1
|
Yao Y, Zhou J, Song J, Chen C. Ruxolitinib enhances gastric cancer to chemotherapy by suppressing JAK/STAT3 and inducing mitochondrial dysfunction and oxidative stress. Immunopharmacol Immunotoxicol 2025; 47:263-271. [PMID: 40009869 DOI: 10.1080/08923973.2025.2470344] [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: 09/30/2024] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
OBJECTIVE Chemoresistance in gastric cancer poses a major challenge in treatment, necessitating the development of novel therapeutic strategies. This study evaluates the efficacy of ruxolitinib, a JAK1/2 inhibitor, in both sensitive and resistant gastric cancer cell lines. MATERIALS AND METHODS Gastric cancer cell lines, including sensitive (N87 and AGS) and resistant (N87-R and AGS-R) variants, were treated with ruxolitinib alone or in combination with chemotherapeutic agents. Apoptosis induction, mitochondrial function, oxidative stress, and key signaling pathways were analyzed. Tumor growth, p-STAT3 levels, and overall survival were evaluated in xenograft models. RESULTS Ruxolitinib induced dose-dependent mitochondrial-mediated apoptosis in resistant cells and enhanced cytotoxicity in combination with chemotherapy in sensitive cells. The treatment inhibited phosphorylation of STAT3, Akt, and mTOR. Additionally, ruxolitinib reduced basal and maximal respiration rates while increasing ROS levels, suggesting mitochondrial dysfunction and oxidative stress. Resistant cells exhibited increased mitochondrial DNA content, elevated respiration rates, and higher ROS levels compared to sensitive cells, indicating alterations in mitochondrial biogenesis and redox homeostasis. These findings were supported by changes in gene expression related to mitochondrial function. In vivo, ruxolitinib significantly inhibited tumor growth and reduced p-STAT3 levels in resistant gastric cancer xenografts without causing significant weight loss. Furthermore, ruxolitinib treatment significantly prolonged overall survival in mice. CONCLUSION Ruxolitinib demonstrates potential in overcoming chemoresistance in gastric cancer by targeting mitochondrial function, oxidative stress, and key survival pathways. These findings support further investigation into its clinical application as an adjunct therapy for chemoresistant gastric cancer.
Collapse
Affiliation(s)
- Yang Yao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Jun Zhou
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Jing Song
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Cheng Chen
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, People's Republic of China
| |
Collapse
|
2
|
Yamahata Y, Gocho T, Furukawa K, Haruki K, Onda S, Shirai Y, Tsunematsu M, Taniai T, Yanagaki M, Matsumoto M, Hamura R, Okui N, Tanji Y, Ikegami T. Prognostic impact of abdominal aortic calcification in patients who underwent hepatectomy for intrahepatic cholangiocarcinoma. Surg Today 2025; 55:544-551. [PMID: 39164424 DOI: 10.1007/s00595-024-02922-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: 07/08/2024] [Accepted: 08/06/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE Abdominal aortic calcification (AAC), an indicator of systemic arteriosclerosis, is associated with short- and long-term outcomes in malignancies. We investigated the prognostic impact of AAC in patients who underwent hepatectomy for intrahepatic cholangiocarcinoma (IHCC). METHODS The study cohort comprised 46 patients who underwent hepatectomy for IHCC between January 2008 and September 2020. The AAC volume measured by preoperative computed tomography was used to construct a model of the calcified segment from the renal artery to the common iliac artery bifurcation. We investigated the relationship between AAC and the long-term outcomes. The AAC volume cutoff value was calculated from a receiver-operating characteristic curve based on the three-year survival. RESULTS According to our cutoff AAC volume of 3,700 mm3, 11 patients (24%) had high AAC volumes. The high-AAC group was significantly older than the low-AAC group (73 vs. 62 years old, p < 0.01). A multivariate analysis of the cancer-specific survival showed that a high serum carbohydrate antigen 19-9 concentration (hazard ratio [HR] 5.57, p = 0.01), high AAC volume (HR 3.03, p = 0.04), and [high?] T3 or T4 levels (HR 9.05, p < 0.01) were independently associated with a poor prognosis. CONCLUSION AAC is a useful predictor of the oncological prognosis in patients undergoing hepatectomy for IHCC.
Collapse
Affiliation(s)
- Yuto Yamahata
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takeshi Gocho
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kenei Furukawa
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan.
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shinji Onda
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yoshihiro Shirai
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Masashi Tsunematsu
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Tomohiko Taniai
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Mitsuru Yanagaki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Michinori Matsumoto
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Ryoga Hamura
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Norimitsu Okui
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Yoshiaki Tanji
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-ku, Tokyo, 105-8461, Japan
| |
Collapse
|
3
|
Souza PFN, Zelaya EAE, da Silva EL, Brasil-Oliveira LL, de Oliveira FL, de Moraes MEA, Montenegro RC, Mesquita FP. PepGAT, a chitinase-derived peptide, alters the proteomic profile of colorectal cancer cells and perturbs pathways involved in cancer survival. Int J Biol Macromol 2025; 299:140204. [PMID: 39848367 DOI: 10.1016/j.ijbiomac.2025.140204] [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: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Colorectal cancer (CRC) affects the population worldwide, occupying the first place in terms of death and incidence. Synthetic peptides (SPs) emerged as alternative molecules due to their activity and low toxicity. Proteomic analysis of PepGAT-treated HCT-116 cells revealed a decreased abundance of proteins involved in ROS metabolism and energetic metabolisms, cell cycle, DNA repair, migration, invasion, cancer aggressiveness, and proteins involved in resistance to 5-FU. PepGAT induced earlier ROS and apoptosis in HCT-116 cells, cell cycle arrest, and inhibited HCT-116 migration. PepGAT enhances the action of 5-FU against HCT-116 cells by dropping down 6-fold the 5-FU toward HCT-116 and reduces its toxicity for non-cancerous cells. These findings strongly suggest the multiple mechanisms of action displayed by PepGAT against CRC cells and its potential to either be studied alone or in combination with 5-FU to develop new studies against CRC and might develop new drugs against it.
Collapse
Affiliation(s)
- Pedro Filho Noronha Souza
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Cearense Foundation to Support Scientific and Technological Development, Brazil.
| | - Elmer Adilson Espino Zelaya
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Emerson Lucena da Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Laís Lacerda Brasil-Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Francisco Laio de Oliveira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Maria Elisabete Amaral de Moraes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Felipe Pantoja Mesquita
- Laboratory of Bioinformatics Applied to Health, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil; Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil.
| |
Collapse
|
4
|
Reiter RJ, Sharma R, Bai Y, Chuffa LGDA, Loh D, Fan L, Cardinali DP. Function of intramitochondrial melatonin and its association with Warburg metabolism. Cell Signal 2025; 131:111754. [PMID: 40122433 DOI: 10.1016/j.cellsig.2025.111754] [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: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Warburg metabolism (aerobic glycolysis) is accompanied by high mitochondrial reactive oxygen species (ROS) generation from the electron transport chain; this is a "Hallmark of Cancer". The elevated ROS sustain the growth and proliferation of the cancer cells. Melatonin is a potent and functionally diverse free radical scavenger and antioxidant that is synthesized in the mitochondria of non-pathological cells and normally aids in keeping mitochondrial ROS levels low and in maintaining redox homeostasis. Because the glucose metabolite, pyruvate, does not enter mitochondria of Warburg metabolizing cells due to the inhibition of pyruvate dehydrogenase complex (PDH), acetyl coenzyme A production is diminished. Acetyl coenzyme A is a necessary co-substrate with serotonin for melatonin synthesis; thus, intramitochondrial melatonin levels become reduced in cancer cells. The hypothesis is that the depressed melatonin levels initiate aerobic glycolysis and allow the exaggerated ROS concentrations to go uncontested; the authors speculate that the elevated mtROS upregulates hypoxia inducible factor 1α (HIF-1α)/pyruvate dehydrogenase kinase (PDK) axis which inhibits PDH, thereby supporting cancer cell proliferation and stimulating cancer biomass. Exposing Warburg metabolizing cancer cells to melatonin elevates intramitochondrial melatonin, thereby reducing mtROS and concurrently interrupting aerobic glycolysis and inhibiting tumor cell proliferation. Mechanistically, higher mitochondrial melatonin levels by supplementation directly upregulates the sirtuin 3 (SIRT3)/FOXO/PDH axis, allowing pyruvate entry into mitochondria and enhancing intrinsic mitochondrial melatonin production as in non-pathological cells. Additionally, melatonin inhibits HIF1α, thereby decreasing PDK activity and disinhibiting PDH, so pyruvate enters mitochondria and is metabolized to acetyl coenzyme A, resulting in reversal of Warburg metabolism.
Collapse
Affiliation(s)
- Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA..
| | - Ramaswamy Sharma
- Applied Biomedical Sciences, University of the Incarnate Word, School of Osteopathic Medicine, San Antonio, TX, USA..
| | - Yidong Bai
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA..
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, UNESP - Saõ Paulo State University, Institute of Biosciences, Botucatu 18618-689, Sao Paulo, Brazil..
| | - Doris Loh
- Independent Researcher, Marble Falls, TX 78654, USA..
| | - Lihong Fan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Institute of Energy Metabolism and Health, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Daniel P Cardinali
- CENECON, Faculty of Medical Sciences, Universidad de Buenos Aires, and, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Akasha R, Enrera JA, Fatima SB, Hegazy AM, Hussein W, Nawaz M, Alshammari MD, Almuntashiri S, Albadari N, Break MKB, Syed RU. Oxidative phosphorylation and breast cancer progression: insights into PGC-1α's role in mitochondrial function. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04018-w. [PMID: 40095051 DOI: 10.1007/s00210-025-04018-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: 12/17/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Breast cancer still ranks high as a leading cause of mortality in women due to its complex relationship with metabolic reprogramming and tumor progression. The peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), a key transcriptional coactivator regulating mitochondrial biogenesis and oxidative phosphorylation (OXPHOS), plays a dual role in breast cancer metabolism. On the one hand, PGC-1α enhances mitochondrial function and energy production, facilitating tumor survival and metastasis, particularly in hypoxic environments. On the other hand, its suppression can limit tumor aggressiveness and energy metabolism. This dual functionality underscores its context-dependent role in cancer progression, where its activation or inhibition varies across tumor subtypes and microenvironmental conditions. The purpose of this review is to provide a comprehensive understanding of PGC-1α's dual roles in breast cancer, elucidating its regulation of mitochondrial function, its contribution to tumor progression, and the therapeutic implications of targeting this key metabolic regulator.
Collapse
Affiliation(s)
- Rihab Akasha
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Hail, Saudi Arabia
| | - Jerlyn Apatan Enrera
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Hail, Saudi Arabia
| | - Syeda Bushra Fatima
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Hail, Kingdom of Saudi Arabia
| | - A M Hegazy
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, 2440, Hail, Saudi Arabia
| | - Weiam Hussein
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Muhammad Nawaz
- Department of Nano-Medicine Research, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, 31441, Dammam, Saudi Arabia
| | - Maali D Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Sultan Almuntashiri
- Department of Clinical Pharmacy, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Najah Albadari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohammed Khaled Bin Break
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha'il, Ha'il, Saudi Arabia.
- Medical and Diagnostic Research Centre, University of Ha'il, 55473, Ha'il, Saudi Arabia.
| | - Rahamat Unissa Syed
- Department of Pharmaceutics, College of Pharmacy, University of Ha'il, 81442, Hail, Saudi Arabia.
| |
Collapse
|
6
|
Zhou J, Shu QJ, Wang T, Huang HD, Zhang SP, Zhang J, Zheng YQ, Zhang C. Piperlongumine induces ROS accumulation to reverse resistance of 5-FU in human colorectal cancer via targeting TrxR. Eur J Pharmacol 2025; 997:177478. [PMID: 40054719 DOI: 10.1016/j.ejphar.2025.177478] [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: 07/26/2024] [Revised: 03/05/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Resistance is a major concern for colorectal cancer patients undergoing chemotherapy. Piperlongumine (PL) has been proven to effectively reverse drug resistance in several types of cancers; however, the mechanisms associated with the reversal effect and the targets of PL in cancer drug resistance are still unclear. In this research, the reversal effects and associated mechanisms of PL in 5-Fluorouracil (5-FU) resistance colorectal cancer were investigated both in vitro and in vivo. Our data revealed that PL acted as a ROS inducer via binding and inhibiting TrxR (IC50 around 10.17 μM). By inducing ROS accumulation, PL reversed resistance to 5-FU in HCT-8/5-FU cells (reversal ratio: 4.9-fold) and enhanced the therapeutic effects of 5-FU through the dephosphorylation of Akt in BALB/c athymic nude mice bearing HCT-8/5-FU tumor xenografts. As a ROS inducer, PL reversed resistance to 5-FU by directly promoting inhibition of Akt phosphorylation, and further inhibited 5-FU efflux and promoted cell apoptosis through affecting the Akt/Foxo3/NRF2/P-gp and Akt/Foxo3/NRF2/BAD signaling pathway.
