1
|
Liu Y, Chen N, He H, Liu L, Sun S. Sodium butyrate alleviates DSS-induced inflammatory bowel disease by inhibiting ferroptosis and modulating ERK/STAT3 signaling and intestinal flora. Ann Med 2025; 57:2470958. [PMID: 40028886 DOI: 10.1080/07853890.2025.2470958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/14/2025] [Accepted: 02/06/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD) and ulcerative colitis (UC), can seriously impact patients' quality of life. Sodium butyrate (NaB), a product of dietary fiber fermentation, has been shown to alleviate IBD symptoms. Some studies have shown that it is related to ferroptosis. However, the precise mechanism linking NaB, IBD, and ferroptosis is not clear. OBJECTIVE This study aimed to demonstrate that NaB suppresses ferroptosis, thereby alleviating inflammatory bowel disease (IBD) through modulation of the extracellular regulated protein kinases/signal transducer and activator of transcription 3 (ERK/STAT3) signaling pathway and intestinal flora. METHODS An IBD model was established using 2.5% (w/v) dextran sulfate sodium (DSS). Mice were orally administered low-dose NaB, high-dose NaB , or 5-aminosalicylic acid (5-ASA). Ferroptosis-related molecules were measured using specific kits, and western blotting (WB) and real-time polymerase chain reaction (RT-qPCR) were used to determine the levels of the target molecules. RESULTS NaB alleviated symptoms in IBD mice, including reduced weight loss, prolonged colon length, reduced disease activity index (DAI), and reduced spleen index and mRNA expression of inflammatory factors. Additionally, NaB reduced the content of Fe2+ and myeloperoxidase (MPO) and increased the content of GSH and the activity of superoxide dismutase (SOD), which reflected NaB-inhibited ferroptosis. Moreover, western blotting showed that NaB enhanced STAT3 and ERK phosphorylation. In addition, NaB regulates the composition and functions of flora related to IBD. CONCLUSION NaB alleviates IBD by inhibiting ferroptosis and modulating ERK/STAT3 signaling and the intestinal flora.
Collapse
Affiliation(s)
- Yingyin Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Nachuan Chen
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Huaxing He
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Lulin Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Suxia Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Yang AY, Kim JY, Gwon MG, Kim K, Kwon HH, Leem J, Kim SW. Protective effects and mechanisms of cynaroside on renal fibrosis in mice with unilateral ureteral obstruction. Redox Rep 2025; 30:2500271. [PMID: 40322965 PMCID: PMC12054570 DOI: 10.1080/13510002.2025.2500271] [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: 05/08/2025] Open
Abstract
Renal fibrosis is a key factor in the progression of chronic kidney disease (CKD), and current treatments remain inadequate. In this study, we investigated the therapeutic effects of cynaroside (Cyn), a natural flavonoid, in a mouse model of renal fibrosis induced by unilateral ureteral obstruction. Cyn treatment significantly ameliorated tubular injury and interstitial fibrosis while improving renal function. Mechanistically, Cyn inhibited the expression of fibrosis-related proteins and suppressed Smad2/3 phosphorylation. Additionally, Cyn reduced myofibroblast accumulation by inhibiting epithelial-mesenchymal transition, as indicated by increased E-cadherin expression and decreased levels of mesenchymal markers. Cyn also reduced oxidative stress by downregulating the prooxidant enzyme NADPH oxidase 4 and restoring antioxidant enzymes. Furthermore, Cyn attenuated ferroptosis by regulating key proteins, including acyl-CoA synthetase long-chain family member 4, transferrin receptor 1, and glutathione peroxidase 4, while also restoring glutathione levels. Cyn alleviated endoplasmic reticulum stress, as evidenced by the downregulation of key markers such as glucose-regulated protein 78 and activating transcription factor 6, and reduced inflammation, as confirmed by decreased macrophage infiltration and lower cytokine production. Overall, Cyn demonstrated broad protective effects against renal fibrosis by modulating oxidative stress, ferroptosis, ER stress, and inflammation, positioning it as a potential therapeutic agent for CKD management.
Collapse
Affiliation(s)
- Ah Young Yang
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jung-Yeon Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Mi-Gyeong Gwon
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kiryeong Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Sung-Woo Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
3
|
Xue X, Wang M, Cui J, Yang M, Ma L, Kang R, Tang D, Wang J. Glutathione metabolism in ferroptosis and cancer therapy. Cancer Lett 2025; 621:217697. [PMID: 40189013 DOI: 10.1016/j.canlet.2025.217697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Glutathione (GSH), a non-enzymatic antioxidant in mammalian cells, plays an essential role in maintaining redox balance, mitigating oxidative stress, and preserving cellular homeostasis. Beyond its well-established function in detoxifying reactive oxygen species (ROS), GSH serves as a critical regulator of ferroptosis-an iron-dependent form of cell death marked by excessive lipid peroxidation. Serving as a cofactor for glutathione peroxidase 4 (GPX4), GSH catalyzes the conversion of lipid peroxides into non-toxic lipid alcohols, thereby preventing the accumulation of deleterious lipid oxidation products and halting the spread of oxidative damage. In cancer cells, upregulated GSH synthesis and GPX4 activity contribute to an enhanced antioxidant defense, countering oxidative stress provoked by increased metabolic demands and exposure to therapeutic agents such as chemotherapy, radiotherapy, and immunotherapy. This ability of cancer cells to modulate their ferroptosis susceptibility through GSH metabolism underscores its potential as a therapeutic target. Additionally, GSH influences several key oncogenic and tumor-suppressive signaling pathways, including NFE2L2/NRF2, TP53/p53, NF-κB, Hippo, and mTOR, which collectively regulate responses to oxidative stress, affect metabolic processes, and modulate sensitivity to ferroptosis in cancer cells. This review explores recent advancements in understanding GSH's multifaceted role in ferroptosis, emphasizing its implications for cancer biology and therapeutic interventions.
