1
|
Wang TH, Chou LF, Shen YW, Lin NC, Shih YH, Shieh TM. Mechanistic insights into temoporfin-based photodynamic therapy: Ferroptosis as a critical regulator under normoxic and hypoxic conditions in head and neck cancer. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 267:113165. [PMID: 40267720 DOI: 10.1016/j.jphotobiol.2025.113165] [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: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Temoporfin is a second-generation photosensitizer used in photodynamic therapy (PDT) for the clinical treatment of head and neck cancer. However, its role in inhibiting cancer cell viability under normoxic and hypoxic conditions remains unclear. The oral squamous cell carcinoma (OSCC) cell lines, SAS and OECM-1 were cultured under normoxic or hypoxic conditions to investigate temoporfin-based PDT-induced cell death and the underlying mechanisms. Cell viability was analyzed using the MTT assay. Intracellular reactive oxygen species (ROS) levels, cell apoptosis, intracellular ROS, iron levels, lipid peroxidation, and glutathione (GSH) levels were assessed by flow cytometry. The expression of proteins related to oxidative stress, apoptosis, autophagy, and ferroptosis was verified by western blotting. Results showed that increasing the temoporfin dose, absorption time, and illumination time was positively correlated with the inhibition of oral cancer cells. Hypoxic conditions attenuated the toxicity of temoporfin in cancer cells. OECM-1 cells were more sensitive to temoporfin than SAS cells. Temoporfin-based PDT-induced ROS exhibited similar trends to oxidative stress-inducing enzymes under both normoxic and hypoxic conditions and triggered cell autophagy and ferroptosis. Administration of the ferroptosis inhibitor BRD4770 under normoxic conditions reversed temoporfin-based PDT-induced reductions in glutathione peroxidase 4 (GPx4), increasing in light chain 3-II (LC3-II) and cleaved poly (ADP-ribose) polymerase (cleaved-PARP). This study confirms that hypoxia weakens the anticancer effects of temoporfin-based PDT and that ferroptosis plays a key role in temoporfin-based PDT-mediated cancer cell inhibition.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Biobank, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Health Industry Technology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, No. 261, Wenhua 1st Rd., Guishan District, Taoyuan 33303, Taiwan; Liver Research Center, Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital at Linkou, No.5, Fuxing Street, Guishan District, Taoyuan 33305, Taiwan; Graduate Institute of Natural Products, Chang Gung University, No.259, Wenhua 1st Rd., Guishan District, Taoyuan 33302, Taiwan.
| | - Li-Fang Chou
- Kidney Research Center, Chang Gung Memorial Hospital, Tao-Yuan 33305, Taiwan
| | - Yen-Wen Shen
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan.
| | - Nan-Chin Lin
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, No.542, Sec 1 Chung-shan Rd., Changhua 500. Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, No.500, Lioufeng Rd., Wufeng, Taichung 413305, Taiwan.
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, No. 100, Section 1, Jingmao Road, Beitun District, Taichung 406040, Taiwan; Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong St. Beitou District, Taipei 112304, Taiwan.
| |
Collapse
|
2
|
Zhang S, Zhu X, Yu W, Yu Y, Qian L, Chen Z, Peng Z, Gao L, Chen L, Chen J. Self-Assembly of Ru3-Aptamer Nanoparticles Triggers Pyroptosis through Photoredox Catalysis of NADH and Lysosomal Disruption. J Med Chem 2025. [PMID: 40372006 DOI: 10.1021/acs.jmedchem.5c00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Photodynamic therapy (PDT) can induce tumor cell death. Ru3, a metal-based photosensitizer, features a high positive charge, a long triplet excited-state lifetime, and an excellent PDT activity. The aptamer AS1411, known for its ability to selectively bind to nucleolin (which is overexpressed in tumor cells), self-assembled with Ru3 into nanoparticles termed Ru3ApNPs. These nanoparticles specifically target SiHa tumor cells. Upon light irradiation, Ru3ApNPs increase intracellular ROS levels, catalyze NADH redox reactions, and induce lysosomal disruption, ultimately triggering pyroptosis in tumor cells. Notably, Ru3ApNPs demonstrate excellent tumor penetration in 3D multicellular spheroids (MCSs) of SiHa cells and effectively inhibit their growth under light exposure. Ru3ApNPs exhibit a mechanism of action distinct from that of traditional PDT. Furthermore, under light irradiation, Ru3ApNPs can effectively inhibit the growth of distant tumors and induce systemic immune responses in mice. Our data suggest that Ru3ApNPs can be developed as promising targeted therapeutic agents in the future.
Collapse
Affiliation(s)
- Shenting Zhang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Wenzhu Yu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Yunjiang Yu
- School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang 524048, P. R. China
| | - Li Qian
- Youjiang Medical College for Nationalities, Baise 533000, P. R. China
| | - Zhikai Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Zitong Peng
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Lijun Gao
- School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang 524048, P. R. China
| | - Lanmei Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Jincan Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| |
Collapse
|
3
|
Mukherjee S, Banik SK, Chakraborty S, Das T, Choudhury MD, Tripathi A. Bryophyllum pinnatum Induces p53-Dependent Apoptosis of Colorectal Cancer Cells via Increased Intracellular ROS and G2/M Cell-Cycle Arrest In Vitro and Validated in Silico by Molecular Docking. Cell Biol Int 2025; 49:534-554. [PMID: 39992739 DOI: 10.1002/cbin.70004] [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: 09/27/2024] [Revised: 01/31/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025]
Abstract
Chemotherapy, radiotherapy and surgical treatments of cancer having several limitations and toxic side-effects, have led researchers to focus towards development of alternative natural plant-based therapeutics that can reduce disease severity. The present research work is mainly focussed towards identifying molecular mechanisms of apoptosis of colorectal cancer cells (HCT116) by perennial herb Bryophyllum pinnatum leaf-extract via both in vitro experimentations and in silico analysis. B. pinnatum leaf extract induced highest cytotoxicity at lowest dose (IC50:0.01 mg/mL) against HCT116 cells with 49.5% (p < 0.0001) cellular death, in comparison to other cancer cell lines. It has arrested HCT116 cell populations at G2/M cell-cycle phase and led to 10 folds (p < 0.0001) and 5.5 folds (p < 0.0001) increased intracellular ROS production in treated groups. ROS production might have led to significant 34.23% and 21.03% (p < 0.0001) apoptosis in treated cells, proved in vitro and in silico, with significant upregulation of p53 (p < 0.0001), BAX (p = 0.0252), CASPASE3 (p < 0.0001) and downregulation of BCL2 (p = 0.0058), leading to increased nuclear p53 (p = 0.0002) accumulation in treated cells, suggesting that the leaf-extract might have induced p53-dependent apoptosis of colorectal cancer cells. The phyto-extract also possess significant gene-modulatory potential as evident from qRT-PCR analysis of oncogenes and tumor suppressor genes. Leaf's bioactive phyto-constituents were elucidated by GC-MS and HPLC-ESI/MS analysis. In silico STITCH analysis provided significant network interactions between these bioactive phyto-compounds and studied proteins. Further Molecular Docking studies revealed strong binding between such docked complexes. Also, predicted major bioactive phyto-constituents of B. pinnatum leaf-extract such as Quercetin, Morin and β-Sitosterol have induced significant (p < 0.0001) apoptosis and increased intracellular ROS, validating their in silico interactions with studied proteins of HCT116 cells. All these studies together demonstrated ability of B. pinnatum to be used as a suitable natural phyto-therapeutic agent for development of chemo-preventive medications against colorectal cancer.
Collapse
Affiliation(s)
- Sumoyee Mukherjee
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, India
| | - Sheuli Kangsa Banik
- Department of Life Science and Bioinformatics, Assam University, Silchar, India
| | | | | | | | - Anusri Tripathi
- Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical, Medicine, Kolkata, India
| |
Collapse
|
4
|
Saha S, Tandon R, Sanku J, Kumari A, Shukla R, Srivastava N. siRNA-based Therapeutics in Hormone-driven Cancers: Advancements and benefits over conventional treatments. Int J Pharm 2025; 674:125463. [PMID: 40081431 DOI: 10.1016/j.ijpharm.2025.125463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Hormone-related cancers, also known as hormone-sensitive or hormone-dependent cancers, rely on hormones such as estrogen, testosterone, and progesterone for growth. These malignancies, including breast, pituitary, thyroid, ovarian, uterine, cervical, and prostate cancers, often exhibit accelerated progression in response to hormonal signaling. Small interfering RNA (siRNA) has emerged as a groundbreaking gene suppression therapy since the FDA approval of its first product in 2018. With over 200 ongoing clinical trials, siRNA is being actively explored as a targeted treatment for hormone-related cancers. Its ability to silence specific oncogenes offers significant advantages over conventional therapies, which are often associated with toxicity, resistance, and non-specific targeting. However, challenges in siRNA delivery remain a major barrier to its clinical translation, limiting its ability to reach target cells effectively. This review evaluates the potential of siRNA in hormone-related cancers, addressing the shortcomings of traditional treatments while examining novel strategies to enhance siRNA delivery and overcome tumor microenvironment obstacles. Notably, no existing literature comprehensively consolidates siRNA-based therapies for these cancers, emphasizing the importance of this manuscript in bridging current knowledge gaps and advancing the translational application of siRNA therapeutics.
Collapse
Affiliation(s)
- Sayani Saha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Jhansi Sanku
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Anchala Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India.
| |
Collapse
|
5
|
Singh S, Singh V, Singh R, Gouri V, Koch B, Samant M. Synergistic combination of doxorubicin with fisetin for the treatment of lymphoma. Eur J Pharmacol 2025; 992:177361. [PMID: 39929420 DOI: 10.1016/j.ejphar.2025.177361] [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: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
Lymphoma is a common cancer of the lymphatic system, and its treatment presents considerable clinical difficulties due to the constraints of existing medicines. Anticancer drug such as Doxorubicin (DOX) is an effective chemotherapeutic drug that is frequently used to treat lymphoma and other cancers; however, it is linked with considerable toxicities. Fisetin, a naturally occurring flavonoid, exhibits anticancer properties and has the potential to augment the therapeutic effects of DOX. This study explores the synergistic effects of combining DOX with fisetin in the treatment of lymphoma. The combination of DOX and fisetin significantly inhibits cell viability, induced membrane blabbing, chromatin condensation, and promoted apoptosis compared to monotherapies. The study also showed that the synergistic effect of fisetin along with DOX significantly promotes apoptosis in DL cells through intracellular ROS generation, mitochondrial aggregation at the periphery of the nucleus and, increased p53, Bax, cytochrome c, caspase 3, caspase 9, and cleaved caspase 9 expression. Additionally, combination therapy not only increased the mean survival of the treated group animals but also reduced the tumor burden. While histopathological parameters have shown overall improvement in combination therapy. This study proposes a novel combinational therapy for the treatment of lymphoma and requires further clinical investigation.
Collapse
Affiliation(s)
- Sumeet Singh
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India
| | - Virendra Singh
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Ranjeet Singh
- Department of Zoology, Soban Singh Jeena University (Bageshwar Campus), Almora, Uttarakhand, India; Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Vinita Gouri
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India; Department of Zoology, Kumaun University, Nainital, Uttarakhand, India
| | - Biplob Koch
- Genotoxicology and Cancer Biology Laboratory, Department of Zoology Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| | - Mukesh Samant
- Cell and Molecular Biology Laboratory, Department of Zoology, Soban Singh Jeena University, Almora, Uttarakhand, India.
| |
Collapse
|
6
|
Lee HJ, Lim SH, Lee H, Han JM, Min DS. Phospholipase D6 activates Wnt/β-catenin signaling through mitochondrial metabolic reprogramming to promote tumorigenesis in colorectal cancer. Exp Mol Med 2025; 57:910-924. [PMID: 40259095 PMCID: PMC12046002 DOI: 10.1038/s12276-025-01446-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/24/2025] [Accepted: 02/18/2025] [Indexed: 04/23/2025] Open
Abstract
Phospholipase D6 (PLD6) is a critical enzyme involved in mitochondrial fusion with a key role in spermatogenesis. However, the role of PLD6 in cancer remains unknown. Notably, Wnt signaling, energy metabolism and mitochondrial function show complex interactions in colorectal cancer (CRC) progression. Here we found that PLD6 is highly expressed in CRC and positively correlated with poor prognosis. We present a novel function of PLD6 in activating Wnt/β-catenin signaling by enhancing mitochondrial metabolism. PLD6 depletion suppresses the oncogenic properties of CRC cells and impairs mitochondrial respiration, leading to reduced mitochondrial length, membrane potential, calcium levels and reactive oxygen species. PLD6 depletion also disrupts mitochondrial metabolic reprogramming by inhibiting the tricarboxylic acid cycle and mitochondrial oxidative phosphorylation, resulting in altered intracellular levels of citrate and acetyl-CoA-both key modulators of Wnt/β-catenin activation. PLD6-mediated acetyl-CoA production enhances β-catenin stability by promoting its acetylation via the acetyltransferases CREB-binding protein and P300/CREB-binding-protein-associated factor. Consequently, PLD6 ablation reduces cancer stem cell-associated gene expression downstream of Wnt/β-catenin signaling, suppressing stem-like traits and chemoresistance to 5-fluorouracil. Furthermore, PLD6 depletion attenuates CRC tumorigenesis in both subcutaneous and orthotopic tumor models. Overall, PLD6 acts as an oncogenic switch by promoting mitochondria-mediated retrograde signaling, thereby regulating Wnt signaling in CRC.
Collapse
Affiliation(s)
- Hyun Ji Lee
- Department of Pharmacy, Yonsei University, Incheon, South Korea
| | - Seong Hun Lim
- Department of Pharmacy, Yonsei University, Incheon, South Korea
| | - Hyesung Lee
- Department of Pharmacy, Yonsei University, Incheon, South Korea
| | - Jung Min Han
- Department of Pharmacy, Yonsei University, Incheon, South Korea
- Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Do Sik Min
- Department of Pharmacy, Yonsei University, Incheon, South Korea.