Collapse
Affiliation(s)
- Ji Zhou
- Center for Reproductive Medicine, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Qing-Ju Shu
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Tian Wang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Hui-Dan Huang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Sheng-Peng Zhang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China
| | - Jing Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, PR China.
| | - Yong-Qiu Zheng
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China.
| | - Chao Zhang
- Provincial Engineering Laboratory for Screening and Re-evaluation of Active Compounds of Herbal Medicines in Southern Anhui, School of Pharmacy, Wannan Medical College, Wuhu, Anhui, 241000, PR China.
| |
Collapse
|
7
|
Zhu J, Jiang C, Wang F, Tao MY, Wang HX, Sun Y, Hui HX. NOX4 Suppresses Ferroptosis Through Regulation of the Pentose Phosphate Pathway in Colorectal Cancer. Curr Med Sci 2025:10.1007/s11596-025-00013-7. [PMID: 40029499 DOI: 10.1007/s11596-025-00013-7] [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] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025]
Abstract
OBJECTIVE Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are known as major sources of reactive oxygen species (ROS), yet their role in regulating cellular antioxidative metabolism and ferroptosis is unclear. This study assessed the expression and clinical relevance of NOXs across pan-cancer and investigated the role of NOX4 in colorectal cancer progression METHODS: We analyzed transcriptomic and survival data from The Cancer Genome Atlas (TCGA) for NOXs across 22 types of solid tumors. A CRISPR library targeting NOXs was developed for potential therapeutic target screening in colorectal cancer cells (CRCs). Techniques such as CRISPR-knockout cell lines, 1,2-13C-glucose tracing, PI staining, BrdU assays, and coimmunoprecipitation were employed to elucidate the function of NOX4 in CRCs. RESULTS NOX4 emerged as a key therapeutic target for colorectal cancer from TCGA data. CRISPR screening highlighted its essential role in CRC survival, with functional experiments confirming that NOX4 upregulation promotes cell survival and proliferation. The interaction of NOX4 with glucose‑6‑phosphate dehydrogenase (G6PD) was found to enhance the pentose phosphate pathway (PPP), facilitating ROS clearance and protecting CRCs against ferroptosis. CONCLUSIONS This study identified NOX4 as a novel ferroptosis suppressor and a therapeutic target for the treatment of colorectal cancer. The findings suggest that a coupling between NADPH oxidase enzyme NOX4 and the PPP regulates ferroptosis and reveal an accompanying metabolic vulnerability for therapeutic targeting in colorectal cancer.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Chao Jiang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Fan Wang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Ming-Yue Tao
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hai-Xiao Wang
- Department of General Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yuan Sun
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hong-Xia Hui
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
8
|
Consoli V, Sorrenti V, Gulisano M, Spampinato M, Vanella L. Navigating heme pathways: the breach of heme oxygenase and hemin in breast cancer. Mol Cell Biochem 2025; 480:1495-1518. [PMID: 39287890 PMCID: PMC11842487 DOI: 10.1007/s11010-024-05119-5] [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: 08/06/2024] [Accepted: 09/07/2024] [Indexed: 09/19/2024]
Abstract
Breast cancer remains a significant global health challenge, with diverse subtypes and complex molecular mechanisms underlying its development and progression. This review comprehensively examines recent advances in breast cancer research, with a focus on classification, molecular pathways, and the role of heme oxygenases (HO), heme metabolism implications, and therapeutic innovations. The classification of breast cancer subtypes based on molecular profiling has significantly improved diagnosis and treatment strategies, allowing for tailored approaches to patient care. Molecular studies have elucidated key signaling pathways and biomarkers implicated in breast cancer pathogenesis, shedding light on potential targets for therapeutic intervention. Notably, emerging evidence suggests a critical role for heme oxygenases, particularly HO-1, in breast cancer progression and therapeutic resistance, highlighting the importance of understanding heme metabolism in cancer biology. Furthermore, this review highlights recent advances in breast cancer therapy, including targeted therapies, immunotherapy, and novel drug delivery systems. Understanding the complex interplay between breast cancer subtypes, molecular pathways, and innovative therapeutic approaches is essential for improving patient outcomes and developing more effective treatment strategies in the fight against breast cancer.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy
| | - Maria Gulisano
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Mariarita Spampinato
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, 95125, Catania, Italy.
- CERNUT - Research Centre on Nutraceuticals and Health Products, University of Catania, 95125, Catania, Italy.
| |
Collapse
|
9
|
Obalola AA, Abrahamse H, Dhilip Kumar SS. Enhanced therapeutic precision using dual drug-loaded nanomaterials for targeted cancer photodynamic therapy. Biomed Pharmacother 2025; 184:117909. [PMID: 39938348 DOI: 10.1016/j.biopha.2025.117909] [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: 10/22/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025] Open
Abstract
Combination therapy has expanded significantly, including dual drug-loaded nanomaterials in drug delivery systems. Cancer therapy can be developed by targeting cancer cells and lessening the adverse consequences of anticancer drugs, which are just two of the numerous intriguing possibilities in this research field. Dual-drug delivery nanosystems that utilize nanotechnology to combine dual-drug administration may overcome the limitations of free drugs, the properties of nanomaterials, and the combined action of two drugs work together to overcome several drug-resistant systems within cancerous cells. It is essential to design dual-drug delivery nanosystems that use various multidrug-resistant techniques to overcome drug resistance mechanisms and enhance the effectiveness of clinical antitumor therapy. In this study, we discuss the use of photosensitizers in cancer photodynamic therapy, nanomaterials with dual-drug loading for targeted drug delivery, and the function and impact of nanomaterials in cancer photodynamic therapy. Furthermore, an overview of the drug-loaded nanomaterials in vitro and in vivo activity for cancer photodynamic treatment is discussed. The commercial and clinical applications of photosensitizer-loaded nanoparticles in cancer photodynamic therapy are also briefly discussed in the study. A key finding of the study is the importance of nanomaterials and dual drugs as effective drug delivery systems in cancer treatment.
Collapse
Affiliation(s)
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | | |
Collapse
|
10
|
Farooq A, Jia G, Rizwan M, Imran M, Wei D. Can WGX-50 be a potential therapy to treat tumor by inhibiting mitochondrial reactive oxidative species? Med Hypotheses 2025; 196:111583. [DOI: 10.1016/j.mehy.2025.111583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
|
11
|
Enoch K, Somasundaram AA. Unravelling the rheological and multifunctionality of Justicia adhatoda-impregnated carboxymethyl cellulose hydrogels for drug delivery systems. Int J Biol Macromol 2025; 306:141419. [PMID: 40024406 DOI: 10.1016/j.ijbiomac.2025.141419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/06/2025] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Herbal medicine harnesses the therapeutic properties of natural compounds, providing a sustainable approach to healthcare. Due to its limited aqueous solubility, therapeutic potential remains largely unexplored. The enhanced therapeutic potential of the herbal drug entails meticulous formulation and an effective drug delivery matrix. In this study, we formulated a hydrogel system based on Carboxymethylcellulose to deliver the herbal drug, J. adhatoda. The phytochemicals in the J. adhatoda extract involved in the crosslinking of the hydrogel via hydrogen bonding resulted from the interaction with the hydroxyl and carboxyl group of the polymeric chains, confirmed by the FT-IR studies. Rheological studies demonstrated improved mechanical properties with increasing extract concentration (20 μg to 100 μg), with yield stress increasing from 58.83 Pa to 121.5 Pa. The hydrogels exhibited significant antimicrobial activity, reducing biofilm formation against S. aureus, E. faecalis, E. coli, and K. pneumoniae by 71 %, 68 %, 61 %, and 57 %. Antioxidant activity improved with extract concentration, increasing from 29 % to 74 %. The optimal anticancer activity of the hydrogels showed IC50 values of 37.4 μL for MCF-7 and 65 μL for A431 cell lines. These findings demonstrate that J. adhatoda-impregnated CMC hydrogels offer enhanced bioactivity and mechanical strength, positioning them as promising candidates for biomedical applications.
Collapse
Affiliation(s)
- Karolinekersin Enoch
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Anbumozhi Angayarkanni Somasundaram
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India.
| |
Collapse
|
12
|
Huang H, Xu Y, Guo Z, Zhang M, Li W, Song Y, Nie J, Hu W, Hei TK, Zhou G. Irradiation-responsive PRDM10-DT modulates the angiogenic response in human NSCLC cells in an SP1-dependent manner via the miR-663a/TGF-β1 axis. J Transl Med 2025; 23:235. [PMID: 40016776 PMCID: PMC11866594 DOI: 10.1186/s12967-025-06273-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Photon radiation has been shown to stimulate the secretion of radioresistant factors from tumor cells, ultimately promoting tumor angiogenesis and metastasis. On the other hand, heavy-ion radiotherapy has been demonstrated to control tumor angiogenesis and metastasis levels. The molecular mechanisms responsible for the different angiogenic responses to photon and heavy-ion irradiation are not fully understood. This study aims to explore the irradiation-responsive genes related to tumor angiogenesis and reveal the regulatory effect. METHODS In order to clarify the potential regulatory mechanisms of tumor angiogenesis after X-ray or carbon ion (C-ion) irradiation, we performed RNA-sequencing (RNA-seq), as well as bioinformatics, public database analysis, Western blotting, immunohistochemistry, and immunofluorescence. RESULTS In this study, we identified the long intergenic noncoding RNA PRDM10 divergent transcript (PRDM10-DT), which was responsive to X-rays but not carbon ions. Mechanistically, PRDM10-DT triggers tumor angiogenesis by upregulating the TGF-β1/VEGF signaling pathway through its competitive binding to miR-663a. Additionally, the transcription factor SP1 facilitated the transcription of PRDM10-DT by binding to its promoter region. It's notable that the DNA-binding activity of SP1 was enhanced by reactive oxygen species (ROS). The knockdown of either PRDM10-DT or SP1 effectively inhibited NSCLC angiogenesis and metastasis. CONCLUSION These results illustrate the proangiogenic function of the PRDM10-DT/miR-663a/TGF-β1 axis and reveal the regulatory role of ROS and SP1 in the upstream response to radiation, with differential ROS production mediating the differential angiogenesis levels after X-ray and C-ion irradiation. Our findings suggest the potential of PRDM10-DT as a nucleic acid biomarker after radiotherapy and that targeting this gene could be a therapeutic strategy to counteract angiogenesis in NSCLC radiotherapy.
Collapse
MESH Headings
- Humans
- Sp1 Transcription Factor/metabolism
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Transforming Growth Factor beta1/metabolism
- Carcinoma, Non-Small-Cell Lung/radiotherapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/blood supply
- Lung Neoplasms/radiotherapy
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/blood supply
- Neovascularization, Pathologic
- Cell Line, Tumor
- Transcription Factors/metabolism
- Signal Transduction
- Animals
- Gene Expression Regulation, Neoplastic/radiation effects
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Mice, Nude
- Base Sequence
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Zi Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Miaomiao Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Wanshi Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Yidan Song
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Jing Nie
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| | - Tom K Hei
- Center for Radiological Research, College of Physician and Surgeons, Columbia University, New York, NY, 10032, USA.
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
13
|
Chen L, Dai C, Peng S, Dong H, Wang X, Liu Y, Wang J. Preoperative integrated oxidative stress score as a prognostic factor for predicting clinical outcomes in breast cancer patients received neoadjuvant chemotherapy: a real-world retrospective study. Int J Med Sci 2025; 22:1437-1449. [PMID: 40084249 PMCID: PMC11898859 DOI: 10.7150/ijms.109915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
Objective: The current study aims to investigate the prognostic value of breast cancer integrated oxidative stress score (BCIOSS) in patients with breast cancer who received neoadjuvant chemotherapy (NACT). Methods: A retrospective analysis of 104 breast cancer patients who underwent NACT from June 2009 to December 2015 was performed. The differences of BCIOSS of breast cancers in regard to variables were analyzed using Chi-square test and Fisher's exact test. The Kaplan-Meier method was used to evaluate survival curve between low BCIOSS group and high BCIOSS group, and the two groups were compared by Log-rank tests at the individual index level. The univariate and multivariate Cox regression analyses were established by the important predictive factors determined based on univariate analysis. The nomograms were further conducted based on the factors by the multivariate analyses. Results: Patients were assigned to low BCIOSS group (BCIOSS≤2.54) or high BCIOSS group (BCIOSS>2.54) via ROC curve. BCIOSS was a latent prognostic factor for patient survival [DFS; hazard ratio (HR): 0.163, 95%CI: 0.045-0.596, P=0.006; and OS; HR: 0.168, 95%CI: 0.056-0.500, P=0.001]. Patients with a high BCIOSS had longer survival time than those with a low BCIOSS (DFS: χ2=7.317, P=0.0068; and OS: χ2=9.407, P=0.0022). Calibration curves shown that the predicted line conformed well to the reference line for the 5-year survival category. DCA revealed that the nomograms conducted had a better clinical predictive application than only by BCIOSS. Conclusion: BCIOSS is a latent prognostic factor, and patients with high oxidative stress scores have a better prognosis and longer survival time.