Collapse
Affiliation(s)
- Xiangfei Xue
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Manyuan Wang
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 200025, China
| | - Jiangtao Cui
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Minying Yang
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, 200025, China
| | - Lifang Ma
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, 75390, USA.
| | - Jiayi Wang
- Department of Clinical Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Shanghai Institute of Thoracic Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 200030, China; Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Inui J, Ueyama-Toba Y, Imamura C, Nagai W, Asano R, Mizuguchi H. Two-dimensionally cultured functional hepatocytes generated from human induced pluripotent stem cell-derived hepatic organoids for pharmaceutical research. Biomaterials 2025; 318:123148. [PMID: 39904185 DOI: 10.1016/j.biomaterials.2025.123148] [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: 09/27/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/06/2025]
Abstract
Human induced pluripotent stem (iPS) cell-derived hepatocyte-like cells (HLCs) are expected to replace primary human hepatocytes (PHHs) as a new stable source of hepatocytes for pharmaceutical research. However, HLCs have lower hepatic functions than PHHs, require a long time for differentiation and cannot be prepared in large quantities because they do not proliferate after their terminal differentiation. To overcome these problems, we here established hepatic organoids (iHOs) from HLCs. We then showed that the iHOs could proliferate approximately 105-fold by more than 3 passages and expressed most hepatic genes more highly than HLCs. In addition, to enable their widespread use for in vitro drug discovery research, we developed a two-dimensional culture protocol for iHOs. Two-dimensionally cultured iHOs (iHO-Heps) expressed most of the major hepatocyte marker genes at much higher levels than HLCs, iHOs, and even PHHs. The iHO-Heps exhibited glycogen storage capacity, the capacity to uptake and release indocyanine green (ICG), albumin and urea secretion, and the capacity for bile canaliculi formation. Importantly, the iHO-Heps had the activity of major drug-metabolizing enzymes and responded to hepatotoxic drugs, much like PHHs. Thus, iHO-Heps overcome the limitations of the current models and promise to provide robust and reproducible pharmaceutical assays.
Collapse
Affiliation(s)
- Jumpei Inui
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Yukiko Ueyama-Toba
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Chiharu Imamura
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Wakana Nagai
- Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Rei Asano
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
| | - Hiroyuki Mizuguchi
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Biochemistry and Molecular Biology, School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan; Laboratory of Functional Organoid for Drug Discovery, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, 567-0085, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, Osaka, 565-0871, Japan; Center for Infectious Disease Education and Research (CiDER), Osaka University, Osaka, 565-0871, Japan.
| |
Collapse
|
5
|
Kreps F, Krepsová Z, Dubaj T. Formation of oxidative and cytotoxic products of tocopherols and their adsorption onto the surface of French fries when fried with rapeseed oil. Food Chem 2025; 478:143701. [PMID: 40056623 DOI: 10.1016/j.foodchem.2025.143701] [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: 11/24/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
This work examines the impact of frying (180 °C) French fries on the degradation of fatty acids and tocopherols in rapeseed oil, with a particular focus on the formation and adsorption of gamma-tocopheryl quinone on the surface of French fries. Gamma-tocopheryl quinone has a cytotoxic effect, and long-term exposure may alter DNA. It was found that gamma-tocopheryl quinone formed 1.2 times faster than alpha-tocopheryl quinone during pan-frying French fries. Addition of French fries also accelerated the hydrolysis of fatty acids, leading to a 1.3-fold increase in the rate of tocopherol degradation compared to heating pure oil. A new finding of this study is that gamma-tocopheryl quinone is the dominant oxidation product adsorbed onto the surface of French fries during frying, with its content being 1.5 times higher than that of alpha-tocopheryl quinone. The study highlights the need to reconsider using oils rich in gamma-tocopherol for frying to minimise health risks.
Collapse
Affiliation(s)
- František Kreps
- Institute of Food Science and Nutrition, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, Bratislava SK-812 37, Slovakia.
| | - Zuzana Krepsová
- Institute of Food Science and Nutrition, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, Bratislava SK-812 37, Slovakia
| | - Tibor Dubaj
- Institute of Physical Chemistry and Chemical Physics, Faculty of Chemical and Food Technology, Slovak University of Technology in Bratislava, Radlinského 9, Bratislava SK-812 37, Slovakia
| |
Collapse
|
6
|
Yang D, Zhang C, Ma J, Tie Y, Wang S. Selenium homeostasis and male reproduction. Biochem Biophys Res Commun 2025; 765:151879. [PMID: 40279800 DOI: 10.1016/j.bbrc.2025.151879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/30/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
As an important micronutrient, selenium exerts antioxidant and anti-inflammatory effects via selenoproteins. Key components of selenium transport include the glutathione peroxidase system, selenoprotein P, metal-regulated transcription factor 1, and zinc transporter protein 8. These proteins regulate selenium homeostasis and maintain appropriate selenium levels, which subsequently influences various organ functions. Selenium is closely linked to male reproduction, as it affects physiological processes, such as spermatogenesis, maturation, and motility by regulating oxidative stress and inflammatory responses. There is a significant correlation between semen selenium levels and quality, and moderate selenium supplementation can improve semen quality in men with infertility. This review describes the general biological and specific role of selenium in male reproduction, and transport mechanism of selenoproteins. Furthermore, it discusses the current research advancements in the capacity of selenium supplementation to mitigate reproductive toxicity and improve semen quality in clinical and experimental settings.