- Yonsei Institute of Pharmaceutical Science, College of Pharmacy, Yonsei University, Incheon, South Korea.
| |
Collapse
|
7
|
Selvaraj NR, Nandan D, Nair BG, Nair VA, Venugopal P, Aradhya R. Oxidative Stress and Redox Imbalance: Common Mechanisms in Cancer Stem Cells and Neurodegenerative Diseases. Cells 2025; 14:511. [PMID: 40214466 PMCID: PMC11988017 DOI: 10.3390/cells14070511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 04/14/2025] Open
Abstract
Oxidative stress (OS) is an established hallmark of cancer and neurodegenerative disorders (NDDs), which contributes to genomic instability and neuronal loss. This review explores the contrasting role of OS in cancer stem cells (CSCs) and NDDs. Elevated levels of reactive oxygen species (ROS) contribute to genomic instability and promote tumor initiation and progression in CSCs, while in NDDs such as Alzheimer's and Parkinson's disease, OS accelerates neuronal death and impairs cellular repair mechanisms. Both scenarios involve disruption of the delicate balance between pro-oxidant and antioxidant systems, which leads to chronic oxidative stress. Notably, CSCs and neurons display alterations in redox-sensitive signaling pathways, including Nrf2 and NF-κB, which influence cell survival, proliferation, and differentiation. Mitochondrial dynamics further illustrate these differences: enhanced function in CSCs supports adaptability and survival, whereas impairments in neurons heighten vulnerability. Understanding these common mechanisms of OS-induced redox imbalance may provide insights for developing interventions, addressing aging hallmarks, and potentially mitigating or preventing both cancer and NDDs.
Collapse
Affiliation(s)
| | | | | | | | - Parvathy Venugopal
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| | - Rajaguru Aradhya
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India; (N.R.S.); (D.N.); (B.G.N.); (V.A.N.)
| |
Collapse
|
8
|
Kozak Y, Finiuk N, Czarnomysy R, Gornowicz A, Pinyazhko R, Lozynskyi A, Holota S, Klyuchivska O, Karkhut A, Polovkovych S, Klishch M, Stoika R, Lesyk R, Bielawski K, Bielawska A. Juglone-Bearing Thiopyrano[2,3-d]thiazoles Induce Apoptosis in Colorectal Adenocarcinoma Cells. Cells 2025; 14:465. [PMID: 40136714 PMCID: PMC11941218 DOI: 10.3390/cells14060465] [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: 02/20/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Colorectal cancer is a major global health challenge, with current treatments limited by toxicity and resistance. Thiazole derivatives, known for their bioactivity, are emerging as promising alternatives. Juglone (5-hydroxy-1,4-naphthoquinone) is a naturally occurring compound with known anticancer properties, and its incorporation into thiopyrano[2,3-d]thiazole scaffolds may enhance their therapeutic potential. This study examined the cytotoxicity of thiopyrano[2,3-d]thiazoles and their effects on apoptosis in colorectal cancer cells. Les-6547 and Les-6557 increased the population of ROS-positive HT-29 cancer cells approximately 10-fold compared with control cells (36.3% and 38.5% vs. 3.8%, respectively), potentially contributing to various downstream effects. Elevated ROS levels were associated with cell cycle arrest, inhibition of DNA biosynthesis, and reduced cell proliferation. A significant shift in the cell cycle distribution was observed, with an increase in S-phase (from 17.3% in the control to 34.7% to 51.3% for Les-6547 and Les-6557, respectively) and G2/M phase (from 24.3% to 39.9% and 28.8%). Additionally, Les-6547 and Les-6557 inhibited DNA biosynthesis in HT-29 cells, with IC50 values of 2.21 µM and 2.91 µM, respectively. Additionally, ROS generation may initiate the intrinsic apoptotic pathway. Les-6547 and Les-6557 activated both intrinsic and extrinsic apoptotic pathways, demonstrated by notable increases in the activity of caspase 3/7, 8, 9, and 10. This study provides a robust basis for investigating the detailed molecular mechanisms of action and therapeutic potential of Les-6547 and Les-6557.
Collapse
Affiliation(s)
- Yuliia Kozak
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Nataliya Finiuk
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Robert Czarnomysy
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland; (R.C.); (K.B.)
| | - Agnieszka Gornowicz
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland;
| | - Roman Pinyazhko
- Department of Normal Physiology, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
| | - Andrii Lozynskyi
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
| | - Olga Klyuchivska
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Andriy Karkhut
- Department of Technology of Biologically Active Substances, Pharmacy and Biotechnology, Lviv Polytecnic National University, Bandera 12, 79013 Lviv, Ukraine; (A.K.); (S.P.)
| | - Svyatoslav Polovkovych
- Department of Technology of Biologically Active Substances, Pharmacy and Biotechnology, Lviv Polytecnic National University, Bandera 12, 79013 Lviv, Ukraine; (A.K.); (S.P.)
| | - Mykola Klishch
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Rostyslav Stoika
- Department of Regulation of Cell Proliferation and Apoptosis, Institute of Cell Biology of National Academy of Sciences of Ukraine, Drahomanov 14/16, 79005 Lviv, Ukraine; (N.F.); (O.K.); (M.K.); (R.S.)
| | - Roman Lesyk
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine; (A.L.); (S.H.); (R.L.)
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszów, Sucharskiego 2, 35-225 Rzeszów, Poland
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland; (R.C.); (K.B.)
| | - Anna Bielawska
- Department of Biotechnology, Faculty of Pharmacy, Medical University of Bialystok, Kilinskiego 1, 15-089 Białystok, Poland;
| |
Collapse
|
9
|
Hachem H, Le Gal Y, Jeannin O, Lorcy D, Scalese G, Pérez-Díaz L, Gambino D, Matos AP, Marques F. Metal (Au, Pt, Pd, Ni) Bis(dithiolene) complexes as dual-action agents combating cancer and trypanosomatid infections. J Inorg Biochem 2025; 264:112788. [PMID: 39642704 DOI: 10.1016/j.jinorgbio.2024.112788] [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: 08/06/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
Cancer and infection diseases pose severe threats to public health worldwide stressing the need for more effective and efficient treatments. Thus, the search for broad-spectrum activity drugs seems justifiable and urgent. Herein, we investigate the anticancer and antitrypanosomatid (anti-Trypanosoma cruzi) activities of eight monoanionic metal bis(dithiolene) complexes, [Ph4P][M(R-thiazdt)2] with Mn+ = Au3+, Pt2+, Pd2+, Ni2+, containing N-alkyl-1,3-thiazoline-2-thione dithiolene ligands (R-thiazdt) with different alkyl groups (R = Et, tBu). Compared to auranofin (AF) and cisplatin (CP), two reference drugs in clinical use, all complexes showed high anticancer activities against A2780 ovarian cancer cells (IC50 values of 0.6-3.8 μM) some also being able to overcome CP resistance in A2780cisR cells. The selectivity index (SI), the IC50 values on normal cells (HDF) vs. A2780 cells, indicated good anticancer specificity (SI > 3) for most of the complexes but with clinical relevance for [Ph4P][Pd(tBu-thiazdt)2] (SI = 10). All complexes showed relevant antitrypanosomatid activities (IC50 values of 2.6-5.8 μM) some even exhibiting lower IC50 values than the reference drug nifurtimox (NFX). The mechanism of cell death seemed to be mediated mainly by the formation of reactive oxygen species (ROS), although to lesser extent for the gold complexes but superior to AF. Although ROS play a role in the main apoptotic pathways, cell death by apoptosis was not evident as shown by the caspase-3/7 assay and the morphological cell features studies by electron microscopy (SEM). Results obtained evidenced that [Ph4P][Pt(tBu-thiazdt)2] and [Ph4P][Pd(tBu-thiazdt)2] complexes might have potential as novel anticancer and antitrypanosomatid agents as alternatives to current therapeutics.
Collapse
Affiliation(s)
- Hadi Hachem
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Yann Le Gal
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Olivier Jeannin
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Dominique Lorcy
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Gonzalo Scalese
- Área Química Inorgánica, Departamento Estrella Campos, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - Leticia Pérez-Díaz
- Sección Genómica Funcional, Instituto de Química Biológica, Facultad de Ciencias, Universidad de la República, 11400 Montevideo, Uruguay
| | - Dinorah Gambino
- Área Química Inorgánica, Departamento Estrella Campos, Facultad de Química, Universidad de la República, 11800 Montevideo, Uruguay
| | - António P Matos
- Egas Moniz School of Health and Science, Egas Moniz Center for Interdisciplinary Research (CiiEM), University Campus, Quinta da Granja, Monte de Caparica, 2829-511 Caparica, Portugal
| | - Fernanda Marques
- Centro de Ciências e Tecnologias Nucleares and Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Estrada Nacional 10, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
10
|
Chmelyuk N, Kordyukova M, Sorokina M, Sinyavskiy S, Meshcheryakova V, Belousov V, Abakumova T. Inhibition of Thioredoxin-Reductase by Auranofin as a Pro-Oxidant Anticancer Strategy for Glioblastoma: In Vitro and In Vivo Studies. Int J Mol Sci 2025; 26:2084. [PMID: 40076706 PMCID: PMC11900239 DOI: 10.3390/ijms26052084] [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: 01/14/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Reactive oxygen species (ROS) play a key role in cancer progression and antitumor therapy. Glioblastoma is a highly heterogeneous tumor with different cell populations exhibiting various redox statuses. Elevated ROS levels in cancer cells promote tumor growth and simultaneously make them more sensitive to anticancer drugs, but further elevation leads to cell death and apoptosis. Meanwhile, various subsets of tumor cells, such a glioblastoma stem cells (GSC) or the cells in tumor microenvironment (TME), demonstrate adaptive mechanisms to excessive ROS production by developing effective antioxidant systems such as glutathione- and thioredoxin-dependent. GSCs demonstrate higher chemoresistance and lower ROS levels than other glioma cells, while TME cells create a pro-oxidative environment and have immunosuppressive effects. Both subpopulations have become an attractive target for developing therapies. Increased expression of thioredoxin reductase (TrxR) is often associated with tumor progression and poor patient survival. Various TrxR inhibitors have been investigated as potential anticancer therapies, including nitrosoureas, flavonoids and metallic complexes. Gold derivatives are irreversible inhibitors of TrxR. Among them, auranofin (AF), a selective TrxR inhibitor, has proven its effectiveness as a drug for the treatment of rheumatoid arthritis and its efficacy as an anticancer agent has been demonstrated in preclinical studies in vitro and in vivo. However, further clinical application of AF could be challenging due to the low solubility and insufficient delivery to glioblastoma. Different delivery strategies for hydrophobic drugs could be used to increase the concentration of AF in the brain. Combining different therapeutic approaches that affect the redox status of various glioma cell populations could become a new strategy for treating brain tumor diseases.
Collapse
Affiliation(s)
- Nelly Chmelyuk
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Laboratory of Biomedical nanomaterials, National Research Technological University “MISIS”, Leninskiy Prospekt 4, 119049 Moscow, Russia
| | - Maria Kordyukova
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Maria Sorokina
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Semyon Sinyavskiy
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Valeriya Meshcheryakova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vsevolod Belousov
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Neurotechnology Laboratory, Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Tatiana Abakumova
- Department of Synthetic Neurotechnologies, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
11
|
Ahmed S, Aschner M, Alsharif KF, Allahyani M, Huang G, Wan C, Khan H. Marine peptides in lymphoma: surgery at molecular level for therapeutic understanding. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03901-w. [PMID: 39992419 DOI: 10.1007/s00210-025-03901-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
Lymphoma, the most common form of blood cancer, affects primarily the intricate network of tissues and organs known as the lymphatic system. Globally, it ranks among the leading causes of cancer-related deaths. Although conventional therapies have led to significant advancements, they are accompanied by adverse side effects and present challenges in cases of multidrug resistance, refractory patients, and relapses. This highlights a pressing need for innovative treatment approaches. Extensive research on the anti-lymphoma properties of natural compounds has particularly focused on marine organisms as valuable sources for potential medicinal agents. Among these, anticancer peptides have garnered attention due to their multiple beneficial effects against cancer, coupled with reduced toxicity to normal cells. This review focuses on the molecular mechanisms underlying the anti-lymphoma effects of marine peptides, examining the diverse pathways through which these peptides impact physiological processes. Key effects include modulation of cell viability, induction of apoptosis, cell cycle arrest, antimitotic activity, immunotherapeutic properties, disruption of mitochondrial function and induction of oxidative stress, cancer cell membrane destruction, and interference with microtubule stability. The review also highlights the antibody-drug conjugates (ADCs) derived from marine peptides and their synergistic effects with other anti-lymphoma medications. This knowledge should inspire future study and development of these prospective therapeutic modalities and hasten the investigation and creation of novel lymphoma remedies derived from marine sources.
Collapse
Affiliation(s)
- Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Mamdouh Allahyani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chunpeng Wan
- Jiangxi Provincial Key Laboratory for Postharvest Storage and Preservation of Fruits & Vegetables, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Khyber Pakhtunkhwa, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| |
Collapse
|
12
|
Catalano T, Selvaggi F, Cotellese R, Aceto GM. The Role of Reactive Oxygen Species in Colorectal Cancer Initiation and Progression: Perspectives on Theranostic Approaches. Cancers (Basel) 2025; 17:752. [PMID: 40075600 PMCID: PMC11899472 DOI: 10.3390/cancers17050752] [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: 01/12/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Altered levels of reactive oxygen species (ROS) are recognized as one of the key factors in mediating tumor cell survival in the tissue microenvironment, where they play a role in the initiation, progression and recurrence/relapse of colorectal cancer (CRC). Tumor cells can adapt to oxidative stress (OS) using genetic or metabolic reprogramming in the long or short term. In addition, tumor cells defend themselves through positive regulation of antioxidant molecules, enhancing ROS-driven proliferation. Balanced oxidative eustress levels can influence chemotherapy resistance, allowing tumor cells to survive treatment. Secondary effects of chemotherapy include increased ROS production and redox stress, which can kill cancer cells and eliminate drug resistance. Anticancer treatments based on manipulating ROS levels could represent the gold standard in CRC therapy. Therefore, exploring the modulation of the response to OS in deregulated signaling pathways may lead to the development of new personalized CRC treatments to overcome therapy resistance. In this review, we explore the role of ROS in the initiation and progression of CRC and their diagnostic implications as biomarkers of disease. Furthermore, we focused on the involvement of ROS in different CRC therapeutic options, such as surgery, radiotherapy, theranostic imaging, chemotherapy and immunotherapy and other precision medicine approaches.
