1
|
Wang W, Wang R, An L, Li L, Xiong H, Li D, Dong F, Lei J, Wang M, Yang Z, Wang H, Ling X, Fountzilas C, Li F, Li Q. Design, synthesis and investigation of biological activity and mechanism of fluoroaryl-substituted derivatives at the FL118 position 7. Eur J Med Chem 2025; 283:117143. [PMID: 39647420 DOI: 10.1016/j.ejmech.2024.117143] [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: 10/14/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
Addition of fluorine atoms into chemical compounds is a validated strategy to enhance their physical, chemical and biological properties. In this study, FL118, a novel camptothecin-related small molecule known for its unique mechanism of action and superior antitumor efficacy, was utilized as a foundational drug platform. By replacing the hydrogen atom at position 7 of FL118 with a fluoroaryl group, a diverse array of FL118 derivatives were synthesized. Our investigations revealed that the majority of these newly synthesized compounds exhibited improved cytotoxicity compared to FL118, with some demonstrating enhanced in vivo antitumor efficacy. Among these derivatives, compound 7h stood out and was subjected to detailed analysis. Compound 7h demonstrated a remarkable ability to inhibit colorectal cancer (CRC) cell colony formation and cell migration, while also promoting reactive oxygen species (ROS) production and CRC cell apoptosis. Notably, our studies unveiled that the presence of DDX5 could modulate Topoisomerase I (Top1) activity, a process effectively reversed by a low concentration of 7h, but not SN38. Moreover, only 7h was able to decrease DDX5 expression, SN38 was not. Molecular docking studies further supported the binding of 7h to DDX5. Interestingly, although both 7h and SN38 exhibited similar inhibitory effects on Top1 activity, only 7h, and not SN38, could inhibit DDX5. These findings not only pave the way for deeper mechanistic explorations of FL118 and its derivatives in cancer research but also position the identified compound 7h as a promising candidate for further development.
Collapse
Affiliation(s)
- Wenchao Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ruojiong Wang
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Lianhao An
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Lei Li
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Haonan Xiong
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Dan Li
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Fangze Dong
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Junrong Lei
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Mengke Wang
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Zhikun Yang
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Hong Wang
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Xiang Ling
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA; Canget BioTekpharma LLC, Buffalo, NY, 14203, USA
| | - Christos Fountzilas
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Fengzhi Li
- Department of Pharmacology & Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| | - Qingyong Li
- College of Pharmaceutical Sciences, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Key Laboratory of Marine Fishery Resources Exploitment & Utilization of Zhejiang Province, Zhejiang University of Technology, Hangzhou, 310014, China.
| |
Collapse
|
2
|
Desai VM, Choudhary M, Chowdhury R, Singhvi G. Reply to "Comment on 'Photodynamic Therapy Induced Mitochondrial Targeting Strategies for Cancer Treatment: Emerging Trends and Insights'". Mol Pharm 2025; 22:585-587. [PMID: 39636785 DOI: 10.1021/acs.molpharmaceut.4c01100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Affiliation(s)
- Vaibhavi Meghraj Desai
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Mahima Choudhary
- Cancer Biology Laboratory, Department of Biological Sciences, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Rajdeep Chowdhury
- Cancer Biology Laboratory, Department of Biological Sciences, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Gautam Singhvi
- Industrial Research Laboratory, Department of Pharmacy, FD-III, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| |
Collapse
|
3
|
Gao XY, Zhang Y, Zhao WP, Tian EJ, Ommati MM, Wang JC, Wang HW, Zhou BH. Molybdenum interferes with MMPs/TIMPs expression to reduce the receptivity of porcine endometrial epithelial cells. Chem Biol Interact 2025; 405:111304. [PMID: 39486568 DOI: 10.1016/j.cbi.2024.111304] [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: 12/20/2023] [Revised: 04/17/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024]
Abstract
To investigate the effect of trace element molybdenum (Mo) on the receptivity of porcine endometrial epithelial cells (PEECs) and evaluate Mo toxicity and its potential molecular mechanisms, Mo-treated PEECs models were established by incubating the cells with various concentrations of medium containing Mo (0, 0.005, 0.020, 0.200, and 5 mmol/L MoNa2O4·2H2O). The results showed that Mo disrupted the morphology and ultrastructure of PEECs, triggered blurred cell edges, cell swelling, cell cycle arrest, and increased apoptosis. At the molecular level, Mo treatment activated the TGF-β1/SMAD2 and PI3K/AKT1 pathways, causing a significant increase in matrix metalloproteinase (MMP)-9 and MMP-2 protein expression. Accompanied by markedly increased tissue inhibitors matrix metalloproteinase (TIMP)-2 and decreased TIMP-1, the balance of MMP2/TIMP-2 and MMP-9/TIMP-1 were disrupted. Ultimately, the receptivity of PEECs was destroyed by excessive Mo, which is revealed by the significant decrease of receptive marker molecules, including leukemia inhibitory factor (LIF), integrins β3 (ITGβ3), heparin-binding epidermal growth factor (HB-EGF), and vascular endothelial growth factor (VEGF). To sum up, the current study demonstrated the potential toxicity of Mo to PEECs, indicating reproductive toxicity at high Mo concentrations and suggesting that the content of Mo should be evaluated as a potential risk factor.
Collapse
Affiliation(s)
- Xiao-Ying Gao
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Yan Zhang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Wen-Peng Zhao
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Er-Jie Tian
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Mohammad Mehdi Ommati
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Ji-Cang Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Hong-Wei Wang
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China
| | - Bian-Hua Zhou
- Henan Key Laboratory of Environmental and Animal Product Safety, College of Animal Science and Technology, Henan University of Science and Technology, Kaiyuan Avenue 263, Luoyang, 471000, Henan, PR China.
| |
Collapse
|
4
|
Gao J, Yin J, Li S, Jia P, Hong R, Chen J, Qu X, Zhang Z, Li M, Zhao H. Discovery of 4-(4-(3-(1-(2-(piperidin-1-yl)ethyl)-1H-benzo[d]imidazole-2-yl)isoxazol-5-yl)phenyl)morpholine as a novel c-Myc inhibitor against lung cancer in vitro and in vivo. Eur J Med Chem 2025; 281:117023. [PMID: 39531932 DOI: 10.1016/j.ejmech.2024.117023] [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: 09/17/2024] [Revised: 10/09/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The critical role of c-Myc as a driving factor in the development and progression of lung cancer establishes it as a pivotal target for anti-lung cancer therapeutic research. In our previous study, we reported on the discovery of D347-2761, a novel small-molecule inhibitor that specifically targets the unstable domain of c-Myc and disrupts the c-Myc/Max heterodimer. To enhance targeted therapies further, we conducted an extensive structural analysis and designed a series of innovative benzimidazole derivatives. The cytotoxic activities of these compounds were assessed using the CCK-8 assay, revealing that compound A1 displayed IC50 values of 6.32 μM and 11.39 μM against the A549 and NCI-H1299 lung cancer cell lines, respectively, while compound A5 exhibited IC50 values of 4.08 μM and 7.86 μM against the same cell lines. Our findings revealed that compounds A1 and A5 exhibited potent anticancer activity by disrupting the interaction between c-Myc and Max proteins, leading to the downregulation of c-Myc protein levels and induction of apoptosis through apoptotic pathways. Notably, compound A5 demonstrated superior inhibitory capacity compared to other compounds tested. Furthermore, in a syngeneic tumor model, compound A5 exhibited excellent efficacy with a tumor growth inhibition rate reaching up to 76.4 %, accompanied by a significant reduction in c-Myc protein expression levels. Therefore, compound A5 holds promise as a potential agent for targeting c-Myc in anti-lung cancer therapy.
Collapse
Affiliation(s)
- Jian Gao
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Jiacheng Yin
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Shihao Li
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Pingting Jia
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Renjie Hong
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Jiahui Chen
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Xinxin Qu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Zihui Zhang
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Mengting Li
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Hui Zhao
- School of Medicine, Anhui University of Science and Technology, Huainan, China.
| |
Collapse
|
5
|
Madbouly NA, Kamal SM, El-Amir AM. Chronic artificial light exposure in daytime and reversed light: Dark cycle inhibit anti-apoptotic cytokines and defect Bcl-2 in peripheral lymphoid tissues during acute systemic inflammatory response to lipopolysaccharide. Int Immunopharmacol 2025; 145:113768. [PMID: 39672023 DOI: 10.1016/j.intimp.2024.113768] [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: 06/17/2024] [Revised: 11/21/2024] [Accepted: 11/30/2024] [Indexed: 12/15/2024]
Abstract
AIMS The disturbed light: dark (LD) cycle has been associated with critical complications, including obesity, diabetes and cancer. In the present study, we investigated the chronic effects of artificial light at daytime (AL) and light at night (RAL) after intraperitoneal (i.p.) injection of saline and 0.5 mg/kg lipopolysaccharide (LPS) in male Wistar rats. METHODS Liver and kidney parameters, fasting blood glucose (FBG), melatonin level, immunohistochemical examinations of B-cell lymphoma-2 (Bcl-2) in spleen and mesenteric lymph and serum antiapoptotic cytokines [interleukin (IL-) 2, 7 and 1]. KEY FINDINGS After 16 weeks of a daily disturbed LD cycle, RAL increased body weight, upgraded FBG and altered liver and kidney functions with surprisingly increased daytime plasma melatonin. AL + LPS and RAL + LPS rats suffered significantly higher oxidative-nitrosative stress compared to NL + LPS. Oxidative-nitrosative stress was associated with multi-organ inflammation in hepatic, renal, pancreatic, splenic and mesenteric lymph node tissues due to LPS-induced endotoxemia. Anti-apoptotic Bcl-2 activity in peripheral lymphoid organs (spleen and mesenteric lymph node) was lowered due to AL and RAL regimens. At the same pattern, lowering of antiapoptotic serum levels of IL-2, IL-7 and IL-15 indicate alteration of cell cycle and the shifted ability of cells to undergo apoptosis due to abnormal light pollution. SIGNIFICANCE Here, the increased lymphocyte apoptosis in lymphoid tissues due to disturbed LD cycle defects the host defense, dysregulates the inflammatory immune response and dysregulates the immune tolerance during acute systemic inflammation due to LPS.
Collapse
|
6
|
P J N, Patil SR, Veeraraghavan VP, Daniel S, Aileni KR, Karobari MI. Oral Cancer Stem Cells: A Comprehensive Review of Key Drivers of Treatment Resistance and Tumor Recurrence. Eur J Pharmacol 2025:177222. [PMID: 39755243 DOI: 10.1016/j.ejphar.2024.177222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/06/2025]
Abstract
Oral squamous cell carcinoma (OSCC) remains a major cause of morbidity and mortality worldwide with high recurrence rates and resistance to conventional therapies. Recent studies have highlighted the pivotal role of oral cancer stem cells (OCSCs) in driving treatment resistance and tumor recurrence. OCSCs possess unique properties, including self-renewal, differentiation potential, and resistance to chemotherapy and radiotherapy, which contribute to their ability to survive treatment and initiate tumor relapse. Several signaling pathways, such as Wnt/β-catenin, Hedgehog, Notch, and PI3K/Akt/mTOR, have been implicated in maintaining OCSC properties, promoting survival, and conferring resistance. Additionally, mechanisms such as drug efflux, enhanced DNA repair, epithelial-mesenchymal transition (EMT), and resistance to apoptosis further contribute to resilience. Targeting these pathways offers promising therapeutic strategies for eliminating OCSCs and improving treatment outcomes. Approaches such as immunotherapy, nanotechnology-based drug delivery, and targeting of the tumor microenvironment are emerging as potential solutions to overcome OCSC-mediated resistance. However, further research is needed to fully understand the molecular mechanisms governing OCSCs and develop effective therapies to prevent tumor recurrence. This review discusses the role of OCSCs in treatment resistance and recurrence and highlights the current and future directions for targeting these cells in OSCC.
Collapse
Affiliation(s)
- Nagarathna P J
- Department of Pediatric Dentistry, Chhattisgarh Dental College and Research Institute, India.
| | - Santosh R Patil
- Department of Oral Medicine and Radiology, Chhattisgarh Dental College and Research Institute, Rajnandgaon, C.G.
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| | - Shikhar Daniel
- Department of Oral Medicine and Radiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India.
| | | | - Mohmed Isaqali Karobari
- Department of Conservative Dentistry & Endodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India.
| |
Collapse
|
7
|
Trory JS, Vautrinot J, May CJ, Hers I. PROTACs in platelets: emerging antithrombotic strategies and future perspectives. Curr Opin Hematol 2025; 32:34-42. [PMID: 39446364 DOI: 10.1097/moh.0000000000000846] [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: 12/07/2024]
Abstract
PURPOSE OF REVIEW Proteolysis-targeted chimeras (PROTACs) are heterobifunctional compounds that selectively target proteins for degradation and are an emerging therapeutic modality to treat diseases such as cancer and neurodegenerative disorders. This review will widen the area of application by highlighting the ability of PROTACs to remove proteins from the anucleate platelets and evaluate their antithrombotic potential. RECENT FINDINGS Proteomic and biochemical studies demonstrated that human platelets possess the Ubiquitin Proteasomal System as well as the E3 ligase cereblon (CRBN) and therefore may be susceptible to PROTAC-mediated protein degradation. Recent findings confirmed that CRBN ligand-based PROTACs targeting generic tyrosine kinases, Btk and/or Fak lead to efficacious and selective protein degradation in human platelets. Downregulation of Btk, a key player involved in signalling to thrombosis, but not haemostasis, resulted in impaired in-vitro thrombus formation. SUMMARY Platelets are susceptible to targeted protein degradation by CRBN ligand-based PROTACs and have limited ability to resynthesise proteins, ensuring long-term downregulation of target proteins. Therefore, PROTACs serve as an additional research tool to study platelet function and offer new therapeutic potential to prevent thrombosis. Future studies should focus on enhancing cell specificity to avoid on-target side effects on other blood cells.
