contribute valuable knowledge to the field, highlighting the potential of these nanomedicine combinations in advancing cancer treatment modalities.
Collapse
Affiliation(s)
- Anandita Mitra
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati, Assam 781039, India
| |
Collapse
|
41
|
Milutinović M, Ćujić Nikolić N, Cvetković T, Šavikin K, Djordjević I, Janković Veličković L, Randjelović M, Miladinović B, Branković S, Kitić D. Chokeberry Products and By-Products as the Potential Pharmaceuticals for Kidney Protection-An Experimental Study in Rats. PLANTS (BASEL, SWITZERLAND) 2024; 13:3136. [PMID: 39599345 PMCID: PMC11597244 DOI: 10.3390/plants13223136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/27/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
The study aimed to investigate the protective effects of chokeberry fruit products and by-products against cisplatin-induced acute nephrotoxicity in rats. Potential mechanisms involving oxidative stress and inflammatory responses were examined through biochemical and histopathological analyses of kidney tissue. Chokeberry waste, along with the whole fruit extract and juice, was evaluated as a potential raw material for pharmaceutical use. The chemical composition of chokeberry juice and extracts was analyzed using spectrophotometry and HPLC. Rats were treated with chokeberry preparations via intragastric tube for ten days, with a single intraperitoneal dose of cisplatin (8 mg/kg BW) administered on the third day. Post-sacrifice, plasma samples were analyzed for biochemical nephrotoxicity markers, oxidative stress, and inflammatory markers. Kidneys were removed for histopathological and biochemical analysis. Cisplatin-induced acute nephrotoxicity was confirmed by elevated plasma creatinine and blood urea nitrogen levels. Additionally, lipid peroxidation was significantly elevated, while reduced glutathione and catalase activity were significantly reduced. Pro-inflammatory mediators IL-1β, TNF-α, and IL-6 levels were significantly increased in the cisplatin group. Treatment with chokeberry extracts and juice significantly mitigated these nephrotoxic effects, as confirmed by histopathological examination and biochemical marker analysis. Notably, the waste extract demonstrated greater efficacy than the whole fruit extract, likely due to its higher concentration of polyphenolic compounds, especially anthocyanins. These results highlight the potential of chokeberry as a therapeutic and preventive agent for kidney protection, emphasizing the value of by-products rich in biologically active compounds.
Collapse
Affiliation(s)
- Milica Milutinović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Nada Ćujić Nikolić
- Institute of Medicinal Plants Research, Dr Josif Pančić, Tadeuša Koščuška 1, 11000 Beograd, Serbia; (N.Ć.N.); (K.Š.)
| | - Tatjana Cvetković
- Institute of Biochemistry, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia;
- Centre of Medical and Clinical Biochemistry, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia
| | - Katarina Šavikin
- Institute of Medicinal Plants Research, Dr Josif Pančić, Tadeuša Koščuška 1, 11000 Beograd, Serbia; (N.Ć.N.); (K.Š.)
| | - Ivana Djordjević
- Pathology and Pathological Anatomy Center, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia; (I.D.); (L.J.V.)
| | - Ljubinka Janković Veličković
- Pathology and Pathological Anatomy Center, University Clinical Centre, Bul. Dr Zorana Đinđića 48, 18000 Niš, Serbia; (I.D.); (L.J.V.)
| | - Milica Randjelović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Bojana Miladinović
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| | - Suzana Branković
- Department of Physiology, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia;
| | - Dušanka Kitić
- Department of Pharmacy, Faculty of Medicine, University of Niš, Bul. Dr Zorana Đinđića 81, 18000 Niš, Serbia; (M.R.); (B.M.)
| |
Collapse
|
42
|
Motie FM, Soltani Howyzeh M, Ghanbariasad A. Synergic effects of DL-limonene, R-limonene, and cisplatin on AKT, PI3K, and mTOR gene expression in MDA-MB-231 and 5637 cell lines. Int J Biol Macromol 2024; 280:136216. [PMID: 39362430 DOI: 10.1016/j.ijbiomac.2024.136216] [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: 09/23/2023] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
The anticancer and cytotoxic effects of DL-Limonene and R-Limonene are well-documented. However, the role of natural compounds in enhancing the efficacy of platinum-based drugs like Cisplatin (CisPt) remains debated. This study aims to boost Cisplatin's impact on breast (MDA-MB-231) and bladder (5637) cancer cells using DL-Limonene and R-Limonene. Different concentrations of DL-Limonene, R-Limonene, and Cisplatin, combined, were used to treat MDA-MB-231 and 5637 cells in this experimental study. The cell's viability was evaluated using an MTT assay. AnnexinV- PI staining was applied to evaluate the percentage of apoptotic cells. Cytotoxicity results showed that combining DL-Limonene, R-Limonene, and Cisplatin significantly improved outcomes in MDA-MB-231 cells (P < 0.05). Annexin/PI staining revealed apoptosis rates of 74 %, 28 %, 43 %, 81 %, and 91 % for Cisplatin40, R-Limonen1000, DL-Limonen1000, R-Limonen1000/DL-Limonen1000, and the combined treatment, respectively, versus 13 % in the control. The combination also resulted in the greatest reduction of AKT, PI3K, and mTOR gene expression. Our results show that R-Limonene and DL-Limonene enhance Cisplatin's cancer-inhibiting effects in breast and bladder cancer cell lines. These compounds may be promising for combination therapy, potentially allowing for lower doses of chemotherapy and reducing side effects like nephrotoxicity.