Collapse
Affiliation(s)
- Li Chen
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Caixia Dai
- Department of Hepatobiliary&Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, P.R. China
| | - Shu Peng
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Hao Dong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| | - Yanfei Liu
- Department of Oncology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, Hubei 430081, P.R. China
| | - Jing Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P.R. China
| |
Collapse
|
14
|
Naderzadeh E, Kargar M, Mokhtari MJ, Farhadi A. Activating transcription factor 3 induces oxidative stress and genotoxicity, transcriptionally modulating metastasis-related gene expression in human papillomavirus-infected cervical cancer. Virol J 2025; 22:46. [PMID: 39994644 PMCID: PMC11849226 DOI: 10.1186/s12985-025-02675-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Activating Transcription Factor 3 (ATF3) is known for its tumor-suppressive properties in cervical cancer, particularly through its role in stress response and interactions with human papillomavirus (HPV) oncogenes. This study investigates ATF3's regulatory impact on metastasis-related genes, oxidative stress, and DNA damage in HPV-positive cervical cancer cells. METHODS HeLa and Ca Ski cell lines were transfected with ATF3-expressing vectors. Western blotting and quantitative reverse transcription polymerase chain reaction (RT-qPCR) were used to confirm ATF3 overexpression following transfection. ROS assays and Comet assays assessed the impact of ATF3 on oxidative stress and DNA damage, while RT-qPCR was used to evaluate changes in HPV E6/E7, SHARP1, and MMP1 gene expression. RESULTS ATF3 overexpression led to elevated ROS levels (p < 0.02), resulting in oxidative DNA damage. These results demonstrate ATF3's cytotoxic impact on cervical cancer cells through oxidative stress and DNA damage. Additionally, ATF3 overexpression significantly decreased MMP1 expression (p < 0.03), indicating a potential anti-metastatic effect, while SHARP1 and HPV E6/E7 expression levels were not significantly altered, indicating selective gene modulation by ATF3. CONCLUSION These findings reveal that ATF3 contributes to tumor suppression in cervical cancer by modulating oxidative stress and DNA damage, selectively targeting genes involved in metastasis. These findings supports ATF3's role in regulating key pathways in HPV-positive cervical cancer cells, providing a basis for further exploration of ATF3 as a target in therapeutic strategies aimed at improving outcomes in cervical cancer.
Collapse
Affiliation(s)
- Elham Naderzadeh
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Mohammad Kargar
- Department of Biology, Zand Institute of Higher Education, Shiraz, Iran
| | | | - Ali Farhadi
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, 7143918596, Iran.
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
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] [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
|
16
|
Liu Z, Lai K, Li P, Gong Y, Fu H, Dong H, Yang Z, Qin R, Guo L. Enhanced Anticancer Selectivity of Cyclometalated Imidazole/Pyrazole-Imine Iridium III Complexes Through the Switch from Cationic to Zwitterionic Forms. Inorg Chem 2025; 64:2837-2856. [PMID: 39895267 DOI: 10.1021/acs.inorgchem.4c04937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cyclometalated iridiumIII complexes have shown promising anticancer properties, with variations in charge and ligand substitution significantly influencing their biological activity. However, zwitterionic iridiumIII complexes remain scarcely explored. Herein, we report a series of zwitterionic cyclometalated imidazole/pyrazole-imine iridiumIII complexes and compare their biological activity to analogous cationic complexes with sulfonate counteranions. X-ray crystallography confirmed the structural differences between the cationic and zwitterionic forms. These complexes exhibited cytotoxicity against A549, HeLa, and HepG2 cancer cells, with IC50 values ranging from 14.35 to 69.12 μM. While cationic complexes showed higher cytotoxicity, zwitterionic complexes demonstrated enhanced selectivity for A549 cancer cells over BEAS-2B normal cells (selectivity index: 3.72-5.90 for zwitterionic forms vs 1.16-1.44 for cationic forms). This selectivity is attributed to distinct cellular uptake mechanisms: zwitterionic complexes use an energy-dependent pathway in cancer cells and an energy-independent pathway in normal cells, leading to differences in cellular accumulation and redox activity. Mechanistic studies revealed that both complex types induce ROS generation and mitochondrial membrane depolarization (MMP), with apoptosis as the primary cell death pathway.
Collapse
Affiliation(s)
- Zhe Liu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Kangning Lai
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Pengwei Li
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Yuwen Gong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Hanxiu Fu
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Heqian Dong
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Zhihao Yang
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Ruixin Qin
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| | - Lihua Guo
- Key Laboratory of Life-Organic Analysis of Shandong Province, Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu 273165, P. R. China
| |
Collapse
|
17
|
Torres J, Valenzuela Oses JK, Rabasco-Álvarez AM, González-Rodríguez ML, García MC. Innovations in Cancer Therapy: Endogenous Stimuli-Responsive Liposomes as Advanced Nanocarriers. Pharmaceutics 2025; 17:245. [PMID: 40006612 PMCID: PMC11858840 DOI: 10.3390/pharmaceutics17020245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/03/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
Recent advancements in nanotechnology have revolutionized cancer therapy-one of the most pressing global health challenges and a leading cause of death-through the development of liposomes (L), lipid-based nanovesicles known for their biocompatibility and ability to encapsulate both hydrophilic and lipophilic drugs. More recent innovations have led to the creation of stimuli-responsive L that release their payloads in response to specific endogenous or exogenous triggers. Dual- and multi-responsive L, which react to multiple stimuli, offer even greater precision, improving therapeutic outcomes while reducing systemic toxicity. Additionally, these smart L can adjust their physicochemical properties and morphology to enable site-specific targeting and controlled drug release, enhancing treatment efficacy while minimizing adverse effects. This review explores the latest advancements in endogenous stimuli-responsive liposomal nanocarriers, as well as dual- and multi-responsive L that integrate internal and external triggers, with a focus on their design strategies, mechanisms, and applications in cancer therapy.
Collapse
Affiliation(s)
- Jazmín Torres
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Johanna Karina Valenzuela Oses
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| | - Antonio María Rabasco-Álvarez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González 2, 41012 Seville, Spain;
| | - Mónica Cristina García
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre and Medina Allende, Ciudad Universitaria, Science Building 2, Córdoba X5000HUA, Argentina; (J.T.); (J.K.V.O.)
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica, UNITEFA, Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Córdoba X5000HUA, Argentina
| |
Collapse
|
18
|
Liu Z, He Y, Wang Y, Ren K, Xia P, Xie B, Wei T. Oxidative stress caused by 3-monochloro-1,2-propanediol provokes intestinal stem cell hyperproliferation and the protective role of quercetin. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 291:117851. [PMID: 39914075 DOI: 10.1016/j.ecoenv.2025.117851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
Recently, the contaminant 3-monochloropropane-1,2-diol (3-MCPD) found in food and the environment has garnered significant global attention due to its detrimental health effects on animals, including reproductive toxicity, neurotoxicity, and nephrotoxicity. However, the specific impacts and mechanisms of 3-MCPD on intestinal health remain elusive. Here, we employed the adult intestine of Drosophila melanogaster, a notable invertebrate model organism, to investigate the intestinal toxicity of 3-MCPD and its underlying mechanisms. Our findings revealed that exposure to 3-MCPD led to a decrease in the number of enterocyte cells and an elevation in apoptosis levels, ultimately disrupting the intestinal epithelial barrier and its function. This disruption subsequently triggered hyperproliferation and differentiation of intestinal stem cells (ISCs). Mechanistically, 3-MCPD induced oxidative stress in the Drosophila intestine, which was likely responsible for ISC hyperproliferation and intestinal damage. Intriguingly, quercetin, a natural antioxidant derived from dietary fruits and vegetables, alleviated 3-MCPD-induced intestinal toxicity by inhibiting the JNK pathway. Our findings uncover a mechanism whereby suppression of undesirable ISC hyperproliferation, caused by 3-MCPD-induced oxidative stress, maintains intestinal homeostasis, and provide a theoretical basis for exploiting quercetin, a natural antioxidant, as a dietary antidote against the intestinal hazards posed by environmental toxicants.
Collapse
Affiliation(s)
- Zongzhong Liu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yanfei He
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Yuhan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Kefeng Ren
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Pengpeng Xia
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Binbin Xie
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Tian Wei
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| |
Collapse
|
19
|
Wu JJ, Zheng X, Wu C, Ma W, Wang Y, Wang J, Wei Y, Zeng X, Zhang S, Guan W, Chen F. Melatonin alleviates high temperature exposure induced fetal growth restriction via the gut-placenta-fetus axis in pregnant mice. J Adv Res 2025; 68:131-146. [PMID: 38382594 PMCID: PMC11785557 DOI: 10.1016/j.jare.2024.02.014] [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: 12/16/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION Global warming augments the risk of adverse pregnancy outcomes in vulnerable expectant mothers. Pioneering investigations into heat stress (HS) have predominantly centered on its direct impact on reproductive functions, while the potential roles of gut microbiota, despite its significant influence on distant tissues, remain largely unexplored. Our understanding of deleterious mechanisms of HS and the development of effective intervention strategies to mitigate the detrimental impacts are still limited. OBJECTIVES In this study, we aimed to explore the mechanisms by which melatonin targets gut microbes to alleviate HS-induced reproductive impairment. METHODS We firstly evaluated the alleviating effects of melatonin supplementation on HS-induced reproductive disorder in pregnant mice. Microbial elimination and fecal microbiota transplantation (FMT) experiments were then conducted to confirm the efficacy of melatonin through regulating gut microbiota. Finally, a lipopolysaccharide (LPS)-challenged experiment was performed to verify the mechanism by which melatonin alleviates HS-induced reproductive impairment. RESULTS Melatonin supplementation reinstated gut microbiota in heat stressed pregnant mice, reducing LPS-producing bacteria (Aliivibrio) and increasing beneficial butyrate-producing microflora (Butyricimonas). This restoration corresponded to decreased LPS along the maternal gut-placenta-fetus axis, accompanied by enhanced intestinal and placental barrier integrity, safeguarding fetuses from oxidative stress and inflammation, and ultimately improving fetal weight. Further pseudo-sterile and fecal microbiota transplantation trials confirmed that the protective effect of melatonin on fetal intrauterine growth under HS was partially dependent on gut microbiota. In LPS-challenged pregnant mice, melatonin administration mitigated placental barrier injury and abnormal angiogenesis via the inactivation of the TLR4/MAPK/VEGF signaling pathway, ultimately leading to enhanced nutrient transportation in the placenta and thereby improving the fetal weight. CONCLUSION Melatonin alleviates HS-induced low fetal weight during pregnancy via the gut-placenta-fetus axis, the first time highlighting the gut microbiota as a novel intervention target to mitigate the detrimental impact of global temperature rise on vulnerable populations.
Collapse
Affiliation(s)
- Jia-Jin Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Henry Fok School of Biology and Agriculture, Shaoguan University, Shaoguan 512005, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Caichi Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wen Ma
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yibo Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jun Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yulong Wei
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture Feed Industry Center, China Agricultural University, Beijing 100193, PR China
| | - Shihai Zhang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
| | - Fang Chen
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China; National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
20
|
Peng BY, Wu CY, Lee CJ, Chang TM, Tsao YT, Liu JF. Nimbolide Induces Cell Apoptosis via Mediating ER Stress-Regulated Apoptotic Signaling in Human Oral Squamous Cell Carcinoma. ENVIRONMENTAL TOXICOLOGY 2025; 40:347-356. [PMID: 39462890 DOI: 10.1002/tox.24436] [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: 03/07/2024] [Revised: 07/29/2024] [Accepted: 10/17/2024] [Indexed: 10/29/2024]
Abstract
Human oral squamous cell carcinoma (OSCC) poses a significant health challenge in Asia, with current therapeutic strategies failing to improve the survival rates for OSCC patients sufficiently. To elucidate the effects of Nimbolide on OSCC cell proliferation and apoptosis, we performed a series of experiments, including cell proliferation assays, annexin V/PI assays, and cell cycle analysis. We further investigated nimbolide's role in modulating endoplasmic reticulum (ER) stress, reactive oxygen species (ROS) production, and mitochondrial dysfunction using flow cytometry. Additionally, Western blotting was used to detect apoptosis-related protein expression. Our findings reveal that nimbolide exerts its anti-proliferative effects on OSCC cells by inducing apoptosis. The nimbolide increased intracellular ROS levels and acceleration of cellular calcium accumulation, respectively promoting endoplasmic reticulum stress and cancer cell apoptosis. Furthermore, nimbolide activates the caspase cascade by altering the mitochondrial membrane potential and apoptotic protein expression, thereby inhibiting the viability of tumor cells. Our data show that Nimbolide suppresses tumor growth through the induction of ROS production, ER stress, and mitochondrial dysfunction, resulting in apoptosis in OSCC cells. Overall, our study highlights nimbolide as a potential natural compound for OSCC therapy.