Collapse
Affiliation(s)
- Justin S Trory
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, UK
| | | | | | | |
Collapse
|
8
|
Gedik D, Eraslan G. Evaluation of the efficacy of diosmin and chrysin against tau-fluvalinate exposure in rats. Food Chem Toxicol 2025; 195:115097. [PMID: 39522795 DOI: 10.1016/j.fct.2024.115097] [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: 08/20/2024] [Revised: 11/04/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Tau-fluvalinate is a type 2 pyrethroid insecticide. Diosmin and chrysin are flavonoids with antioxidant and anti-apoptotic effects. Role of diosmin and chrysin against infavorable toxic effects caused by tau-fluvalinate and the underlying mechanisms of these effects were investigated. Six groups were formed and diosmin, chrysin, tau-fluvalinate, tau-fluvalinate + diosmin and tau-fluvalinate + chrysin were administered orally to rats at a dose of 20 mg/kg.bw except for the control group, once a day for 21 days, respectively. Tau-fluvalinate elevated MDA and NO levels while diminishing the activities of antioxidant enzymes (SOD, CAT, GSH-Px, GR, GST, G6PD) and GSH levels in the majority of the analyzed blood and tissues, statistically significant. Serum triglyceride, cholesterol, total protein and albumin levels as well as LDH and PChE activities decreased. Conversely, serum creatinine, AST, ALT and ALP levels/activities increased. Elevated protein levels of caspase 3, caspase 9, p53 and Bax and decreased protein levels of Bcl-2 were observed in the liver. There were negative changes in body/some organ weights. Diosmin and chrysin administration resulted in a marked recovery in tau-fluvalinate-induced toxic effects, but this improvement was not complete. These flavonoids may be considered as promising potential therapeutic options to alleviate the adverse effects associated with tau-fluvalinate intoxication.
Collapse
Affiliation(s)
- Didem Gedik
- Department of Veterinary Pharmacology and Toxicology, Institute of Health Science, Erciyes University, Kayseri, Turkey
| | - Gökhan Eraslan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
9
|
Xu X, Liu J, Fang C, Deng X, Zhu D, Jiang J, Wu C. NAALADL2-AS2 functions as a competing endogenous RNA to regulate apoptosis and drug resistance in DLBCL. Cancer Biol Ther 2024; 25:2432690. [PMID: 39575888 PMCID: PMC11587827 DOI: 10.1080/15384047.2024.2432690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 11/05/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024] Open
Abstract
To explore role of NAALADL2-AS2 as ceRNA in DLBCL. Fluorescence in situ hybridization was used to determine location of NAALADL2-AS2 in cells and to verify its expression in DLBCL tissues. The miRNAs interacting with NAALADL2-AS2 and related regulatory genes were identified by small interfering RNA (siRNA) assay, luciferase reporter assay, fluorescent quantitative polymerase chain reaction, western blotting. DLBCL cells transfected with NAALADL2-AS2 siRNA or control siRNA were treated with doxorubicin, rituximab at different concentrations alone or in combination. The growth curves, drug sensitivity changes of cells before and after transfection were detected by MTT assay, ATP-TCA drug sensitivity test. Cell proliferation was detected by BrdU cell proliferation assay, and apoptosis was detected by Annexin V-fluorescein isothiocyanate/propidium iodide staining. The effects and mechanisms of NAALADL2-AS2 on proliferation, apoptosis, drug resistance of DLBCL cells were studied at cellular level. We confirmed expression of NAALADL2-AS2 in both cytoplasm and nuclei of DLBCL cells. Additionally, we observed elevated levels of NAALADL2-AS2 in DLBCL tissues. We discovered that NAALADL2-AS2 functions as ceRNA to inhibit expression of miR-34a, miR-125a, whereas overexpression of NAALADL2-AS2 indirectly upregulates expression of BCL-2. Interfering with NAALADL2-AS2 promoted apoptosis in DLBCL cells, resulting in approximately a 40% increase in sensitivity to doxorubicin and rituximab. In vivo experiments further confirmed that targeting NAALADL2-AS2 effectively suppressed tumor growth, leading to upregulation of miR-34a and miR-125a, downregulation of BCL-2, and enhanced apoptosis in DLBCL cells, which significantly improved their sensitivity to doxorubicin and rituximab by approximately 50%. These results indicate that NAALADL2-AS2/miR-34a, miR-125a/BCL-2 networks hold promise as therapeutic targets for treatment of DLBCL.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Apoptosis/drug effects
- Drug Resistance, Neoplasm/genetics
- Animals
- Cell Line, Tumor
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Mice
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Cell Proliferation/drug effects
- Rituximab/pharmacology
- Rituximab/therapeutic use
- Gene Expression Regulation, Neoplastic/drug effects
- Xenograft Model Antitumor Assays
- RNA, Competitive Endogenous
Collapse
Affiliation(s)
- Xiaoli Xu
- Department of Integrated Chinese and Western Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Juan Liu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Cheng Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xu Deng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Danxia Zhu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
10
|
Rafiee J, Jamialahmadi K, Bazyari MJ, Aghaee-Bakhtiari SH. Drug repositioning in castration-resistant prostate cancer using systems biology and computational drug design techniques. Comput Biol Chem 2024; 115:108329. [PMID: 39731827 DOI: 10.1016/j.compbiolchem.2024.108329] [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: 04/06/2024] [Revised: 05/07/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND AND OBJECTIVE Castration-resistant prostate cancer (CRPC) is caused by resistance to androgen deprivation treatment and leads to the death of patients and there is almost no chance of survival. Therefore, finding a cure to overcome CRPC is challenging and important, but discovering a new drug is very time-consuming and expensive. To overcome these problems, we used Drug repositioning (drug repurposing) strategy in this study. METHODS Gene expression data of CRPC and primary prostate samples were extracted from the GEO database to identify DEGs. Pathway enrichment was performed to find the role of DEGs in signaling pathways. To identify hub proteins, the PPI network was reconstructed and analyzed. drug candidates were identified and to select the most effective drug, molecular docking analysis, and molecular dynamics simulation were performed. Then MTT and qRT-PCR tests were performed to check the effectiveness of the selected drug. RESULTS A total of 152 upregulated DEGs and 343 downregulated DEGs were identified, and after PPI network analysis, IKBKB, SNAP23, MYC, and NOTCH1 genes were introduced as hubs. drug candidates for IKBKB were identified and by examining the results of docking screening and molecular dynamics, sulfasalazine was selected as the most effective drug. Laboratory analyses proved the effectiveness of this drug and a decrease in the expression of all target genes was observed. CONCLUSION In this study, IKBKB key protein were identified in CRPC, and sulfasalazine was selected as a suitable candidate for drug repositioning and its effectiveness was confirmed through tests.
Collapse
Affiliation(s)
- Javad Rafiee
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khadijeh Jamialahmadi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Javad Bazyari
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Hamid Aghaee-Bakhtiari
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Mfotie Njoya E, van Dyk H, Nambooze J, Chukwuma CI, Brink A, Makhafola TJ. Insight into the molecular mechanism of anti-breast cancer therapeutic potential of substituted salicylidene-based compounds using cell-based assays and molecular docking studies. Eur J Pharmacol 2024; 985:177129. [PMID: 39542411 DOI: 10.1016/j.ejphar.2024.177129] [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: 09/06/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Targeting oxidative stress and inflammatory signaling pathways is an effective cancer prevention and therapy approach. The mechanism of action of synthesized salicylidene-based compounds was investigated in regulating key molecular targets of breast cancer development. Compounds (1), (4), (5), and (7) were found to be more cytotoxic to MCF-7 and 4T1 cells compared to non-cancerous Chang liver cells, while these compounds were cytotoxic to MDA-MB-231 cells, but with poor selectivity. The colony formation assay indicated that bioactive compounds induced significant damage to breast cancer cells, as observed by a reduction in the number of colonies compared to control cells. By inducing a concentration and time-dependent increase of luminescence and fluorescence of phosphatidylserine, and activating the expression of caspases-3, -7, -8, -9 in breast cancer cells, (1) and (7) have shown to induce caspase-dependent apoptosis. The downregulation of NF-kB-p65 and an upregulation of TP53 expression after exposure to bioactive compounds, demonstrated the suppression of two key targets of breast cancer development. Molecular docking studies revealed that selected protein targets strongly interact with bioactive compounds, and the estimated inhibition constants (Ki) of JAK2, STAT3, COX-2, HPV31 E6, EGFR1, TP53, and PARP1 were significantly decreased compared to acetylsalicylic acid. This could be a clear indication that these protein targets are implicated with antiproliferative efficacy, thereby warranting the potential of (1) and (7) to be used as anti-breast cancer drug candidates.
Collapse
Affiliation(s)
- Emmanuel Mfotie Njoya
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, 9301, Free State, South Africa
| | - Hannah van Dyk
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, PO Box 339, Bloemfontein, 9301, Free State, South Africa
| | - Jennifer Nambooze
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, PO Box 339, Bloemfontein, 9301, Free State, South Africa
| | - Chika I Chukwuma
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, 9301, Free State, South Africa
| | - Alice Brink
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of the Free State, PO Box 339, Bloemfontein, 9301, Free State, South Africa
| | - Tshepiso Jan Makhafola
- Centre for Quality of Health and Living (CQHL), Faculty of Health and Environmental Sciences, Central University of Technology, Bloemfontein, 9301, Free State, South Africa.
| |
Collapse
|
12
|
Wu X, Wei D, Zhou Y, Cao Q, Han G, Han E, Chen Z, Guo Y, Huo W, Wang C, Huang S, Zeng X, Wang X, Mao Z. Pesticide exposures and 10-year atherosclerotic cardiovascular disease risk: Integrated epidemiological and bioinformatics analysis. JOURNAL OF HAZARDOUS MATERIALS 2024; 485:136835. [PMID: 39673955 DOI: 10.1016/j.jhazmat.2024.136835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/01/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND AND PURPOSE Recent studies link pesticide exposures to cardiovascular disease risk factors. However, research on the combined effects of multiple pesticides on atherosclerotic cardiovascular disease (ASCVD) is limited, particularly in rural areas. Despite advances in toxicogenomics, the mechanisms underlying these effects remain unclear. This study aims to investigate the combined effects and mechanisms of pesticide exposures on ASCVD. METHODS In the cross-sectional study section, 2291 participants were included. Variables were filtered using machine learning models, and associations between mixed exposure to multiple pesticides and ASCVD were explored using environmental mixed exposure models (weighted quartile sum (WQS) regression and quantile-based g-computation (QGC)). In the bioinformatics analysis section, the GEO, CTD, Malacards, and GeneCards databases were used to retrieve target genes for pesticide exposure and atherosclerotic diseases. Enrichment analysis was then performed to identify the biological pathways associated with these genes. RESULTS Three machine models screened 34 pesticides. Single pesticide exposures, such as atrazine, oxadiazon, p,p'-DDE, α-BHC, β-BHC, fenitrothion, malathion, fenitrothion, cypermethrin, cypermethrin, and cypermethrin might increase the 10-year ASCVD risk (all P < 0.05). Total mixed pesticide exposure was positively associated with 10-year ASCVD risk in both the QGC (3.223(2.196, 4.730)) and WQS models (4.642(3.070, 7.020)). Notably, there was a linear relationship between totalQGC (P_overal < 0.001; P_nonlinearity = 0.864) and high 10-year ASCVD risk. In toxicogenomic bioinformatics analysis, we identified 112 potential atherosclerosis target genes affected by pesticide exposure. Pathway enrichment analysis suggests pesticide-induced atherosclerosis is linked to pathways such as metabolic pathways, lipid metabolism, MAPK, AMPK, FoxO signaling, apoptosis, fluid shear stress, endocrine resistance, TNF, and PI3K-Akt. Key genes were identified based on maximal clique centrality, including AKT1, TP53, IL6, BCL2, TNF, JUN, PTGS2, CASP3, MAPK3, and CASP9. CONCLUSION Individual and combined exposure to pesticides increased the 10-year ASCVD risk, especially in patients with T2DM. Mixed levels of pesticide exposure were linearly and positively associated with high 10-year ASCVD risk. The mechanism of atherogenesis by mixed pesticide exposure may involve pathways such as lipid metabolism, MAPK, AMPK, FoxO signaling, apoptosis, fluid shear stress, endocrine resistance, TNF, and PI3K-Akt.
Collapse
Affiliation(s)
- Xueyan Wu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Dandan Wei
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yilin Zhou
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Qingqing Cao
- Department of Occupational and Environmental Health Sciences, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Guozhen Han
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Erbao Han
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Zhiwei Chen
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yao Guo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Wenqian Huo
- Department of Occupational and Environmental Health Sciences, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Shan Huang
- Henan Institute of Food and Salt Industry Inspection Technology, Zhengzhou, Henan, PR China
| | - Xin Zeng
- School of Public Health, Zhengzhou University, Henan, PR China
| | - Xinlu Wang
- Collaborative Innovation Center of Prevention and Treatment of Major Diseases by Chinese and Western Medicine, Henan Province, PR China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| |
Collapse
|
13
|
Aisanjiang M, Dai W, Wu L, Yuan Y, Liu S, Liao G, Li L, Tong X, Zhang H, Chen Y, Liu J, Cheng J, Wang C, Lu Y. Ameliorating lung fibrosis and pulmonary function in diabetic mice: Therapeutic potential of mesenchymal stem cell. Biochem Biophys Res Commun 2024; 737:150495. [PMID: 39126861 DOI: 10.1016/j.bbrc.2024.150495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
This study aimed to investigate the potential of mesenchymal stem cells (MSCs) in alleviating diabetic lung injury by decreasing inflammation, fibrosis and recovering tissue macrophage homeostasis. To induce pulmonary injuries in an in vivo murine model, we utilized a streptozotocin (STZ), and high-fat diet (HFD) induced diabetic C57 mouse model. Subsequently, human umbilical cord-derived MSCs (hUC-MSCs) were administered through the tail vein on a weekly basis for a duration of 4 weeks. In addition, in vitro experiments involved co-culturing of isolated primary abdominal macrophages from diabetic mice and high glucose-stimulated MLE-12 cells with hUC-MSCs. The objective was to evaluate if hUC-MSCs co-culturing could effectively mitigate cell inflammation and fibrosis. Following hUC-MSCs injection, diabetic mice displayed enhanced pulmonary functional parameters, reduced pulmonary fibrosis, and diminished inflammation. Notably, the dynamic equilibrium of lung macrophages shifted from the M1 phenotype to the M2 phenotype, accompanied by a notable reduction in various indicators associated with inflammation and fibrosis. Results from cell co-culturing experiments further supported this trend, demonstrating a reduction in inflammatory and fibrotic indicators. In conclusion, our findings suggest that hUC-MSCs treatment holds promise in mitigating diabetic pulmonary injury by significantly reducing inflammation, fibrosis and maintain tissue macrophage homeostasis within the lungs. This study sheds light on the therapeutic potential of hUC-MSCs in managing diabetic complications affecting the pulmonary system.