Collapse
Affiliation(s)
- Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Federico Selvaggi
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Roberto Cotellese
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Gitana Maria Aceto
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
- Department of Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
13
|
Verma M, Fatima S, Syed A, Elgorban AM, Abid I, Wong LS, Khan MS, Ansari IA. 3-Acetyl-11-keto-β-boswellic acid (AKBA) induced antiproliferative effect by suppressing Notch signaling pathway and synergistic interaction with cisplatin against prostate cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03899-1. [PMID: 39985578 DOI: 10.1007/s00210-025-03899-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Studies on the assessment of anticancer efficacy of plant-derived phytochemicals by targeting signaling pathways have drawn a lot of attention recently for human health. Multiple investigations have proposed an involvement of Notch pathway in the processes of cancer angiogenesis and metastasis, and drug resistance. Moreover, overexpression of Notch signaling is associated with increased prostate cancer (PrCa) cell growth and development. A number of chemotherapeutic agents are reported to become resistant over a period of time and have severe side effects. To increase efficacy and lessen drug-induced toxicity, a variety of bioactive compounds have been utilized alone or as adjuncts to traditional chemotherapy. Therefore, in the present study, the potential of AKBA in inhibiting the proliferation of PrCa cells by modulating Notch signaling components and its efficacy in combination with cisplatin was investigated. The results exhibited a substantial reduction in cell survival (IC50 = 25.28 µM at 24 h and 16.50 µM at 48 h) and cellular alterations in AKBA-treated PrCa cells. Additionally, AKBA caused nuclear condensation, increased reactive oxygen species (ROS) generation, mitochondrial membrane depolarization, and caspase activation, ultimately leading to apoptosis in PrCa cells. Moreover, AKBA-elicited apoptosis was evidenced by an augmentation in the Bax to Bcl2 ratio. AKBA was also found to induce G0/G1 arrest which was substantiated by reduced cyclin D1 and CDK4 expression levels concomitantly with increased expression of p21 and p27 genes. Intriguingly, AKBA demonstrated significant downregulation of Notch signaling mediators. Furthermore, the isobolograms of the combination treatment indicated that AKBA has the potential to synergistically enhance the cytotoxic efficacy of cisplatin in DU145 cells, as evidenced by CI < 1 across all tested combinations. Overall, the results of this study suggest strong antiproliferative, apoptotic, and chemo-sensitizing potential of AKBA. Thus, AKBA holds a promising drug candidature warranting further investigation as a probable therapeutic option for both the prevention and treatment of PrCa and other solid tumors.
Collapse
Affiliation(s)
- Mahima Verma
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India
- Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Shireen Fatima
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India
- Department of Biosciences, Integral University, Lucknow, 226026, India
| | - Asad Syed
- Department of Botany and Microbiology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Abdallah M Elgorban
- Centre of Excellence in Biotechnology Research (CEBR), King Saud University, Riyadh, Saudi Arabia
| | - Islem Abid
- Centre of Excellence in Biotechnology Research (CEBR), King Saud University, Riyadh, Saudi Arabia
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Putra Nilai, 71800, Nilai, Negeri Sembilan, Malaysia
| | - Mohd Sajid Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Irfan Ahmad Ansari
- Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, 226026, India.
- Department of Biosciences, Integral University, Lucknow, 226026, India.
| |
Collapse
|
14
|
Altwaijry NA, Omar MA, Mohamed HS, Mounier MM, Afifi AH, Srour AM. Design, synthesis, molecular docking and anticancer activity of benzothiazolecarbohydrazide-sulfonate conjugates: insights into ROS-induced DNA damage and tubulin polymerization inhibition. RSC Adv 2025; 15:5895-5905. [PMID: 39990814 PMCID: PMC11843914 DOI: 10.1039/d4ra07810a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/07/2025] [Indexed: 02/25/2025] Open
Abstract
A series of novel benzothiazolecarbohydrazide-sulfonate conjugates 6a-l were designed, synthesized, and then assessed as potential antiproliferative agents in three distinct human cancer cell lines: MCF-7 (breast cancer), HCT-116 (colon cancer), and PC3 (prostate cancer), along with a normal cell line (BJ-1). The reference standard used was 5-fluorouracil. The results obtained reveal that the newly synthesized analogs demonstrate varying degrees of cytotoxicity against the targeted cell lines; however, compounds 6i and 6e exhibited the highest efficacy against MCF-7, HCT-116, and PC3 cells with IC50 values of 78.8 ± 2.6, 81.4 ± 1.9, and 90.6 ± 2.7 μM, respectively, compared to an IC50 of 78.4 ± 4.2 μM for 5-FU in MCF-7 cells, 29.2 ± 1.7 μM in HCT-116 cells and >200 μM in PC3 cells. Moreover, the most potent compounds demonstrated acceptable safety profiles when evaluated aganist BJ-1 cells. Consequently, compound 6i, which possesses no cytotoxicity towards BJ-1 cells and displays promising anticancer activity, was further investigated for its impact on tubulin polymerization compared to control MCF-7 cells, 210.3 and 632.9 pg ml-1, respectively. Compound 6i was found to significantly elevate the ROS levels in treated cancer cells, resulting in an 8.3-fold increase in DNA fragmentation compared to untreated cells. Additionally, it raised the percentage of accumulated cells in the G2 phase from 6.85% to 18.27% in MCF-7 cells. A molecular docking technique was conducted to elucidate the binding energy, binding pose, and binding interactions of compound 6i, revealing a strong fit within the active sites of the tubulin-colchicine binding site (CBS). This study provides valuable insights into the design and synthesis of novel anticancer agents targeting tubulin polymerization.
Collapse
Affiliation(s)
- Najla A Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University Riyadh Saudi Arabia
| | - Mohamed A Omar
- Chemistry of Natural and Microbial Products Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Hanaa S Mohamed
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Marwa M Mounier
- Department of Pharmacognosy, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Ahmed H Afifi
- Department of Pharmacognosy, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| | - Aladdin M Srour
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre Dokki 12622 Giza Egypt
| |
Collapse
|
15
|
Kurhaluk N, Tkaczenko H. Recent Issues in the Development and Application of Targeted Therapies with Respect to Individual Animal Variability. Animals (Basel) 2025; 15:444. [PMID: 39943214 PMCID: PMC11815764 DOI: 10.3390/ani15030444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
This literature review explores the impact of molecular, genetic, and environmental factors on the efficacy of targeted therapies in veterinary medicine. Relevant studies were identified through systematic searches of PubMed, Web of Science, Scopus, and ScienceDirect using keywords such as "species-specific treatment strategies", "signalling pathways", "epigenetic and paragenetic influences", "targeted therapies", "veterinary medicine", "genetic variation", and "free radicals and oxidative stress". Inclusion criteria included studies focusing on species-specific therapeutic responses, genetic influences, and oxidative stress. To ensure that only the most recent and relevant evidence was included, only peer-reviewed publications from the last two decades were considered. Each study selected for analysis was critically appraised, with a particular emphasis on methodological quality, experimental design, and scientific contribution to the understanding of how environmental and biological factors influence therapeutic outcomes. A special emphasis was placed on studies that used a comparative, cross-species approach to assess variability in therapeutic responses and potential adverse effects. The review synthesises evidence on the role of epigenetic and paragenetic factors and highlights the importance of cross-species studies to understand how environmental and biological factors influence treatment outcomes. By highlighting genetic variation, oxidative stress, and individual species differences, the review argues for personalised and species-specific therapeutic approaches. The review emphasises that such an approach would improve veterinary care and inform future research aimed at optimising targeted therapies, ultimately leading to better animal health and treatment efficacy. A key contribution of the review is its emphasis on the need for more personalised treatment protocols that take into account individual genetic profiles and environmental factors; it also calls for a greater integration of cross-species studies.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22b, 76-200 Słupsk, Poland;
| | | |
Collapse
|
16
|
Barman D, Rajamalli P, Bidkar AP, Sarmah T, Ghosh SS, Zysman-Colman E, Iyer PK. Modulation of Donor in Purely Organic Triplet Harvesting AIE-TADF Photosensitizer for Image-guided Photodynamic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409533. [PMID: 39780649 DOI: 10.1002/smll.202409533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Image-guided photodynamic therapy is acknowledged as one of the most demonstrative therapeutic modalities for cancer treatment because of its high precision, non-invasiveness, and improved imaging ability. A series of purely organic photosensitizers denoted as BTMCz, BTMPTZ, and BTMPXZ, have been designed and synthesized and are found to exhibit both thermally activated delayed fluorescence and aggregation-induced emission simultaneously. Experimental and theoretical studies are combined to reveal that modulation of the donor of the photosensitizer enables distinct thermally activated delayed fluorescence via a second-order spin-orbit perturbation mechanism involving lowest singlet charge-transfer and higher-lying triplet locally excited states, respectively. Further, different donor strengths and unique aggregations (H-, J- and X-type packings) greatly influence their color-tunable up-converted luminescence and endow them with superb dispersibility in water. The confocal microscopy-based cellular uptake study confirms the successful internalization of the nano-probes, while BTMCz enables the generation of reactive oxygen species (singlet oxygen) under white-light irradiation, enabling the efficient killing of cancer cells.
Collapse
Affiliation(s)
- Debasish Barman
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| | - Pachaiyappan Rajamalli
- Materials Research Centre, Indian Institute of Science, Bangalore, 560012, India
- Organic Semiconductor Centre, EaStCHEM School of Chemistry, University of St Andrews, St Andrews, Fife, KY16 9ST, UK
| | - Anil Parsram Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, 94143, USA
- Department of Bioscience and Bioengineering IIT Guwahati, Guwahati, Assam, 781039, India
| | - Tapashi Sarmah
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| | - Siddhartha Sankar Ghosh
- Department of Bioscience and Bioengineering IIT Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| | - Eli Zysman-Colman
- Organic Semiconductor Centre, EaStCHEM School of Chemistry, University of St Andrews, St Andrews, Fife, KY16 9ST, UK
| | - Parameswar Krishnan Iyer
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, 781039, India
| |
Collapse
|
17
|
Malarz K, Borzęcka W, Ziola P, Domiński A, Rawicka P, Bialik-Wąs K, Kurcok P, Torres T, Mrozek-Wilczkiewicz A. pH-sensitive phthalocyanine-loaded polymeric nanoparticles as a novel treatment strategy for breast cancer. Bioorg Chem 2025; 155:108127. [PMID: 39798455 DOI: 10.1016/j.bioorg.2025.108127] [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: 12/15/2024] [Accepted: 01/01/2025] [Indexed: 01/15/2025]
Abstract
Novel pH-sensitive polymeric photosensitizer carriers from the phthalocyanine (Pc) group were investigated as potential photodynamic therapy drugs for the treatment of breast cancer. Their high antiproliferative activity was confirmed by photocytotoxicity studies, which indicated their high efficacy and specificity toward the SK-BR-3 cell line. Importantly, the Pcs encapsulated in the polymeric nanoparticle (NP) carrier exhibited a much better penetration into the acidic environment of tumor cells than their free form. The investigated Pc4-NPs and TT1-NPs exhibited a high selectivity to healthy fibroblasts as well as non-toxicity without irradiation. This paper describes the detailed mechanism of action of the evaluated compounds by measuring reactive oxygen species (ROS), including singlet oxygen; imaging cellular localization; and analyzing key signaling pathway proteins. An additional advantage of the evaluated compounds is their ability to inhibit the Akt protein expression, including its phosphorylation, which the Western blot test confirmed. This is particularly important because breast cancers often overexpress the HER-2 receptor-related signaling proteins. Moreover, an analysis of proteins such as GLUT-1, HO-1, phospho-p42/44, and BID revealed the significant involvement of ROS in disrupting cellular homeostasis, thereby leading to the induction of oxidative stress and resulting in apoptotic cell death.
Collapse
Affiliation(s)
- Katarzyna Malarz
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland; A. Chełkowski Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland
| | - Wioleta Borzęcka
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Marii Skłodowskiej-Curie 34, 41-819 Zabrze, Poland.
| | - Patryk Ziola
- A. Chełkowski Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland
| | - Adrian Domiński
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Marii Skłodowskiej-Curie 34, 41-819 Zabrze, Poland
| | - Patrycja Rawicka
- A. Chełkowski Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland
| | - Katarzyna Bialik-Wąs
- Department of Chemistry and Technology of Polymers, Faculty of Chemical Engineering and Technology, Cracow University of Technology, Warszawska 24, 31-155 Cracow, Poland
| | - Piotr Kurcok
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Marii Skłodowskiej-Curie 34, 41-819 Zabrze, Poland
| | - Tomas Torres
- Department of Organic Chemistry, Autonoma University of Madrid, 28049 Madrid, Spain; IMDEA-Nanociencia, Campus de Cantoblanco, c/Faraday 9, 28049 Madrid, Spain; Institute for Advanced Research in Chemical Sciences (IAdChem), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - Anna Mrozek-Wilczkiewicz
- Department of Systems Biology and Engineering, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland; A. Chełkowski Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1a, 41-500 Chorzów, Poland.
| |
Collapse
|
18
|
Verma M, Fatima S, Saeed M, Ansari IA. Anti-proliferative, Pro-apoptotic, and Chemosensitizing Potential of 3-Acetyl-11-keto-β-boswellic Acid (AKBA) Against Prostate Cancer Cells. Mol Biotechnol 2025; 67:746-761. [PMID: 38502429 DOI: 10.1007/s12033-024-01089-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/26/2024] [Indexed: 03/21/2024]
Abstract
Prostate cancer incidences are rising worldwide at an alarming rate. Drug resistance and relapse are two major challenges in the treatment of prostate cancer. Therefore, new multimodal, safe, and effective therapeutic agents are urgently required which could effectively mitigate the menace of tumor recurrence and chemo-resistance. Plant-derived products are increasingly being utilized due to their antioxidant, antibacterial, and anti-tumor potential. In the current study, 3-acetyl-11-keto-β-boswellic acid, a triterpenoid isolated from plant Boswellia, was utilized to ascertain its chemotherapeutic potential against human prostate cancer cells. Various in vitro assays including cell viability, nuclear staining, mitochondria potential, reactive oxygen species (ROS) generation, and quantification of apoptosis, were performed for the evaluation of the cytotoxic potential of AKBA. We observed that AKBA (10-50 µM) dose-dependently suppressed cell proliferation and caused programmed cell death in PC3 cells via both intrinsic and extrinsic pathway. Intriguingly, AKBA was also found to chemosensitize PC3 cells in synergistic combination with doxorubicin. To the best of our knowledge, this is the first study to document the synergistic chemosensitizing impact of AKBA when combined with doxorubicin in prostate cancer cells.This showcases the potential of AKBA in combinatorial therapy or adjuvant therapy for the management of prostate cancer. In sum, our results suggested that AKBA is a promising drug-like molecule against prostate cancer. Our investigation introduces a novel perspective, elucidating a previously unexplored dimension, and uncovering a compelling chemosensitizing phenomenon along with a strong synergistic effect arising from the concurrent application of these two agents.