Collapse
Affiliation(s)
- Bou-Yue Peng
- Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Yu Wu
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Taipei Medical University Hospital, Taipei, Taiwan, ROC
| | - Chia-Jung Lee
- Department of Otolaryngology Head and Neck Surgery, Shin-Kong Wu-Ho-Su Memorial Hospital, Taipei, Taiwan, ROC
- School of Medicine, Fu-Jen Catholic University, Taipei, Taiwan, ROC
| | - Tsung-Ming Chang
- School of Dental Technology, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Ya-Ting Tsao
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Ju-Fang Liu
- Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, ROC
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan, ROC
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
| |
Collapse
|
21
|
Kumar N, Samanta B, Km J, Raghunathan V, Sen P, Bhat R. Demonstration of Enhancement of Tumor Intravasation by Dicarbonyl Stress Using a Microfluidic Organ-on-chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405998. [PMID: 39745135 DOI: 10.1002/smll.202405998] [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: 07/17/2024] [Revised: 11/28/2024] [Indexed: 02/13/2025]
Abstract
Cancer metastasis involves cell migration from their primary organ foci into vascular channels, followed by dissemination to prospective colonization sites. Vascular entry of tumor cells or intravasation involves their breaching stromal and endothelial extracellular matrix (ECM) and the endothelial barriers. How the kinetics of this breach are confounded by chronic inflammatory stresses seen in diabetes and aging remains ill-investigated. To study the problem, a histopathology-motivated, imaging-tractable, microfluidic multi-organ-on-chip platform is constructed, that seamlessly integrates a breast tumor-like compartment: invasive MDA-MB-231 in a 3D Collagen I scaffold, and a flow-implemented vascular channel: immortalized human aortic endothelia (TeloHAEC) on laminin-rich basement membrane (lrBM). The chip showcases the complexity of intravasation, wherein tumor cells and endothelia cooperate to form anastomotic structures, which facilitate cancer cell migration into the vascular channel. Upon entry, cancer cells adhere to and flow within the vascular channel. Exposure to methylglyoxal (MG), a dicarbonyl stressor associated with diabetic circulatory milieu increases cancer cell intravasation and adhesion through the vascular channel. This can be driven by MG-induced endothelial senescence and shedding, but also by the ability of MG to degrade lrBM and pathologically cross-link Collagen I, diminishing cell-ECM adhesion. Thus, dicarbonyl stress attenuates homeostatic barriers to cancer intravasation, exacerbating metastasis.
Collapse
Affiliation(s)
- Nilesh Kumar
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Bidita Samanta
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Jyothsna Km
- Department of Electrical and Communications Engineering, Bengaluru, 560012, India
| | - Varun Raghunathan
- Department of Electrical and Communications Engineering, Bengaluru, 560012, India
| | - Prosenjit Sen
- Centre for Nano Science and Engineering, Indian Institute of Science, Bengaluru, 560012, India
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
| | - Ramray Bhat
- Department of Bioengineering, Indian Institute of Science, Bengaluru, 560012, India
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru, 560012, India
| |
Collapse
|
22
|
Jang JH, Kim DH, Chun KS. Tumor microenvironment regulation by reactive oxygen species-mediated inflammasome activation. Arch Pharm Res 2025; 48:115-131. [PMID: 39888519 DOI: 10.1007/s12272-025-01532-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Tumor microenvironment (TME) is composed of diverse cell types whose interactions, both direct and indirect, significantly influence tumorigenesis and therapeutic outcomes. Within TME, reactive oxygen species (ROS) are produced by various cells and exhibit a dual role: moderate ROS levels promote tumor initiation and progression, whereas excessive levels induce cancer cell death, influencing the efficacy of anticancer therapies. Inflammasomes, cytosolic multiprotein complexes, are pivotal in multiple stages of tumorigenesis and play a crucial role in establishing the inflammatory TME. By releasing cytokines such as IL-1β and IL-18, inflammasomes contribute to immune cell recruitment and sustain a chronic inflammatory state that supports tumor growth. ROS are critical regulators of inflammasome activation, with the impact of ROS-mediated activation differing across cell types, leading to distinct influences on tumor progression and therapeutic responses. This review explores how ROS drive inflammasome activation in various TME-associated cells and the reciprocal ROS generation induced by inflammasomes, examining their multifaceted impact on tumorigenesis and therapeutic efficacy. By elucidating the complex interplay between ROS and inflammasomes in TME, we provide insights into potential therapeutic approaches that could modulate cancer progression and enhance treatment outcomes.
Collapse
Affiliation(s)
- Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan-si, Gyeongbuk, 38430, Republic of Korea
| | - Do-Hee Kim
- Department of Chemistry, Kyonggi University, Suwon, 16227, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu, 42601, Republic of Korea.
| |
Collapse
|
23
|
Song B, Wu M, Qin L, Liang W, Wang X. Smart Design of Targeted Drug Delivery System for Precise Drug Delivery and Visual Treatment of Brain Gliomas. Adv Healthc Mater 2025; 14:e2402967. [PMID: 39707642 DOI: 10.1002/adhm.202402967] [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: 08/09/2024] [Revised: 12/04/2024] [Indexed: 12/23/2024]
Abstract
In the treatment of glioma, which is one of the malignant tumors, although chemotherapy is used as the most common treatment method, it often suffers from low bioavailability. Therefore, improving the precision and efficiency of drugs is crucial in treating gliomas and a great challenge. Here, an advanced drug delivery system is reported for gliomas (CZQD@HA@DOX), which aggregates multiple features such as the susceptible imaging tracer property due to the use of CZQD and the targeting of HA to the receptor cluster 44 (CD44) of glioma cells, which provides the system with the functions of targeted enrichment and precise drug delivery at the tumor site. The pH-responsive drug delivery system has not only an excellent encapsulation rate but also a high drug loading capacity, and the doxorubicin loaded on it can be released centrally at the tumor microenvironment site and causes an increase of reactive oxygen species in the mitochondria and trigger oxidative stress, which leads to high expression of Bax apoptotic proteins, ultimately activating the mitochondrial pathway-mediated apoptotic process in glioma cells. Overall, this drug delivery system has great potential for application in precision targeted therapy and visual tracer imaging of gliomas.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Mengru Wu
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Lijing Qin
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences and Institute of Materia Medica, Shandong First Medical University and Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery System, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare and Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, China
| |
Collapse
|
24
|
Zhou W, Lyu SB, Li H, Li SX, Yao WH, Shan SL, Tang H, Zhang J, Sun CH, Wen CL, Yang F, Guo J, Xu LJ, Yan Y, Yan ZQ, He QL, Cheng D. Toxic effects and safety assessment of Xanthoceras sorbifolium bunge seed kernels. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119242. [PMID: 39694427 DOI: 10.1016/j.jep.2024.119242] [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: 09/06/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xanthoceras sorbifolium Bunge (X. sorbifolia), an oil crop native to northern China, is valued for both its edible and medicinal uses. It has various applications, including the production of edible and bioactive oils, and is used in traditional medicine for its antioxidant and anti-inflammatory properties. However, the toxicity of X. sorbifolia, particularly its widely used seed kernels, remains unclear. AIM OF THE STUDY This study aimed to evaluate the acute toxicity and safety risks of X. sorbifolia seed kernels based on human-recommended doses by in vitro or in vivo experiments, and integrating network analysis. MATERIALS AND METHODS In this study, rats and mice were employed as model organisms to investigate the acute toxicity of X. sorbifolia seed kernels. The experiments included the Salmonella typhimurium reverse mutation test, red blood cell micronucleus test, spermatocyte chromosome aberration test in mice, and a 90-day exposure study in rats to assess the potential toxicity and safety risks of the seed kernels. Based on this, combined with The Comparative Toxicogenomics Database (CTD), the biological functions of the main active ingredients of X. sorbifolia were further explored through integrated network analysis, and the anti-inflammatory effect of X. sorbifolia was explored through cotton ball granuloma inflammation experiment. RESULTS During the experimental period, animals in all treatment groups demonstrated normal growth and development. Although some detection indicators showed significant differences in different treatment groups, the results were still within a reasonable range. In addition, by screening the CTD, 120 target genes with potential interactions of the main active ingredients in the kernel of X. sorbifolia were obtained for analysis, and it was found that these genes were involved in important biological processes such as response to oxidative stress, response to reactive oxygen species, and regulation of inflammatory response. The cotton ball granuloma inflammation experiment in rats also suggested that X. sorbifolia tended to inhibit the proliferation of granulomas, indicating that the kernel of X. sorbifolia has potential anti-inflammatory and antioxidant properties. CONCLUSION The findings suggested that X. sorbifolia seed kernels were safe within the recommended dosage range. As a traditional Chinese medicine prescription, it has certain anti-inflammatory and antioxidant effects. This study provides valuable reference guidelines for the clinical application of X. sorbifolia seed kernels and encourages further research into its potential uses and safety.
Collapse
Affiliation(s)
- Wen Zhou
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Shi-Bo Lyu
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Hui Li
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Shu-Xian Li
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Wen-Huan Yao
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Shu-Lin Shan
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Hui Tang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Jing Zhang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Chang-Hua Sun
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Cheng-Li Wen
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Fei Yang
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Jie Guo
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Long-Jin Xu
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Yan Yan
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China
| | - Zhi-Qiang Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China; Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
| | - Qi-Long He
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China.
| | - Dong Cheng
- Division of Toxicology, Shandong Center for Disease Control and Prevention, Jinan, 250014, China; Shandong Provincial Key Medical And Health Discipline of Sanitary Analysis (Shandong Center For Disease Control And Prevention), Jinan, 250014, China.
| |
Collapse
|
25
|
Barkat MA, Fatima A, Riaz B, Hassan MZ, Ahamad T, Alanezi AA, Barkat H, Almuqati AF, Asiri YI, Siddiqui S. Bidirectional approach of Punica granatum natural compounds: reduction in lung cancer and SARS-CoV-2 propagation. BMC Complement Med Ther 2025; 25:32. [PMID: 39885485 PMCID: PMC11781039 DOI: 10.1186/s12906-024-04738-1] [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: 11/01/2023] [Accepted: 12/19/2024] [Indexed: 02/01/2025] Open
Abstract
The spreading of COVID-19 has posed a risk to global health, especially for lung cancer patients. An investigation is needed to overcome the challenges of COVID-19 pathophysiology and lung cancer disease. This study was designed to evaluate the phytoconstituents in Punica granatum peel (PGP), its anti-lung cancer activity, and in silico evaluation for antiviral potential. GC-MS technique was used to detect the phytoconstituents. Cytotoxicity was analyzed using MTT dye, followed by apoptosis, ROS generation, and cell cycle phase detection in human lung cancer cells (A549). The glide module of Maestro software was used to investigate the molecular-docking interaction of the constituents against main protease (Mpro) and papain-like protease (PLpro) of SARS-CoV-2. GROMACS 2023.2 was utilized to evaluate the complex stability. A total of nineteen phytocomponents were detected in the PGP extract through GC-MS analysis. PGP has shown a potential to reduce lung cancer cell proliferation while evading normal cell death. PGP induced apoptosis by arresting cells in the G0/G1 phase and generating ROS. A total of six and eight phytocomponents had a high affinity for PLpro and Mpro proteins, respectively. The top docked complex, ethyl 5-oxo-2-pyrrolidinecarboxylate, with PLpro and Mpro proteins, showed likely stable interaction throughout 100 ns simulation. This finding raises the possibility of top-eight hits (docking score ≥ -1.0 kcal/mol) preventing SARS-CoV-2 severity. The phytoconstituents exhibited orally active drugs with no more than one violation and drug-likeness activity. The PGP phytoconstituents are suggested to be dual agents for lung cancer and SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, 39524, Hafr Al Batin, Saudi Arabia
| | - Afreen Fatima
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39524, Hafr Al Batin, Saudi Arabia
| | - Bushra Riaz
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39524, Hafr Al Batin, Saudi Arabia
| | - Mohd Zaheen Hassan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Asir, Saudi Arabia
| | - Tanveer Ahamad
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India
| | - Abdulkareem A Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, 39524, Hafr Al Batin, Saudi Arabia
| | - Harshita Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, 39524, Hafr Al Batin, Saudi Arabia
| | - Afaf F Almuqati
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Hafr Al-Batin, Al Jamiah, 39524, Hafr Al Batin, Saudi Arabia
| | - Yahya I Asiri
- Department of Pharmacology, College of Pharmacy, King Khalid University, Asir, Saudi Arabia
| | - Sahabjada Siddiqui
- Department of Biotechnology, Era's Lucknow Medical College and Hospital, Era University, Lucknow, India.