Collapse
Affiliation(s)
- Maikeliya Aisanjiang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Wenshu Dai
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Luna Wu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Yuan
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyun Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Guangneng Liao
- Animal experimental center of West China hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Heteng Zhang
- Sichuan Neo-Life Stem Cell Biotech Inc., Chengdu, China
| | - Younan Chen
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Chengshi Wang
- Department of Endocrinology and Metabolism, Center for Diabetes and Metabolism Research, West China Hospital, Sichuan University, Chengdu, China.
| | - Yanrong Lu
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Transplant Engineering and Immunology, NHFPC, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
14
|
Zhang H, Tang M, Liu Q, Wu D, Sun B, Dong J, Guan L, Luo J, Zeng M. PAT exposure caused human hepatocytes apoptosis and induced mice subacute liver injury by activating oxidative stress and the ERS-associated PERK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:177003. [PMID: 39433224 DOI: 10.1016/j.scitotenv.2024.177003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
With the widespread use of antimony compounds in synthetic materials and processing, the occupational exposure and environmental pollution caused by antimony have attracted the attention of researchers. Studies have shown that antimony compounds can cause liver damage, but the mechanism has not yet been elucidated. In this study, we used the trivalent potassium antimony tartrate (PAT) to infect L02 hepatocytes and Kunming (KM) mice to establish an antimony-induced apoptosis model of L02 cells and a subacute liver injury model of KM mice. We found that PAT exposure caused hepatocyte apoptosis and was accompanied by oxidative stress and endoplasmic reticulum stress (ERS), and the ERS-associated PERK pathway was activated. Further experimental results showed that N-acetyl-l-cysteine (NAC) pretreatment or silencing of the PERK gene in L02 cells reduced PAT-induced apoptosis. The activity of SOD and CAT in treated L02 cells was increased, the malondialdehyde content in L02 cells and liver tissues was decreased, and the content of ERS-related proteins GRP78 and CHOP, as well as the content of PERK-pathway-related proteins p-PERK/PERK, p-eif2α/eif2α and ATF4 protein were significantly reduced. Overall, PAT exposure triggered hepatocyte apoptosis and liver injury by inducing oxidative stress and activating the ERS-associated PERK pathway; however, this effect could be alleviated by NAC intervention or silencing of PERK in hepatocytes.
Collapse
Affiliation(s)
- Hualing Zhang
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Meng Tang
- Center for Disease Control and Prevention, Jiulongpo District, Chongqing 400050, PR China
| | - Qin Liu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Die Wu
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Bing Sun
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jingbang Dong
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Lan Guan
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China
| | - Jianlan Luo
- Institute of Geophysical & Geochemical Exploration of Hunan, Changsha 411100, PR China
| | - Ming Zeng
- Department of Health Toxicology, Xiangya School of Public Health, Central South University, Changsha 410078, PR China.
| |
Collapse
|
15
|
Magalhães M, Domínguez-Martín EM, Jorge J, Gonçalves AC, Massenzio F, Spigarelli R, Ribeiro-Rodrigues T, Catarino S, Girão H, Monti B, Spisni E, Ferreira L, Oliveira PJ, Efferth T, Rijo P, Cabral C. Unveiling the antitumor mechanism of 7α-acetoxy-6β-hydroxyroyleanone from Plectranthus hadiensis in glioblastoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118689. [PMID: 39128799 DOI: 10.1016/j.jep.2024.118689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glioblastoma (GB) is the most aggressive and prevalent glioma within the central nervous system. Despite considerable efforts, GB continues to exhibit a dismal 5-year survival rate (∼6%). This is largely attributed to unfavorable prognosis and lack of viable treatment options. Therefore, novel therapies centered around plant-derived compounds emerge as a compelling avenue to enhance patient survival and well-being. The South African species, Plectranthus hadiensis Schweinf. (P. hadiensis), a member of the Lamiaceae family, has a history of use in traditional medicine for treating a range of diseases, including respiratory, digestive, and liver disorders. This species exhibits diverse biological activities, such as anti-inflammatory and antitumoral properties, likely attributed to its rich composition of naturally occurring diterpenes, like the abietane diterpene, 7α-acetoxy-6β-hydroxyroyleanone (Roy). Roy has demonstrated promising antitumor effects in various cancer cell lines, making it a compelling candidate for further investigation into its mechanisms against GB. AIM OF THE STUDY This study aims to investigate the antitumor activity and potential mechanism of Roy, a natural lead compound, in GB cells. MATERIAL AND METHODS Roy was isolated from the acetonic extract of P. hadiensis and its antitumor mechanism was assessed in a panel of human GB cell lines (U87, A172, H4, U373, and U118) to mimic tumor heterogeneity. Briefly, the impact of Roy treatment on the metabolic activity of cells was evaluated by Alamar Blue® assay, while cell death, cell cycle regulation, mitochondrial membrane potential, and activated caspase-3 activity were evaluated by flow cytometry. Measurement of mRNA levels of target genes was performed by qPCR, while protein expression was assessed by Western blotting. Cell uptake and impact on mitochondrial morphology were evaluated by confocal microscopy. RESULTS Roy induced G2/M cell cycle arrest, mitochondrial fragmentation, and apoptosis by inhibiting the expression of anti-apoptotic proteins and increasing the levels of activated caspase-3. The concentrations of Roy needed to achieve significant inhibitory outcomes were notably lower (6-9 fold) than those of temozolomide (TMZ), the standard first-line treatment, for achieving comparable effects. In addition, at low concentrations (16 μM), Roy affected the metabolic activity of tumor cells while having no significant impact on non-tumoral cells (microglia and astrocytes). CONCLUSION Overall, Roy demonstrated a robust antitumor activity against GB cells offering a promising avenue for the development of novel chemotherapeutic approaches.
Collapse
Affiliation(s)
- Mariana Magalhães
- University of Coimbra, Institute for Interdisciplinary Research, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Portugal; University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Eva María Domínguez-Martín
- CBIOS-Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Departamento de Ciencias Biomédicas, Facultad de Farmacia, Universidad de Alcalá de Henares, Madrid, Spain
| | - Joana Jorge
- University of Coimbra, Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, ICBR, Group of Environment Genetics and Oncobiology (CIMAGO)-Faculty of Medicine, Coimbra, Portugal
| | - Ana Cristina Gonçalves
- University of Coimbra, Laboratory of Oncobiology and Hematology, University Clinic of Hematology and Applied Molecular Biology, Faculty of Medicine, Coimbra, Portugal; University of Coimbra, ICBR, Group of Environment Genetics and Oncobiology (CIMAGO)-Faculty of Medicine, Coimbra, Portugal
| | - Francesca Massenzio
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Renato Spigarelli
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Teresa Ribeiro-Rodrigues
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Steve Catarino
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Henrique Girão
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Enzo Spisni
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Lino Ferreira
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; University of Coimbra, Faculty of Medicine, Coimbra, Portugal
| | - Paulo J Oliveira
- University of Coimbra, CNC-Center for Neuroscience and Cell Biology, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Patrícia Rijo
- CBIOS-Universidade Lusófona's Research Center for Biosciences & Health Technologies, Lisbon, Portugal; Faculty of Pharmacy, Instituto de Investigação Do Medicamento (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Célia Cabral
- University of Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Clinic Academic Center of Coimbra (CACC), Faculty of Medicine, Coimbra, Portugal; University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; University of Coimbra, Centre for Functional Ecology, Department of Life Sciences, Coimbra, Portugal.
| |
Collapse
|
16
|
Pal N, Banerjee K, Sarkar S, Mandal TK, Bhabak KP. Synthesis of Thiazolidinedione- and Triazole-Linked Organoselenocyanates and Evaluation of Anticancer Activities Against Breast Cancer with Mechanistic Investigations. Chemistry 2024:e202403026. [PMID: 39630055 DOI: 10.1002/chem.202403026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 12/13/2024]
Abstract
Organoselenocyanates are important classes of organoselenium compounds having potential pharmaceutical applications in cancer biology. In the present study, two different series of organoselenocyanates (15 a-15 c and 16 a-16 c) incorporating crucial heterocyclic pharmacophores such as 2,4-thiazolidine-1,3-dione and 1,2,3-triazole were rationally designed. The organoselenocyanates were synthesized using multi-step organic synthesis and investigated for their anticancer activities against triple-negative breast cancer cells. Based on the preliminary anti-proliferative activities and the selectivity index towards cancer cells over the normal cells, 2,4-thiazolidine-1,3-dione-based selenocyanate 15 a was identified as the lead analogue for detailed investigations. In addition to the anti-migratory activity, compound 15 a induced G1-phase arrest of the cell cycle and led to early apoptosis. Further studies on the redox balance of MDA-MB-231 cells indicated the antioxidant nature of 15 a with the quenching of ROS level and upregulation of TrxR1 expression. Detailed mechanistic investigations with the expression levels of key-cancer marker proteins revealed that the selenocyanate 15 a induced the activation of ERK pathway by upregulating p-ERK expression with the subsequent downregulation of p-Akt and c-Myc levels leading to the inhibition of cellular proliferation. Therefore, the primary outcomes of the study would be valuable in the development of chemotherapeutic agents towards the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Nikita Pal
- Department of Chemistry, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Kaustav Banerjee
- Department of Chemistry, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Shilpi Sarkar
- Department Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Tapas K Mandal
- Department of Chemical Engineering, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Krishna P Bhabak
- Department of Chemistry, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| |
Collapse
|
17
|
Park MJ, Kim YH. Anti-Cancer Effect of Sulforaphane in Human Pancreatic Cancer Cells Mia PaCa-2. Cancer Rep (Hoboken) 2024; 7:e70074. [PMID: 39632551 PMCID: PMC11617590 DOI: 10.1002/cnr2.70074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 10/28/2024] [Accepted: 11/21/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Pancreatic cancer is difficult to treat early as it has no early symptoms. The presence of sulforaphane (SFN) in cruciferous vegetables has been found to possess anti-cancer effects in gastric and colon cancers. Glycogen synthase kinase-3 beta (GSK-3β), a serine/threonine kinase, plays a significant role in pancreatic cancer progression, influencing tumor growth, metastasis, and treatment resistance. Targeting GSK-3β has shown potential to enhance the efficacy of chemotherapy. However, the mechanism underlying the anticancer effects of SFN on pancreatic cancer through GSK-3β is unclear. AIMS In this study, we examined the anticancer effects of SFN in human pancreatic cancer cell line Mia PaCa-2 and evaluated its molecular mechanisms with respect to the GSK-3β-related pathway. METHODS AND RESULTS SFN increased the protein expression of the phosphorylated form of GSK3β (Ser9). In the Wingless Int-1 homolog/β-catenin pathway, GSK3β induced apoptosis by phosphorylating β-catenin. However, in mutant Kirsten rat sarcoma viral oncogene homolog-like-dependent cells such as Mia PaCa-2, GSK3β was suppressed and the β-catenin level was increased, thus inducing apoptosis. Indeed, SFN increased the protein expression of β-catenin in the cytoplasm and nucleus. Subsequently, we measured the level of cMyc, the target gene of β-catenin. SFN decreased cMyc expression despite an increase in the β-catenin. We measured the expression of nuclear factor (NF)-κB, a downstream factor of GSK3β and an upstream factor of cMyc. SFN decreased the expression of NF-κB and cMyc, indicating that SFN inhibits cell proliferation by suppressing the GSK3β/NF-κB/cMyc pathway. As the suppression of NF-κB results in a decrease in B-cell lymphoma 2 (BCL-2) which is the anti-apoptotic gene, we tested the effect of SFN in the expression of BCL-2. SFN inhibited the expression of BCL-2 and increased the ratio of the apoptotic regulator gene BCL-2 associated X (BAX), where SFN induced the cleaved cysteine aspartase-3 and poly-adenosine diphosphate ribose polymerase. CONCLUSION These results indicate that SFN may have therapeutic potential in the inhibition of pancreatic cancer.
Collapse
Affiliation(s)
- Min Ju Park
- Department of Food and Nutrition, College of EngineeringDaegu UniversityGyeongsan‐SiGyeongsangbuk‐doRepublic of Korea
| | - Yoon Hee Kim
- Department of Food and Nutrition, College of EngineeringDaegu UniversityGyeongsan‐SiGyeongsangbuk‐doRepublic of Korea
| |
Collapse
|
18
|
Sato K, Sato T, Hirotani R, Bam M. Effect of combined blue light and 5-ALA on mitochondrial functions and cellular responses in B16F1 melanoma and HaCaT cells. Cytotechnology 2024; 76:795-816. [PMID: 39435424 PMCID: PMC11490642 DOI: 10.1007/s10616-024-00654-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 08/21/2024] [Indexed: 10/23/2024] Open
Abstract
In this study, we investigated the effects of blue light and 5-aminolevulinic acid (5-ALA) co-treatment on B16F1 melanoma cells and HaCaT keratinocytes. We focused on cellular responses, including mitochondrial function, DNA integrity, and gene expression. Co-treatment significantly damaged the mitochondria, altered their morphology, induced mitochondrial membrane depolarization, increased intracellular reactive oxygen species, and led to cardiolipin peroxidation in both cell types. This approach promoted DNA fragmentation and apoptosis. However, blue light and co-treatment with 5-ALA did not enhance the formation of cyclobutane pyrimidine dimers, 6-4 photoproducts, or Dewar photoproducts. Moreover, it triggered complex, time-dependent changes in gene expression, particularly the upregulation of MMP-1 and p21 in HaCaT cells. Our findings revealed that blue light and 5-ALA co-treatment caused substantial cellular stress and damage, suggesting their therapeutic potential against melanoma and highlighting the need for caution and precision in their application to avoid harming normal cells. This underscores the necessity for further research to refine therapeutic approaches. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00654-x.