Collapse
Affiliation(s)
- Mahima Verma
- Department of Biosciences, Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, India
| | - Shireen Fatima
- Department of Biosciences, Integral Centre of Excellence for Interdisciplinary Research (ICEIR), Integral University, Lucknow, India
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | | |
Collapse
|
19
|
Chiaramonte R, Sauro G, Giannandrea D, Limonta P, Casati L. Molecular Insights in the Anticancer Activity of Natural Tocotrienols: Targeting Mitochondrial Metabolism and Cellular Redox Homeostasis. Antioxidants (Basel) 2025; 14:115. [PMID: 39857449 PMCID: PMC11760857 DOI: 10.3390/antiox14010115] [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: 11/27/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
The role of mitochondria as the electric engine of cells is well established. Over the past two decades, accumulating evidence has pointed out that, despite the presence of a highly active glycolytic pathway (Warburg effect), a functional and even upregulated mitochondrial respiration occurs in cancer cells to meet the need of high energy and the biosynthetic demand to sustain their anabolic growth. Mitochondria are also the primary source of intracellular ROS. Cancer cells maintain moderate levels of ROS to promote tumorigenesis, metastasis, and drug resistance; indeed, once the cytotoxicity threshold is exceeded, ROS trigger oxidative damage, ultimately leading to cell death. Based on this, mitochondrial metabolic functions and ROS generation are considered attractive targets of synthetic and natural anticancer compounds. Tocotrienols (TTs), specifically the δ- and γ-TT isoforms, are vitamin E-derived biomolecules widely shown to possess striking anticancer properties since they regulate several intracellular molecular pathways. Herein, we provide for the first time an overview of the mitochondrial metabolic reprogramming and redox homeostasis perturbation occurring in cancer cells, highlighting their involvement in the anticancer properties of TTs. This evidence sheds light on the use of these natural compounds as a promising preventive or therapeutic approach for novel anticancer strategies.
Collapse
Affiliation(s)
- Raffaella Chiaramonte
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (R.C.); (G.S.); (D.G.)
| | - Giulia Sauro
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (R.C.); (G.S.); (D.G.)
| | - Domenica Giannandrea
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (R.C.); (G.S.); (D.G.)
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences “R. Paoletti”, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Lavinia Casati
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy; (R.C.); (G.S.); (D.G.)
| |
Collapse
|
20
|
Jafary B, Akbarzadeh-Khiavi M, Farzi-Khajeh H, Safary A, Adibkia K. EGFR-targeting RNase A-cetuximab antibody-drug conjugate induces ROS-mediated apoptosis to overcome drug resistance in KRAS mutant cancer cells. Sci Rep 2025; 15:1483. [PMID: 39789190 PMCID: PMC11718297 DOI: 10.1038/s41598-025-85856-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
Antibody-drug conjugates (ADCs) are an emerging strategy in cancer therapy, enhancing precision and efficacy by linking targeted antibodies to potent cytotoxic agents. This study introduces a novel ADC that combines ribonuclease A (RNase A) with cetuximab (Cet), an anti-EGFR monoclonal antibody, through a polyethylene glycol (PEG) linker (RN-PEG-Cet), aimed to induce apoptosis in KRAS mutant colorectal cancer (CRC) via a ROS-mediated pathway. RN-PEG-Cet was successfully synthesized and characterized for its physicochemical properties, retaining full enzymatic activity in RNA degradation and high binding affinity to EGFR. In KRAS mutant SW-480 cells, RN-PEG-Cet significantly reduced cell viability at lower doses, with an IC50 of 11.7 µg/mL at 72 h. Compared to free Cet, RN-PEG-Cet demonstrated a ~ 2-fold increase in apoptosis and a ~ 3.5-fold increase in ROS production. The conjugate also disrupted the Nrf2/Keap1 pathway, with a significant upregulation of Keap1 (FC = 3.7, p ≤ 0.01) and downregulation of Nrf2 (FC = 3.3, p < 0.01), highlighting its role in impairing antioxidant defenses and promoting ROS-mediated cytotoxicity. These findings emphasize the potential of RN-PEG-Cet as a novel therapeutic approach for KRAS mutant CRC, offering superior apoptosis induction and targeted cytotoxicity compared to conventional therapies. This ADC could represent a new strategy for improving CRC treatment outcomes by effectively overcoming resistance mechanisms.
Collapse
Affiliation(s)
- Bita Jafary
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Akbarzadeh-Khiavi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hamed Farzi-Khajeh
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
22
|
Chinnapaka S, Bakthavachalam V, Dasari S, Kannan J, Sapkota S, Kumar R, Munirathinam G. Vitamin K3 derivative inhibits androgen receptor signaling in targeting aggressive prostate cancer cells. Biofactors 2025; 51:e2117. [PMID: 39225404 DOI: 10.1002/biof.2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Prostate cancer (PCa) is the second critical cause of cancer-related deaths, with African Americans dying at higher rates in the U.S. The main reasons for the higher mortality rate are ethnic differences and lack of understanding of prostate cancer biology and affordable treatments, as well as the financial burden of African American men to obtain the most effective and safe treatments. The effect of micronutrients, including Vitamin K, on various cancer cell lines has been widely studied, but the potential anticancer effect of VK3-OCH3, an analog of vitamin K3 (Menadione), on African American prostate cancer has not been evaluated. In this study, we compared the anticancer effect of VK3-OCH3 on targeting African American derived PCa cell lines namely RC77-T and MDA-PCa-2b. Our results show that VK3-OCH3 significantly inhibits the proliferation of both RC77-T and MDA-PCa-2b African American PCa cells and promotes apoptosis, and the underlying mechanism of cell death appears to be similar in both the cell lines. Notably, VK3-OCH3 inhibits colony-forming ability and induces apoptosis by blocking the cell cycle at G0 in African American PCa cells. VK3-OCH3 also acts as an anti-metastatic agent by inhibiting the migration ability of the metastatic properties of African American PCa cells. The cell death of African American PCa cells mediated by VK3-OCH3 is associated with the production of free radicals, such as intracellular and mitochondrial reactive oxygen species (ROS). Interestingly, antioxidants such as N-Acetylcysteine (NAC) and Glutathione (GSH) effectively negated the oxidative stress induced by VK3-OCH3 on PCa cell lines derived from African American patients. Of note, VK3-OCH3 reduces androgen receptor and prostate-specific antigen expression in these PCa cells. Furthermore, molecular dynamic studies reiterated that VK3-OCH3 strongly binds to the androgen receptor, suggesting that the androgen receptor is the potential molecular target of VK3-OCH3. In addition, Western blot analysis showed that VK3-OCH3 reduces the expression of androgen receptor, TRX2, and anti-apoptotic signaling molecules such as Bcl-2 and TCTP in the MDA-PCa-2b metastatic PCa cellular model. In conclusion, our results suggested that VK3-OCH3 is a promising anticancer agent that could potentially reduce the mortality rates of African American PCa patients, warranting further preclinical and translational studies.
Collapse
Affiliation(s)
- Somaiah Chinnapaka
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Velavan Bakthavachalam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | | | - Jhishnuraj Kannan
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Sworaj Sapkota
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| | - Raj Kumar
- Department of Biotechnology and Bioinformatics, Jaypee University of Information Technology, Solan, Himachal Pradesh, India
| | - Gnanasekar Munirathinam
- Department of Biomedical Sciences, University of Illinois College of Medicine, Rockford, Illinois, USA
| |
Collapse
|
23
|
Zheng Z, Zeng Y, Bao X, Huang C, Guo F, Xu F, Luo Z. OTULIN confers cisplatin resistance in osteosarcoma by mediating GPX4 protein homeostasis to evade the mitochondrial apoptotic pathway. J Exp Clin Cancer Res 2024; 43:330. [PMID: 39721999 DOI: 10.1186/s13046-024-03249-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Osteosarcoma (OS), the most prevalent primary malignant bone tumor in children and adolescents, arises from bone-forming mesenchymal cells. Despite advancements in surgical resection and neoadjuvant chemotherapy (cisplatin, doxorubicin, and methotrexate), chemotherapy resistance remains a significant challenge, leading to poor survival rates in patients with metastatic or recurrent OS. METHODS In this study, we focused on the role of OTULIN, a key linear deubiquitinating enzyme, in OS chemoresistance. In addition, mechanistic investigations were carried out to identify potential downstream targets of OTULIN involved in cisplatin resistance. RESULTS Our results demonstrated that OTULIN expression was significantly upregulated in OS tissues and cell lines following cisplatin treatment but not in response to doxorubicin or methotrexate. High OTULIN expression was associated with reduced survival in sarcoma patients. Furthermore, immunohistochemical analysis of prechemotherapy and postchemotherapy OS tissues revealed increased OTULIN expression in postchemotherapy samples. In vitro results demonstrated that OTULIN plays a critical role in mediating cisplatin resistance in OS. Mechanistically, GPX4 could be a downstream target of OTULIN, conferring cisplatin resistance to OS by blocking the mitochondrial apoptotic pathway but not ferroptosis. Specifically, OTULIN prevents the proteasomal degradation of GPX4 by reducing its ubiquitin level, thereby conferring resistance to cisplatin in OS cells. CONCLUSION This study highlights the importance of OTULIN in OS chemoresistance and provides a promising approach for targeting the OTULIN-GPX4 axis to improve the prognosis of OS patients. Our findings offer new insights into the molecular mechanisms underlying OS chemoresistance and suggest potential therapeutic targets for future clinical interventions.
Collapse
Affiliation(s)
- Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunhao Zeng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing Bao
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuang Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Fei Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhengqiang Luo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
24
|
Mavroeidi D, Georganta A, Stefanou DT, Papanikolaou C, Syrigos KN, Souliotis VL. DNA Damage Response Network and Intracellular Redox Status in the Clinical Outcome of Patients with Lung Cancer. Cancers (Basel) 2024; 16:4218. [PMID: 39766117 PMCID: PMC11726754 DOI: 10.3390/cancers16244218] [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: 11/25/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Background/Objectives: DNA damage response (DDR) is a network of molecular pathways associated with the pathogenesis and progression of several diseases, as well as the outcome of chemotherapy. Moreover, the intracellular redox status is essential for maintaining cell viability and controlling cellular signaling. Herein, we analyzed DDR signals and redox status in peripheral blood mononuclear cells (PBMCs) from patients with lung cancer with different response rates to platinum-based chemotherapy. Methods: Several DDR-associated signals and redox status, expressed as the GSH/GSSG ratio, were measured in two lung cancer cell lines (A549, H1299), two normal fibroblast cell lines (WS1, 1BR3hT), and PBMCs from 20 healthy controls and 32 patients with lung cancer at baseline (17 responders and 15 non-responders to subsequent platinum-based chemotherapy). Results: Higher levels of endogenous/baseline DNA damage, decreased GSH/GSSG ratios, and augmented apurinic/apyrimidinic sites, as well as lower nucleotide excision repair (NER) and increased interstrand cross-links (ICLs) repair efficiencies, were observed in lung cancer cell lines compared with normal ones (all p < 0.05). Moreover, PBMCs from patients with lung cancer showed reduced GSH/GSSG ratios, augmented apurinic/apyrimidinic sites, decreased NER and ICL repair capacities, and lower apoptosis rates, compared with healthy controls (all p < 0.001). Interestingly, PBMCs from patients who are responders are characterized by reduced GSH/GSSG ratios, augmented apurinic/apyrimidinic sites, decreased NER and ICL repair capacities, and higher apoptosis rates compared with patients who are non-responders (all p < 0.01). Conclusions: Together, DDR-associated parameters and redox status measured in PBMCs from patients with lung cancer at baseline are associated with the therapeutic benefit of platinum-based chemotherapy.
Collapse
Affiliation(s)
- Dimitra Mavroeidi
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.M.); (C.P.)
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.G.); (K.N.S.)
| | - Anastasia Georganta
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.G.); (K.N.S.)
| | - Dimitra T. Stefanou
- First Department of Internal Medicine, Laikon General Hospital, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christina Papanikolaou
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.M.); (C.P.)
| | - Konstantinos N. Syrigos
- Third Department of Medicine, Sotiria General Hospital for Chest Diseases, National and Kapodistrian University of Athens, 11527 Athens, Greece; (A.G.); (K.N.S.)
| | - Vassilis L. Souliotis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.M.); (C.P.)
| |
Collapse
|
25
|
Ali BA, Allam RM, Hasanin MS, Hassabo AA. Biosynthesis of selenium nanoparticles as a potential therapeutic agent in breast cancer: G2/M arrest and apoptosis induction. Toxicol Rep 2024; 13:101792. [PMID: 39554610 PMCID: PMC11565031 DOI: 10.1016/j.toxrep.2024.101792] [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: 08/27/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/19/2024] Open
Abstract
The drawbacks and adverse reactions of conventional breast cancer (BC) medications have prompted researchers to seek novel therapeutic approaches. This study aimed to study the impact of biosynthesized selenium nanoparticles by yeast on breast cancer (MCF-7) cells and to find potential underlying mechanisms. Therefore, marine yeast isolates were screened for their ability to biosynthesis selenium nanoparticles (SeNPs). The most potent isolate was identified as Candida pseudojiufengensis based on 18 S rRNA gene sequencing. Incubation of cell-free extract with 0.8 mM of SeO2 for 48 h at 40°C in pH of 7.0 were optimal conditions for the biosynthesis of SeNPs. The biosynthesized SeNPs were characterized by UV-Vis spectroscopy, X-ray diffraction (XRD), transmission electron microscopy (TEM), and dynamic light scattering (DLS) measurements including average particle size distribution and average zeta potential. The results showed that the biosynthesized SeNPs displayed a maximum absorbance peak in the UV-Vis spectrum at 560 nm due to surface plasmon resonance. TEM image elevated spherical shape particles with an average size of 12 nm. SRB assay, flow cytometry, and other biochemical methods were employed to assess SeNPs anti-proliferative effects on MCF-7 cells. SeNPs showed superior anticancer efficacy against MCF-7 cells compared to colon (HCT-116) and liver (HepG2) cancer cells, as evidenced by lower IC50 values (19.59 µg/ml) against 36.36 µg/ml and 27.81 ±1.4 µg/ml, respectively. However, SeNPs demonstrated no cytotoxic effects against HSF cells. Moreover, treatment with SeNPs induces G2/M arrest along with triggering apoptosis in MCF-7 cells. Furthermore, MCF-7 cells treated with SeNPs showed increased oxidative stress, as indicated by observable rises in LPO and 8-OHDG, accompanied by considerable exhaustion in antioxidant enzyme activity. These findings demonstrated that Se nanoparticles synthesized from yeast have therapeutic promise in BC treatment.
Collapse
Affiliation(s)
- Basant A. Ali
- Microbial Chemistry Department, Biotechology Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Rasha Mosa Allam
- Pharmacology Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Mohamed S. Hasanin
- Cellulose and Paper Department, National Research Centre, Dokki, Cairo 12622, Egypt
| | - Amany A. Hassabo
- Microbial Chemistry Department, Biotechology Research Institute, National Research Centre, Dokki, Cairo 12622, Egypt
| |
Collapse
|
26
|
Ali Ibrahim Mze A, Abdul Rahman A. Repurposing the antipsychotic drug penfluridol for cancer treatment (Review). Oncol Rep 2024; 52:174. [PMID: 39513619 PMCID: PMC11541647 DOI: 10.3892/or.2024.8833] [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: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Cancer is one of the most prevalent diseases and the leading cause of death worldwide. Despite the improved survival rates of cancer in recent years, the current available treatments often face resistance and side effects. Drug repurposing represents a cost‑effective and efficient alternative to cancer treatment. Recent studies revealed that penfluridol (PF), an antipsychotic drug, is a promising anticancer agent. In the present study, a scoping review was conducted to ascertain the anticancer properties of PF. For this, a literature search was performed using the Scopus, PubMed and Web of Science databases with the search string 'penfluridol' AND 'cancer'. A total of 23 original articles with in vivo and/or in vitro studies on the effect of PF on cancer were included in the scoping review. The outcome of the analysis demonstrated the anticancer potential of PF. PF significantly inhibited cell proliferation, metastasis and invasion while inducing apoptosis and autophagy in vivo and across a spectrum of cancer cell lines, including breast, lung, pancreatic, glioblastoma, gallbladder, bladder, oesophageal, leukaemia and renal cancers. However, research on PF derivatives with high anticancer activities and reduced neurological side effects may be necessary.