Collapse
Affiliation(s)
- Fatemeh Malek Motie
- Department of Genetics, Khuzestan Science and Research Branch, Islamic Azad University, Ahvaz, Iran; Department of Genetics, Ahvaz Branch, Islamic Azad University, Ahvaz, Iran
| | - Mehdi Soltani Howyzeh
- Department of Genetics and Plant Breeding, Ahvaz Branch, Islamic Azad University, Ahvaz, Iran.
| | - Ali Ghanbariasad
- Department of Medical Biotechnology, School of Advanced Technologies, Fasa University of Medical Sciences, Iran.
| |
Collapse
|
43
|
Ciftel E, Mercantepe F, Mercantepe T, Akyildiz K, Yilmaz A, Ciftel S. Comparative Analysis of Epigallocatechin-3-Gallate and TNF-Alpha Inhibitors in Mitigating Cisplatin-Induced Pancreatic Damage Through Oxidative Stress and Apoptosis Pathways. Biol Trace Elem Res 2024; 202:5190-5207. [PMID: 38776022 PMCID: PMC11442533 DOI: 10.1007/s12011-024-04239-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/15/2024] [Indexed: 10/01/2024]
Abstract
Oxidative stress and inflammation caused by cisplatin, which is frequently used in the treatment of many cancers, damage healthy tissues as well as cancer cells. In this study, we aimed to investigate the effect of epigallocatechin-3-gallate (EGCG) and infliximab (INF) administration on pancreatic endocrine cells in rats treated with systemic cisplatin (CDDP). The rats were randomly divided into 6 groups: group 1 (control group), group 2 (EGCG group), group 3 (CDDP group), group 4 (EGCG + CDDP group), group 5 (CDDP + INF group), and group 6 (EGCG + CDDP + INF group). The study's findings demonstrated that EGCG and INF effectively reduced the cellular damage induced by CDDP in histopathologic investigations of the pancreas. EGCG and INF, whether used individually or in combination, demonstrated a significant reduction in malondialdehyde (MDA) levels and an increase in glutathione (GSH) levels in the rat pancreas compared to the CDDP group. Immunohistochemically, the enhanced presence of insulin and glucagon positivity in the EGCG and INF groups, along with the absence of TUNEL immunopositivity, indicate that both treatments reduced CDDP-induced apoptosis. Furthermore, the observed lack of immunopositivity in TNF-α and 8-OHdG in the groups treated with EGCG and INF, compared to those treated with CDDP, indicates that these substances can inhibit inflammation. EGCG and INF, whether provided alone or together, can potentially reduce the damage caused to pancreatic islet cells by cisplatin. This effect is achieved through their anti-inflammatory and antioxidant properties during the early stages of the condition.
Collapse
Affiliation(s)
- Enver Ciftel
- Department of Endocrinology and Metabolism, Sivas Numune Hospital, Sivas, Turkey
| | - Filiz Mercantepe
- Department of Endocrinology and Metabolism, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, 53010, Turkey.