Collapse
Affiliation(s)
- Kazuomi Sato
- Graduate School of Agriculture, Tamagawa University, 6-1-1 Machida, Tokyo, 1940-8610 Japan
- Biosystems and Biofunctions Research Center, Tamagawa University Research Institute, 6-1-1 Machida, Tokyo, 194-8610 Japan
| | - Taiki Sato
- Graduate School of Agriculture, Tamagawa University, 6-1-1 Machida, Tokyo, 1940-8610 Japan
| | - Riku Hirotani
- Graduate School of Agriculture, Tamagawa University, 6-1-1 Machida, Tokyo, 1940-8610 Japan
| | - Munetsugu Bam
- Department of Anatomy and Structural Biology, Graduate School of Medicine, University of Yamanashi, 1110 Shimokato, Chuo, 409-3998 Yamanashi Japan
| |
Collapse
|
19
|
Gosálvez J, Johnston SD, Prado A, López-Fernández C, Contreras P, Bartolomé-Nebreda J, González-Martínez M, Fernández JL, de la Vega CG, Góngora A. Strong Correlation Between Double-Strand DNA Breaks and Total Sperm DNA Fragmentation in the Human Ejaculate. Arch Med Res 2024; 55:103122. [PMID: 39566167 DOI: 10.1016/j.arcmed.2024.103122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/30/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Double- and single-strand DNA breaks (DSBs and SSBs, respectively) in spermatozoa, which emerge from intrinsic and extrinsic degenerative processes, are likely related to the underlying male pathology. AIM To determine whether the incidence of DSBs in the human ejaculate is a consistent predictor of whole sperm DNA fragmentation (W-SDF = SSBs + DSBs). METHODS A correlation between the proportion of spermatozoa that showed whole W-SDF and those displaying only DSBs in DNA. Two patient cohorts were established: W-SDF ≤30% (low SDF; n = 153) and W-SDF ≥30% (high SDF; n = 222). RESULTS An increasing level of W-SDF is associated with an increased incidence of DSBs in the ejaculate. When data from both the low and high W-SDF groups were combined, a linear relationship was observed, with DSBs increasing by 0.799 units for each unit increase in W-SDF. However, when the cohorts were analyzed separately, the relationships differed. In the low SDF group, DSBs increased linearly by 0.559 units for each unit increase in W-SDF. In the high SDF group, DSBs increased exponentially by 0.602 units per unit of W-SDF. Furthermore, the data dispersion between the two variables was significantly different between the cohorts, with the high SDF group showing 0.8 times greater variability than the low SDF group. CONCLUSIONS While the presence of DSBs in sperm is correlated with the W-SDF present in raw semen samples, the biological mechanisms responsible for DSBs are expressed in different proportions and/or at different levels in ejaculates with higher levels of DNA damage.
Collapse
Affiliation(s)
- Jaime Gosálvez
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain.
| | - Stephen D Johnston
- School of Environment, University of Queensland, Gatton, Queensland, Australia; School of Veterinary Science, University of Queensland, Gatton, Queensland, Australia; Halotech DNA, Calle Faraday SN, Parque Científico de Madrid, Madrid, Spain
| | - Ahinoa Prado
- School of Veterinary Science, University of Queensland, Gatton, Queensland, Australia; Halotech DNA, Calle Faraday SN, Parque Científico de Madrid, Madrid, Spain
| | | | - Pablo Contreras
- School of Veterinary Science, University of Queensland, Gatton, Queensland, Australia; Halotech DNA, Calle Faraday SN, Parque Científico de Madrid, Madrid, Spain
| | - Javier Bartolomé-Nebreda
- Department of Biology, Universidad Autónoma de Madrid, Madrid, Spain; School of Veterinary Science, University of Queensland, Gatton, Queensland, Australia; Halotech DNA, Calle Faraday SN, Parque Científico de Madrid, Madrid, Spain
| | | | - José Luis Fernández
- Unidad de Genética, Complejo Hospitalario Universitario A Coruña, Instituto de Investigación Biomédica de A Coruña, A Coruña, Spain; Laboratorio de Genética Molecular y Radiobiología, Centro Oncológico de Galicia. A Coruña, Spain
| | | | | |
Collapse
|
20
|
Alotaibi B, A El-Masry T, Elekhnawy E, Mokhtar FA, El-Seadawy HM, A Negm W. Studying the effects of secondary metabolites isolated from Cycas thouarsii R.Br. leaves on MDA-MB-231 breast cancer cells. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:103-113. [PMID: 38279824 DOI: 10.1080/21691401.2024.2306529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/03/2024] [Indexed: 01/29/2024]
Abstract
The various therapeutic drugs that are currently utilized for the management of cancer, especially breast cancer, are greatly challenged by the augmented resistance that is either acquired or de novo by the cancer cells owing to the long treatment periods. So, this study aimed at elucidating the possible anticancer potential of four compounds 7, 4', 7'', 4'''-tetra-O-methyl amentoflavone, hesperidin, ferulic acid, and chlorogenic acid that are isolated from Cycas thouarsii leaves n-butanol fraction for the first time. The MTT assay evaluated the cytotoxic action of four isolated compounds against MDA-MB-231 breast cancer cells and oral epithelial cells. Interestingly, ferulic acid revealed the lowest IC50 of 12.52 µg/mL against MDA-MB-231 cells and a high IC50 of 80.2 µg/mL against oral epithelial cells. Also, using an inverted microscope, the influence of ferulic acid was studied on the MDA-MB-231, which revealed the appearance of apoptosis characteristics like shrinkage of the cells and blebbing of the cell membrane. In addition, the flow cytometric analysis showed that the MDA-MB-231 cells stained with Annexin V/PI had a rise in the count of the cells in the early and late apoptosis stages. Moreover, gel electrophoresis detected DNA fragmentation in the ferulic acid-treated cells. Finally, the effect of the compound was tested at the molecular level by qRT-PCR. An upregulation of the pro-apoptotic genes (BAX and P53) and a downregulation of the anti-apoptotic gene (BCL-2) were observed. Consequently, our study demonstrated that these isolated compounds, especially ferulic acid, may be vital anticancer agents, particularly for breast cancer, through its induction of apoptosis through the P53-dependent pathway.
Collapse
Affiliation(s)
- Badriyah Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Tanta University, Tanta, Egypt
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Tanta University, Tanta, Egypt
| | - Fatma A Mokhtar
- Department of Pharmacognosy, El Saleheya El Gadida University, Sharkia, Egypt
| | | | - Walaa A Negm
- Department of Pharmacognosy, Tanta University, Tanta, Egypt
| |
Collapse
|
21
|
Gungordu S, Aptullahoglu E. Targeting MDM2-mediated suppression of p53 with idasanutlin: a promising therapeutic approach for acute lymphoblastic leukemia. Invest New Drugs 2024; 42:603-611. [PMID: 39305365 DOI: 10.1007/s10637-024-01473-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 12/08/2024]
Abstract
Despite available treatments for acute lymphoblastic leukemia (ALL), the disease's high clinical variability necessitates new therapeutic strategies, particularly for patients with high-risk features. The tumor suppressor protein p53, encoded by the TP53 gene and known as the guardian of the genome, plays a crucial role in preventing tumor development. Over 90% of ALL cases initially harbor wild-type TP53. Reactivation of p53, which is encoded from the wild type TP53 but lost its function for several reasons, is an attractive therapeutic approach in cancer treatment. p53 can be activated in a non-genotoxic manner by targeting its primary repressor, the MDM2 protein. Clinical trials involving MDM2 inhibitors are currently being conducted in a growing body of investigation, reflecting of the interest in incorporating these treatments into cancer treatment strategies. Early-phase clinical trials have demonstrated the promise of idasanutlin (RG7388), one of the developed compounds. It is a second-generation MDM2-p53 binding antagonist with enhanced potency, selectivity, and bioavailability. The aim of this study is to evaluate the efficacy of RG7388 as a therapeutic strategy for ALL and to investigate its potential impact on improving treatment outcomes for high-risk patients. RG7388 potently decreased the viability in five out of six ALL cell lines with diverse TP53 mutation profiles, whereas only one cell line exhibited high resistance. RG7388 induced a pro-apoptotic gene expression signature with upregulation of p53-target genes involved in the intrinsic and extrinsic pathways of apoptosis. Consequently, RG7388 led to a concentration-dependent increase in caspase-3/7 activity and cleaved poly (ADP-ribose) polymerase. In this research, RG7388 was investigated with pre-clinical methods in ALL cells as a novel treatment strategy. This study suggests further functional research and in-vivo evaluation, and it highlights the prospect of treating p53-functional ALL with MDM2 inhibitors.
Collapse
Affiliation(s)
- Seyda Gungordu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey
| | - Erhan Aptullahoglu
- Biotechnology Application and Research Centre, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
- Department of Molecular Biology and Genetics, Faculty of Science, Bilecik Şeyh Edebali University, 11100, Bilecik, Turkey.
| |
Collapse
|
22
|
Ghosh S, Dey A, Chakrabarti A, Bhuniya T, Indu N, Hait A, Chowdhury A, Paul A, Mahajan AA, Papadakis M, Alexiou A, Jha SK. The theragnostic advances of exosomes in managing leukaemia. J Cell Mol Med 2024; 28:e70052. [PMID: 39659020 PMCID: PMC11632122 DOI: 10.1111/jcmm.70052] [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: 07/21/2024] [Accepted: 08/20/2024] [Indexed: 12/12/2024] Open
Abstract
Leukaemia, a group of haematological malignancies, presents ongoing diagnosis, prognosis, and treatment challenges. A major obstacle in treating this disease is the development of drug resistance. Overcoming drug resistance poses a significant barrier to effective leukaemia treatment. The emergence of exosome research has unveiled new insights into the probable theragnostic implementations in leukaemia. Various research has exhibited the diagnostic possibilities of exosomes in identifying leukaemia-specific biomarkers, including genetic mutations and fusion transcripts. Additionally, exosomes have been implicated in disease progression and treatment response, rendering them appealing targets for therapeutics. Exosomes, originating from diverse cell types, are instrumental in intercellular communication as they participate in the functional transportation of molecules like proteins, nucleic acids and lipids across space. Exosomes have a dual role in immune regulation, mediating immune suppression and modulating anti-leukaemia immune responses. Interestingly, exosomes can even act as drug transport vehicles. This review delves into the intricate process of exosome biogenesis, shedding light on their formation and release from donor cells. The key mechanisms engaged in exosome biogenesis, for instance, the endosomal sorting complexes required for transport (ESCRT) machinery and ESCRT-independent pathways, are thoroughly discussed. Looking ahead, future approaches that leverage innovative technologies hold the promise of revolutionizing disease management and improving patient outcomes.
Collapse
Affiliation(s)
- Subhrojyoti Ghosh
- Department of BiotechnologyIndian Institute of Technology MadrasChennaiTamil NaduIndia
| | - Anuvab Dey
- Department of Biosciences and BioengineeringIndian Institute of Technology GuwahatiGuwahatiAssamIndia
| | - Aneshwa Chakrabarti
- Department of Chemistry and Chemical BiologyIndian Institute of Technology, Indian School of Mines DhanbadDhanbadIndia
| | - Tiyasa Bhuniya
- Department of BiotechnologyNIT DurgapurDurgapurWest BengalIndia
| | - Neelparna Indu
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Anirban Hait
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Ankita Chowdhury
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | - Aritra Paul
- Department of BiotechnologyHeritage Institute of TechnologyKolkataIndia
| | | | - Marios Papadakis
- Department of Surgery IIUniversity Hospital Witten‐HerdeckeWuppertalGermany
| | - Athanasios Alexiou
- University Centre for Research & DevelopmentChandigarh UniversityMohaliPunjabIndia
- Department of Research & DevelopmentFunogenAthensGreece
- Department of Research & DevelopmentAFNP MedWienAustria
- Department of Science and EngineeringNovel Global Community Educational FoundationHebershamNew South WalesAustralia
| | | |
Collapse
|
23
|
Mota IDS, Cardoso M, Bueno J, da Silva IGM, Gonçalves J, Bao SN, Neto BAD, Brand G, Corrêa JR, Leite JRSA, Saldanha-Araujo F. Intragenic antimicrobial peptide Hs02 toxicity against leukemia cell lines is associated with increased expression of select pyroptotic components. Toxicol In Vitro 2024; 101:105945. [PMID: 39343072 DOI: 10.1016/j.tiv.2024.105945] [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: 07/22/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
The anticancer potential of some antimicrobial peptides has been reported. Hs02 is a recently characterized Intragenic Antimicrobial Peptide (IAP), which was able to exhibit potent antimicrobial and anti-inflammatory action. In this study, we evaluate for the first time the antineoplastic potential of the Hs02 IAP using cell lines representing the main types of leukemia as cancer models. Interestingly, this peptide decreased the viability of several leukemic cell lines, without compromising the viability of PBMCs in the same concentration. In the HL-60 line, treatment with Hs02 controlled cell division, leading to cell arrest in the G1 phase of the cell cycle. More importantly, HL-60 cells treated with Hs02 undergo cell death, with the formation of pores in the plasma membrane and the release of LDH. Accordingly, Hs02 treatment stimulated the expression of components involved in pyroptosis, such as NLRP1, CASP-1, GSDME, and IL-1β. Taken together, our data characterize the antineoplastic potential of Hs02 and open an opportunity for both evaluating the peptide's antineoplastic potential in other cancer models and using this molecule as a template for new peptides with therapeutic potential against cancer.