Collapse
Affiliation(s)
- Asma Ali Ibrahim Mze
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, Selangor 47000, Malaysia
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, Selangor 47000, Malaysia
| | - Amirah Abdul Rahman
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Universiti Teknologi MARA (UiTM), Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, Selangor 47000, Malaysia
- Institute of Medical Molecular Biotechnology, Faculty of Medicine, Universiti Teknologi MARA (UiTM) Cawangan Selangor, Kampus Sungai Buloh, Sungai Buloh, Selangor 47000, Malaysia
| |
Collapse
|
27
|
Mackova V, Raudenska M, Polanska HH, Jakubek M, Masarik M. Navigating the redox landscape: reactive oxygen species in regulation of cell cycle. Redox Rep 2024; 29:2371173. [PMID: 38972297 PMCID: PMC11637001 DOI: 10.1080/13510002.2024.2371173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Objectives: To advance our knowledge of disease mechanisms and therapeutic options, understanding cell cycle regulation is critical. Recent research has highlighted the importance of reactive oxygen species (ROS) in cell cycle regulation. Although excessive ROS levels can lead to age-related pathologies, ROS also play an essential role in normal cellular functions. Many cell cycle regulatory proteins are affected by their redox status, but the precise mechanisms and conditions under which ROS promote or inhibit cell proliferation are not fully understood.Methods: This review presents data from the scientific literature and publicly available databases on changes in redox state during the cell cycle and their effects on key regulatory proteins.Results: We identified redox-sensitive targets within the cell cycle machinery and analysed different effects of ROS (type, concentration, duration of exposure) on cell cycle phases. For example, moderate levels of ROS can promote cell proliferation by activating signalling pathways involved in cell cycle progression, whereas excessive ROS levels can induce DNA damage and trigger cell cycle arrest or cell death.Discussion: Our findings encourage future research focused on identifying redox-sensitive targets in the cell cycle machinery, potentially leading to new treatments for diseases with dysregulated cell proliferation.
Collapse
Affiliation(s)
- Viktoria Mackova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Martina Raudenska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Hana Holcova Polanska
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
| | - Michal Masarik
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czech Republic
- Institute of Pathophysiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
28
|
Lee HY, Park JS, Kim TG, Kim T, Kim DH, Yun J, Jeong YI. Reactive Oxygen Species-Sensitive Nanophotosensitizers Composed of Buthionine Sulfoximine-Conjugated Chitosan Oligosaccharide for Enhanced Photodynamic Treatment of Cancer Cells. Int J Mol Sci 2024; 25:12609. [PMID: 39684320 DOI: 10.3390/ijms252312609] [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: 10/17/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
The efficacy of photodynamic therapy (PDT) based on traditional photosensitizers is generally limited by the cellular redox homeostasis system due to the reactive oxygen species (ROS) scavenging effect of glutathione (GSH). In this study, buthionine sulfoximine (BSO), a GSH inhibitor, was conjugated with the amine group of chitosan oligosaccharide (COS) using a thioketal linker (COSthBSO) to liberate BSO and chlorine e6 (Ce6) under oxidative stress, and then, Ce6-COSthBSO NP (Ce6-COSthBSO NP), fabricated by a dialysis procedure, showed an accelerated release rate of BSO and Ce6 by the addition of hydrogen peroxide, indicating that nanophotosensitizers have ROS sensitivity. In the in vitro cell culture study using HCT116 colon carcinoma cells, a combination of BSO and Ce6 efficiently suppressed the intracellular GSH and increased ROS production compared to the sole treatment of Ce6. In particular, Ce6-COSthBSO NP showed higher efficacy in the suppression of GSH levels and ROS production compared to the free Ce6 and Ce6/BSO combination. These results were due to the fact that Ce6-COSthBSO NP was efficiently delivered to the intracellular region, suppressed intracellular GSH levels, and elevated ROS levels. The in vivo animal tumor xenograft study demonstrated Ce6-COSthBSO NP being efficiently delivered to the tumor tissue, i.e., the fluorescence intensity in the tumor tissue was higher than those of other organs. The combination of Ce6 and BSO efficiently suppressed tumor growth compared to the sole treatment of Ce6, indicating that BSO might efficiently suppress GSH levels and increase ROS levels in the tumor microenvironment. Specifically, Ce6-COSthBSO NP showed the strongest performance in inhibition of tumor growth than those of Ce6 or the CE6/BSO combination, indicating that they were efficiently delivered to tumor tissue, increased ROS levels, and then efficiently inhibited tumor growth. We suggest that COSthBSO nanophotosensitizers are promising candidates for PDT treatment of cancer cells.
Collapse
Affiliation(s)
- Hyo Young Lee
- Department of Radiological Science, Dong-Eui University, Pusan 47340, Republic of Korea
| | - Ji-Sun Park
- Interdisciplinary Program of Perfume and Cosmetic, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Taeyu Grace Kim
- Wellesley College, 106 Central Street, Wellesley, MA 02481, USA
| | - Taeyeon Kim
- College of Arts and Sciences, University of Pennsylvania, 20 Cohen Hall, 249 South 36th St, Philadelphia, PA 19104, USA
| | - Do Hoon Kim
- Tyros Biotechnology Inc., 75 Kneeland St. 14 Floors, Boston, MA 02111, USA
| | - Jejung Yun
- Jeonnam Bio Foundation (JBF), Food Research Center, Naju City 58275, Jeonnam, Republic of Korea
| | - Young-Il Jeong
- Department of Dental Materials, College of Dentistry, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
29
|
Minisini M, Mascaro M, Brancolini C. HDAC-driven mechanisms in anticancer resistance: epigenetics and beyond. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:46. [PMID: 39624079 PMCID: PMC11609180 DOI: 10.20517/cdr.2024.103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 01/03/2025]
Abstract
The emergence of drug resistance leading to cancer recurrence is one of the challenges in the treatment of cancer patients. Several mechanisms can lead to drug resistance, including epigenetic changes. Histone deacetylases (HDACs) play a key role in chromatin regulation through epigenetic mechanisms and are also involved in drug resistance. The control of histone acetylation and the accessibility of regulatory DNA sequences such as promoters, enhancers, and super-enhancers are known mechanisms by which HDACs influence gene expression. Other targets of HDACs that are not histones can also contribute to resistance. This review describes the contribution of HDACs to the mechanisms that, in some cases, may determine resistance to chemotherapy or other cancer treatments.
Collapse
Affiliation(s)
| | | | - Claudio Brancolini
- Laboratory of Epigenomics, Department of Medicine, Università degli Studi di Udine, Udine 33100, Italy
| |
Collapse
|
30
|
Rogalewicz B, Sierański T, Szczesio M, Olczak A, Gobis K, Orlewska C, Korona-Głowniak I, Korga-Plewko A, Iwan M, Michalczuk M, Kubik J, Adamczuk G, Korga M, Rutkowska N, Boruta T, Gas K, Sawicki M, Poleszak E, Maniukiewicz W, Świątkowski M, Czylkowska A. Physicochemical properties and mechanism of action of a new copper(ii) pyrazine-based complex with high anticancer activity and selectivity towards cancer cells. RSC Adv 2024; 14:36295-36307. [PMID: 39534047 PMCID: PMC11556459 DOI: 10.1039/d4ra06874b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Two compounds, benzyl-2-(amino(pyrazin-2-yl)methylene)-1-methylhydrazine-1-carbodithioate (L) and its copper(ii) complex Cu(L) were synthesized and studied in terms of their physicochemical properties, including single crystal, spectroscopic and magnetic properties; in silico simulations, including DFT calculations and pharmacokinetic profile analysis; and in vitro biological activity. The Cu(L) compound was found to exhibit good anticancer activity against A375, PANC-1, MKN-74, T-47D, HeLa, and NCI-H1563 cells, with the IC50 value against the HeLa cell line reaching 17.50 μM, significantly surpassing the activity of the organic ligand. Moreover, at the same time, the Cu(L) complex did not exhibit significant toxicity towards healthy cells. Mechanism of action studies revealed that its activity is connected with the oxidative stress and redox imbalance caused by the upregulation of genes encoding superoxide dismutase (SOD2) and catalase (CAT) antioxidant enzymes. The reported results further underscore the anticancer potential of pyrazine-based copper(ii) complexes.
Collapse
Affiliation(s)
- B Rogalewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - T Sierański
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - M Szczesio
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - A Olczak
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - K Gobis
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Gdansk Gen. Hallera 107 Gdańsk 80-416 Poland
| | - C Orlewska
- Department of Organic Chemistry, Faculty of Pharmacy, Medical University of Gdansk Gen. Hallera 107 Gdańsk 80-416 Poland
| | - I Korona-Głowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin Lublin 20-093 Poland
| | - A Korga-Plewko
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - M Iwan
- Department of Toxicology, Medical University of Lublin Chodźki 8b Lublin 20-093 Poland
| | - M Michalczuk
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - J Kubik
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - G Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - M Korga
- Independent Medical Biology Unit, Medical University of Lublin Jaczewskiego 8b Lublin 20-093 Poland
| | - N Rutkowska
- Institute of Molecular and Industrial Biotechnology, Lodz University of Technology Stefanowskiego 2/22 Lodz 90-537 Poland
| | - T Boruta
- Faculty of Process and Environmental Engineering, Department of Bioprocess Engineering, Lodz University of Technology ul. Wolczanska 213 Lodz 93-005 Poland
| | - K Gas
- Institute of Physics, Polish Academy of Sciences Aleja Lotnikow 32/46 Warsaw PL-02668 Poland
| | - M Sawicki
- Institute of Physics, Polish Academy of Sciences Aleja Lotnikow 32/46 Warsaw PL-02668 Poland
| | - E Poleszak
- Laboratory of Preclinical Testing, Chair and Department of Applied and Social Pharmacy, Medical University of Lublin Chodzki 1 Lublin 20-093 Poland
| | - W Maniukiewicz
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - M Świątkowski
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| | - A Czylkowska
- Institute of General and Ecological Chemistry, Faculty of Chemistry, Lodz University of Technology Żeromskiego 116 Lodz 90-924 Poland
| |
Collapse
|
31
|
Xie H, Tang J, Song L, Xu G, Li W, Zhu J, Liu Y, Ma H, Cai L, Han XX. Mitochondria-endoplasmic reticulum crosstalk in apoptosis: The interactions of cytochrome c with monooxygenase and its reductase. Int J Biol Macromol 2024; 279:135160. [PMID: 39214221 DOI: 10.1016/j.ijbiomac.2024.135160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The crosstalk between endoplasmic reticulum and mitochondria is of significance in apoptosis, in which cytochrome b5 (Cyt b5) is thought to be a major target for cytochrome c (Cyt c) upon its release from the mitochondria. In the absence of Cyt b5, the role of interactions of Cyt c with CYP-dependent monooxygenase system in apoptotic regulation was explored in this study. NADPH-dependent and Cyt c-induced formation of reactive oxygen species (ROS) and NADPH-independent Cyt c unfolding were revealed. With the aid of a CPR inhibitor and CYP antibodies, the interactions among Cyt c, cytochrome P450 reductase (CPR) and cytochrome P450 (CYP) are evidenced, which are found crucial for monooxygenase-derived ROS formation. The underlying structural basis of Cyt c-CYP complex was unveiled by molecular dynamics simulations. This study provides novel insights into how Cyt c regulates ROS formation through the interactions with CPR and CYP, and is implicated for a deeper understanding of the regulation mechanism in the mitochondria-endoplasmic reticulum apoptotic pathway.
Collapse
Affiliation(s)
- Han Xie
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Jinping Tang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Li Song
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Guangyang Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Wei Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Jinyu Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Yawen Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China
| | - Hao Ma
- State Key Laboratory of Physical Chemistry Solid Surfaces, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, PR China.
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, 130012, PR China
| | - Xiao Xia Han
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, PR China.
| |
Collapse
|
32
|
Zheng YY, Tong XY, Zhang DY, Ouyang JM. Enhancement of Antioxidative and Anti-Inflammatory Activities of Corn Silk Polysaccharides After Selenium Modification. J Inflamm Res 2024; 17:7965-7991. [PMID: 39502937 PMCID: PMC11537195 DOI: 10.2147/jir.s467665] [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: 05/17/2024] [Accepted: 09/07/2024] [Indexed: 11/08/2024] Open
Abstract
Objective This study aimed to study the effect of selenium modification on the bioactivity of corn silk polysaccharides, particularly its antioxidant and anti-inflammatory functions. Methods HNO3-NaSeO3 was used to selenize degraded corn silk polysaccharides (DCSP). The structure and physicochemical properties of DCSP and selenized corn silk polysaccharides (Se-DCSP) were characterized by inductively coupled plasma emission spectroscopy, Fourier-transform infrared, ultraviolet-visible spectroscopy, nuclear magnetic resonance, nanometer, scanning electron microscopy, and thermogravimetric analysis. The protective effects of DCSP and Se-DCSP on HK-2 cells damaged by nano-calcium oxalate and the changes of inflammatory factors were detected by laser confocal microscopy, flow cytometry, and fluorescence microscopy. Results The selenium content of DCSP and Se-DCSP were 19.5 and 1226.7 μg/g, respectively. Compared with DCSP, Se-DCSP showed significantly improved biological activity, including the scavenging ability of various free radicals (increased by about 2-3 times), the intracellular reactive oxygen content (decreased by about 1.5 times), and the mitochondrial membrane potential (decreased by about 2.5 times). Moreover, cell viability and morphological recovery ability were improved. Compared with DCSP, Se-DCSP significantly down-regulated HK-2 cell inflammatory factors MCP-1 (about 1.7 times), NLRP3, and NO (about 1.5 times). Conclusion The antioxidant activity and the ability to down-regulate the expression of inflammatory factors of Se-DCSP were significantly enhanced compared with DCSP, and Se-DCSP can better protect HK-2 cells from oxidative damage, indicating that Se-DCSP has a stronger potential ability to inhibit kidney stone formation.