| | - Tolga Mercantepe
- Department of Histology and Embryology, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Kerimali Akyildiz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Adnan Yilmaz
- Department of Biochemistry, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| | - Serpil Ciftel
- Department of Endocrinology and Metabolism, Erzurum Education and Research Hospital, Erzurum, Turkey
| |
Collapse
|
44
|
Bhat AA, Gupta G, Afzal M, Thapa R, Ali H, Alqahtani SM, almalki WH, Kazmi I, Alzarea SI, Saleem S, Subramaniyan V. Polyphenol-Loaded Nano-carriers for Breast Cancer Therapy: A Comprehensive Review. BIONANOSCIENCE 2024; 14:4219-4237. [DOI: 10.1007/s12668-023-01288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 11/22/2024]
|
45
|
Adewale OO, Oyelola RF, Adetuyi OA, Adebisi OA, Adekomi DA, Oladele JO. Water-soluble phenolics from Phoenix dactylifera fruits as potential reno-protective agent against cisplatin-induced toxicity: pre- and post-treatment strategies. Drug Chem Toxicol 2024; 47:1058-1071. [PMID: 38529813 DOI: 10.1080/01480545.2024.2329762] [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: 12/14/2023] [Accepted: 03/07/2024] [Indexed: 03/27/2024]
Abstract
Nephrotoxicity is the major side effect of cisplatin, an effective platinum-based chemotherapeutic drug that is applicable in the treatment of several solid-tissue cancers. Studies have indicated that certain water-soluble phenolics offer renal protection. Thus, this study investigates the role of pre and post-treatment of rats with water-soluble phenolics from Phoenix dactylifera (PdP) against nephrotoxicity induced by cisplatin. Rats were either orally pretreated or post-treated with 200 mg/kg body weight of PdP before or after exposure to a single therapeutic dose of cisplatin (5 mg/kg body weight) for 7 successive days intraperitoneally. The protective effects of PdP against Cisplatin-induced nephrotoxicity was based on the evaluation of various biochemical and redox biomarkers, together with histopathological examination of kidney tissues. The composition, structural features, and antioxidative influence of PdP were determined based on chromatographic, spectroscopic, and in vitro antioxidative models. Cisplatin single exposure led to a substantial increase in the tested renal function biomarkers (uric acid, creatinine, and urea levels), associated with an increase in malondialdehyde indicating lipid peroxidation and a significant decline (p < 0.05) in reduced glutathione (GSH) levels in the renal tissue when compared with the control group. A marked decline exists in the kidney antioxidant enzymes (catalase, SOD, and GPx). Nevertheless, treatment with PdP significantly suppressed the heightened renal function markers, lipid peroxidation, and oxidative stress. Spectroscopic analysis revealed significant medicinal phenolics, and in vitro tests demonstrated antioxidative properties. Taken together, results from this study indicate that pre- and/or post-treatment strategies of PdP could serve therapeutic purposes in cisplatin-induced renal damage.
Collapse
Affiliation(s)
| | | | - Oluwatosin Adefunke Adetuyi
- Department of Biochemistry, Osun State University, Osogbo, Nigeria
- Department of Chemistry and Biochemistry, Wichita State University, Wichita, KS, USA
| | - Oluwaseun Abraham Adebisi
- Department of Biochemistry, Osun State University, Osogbo, Nigeria
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Damilare Adedayo Adekomi
- Department of Anatomy, Faculty of Basic Medical Sciences, Osun State University, Osogbo, Nigeria
| | - Johnson Olaleye Oladele
- Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
- Phytochemical research unit, Royal Scientific Research Institute, Osogbo, Nigeria
| |
Collapse
|
46
|
Sianipar NF, Muflikhati Z, Mangindaan D, Assidqi K. Anticancer Potential of Tocopherols-Containing Plants and Semi-Synthetic Tocopherols. PLANTS (BASEL, SWITZERLAND) 2024; 13:2994. [PMID: 39519912 PMCID: PMC11548436 DOI: 10.3390/plants13212994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 09/23/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Tocopherols, potent bioactive compounds with anticancer properties, remain understudied in herbal medicinal plants, presenting a significant knowledge gap in the field of natural anticancer agents. This review evaluates tocopherol-containing plants for their anticancer potential, analyzing Scopus publications from 2016 to 2024. Fifteen herbal medicinal plants were identified as promising candidates, including Bulbine anguistifolia Poelln, Punica granatum L., Moringa oleifera, Kigelia pinnata, and Typhonium flagelliforme Lodd. The review explores tocopherols' anticancer mechanisms, including apoptosis induction and cell cycle arrest. Factors influencing tocopherols' anticancer effects are examined, such as their forms (α, β, γ, δ), concentrations, plant parts utilized, and their stability in various plants. Additionally, emerging research on semi-synthetic tocopherol derivatives is analyzed, highlighting their potential as adjuvants in chemotherapy and their role in enhancing drug delivery and reducing side effects. This comprehensive analysis aims to advance the development of plant-based anticancer pharmaceuticals and improve cancer treatment strategies. By elucidating the mechanisms and potential of tocopherol-containing plants, this review provides a foundation for future research in plant-based anticancer therapies. It emphasizes the need for further investigation into these plants' anticancer properties, potentially leading to novel, more effective, and less toxic cancer therapies. The findings presented here contribute to a nuanced understanding of how tocopherol-containing plants can be leveraged in the development of future anticancer drugs.