| |
Collapse
|
26
|
Polloni L, Costa TR, Morais LP, Borges BC, Teixeira SC, de Melo Fernandes TA, Correia LIV, Bastos LM, Soares AM, Silva MJB, Amália Vieira Ferro E, Lopes DS, Ávila VDMR. Pollonein-LAAO unveiling anti-angiogenic effects through oxidative stress: Insights from mimetic tumor angiogenesis environment in a 3D co-culture model. Chem Biol Interact 2025; 406:111361. [PMID: 39716533 DOI: 10.1016/j.cbi.2024.111361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/22/2024] [Accepted: 12/19/2024] [Indexed: 12/25/2024]
Affiliation(s)
- Lorena Polloni
- Institute of Biotechnology, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil.
| | - Tássia Rafaella Costa
- Institute of Biotechnology, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Lorena Pinheiro Morais
- Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Bruna Cristina Borges
- Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Samuel Cota Teixeira
- Institute of Biomedical Sciences, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil
| | | | | | - Luciana Machado Bastos
- Institute of Biotechnology, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil
| | - Andreimar Martins Soares
- Oswaldo Cruz Foundation (FIOCRUZ) Rondônia, Federal University of Rondônia (UNIR), National Institute of Science and Technology of Epidemiology of the Western Amazon (INCT-EPIAMO), Porto Velho-RO, Brazil; Network of Research and Knowledge of Excellence in the Western/Eastern Amazon (RED-CONEXAO), Brazil
| | | | | | - Daiana Silva Lopes
- Multidisciplinary Institute for Health, Federal University of Bahia - UFBA, Vitoria da Conquista, BA, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Institute of Biotechnology, Federal University of Uberlândia - UFU, Uberlândia, MG, Brazil; Network of Research and Knowledge of Excellence in the Western/Eastern Amazon (RED-CONEXAO), Brazil.
| |
Collapse
|
27
|
Seven D, Tecimel D, Özbey U, Kızılilsoley N, Nikerel E, Dalan AB, Türe U, Bayrak ÖF. Silencing superoxide dismutases (SOD1&SOD2) potentiates ROS-induced apoptosis in chordoma cells. Mol Biol Rep 2025; 52:157. [PMID: 39853601 DOI: 10.1007/s11033-025-10239-2] [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: 06/24/2024] [Accepted: 01/08/2025] [Indexed: 01/26/2025]
Abstract
BACKGROUND Chordoma, characterized as a slow growing yet locally invasive and destructive bone tumor mainly emerging in the sacrum and clivus, presents a unique challenge due to its rarity, hampering the development of effective treatment strategies. Comprehensive understanding of tumor biology is crucial to suggest novel treatment modalities. Reactive oxygen species (ROS), a family of chemically reactive and unstable oxygen derivatives, are controlled by an intracellular antioxidant system to maintain homeostasis. Higher doses of ROS levels have been associated with the oxidative stress-induced tumor cell death, highlighting the potential of fine-tuning ROS regulation as a target for cancer therapies. The association of ROS mechanism and chordoma remains to be elucidated. In this study, we investigated the effect of targeting the ROS mechanism in chordoma, focusing on superoxide dismutase 1 and superoxide dismutase 2. METHODS Two different chordoma datasets were used to assess oxidative stress-related genes. ROS levels and mitochondrial membrane potential (mtMP) in chordoma cells were measured. The gene expression levels of SOD1 and SOD2 in chordoma patients were also evaluated. SOD2 and SOD1 targeted siRNAs were used to silence gene expression in chordoma cells, and quantitative real-time PCR (qRT-PCR) was used to compare gene expression levels. Apoptotic cell populations were determined using flow cytometry. RESULTS The levels of ROS and mtMP were increased in chordoma cell lines compared to healthy nucleus pulposus cells. The chordoma omics data showed induced levels of SOD2. Chordoma tissues also showed high levels of the SOD2 gene. Silencing SOD2 and combined silencing of SOD2 and SOD1 expression increased ROS levels or mtMP, and both induced apoptosis in chordoma cells. ROS imbalance plays a role in chordoma pathogenesis. CONCLUSION SOD2 and SOD1 might be key enzymes in chordoma to modulate ROS levels, and inhibiting the SOD2 and SOD1 activity might be a potential therapeutic target for chordoma treatment.
Collapse
Affiliation(s)
- Didem Seven
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey
| | - Didem Tecimel
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Utku Özbey
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Nehir Kızılilsoley
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Emrah Nikerel
- Faculty of Engineering, Genetics and Bioengineering Department, Yeditepe University, İstanbul, 34755, Turkey
| | - Altay Burak Dalan
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey
| | - Uğur Türe
- Department of Neurosurgery, Yeditepe University Medical School, Yeditepe University, Istanbul, 34755, Turkey
| | - Ömer Faruk Bayrak
- Department of Medical Genetics, School of Medicine, Yeditepe University, İstanbul, 34755, Turkey.
| |
Collapse
|
28
|
Al-Najjar BO, Helal M, Saqallah FG, Bandy B. Isozyme-specific inhibition of GSTP1-1: a crucial element in cancer-targeting drugs. RSC Med Chem 2025:d4md00872c. [PMID: 39917632 PMCID: PMC11795191 DOI: 10.1039/d4md00872c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/18/2025] [Indexed: 02/09/2025] Open
Abstract
Selectively targeting cancer cells has been a main challenge in cancer therapy. The purpose is to spare normal cells and minimize side effects. Targeting the antioxidant enzymes (i.e. GST) for the purpose of selectively killing cancer cells has attracted much attention in the past few decades. The intention of lowering the antioxidant enzymes is "tipping" the ROS concentrations to levels above the cytotoxic threshold. This would result in extensive damage to the cellular macromolecules and organelles leading to cell death. Here we focused on the glutathione S-transferase pi 1 (GSTP1), because it is one of the overexpressed antioxidant enzymes in cancer and has been targeted for the purpose of killing cancer cells. However, most available GSTP1 inhibitors do not show selectivity towards the isozyme. This can potentially lead to many side effects. Therefore, the search for optimal selective GSTP1 inhibitors is still underway. The novelty of this review stems from highlighting the significance of selectively targeting GSTP1. We also addressed the structural feature of the enzyme which challenges the design of novel selective GSTP1 inhibitors. We then provide guidelines to help resolve these challenges to help design future compounds. The first objective of this review is to present a brief literature review to highlight the importance of selectively targeting GSTP1. Briefly, the lack of selectivity towards GSTP1 has resulted in extensive side effects which limited reaching advanced clinical trials. We screened publications on many potential inhibitors, including some that reached phase I and II clinical trials, for their ability to bind with GSTP1, GSTM, and GSTA. All compounds appear to bind different GST isozymes (at least to some extent). The second objective is to present differences in the structures of GST isotypes (GSTP1, GSTM, GSTA) which could allow selectively targeting a certain isotype. Our modelling results highlight the importance of certain structural moieties for better selective binding to GSTP1.
Collapse
Affiliation(s)
- Belal O Al-Najjar
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University Amman 19328 Jordan
| | - M Helal
- Physiology, Pharmacology, and Toxicology Division, Biomedical Sciences Department, Faculty of Medicine and Health Sciences, An-Najah National University Palestine
| | - Fadi G Saqallah
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan Amman 11733 Jordan
| | - B Bandy
- College of Pharmacy and Nutrition, University of Saskatchewan Saskatoon Canada
| |
Collapse
|
29
|
Horta M, Soares P, Leite Pereira C, Lima RT. Emerging Approaches in Glioblastoma Treatment: Modulating the Extracellular Matrix Through Nanotechnology. Pharmaceutics 2025; 17:142. [PMID: 40006509 PMCID: PMC11859630 DOI: 10.3390/pharmaceutics17020142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Glioblastoma's (GB) complex tumor microenvironment (TME) promotes its progression and resistance to therapy. A critical component of TME is the extracellular matrix (ECM), which plays a pivotal role in promoting the tumor's invasive behavior and aggressiveness. Nanotechnology holds significant promise for GB treatment, with the potential to address challenges posed by both the blood-brain barrier and the GB ECM. By enabling targeted delivery of therapeutic and diagnostic agents, nanotechnology offers the prospect of improving treatment efficacy and diagnostic accuracy at the tumor site. This review provides a comprehensive exploration of GB, including its epidemiology, classification, and current treatment strategies, alongside the intricacies of its TME. It highlights nanotechnology-based strategies, focusing on nanoparticle formulations such as liposomes, polymeric nanoparticles, and gold nanoparticles, which have shown promise in GB therapy. Furthermore, it explores how different emerging nanotechnology strategies modulate the ECM to overcome the challenges posed by its high density, which restricts drug distribution within GB tumors. By emphasizing the intersection of nanotechnology and GB ECM, this review underscores an innovative approach to advancing GB treatment. It addresses the limitations of current therapies, identifies new research avenues, and emphasizes the potential of nanotechnology to improve patient outcomes.
Collapse
Affiliation(s)
- Miguel Horta
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Paula Soares
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Catarina Leite Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- INEB—Instituto Nacional de Engenharia Biomédica, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Raquel T. Lima
- i3S—Instituto de Investigação e Inovação em Saúde, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; (M.H.); (P.S.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular, University of Porto, Rua Júlio Amaral de Carvalho 45, 4200-135 Porto, Portugal
- FMUP—Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
30
|
Shah V, Lam HY, Leong CHM, Sakaizawa R, Shah JS, Kumar AP. Epigenetic Control of Redox Pathways in Cancer Progression. Antioxid Redox Signal 2025. [PMID: 39815993 DOI: 10.1089/ars.2023.0465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Significance: Growing evidence indicates the importance of redox reactions homeostasis, mediated predominantly by reactive oxygen species (ROS) in influencing the development, differentiation, progression, metastasis, programmed cell death, tumor microenvironment, and therapeutic resistance of cancer. Therefore, reviewing the ROS-linked epigenetic changes in cancer is fundamental to understanding the progression and prevention of cancer. Recent Advances: We review in depth the molecular mechanisms involved in ROS-mediated epigenetic changes that lead to alteration of gene expression by altering DNA, modifying histones, and remodeling chromatin and noncoding RNA. Critical Issues: In cancerous cells, alterations of the gene-expression regulatory elements could be generated by the virtue of imbalance in tumor microenvironment. Various oxidizing agents and mitochondrial electron transport chain are the major pathways that generate ROS. ROS plays a key role in carcinogenesis by activating pro-inflammatory signaling pathways and DNA damage. This loss of ROS-mediated epigenetic regulation of the signaling pathways may promote tumorigenesis. We address all such aspects in this review. Future Directions: Developments in this growing field of epigenetics are expected to contribute to further our understanding of human health and diseases such as cancer and to test the clinical applications of redox-based therapy. Recent studies of the cancer-epigenetic landscape have revealed pervasive deregulation of the epigenetic factors in cancer. Thus, the study of interaction between ROS and epigenetic factors in cancer holds a great promise in the development of effective and targeted treatment modalities. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Hoi-Mun Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigna S Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
31
|
Taibi M, Elbouzidi A, Haddou M, Baraich A, Gharsallaoui A, Mothana RA, Alqahtani AM, Asehraou A, Bellaouchi R, Addi M, El Guerrouj B, Chaabane K. Evaluation of the Interaction Between Menthol and Camphor, Major Compounds of Clinopodium nepeta Essential Oil: Antioxidant, Anti-inflammatory and Anticancer Activities Against Breast Cancer Cell Lines. Chem Biodivers 2025:e202403098. [PMID: 39803778 DOI: 10.1002/cbdv.202403098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/14/2025]
Abstract
This study evaluates the antioxidant, anti-inflammatory, and anticancer activities of camphor, menthol, and their equimolar combination. In silico toxicity analysis confirmed the absence of toxic effects for both compounds. Antioxidant activity, assessed by 1,1-diphenyl-2-picrylhydrazyl (DPPH) and 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) assays, revealed a synergistic effect of the equimolar combination with half-maximal inhibitory concentration (IC50) values of 10.3 µg/mL (DPPH) and 8.9 µg/mL (ABTS), surpassing the efficacy of ascorbic acid (IC50 = 12.4 µg/mL). Evaluation of anti-inflammatory activity showed that the combination more effectively inhibited 5-lipoxygenase (72.5% vs. 48.3% for camphor and 52.9% for menthol) and COX-1 and COX-2 cyclooxygenases (78.1% and 79.4% respectively, vs. 60.4% and 62.7% for camphor, 64.2% and 66.3% for menthol). Anticancer activity, tested on MCF-7, MDA-MB-231, and MDA-MB-436 breast cancer lines, revealed that the equimolar combination exhibited IC50 of 27.6, 31.2, and 36.5 µg/mL, respectively, with an IC50 of 52.3 µg/mL on normal cells, demonstrating remarkable selectivity for cancer cells. These results suggest that the camphor-menthol combination represents a promising therapeutic approach against pathologies associated with oxidative stress, inflammation, and carcinogenesis.