Collapse
Affiliation(s)
- Yu-Yun Zheng
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science; Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xin-Yi Tong
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science; Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Da-Ying Zhang
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science; Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science; Jinan University, Guangzhou, 510632, People’s Republic of China
| |
Collapse
|
33
|
Gonçalves B, Aires A, Oliveira I, Baltazar M, Cosme F, Afonso S, Pinto T, Anjos MR, Inês A, Morais MC, Vilela A, Silva AP. From Orchard to Wellness: Unveiling the Health Effects of Sweet Cherry Nutrients. Nutrients 2024; 16:3660. [PMID: 39519493 PMCID: PMC11547742 DOI: 10.3390/nu16213660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
This review paper explores the multifaceted relationship between sweet cherry nutrients and human health, aiming to uncover the comprehensive impact of these bioactive compounds from orchard to wellness. Furthermore, it highlights how advanced crop techniques can be pivotal in optimizing these beneficial compounds. Synthesizing existing literature, the paper examines the diverse bioactive nutrients in sweet cherries, including antioxidants, polyphenols, vitamins, and minerals, and elucidating their mechanisms of action and potential health benefits. From antioxidant properties to anti-inflammatory effects, the paper elucidates how these nutrients may mitigate chronic diseases such as cardiovascular disorders, diabetes, and neurodegenerative conditions. Additionally, it explores their role in promoting gastrointestinal health, enhancing exercise recovery, and modulating sleep patterns. The review discusses emerging research on the potential anti-cancer properties of sweet cherry compounds, highlighting their promising role in cancer prevention and treatment. Furthermore, it delves into the impact of sweet cherry consumption on metabolic health, weight management, and skin health. By providing a comprehensive overview of the current understanding of sweet cherry nutrients and their health effects, this paper offers valuable insights for researchers, healthcare professionals, and consumers interested in utilizing nature's bounty for holistic wellness.
Collapse
Affiliation(s)
- Berta Gonçalves
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Alfredo Aires
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Ivo Oliveira
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Miguel Baltazar
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Fernanda Cosme
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Sílvia Afonso
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Teresa Pinto
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Maria Rosário Anjos
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - António Inês
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Maria Cristina Morais
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| | - Alice Vilela
- Chemistry Research Centre-Vila Real (CQ-VR), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (F.C.); (A.I.); (A.V.)
| | - Ana Paula Silva
- Centre for the Research and Technology of Agro-Environmental and Biological Sciences (CITAB), Institute for Innovation, Capacity Building and Sustainability of Agri-Food Production (Inov4Agro), University of Trás-of-Montes e Alto Douro, Quinta de Prados, 5000-801 Vila Real, Portugal; (A.A.); (I.O.); (M.B.); (S.A.); (T.P.); (M.R.A.); (M.C.M.); (A.P.S.)
| |
Collapse
|
34
|
Khan SH, Anees M, Zofair SFF, Rasool F, Khan MA, Moin S, Younus H. Fucoidan based polymeric nanoparticles encapsulating epirubicin: A novel and effective chemotherapeutic formulation against colorectal cancer. Int J Pharm 2024; 664:124622. [PMID: 39197799 DOI: 10.1016/j.ijpharm.2024.124622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/26/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
Colorectal cancer (CRC) is one of the most common and challenging malignancy that needs some effective and safer chemotherapeutic agents for the treatment. In this study, anticancer agent epirubicin (Epi) was loaded in polymeric polyethylene glycol-polylactic acid-nanoparticles (mPEG-PLA-NPs) coated with a marine anti-cancer non-toxic polysaccharide fucoidan (FC), to achieve a synergistic activity against CRC. The characterization of the NPs revealed that they were spherical, monodispersed, stable, with a negative zeta potential, and exhibited good biocompatibility and controlled release. In vitro anti-cancer activity of the NPs on HCT116 cell line was found to be promising, and corroborated well with in vivo studies involving BALB/C mice injected with C26 murine cancer cells. The outcome of MTT assay demonstrated that IC50 value of free Epi was 3.72 µM, and that of non-coated and coated Epi nano-formulations was 33.67 and 10.19 µM, respectively. Higher tumor regression, better survival and reduced off-side cardiotoxicity were observed when this novel NPs formulation was used to treat tumor-bearing mice. Free FC and Epi treated mice showed 37.73 % and 61.49 % regression in tumor size, whereas there was 79.76 % and 90.34 % tumor regression in mice treated with non-coated Epi NPs and coated Epi NPs, respectively. Therefore, mPEG-PLA-FC-Epi-NPs hold a potential to be used as an effective chemotherapeutic formulation against CRC, since it exhibited better efficacy and lower toxicity.
Collapse
Affiliation(s)
- Shaheer Hasan Khan
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Mohd Anees
- Centre for Biomedical Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Syeda Fauzia Farheen Zofair
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Fayyaz Rasool
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Greater Noida 201314, India
| | - Masood Alam Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Shagufta Moin
- Department of Biochemistry, J.N.M.C., Faculty of Medicine, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Hina Younus
- Interdisciplinary Biotechnology Unit, Faculty of Life Sciences, Aligarh Muslim University, Aligarh 202002, UP, India.
| |
Collapse
|
35
|
Dai X, Feng S, Zheng Y. Cold Atmospheric Plasma: Possible Cure of Autoimmune Disorders and Cancer via Attenuating Inflammation. Int J Biol Sci 2024; 20:5436-5449. [PMID: 39494336 PMCID: PMC11528447 DOI: 10.7150/ijbs.102445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Autoimmune diseases and cancers, two seemingly unrelated diseases, have been threatening human health, and many of them have no cure. By identifying pathological inflammation as the driving cause of uncontrolled cell proliferation in both classes of diseases, and differentiating autoimmune disorders and cancers by whether the cell death programs are under control, we propose the attenuation of prolonged inflammation via maintaining mitochondrial reduction-oxidation (redox) homeostasis being a possible cure of both diseases. Importantly, we propose the feasibility of applying cold atmospheric plasma (CAP) in treating autoimmune disorders and cancers given its redox-modulatory nature, which not only extends the medical utilities of CAP to autoimmune diseases and all other inflammation-driven disorders, but also positions the efficacy of CAP against cancer cells to its suppressive role on prolonged inflammation. Our insights may open an innovative avenue towards a unified view on the molecular mechanism driving the diversified types of medical miracles of CAP and what CAP can do in the field of plasma medicine.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Shuo Feng
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Yan Zheng
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
36
|
Yun HM, Kim SH, Kwon YJ, Park KR. Effect of Spicatoside a on Anti-Osteosarcoma MG63 Cells through Reactive Oxygen Species Generation and the Inhibition of the PI3K-AKT-mTOR Pathway. Antioxidants (Basel) 2024; 13:1162. [PMID: 39456416 PMCID: PMC11505237 DOI: 10.3390/antiox13101162] [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: 08/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Osteosarcoma is a primary malignant tumor found in the bones of children and adolescents. Unfortunately, many patients do not respond well to treatment and succumb to the illness. Therefore, it is necessary to discover novel bioactive compounds to overcome therapeutic limitations. Liriope platyphylla Wang et Tang is a well-known herb used in oriental medicine. Studies have shown that metabolic diseases can be clinically treated using the roots of L. platyphylla. Recent studies have demonstrated the anticarcinoma potential of root extracts; however, the exact mechanism remains unclear. The aim of this study was to examine the anti-osteosarcoma activity of a single compound extracted from the dried roots of L. platyphylla. We purified Spicatoside A (SpiA) from the dried roots of L. platyphylla. SpiA significantly inhibited the proliferation of human osteosarcoma MG63 cells in a dose- and time-dependent manner. SpiA also regulated the expression of various downstream proteins that mediate apoptosis (PARP, Bcl-2, and Bax), cell growth (cyclin D1, Cdk4, and Cdk6), angiogenesis (VEGF), and metastasis (MMP13). The Proteome Profiler Human Phospho-Kinase Array Kit showed that the AKT signaling protein was a target of SpiA in osteosarcoma cells. We also found that SpiA suppressed the constitutive activation of the PI3K-AKT-mTOR-p70S6K1 signaling pathway. We further validated the effects of SpiA on the AKT signaling pathway. SpiA induced autophagosome formation and suppressed necroptosis (a form of programmed cell death). SpiA increased the generation of reactive oxygen species (ROS) and led to the loss of mitochondrial membrane potential. N-acetylcysteine (NAC)-induced inhibition of ROS generation reduced SpiA-induced AKT inhibition, apoptotic cell death, and anti-metastatic effects by suppressing cell migration and invasion. Overall, these results highlight the anti-osteosarcoma effect of SpiA by inhibiting the AKT signaling pathway through ROS generation, suggesting that SpiA may be a promising compound for the treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Soo Hyun Kim
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.-J.K.)
| | - Yoon-Ju Kwon
- National Development Institute for Korean Medicine, Gyeongsan 38540, Republic of Korea; (S.H.K.); (Y.-J.K.)
| | - Kyung-Ran Park
- Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea
| |
Collapse
|
37
|
Napiórkowska M, Otto-Ślusarczyk D, Kurpios-Piec D, Stukan I, Gryzik M, Wojda U. BM7, a derivative of benzofuran, effectively fights cancer by promoting cancer cell apoptosis and impacting IL-6 levels. Eur J Pharmacol 2024; 978:176751. [PMID: 38897442 DOI: 10.1016/j.ejphar.2024.176751] [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: 02/07/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/21/2024]
Abstract
The BM7 compound, a bromo derivative of methyl 6-acetyl-5-hydroxy-2-methyl-1-benzofuran-3-carboxylate, was previously identified as cytotoxic to human leukaemia cells (K562 and HL60) and human cervical cancer (HeLa), while showing no toxicity to non-cancerous primary endothelial cells (HUVEC). In this study, we present the first demonstration of BM7's anticancer efficacy in vivo using a mouse chronic myeloid leukaemia xenograft model. Administered intraperitoneally in a mixture of 10% Solutol HS 15/10% ethanol, BM7 exhibited no visible toxicity and significantly reduced tumor weight, comparable to standard drugs imatinib and hydroxyurea. Further supporting its anticancer potential, a multi-model in vitro study involving seven human cancer cell lines revealed the most promising responses in colon cancer (SW480, SW620, HCT116), liver cancer (HEPG2), and breast adenocarcinoma (MDA-MB-231) cells. BM7 demonstrated multifaceted anticancer mechanisms, inducing apoptosis while elevating reactive oxygen species (ROS) levels and suppressing interleukin-6 (IL-6) release in these cell lines. These findings position BM7 as a candidate of significant interest for cancer therapy. Its ability to not only induce apoptosis but also modulate cellular processes such as ROS levels and immune responses, specifically IL-6 suppression, makes BM7 a versatile and promising agent for further exploration in the realm of cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Iga Stukan
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland; Department of General Pathology, Faculty of Medicine and Dentistry, Pomeranian Medical University in Szczecin, 1 Rybacka Street, 70-204, Szczecin, Poland
| | - Marek Gryzik
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, 3 Pasteur Street, 02-093, Warsaw, Poland
| |
Collapse
|
38
|
Kazemi KS, Kazemi P, Mivehchi H, Nasiri K, Eshagh Hoseini SS, Nejati ST, Pour Bahrami P, Golestani S, Nabi Afjadi M. Photodynamic Therapy: A Novel Approach for Head and Neck Cancer Treatment with Focusing on Oral Cavity. Biol Proced Online 2024; 26:25. [PMID: 39154015 PMCID: PMC11330087 DOI: 10.1186/s12575-024-00252-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/31/2024] [Indexed: 08/19/2024] Open
Abstract
Oral cancers, specifically oral squamous cell carcinoma (OSCC), pose a significant global health challenge, with high incidence and mortality rates. Conventional treatments such as surgery, radiotherapy, and chemotherapy have limited effectiveness and can result in adverse reactions. However, as an alternative, photodynamic therapy (PDT) has emerged as a promising option for treating oral cancers. PDT involves using photosensitizing agents in conjunction with specific light to target and destroy cancer cells selectively. The photosensitizers accumulate in the cancer cells and generate reactive oxygen species (ROS) upon exposure to the activating light, leading to cellular damage and ultimately cell death. PDT offers several advantages, including its non-invasive nature, absence of known long-term side effects when administered correctly, and cost-effectiveness. It can be employed as a primary treatment for early-stage oral cancers or in combination with other therapies for more advanced cases. Nonetheless, it is important to note that PDT is most effective for superficial or localized cancers and may not be suitable for larger or deeply infiltrating tumors. Light sensitivity and temporary side effects may occur but can be managed with appropriate care. Ongoing research endeavors aim to expand the applications of PDT and develop novel photosensitizers to further enhance its efficacy in oral cancer treatment. This review aims to evaluate the effectiveness of PDT in treating oral cancers by analyzing a combination of preclinical and clinical studies.
Collapse
Affiliation(s)
- Kimia Sadat Kazemi
- Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Kazemi
- Faculty of Dentistry, Ilam University of Medical Sciences, Ilam, Iran
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Kamyar Nasiri
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | | | | | | | - Shayan Golestani
- Department of Oral and Maxillofacial Surgery, Dental School, Islamic Azad University, Isfahan, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
39
|
Majirská M, Pilátová MB, Kudličková Z, Vojtek M, Diniz C. Targeting hematological malignancies with isoxazole derivatives. Drug Discov Today 2024; 29:104059. [PMID: 38871112 DOI: 10.1016/j.drudis.2024.104059] [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: 05/18/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Compounds with a heterocyclic isoxazole ring are well known for their diverse biologic activities encompassing antimicrobial, antipsychotic, immunosuppressive, antidiabetic and anticancer effects. Recent studies on hematological malignancies have also shown that some of the isoxazole-derived compounds feature encouraging cancer selectivity, low toxicity to normal cells and ability to overcome cancer drug resistance of conventional treatments. These characteristics are particularly promising because patients with hematological malignancies face poor clinical outcomes caused by cancer drug resistance or relapse of the disease. This review summarizes the knowledge on isoxazole-derived compounds toward hematological malignancies and provides clues on their mechanism(s) of action (apoptosis, cell cycle arrest, ROS production) and putative pharmacological targets (c-Myc, BET, ATR, FLT3, HSP90, CARM1, tubulin, PD-1/PD-L1, HDACs) wherever known.