Collapse
Affiliation(s)
- Nesti Fronika Sianipar
- Biotechnology Department, Faculty of Engineering, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia;
- Food Biotechnology Research Center, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia;
| | - Zidni Muflikhati
- Food Biotechnology Research Center, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia;
| | - Dave Mangindaan
- Waste-Food-Environmental Nexus Research Interest Group, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia
- Civil Engineering Department, Faculty of Engineering, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia
- Professional Engineering Program Department, Faculty of Engineering, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia
| | - Khoirunnisa Assidqi
- Biotechnology Department, Faculty of Engineering, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia;
- Food Biotechnology Research Center, Bina Nusantara University, Jl. KH Syahdan No. 9, Jakarta 11480, Indonesia;
| |
Collapse
|
47
|
Lin JG, Jiang WP, Tsai YS, Lin SW, Chen YL, Chen CC, Huang GJ. Dietary Probiotic Pediococcus acidilactici GKA4, Dead Probiotic GKA4, and Postbiotic GKA4 Improves Cisplatin-Induced AKI by Autophagy and Endoplasmic Reticulum Stress and Organic Ion Transporters. Nutrients 2024; 16:3532. [PMID: 39458526 PMCID: PMC11510457 DOI: 10.3390/nu16203532] [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: 08/07/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Acute kidney injury (AKI) syndrome is distinguished by a quick decline in renal excretory capacity and usually diagnosed by the presence of elevated nitrogen metabolism end products and/or diminished urine output. AKI frequently occurs in hospital patients, and there are no existing specific treatments available to diminish its occurrence or expedite recovery. For an extended period in the food industry, Pediococcus acidilactici has been distinguished by its robust bacteriocin production, effectively inhibiting pathogen growth during fermentation and storage. METHODS In this study, the aim is to assess the effectiveness of P. acidilactici GKA4, dead probiotic GKA4, and postbiotic GKA4 against cisplatin-induced AKI in an animal model. The experimental protocol involves a ten-day oral administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 to mice, with a cisplatin intraperitoneal injection being given on the seventh day to induce AKI. RESULTS The findings indicated the significant alleviation of the renal histopathological changes and serum biomarkers of GKA4, dead probiotic GKA4, and postbiotic GKA4 in cisplatin-induced nephrotoxicity. GKA4, dead probiotic GKA4, and postbiotic GKA4 elevated the expression levels of HO-1 and decreased the expression levels of Nrf-2 proteins. In addition, the administration of GKA4, dead probiotic GKA4, and postbiotic GKA4 significantly reduced the expression of apoptosis-related proteins (Bax, Bcl-2, and caspase 3), autophagy-related proteins (LC3B, p62, and Beclin1), and endoplasmic reticulum (ER) stress-related proteins (GRP78, PERK, ATF-6, IRE1, CHOP, and Caspase 12) in kidney tissues. Notably, GKA4, dead probiotic GKA4, and postbiotic GKA4 also upregulated the levels of proteins related to organic anion transporters and organic cation transporters. CONCLUSIONS Overall, the potential therapeutic benefits of GKA4, dead probiotic GKA4, and postbiotic GKA4 are significant, particularly after cisplatin treatment. This is achieved by modulating apoptosis, autophagy, ER stress, and transporter proteins to alleviate oxidative stress.
Collapse
Affiliation(s)
- Jaung-Geng Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan;
- Chinese Medicine Research Center, China Medical University, Taichung 404, Taiwan
| | - Wen-Ping Jiang
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, Taiwan;
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City 325, Taiwan (Y.-L.C.)
| | - Shih-Wei Lin
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City 325, Taiwan (Y.-L.C.)
| | - Yen-Lien Chen
- Biotech Research Institute, Grape King Bio Ltd., Taoyuan City 325, Taiwan (Y.-L.C.)