Collapse
Affiliation(s)
- Mohamed Taibi
- Laboratoire d'Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Département de Biologie, Faculté des Sciences, Université Mohammed Premier, Oujda, Morocco
| | - Amine Elbouzidi
- Laboratoire d'Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Département de Biologie, Faculté des Sciences, Université Mohammed Premier, Oujda, Morocco
| | - Mounir Haddou
- Laboratoire d'Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Département de Biologie, Faculté des Sciences, Université Mohammed Premier, Oujda, Morocco
| | - Abdellah Baraich
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Biology Departement, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Adem Gharsallaoui
- LAGEPP UMR 5007, University of Lyon, Université Claude Bernard Lyon 1, CNRS, Villeurbanne, France
| | - Ramzi A Mothana
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulaziz M Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdeslam Asehraou
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Biology Departement, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Reda Bellaouchi
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Biology Departement, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Mohamed Addi
- Laboratoire d'Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Département de Biologie, Faculté des Sciences, Université Mohammed Premier, Oujda, Morocco
| | - Bouchra El Guerrouj
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Biology Departement, Faculty of Sciences, Mohammed First University, Oujda, Morocco
| | - Khalid Chaabane
- Laboratoire d'Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Département de Biologie, Faculté des Sciences, Université Mohammed Premier, Oujda, Morocco
| |
Collapse
|
32
|
Yamada Y, Furukawa K, Haruki K, Okui N, Shirai Y, Tsunematsu M, Yanagaki M, Yasuda J, Onda S, Ikegami T. Abdominal aortic calcification volume as a preoperative prognostic predictor for pancreatic cancer. Surg Today 2025; 55:70-77. [PMID: 38880804 DOI: 10.1007/s00595-024-02882-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Atherosclerosis and cancer may progress through common pathological factors. This study was performed to investigate the association between the abdominal aortic calcification (AAC) volume and outcomes following surgical treatment for pancreatic cancer. METHODS The subjects of this retrospective study were 194 patients who underwent pancreatic cancer surgery between 2007 and 2020. The AAC volume was assessed through routine preoperative computed tomography. Univariate and multivariate analyses were performed to evaluate the impact of the AAC volume on oncological outcomes. RESULTS A higher AAC volume (≥ 312 mm3) was identified in 66 (34%) patients, who were significantly older and had a higher prevalence of diabetes and sarcopenia. Univariate analysis revealed several risk factors for overall survival (OS), including male sex, an AAC volume ≥ 312 mm3, elevated carbohydrate antigen 19-9, prolonged operation time, increased intraoperative bleeding, lymph node metastasis, poor differentiation, and absence of adjuvant chemotherapy. Multivariate analysis identified an AAC volume ≥ 312 mm3, prolonged operation time, lymph node metastasis, poor differentiation, and absence of adjuvant chemotherapy as independent OS risk factors. The OS rate was significantly lower in the high AAC group than in the low AAC group. CONCLUSION The AAC volume may serve as a preoperative prognostic indicator for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yuta Yamada
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Kenei Furukawa
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Norimitsu Okui
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Yoshihiro Shirai
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Masashi Tsunematsu
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Mitsuru Yanagaki
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Jungo Yasuda
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Shinji Onda
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, 3-25-8, Nishi-Shinbashi, Minato-Ku, Tokyo, 105-8461, Japan
| |
Collapse
|
33
|
Chattopadhyay M, Nath N, Kodela R, Metkar S, Soyemi SA, Kashfi K. NOSH-aspirin (NBS-1120) inhibits estrogen receptor-negative breast cancer in vitro and in vivo by modulating redox-sensitive signaling pathways. J Pharmacol Exp Ther 2025; 392:100019. [PMID: 39892987 DOI: 10.1124/jpet.124.002240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 06/29/2024] Open
Abstract
Estrogen receptor (ER)-negative breast cancers are known to be aggressive and unresponsive to antiestrogen therapy, and triple-negative breast cancers are associated with poor prognosis and metastasis. Thus, new targeted therapies are needed. Forkhead box M1 (FOXM1) is abundantly expressed in human cancers and implicated in protecting tumor cells from oxidative stress by reducing the levels of intracellular reactive oxygen species (ROS). Aspirin, a prototypical anticancer agent with deleterious side effects that has been modified to release nitric oxide and hydrogen sulfide is called nitric oxide-hydrogen sulfide-releasing aspirin (NOSH-aspirin, NOSH-ASA), generating a "safer" class of new anti-inflammatory agents. We evaluated NOSH-ASA against ER-negative breast cancer using cell lines and a xenograft mouse model. NOSH-ASA strongly inhibited growth of MDA-MB-231 and SKBR3 breast cancer cells with low IC50s of 90 ± 5 and 82 ± 5 nM, respectively, with marginal effects on a normal breast epithelial cell line. NOSH-ASA inhibited cell proliferation, caused G0/G1 phase arrest, increased apoptosis, and was associated with increases in ROS. In MDA-MB-231 cell xenografts, NOSH-ASA reduced tumor size markedly, which was associated with reduced proliferation (decreased proliferating cell nuclear antigen expression), induction of apoptosis (increased terminal deoxynucleotidyl transferase dUTP nick end labeling-positive cells), and increased ROS, whereas nuclear factor κ-light-chain-enhancer of activated B cells and FoxM1 that were high in untreated xenografts were significantly reduced. mRNA data for FoxM1, p21, and cyclin D1 corroborated with the respective protein expressions and arrest of cells. Taken together, these molecular events contribute to NOSH-ASA-mediated growth inhibition and apoptotic death of ER-negative breast cells in vitro and in vivo. Additionally, as a ROS inducer and FOXM1 inhibitor, NOSH-ASA has potential as a targeted therapy. SIGNIFICANCE STATEMENT: We examined the cellular effects and xenograft tumor inhibitory potential of NOSH-aspirin, a nitric oxide- and hydrogen sulfide-donating hybrid, against estrogen receptor-negative breast cancer, which currently lacks effective therapeutic options. Inducing reactive oxygen species and downregulating forkhead box M1 are plausible mechanisms contributing to decreased cell proliferation and increased apoptosis. NOSH-aspirin reduced tumor size by 90% without inducing any observable gross toxicity, underscoring its promising translational potential.
Collapse
Affiliation(s)
- Mitali Chattopadhyay
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Niharika Nath
- Department of Biological and Chemical Sciences, New York Institute of Technology, New York, New York
| | - Ravinder Kodela
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Shalaka Metkar
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Sarin A Soyemi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York
| | - Khosrow Kashfi
- Department of Molecular, Cellular, and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, New York; Graduate Program in Biology, City University of New York Graduate Center, New York, New York.
| |
Collapse
|
34
|
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
|
35
|
Nandha SR, Checker R, Patwardhan RS, Sharma D, Sandur SK. Anti-oxidants as therapeutic agents for oxidative stress associated pathologies: future challenges and opportunities. Free Radic Res 2025; 59:61-85. [PMID: 39764687 DOI: 10.1080/10715762.2025.2450504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/13/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025]
Abstract
Free radicals have been implicated in the pathogenesis of cancer along with cardiovascular, neurodegenerative, pulmonary and inflammatory disorders. Further, the relationship between oxidative stress and disease is distinctively established. Clinical trials using anti-oxidants for the prevention of disease progression have indicated some beneficial effects. However, these trials failed to establish anti-oxidants as therapeutic agents due to lack of efficacy. This is attributed to the fact that living systems are under dynamic redox control wherein their redox behavior is compartmentalized and simple aggregation of redox couples, distributed throughout the system, is of miniscule importance while determining their overall redox state. Further, free radical metabolism is intriguingly complex as they play plural roles segregated in a spatio-temporal manner. Depending on quality, quantity and site of generation, free radicals exhibit beneficial or harmful effects. Use of nonspecific, non-targeted, general ROS scavengers lead to systemic elimination of all types of ROS and interferes in cellular signaling. Failure of anti-oxidants to act as therapeutic agents lies in this oversimplification of extremely dynamic cellular redox environment as a static and non-compartmentalized redox state. Rather than generalizing the term "oxidative stress" if we can identify the "type of oxidative stress" in different types of diseases, a targeted and more specific anti-oxidant therapy may be developed. In this review, we discuss the concept of redox dynamics, role and type of oxidative stress in disease conditions, and current status of anti-oxidants as therapeutic agents. Further, we probe the possibility of developing novel, targeted and efficacious anti-oxidants with drug-like properties.
Collapse
Affiliation(s)
- Shivani R Nandha
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rahul Checker
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Bio-science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
36
|
Jun M, Vijayan V, Shin S, Nam HY, Song D, Choi J, Vasvani S, Cho SK, Park IK, Seo J. A bleomycin-mimicking manganese-porphyrin-conjugated mitochondria-targeting peptoid for cancer therapy. Bioorg Med Chem 2025; 117:118023. [PMID: 39602865 DOI: 10.1016/j.bmc.2024.118023] [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: 08/13/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Bleomycin (BLM) is a natural product with established anticancer activity, attributed to its ability to cleave intracellular DNA. BLM complexes with iron (BLM-Fe3+) exhibit peroxidase-like activity, generate reactive oxygen species (ROS), and cause DNA cleavage. Inspired by the mechanism of BLM, we synthesized a novel conjugate of manganese tetraphenylporphyrin (MnTPP) with a biomimetic peptoid (i.e., oligo-N-substituted glycines); this conjugate harnesses the oxidative capabilities of manganese porphyrins combined with the cell-penetrating ability of a previously reported mitochondria-targeting peptoid (MTP). UV-vis spectroscopy showed the formation of Mn(V)-oxo porphyrin, a potent oxidative species, in the presence of hydrogen peroxide, simulating metallobleomycin reactivity. Biological assays demonstrated that MnTPP-MTP significantly boosted ROS production and induced cytotoxicity toward cancer cells, while sparing normal fibroblasts. Tetramethylrhodamine ethyl ester (TMRE) assay revealed reversible, dose-dependent impairment of the mitochondrial membrane potential by MnTPP-MTP treatment. DNA cleavage assays showed that MnTPP-MTP, specifically in the presence of hydrogen peroxide, could elicit substantial DNA damage, in a similar way to BLM. In vivo studies using liposome-encapsulated MnTPP-MTP (lipo-peptoid) indicated superior tumor suppression, without systemic toxicity, when administered locally. Immunofluorescence staining for Ki67 and TUNEL confirmed reduced cell proliferation and increased apoptosis, respectively, validating the anticancer efficacy of lipo-peptoid. These results suggest that MnTPP-MTP, particularly in a liposomal formulation, is a promising new chemotherapeutic agent with robust oxidative mechanisms, poised for further development and application against diverse cancers.
Collapse
Affiliation(s)
- Minjae Jun
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Seungheon Shin
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Ho Yeon Nam
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Dasom Song
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Shyam Vasvani
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Steve K Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea.
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea.
| |
Collapse
|
37
|
Üremiş N, Üremiş MM. Oxidative/Nitrosative Stress, Apoptosis, and Redox Signaling: Key Players in Neurodegenerative Diseases. J Biochem Mol Toxicol 2025; 39:e70133. [PMID: 39799559 PMCID: PMC11725306 DOI: 10.1002/jbt.70133] [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: 10/24/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
Neurodegenerative diseases are significant health concerns that have a profound impact on the quality and duration of life for millions of individuals. These diseases are characterized by pathological changes in various brain regions, specific genetic mutations associated with the disease, deposits of abnormal proteins, and the degeneration of neurological cells. As neurodegenerative disorders vary in their epidemiological characteristics and vulnerability of neurons, treatment of these diseases is usually aimed at slowing disease progression. The heterogeneity of genetic and environmental factors involved in the process of neurodegeneration makes current treatment methods inadequate. However, the existence of common molecular mechanisms in the pathogenesis of these diseases may allow the development of new targeted therapeutic strategies. Oxidative and nitrosative stress damages membrane components by accumulating ROS and RNS and disrupting redox balance. This process results in the induction of apoptosis, which is important in the pathogenesis of neurodegenerative diseases through oxidative stress. Studies conducted using postmortem human samples, animal models, and cell cultures have demonstrated that oxidative stress, nitrosative stress, and apoptosis are crucial factors in the development of diseases such as Alzheimer's, Parkinson's, Multiple Sclerosis, amyotrophic lateral sclerosis, and Huntington's disease. The excessive production of reactive oxygen and nitrogen species, elevated levels of free radicals, heightened mitochondrial stress, disturbances in energy metabolism, and the oxidation and nitrosylation of cellular macromolecules are recognized as triggers for neuronal cell death. Challenges in managing and treating neurodegenerative diseases require a better understanding of this field at the molecular level. Therefore, this review elaborates on the molecular mechanisms by which oxidative and nitrosative stress are involved in neuronal apoptosis.