Collapse
Affiliation(s)
- Isabella de Souza Mota
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - Miguel Cardoso
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, Universidade de Brasília-DF, 70910-900, Brazil; iMed.ULisboa-Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon 1649-003, Portugal
| | - João Bueno
- Laboratório de Síntese e Análise de Biomoléculas, Instituto de Química, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - Ingrid Gracielle Martins da Silva
- Laboratório de Microscopia e Microanálise, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - João Gonçalves
- iMed.ULisboa-Research Institute for Medicines, Faculty of Pharmacy, University of Lisbon, Lisbon 1649-003, Portugal
| | - Sonia N Bao
- Laboratório de Microscopia e Microanálise, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - Brenno A D Neto
- Laboratório de Química Medicinal e Tecnológica, Instituto de Química, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - Guilherme Brand
- Laboratório de Síntese e Análise de Biomoléculas, Instituto de Química, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - José Raimundo Corrêa
- Laboratório de Microscopia e Microanálise, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, DF 70910-900, Brazil
| | - José Roberto S A Leite
- Núcleo de Pesquisa em Morfologia e Imunologia Aplicada, NuPMIA, Faculdade de Medicina, Universidade de Brasília-DF, 70910-900, Brazil
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, DF 70910-900, Brazil.
| |
Collapse
|
24
|
Hussein Z, Michel HE, El-Naga RN, El-Demerdash E, Mantawy EM. Coenzyme Q10 ameliorates cyclophosphamide-induced chemobrain by repressing neuronal apoptosis and preserving hippocampal neurogenesis: Mechanistic roles of Wnt/ β-catenin signaling pathway. Neurotoxicology 2024; 105:21-33. [PMID: 39209270 DOI: 10.1016/j.neuro.2024.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Deterioration in the neurocognitive function of cancer patients referred to as "Chemobrain" is a devastating obstacle associated with cyclophosphamide (CYP). CYP is an alkylating agent, clinically utilized as an efficient anticancer and immunosuppressant. Coenzyme Q10 (CoQ10) is a worthwhile micronutrient with diverse biological activities embracing antioxidant, anti-apoptotic, and neuroprotective effects. The current experiment was designed for investigating the neuroprotective capability of CoQ10 versus CYP-elicited chemobrain in rats besides elucidating the causal molecular mechanisms. Male Sprague Dawley rats received CoQ10 (10 mg/kg, orally, once daily, for 10 days) and/or a single dose of CYP (200 mg/kg i.p. on day 7). CoQ10 counteracted CYP-induced cognitive and motor dysfunction as demonstrated by the findings of neurobehavioral tests (passive avoidance, Y maze, locomotion, and rotarod tests). Histopathological analysis further affirmed the neuroprotective abilities of CoQ10. CoQ10 effectually diminished CYP-provoked oxidative injury by restoring the antioxidant activity of catalase (CAT) enzyme while reducing malondialdehyde (MDA) levels. Besides, CoQ10 efficiently repressed CYP-induced neuronal apoptosis by downregulating the expression of Bax and caspase-3 while upregulating the Bcl-2 expression. Moreover, CoQ10 hampered CYP-provoked upregulation in acetylcholinesterase (AChE) activity. Furthermore, CoQ10 considerably augmented hippocampal neurogenesis by elevating the expressions of brain-derived neurotrophic factor (BDNF) and Ki-67. These promising neuroprotective effects can be credited to upregulating Wnt/β-catenin pathway as evidenced by the elevated expressions of Wnt-3a, β-catenin, and Phoshpo-glycogen synthase kinase-3 β (p-GSK-3β). Collectively, these findings proved the neuroprotective capabilities of CoQ10 against CYP-induced chemobrain through combating oxidative injury, repressing intrinsic apoptosis, boosting neurogenesis, and eventually upregulating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Zeina Hussein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ebtehal El-Demerdash
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman M Mantawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt; Preclinical and Translational Research Center, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
25
|
Kumar A, Jeyakumar A, Lam AK, Gopalan V. Epidemiological and genetic insights into the co-occurrence of cutaneous melanoma and hematologic malignancies: A meta-analytic review. Leuk Res 2024; 147:107610. [PMID: 39476615 DOI: 10.1016/j.leukres.2024.107610] [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: 09/04/2024] [Revised: 10/13/2024] [Accepted: 10/23/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND The number of cancer survivors has been increasing in recent years due to advancements in early diagnosis and prolonged survival. Existing literature suggests a connection between cutaneous melanoma (CM) and hematologic malignancies (HM). AIM This study aims to examine epidemiological research on the link between CM and HM and explore genetic, biological, and environmental factors contributing to this association. METHODOLOGY A literature review and meta-analysis were performed to evaluate the risk of CM following HM and vice versa. Data from included studies, which reported standardized incidence ratios (SIR) or hazard ratios (HR) with 95 % confidence intervals (CI), were pooled using a random effects model. Heterogeneity among studies was assessed using I² and Cochrane Q test statistics. The incidence data were pooled using a random effects model. This review is registered on PROSPERO (CRD42022359887). RESULTS Ten studies focused on HM diagnosis in CM patients, comprising a combined cohort of 189,094 individuals and 11 focused on CM diagnosis in HM patients in a cohort of 306,967 individuals. The SIR for HM after CM ranged from 1.25 to 3.12, while the SIR for CM after HM ranged from 0.83 to 4.12. The pooled proportion of HM in CM patients was 62.4 %, and the proportion of CM in HM patients was 19.6 %. Statistical heterogeneity was high, with I² values of 99.19 % and 89.15 %, respectively. CONCLUSION This review confirms an association between CM and HM within the same patient. The link is primarily attributed to genetic factors involving BRAF-V600K, tyrosine kinase pathway genes, CDKN2A (P16), and BCL-2. Additionally, risk factors such as ultraviolet radiation and compromised immune function are associated with the incidence of these cancers.
Collapse
Affiliation(s)
- Ashmitha Kumar
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast 4222, Australia
| | - Arunan Jeyakumar
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast 4222, Australia
| | - Alfred K Lam
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast 4222, Australia.
| | - Vinod Gopalan
- School of Medicine and Dentistry, Griffith University, Gold Coast Campus, Gold Coast 4222, Australia.
| |
Collapse
|
26
|
Paul S, Chatterjee A, Das K, Ray A, Basu A, Mukhopadhyay S, Sen P. Thrombin confers chemotherapeutic resistance by promoting transcriptional induction and post-translational stabilization of pro-survival MCL1 in TNBC. J Biol Chem 2024; 301:108025. [PMID: 39608719 DOI: 10.1016/j.jbc.2024.108025] [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: 04/25/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/30/2024] Open
Abstract
The association between idiopathic venous thrombosis and occult cancer is widely recognized. However, the comprehensive understanding of how thrombin, generated during the process of thrombosis, possesses the potential to augment the malignant phenotype is still not well understood. The coagulation protease thrombin mediates its effects by cleaving protease-activated receptor 1 (PAR1), a receptor abundantly expressed on the surface of triple-negative breast cancer (TNBC) cells. While emerging evidence implicates coagulation proteases in facilitating cancer progression, the precise molecular pathways underlying thrombin-mediated induction of chemoresistance remain poorly defined. Here, we demonstrate that thrombin-induced PAR1 activation in TNBC cells promotes the development of a multidrug-resistant phenotype, mechanistically linked to the upregulation of the pro-survival protein MCL1. Genetic ablation of MCL1 sensitizes TNBC cells to cytotoxic drugs despite thrombin exposure, affirming MCL1's functional importance. Chromatin immunoprecipitation analyses reveal thrombin triggers protein kinase A-dependent phosphorylation of serine 133 residues of cAMP-responsive element-binding protein (CREB), enhancing CREB's affinity for the co-activators CBP and p300. Furthermore, thrombin treatment induces the nuclear translocation of CREB-regulated transcription coactivator 2 (CRTC2) in a calcium-dependent manner, which collectively interacts with CREB/CBP-P300. The coordinated action of these transcriptional co-activators facilitates the transcriptional induction of MCL1. We further report that PAR1 activation augments MCL1 binding to the deubiquitinase USP9X, reducing MCL1 turnover. Our pre-clinical breast cancer murine model also shows that genetic deletion of PAR1 sensitizes breast cancer cells to chemotherapeutic drugs in vivo. Collectively, these findings emphasize the thrombin-PAR1 axis as a novel driver of chemoresistance. Utilizing FDA-approved oral anticoagulants for selective blocking of thrombin action may serve as a potential therapeutic adjunct for the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Akash Chatterjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | - Kaushik Das
- National Institute of Biomedical Genomics, Kalyani, India
| | - Anushka Ray
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India
| | | | | | - Prosenjit Sen
- School of Biological Sciences, Indian Association for the Cultivation of Science, Kolkata, India.
| |
Collapse
|
27
|
Hsu PC, Hsu YH, Chang CH, Shih TL. Design and Synthesis of Unsymmetric Benzils, Quinoxalines, and Evaluations of their Anticancer Activities against Human Non-Small Lung Cancer Cells. ChemMedChem 2024:e202400847. [PMID: 39592439 DOI: 10.1002/cmdc.202400847] [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: 10/24/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 11/28/2024]
Abstract
Quinoxaline and its derivatives exhibit a broad spectrum of biological activity, making them valuable for various therapeutic applications. However, most quinoxalines are synthetically produced due to their scarcity in nature. In this article, a series of unsymmetric benzils were synthesized and subsequently condensed with 1,2-diaminobenzene to produce unsymmetric quinoxalines. The novel synthetic benzils and quinoxalines were evaluated for their anticancer activities against human non-small-cell lung cancer (NSCLC) cells harboring different gene mutations, to explore their potential as anticancer agents. Among these synthesized molecules, compound 5 g demonstrated inhibitory effects comparable to those of cisplatin.
Collapse
Affiliation(s)
- Ping-Chih Hsu
- Division of Thoracic Medicine, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333423, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, 333323, Taiwan
| | - Yu-Hsin Hsu
- Department of Chemistry, Tamkang University, 251301, Tamsui Dist., New Taipei City, Taiwan
| | - Chuan-Hsin Chang
- Research Center for Chinese Herbal Medicine, Graduate Institute of Healthy Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, 333324, Taoyuan, Taiwan
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, 231405, Taiwan
| | - Tzenge-Lien Shih
- Department of Chemistry, Tamkang University, 251301, Tamsui Dist., New Taipei City, Taiwan
| |
Collapse
|
28
|
Kędzierska M, Bańkosz M. Role of Proteins in Oncology: Advances in Cancer Diagnosis, Prognosis, and Targeted Therapy-A Narrative Review. J Clin Med 2024; 13:7131. [PMID: 39685591 DOI: 10.3390/jcm13237131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Modern oncology increasingly relies on the role of proteins as key components in cancer diagnosis, prognosis, and targeted therapy. This review examines advancements in protein biomarkers across several cancer types, including breast cancer, lung cancer, ovarian cancer, and hepatocellular carcinoma. These biomarkers have proven critical for early detection, treatment response monitoring, and tailoring personalized therapeutic strategies. The article highlights the utility of targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, in improving treatment efficacy while minimizing systemic toxicity. Despite these advancements, challenges like tumor resistance, variability in protein expression, and diagnostic heterogeneity persist, complicating universal application. The review underscores future directions, including the integration of artificial intelligence, advanced protein analysis technologies, and the development of combination therapies to overcome these barriers and refine personalized cancer treatment.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, Copernicus Memorial Hospital of Lodz, 90-549 Lodz, Poland
| | - Magdalena Bańkosz
- CUT Doctoral School, Faculty of Materials Engineering and Physics, Department of Material Engineering, Cracow University of Technology, 37 Jana Pawla II Av., 31-864 Krakow, Poland
| |
Collapse
|
29
|
Elizalde-Velázquez GA, Gómora-Martínez O, Raldua D, Herrera-Vázquez SE, Gómez-Oliván LM. Understanding the impact of environmentally relevant alkyl C12-16 dimethylbenzyl ammonium chloride concentrations on zebrafish health. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 953:175984. [PMID: 39244042 DOI: 10.1016/j.scitotenv.2024.175984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alkyldimethylbenzylammonium chlorides (ADBACs), classified as second-generation quaternary ammonium compounds, are extensively employed across various sectors, encompassing veterinary medicine, food production, pharmaceuticals, cosmetics, ophthalmology, and agriculture. Consequently, significant volumes of ADBAC C12-C16 are discharged into the environment, posing a threat to aquatic organisms. Regrettably, comprehensive data regarding the toxicological characteristics of these compounds remain scarce. This research aimed to determine whether or not ADBAC C12-C16, at environmentally relevant concentrations (0.4, 0.8, and 1.6 μg/L), may instigate oxidative stress and alter the expression of apoptosis-related genes in the liver, brain, gut, and gills of Danio rerio adults (5-6 months). The findings revealed that ADBAC C12-C16 elicited an oxidative stress response across all examined organs following 96 h of exposure. Nonetheless, the magnitude of this response varied among organs, with the gills exhibiting the highest degree of susceptibility, followed by the gut, liver, and brain, in descending order. Only the gut and gills of the examined organs displayed a concentration-dependent reduction in the activity of superoxide dismutase (SOD) and catalase (CAT). Akin to the oxidative stress response, all organs exhibited a marked increase in bax, blc2, casp3, and p53 expression levels. However, the gills and gut manifested a distinctive suppression in the expression of nrf1 and nrf2. Our Principal Component Analysis (PCA) confirmed that SOD, CAT, nrf1, and nrf2 were negatively correlated to oxidative damage biomarkers and apoptosis-related genes in the gills and gut; meanwhile, in the remaining organs, all biomarkers were extensively correlated. From the above, it can be concluded that ADBAC C12-C16 in low and environmental concentrations may threaten the health of freshwater fish.
Collapse
Affiliation(s)
- Gustavo Axel Elizalde-Velázquez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Omar Gómora-Martínez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Demetrio Raldua
- Institute of Environmental Assessment and Water Research (IDAEA-CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Selene Elizabeth Herrera-Vázquez
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico.
| |
Collapse
|
30
|
Ebadi M, Kavousi M, Farahmand M. Investigation of the Apoptotic and Antimetastatic Effects of Nano-Niosomes Containing the Plant Extract Anabasis setifera on HeLa: In Vitro Cervical Cancer Study. Chem Biodivers 2024:e202402599. [PMID: 39575851 DOI: 10.1002/cbdv.202402599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
The present study focuses on the preparation of niosomes containing an extract of Anabasis setifera and evaluates their efficacy in inhibiting the growth and proliferation of HeLa cells. Thin-layer hydration technique was used to prepare niosomes/extract nanoparticles (NPs). The physicochemical properties of the synthesized NPs were confirmed by scanning electron microscope (SEM), dynamic light scattering (DLS), zeta potential analysis, and FTIR. The cytotoxicity of the free extract, free niosome, and NPs was investigated by MTT (3-(4, 5-diMethylThiazol-2-yl)-2,5-diphenylTetrazolium bromide) assay. For this purpose, solutions of the three mentioned agents were prepared and diluted in 400, 200, 100, 50, 25, 12.5, and 6.25 µg/mL concentrations and incubated for 24, 48, and 72 h. After calculating the IC50 concentration and treating the cells with this concentration, real-time polymerase chain reaction (PCR) (to measure changes in the expression of apoptosis and metastasis genes), flow cytometry (to determine the amount of early and late induced apoptosis), and cell cycle test (to determine the stopping stage of the cancer cell division cycle) were performed. Moreover, the scratch test (the ability to inhibit cell metastasis after treatment) was used to evaluate cell migration. The MTT assay results showed that 72 h of treatment with NPs has the greatest effect on the death of cancer cells. Real-time PCR showed that the expression of the Bad gene increased dramatically and the expression of the BCL-XL, integrin alpha 5 (ITGA5), and zinc finger E-box-binding homeobox 1 (ZEB-1) genes decreased significantly. The flow cytometry results showed that 48.64% of HeLa cells underwent apoptosis after treatment with synthesized NPs. The scratch test results showed that cancer cell metastasis stopped after treatment with NPs. The research demonstrates the significant potential of plant extract-loaded niosomes, as highly efficient drug carriers for cancer therapy.