Collapse
Affiliation(s)
- Dan Yang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, PR China.
| | - Chenhui Zhang
- North China University of Science and Technology, Tangshan, Hebei Province, 063210, PR China.
| | - Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, PR China.
| | - Yanqing Tie
- Hebei Provincial People's Hospital, Shijiazhuang, Hebei Province, 050057, PR China.
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Reproductive Health Hospital, Shijiazhuang, Hebei Province, 050071, PR China.
| |
Collapse
|
7
|
Mercola J. Reductive stress and mitochondrial dysfunction: The hidden link in chronic disease. Free Radic Biol Med 2025; 233:118-131. [PMID: 40127851 DOI: 10.1016/j.freeradbiomed.2025.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 03/26/2025]
Abstract
Conventional theories of oxidative stress have long focused on the deleterious consequences of excessive reactive oxygen species (ROS) formation. However, growing evidence reveals that an overload of reducing equivalents-termed reductive stress-may be equally pivotal in driving mitochondrial dysfunction and chronic disease. In this paradigm, abnormally high concentrations of NADH and NADPH create an electron "traffic jam" in the mitochondrial electron transport chain (ETC), leading to partial inhibition or reverse electron flow at upstream complexes. Paradoxically, this hyper-reduced environment promotes ROS generation by increasing electron leakage to molecular oxygen, thereby intensifying oxidative damage to lipids, proteins, and mitochondrial DNA. This review explores the intertwined nature of reductive and oxidative stress, showing how a surplus of reducing equivalents can potentiate metabolic derangements in conditions such as type 2 diabetes, nonalcoholic fatty liver disease, and neurodegenerative disorders. The review discusses common drivers of reductive overload, including chronic hyperglycemia, high-fat diets, and specific dietary patterns-particularly those enriched in polyunsaturated omega-6 fatty acids-that inundate mitochondria with electron donors. The review also highlights emerging evidence that targeted assessment of redox biomarkers (e.g., lactate:pyruvate, β-hydroxybutyrate:acetoacetate ratios) can provide clinically relevant indicators of reductive stress. Finally, the review examines how novel therapeutic strategies can address the underlying reductive imbalance, from rational nutrient modulation to pharmacologic interventions that restore NAD+ levels or optimize ETC flux. Recognizing reductive stress as a critical inflection point in mitochondrial pathophysiology underscores the need for a refined redox framework, one that moves beyond conventional oxidative paradigms to embrace the full spectrum of redox dysregulation in chronic degenerative disease.
Collapse
|
8
|
Trigueira PDC, Leal VDO, Cardoso BR, Mafra D, Araujo MC, Stockler-Pinto MB. Selenium supplementation in chronic kidney disease patients undergoing haemodialysis: a systematic review of the effects on plasma selenium, antioxidant and inflammatory markers, immunological parameters and thyroid hormones. Nutr Res Rev 2025; 38:294-305. [PMID: 39320843 DOI: 10.1017/s0954422424000222] [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: 09/26/2024]
Abstract
Selenium (Se) is a mineral with several biological functions, and studies have shown that its deficiency can be linked to many complications in patients with chronic kidney disease (CKD). This study aims to systematically review the effects of Se supplementation in patients with CKD undergoing haemodialysis (HD). This systematic review was carried out according to the PRISMA statement. Clinical trials were searched in PubMed, Lilacs, Embase, Scopus and Cochrane Library databases from inception to July 2021 and updated in July 2024. The protocol was registered on PROSPERO (CRD42021231444). Two independent reviewers performed the study screening and data extraction, and the risk of bias was evaluated using the Cochrane Collaboration tool. Thirteen studies were included in this review. Only nine studies showed results on Se levels; in all, reduced Se levels were observed before supplementation. A positive effect of supplementation on plasma Se level was demonstrated. Of the ten studies analysed, six demonstrated positive effects on antioxidant and inflammatory markers. Only one study analysed immunological parameters, showing a positive impact. From two studies that analysed thyroid hormones, only one showed positive results. All studies were classified as high risk of bias. The findings suggest that Se supplementation significantly increases plasma Se levels in these patients; however, there are still not enough studies to clarify the effects of Se supplementation on the antioxidant and inflammatory markers, immune system and thyroid hormones. Further studies are needed to elucidate the effects of Se supplementation and to provide a recommendation for patients with CKD undergoing HD.