Collapse
Affiliation(s)
- Nuray Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| | - Muhammed Mehdi Üremiş
- Department of Medical BiochemistryFaculty of Medicine, Kahramanmaraş Sütçü İmam UniversityKahramanmaraşTurkey
| |
Collapse
|
38
|
Elhinnawi MA, Boushra MI, Hussien DM, Hussein FH, Abdelmawgood IA. Mitochondria's Role in the Maintenance of Cancer Stem Cells in Hepatocellular Carcinoma. Stem Cell Rev Rep 2025; 21:198-210. [PMID: 39422808 DOI: 10.1007/s12015-024-10797-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and is recognized as a major contributor to cancer-related mortality worldwide. Cancer stem cells (CSCs) are a tiny group of cancer cells that possess a significant ability to regenerate themselves, form tumors, and undergo differentiation. CSCs have a pivotal role in the initiation, spread, recurrence, and resistance to treatment of cancer. As a result, they are very susceptible to being targeted for therapeutic intervention. The potential to cure HCC may be achieved by efficiently targeting drugs that eradicate cancer stem cells. Mitochondria have a crucial function in granting drug resistance to cancer stem cells by means of mitochondrial metabolism, biogenesis, and dynamics. Dysfunction in mitochondrial metabolic processes, such as mitochondrial oxidative phosphorylation (OXPHOS), calcium signaling, and reactive oxygen species (ROS) generation, contributes to the initiation and progression of human malignancies, including HCC. ROS have both beneficial and detrimental effects depending on their concentration. Consequently, ROS have become a prominent subject in the study of the fundamental mechanisms of HCC. Furthermore, an imbalance in the process of creating new mitochondria is a characteristic feature of CSCs, and an increase in mitochondrial biogenesis is associated with the heightened resistance observed in CSCs. This article provides a detailed examination of the involvement of mitochondria in the preservation of CSCs, as well as the spread of HCC. A deeper understanding of how mitochondria participate in tumorigenesis and drug resistance could result in the discovery of novel cancer treatments.
Collapse
Affiliation(s)
- Manar A Elhinnawi
- Experimental Pathology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Giza, Egypt
| | | | | | | | | |
Collapse
|
39
|
Ntekoumes D, Song J, Liu H, Amelung C, Guan Y, Gerecht S. Acute Three-Dimensional Hypoxia Regulates Angiogenesis. Adv Healthc Mater 2025; 14:e2403860. [PMID: 39623803 PMCID: PMC11729260 DOI: 10.1002/adhm.202403860] [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: 10/06/2024] [Indexed: 01/15/2025]
Abstract
Hypoxia elicits a multitude of tissue responses depending on the severity and duration of the exposure. While chronic hypoxia is shown to impact development, regeneration, and cancer, the understanding of the threats of acute (i.e., short-term) hypoxia is limited mainly due to its transient nature. Here, a novel gelatin-dextran (Gel-Dex) hydrogel is established that decouples hydrogel formation and oxygen consumption and thus facilitates 3D sprouting from endothelial spheroids and, subsequently, induces hypoxia "on-demand." The Gel-Dex platform rapidly achieves acute moderate hypoxic conditions without compromising its mechanical properties. Acute exposure to hypoxia leads to increased endothelial cell migration and proliferation, promoting the total length and number of vascular sprouts. This work finds that the enhanced angiogenic response is mediated by reactive oxygen species, independently of hypoxia-inducible factors. Reactive oxygen species-dependent matrix metalloproteinases activity mediated angiogenic sprouting is observed following acute hypoxia. Overall, the Gel-Dex hydrogel offers a novel platform to study how "on-demand" acute moderate hypoxia impacts angiogenesis, with broad applicability to the development of novel sensing technologies.
Collapse
Affiliation(s)
- Dimitris Ntekoumes
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
- Department of Chemical and Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jiyeon Song
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Haohao Liu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Connor Amelung
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Ya Guan
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Sharon Gerecht
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
40
|
Russo M, Martella N, Gargano D, Fantasma F, Marcovecchio C, Russo V, Oliva MA, Segatto M, Saviano G, Di Bartolomeo S, Arcella A. Lavender Essential Oil and Its Terpenic Components Negatively Affect Tumor Properties in a Cell Model of Glioblastoma. Molecules 2024; 29:6044. [PMID: 39770132 PMCID: PMC11676467 DOI: 10.3390/molecules29246044] [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: 11/14/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive form of brain cancer in adults, characterized by extensive growth, a high recurrence rate, and resistance to treatment. Growing research interest is focusing on the biological roles of natural compounds due to their potential beneficial effects on health. Our research aimed to investigate the effects of lavender essential oil (LEO) on a GBM cell model. Chemical characterization using GC-MS analysis indicated that LEO contains several terpenes, compounds that have been found to exhibit anticancer properties by interfering with key cancer-related pathways in several cancer models. By means of cell biology assays, we demonstrated that LEO impairs cell proliferation and migration, and also reduces oxidative stress in U87 cells. We further observed that Terpinen-4-ol, contained in LEO, was capable of reproducing the effects of the oil on GBM cells. Our results suggest that the terpenic molecules present in LEO could be considered valuable allies alongside conventional therapies against GBM.
Collapse
Affiliation(s)
- Miriam Russo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Deborah Gargano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Francesca Fantasma
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Chiara Marcovecchio
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Veronica Russo
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Maria Antonietta Oliva
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Gabriella Saviano
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Sabrina Di Bartolomeo
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (M.R.); (N.M.); (D.G.); (F.F.); (C.M.); (M.S.)
| | - Antonietta Arcella
- IRCCS Istituto Neurologico Mediterraneo NEUROMED, Via Atinense 18, 86077 Pozzilli, Italy; (V.R.); (M.A.O.); (A.A.)
| |
Collapse
|
41
|
Dong J, Wang Z, Fei F, Jiang Y, Jiang Y, Guo L, Liu K, Cui L, Meng X, Li J, Wang H. Selenium Enhances the Growth of Bovine Endometrial Stromal Cells by PI3K/AKT/GSK-3β and Wnt/β-Catenin Pathways. Vet Sci 2024; 11:674. [PMID: 39729014 DOI: 10.3390/vetsci11120674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
The bovine uterus is susceptible to bacterial infections after calving, particularly from Escherichia coli (E. coli), which often results in endometritis. Additionally, postpartum stress in cows can elevate cortisol levels in the body, inhibiting endometrial regeneration and reducing immune function, thereby further increasing the risk of infection. Selenium (Se) is a common feed additive in dairy farming, known for its anti-inflammatory and antioxidant effects. The aim of this study was to investigate the regulatory role of Se in the growth of bovine endometrial stromal cells (BESCs) under the conditions of LPS-induced inflammatory damage at high cortisol levels. BESCs were treated with 1, 2, 4 μM Se in combination with co-treatment of LPS and cortisol. The results indicated that LPS inhibited the cell viability and reduced the mRNA expression of CTGF, TGF-β1, and TGF-β3. Additionally, LPS increased apoptosis, hindered the cell cycle progression by blocking it in the G0/G1 phase, and suppressed the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways. Furthermore, increased concentrations of cortisol can exacerbate the impacts of LPS on cell proliferation and apoptosis. Conversely, the supplementation of Se promoted cell viability, increased the mRNA expression of TGF-β1 and TGF-β3, and enhanced cell cycle progression, while simultaneously repressing cell apoptosis as well as activating the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways. The above findings demonstrated that Se can promote cell proliferation, reduce cell apoptosis, and aid in the growth of BESCs damaged by LPS under high levels of cortisol. The potential mechanisms may be associated with the regulation of the PI3K/AKT/GSK-3β and Wnt/β-catenin signaling pathways.
Collapse
Affiliation(s)
- Junsheng Dong
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Zi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Fan Fei
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Yeqi Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Yongshuai Jiang
- Guangling College, Yangzhou University, Yangzhou 225009, China
- School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Long Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Kangjun Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Luying Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Xia Meng
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Jianji Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| | - Heng Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education, Yangzhou 225009, China
- International Research Laboratory of Prevention and Control of Important Animal infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
42
|
Quilaqueo-Millaqueo N, Brown-Brown DA, Vidal-Vidal JA, Niechi I. NOX proteins and ROS generation: role in invadopodia formation and cancer cell invasion. Biol Res 2024; 57:98. [PMID: 39696702 DOI: 10.1186/s40659-024-00577-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
NADPH oxidases (NOX) are membrane-bound proteins involved in the localized generation of reactive oxygen species (ROS) at the cellular surface. In cancer, these highly reactive molecules primarily originate in mitochondria and via NOX, playing a crucial role in regulating fundamental cellular processes such as cell survival, angiogenesis, migration, invasion, and metastasis. The NOX protein family comprises seven members (NOX1-5 and DUOX1-2), each sharing a catalytic domain and an intracellular dehydrogenase site. NOX-derived ROS promote invadopodia formation, aberrant tyrosine kinase activation, and upregulation of matrix metalloproteinases (MMPs). Specifically, NOX5 modulates adhesion, motility, and proteolytic activation, while NOX1 likely contributes to invadopodia formation and adhesive capacity. NOX2 and NOX4 are implicated in regulating the invasive phenotype, expression of MMPs and EMT markers. DUOX1-2 participate in epithelial-mesenchymal transition (EMT), crucial for invasive phenotype development. Soluble molecules such as TGF-β and EGF modulate NOX protein activation, enhancing cell invasion through localized ROS production. This review focuses on elucidating the specific role of NOX proteins in regulating signaling pathways promoting cancer cell spread, particularly EMT, invadopodia formation and invasive capacity.
Collapse
Affiliation(s)
- Nelson Quilaqueo-Millaqueo
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - David A Brown-Brown
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - Jetzabel A Vidal-Vidal
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile.
| |
Collapse
|
43
|
Chauhan A, Kamal R, Bhatia R, Singh TG, Awasthi A. From Bench to Bedside: ROS-Responsive Nanocarriers in Cancer Therapy. AAPS PharmSciTech 2024; 26:10. [PMID: 39668268 DOI: 10.1208/s12249-024-03011-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 11/25/2024] [Indexed: 12/14/2024] Open
Abstract
Reactive oxygen species (ROS) play a dual role in cancer, acting as both signaling molecules that promote tumour growth and as agents that can inhibit tumour progression through cytotoxic effects. In cancer therapy, ROS-responsive drug delivery systems take advantage of the elevated ROS levels found in tumors compared to healthy tissues. These systems are engineered to release drugs precisely in response to increased ROS levels in tumour cells, allowing targeted and controlled treatment, minimizing side effects, and enhancing therapeutic outcomes. ROS generation in cancer cells is linked to metabolic changes, mitochondrial dysfunction, and oncogenic signaling, leading to increased oxidative stress. Tumour cells manage this by upregulating antioxidant defenses to prevent ROS from reaching harmful levels. This balance between ROS production and neutralization is critical for cancer cell survival, making ROS both a challenge and an opportunity for targeted therapies. ROS also connect inflammation and cancer. Chronic inflammation leads to elevated ROS, which can damage DNA and proteins, promoting mutations and cancer development. Additionally, ROS contribute to protein degradation, affecting essential cellular functions. Therapeutic strategies targeting ROS aim to either increase ROS beyond tolerable levels for cancer cells or inhibit their antioxidant defenses. Nanocarriers responsive to ROS show great potential in improving the precision of cancer treatments by releasing drugs specifically in high ROS environments, like tumors. This review discusses the mechanisms of ROS in cancer, its role in inflammation and protein degradation, and the advances in ROS-targeted nanocarrier therapies across different cancer types.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Raj Kamal
- School of Pharmacy, Desh Bhagat University, 147301, Punjab, India, Mandi Gobindgarh
| | - Rohit Bhatia
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
| | | | - Ankit Awasthi
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India.
| |
Collapse
|
44
|
Young M, Booth DM, Smith D, Tigano M, Hajnόczky G, Joseph SK. Transcriptional regulation in the absence of inositol trisphosphate receptor calcium signaling. Front Cell Dev Biol 2024; 12:1473210. [PMID: 39712573 PMCID: PMC11659226 DOI: 10.3389/fcell.2024.1473210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The activation of IP3 receptor (IP3R) Ca2+ channels generates agonist-mediated Ca2+ signals that are critical for the regulation of a wide range of biological processes. It is therefore surprising that CRISPR induced loss of all three IP3R isoforms (TKO) in HEK293 and HeLa cell lines yields cells that can survive, grow and divide, albeit more slowly than wild-type cells. In an effort to understand the adaptive mechanisms involved, we have examined the activity of key Ca2+ dependent transcription factors (NFAT, CREB and AP-1) and signaling pathways using luciferase-reporter assays, phosphoprotein immunoblots and whole genome transcriptomic studies. In addition, the diacylglycerol arm of the signaling pathway was investigated with protein kinase C (PKC) inhibitors and siRNA knockdown. The data showed that agonist-mediated NFAT activation was lost but CREB activation was maintained in IP3R TKO cells. Under base-line conditions transcriptome analysis indicated the differential expression of 828 and 311 genes in IP3R TKO HEK293 or HeLa cells, respectively, with only 18 genes being in common. Three main adaptations in TKO cells were identified in this study: 1) increased basal activity of NFAT, CREB and AP-1; 2) an increased reliance on Ca2+- insensitive PKC isoforms; and 3) increased production of reactive oxygen species and upregulation of antioxidant defense enzymes. We suggest that whereas wild-type cells rely on a Ca2+ and DAG signal to respond to stimuli, the TKO cells utilize the adaptations to allow key signaling pathways (e.g., PKC, Ras/MAPK, CREB) to transition to the activated state using a DAG signal alone.