Collapse
Affiliation(s)
- Monika Majirská
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martina Bago Pilátová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia.
| | - Zuzana Kudličková
- NMR Laboratory, Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
40
|
Shakeel I, Haider S, Khan S, Ahmed S, Hussain A, Alajmi MF, Chakrabarty A, Afzal M, Imtaiyaz Hassan M. Thymoquinone, artemisinin, and thymol attenuate proliferation of lung cancer cells as Sphingosine kinase 1 inhibitors. Biomed Pharmacother 2024; 177:117123. [PMID: 39004062 DOI: 10.1016/j.biopha.2024.117123] [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: 05/03/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024] Open
Abstract
Sphingosine-1-phosphate (S1P) formed via catalytic actions of sphingosine kinase 1 (SphK1) behaves as a pro-survival substance and activates downstream target molecules associated with various pathologies, including initiation, inflammation, and progression of cancer. Here, we aimed to investigate the SphK1 inhibitory potentials of thymoquinone (TQ), Artemisinin (AR), and Thymol (TM) for the therapeutic management of lung cancer. We implemented docking, molecular dynamics (MD) simulations, enzyme inhibition assay, and fluorescence measurement studies to estimate binding affinity and SphK1 inhibitory potential of TQ, AR, and TM. We further investigated the anti-cancer potential of these compounds on non-small cell lung cancer (NSCLC) cell lines (H1299 and A549), followed by estimation of mitochondrial ROS, mitochondrial membrane potential depolarization, and cleavage of DNA by comet assay. Enzyme activity and fluorescence binding studies suggest that TQ, AR, and TM significantly inhibit the activity of SphK1 with IC50 values of 35.52 µM, 42.81 µM, and 53.68 µM, respectively, and have an excellent binding affinity. TQ shows cytotoxic effect and anti-proliferative potentials on H1299 and A549 with an IC50 value of 27.96 µM and 54.43 µM, respectively. Detection of mitochondrial ROS and mitochondrial membrane potential depolarization shows promising TQ-induced oxidative stress on H1299 and A549 cell lines. Comet assay shows promising TQ-induced oxidative DNA damage. In conclusion, TQ, AR, and TM act as potential inhibitors for SphK1, with a strong binding affinity. In addition, the cytotoxicity of TQ is linked to oxidative stress due to mitochondrial ROS generation. Overall, our study suggests that TQ is a promising inhibitor of SphK1 targeting lung cancer therapy.
Collapse
Affiliation(s)
- Ilma Shakeel
- Department of Zoology, Aligarh Muslim University, Aligarh, UP 202001, India; Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Shaista Haider
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Deemed to be University, Gautam Buddha Nagar, UP 201314, India
| | - Shama Khan
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Science, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Shahbaz Ahmed
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Anindita Chakrabarty
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence, Deemed to be University, Gautam Buddha Nagar, UP 201314, India
| | - Mohammad Afzal
- Department of Zoology, Aligarh Muslim University, Aligarh, UP 202001, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
41
|
Elias MG, Aputen AD, Fatima S, Mann TJ, Karan S, Mikhael M, de Souza P, Gordon CP, Scott KF, Aldrich-Wright JR. Chemotherapeutic Potential of Chlorambucil-Platinum(IV) Prodrugs against Cisplatin-Resistant Colorectal Cancer Cells. Int J Mol Sci 2024; 25:8252. [PMID: 39125821 PMCID: PMC11312340 DOI: 10.3390/ijms25158252] [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: 06/12/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Chlorambucil-platinum(IV) prodrugs exhibit multi-mechanistic chemotherapeutic activity with promising anticancer potential. The platinum(II) precursors of the prodrugs have been previously found to induce changes in the microtubule cytoskeleton, specifically actin and tubulin of HT29 colon cells, while chlorambucil alkylates the DNA. These prodrugs demonstrate significant anticancer activity in 2D cell and 3D spheroid viability assays. A notable production of reactive oxygen species has been observed in HT29 cells 72 h post treatment with prodrugs of this type, while the mitochondrial membrane potential was substantially reduced. The cellular uptake of the chlorambucil-platinum(IV) prodrugs, assessed by ICP-MS, confirmed that active transport was the primary uptake mechanism, with platinum localisation identified primarily in the cytoskeletal fraction. Apoptosis and necrosis were observed at 72 h of treatment as demonstrated by Annexin V-FITC/PI assay using flow cytometry. Immunofluorescence measured via confocal microscopy showed significant changes in actin and tubulin intensity and in architecture. Western blot analysis of intrinsic and extrinsic pathway apoptotic markers, microtubule cytoskeleton markers, cell proliferation markers, as well as autophagy markers were studied post 72 h of treatment. The proteomic profile was also studied with a total of 1859 HT29 proteins quantified by mass spectroscopy, with several dysregulated proteins. Network analysis revealed dysregulation in transcription, MAPK markers, microtubule-associated proteins and mitochondrial transport dysfunction. This study confirms that chlorambucil-platinum(IV) prodrugs are candidates with promising anticancer potential that act as multi-mechanistic chemotherapeutics.
Collapse
Affiliation(s)
- Maria George Elias
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| | - Angelico D. Aputen
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Shadma Fatima
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Timothy J. Mann
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Shawan Karan
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Meena Mikhael
- Mass Spectrometry Facility, Western Sydney University, Sydney, NSW 2751, Australia;
| | - Paul de Souza
- Nepean Clinical School, Faculty of Medicine and Health, University of Sydney, Kingswood, NSW 2747, Australia;
| | - Christopher P. Gordon
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
| | - Kieran F. Scott
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
- School of Medicine, Western Sydney University, Sydney, NSW 2751, Australia
| | - Janice R. Aldrich-Wright
- School of Science, Western Sydney University, Sydney, NSW 2751, Australia; (M.G.E.); (A.D.A.); (S.K.); (C.P.G.)
- Medical Oncology, Ingham Institute for Applied Medical Research, Liverpool Hospital, Liverpool, NSW 2170, Australia; (S.F.); (T.J.M.); (K.F.S.)
| |
Collapse
|
42
|
Iskandar A, Kim SK, Wong TW. “Drug-Free” chitosan nanoparticles as therapeutic for cancer treatment. POLYM REV 2024; 64:818-871. [DOI: 10.1080/15583724.2024.2323943] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 11/22/2023] [Accepted: 02/19/2024] [Indexed: 01/06/2025]
Affiliation(s)
- Athirah Iskandar
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
| | - Se-Kwon Kim
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Seoul, Republic of Korea
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam, Malaysia
- Sino-Malaysia Molecular Oncology and Traditional Chinese Medicine Delivery Joint Research Centre, Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
43
|
Jiang L, Liu Y, Tumbath S, Boudreau MW, Chatkewitz LE, Wang J, Su X, Zahid KR, Li K, Chen Y, Yang K, Hergenrother PJ, Huang X. Isopentyl-Deoxynboquinone Induces Mitochondrial Dysfunction and G2/M Phase Cell Cycle Arrest to Selectively Kill NQO1-Positive Pancreatic Cancer Cells. Antioxid Redox Signal 2024; 41:74-92. [PMID: 37950707 PMCID: PMC11321107 DOI: 10.1089/ars.2022.0224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 11/13/2023]
Abstract
Aims: Pancreatic cancer is among the top five leading causes of cancer-related deaths worldwide, with poor overall survival rates. Current therapies for pancreatic cancer lack tumor specificity, resulting in harmful effects on normal tissues. Therefore, developing tumor-specific agents for the treatment of pancreatic cancer is critical. NAD(P)H:quinone oxidoreductase 1 (NQO1), highly expressed in pancreatic cancers but not in associated normal tissues, makes NQO1 bioactivatable drugs a potential therapy for selectively killing NQO1-positive cancer cells. Our previous studies have revealed that the novel NQO1 bioactivatable drug deoxynyboquinone (DNQ) is 10-fold more potent than the prototypic NQO1 bioactivatable drug β-lapachone in killing of NQO1-positive cancer cells. However, DNQ treatment results in high-grade methemoglobinemia, a significant side effect that limits clinical development. Results: Here, we report for the first time on a DNQ derivative, isopentyl-deoxynboquinone (IP-DNQ), which selectively kills pancreatic ductal adenocarcinoma (PDAC) cells in an NQO1-dependent manner with equal potency to the parent DNQ. IP-DNQ evokes massive reactive oxygen species (ROS) production and oxidative DNA lesions that result in poly(ADP-ribose)polymerase-1 (PARP1) hyperactivation, mitochondrial catastrophe, and G2/M phase cell cycle arrest, leading to apoptotic and necrotic programmed cell death. Importantly, IP-DNQ treatment causes only mild methemoglobinemia in vivo, with a threefold improvement in the maximum tolerated dose (MTD) compared with DNQ, while it significantly suppresses tumor growth and extends the life span of mice in subcutaneous and orthotopic pancreatic cancer xenograft models. Innovation and Conclusion: Our study demonstrates that IP-DNQ is a promising therapy for NQO1-positive pancreatic cancers and may enhance the efficacy of other anticancer drugs. IP-DNQ represents a novel approach to treating pancreatic cancer with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Lingxiang Jiang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yingchun Liu
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University/School of Basic Medical Sciences, Fujian Medical University, Fuzhou, P.R. China
| | - Soumya Tumbath
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Matthew W. Boudreau
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Lindsay E. Chatkewitz
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jiangwei Wang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Xiaolin Su
- Department of Biochemistry and Molecular Biology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kashif Rafiq Zahid
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Katherine Li
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Yaomin Chen
- Indiana University Health Pathology Laboratory, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Kai Yang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- The Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul J. Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Xiumei Huang
- Department of Radiation Oncology, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
44
|
Wang Y, Zhang X, Ma Y, Zhou X, Xu W, Qin S, Yang C. Self-assembled copper-based nanoparticles for enzyme catalysis-enhanced chemodynamic/photodynamic/antiangiogenic tritherapy against hepatocellular carcinoma. J Nanobiotechnology 2024; 22:375. [PMID: 38926721 PMCID: PMC11202248 DOI: 10.1186/s12951-024-02626-x] [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: 02/12/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
As an emerging cancer treatment strategy, reactive oxygen species-based tumor catalytic therapies face enormous challenges due to hypoxia and the overexpression of glutathione (GSH) in the tumor microenvironment. Herein, a self-assembled copper-based nanoplatform, TCCHA, was designed for enzyme-like catalysis-enhanced chemodynamic/photodynamic/antiangiogenic tritherapy against hepatocellular carcinoma. TCCHA was fabricated from Cu2+, 3,3'-dithiobis (propionohydrazide), and photosensitizer chlorine e6 via a facile one-pot self-assembly strategy, after which an aldehyde hyaluronic acid was coated, followed by loading of the antivascular drug AL3818. The obtained TCCHA nanoparticles exhibited pH/GSH dual-responsive drug release behaviors and multienzymatic activities, including Fenton, glutathione peroxidase-, and catalase-like activities. TCCHA, a redox homeostasis disruptor, promotes ⋅OH generation and GSH depletion, thus increasing the efficacy of chemodynamic therapy. TCCHA, which has catalase-like activity, can also reinforce the efficacy of photodynamic therapy by amplifying O2 production. In vivo, TCCHA efficiently inhibited tumor angiogenesis and suppressed tumor growth without apparent systemic toxicity. Overall, this study presents a facile strategy for the preparation of multienzyme-like nanoparticles, and TCCHA nanoparticles display great potential for enzyme catalysis-enhanced chemodynamic/photodynamic/antiangiogenic triple therapy against cancer.
Collapse
Affiliation(s)
- Yaping Wang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xun Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yunfeng Ma
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaobo Zhou
- Department of Pathogenic Biology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Sida Qin
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Chengcheng Yang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
45
|
Kosińska K, Skóra B, Holota S, Shepeta Y, Tabęcka-Łonczyńska A, Lesyk R, Szychowski KA. Role of 4-Thiazolidinone-Pyrazoline/Indoline Hybrids Les-4369 and Les-3467 in BJ and A549 Cell Lines. Cells 2024; 13:1007. [PMID: 38920636 PMCID: PMC11202306 DOI: 10.3390/cells13121007] [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: 05/08/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Cancer is one of the most important problems of modern societies. Recently, studies have reported the anticancer properties of rosiglitazone related to its ability to bind peroxisome proliferator receptor γ (PPARγ), which has various effects on cancer and can inhibit cell proliferation. In this study, we investigated the effect of new 4-thiazolidinone (4-TZD) hybrids Les-4369 and Les-3467 and their effect on reactive oxygen species (ROS) production, metabolic activity, lactate dehydrogenase (LDH) release, caspase-3 activity, and gene and protein expression in human foreskin fibroblast (BJ) cells and lung adenocarcinoma (A549) cells. The ROS production and caspase-3 activity were mainly increased in the micromolar concentrations of the studied compounds in both cell lines. Les-3467 and Les-4369 increased the mRNA expression of PPARG, P53 (tumor protein P53), and ATM (ATM serine/threonine kinase) in the BJ cells, while the mRNA expression of these genes (except PPARG) was mainly decreased in the A549 cells treated with both of the tested compounds. Our results indicate a decrease in the protein expression of AhR, PPARγ, and PARP-1 in the BJ cells exposed to 1 µM Les-3467 and Les-4369. In the A549 cells, the protein expression of AhR, PPARγ, and PARP-1 increased in the treatment with 1 µM Les-3467 and Les-4369. We have also shown the PPARγ modulatory properties of Les-3467 and Les-4369. However, both compounds prove weak anticancer properties evidenced by their action at high concentrations and non-selective effects against BJ and A549 cells.