Collapse
Affiliation(s)
- Pricilla de C Trigueira
- Post-Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Post-Graduate Program in Pathology, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| | - Viviane de O Leal
- Pedro Ernesto University Hospital (HUPE), University of the State of Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Barbara R Cardoso
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne, VIC, Australia
- Victorian Heart Institute, Monash University, Clayton, VIC, Australia
| | - Denise Mafra
- Post-Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Post-Graduate program in Medical Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Post-Graduate Program in Biological Sciences-Physiology, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Marina C Araujo
- Sergio Arouca National School of Public Health-Oswaldo Cruz Foundation/Ministry of Health, Rio de Janeiro, RJ, Brazil
| | - Milena B Stockler-Pinto
- Post-Graduate Program in Nutrition Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Post-Graduate Program in Pathology, Fluminense Federal University (UFF), Niterói, RJ, Brazil
- Post-Graduate Program in Cardiovascular Sciences, Fluminense Federal University (UFF), Niterói, RJ, Brazil
| |
Collapse
|
9
|
Han L, Tian X, Yang X, Li T, Wang S, Bao Y, Meng X. The pathogenesis of hepatocellular carcinoma: ERK/ULK1/NCOA4-mediated inhibition of iron autophagy, and Epimedium extract targeted modulation of this pathway to treat hepatocellular carcinoma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156666. [PMID: 40121885 DOI: 10.1016/j.phymed.2025.156666] [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: 10/11/2024] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND The pathogenesis of hepatocellular carcinoma (HCC) is characterized by its complexity and diversity, involving processes such as glycolysis, autophagy, and cellular immunity. Notably, the role of ERK/ULK1/NCOA4-mediated inhibition of iron autophagy in HCC pathogenesis has not been previously reported. This study provides a novel elucidation of HCC pathogenesis and identifies the clinical adjuvant therapy drug, Epimedium, as a potential treatment based on this mechanism. The research clarifies the regulatory effects of Epimedium on the ERK/ULK1/NCOA4-mediated inhibition of iron autophagy pathway in the treatment of HCC, thereby offering a scientific foundation for clinical treatment strategies and the development of innovative drugs. PURPOSE The objective of this study is to uncover a new aspect of HCC pathogenesis, ERK/ULK1/NCOA4-mediated inhibition of iron autophagy, and to screen for clinical targeted adjuvant therapy drugs based on this mechanism. METHODS A HCC rat model was induced with N-Nitrosodiethylamine (DEN). The physiological status of the HCC rats was assessed through indicators such as body weight and organ index. Liver damage in HCC rats was evaluated using hematoxylin and eosin (HE) staining and biochemical markers. Additionally, untargeted metabolomics was employed to explore the pathogenesis of HCC. UPLC-Q-TOF-MS combined with network pharmacology was employed to elucidate novel mechanisms, predict pathway targets, filtrate active ingredients and analyze the biological processes and signaling pathways modulated by EPME. DEN liver cancer rats were treated with different concentrations of EPME and protein expression levels were assessed by Western blot analysis. Molecular docking techniques were utilized to assess the binding affinity between the core components of EPME and target proteins. A HepG2 liver cancer in vitro model, in combination with inhibitor (SBI-0206965), was employed to verify the modulatory effects of EPME and its active ingredients on the ERK/ULK1/NCOA4 signaling pathway. Microscale thermophoretic (MST) was employed to verify the binding ability of the EPME core components to the ULK1 protein. RESULTS Metabolomics combined with network pharmacology revealed a novel pathogenesis of HCC, which is ERK/ULK1/NCOA4-mediated iron autophagy inhibition. EPME can activate iron autophagy mediated by ERK/ULK1/NCOA4 through active ingredients such as icaritin, astragalin, and emodin, thereby enhancing the survival conditions of HCC-afflicted rats and mitigating liver damage and carcinogenesis, ultimately achieving therapeutic outcomes in HCC treatment. CONCLUSION The ERK/ULK1/NCOA4-mediated iron autophagy inhibition represents a novel therapeutic mechanism for HCC. The clinical adjuvant drug EPME may exert therapeutic effects on HCC by activating ERK/ULK1/NCOA4-mediated iron autophagy.
Collapse
Affiliation(s)
- Liying Han
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Xiangmu Tian
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Xinxin Yang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Tianjiao Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| | - Shuai Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Yongrui Bao
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China; Shenyang Key Laboratory for Causes and Drug Discovery of Chronic Diseases, Shenyang 110036, China.
| | - Xiansheng Meng
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China; Liaoning Multi-dimensional Analysis of Traditional Chinese Medicine Technical Innovation Center, Dalian 116600, China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian 116600, China.
| |
Collapse
|
10
|
Li Y, Zhou F, Xu Z. TRIM25 facilitates ferroptosis in ovarian cancer through promoting PIEZO1 K63-linked ubiquitination and degradation. Transl Oncol 2025; 56:102386. [PMID: 40250035 PMCID: PMC12033990 DOI: 10.1016/j.tranon.2025.102386] [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: 11/04/2024] [Revised: 01/13/2025] [Accepted: 04/05/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Ovarian cancer represents a significant threat to women's health. and ferroptosis is recognized as a potential natural inhibitor in cancer therapy, the regulatory mechanism of TRIM25 in ovarian cancer and its potential for regulating ferroptosis as a treatment remain unclear. METHODS The role of TRIM25 in ovarian cancer was examined through functional gain- and loss-of-function assays both in vitro and in vivo, while its target genes were identified. The stability and ubiquitination sites of PIEZO1 were analyzed using protein docking and ubiquitination experiments. RESULTS TRIM25 is highly expressed in ovarian cancer and promotes the growth and metastasis of ovarian cancer cells both in vivo and in vitro. Mechanistically, it facilitates PIEZO1 degradation through ubiquitination-dependent proteasome activity, inhibits ferroptosis, and stimulates ovarian cancer cell growth. CONCLUSION Our study clearly shows that TRIM25 stimulates ovarian cancer by inducing K63-linked ubiquitination of PIEZO1, which suppresses ferroptosis and promotes excessive proliferation of ovarian cancer cells. Further research identified the ubiquitination modification site on PIEZO1, providing insights for ovarian cancer treatment.