Collapse
Affiliation(s)
- Mahya Ebadi
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Kavousi
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mahnaz Farahmand
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
31
|
Saleh DO, Abo El Nasr NME, Hussien YA, El-Baset MA, Ahmed KA. Cyclophosphamide-induced testicular injury: the role of chrysin in mitigating iron overload and ferroptosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03519-4. [PMID: 39565397 DOI: 10.1007/s00210-024-03519-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/06/2024] [Indexed: 11/21/2024]
Abstract
This study evaluated the beneficial effects of chrysin against cyclophosphamide (CP)-induced testicular toxicity in rats across several parameters, including hormones, oxidative stress, inflammation, apoptosis, and protein expression. Rats were pretreated with oral doses of chrysin at 25, 50, or 100 mg/kg daily for 7 days. On the 8th day, all groups except controls received CP (200 mg/kg) injection. Chrysin doses continued for 7 more days. Hormones, oxidative stress markers, inflammatory cytokines, apoptosis regulators, and iron regulatory proteins were assessed. CP decreased testosterone, inhibin B, GSH, and GPx4 and increased FSH, cholesterol, MDA, IL-6, and BAX. It also drastically reduced TfR1, liprin, and IREB2. Chrysin dose-dependently counteracted these effects. The highest 100 mg/kg chrysin dose increased testosterone, inhibin B, GSH, GPx4, BCL2, TfR1, liprin, and IREB2 while decreasing FSH, cholesterol, MDA, IL-6, and BAX close to control levels. There were also significant incremental benefits for testosterone, inhibin B, and other parameters with higher chrysin doses. Chrysin dose-dependently attenuated CP-induced hormonal dysfunction, oxidative stress, inflammation, apoptosis, and iron-regulatory protein suppression. The maximum dose showed the most optimal protective effects in restoring the testicular toxicity markers. These results validate the promising spermatoprotective properties of chrysin against chemotherapeutic germ cell damage.
Collapse
Affiliation(s)
- Dalia O Saleh
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Nesma M E Abo El Nasr
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt.
| | - Yosra A Hussien
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Marawan Abd El-Baset
- Pharmacology Depatrment, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
32
|
Ju S, Singh MK, Han S, Ranbhise J, Ha J, Choe W, Yoon KS, Yeo SG, Kim SS, Kang I. Oxidative Stress and Cancer Therapy: Controlling Cancer Cells Using Reactive Oxygen Species. Int J Mol Sci 2024; 25:12387. [PMID: 39596452 PMCID: PMC11595237 DOI: 10.3390/ijms252212387] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer is a multifaceted disease influenced by various mechanisms, including the generation of reactive oxygen species (ROS), which have a paradoxical role in both promoting cancer progression and serving as targets for therapeutic interventions. At low concentrations, ROS serve as signaling agents that enhance cancer cell proliferation, migration, and resistance to drugs. However, at elevated levels, ROS induce oxidative stress, causing damage to biomolecules and leading to cell death. Cancer cells have developed mechanisms to manage ROS levels, including activating pathways such as NRF2, NF-κB, and PI3K/Akt. This review explores the relationship between ROS and cancer, focusing on cell death mechanisms like apoptosis, ferroptosis, and autophagy, highlighting the potential therapeutic strategies that exploit ROS to target cancer cells.
Collapse
Affiliation(s)
- Songhyun Ju
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jyotsna Ranbhise
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Wonchae Choe
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Sik Yoon
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seung Geun Yeo
- Department of Otorhinolaryngology—Head and Neck Surgery, College of Medicine, Kyung Hee University Medical Center, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.); (M.K.S.); (S.H.); (J.R.); (J.H.); (W.C.); (K.-S.Y.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
33
|
AlDoughaim M, AlSuhebany N, AlZahrani M, AlQahtani T, AlGhamdi S, Badreldin H, Al Alshaykh H. Cancer Biomarkers and Precision Oncology: A Review of Recent Trends and Innovations. Clin Med Insights Oncol 2024; 18:11795549241298541. [PMID: 39559827 PMCID: PMC11571259 DOI: 10.1177/11795549241298541] [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: 07/08/2024] [Accepted: 10/22/2024] [Indexed: 11/20/2024] Open
Abstract
The discovery of cancer-specific biomarkers has resulted in major advancements in the field of cancer diagnostics and therapeutics, therefore significantly lowering cancer-related morbidity and mortality. Cancer biomarkers can be generally classified as prognostic biomarkers that predict specific disease outcomes and predictive biomarkers that predict disease response to targeted therapeutic interventions. As research in the area of predictive biomarkers continues to grow, precision medicine becomes far more integrated in cancer treatment. This article presents a general overview on the most recent advancements in the area of cancer biomarkers, immunotherapy, artificial intelligence, and pharmacogenomics of the Middle East.
Collapse
Affiliation(s)
- Maha AlDoughaim
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Nada AlSuhebany
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Mohammed AlZahrani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Tariq AlQahtani
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Sahar AlGhamdi
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hisham Badreldin
- College of Pharmacy, King Saud Bin Abdul Aziz University for Health Sciences (KSAU-HS), King Abdullah International Medical Research Center (KAIMRC), Riyadh, Saudi Arabia
| | - Hana Al Alshaykh
- Pharmaceutical Care Devision, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia
| |
Collapse
|
34
|
Granados-Aparici S, Vieco-Martí I, López-Carrasco A, Navarro S, Noguera R. Real-time morphometric analysis of targeted therapy for neuroblastoma cells in monolayer and 3D hydrogels using digital holographic microscopy. iScience 2024; 27:111231. [PMID: 39569369 PMCID: PMC11576390 DOI: 10.1016/j.isci.2024.111231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/26/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
High-risk neuroblastoma (HR-NB) patient treatment is currently insufficient and challenging due to its high clinical, morphological, and genetic heterogeneity as well as the scarcity of available samples for research. We used a gelatin- and silk fibroin-based hydrogel system with cross-linked vitronectin (VN) as an artificial biomimetic three-dimensional (3D) environment to mirror aggressive neuroblastoma (NB) tumors and tested long-term cell response to Cilengitide (CLG). Based on our previous studies and others using the integrin inhibitor CLG as a potential mechanotherapy drug, we show that CLG caused cell detachment in monolayer cultures of MYCN-amplified SK-N-BE (2) and ALK-mutated SH-SY5Y human neuroblastoma cell lines. Cell detachment and aggregation were maintained in hydrogel-free monolayer cells whereas cells embedded in hydrogels presented different responses to treatment, suggesting differential anoikis resistance between the two cell lines. This underscores the advantages of testing therapeutic approaches using real-time imaging of tumor cells in 3D biomimetic models and its contribution to precision medicine.
Collapse
Affiliation(s)
- Sofia Granados-Aparici
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Isaac Vieco-Martí
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amparo López-Carrasco
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Samuel Navarro
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rosa Noguera
- Pathology Department, Medical School, University of Valencia-INCLIVA, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
35
|
Kuttikrishnan S, Ansari AW, Suleman M, Ahmad F, Prabhu KS, El-Elimat T, Alali FQ, Al Shabeeb Akil AS, Bhat AA, Merhi M, Dermime S, Steinhoff M, Uddin S. The apoptotic and anti-proliferative effects of Neosetophomone B in T-cell acute lymphoblastic leukaemia via PI3K/AKT/mTOR pathway inhibition. Cell Prolif 2024:e13773. [PMID: 39542458 DOI: 10.1111/cpr.13773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/16/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
The phosphatidylinositol 3-kinase/Protein Kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signalling pathway is pivotal in various cancers, including T-cell acute lymphoblastic leukaemia (T-ALL), a particularly aggressive type of leukaemia. This study investigates the effects of Neosetophomone B (NSP-B), a meroterpenoid fungal metabolite, on T-ALL cell lines, focusing on its anti-cancer mechanisms and therapeutic potential. NSP-B significantly inhibited the proliferation of T-ALL cells by inducing G0/G1 cell cycle arrest and promoting caspase-dependent apoptosis. Additionally, NSP-B led to the dephosphorylation and subsequent inactivation of the PI3K/AKT/mTOR signalling pathway, a critical pathway in cell survival and growth. Molecular docking studies revealed a strong binding affinity of NSP-B to the active site of AKT, primarily involving key residues crucial for its activity. Interestingly, NSP-B treatment also induced apoptosis and significantly reduced proliferation in phytohemagglutinin-activated primary human CD3+ T cells, accompanied by a G0/G1 cell cycle arrest. Importantly, NSP-B did not affect normal primary T cells, indicating a degree of selectivity in its action, targeting only T-ALL cells and activated T cells. In conclusion, our findings highlight the potential of NSP-B as a novel therapeutic agent for T-ALL, specifically targeting the aberrantly activated PI3K/AKT/mTOR pathway and being selective in action. These results provide a strong basis for further investigation into NSP-B's anti-cancer properties and potential application in T-ALL clinical therapies.
Collapse
Affiliation(s)
- Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Abdul W Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Muhammad Suleman
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Tamam El-Elimat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Qatar
| | - Feras Q Alali
- College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Ammira S Al Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
- College of Health Sciences, Qatar University, Doha, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Department of Dermatology & Venereology, Hamad Medical Corporation, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine-Qatar, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Laboratory of Animal Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
36
|
Kamala K, Ganapathy D, Sivaperumal P. Advancements in Cancer Therapy: Mycoviruses and Their Oncolytic Potential. Cell Biochem Biophys 2024:10.1007/s12013-024-01608-y. [PMID: 39535660 DOI: 10.1007/s12013-024-01608-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Recent advancements in cancer research focus on reducing treatment side effects while enhancing efficacy against medication resistance and tumor antigen detection. Genetic therapies utilizing microbes like bacteria, fungi, and viruses have garnered attention, with mycoviruses emerging as promising candidates. Particularly, the smallest fungal virus, Myco-phage, exhibits oncolytic properties by lysing cancer cells in the mouth, oral cavity, head, and neck without adverse effects. Genetically Modified Myco-phage (GmMP) adapts quickly to target cancer cells through cell membrane damage, inducing apoptosis and dendritic cell activation. Additionally, GmMP inhibits angiogenesis and modulates immune responses via CAR cells and immune checkpoints, potentially transforming cancer treatment paradigms with enhanced specificity and efficacy.
Collapse
Affiliation(s)
- Kannan Kamala
- Marine Microbial Research Lab, Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India
| | - Dhanraj Ganapathy
- Department of Prosthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Pitchiah Sivaperumal
- Centre for Marine and Aquatic Research (CMAR), Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 602105, India.
- Marine Biomedical Research Lab & Environmental Toxicology Unit, Cellular and Molecular Research Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India.
| |
Collapse
|
37
|
Bidgoli AD, Farmany A, Taheri M, Soleimani M, Nouri F. Preparation and characterization of calcium-doped graphene oxide-chitosan Nanocarrier to enhance the gene delivery in MCF-7 cell line. Sci Rep 2024; 14:27434. [PMID: 39521829 PMCID: PMC11550409 DOI: 10.1038/s41598-024-78958-3] [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: 03/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Gene delivery has emerged as a novel and effective method in the treatment of malignancies within medical interventions by applying nanotechnology. Consequently, the development of appropriate nanocarriers is a key focus of this research. Dynamic light scattering (DLS), fourier transform infrared (FT-IR) spectroscopy, x-ray diffraction (XRD), and thermal gravimetric analysis (TGA) were employed for the characterization of the synthesized nanocarrier. Furthermore, to assess the gene transfer capability of the nanocarrier, various techniques such as gel retardation assay, nuclease resistance assay, cytotoxicity assay, flow cytometry, and transfection were employed. The average particle size and zeta potential of the GO-CS@Ca nanocarrier were obtained as 319.8 nm and + 92.8 mv, respectively. In the gel retardation test, it was observed that pDNA was effectively condensed by the GO-CS@Ca nanocarrier. The results of the MTT assay indicated that both GO-CS@Ca nanocarrier and the GO-CS@Ca/pDNA nanoplex with low toxicity. In flow cytometry analysis, it was observed that the complexation of pDNA with the GO-CS@Ca nanocarrier resulted in effective gene delivery to the MCF-7 cell line and consequently increased apoptosis induction.