Collapse
Affiliation(s)
- Michael Young
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David M. Booth
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David Smith
- Center for Single Cell Biology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marco Tigano
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gyӧrgy Hajnόczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Suresh K. Joseph
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
45
|
Gao J, Zhang C, Hu LJ, Lin K, Zhou M, Zhu SX, Sun BL, Liu W, Shu XJ, Gan GP, Ye XS, Yang DS. Isolation and Characterization of Sesquiterpene Lactones From Syneilesis aconitifolia and Evaluation of Their Anti-Breast Cancer Activity. Chem Biodivers 2024:e202402891. [PMID: 39641744 DOI: 10.1002/cbdv.202402891] [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: 11/05/2024] [Revised: 12/05/2024] [Accepted: 12/06/2024] [Indexed: 12/07/2024]
Abstract
Two undescribed sesquiterpene lactones (1 and 2), along with two known analogs (3 and 4), were isolated from Syneilesis aconitifolia. Their structures and absolute configurations were elucidated using spectroscopic analysis, ECD calculations, and single crystal x-ray diffraction. Compound 3 exhibited the highest cytotoxic activity against 4T1 cells, with an IC50 value of 10.89 µM. In addition, compound 3 significantly induced the apoptosis of 4T1 cells at the concentration of 20 µM. Further anticancer study showed that compound 3 distinctly increased the production of intracellular reactive oxygen species.
Collapse
Affiliation(s)
- Jie Gao
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Cong Zhang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Li-Juan Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Kuan Lin
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Mei Zhou
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Shu-Xiu Zhu
- Department of Traditional Chinese Medicine, School of Medicine, Jianghan University, Wuhan, China
| | - Bin-Lian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Wei Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Xi-Ji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - Guo-Ping Gan
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Chinese Materia Medica Processing Engineering Center of Hubei Province, Hubei University of Chinese Medicine, Wuhan, China
| | - Xian-Sheng Ye
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, China
| | - De-Sen Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
- Chinese Materia Medica Processing Engineering Center of Hubei Province, Hubei University of Chinese Medicine, Wuhan, China
- Hubei Shizhen Laboratory, Donghu New Technology Development Zone, Wuhan, China
| |
Collapse
|
46
|
Wang Y, Wang S, Ma C, Qi W, Lv J, Zhang M, Wang S, Wang R, Lu Y, Qiu W. Nrf2 depletion enhanced curcumin therapy effect in gastric cancer by inducing the excessive accumulation of ROS. Sci Rep 2024; 14:30165. [PMID: 39627516 PMCID: PMC11615379 DOI: 10.1038/s41598-024-81375-1] [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: 03/07/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
Gastric cancer (GC) is the most common malignant tumor of the gastrointestinal tract and currently has a poor clinical outcome. Turmeric's rhizome contains a polyphenolic component called curcumin (Cur), which has been demonstrated to inhibit a variety of tumor cells, such as pancreatic, colon, lung and gastric cancers. However, it remains to be elucidated how Cur functions in GC and what molecular processes underlie it. Here, Cur showed a stronger inhibitory effect on GC cells AGS and HGC27. In addition, Cur's inhibition of GC cells growth was accompanied by increased ROS production, triggering of the Keap1-Nrf2 signaling pathway, and increased transcription of its downstream antioxidant genes HO-1, GCLM, and NQO1. However, when a ROS scavenger NAC was used, the inhibitory effect of Cur on GC cells was reversed. Nuclear factor erythroid 2-related factor 2 (Nrf2) is overexpressed or activated in cancers to shield cancer cells from oxidative damage by responding to oxidative stress (OS). Cur has been found to act as an activator of Nrf2. Notably, compared with Nrf2 knockdown and Cur alone, the combination of the two dramatically increased Cur-induced ROS overaccumulation and inhibition of GC cells proliferation, migration, and invasive abilities. Consistent with in vitro experiments, Cur combined with Nrf2 knockdown significantly inhibited tumor growth in nude mice transplanted with AGS cells. Therefore, we concluded that Nrf2 depletion enhanced Cur therapy effect in GC by inducing the excessive accumulation of ROS, indicating that this is a promising treatment strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenchen Ma
- Spine Surgery, Qingdao Municipal Hospital, Qingdao, China
| | - Weiwei Qi
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jing Lv
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China
| | | | | | - Rui Wang
- Qingdao University, Qingdao, China
| | | | - Wensheng Qiu
- Department of oncology, Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
47
|
Soulier M, Lekbaby B, Houari I, Decauchy H, Pavy A, Coumes A, Morichon R, Dufour T, Fouassier L. Targeting cholangiocarcinoma cells by cold piezoelectric plasmas: in vitro efficacy and cellular mechanisms. Sci Rep 2024; 14:30178. [PMID: 39632956 PMCID: PMC11618313 DOI: 10.1038/s41598-024-81664-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
Cold piezoelectric plasma (CPP) is a novel approach in cancer therapy, enabling the development of portable treatment devices capable of triggering cancer cell death. While its effectiveness remains underexplored, this research focuses on its application against cholangiocarcinoma (CCA), an aggressive cancer of the biliary tract. A CPP device is utilized to generate either a corona discharge (Pz-CD) or a dielectric barrier discharge (Pz-DBD) for in vitro experiments. Notably, Pz-CD can deliver more power than Pz-DBD, although both sources produce significant levels of reactive species in plasma and liquid phases. This work shows that CPP causes a gradient increase in medium temperature from the center towards the edges of the culture well, especially for longer treatment times. Although Pz-CD heats more significantly, it cools quickly after plasma extinction. When applied to human CCA cells, CPP shows immediate and long-term effects, more localized for Pz-CD, while more uniform for Pz-DBD. Immediate effects result also in actin cytoskeleton remodeling without alteration of the cell membrane permeability. Long-term effects of CPP, although the antioxidant system is engaged, include activation of the DNA damage response pathway leading to cell death. In conclusion, CPP should be recognized as a promising antitumor therapy.
Collapse
Affiliation(s)
- Manon Soulier
- Laboratoire de Physique des Plasmas (LPP), Sorbonne Université, Ecole Polytechnique, CNRS, Univ. Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, 75252, Paris, France.
| | - Bouchra Lekbaby
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Imane Houari
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Henri Decauchy
- Laboratoire de Physique des Plasmas (LPP), Sorbonne Université, Ecole Polytechnique, CNRS, Univ. Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, 75252, Paris, France
| | - Allan Pavy
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France
| | - Alexia Coumes
- Laboratoire de Physique des Plasmas (LPP), Sorbonne Université, Ecole Polytechnique, CNRS, Univ. Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, 75252, Paris, France
| | - Romain Morichon
- Cytometry and Imagery Platform Saint-Antoine (CISA), Sorbonne Université, 75012, Paris, France
| | - Thierry Dufour
- Laboratoire de Physique des Plasmas (LPP), Sorbonne Université, Ecole Polytechnique, CNRS, Univ. Paris-Sud, Observatoire de Paris, Université Paris-Saclay, PSL Research University, 75252, Paris, France
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, F-75012, Paris, France.
| |
Collapse
|
48
|
El Boustani M, Mouawad N, Abou Alezz M. AP3M2: A key regulator from the nervous system modulates autophagy in colorectal cancer. Tissue Cell 2024; 91:102593. [PMID: 39488930 DOI: 10.1016/j.tice.2024.102593] [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: 08/08/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Colorectal cancer (CRC) affects approximately a million people annually with a mortality rate of 50 %, accounting for 8 % of cancer-related deaths globally. Molecular characterization by The Cancer Genome Atlas could be useful in these tumor subtypes to reveal "druggable" genes. Our study focuses on the significance of the AP3M2 gene (adaptor-related protein complex 3 subunit mu 2) as a potential oncogene by employing RNA interference to inactivate AP3M2. AP3M2, inplicated in protein trafficking to lysosomes pathway and specialized organelles in neuronal cells, was amplified in CRC cell lines. The Knockdown of AP3M2 significantly reduced the viability of three CRC cell lines HCT-116, CACO2, and HT29. Intriguingly, our findings revealed an interaction between AP3M2 expression and autophagy-related genes, as well as reactive oxygen species (ROS) levels in CRC cell lines. These results suggest that targeting AP3M2 could provide a powerful strategy for CRC treatment through autophagy-ROS mechanism.
Collapse
Affiliation(s)
- Maguie El Boustani
- Nephrology and Dialysis Unit, Genomics of Renal Diseases and Hypertension Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Nayla Mouawad
- Hematology and Clinical Immunology Unit, Department of Medicine, University of Padova, Padua, Italy
| | - Monah Abou Alezz
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
49
|
Shan X, Huang R, Wang K, Yang P. A reactive oxygen species-related signature predicts the prognosis and immunosuppressive microenvironment in gliomas. Redox Rep 2024; 29:2433396. [PMID: 39607823 DOI: 10.1080/13510002.2024.2433396] [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: 11/30/2024] Open
Abstract
OBJECTIVE Intracellular redox homeostasis is crucial for a series of physiological processes. Reactive oxygen species (ROS) play important roles in redox processes. ROS can maintain cell reproduction and survival at moderate levels while promoting the initiation and progression of tumors at high levels. METHODS Based on a comprehensive analysis of ROS-related gene expression profiles, we established a gene signature associated with ROS to explore its influence on prognosis and immune microenvironment in gliomas. RESULTS The ROS-related gene expression profile dichotomized patients into two groups with different clinicopathological features and prognoses. A 19-gene ROS-related signature was used to robustly predict prognosis in both training and validation datasets. Functional analysis indicated an association between ROS levels and the immune microenvironment. The expression of immune checkpoints and M2-type markers was upregulated in the high-risk group, which suggested the immunosuppressive function of ROS. CONCLUSION ROS-related signature is an independent prognostic factor in gliomas and could potentially exert immunosuppressive effects on the tumor microenvironment.
Collapse
Affiliation(s)
- Xia Shan
- Department of Radiotherapy, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ruoyu Huang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Kuanyu Wang
- Department of Stereotactic Radiosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Pei Yang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
50
|
Jung YY, Suresh RN, Mohan CD, Harsha KB, Shivakumara CS, Rangappa KS, Ahn KS. A new isoxazolyl-urea derivative induces apoptosis, paraptosis, and ferroptosis by modulating MAPKs in pancreatic cancer cells. Biochimie 2024; 227:262-272. [PMID: 39098374 DOI: 10.1016/j.biochi.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/06/2024]
Abstract
MAPK pathway regulates the major events including cell division, cell death, migration, invasion, and angiogenesis. Small molecules that modulate the MAPK pathway have been demonstrated to impart cytotoxicity in cancer cells. Herein, the synthesis of a new isoxazolyl-urea derivative (QR-4) has been described and its effect on the growth of pancreatic cancer cells has been investigated. QR-4 reduced the cell viability in a panel of pancreatic cancer cells with minimal effect on normal hepatocytes. QR-4 induced the cleavage of PARP and procaspase-3, reduced the expression of antiapoptotic proteins, increased SubG1 cells, and annexin V/PI-stained cells indicating the induction of apoptosis. QR-4 also triggered paraptosis as witnessed by the reduction of mitochondrial membrane potential, decrease in the expression of Alix, increase in the levels of ATF4 and CHOP, and enhanced ER stress. QR-4 also modulated ferroptosis-related events such as elevation in iron levels, alteration in GSH/GSSG ratio, and increase in the expression of TFRC with a parallel decrease in the expression of GPX4 and SLC7A11. The mechanistic approach revealed that QR-4 increases the phosphorylation of all three forms of MAPKs (JNK, p38, and ERK). Independent application of specific inhibitors of these MAPKs resulted in a partial reversal of QR-4-induced effects. Overall, these reports suggest that a new isoxazolyl-urea imparts cell death via apoptosis, paraptosis, and ferroptosis by regulating the MAPK pathway in pancreatic cancer cells.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdae-mun-gu, Seoul, 02447, Republic of Korea
| | - Rajaghatta N Suresh
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore, 570006, Karnataka, India
| | - Chakrabhavi Dhananjaya Mohan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226 001, Uttar Pradesh, India
| | - Kachigere B Harsha
- Department of Chemistry, School of Engineering, University of Mysore, Mysuru, 570006, India
| | - Chilkunda Sannaiah Shivakumara
- Department of Clinical Nutrition and Dietetics, Sri Devaraj Urs Academy of Higher Education and Research, Kolar, Karnataka, 563101, India
| | | | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdae-mun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|