Collapse
Affiliation(s)
- Ya Li
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China
| | - Fei Zhou
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China
| | - Zhengmei Xu
- Department of Gynecology, Affiliated Hengyang Hospital of Hunan Normal University & Hengyang Central Hospital, Hengyang, No.12 Yancheng Road, Hengyang city, Hunan province, 421000, PR China.
| |
Collapse
|
11
|
Catapano A, Cimmino F, Petrella L, Pizzella A, D'Angelo M, Ambrosio K, Marino F, Sabbatini A, Petrelli M, Paolini B, Lucchin L, Cavaliere G, Cristino L, Crispino M, Trinchese G, Mollica MP. Iron metabolism and ferroptosis in health and diseases: The crucial role of mitochondria in metabolically active tissues. J Nutr Biochem 2025; 140:109888. [PMID: 40057002 DOI: 10.1016/j.jnutbio.2025.109888] [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: 05/17/2024] [Revised: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 03/30/2025]
Abstract
Iron is essential in various physiological processes, but its accumulation leads to oxidative stress and cell damage, thus iron homeostasis has to be tightly regulated. Ferroptosis is an iron-dependent non-apoptotic regulated cell death characterized by iron overload and reactive oxygen species accumulation. Mitochondria are organelles playing a crucial role in iron metabolism and involved in ferroptosis. MitoNEET, a protein of mitochondrial outer membrane, is a key element in this process. Ferroptosis, altering iron levels in several metabolically active organs, is linked to several non-communicable diseases. For example, iron overload in the liver leads to hepatic fibrosis and cirrhosis, accelerating non-alcholic fatty liver diseases progression, in the muscle cells contributes to oxidative damage leading to sarcopenia, and in the brain is associated to neurodegeneration. The aim of this review is to investigate the intricate balance of iron regulation focusing on the role of mitochondria and oxidative stress, and analyzing the ferroptosis implications in health and disease.
Collapse
Affiliation(s)
- Angela Catapano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Fabiano Cimmino
- Department of Biology, University of Naples Federico II, Naples, Italy; Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Lidia Petrella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Amelia Pizzella
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Katia Ambrosio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Francesca Marino
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Annarita Sabbatini
- Dietetic and Clinical Nutrition Unit, IEO European Institute of Oncology IRCSS, Milan, Italy
| | - Massimiliano Petrelli
- Department of Clinical and Molecular Sciences, Clinic of Endocrinology and Metabolic Diseases, Università Politecnica delle Marche, Ancona, Italy
| | - Barbara Paolini
- Department of Innovation, experimentation and clinical research, Unit of dietetics and clinical nutrition, S. Maria Alle Scotte Hospital, University of Siena, Siena, Italy
| | - Lucio Lucchin
- Dietetics and Clinical Nutrition, Bolzano Health District, Bolzano, Italy
| | - Gina Cavaliere
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, National Research Council of Italy, Pozzuoli, Naples, Italy
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | | | | |
Collapse
|
12
|
Gencheva R, Coppo L, Arnér ESJ, Ren X. Selenium supplementation protects cancer cells from the oxidative stress and cytotoxicity induced by the combination of ascorbate and menadione sodium bisulfite. Free Radic Biol Med 2025; 233:317-329. [PMID: 40180024 DOI: 10.1016/j.freeradbiomed.2025.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/28/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
The combination of ascorbate (vitamin C) and menadione sodium bisulfite (MSB, vitamin K3), here called VC/VK3 (also named Apatone®, or M/A), has shown selective cytotoxicity in cancer cells and is under clinical investigation as a cancer therapy. However, the mechanisms of VC/VK3-induced cell death are not fully understood. In this in vitro study using human glioblastoma and non-transformed glial cell lines, we found that VC/VK3 caused higher toxicity in cancer cells in an H2O2- and iron-dependent manner, suggesting that ferroptosis may play a role in the cell death process. Furthermore, selenium supplementation significantly protected cancer cells from VC/VK3 treatment concomitantly with enhanced expression levels and enzymatic activity of antioxidant selenoproteins, including thioredoxin reductases (TXNRDs) and glutathione reductases (GPXs). We also found that VC/VK3 competes for electrons with thioredoxin (TXN), impairing peroxiredoxin 1 (PRDX1) in cells. Finally, chemically inhibiting TXNRDs or the glutathione-dependent antioxidant systems exaggerated the toxicity of VC/VK3. Overall, this study elucidated parts of the cell death mechanisms of VC/VK3 and identified combination strategies to overcome selenium-mediated resistance, advancing the translational potential of this prooxidant treatment.
Collapse
Affiliation(s)
- Radosveta Gencheva
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Lucia Coppo
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; Department of Selenoprotein Research, National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary
| | - Xiaoyuan Ren
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 17177, Sweden; IC-MedTech Corporation, Las Vegas, NV, USA.