Collapse
Affiliation(s)
- Amirreza Diari Bidgoli
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abbas Farmany
- Dental Implant Research Center, Dental School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Taheri
- Department of Medical Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Meysam Soleimani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fatemeh Nouri
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
38
|
Nazari A, Osati P, Seifollahy Fakhr S, Faghihkhorasani F, Ghanaatian M, Faghihkhorasani F, Rezaei-Tazangi F, Pazhouhesh Far N, Shourideh A, Ebrahimi N, Aref AR. New Emerging Therapeutic Strategies Based on Manipulation of the Redox Regulation Against Therapy Resistance in Cancer. Antioxid Redox Signal 2024. [PMID: 39506926 DOI: 10.1089/ars.2023.0491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Background: Resistance to standard therapeutic methods, including chemotherapy, immunotherapy, and targeted therapy, remains a critical challenge in effective cancer treatment. Redox homeostasis modification has emerged as a promising approach to address medication resistance. Objective: This review aims to explore the mechanisms of redox alterations and signaling pathways contributing to treatment resistance in cancer. Methods: In this study, a comprehensive review of the molecular mechanisms underlying drug resistance governed by redox signaling was conducted. Emphasis was placed on understanding how tumor cells manage increased reactive oxygen species (ROS) levels through upregulated antioxidant systems, enabling resistance across multiple therapeutic pathways. Results: Key mechanisms identified include alterations in drug efflux, target modifications, metabolic changes, enhanced DNA damage repair, stemness preservation, and tumor microenvironment remodeling. These pathways collectively facilitate tumor cells' adaptive response and resistance to various cancer treatments. Conclusion: Developing a detailed understanding of the interrelationships between these redox-regulated mechanisms and therapeutic resistance holds potential to improve treatment effectiveness, offering valuable insights for both fundamental and clinical cancer research. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Ahmad Nazari
- Tehran University of Medical Science, Tehran, Iran
| | - Parisa Osati
- Department of Chemical Engineering, Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Siavash Seifollahy Fakhr
- Department of Biotechnology, Faculty of Applied Ecology, Agricultural Science and Biotechnology, Campus Hamar, Norway
| | - Ferdos Faghihkhorasani
- Department of Cardiology, Internal Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xian, Shaanxi Province, 710061, China
| | - Masoud Ghanaatian
- Master 1 Bio-Santé-Parcours Toulouse Graduate School of Cancer, Ageing and Rejuvenation (CARe), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Fereshteh Faghihkhorasani
- General Physician in Medicine Program,General Doctorate Degree of Yazd Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Science, Fasa, Iran
| | - Nazanin Pazhouhesh Far
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Amir Shourideh
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA and Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
39
|
Mustafa M, Ahmad R, Tantry IQ, Ahmad W, Siddiqui S, Alam M, Abbas K, Moinuddin, Hassan MI, Habib S, Islam S. Apoptosis: A Comprehensive Overview of Signaling Pathways, Morphological Changes, and Physiological Significance and Therapeutic Implications. Cells 2024; 13:1838. [PMID: 39594587 PMCID: PMC11592877 DOI: 10.3390/cells13221838] [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: 09/18/2024] [Revised: 10/16/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Cell survival and death are intricately governed by apoptosis, a meticulously controlled programmed cell death. Apoptosis is vital in facilitating embryonic development and maintaining tissue homeostasis and immunological functioning. It is a complex interplay of intrinsic and extrinsic signaling pathways that ultimately converges on executing the apoptotic program. The extrinsic pathway is initiated by the binding of death ligands such as TNF-α and Fas to their respective receptors on the cell surface. In contrast, the intrinsic pathway leads to increased permeability of the outer mitochondrial membrane and the release of apoptogenic factors like cytochrome c, which is regulated by the Bcl-2 family of proteins. Once activated, these pathways lead to a cascade of biochemical events, including caspase activation, DNA fragmentation, and the dismantling of cellular components. Dysregulation of apoptosis is implicated in various disorders, such as cancer, autoimmune diseases, neurodegenerative disorders, and cardiovascular diseases. This article focuses on elucidating the molecular mechanisms underlying apoptosis regulation, to develop targeted therapeutic strategies. Modulating apoptotic pathways holds immense potential in cancer treatment, where promoting apoptosis in malignant cells could lead to tumor regression. This article demonstrates the therapeutic potential of targeting apoptosis, providing options for treating cancer and neurological illnesses. The safety and effectiveness of apoptosis-targeting drugs are being assessed in ongoing preclinical and clinical trials (phase I-III), opening the door for more effective therapeutic approaches and better patient outcomes.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India; (M.M.); (R.A.); (S.S.); (M.)
| | - Rizwan Ahmad
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India; (M.M.); (R.A.); (S.S.); (M.)
| | - Irfan Qadir Tantry
- Department of Biochemistry, School of Biological Sciences, University of Kashmir, Srinagar 190006, India;
| | - Waleem Ahmad
- Department of Medicine, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India;
| | - Sana Siddiqui
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India; (M.M.); (R.A.); (S.S.); (M.)
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202001, India; (M.A.); (K.A.)
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202001, India; (M.A.); (K.A.)
| | - Moinuddin
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India; (M.M.); (R.A.); (S.S.); (M.)
| | - Md. Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India;
| | - Safia Habib
- Department of Biochemistry, Faculty of Medicine, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India; (M.M.); (R.A.); (S.S.); (M.)
| | - Sidra Islam
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
40
|
Bogdanov FB, Balakhonov RY, Volkov ES, Sonin IV, Andreeva OE, Sorokin DV, Piven YA, Scherbakov AM, Shirinian VZ. Photochemical Metal-Free synthesis and biological Assessment of isocryptolepine analogues targeting estrogen receptor Alpha in breast cancer cells. Bioorg Chem 2024; 153:107942. [PMID: 39515131 DOI: 10.1016/j.bioorg.2024.107942] [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: 08/23/2024] [Revised: 11/01/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
The aim of this study was to develop a new series of isocryptolepines and evaluate their antiproliferative and antiestrogenic activities on cancer cells. A series of isocryptolepine derivatives were synthesized using developed one-pot photochemical, metal-free protocol, employing readily available 2-arylindoles as starting compounds. The resulting isocryptolepines demonstrated (sub)micromolar inhibitory activity against selected breast cancer cell lines. The IC50 values of lead compound 3c against hormone-dependent breast cancer types (MCF7 and T47D) were 0.3 μM and 0.12 μM, respectively, and significantly greater than 3 μM against estrogen receptor α (ERα)-deficient cell lines, MDA-MB-231 and HCC1954, respectively. To assess the antiestrogenic potency of compound 3c, MCF7 cells were transfected with a plasmid containing a luciferase reporter gene under the control of an estrogen-responsive element (ERE), creating the MCF7/ERE-LUC cell subline. In these cells, luciferase activity was induced by the natural ERα ligand, 17β-estradiol (E2). Compound 3c inhibited luciferase activity by 50 % at a concentration of 0.12 μM, highlighting its potent inhibitory effect on ERα. Molecular modeling further indicated that compound 3c could directly bind to ERα. Compound 3c induced apoptosis, as evidenced by PARP cleavage and downregulation of p-Bcl-2 and Bcl-2, and demonstrated synergistic effects in combination with the chemotherapeutic agent 5-fluorouracil. Compound 3c also showed selectivity towards hormone-dependent breast cancer cells, likely targeting ERα - a key driver in this cancer subtype. In summary, we report the development of a first-in-class antiestrogenic isocryptolepine with notable pro-apoptotic efficacy.
Collapse
Affiliation(s)
- F B Bogdanov
- Department of Experimental Tumor Biology, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, 115522 Moscow, Russia; Faculty of Medicine, Moscow State University, Lomonosov prospect 27 bldg.1, 119991 Moscow, Russia.
| | - R Yu Balakhonov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia.
| | - E S Volkov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia.
| | - I V Sonin
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia.
| | - O E Andreeva
- Department of Experimental Tumor Biology, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, 115522 Moscow, Russia.
| | - D V Sorokin
- Department of Experimental Tumor Biology, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, 115522 Moscow, Russia.
| | - Yu A Piven
- Institute of Bioorganic Chemistry, National Academy of Sciences of Belarus, Akad. Kuprevicha st. 5/2, Minsk 220084, Belarus.
| | - A M Scherbakov
- Department of Experimental Tumor Biology, N.N. Blokhin National Medical Research Center of Oncology, Kashirskoye shosse 24 bldg.15, 115522 Moscow, Russia; Gause Institute of New Antibiotics, Bol'shaya Pirogovskaya ulitsa 11, 119021 Moscow, Russia.
| | - V Z Shirinian
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Leninsky prospect 47, 119991 Moscow, Russia.
| |
Collapse
|
41
|
Areewong S, Suppramote O, Prasopporn S, Jirawatnotai S. Exploiting acquired vulnerability to develop novel treatments for cholangiocarcinoma. Cancer Cell Int 2024; 24:362. [PMID: 39501277 PMCID: PMC11539612 DOI: 10.1186/s12935-024-03548-2] [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: 07/14/2024] [Accepted: 10/26/2024] [Indexed: 11/08/2024] Open
Abstract
Cholangiocarcinoma (CCA) presents a formidable therapeutic challenge due to its extensive heterogeneity and plasticity, which inevitably lead to acquired resistance to current treatments. However, recent evidence suggests that acquired drug resistance is associated with a fitness cost resulting from the myriad of acquired alterations under the selective pressure of the primary treatment. Consequently, CCA patients with acquired resistance are more susceptible to alternative therapies that are ineffective as monotherapies. This phenomenon, termed "acquired vulnerability," has garnered significant interest in drug development, as the acquired alterations could potentially be exploited therapeutically. This review elucidates the modes of acquired vulnerability, methods for identifying and exploiting acquired vulnerabilities in cancer (particularly in CCA), and strategies to enhance the clinical efficacy of drug combinations by leveraging the principle of acquired vulnerability. Identifying acquired vulnerabilities may pave the way for novel drug combinations to effectively treat highly heterogeneous and adaptable malignancies such as CCA.
Collapse
Affiliation(s)
- Sirayot Areewong
- Siriraj Center of Research Excellence (SiCORE) for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, 2 Wanglang Rd., 11th Floor Srisavarindhira Building, Bangkok Noi, 10700, Bangkok, Thailand
| | - Orawan Suppramote
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, 906 Kampangpetch 6 Rd., Talat Bang Khen, Lak Si, 10210, Bangkok, Thailand
| | - Sunisa Prasopporn
- Siriraj Center of Research Excellence (SiCORE) for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, 2 Wanglang Rd., 11th Floor Srisavarindhira Building, Bangkok Noi, 10700, Bangkok, Thailand
| | - Siwanon Jirawatnotai
- Siriraj Center of Research Excellence (SiCORE) for Systems Pharmacology, Department of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, 2 Wanglang Rd., 11th Floor Srisavarindhira Building, Bangkok Noi, 10700, Bangkok, Thailand.
- Faculty of Pharmacy, Silpakorn University, 6 Ratchamankanai Road., Phra Pathom Chedi Sub-district, Mueang District, 73000, Nakhon Pathom, Thailand.
| |
Collapse
|
42
|
Cai JL, Wang JJ, Zhang Y, Gao H, Huang W, Cai YJ, Jia WX, Chen X, Sun HY. Characterization, expression and functional analysis of Hsp40 during LPS challenge in blood parrot Amphilophus citrinellus ×Vieja melanura. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109910. [PMID: 39299406 DOI: 10.1016/j.fsi.2024.109910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/27/2024] [Accepted: 09/14/2024] [Indexed: 09/22/2024]
Abstract
Heat shock protein 40 belonging to heat shock protein family plays an important role in the immune responses of organisms. In this study, the full length cDNA of Hsp40 was 2426 bp including a 1368 bp open reading frame (ORF) encoding 455 amino acids with a molecular weight of 49.16 kDa and a theoretical isoelectric point of 9.34 in blood parrot Vieja synspila ♀ × Amphilophus citrinellus ♂, an important ornamental fish in China. It had three conserved domains DnaJ, CRR and DnaJ_C. Phylogenetic analysis showed that the sequence of Hsp40 among species was conserved, and the blood parrot Hsp40 was closely related to Neolamprologus brichardi. Blood parrot Hsp40 mRNA could be detected in all of the tissues examined and mainly distributed in the cytoplasm. The expression of Hsp40 was upregulated during lipopolysaccharide (LPS) challenge. Upregulated Hsp40 inhibited the activity of nuclear factor κB (NF-κB) and activated protein 1 (AP-1) and reduced the ratio of Bax/Bcl-2 mRNA expression. This study provides a theoretical basis for further exploring the role of Hsp40 gene in the anti-bacterial immunity of blood parrot.
Collapse
Affiliation(s)
- Jie-Li Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jun-Jie Wang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China
| | - Yue Zhang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hui Gao
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei Huang
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Yi-Jie Cai
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China
| | - Wei-Xin Jia
- School of Life Sciences, South China Normal University, Guangzhou, Guangdong, China
| | - Xiao Chen
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Hong-Yan Sun
- College of Marine Sciences, South China Agricultural University, Guangzhou, Guangdong, China; College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Gholami F, Seyedalipour B, Heidari-Kalvani N, Nabi-Afjadi M, Yaghoubzad-Maleki M, Fathi Z, Alipourfard I, Barjesteh F, Bahreini E. Catharanthine, an anticancer vinca alkaloid: an in silico and in vitro analysis of the autophagic system as the major mechanism of cell death in liver HepG2 cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8879-8892. [PMID: 38856913 DOI: 10.1007/s00210-024-03191-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/27/2024] [Indexed: 06/11/2024]
Abstract
Catharanthine, a component of the anticancer drug vinblastine along with vindoline, disrupts the cell cycle by interfering with mitotic spindle formation. Apart from their antioxidant properties, vinca alkaloids like catharanthine inhibit phosphodiesterase activity and elevate intracellular cAMP levels. The aim of this study was to investigate how catharantine affects apoptosis and autophagy. This study conducted experiments on HepG2 liver carcinoma cells with varying doses of catharanthine to evaluate cell death rates and viability and determine the IC50 concentration via MTT assays. The apoptotic and autophagic effects of catharanthine were assessed using flow cytometry with annexin V and PI staining, while the expression of autophagy-related genes was analyzed through quantitative PCR. Additionally, molecular docking and molecular dynamics simulations were employed to further investigate catharanthine's impact on autophagy mechanisms. The study showed that catharanthine reduced oxidative stress and triggered apoptosis in HepG2 cells in a dose-dependent manner. Catharanthine also upregulated the expression of autophagy-related genes like LC3, Beclin1, and ULK1. Notably, catharanthine increased sirtuin-1 levels, a known autophagy inducer, while decreasing Akt expression compared to untreated cells. Molecular docking results indicated rapamycin had a stronger binding affinity with FRB (-10.7 KJ/mol-1) than catharanthine (-7.3 KJ/mol-1). Additionally, molecular dynamics simulations revealed that catharanthine interacted effectively with the FRB domain of mTOR, displaying stability and a strong binding affinity, although not as potent as rapamycin. In summary, besides its cytotoxic and pro-apoptotic effects, catharanthine activates autophagy signaling pathways and induces autophagic necrosis by inhibiting mTOR.