| | - Chin-Chu Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei 106, Taiwan;
- Department of Food Sciences, Nutrition, and Nutraceutical Biotechnology, Shih Chien University, Taipei 104, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan
| | - Guan-Jhong Huang
- Department of Food Nutrition and Healthy Biotechnology, Asia University, Taichung 413, Taiwan
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan
| |
Collapse
|
48
|
Nowotarska P, Janeczek M, Wiatrak B. Cytotoxic activity of Fomitopsis betulina against normal and cancer cells - a comprehensive literature review. Contemp Oncol (Pozn) 2024; 28:191-200. [PMID: 39512529 PMCID: PMC11538974 DOI: 10.5114/wo.2024.144223] [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: 04/24/2024] [Accepted: 09/20/2024] [Indexed: 11/15/2024] Open
Abstract
In recent years, there has been increasing interest in research on fungi, including Fomitopsis betulina, known for its potential therapeutic properties. The aim of this paper is to systematically review the literature on the cytotoxic effects of Fomitopsis betulina on normal and cancer cell lines. The review was conducted by searching PubMed, Google Scholar, and Web of Science databases up to July 2024, using specific MeSH terms and keywords related to cytotoxicity, cancer cells, and normal cells. Articles were included if they investigated both cancer and normal cell cultures, reported IC50 values, and used validated cytotoxicity assays (e.g. MTT, LDH). Out of 450 articles screened, 5 met the inclusion criteria. The analysed studies demonstrated that Fomitopsis betulina extracts, particularly methanolic and ethanolic, exhibited selective cytotoxicity against cancer cells while having minimal impact on healthy cells. The strongest effects were observed in prostate cancer and melanoma cell lines. The cytotoxic effects were largely attributed to bioactive compounds such as triterpenes and glucans. Fomitopsis betulina extracts, particularly those derived from fruiting bodies and mycelial cultures, show promising cytotoxic properties against cancer cells, with limited impact on healthy cells. However, further research is needed to fully understand the mechanisms of action and to optimise extraction methods.
Collapse
Affiliation(s)
- Paulina Nowotarska
- Department of Biostructure and Animal Physiology, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Maciej Janeczek
- Department of Biostructure and Animal Physiology, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Benita Wiatrak
- Department of Pharmacology, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
49
|
Zhao Z, Wu Q, Xu Y, Qin Y, Pan R, Meng Q, Li S. Groenlandicine enhances cisplatin sensitivity in cisplatin-resistant osteosarcoma cells through the BAX/Bcl-2/Caspase-9/Caspase-3 pathway. J Bone Oncol 2024; 48:100631. [PMID: 39263651 PMCID: PMC11388767 DOI: 10.1016/j.jbo.2024.100631] [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: 05/06/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024] Open
Abstract
Groenlandicine is a protoberberine alkaloid isolated from Coptidis Rhizoma, a widely used traditional Chinese medicine known for its various biological activities. This study aims to validate groenlandicine's effect on both cisplatin-sensitive and cisplatin-resistant osteosarcoma (OS) cells, along with exploring its potential molecular mechanism. The ligand-based virtual screening (LBVS) method and molecular docking were employed to screen drugs. CCK-8 and FCM were used to measure the effect of groenlandicine on the OS cells transfected by lentivirus with over-expression or low-expression of TOP1. Cell scratch assay, CCK-8, FCM, and the EdU assay were utilized to evaluate the effect of groenlandicine on cisplatin-resistant cells. WB, immunofluorescence, and PCR were conducted to measure the levels of TOP1, Bcl-2, BAX, Caspase-9, and Caspase-3. Additionally, a subcutaneous tumor model was established in nude mice to verify the efficacy of groenlandicine. Groenlandicine reduced the migration and proliferation while promoting apoptosis in OS cells, effectively damaging them. Meanwhile, groenlandicine exhibited weak cytotoxicity in 293T cells. Combination with cisplatin enhanced tumor-killing activity, markedly activating BAX, cleaved-Caspase-3, and cleaved-Caspase-9, while inhibiting the Bcl2 pathway in cisplatin-resistant OS cells. Moreover, the level of TOP1, elevated in cisplatin-resistant OS cells, was down-regulated by groenlandicine both in vitro and in vivo. Animal experiments confirmed that groenlandicine combined with cisplatin suppressed OS growth with lower nephrotoxicity. Groenlandicine induces apoptosis and enhances the sensitivity of drug-resistant OS cells to cisplatin via the BAX/Bcl-2/Caspase-9/Caspase-3 pathway. Groenlandicine inhibits OS cells growth by down-regulating TOP1 level.Therefore, groenlandicine holds promise as a potential agent for reversing cisplatin resistance in OS treatment.
Collapse
Affiliation(s)
- Zihao Zhao
- Clincal Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qihong Wu
- The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou Province, China
| | - Yangyang Xu
- Clincal Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yuhuan Qin
- Beijing Jinshuitan Hospital Guizhou Hospital, Guiyang, Guizhou Province, China
| | - Runsang Pan
- Basic Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qingqi Meng
- Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Siming Li
- Clincal Medical College, Guizhou Medical University, Guiyang, Guizhou Province, China
- Guangzhou Red Cross Hospital of Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
50
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|