| |
Collapse
|
13
|
Mishima E, O'Neill TJ, Hoefig KP, Chen D, Behrens G, Henkelmann B, Ito J, Nakagawa K, Heissmeyer V, Conrad M, Krappmann D. MALT1 inhibitor MI-2 induces ferroptosis by direct targeting of GPX4. Proc Natl Acad Sci U S A 2025; 122:e2507997122. [PMID: 40343971 DOI: 10.1073/pnas.2507997122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
Affiliation(s)
- Eikan Mishima
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Thomas J O'Neill
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
| | - Kai P Hoefig
- Research Unit Molecular Immune Regulation, Molecular Targets and Therapeutics Center, Helmholtz Munich, Munich 81377, Germany
| | - Deng Chen
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
| | - Gesine Behrens
- Research Unit Molecular Immune Regulation, Molecular Targets and Therapeutics Center, Helmholtz Munich, Munich 81377, Germany
| | - Bernhard Henkelmann
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
| | - Junya Ito
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Kiyotaka Nakagawa
- Laboratory of Food Function Analysis, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
| | - Vigo Heissmeyer
- Research Unit Molecular Immune Regulation, Molecular Targets and Therapeutics Center, Helmholtz Munich, Munich 81377, Germany
- Institute for Immunology, Medical Faculty, Biomedical Center, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Translational Redox Biology, Natural School of Sciences, Technical University of Munich, Garching 85748, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg 85764, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried 82152, Germany
| |
Collapse
|
14
|
Chan KY, Yu Y, Kong Y, Cheng L, Yao R, Yin Chair PS, Wang P, Wang R, Sun WY, He RR, Min J, Wang F, Björklund M. GPX4-dependent ferroptosis sensitivity is a fitness trade-off for cell enlargement. iScience 2025; 28:112363. [PMID: 40330887 PMCID: PMC12053632 DOI: 10.1016/j.isci.2025.112363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Despite wide variation, each cell type has an optimal size. Maintaining optimal size is essential for cellular fitness and function but the biological basis for this remains elusive. Here, we performed fitness analysis involving genome-wide CRISPR-Cas9 knockout data from tens of human cell lines and identified that cell size influences the essentiality of genes related to mitochondria and membrane repair. These genes also included glutathione peroxidase 4 (GPX4), which safeguards membranes from oxidative damage and prevents ferroptosis-iron-dependent death. Growth beyond normal size, with or without cell-cycle arrest, increased lipid peroxidation, resulting in a ferroptosis-sensitive state. Proteomic analysis revealed cell-cycle-independent superscaling of endoplasmic reticulum, accumulation of iron, and lipidome remodeling. Even slight increases from normal cell size sensitized proliferating cells to ferroptosis as evidenced by deep-learning-based single-cell analysis. Thus, lipid peroxidation may be a fitness trade-off that constrains cell enlargement and contributes to the establishment of an optimal cell size.
Collapse
Affiliation(s)
- Kuan Yoow Chan
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Yini Yu
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Yidi Kong
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ling Cheng
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Renzhi Yao
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Phoebe Sha Yin Chair
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Ping Wang
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
| | - Rong Wang
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Wan-Yang Sun
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Rong-Rong He
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Junxia Min
- The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Second Affiliated Hospital, School of Public Health, State Key Laboratory of Experimental Hematology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mikael Björklund
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) Institute, 718 East Haizhou Road, Haining 314400, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh EH8 9JZ, UK
| |
Collapse
|
15
|
Yin Y, Wang N, Hu B, Guo J, Chen Q, Chen Z, Shahbazi MA, Agüero L, Wang S, Li C. Thermo-responsive and biodegradable MoS 2-based nanoplatform for tumor therapy and postoperative wound management. J Colloid Interface Sci 2025; 686:634-649. [PMID: 39914308 DOI: 10.1016/j.jcis.2025.01.257] [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: 12/10/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025]
Abstract
Inorganic nanoparticles serve as versatile nanoplatforms for efficient cancer diagnosis and therapy. However, their limited in vivo degradability and excretion rates may lead to various adverse effects. Furthermore, the cascade-controlled release of drugs remains a challenge. In this study, we developed a free-radical triggered degradable MoS2-AIPH@LA nanoplatform for tumor photothermal and oxygen-independent thermodynamic therapy. This was achieved by loading the free radical initiator (2,2'-azobis[2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH)) onto MoS2 nanoparticles and encapsulating them with thermo-responsive lauric acid (LA). Upon laser irradiation, the hyperthermia generated by MoS2 induces cancer cell death and releases AIPH, an oxygen-independent and thermal-responsive radical initiator capable of producing toxic alkyl free radicals for tumor therapy and inhibiting bacterial growth. Importantly, these free radicals promote the degradation rate of MoS2-AIPH@LA, further facilitating a rapid AIPH release and improving the biocompatibility of the MoS2-AIPH@LA nanoplatform. In particular, the thermo-responsive nature of LA in this formulation effectively regulates the release of AIPH, thus reducing potential AIPH leakage into the bloodstream and minimizing safety risks. With its free-radical-triggered degradation and cascade-controlled release capabilities, MoS2-AIPH@LA shows significant promise for inhibiting tumor proliferation and managing postoperative bacterial infection.
Collapse
Affiliation(s)
- Yichen Yin
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093 PR China
| | - Nan Wang
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434 PR China
| | - Bin Hu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093 PR China
| | - Jiaxuan Guo
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093 PR China
| | - Qian Chen
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434 PR China.
| | - Zheng Chen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093 PR China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, Groningen 9713 AV The Netherlands
| | - Lissette Agüero
- Departamento de Biomateriales Poliméricos, Centro de Biomateriales, Universidad de La Habana, Ave. Universidad entre Calle Ronda y Calle G, Municipio Plaza de la Revolución 10400, Cuba
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093 PR China.
| | - Cheng Li
- Department of Anesthesiology and Perioperative Medicine, Shanghai Key Laboratory of Anesthesiology and Brain Functional Modulation, Clinical Research Center for Anesthesiology and Perioperative Medicine, Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434 PR China.