Collapse
Affiliation(s)
- Karolina Kosińska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Bartosz Skóra
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Serhii Holota
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
- Department of Organic Chemistry and Pharmacy, Lesya Ukrainka Volyn National University, Volya Avenue 13, 43025 Lutsk, Ukraine
| | - Yulia Shepeta
- Department of Pharmaceutical Chemistry, National Pirogov Memorial Medical University, Pirogov 56, 21018 Vinnytsia, Ukraine;
| | - Anna Tabęcka-Łonczyńska
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| | - Roman Lesyk
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
- Department of Pharmaceutical, Organic and Bioorganic Chemistry, Danylo Halytsky Lviv National Medical University, Pekarska 69, 79010 Lviv, Ukraine;
| | - Konrad A. Szychowski
- Department of Biotechnology and Cell Biology, Medical College, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225 Rzeszow, Poland; (B.S.); (A.T.-Ł.); (R.L.); (K.A.S.)
| |
Collapse
|
46
|
Ali T, Li D, Ponnamperumage TNF, Peterson AK, Pandey J, Fatima K, Brzezinski J, Jakusz JAR, Gao H, Koelsch GE, Murugan DS, Peng X. Generation of Hydrogen Peroxide in Cancer Cells: Advancing Therapeutic Approaches for Cancer Treatment. Cancers (Basel) 2024; 16:2171. [PMID: 38927877 PMCID: PMC11201821 DOI: 10.3390/cancers16122171] [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: 05/02/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Cancer cells show altered antioxidant defense systems, dysregulated redox signaling, and increased generation of reactive oxygen species (ROS). Targeting cancer cells through ROS-mediated mechanisms has emerged as a significant therapeutic strategy due to its implications in cancer progression, survival, and resistance. Extensive research has focused on selective generation of H2O2 in cancer cells for selective cancer cell killing by employing various strategies such as metal-based prodrugs, photodynamic therapy, enzyme-based systems, nano-particle mediated approaches, chemical modulators, and combination therapies. Many of these H2O2-amplifying approaches have demonstrated promising anticancer effects and selectivity in preclinical investigations. They selectively induce cytotoxicity in cancer cells while sparing normal cells, sensitize resistant cells, and modulate the tumor microenvironment. However, challenges remain in achieving selectivity, addressing tumor heterogeneity, ensuring efficient delivery, and managing safety and toxicity. To address those issues, H2O2-generating agents have been combined with other treatments leading to optimized combination therapies. This review focuses on various chemical agents/approaches that kill cancer cells via H2O2-mediated mechanisms. Different categories of compounds that selectively generate H2O2 in cancer cells are summarized, their underlying mechanisms and function are elucidated, preclinical and clinical studies as well as recent advancements are discussed, and their prospects as targeted therapeutic agents and their therapeutic utility in combination with other treatments are explored. By understanding the potential of these compounds, researchers can pave the way for the development of effective and personalized cancer treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xiaohua Peng
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, 3210 N. Cramer Street, Milwaukee, WI 53211, USA; (T.A.); (D.L.); (T.N.F.P.); (A.K.P.); (J.P.); (K.F.); (J.B.); (J.A.R.J.); (H.G.); (G.E.K.); (D.S.M.)
| |
Collapse
|
47
|
Lakshmi Anvitha N, A G, S V, S B, I G K I. Facile Fabrication of Titanium Carbide (Ti3C2)-Bismuth Vanadate (BiVO4) Nano-Coupled Oxides for Anti-cancer Activity. Cureus 2024; 16:e61492. [PMID: 38952587 PMCID: PMC11216123 DOI: 10.7759/cureus.61492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/01/2024] [Indexed: 07/03/2024] Open
Abstract
Background MXene is a newly discovered substance consisting of 2D transition metal carbides or nitrides, produced through the disintegration and etching of aluminum layers. It possesses numerous properties, including a high surface area, conductivity, strength, stiffness, negative zeta potential, and excellent volumetric capacitance. MXene is utilized in detecting anti-cancer medicine, while bismuth vanadate (BiVO4) is synthesized to form an optimized material for anti-cancer activity applications. BiVO4 exhibits visible light absorption, strong chemical stability, and non-toxic properties. However, when loaded onto target stem cells, it can cause skin and respiratory irritation. Aim This study aimed to evaluate the facile fabrication of titanium carbide (Ti3C2)-BiVO4 nanomaterials coupled with oxides for anti-cancer activity. Moreover, it aimed to create Ti3C2-BiVO4 nanomaterials in combination with oxides using X-ray diffraction (XRD) and scanning electron microscopy (SEM) to assess their potential as efficient and targeted anti-cancer agents. Methods and materials To prepare the 2D Ti3C2 MXene, 2.5 g of titanium aluminum carbide (Ti3AlC2) powder was dissolved in 60 mL of a 40% hydrofluoric acid (HF) solution in a polytetrafluoroethylene(PTFE) container. The etching process was made more efficient and completed in 24 hours by using a magnetic stirring system to keep the mixture stirred and heated continuously. The centrifugation was performed at 4000 rpm for five minutes. Subsequently, deionized water was used to wash the solution many times until its pH reached around 7. The appropriate Ti3C2 powder was made by vacuum drying the acquired sediment at 80°C for 24 hours. Monoclinic BiVO4 samples were synthesized via a hydrothermal method. Typically, 10 mmol of Bi(NO3)3.5H2O was dissolved in 100 mL of a 2 mol/L HNO3 solution and stirred uniformly. Subsequently, 10 mmol of ammonium metavanadate (NH4VO3) was added to the mixed solution. After being stirred for one hour, the mixture was transferred into a 100 mL sealed Teflon-lined stainless steel autoclave at 180°C for 16 hours. After cooling to room temperature, the sediment was washed three times with deionized water, ethanol, and acetone, respectively. Finally, the suspension was dried at 80°C, followed by calcination at 450°C for three hours to obtain BiVO4. Ti3C2-BiVO4 heterostructures were prepared by surface modification Ti3C2 using BiVO4 suspensions by a simple, cost-effective approach. Results Ti3C2 nanosheets were observed with BiVO4 particles, and the high crystalline nature of the compound was confirmed after XRD analysis and energy-dispersive spectroscopy (EDS) analysis. The compound was found to be pure without any impurities and exhibited anti-cancer activity. Conclusion The XRD, field emission scanning electron microscopy(FESEM), and EDS investigations provide an in-depth analysis of the structural, morphological, and compositional characteristics of Ti3C2-BiVO4 sheets. The XRD analysis proves the successful combination of different materials and the presence of crystalline phases. The FESEM imaging technique exposes the shape and arrangement of particles in sheets, while the EDS analysis verifies the elemental composition and uniform distribution. These investigations show that Ti3C2-BiVO4 composites have been successfully synthesized, indicating their potential for use in anti-cancer applications.
Collapse
Affiliation(s)
- Nagubandi Lakshmi Anvitha
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Geetha A
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Vasugi S
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Balachandran S
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Ilangovar I G K
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| |
Collapse
|
48
|
Takahashi K, Tanaka T, Ishihara A, Ohta T. Strobilurin X acts as an anticancer drug by inhibiting protein synthesis and suppressing mitochondrial respiratory chain activity. Discov Oncol 2024; 15:177. [PMID: 38769217 PMCID: PMC11106052 DOI: 10.1007/s12672-024-01041-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/17/2024] [Indexed: 05/22/2024] Open
Abstract
PURPOSE Strobilurins act as antifungal agents by inhibiting the mitochondrial respiratory chain. The cytotoxic activity of strobilurins, focusing on its anticancer activities, has been reported. However, the mechanisms involved in these activities remain unclear. METHODS The cytotoxic effects of strobilurin X isolated from the mycelium of Mucidula. venosolamellata were examined in human cancer cell lines (A549 and HeLa) and normal fibroblasts (WI-38). RESULTS Strobilurin X significantly decreased the viability of A549 and HeLa cells compared to that in the WI-38 cells after 48 h of exposure. The EC50 values for cytotoxicity in the A549, HeLa, and WI-38 cells were 3.4, 5.4, and 16.8 μg/mL, respectively. Strobilurin X inhibited the mitochondrial respiratory chain and enhanced the release of lactate in the A549 cells. The IC50 value of strobilurin X against the mitochondrial respiratory chain complex III activity was 139.8 ng/mL. The cytotoxicity induced by strobilurin X was not completely rescued after adding uridine, methyl pyruvate, or N-acetyl cysteine. Furthermore, pharmacological approaches demonstrated that strobilurin X failed to modulate the mitogen-activated protein kinase family and phosphoinositide 3-kinase-Akt pathways; alternatively, it suppressed protein synthesis independent of uridine. CONCLUSION Strobilurin X induced cytotoxicity by blocking the mitochondrial respiratory chain and suppressing protein synthesis. These findings may aid in the development of novel anticancer drugs using strobilurins.
Collapse
Affiliation(s)
- Kenji Takahashi
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
- Division of Functional Fungal Physiology and Pharmacology, Fungus/Mushroom Resource and Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Tomoya Tanaka
- Graduate School of Sustainability Sciences, Tottori University, Tottori, 680-8553, Japan
| | - Atsushi Ishihara
- Division of Applied Fungal Chemistry, Fungus/Mushroom Resource and Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan
| | - Toshio Ohta
- Department of Veterinary Pharmacology, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan.
- Division of Functional Fungal Physiology and Pharmacology, Fungus/Mushroom Resource and Research Center, Faculty of Agriculture, Tottori University, Tottori, 680-8553, Japan.
| |
Collapse
|
49
|
Horvat N, Chocarro S, Marques O, Bauer TA, Qiu R, Diaz-Jimenez A, Helm B, Chen Y, Sawall S, Sparla R, Su L, Klingmüller U, Barz M, Hentze MW, Sotillo R, Muckenthaler MU. Superparamagnetic Iron Oxide Nanoparticles Reprogram the Tumor Microenvironment and Reduce Lung Cancer Regrowth after Crizotinib Treatment. ACS NANO 2024; 18:11025-11041. [PMID: 38626916 PMCID: PMC11064219 DOI: 10.1021/acsnano.3c08335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 05/01/2024]
Abstract
ALK-positive NSCLC patients demonstrate initial responses to ALK tyrosine kinase inhibitor (TKI) treatments, but eventually develop resistance, causing rapid tumor relapse and poor survival rates. Growing evidence suggests that the combination of drug and immune therapies greatly improves patient survival; however, due to the low immunogenicity of the tumors, ALK-positive patients do not respond to currently available immunotherapies. Tumor-associated macrophages (TAMs) play a crucial role in facilitating lung cancer growth by suppressing tumoricidal immune activation and absorbing chemotherapeutics. However, they can also be programmed toward a pro-inflammatory tumor suppressive phenotype, which represents a highly active area of therapy development. Iron loading of TAMs can achieve such reprogramming correlating with an improved prognosis in lung cancer patients. We previously showed that superparamagnetic iron oxide nanoparticles containing core-cross-linked polymer micelles (SPION-CCPMs) target macrophages and stimulate pro-inflammatory activation. Here, we show that SPION-CCPMs stimulate TAMs to secrete reactive nitrogen species and cytokines that exert tumoricidal activity. We further show that SPION-CCPMs reshape the immunosuppressive Eml4-Alk lung tumor microenvironment (TME) toward a cytotoxic profile hallmarked by the recruitment of CD8+ T cells, suggesting a multifactorial benefit of SPION-CCPM application. When intratracheally instilled into lung cancer-bearing mice, SPION-CCPMs delay tumor growth and, after first line therapy with a TKI, halt the regrowth of relapsing tumors. These findings identify SPIONs-CCPMs as an adjuvant therapy, which remodels the TME, resulting in a delay in the appearance of resistant tumors.
Collapse
Affiliation(s)
- Natalie
K. Horvat
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Sara Chocarro
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Oriana Marques
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Tobias A. Bauer
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ruiyue Qiu
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Alberto Diaz-Jimenez
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Ruprecht
Karl University of Heidelberg, 69120, Heidelberg, Germany
| | - Barbara Helm
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
| | - Yuanyuan Chen
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Stefan Sawall
- X-ray
Imaging and CT, German Cancer Research Center
(DKFZ), Im Neuenheimer
Feld 280, 69120, Heidelberg, Germany
| | - Richard Sparla
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
| | - Lu Su
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - Ursula Klingmüller
- Division
of Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Matthias Barz
- Leiden
Academic Centre for Drug Research (LACDR), Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
- Department
of Dermatology, University Medical Center
of the Johannes Gutenberg University Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias W. Hentze
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- European Molecular Biology Laboratory (EMBL), Meyerhofstr.1, 69117, Heidelberg, Germany
| | - Rocío Sotillo
- Division
of Molecular Thoracic Oncology, German Cancer
Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German
Consortium for Translational Cancer Research (DKTK), 69120, Heidelberg, Germany
| | - Martina U. Muckenthaler
- Department
of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- Molecular
Medicine Partnership Unit (MMPU), Otto-Meyerhof-Zentrum, Im Neuenheimer Feld 350, 69120, Heidelberg, Germany
- German
Center for Lung Research (DZL) and Translational Lung Research Center
Heidelberg (TRLC), 69120, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site, 69120, Heidelberg/Mannheim, Germany
| |
Collapse
|
50
|
Janthamala S, Promraksa B, Thanee M, Duenngai K, Jusakul A, Kongpetch S, Kraiklang R, Thanee K, Pinlaor P, Namwat N, Saya H, Techasen A. Anticancer properties and metabolomic profiling of Shorea roxburghii extracts toward gastrointestinal cancer cell lines. BMC Complement Med Ther 2024; 24:178. [PMID: 38689275 PMCID: PMC11061966 DOI: 10.1186/s12906-024-04479-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 04/22/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Gastrointestinal cancer (GIC) ranks as the highest cause of cancer-related deaths globally. GIC patients are often diagnosed at advanced stages, limiting effective treatment options. Chemotherapy, the common GIC recommendation, has significant disadvantages such as toxicity and adverse effects. Natural products contain substances with diverse pharmacological characteristics that promise for use in cancer therapeutics. In this study, the flower of renowned Asian medicinal plant, Shorea roxburghii was collected and extracted to investigate its phytochemical contents, antioxidant, and anticancer properties on GIC cells. METHODS The phytochemical contents of Shorea roxburghii extract were assessed using suitable methods. Phenolic content was determined through the Folin-Ciocalteu method, while flavonoids were quantified using the aluminum chloride (AlCl3) method. Antioxidant activity was evaluated using the FRAP and DPPH assays. Cytotoxicity was assessed in GIC cell lines via the MTT assay. Additionally, intracellular ROS levels and apoptosis were examined through flow cytometry techniques. The correlation between GIC cell viability and phytochemicals, 1H-NMR analysis was conducted. RESULTS Among the four different solvent extracts, ethyl acetate extract had the highest phenolic and flavonoid contents. Water extract exhibited the strongest reducing power and DPPH scavenging activity following by ethyl acetate. Interestingly, ethyl acetate extract demonstrated the highest inhibitory activity against three GIC cell lines (KKU-213B, HepG2, AGS) with IC50 values of 91.60 µg/ml, 39.38 µg/ml, and 35.59 µg/ml, while showing less toxicity to normal fibroblast cells. Ethyl acetate extract induced reactive oxygen species and apoptosis in GIC cell lines by downregulating anti-apoptotic protein Bcl-2. Metabolic profiling-based screening revealed a positive association between reduced GIC cell viability and phytochemicals like cinnamic acid and its derivatives, ferulic acid and coumaric acid. CONCLUSIONS This study highlights the potential of natural compounds in Shorea roxburghii in the development of more effective and safer anticancer agents as options for GIC as well as shedding light on new avenues for cancer treatment.
Collapse
Affiliation(s)
- Sutthiwan Janthamala
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, Thailand
| | - Bundit Promraksa
- Regional Medical Sciences Center 2 Phitsanulok, Department of Medical Sciences, Ministry of Public Health, Phitsanulok, Thailand
| | - Malinee Thanee
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kunyarat Duenngai
- Department of Thai Traditional Medicine, Faculty of Science and Technology, Phetchabun Rajabhat University, Phetchabun, Thailand
| | - Apinya Jusakul
- Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Ratthaphol Kraiklang
- Nutrition for Health Program, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand
| | - Kidsada Thanee
- Faculty of Public Health, Ubon Ratchathani Rajabhat University, Ubon Ratchathani, Thailand
| | - Porntip Pinlaor
- Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Systems Biosciences and Computational Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Hideyuki Saya
- Cancer Center, Fujita Health University, Toyoake, Aichi, Japan
| | - Anchalee Techasen
- Centre for Research and Development of Medical Diagnostic Laboratories (CMDL), Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|