Collapse
Affiliation(s)
- Farnoosh Gholami
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Bagher Seyedalipour
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Nafiseh Heidari-Kalvani
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Yaghoubzad-Maleki
- Division of Biochemistry, Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zeinab Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Fereshte Barjesteh
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elham Bahreini
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
44
|
Anastasiou IA, Sarantis P, Rebelos E, Eleftheriadou I, Tentolouris KN, Katsaouni A, Koustas E, Kokala V, Karamouzis MV, Tentolouris N. l-Securinine Induces ROS-Dependent Apoptosis on Pancreatic Cancer Cells via the PI3K/AKT/mTOR Signaling Pathway. J Biochem Mol Toxicol 2024; 38:e70036. [PMID: 39467148 DOI: 10.1002/jbt.70036] [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: 04/30/2024] [Revised: 09/03/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
Accumulating evidence has shown that l-securinine can, in certain circumstances, suppress tumor development by elevating reactive oxygen species (ROS) levels. The current work set out to examine l-securinine's apoptotic effects on HuP-T3 cells as well as any potential underlying molecular mechanism(s) that could explain its action as an anticancer agent. In this study, we used 1.2B4 cells as a control human cell line to verify our findings. Hup-T3 and 1.2B4 cells were cultured with a medium containing the following dilutions of l-securinine: 1-10 μΜ for up to 72 h. We examined the viability and proliferation levels of cells in both cell lines. Then, we measured only 1.2B4 insulin levels and content. We also quantified cell apoptosis, cell cycle levels, and the intracellular reactive oxygen species on HuP-T3 and 1.2B4. Afterwards, we performed a real-time quantitative polymerase chain reaction and western blot analysis. Our results demonstrated that l-securinine inhibited both proliferation and growth of Hup-T3 cells, showing inhibitory and antiproliferative activity in comparison with the control group. In addition, l-securinine had no impact on the proliferation and growth of 1.2B4 cells, nor on their insulin levels and content. By boosting ROS production, and inhibiting the PI3K/AKT/mTOR signaling pathway, l-securinine induced apoptosis on HuP-T3 cells. Pancreatic cancer was successfully inhibited by l-securinine in vitro. l-securinine triggers ROS-dependent apoptosis on pancreatic cancer cells while inhibiting the PI3K/AKT/mTOR signaling pathway. These findings suggest that l-securinine holds promise as a potential lead for future drug development in the fight against pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Ioanna A Anastasiou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Eleni Rebelos
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Turku PET Centre, University of Turku, Turku, Finland
| | - Ioanna Eleftheriadou
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Konstantinos N Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasia Katsaouni
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Vasileia Kokala
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic Internal Medicine, Diabetes Center, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| |
Collapse
|
45
|
Raposo A, Raheem D, Zandonadi RP, Suri N, Olukosi A, de Lima BR, Carrascosa C, Sharifi-Rad J, Ryu HB, Han H, Calina D. Anethole in cancer therapy: Mechanisms, synergistic potential, and clinical challenges. Biomed Pharmacother 2024; 180:117449. [PMID: 39326099 DOI: 10.1016/j.biopha.2024.117449] [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: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer remains a major global health challenge, prompting the search for effective and less toxic treatments. Anethole, a bioactive compound found in essential oils of anise and fennel, commonly used as a food preservative, has recently garnered attention for its potential anti-cancer properties. This comprehensive review aims to systematically assess the anti-cancer effects of anethole, elucidating its mechanisms of action, pharmacokinetics, bioavailability, and synergistic potential with conventional cancer therapies. A detailed literature search was conducted across databases including PubMed, Embase, Scopus, Science Direct, Web of Science, and Google Scholar. Criteria for inclusion were experimental studies in peer-reviewed journals focusing on the anti-cancer properties of anethole. Extracted data included study design, intervention specifics, measured outcomes, and mechanistic insights. Anethole demonstrates multiple anti-cancer mechanisms, such as inducing apoptosis, causing cell cycle arrest, exhibiting anti-proliferative and anti-angiogenic effects, and modulating critical signaling pathways including NF-κB, PI3K/Akt/mTOR, and caspases. It enhances the efficacy of chemotherapeutic agents like cisplatin and doxorubicin while reducing their toxicity. In vitro and in vivo studies have shown its effectiveness against various cancers, including breast, prostate, lung, and colorectal cancers. Anethole shows significant potential as an anti-cancer agent, with its multi-faceted mechanisms of action and ability to synergize with existing chemotherapy. Further clinical research is essential to fully understand its therapeutic potential and application in oncology.
Collapse
Affiliation(s)
- António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Dele Raheem
- Arctic Centre, University of Lapland, Rovaniemi 96101, Finland
| | - Renata Puppin Zandonadi
- University of Brasília, Faculty of Health Sciences, Nutrition Department, Campus Universitário Darcy Ribeiro, Brasília 70910-900, Brazil
| | - Narinder Suri
- Department of Chemistry, Moi University, P.O. Box 4606, Eldoret 30100, Kenya
| | - Adeola Olukosi
- Department of Medical Biochemistry, Eko University of Medical Sciences, Lagos 102004, Nigeria
| | - Bernardo Romão de Lima
- University of Brasília, Faculty of Health Sciences, Nutrition Department, Campus Universitário Darcy Ribeiro, Brasília 70910-900, Brazil
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, Arucas 35413, Spain
| | - Javad Sharifi-Rad
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico; Department of Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea.
| | - Hyungseo Bobby Ryu
- Foodservice & Culinary Art, Department of the College of Health Sciences, Kyungnam University, 7 Kyungnamdaehak-ro, Masanhappo-gu, Changwon-si, Gyeongsangnam-do 51767, Republic of Korea.
| | - Heesup Han
- College of Hospitality and Tourism Management, Sejong University, 209 Neungdong-ro, Gwangjin-gu, Seoul 05006, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova 200349, Romania
| |
Collapse
|
46
|
El-Masry TA, El-Nagar MMF, Oriquat GA, Alotaibi BS, Saad HM, El Zahaby EI, Ibrahim HA. Therapeutic efficiency of Tamoxifen/Orlistat nanocrystals against solid ehrlich carcinoma via targeting TXNIP/HIF1-α/MMP-9/P27 and BAX/Bcl2/P53 signaling pathways. Biomed Pharmacother 2024; 180:117429. [PMID: 39293373 DOI: 10.1016/j.biopha.2024.117429] [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: 06/27/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/20/2024] Open
Abstract
BACKGROUND Orlistat (Orli) is an anti-obesity medication that has been approved by the US Food and Drug Administration. It has relatively limited oral bioavailability with promising inhibitory effects on cell proliferation as well as reducing the growth of tumors. AIMS This investigation was done to evaluate the potential protective effect of Tamoxifen/Orlistat nanocrystals alone or in combination against Solid Ehrlich Carcinoma (SEC) and to clarify the possible underlying influences. MATERIALS AND METHODS The liquid antisolvent precipitation technique (bottom-up technology) was utilized to manufacture Orlistat Nanocrystals. To explore potential causes for the anti-tumor action, female Swiss Albino mice bearing SEC were randomly assigned into five equal groups (n = 6). Group 1: Tumor control group, group 2: Tam group: tamoxifen (0.01 g/kg, IP), group 3: Free-Orli group: orlistat (0.24 g/kg, IP), group 4: Nano-Orli: orlistat nanocrystals (0.24 g/kg, IP), group 5: Tam-Nano-Orli: Both doses of Tam and Nano-Orli. All treatments were administered for 16 days. KEY FINDINGS The untreated mice showed development in the tumor volume and weight. As well as histopathology results from these mice revealed many tumor large cells as well as solid sheets of malignant cells. Also, untreated mice showed raised VEGF and TGF-1beta content. Moreover, results of gene expression in the SEC-bearing mice noted upregulation in HIF-1α, MMP-9, Bcl-2, and P27 gene expression and downregulation of TXNIP, BAX, and P53 gene expression. On the other hand, administrated TAM, Free-Orli, Nano-Orli, and a combination of Tam-Nano-Orli distinctly suppressed the tumor effects on estimated parameters with special reference to Tam-Nano-Orli. SIGNIFICANCE The developed Tamoxifen/Orlistat nanocrystals combination could be considered a promising approach to augment antitumor effects.
Collapse
Affiliation(s)
- Thanaa A El-Masry
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Maysa M F El-Nagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Ghaleb Ali Oriquat
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan.
| | - Badriyah S Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Cairo 51511, Egypt.
| | - Enas I El Zahaby
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt.
| | - Hanaa A Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| |
Collapse
|
47
|
Fernández-Maestre I, Cai SF, Levine RL. A View of Myeloid Transformation through the Hallmarks of Cancer. Blood Cancer Discov 2024; 5:377-387. [PMID: 39422551 PMCID: PMC11528188 DOI: 10.1158/2643-3230.bcd-24-0009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/30/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
The development of myeloid malignancies is influenced by a range of cell-intrinsic and cell-extrinsic factors, which can be conceptualized using the hallmarks of cancer. Although many facets of myeloid transformation are similar to those in solid tumors, there are also notable differences. Unlike solid tumors, hematologic malignancies typically exhibit fewer genetic mutations, which have been well characterized. However, understanding the cell-extrinsic factors contributing to myeloid malignancies can be challenging due to the complex interactions in the hematopoietic microenvironment. Researchers need to focus on these intricate factors to prevent the early onset of myeloid transformation and develop appropriate interventions. Significance: Myeloid malignancies are common in the elderly, and acute myeloid leukemia has an adverse prognosis in older patients. Investigating cell-extrinsic factors influencing myeloid malignancies is crucial to developing approaches for preventing or halting disease progression and predicting clinical outcomes in patients with advanced disease. Whereas successful intervention may require targeting various mechanisms, understanding the contribution of each cell-extrinsic factor will help prioritize clinical targets.
Collapse
Affiliation(s)
- Inés Fernández-Maestre
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Louis V. Gerstner Jr Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sheng F. Cai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering, Cancer Center, New York, New York
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
- Leukemia Service, Department of Medicine and Center for Hematologic Malignancies, Memorial Sloan Kettering, Cancer Center, New York, New York
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
48
|
Rahmani F, Ajoudanifar H, Arbab Soleimani N, Imani Fooladi AA. Targeted therapies in HER2-positive breast cancer with receptor-redirected Arazyme-linker-Herceptin as a novel fusion protein. Breast Cancer 2024; 31:1101-1113. [PMID: 39122876 DOI: 10.1007/s12282-024-01625-y] [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: 12/15/2023] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Targeted treatment of different types of cancers through highly expressed cancer cell surface receptors by fusion proteins is an efficient method for cancer therapy. The HER2 receptor is a member of the tyrosine kinase receptors family, which plays a notable role in breast cancer tumor development. About 25-30% of breast cancers overexpress human epidermal growth factor receptor 2 (HER2). METHODS In this study, we evaluated the particulars of a designed recombinant protein formed by HER2-specific Mab Herceptin linked with Arazyme on a HER2-overexpressing breast cancer cell line (SKBR3). Arazyme, a metalloprotease produced by Serratia proteamaculans was fused to the variable area of light and heavy chains of the Herceptin. The cytotoxic assay of the Arazyme-linker-Herceptin in the SKBR3 and MDA-MB-468 cells was evaluated by the MTT and flow cytometry techniques. The Caspase‑3 activity determination and adhesion assay were performed to evaluate the antitumor activity of the Arazyme-linker-Herceptin against SKBR3 cells. Furthermore, RT-PCR was used to measure the expression levels of the Bcl-2, Bax, MMP2, MMP9, and RIP3 genes. RESULTS The Arazyme-linker-Herceptin showed higher cytotoxicity in SKBR3 cells compared to MDA-MB-468 cells. In addition, flow cytometry results revealed that the Arazyme-linker-Herceptin can significantly induce apoptosis in the HER2-overexpressing breast cancer cell line (SKBR3), which was confirmed by Bax upregulation and the decrease in adhesion of tumor cells and MMP2/MMP9. CONCLUSION The findings of this study demonstrated that the Arazyme-linker-Herceptin induced apoptosis and decreased metastatic genes in SKBR3 cells; however, further research is required to confirm the effectiveness of the fusion protein.
Collapse
Affiliation(s)
- Farideh Rahmani
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | - Hatef Ajoudanifar
- Department of Microbiology, Damghan Branch, Islamic Azad University, Damghan, Iran
| | | | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Kamel AG, Sabet S, El-Shibiny A. Potential mitochondrial ROS-mediated damage induced by chitosan nanoparticles bee venom-loaded on cancer cell lines. Int J Biol Macromol 2024; 279:135362. [PMID: 39245116 DOI: 10.1016/j.ijbiomac.2024.135362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Recently, numerous studies have confirmed the importance of chitosan nanoparticles (CNP) as a viable drug delivery carrier for increasing the efficacy of anticancer drugs in cancer treatment. It is a macromolecule and natural biopolymer compound, more stable and safer in use than metal nanoparticles. Bee venom (BV), a form of defense venom, has been shown to have anti-tumor, neuroprotective, anti-inflammatory, analgesic, and anti-infectivity properties. Moreover, the regulation of cell death has been linked to reactive oxygen species (ROS)-mediated cell apoptosis, which induces mitochondrial damage and ER stress through oxidative stress events. Therefore, this study aimed to illustrate the ROS-mediated effect on the cancer cells treatment with CNP-loaded BV (CNP-BV) and explained the adverse effects of ROS generation on Mitochondria and ER. We have found that the targeted CNP-BV were high in cytotoxicity against MCF-7 (IC50 437.2 μg/mL) and HepG2 (IC50 109.5 μg/mL) through the induction of massive generation of ROS, which in turn results in activating the mitochondrial cascade and ER stress. These results highlighted the role of ROS generation in inducing apoptosis in cancer cells.
Collapse
Affiliation(s)
- Azza G Kamel
- Center for Microbiology and Phage Therapy, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Salwa Sabet
- Department of Zoology, Faculty of Science, Cairo University, Giza, Egypt
| | - Ayman El-Shibiny
- Center for Microbiology and Phage Therapy, Zewail City of Science and Technology, Giza 12578, Egypt; Faculty of Environmental Agricultural Sciences, Arish University, Arish 45511, Egypt.
| |
Collapse
|
50
|
Majumder A, Bano S, Nayak KB. The Pivotal Role of One-Carbon Metabolism in Neoplastic Progression During the Aging Process. Biomolecules 2024; 14:1387. [PMID: 39595564 PMCID: PMC11591851 DOI: 10.3390/biom14111387] [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: 09/18/2024] [Revised: 10/29/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
One-carbon (1C) metabolism is a complex network of metabolic reactions closely related to producing 1C units (as methyl groups) and utilizing them for different anabolic processes, including nucleotide synthesis, methylation, protein synthesis, and reductive metabolism. These pathways support the high proliferative rate of cancer cells. While drugs that target 1C metabolism (like methotrexate) have been used for cancer treatment, they often have significant side effects. Therefore, developing new drugs with minimal side effects is necessary for effective cancer treatment. Methionine, glycine, and serine are the main three precursors of 1C metabolism. One-carbon metabolism is vital not only for proliferative cells but also for non-proliferative cells in regulating energy homeostasis and the aging process. Understanding the potential role of 1C metabolism in aging is crucial for advancing our knowledge of neoplastic progression. This review provides a comprehensive understanding of the molecular complexities of 1C metabolism in the context of cancer and aging, paving the way for researchers to explore new avenues for developing advanced therapeutic interventions for cancer.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Shabana Bano
- Department of Medicine, University of California, San Francisco, CA 94158, USA
| | - Kasturi Bala Nayak
- Quantitative Biosciences Institute, Department of Medicine, University of California, San Francisco, CA 94158, USA
| |
Collapse
|