| |
Collapse
|
16
|
Dash BK, Urano Y, Mita Y, Ashida Y, Hirose R, Noguchi N. Unconventional secretion of PARK7 requires lysosomal delivery via chaperone-mediated autophagy and specialized SNARE complex. Proc Natl Acad Sci U S A 2025; 122:e2414790122. [PMID: 40327696 DOI: 10.1073/pnas.2414790122] [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: 07/23/2024] [Accepted: 03/09/2025] [Indexed: 05/08/2025] Open
Abstract
PARK7/DJ-1, a redox-sensitive protein implicated in neurodegeneration, cancer, and inflammation, exhibits increased secretion under stress. We previously demonstrated that, as a leaderless protein, PARK7 relies on an unconventional autophagy pathway for stress-induced secretion. The current study delves deeper into the mechanisms governing PARK7 secretion under oxidative stress triggered by the neurotoxin 6-hydroxydopamine (6-OHDA). Here, we revealed that 6-OHDA-induced autophagic flux is critical for PARK7 secretion. Downregulation of syntaxin 17 (STX17), a SNARE protein crucial for autophagosome-lysosome fusion and cargo degradation, hindered PARK7 secretion. Likewise, impairing lysosomal function with bafilomycin A1 (BafA1) or chloroquine (CQ) diminished PARK7 release, highlighting the importance of functional lysosomes, potentially in the form of secretory autolysosomes, in PARK7 release. We also found that 6-OHDA appeared to promote the unfolding of PARK7, allowing its selective recognition by the chaperone HSPA8 via KFERQ-like motifs, leading to PARK7 translocation to the lysosomal membrane through LAMP2 via chaperone-mediated autophagy (CMA). Additionally, a dedicated SNARE complex comprising Qabc-SNAREs (STX3/4, VTI1B, and STX8) and R-SNARE SEC22B mediates the fusion of PARK7-containing autolysosomes with the plasma membrane, facilitating the extracellular release of PARK7. Hence, this study uncovers a mechanism where 6-OHDA-induced autophagic flux drives the unconventional secretion of PARK7, involving CMA for PARK7 translocation to lysosomes and specialized SNARE complexes for membrane fusion events.
Collapse
Affiliation(s)
- Biplab Kumar Dash
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| | - Yasuomi Urano
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| | - Yuichiro Mita
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| | - Yuki Ashida
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| | - Ryoma Hirose
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| | - Noriko Noguchi
- Systems Life Sciences Laboratory, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe 610-0394, Kyoto, Japan
| |
Collapse
|
17
|
Yu Y, Hu J, Wang W, Lei H, Xi Z, Zhang P, Zhao E, Lu C, Chen H, Liu C, Li L. Targeting PSMD14 combined with arachidonic acid induces synthetic lethality via FADS1 m 6A modification in triple-negative breast cancer. SCIENCE ADVANCES 2025; 11:eadr3173. [PMID: 40344056 DOI: 10.1126/sciadv.adr3173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/03/2025] [Indexed: 05/11/2025]
Abstract
Dysregulation of deubiquitination is essential for cancer growth. However, the role of 26S proteasome non-ATPase regulatory subunit 14 (PSMD14) in the progression of triple-negative breast cancer (TNBC) remains to be determined. Gain- and loss-of-function experiments showed that silencing PSMD14 notably attenuated the growth, invasion, and metastasis of TNBC cells in vitro and in vivo. Overexpression of PSMD14 produced the opposite results. Mechanistically, PSMD14 decreased K63-linked ubiquitination on SF3B4 protein to de-ubiquitin and stabilize SF3B4 protein. Then, SF3B4/HNRNPC complex bound to FADS1 mRNA and promoted exon inclusion in the target mRNA through m6A site on FADS1 mRNA recognized by HNRNPC, thereby up-regulating the expression of FADS1 and activating Akt/mTOR signaling. Exogenous arachidonic acid supplementation combined with PSMD14 knockdown induced synthetic lethality, which was further confirmed in TNBC organoid (PDO) and TNBC patient-derived xenograft (PDX) mouse models. Overall, our findings reveal an oncogenic role of PSMD14 in TNBC progression, which indicates a potential biomarker and ferroptosis-mediated therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Yuanhang Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jin Hu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenwen Wang
- Department of Gynecology and Obstetrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hao Lei
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Zihan Xi
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Peiyi Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Ende Zhao
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chong Lu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hengyu Chen
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Hainan Medical University and Key Laboratory of Tropical Translational Medicine of Ministry of Education and School of Tropical Medicine, Hainan Medical University, Haikou 570311, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lei Li
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
18
|
Hayakawa R, Ishii T, Fushimi T, Kamei Y, Yamaguchi A, Sugimoto K, Ashida H, Akagawa M. Luteolin protects human ARPE-19 retinal pigment epithelium cells from blue light-induced phototoxicity through activation of Nrf2/Keap1 signaling. Free Radic Res 2025:1-21. [PMID: 40340707 DOI: 10.1080/10715762.2025.2503832] [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: 02/26/2025] [Revised: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 05/10/2025]
Abstract
Age-related macular degeneration (AMD), a serious physical and mental health problem worldwide, is the leading cause of irreversible, severe vision impairment and loss in older people. AMD is associated with multiple risk factors, many of which are closely linked to increased oxidative stress. Some studies have suggested that long-term and excessive exposure to blue light may be a potential risk factor for the development or progression of AMD. Recently, we demonstrated that blue light irradiation caused oxidative stress in all-trans-retinal (atRAL)-exposed human ARPE-19 retinal pigment epithelium cells by generating singlet oxygen (1O2), leading to apoptotic cell death. Luteolin, a flavonoid found in various edible plants, has been reported to possess divergent health-promoting properties including anti-oxidative and chemopreventive effects by up-regulating anti-oxidative and phase II detoxifying enzymes through activation of Keap1/Nrf2 signaling. Herein, we verified the cytoprotective action of luteolin against blue light irradiation using atRAL-exposed ARPE-19 cells. Our results established that luteolin effectively prevented blue light-induced apoptosis of ARPE-19 cells by mitigating oxidative stress. We also confirmed that luteolin suppressed intracellular accumulation of 1O2 and formation of atRAL-derived lipofuscin by increased expression of heme oxygenase-1 and aldehyde dehydrogenase 1A1 through activation of Keap1/Nrf2 signaling. Furthermore, our data implied that the luteolin-provoked activation of Keap1/Nrf2 signaling might be due to covalent binding of luteolin o-quinone to the critical cysteinyl thiol in Keap1. The present results suggest that luteolin could be helpful in the prevention and amelioration of blue light-induced retinal degeneration, including AMD.
Collapse
Affiliation(s)
- Ryo Hayakawa
- Department of Biological Chemistry, Division of Applied Life Science, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Sakai, Japan
| | - Takeshi Ishii
- Faculty of Nutrition, Kobe Gakuin University, Kobe, Japan
| | - Taiki Fushimi
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yuki Kamei
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Ai Yamaguchi
- Live Cell Imaging Institute, Osaka, Japan
- Research Institute for Cellular Damage Evaluation of Ionizing Radiation and Blue Light, Osaka Prefecture University, Sakai, Japan
| | - Kenji Sugimoto
- Live Cell Imaging Institute, Osaka, Japan
- Research Institute for Cellular Damage Evaluation of Ionizing Radiation and Blue Light, Osaka Prefecture University, Sakai, Japan
| | - Hitoshi Ashida
- Faculty of Food and Nutrition, Mukogawa Women's University, Nishinomiya, Japan
| | - Mitsugu Akagawa
- Department of Food and Nutrition, Institute of Biomedical sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
19
|
Agbadua OG, Kúsz N, Berkecz R, Vass E, Csámpai A, Tóth G, Balogh GT, Marcourt L, Wolfender JL, Queiroz EF, Hunyadi A. New Insights into the French Paradox: Free Radical Scavenging by Resveratrol Yields Cardiovascular Protective Metabolites. J Med Chem 2025. [PMID: 40331971 DOI: 10.1021/acs.jmedchem.4c03061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Resveratrol was subjected to a diversity-oriented synthesis using oxidative transformations by various biorelevant, biomimetic, or biomimetic-related chemical reagents. Using a combined strategy of ultrahigh-resolution profiling, bioactivity screening, and bioactivity-guided isolation, 19 metabolites were obtained. The compounds were tested for their in vitro enzyme inhibitory activity on angiotensin-1 converting enzyme (ACE), cyclooxygenase-1 and -2, and 15-lipoxygenase (LOX), and evaluated for their relevant drug-like properties in silico. The compounds demonstrated a generally increased cardiovascular protective and anti-inflammatory potential and better drug-likeness compared to resveratrol. Trans-δ-viniferin (6) was identified as a competitive, C-domain-selective ACE inhibitor that is over 20 times more potent than resveratrol. Further, trans-ε-viniferin (2) acted as an over 40 times stronger LOX inhibitor than resveratrol. While our results cannot be directly translated to the health benefits of dietary resveratrol consumption without further studies, it is demonstrated that biologically relevant oxidative environments transform resveratrol into potent cardiovascular protective and anti-inflammatory metabolites.
Collapse
Affiliation(s)
- Orinamhe G Agbadua
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
| | - Norbert Kúsz
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
| | - Róbert Berkecz
- Institute of Pharmaceutical Analysis, University of Szeged, Somogyi str. 4, H-6720 Szeged, Hungary
| | - Elemér Vass
- Department of Organic Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/a, H-1117 Budapest, Hungary
| | - Antal Csámpai
- Department of Organic Chemistry, Eötvös Loránd University, Pázmány Péter sétány 1/a, H-1117 Budapest, Hungary
| | - Gábor Tóth
- NMR Group, Department of Inorganic and Analytical Chemistry, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - György T Balogh
- Department of Pharmaceutical Chemistry, Semmelweis University, H-1092 Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, H-1085 Budapest, Hungary
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, H-1111 Budapest, Hungary
| | - Laurence Marcourt
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Jean-Luc Wolfender
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Emerson Ferreira Queiroz
- School of Pharmaceutical Sciences, University of Geneva, CMU, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, CMU, 1211 Geneva, Switzerland
| | - Attila Hunyadi
- Institute of Pharmacognosy, University of Szeged, Eötvös str. 6, H-6720 Szeged, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Eötvös str. 6, H-6720 Szeged, Hungary
- Graduate Institute of Natural Products, Shih-Chuan first Rd. 100, Kaohsiung 807, Taiwan
| |
Collapse
|
20
|
Li Q, Li C, Liu X, Guo Z, Li X, Zhang X. The key role of Piezo1 channels in ferroptosis after spinal cord injury and the therapeutic potential of Piezo1 inhibitors. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:132-140. [PMID: 40339662 DOI: 10.1016/j.pbiomolbio.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/07/2025] [Accepted: 05/04/2025] [Indexed: 05/10/2025]
Abstract
BACKGROUND Ferroptosis has been confirmed to be one of the key mechanisms of neuronal injury and dysfunction after spinal cord injury (SCI). Mechanical stresses such as deformation, compression, and stretching not only directly cause physical damage to spinal cord tissue at the moment of SCI, but also promote the development of ferroptosis through various pathways. However, the mechanism of ferroptosis after SCI remains unclear, which hinders the development of therapeutic methods. OBJECTIVE This article aims to review the key mechanisms by which mechanical stress affects ferroptosis after SCI, including its impact on the structure and function of the endoplasmic reticulum (ER) and mitochondria, its role in triggering inflammatory responses, and its activation of mechanosensitive channels. Special emphasis is placed on the role of Piezo1 channels, which are key factors in cell mechanosensation and ion homeostasis regulation. The review explores how Piezo1 channels are upregulated by mechanical stress after SCI and participate in the ferroptosis process by mediating ion flow and other mechanisms. CONCLUSIONS Inhibiting Piezo1 channels may be a potential therapeutic strategy for SCI. This review summarizes the therapeutic potential of Piezo1 inhibitors by sorting out existing studies, hoping to provide a theoretical basis for effective therapeutic strategies targeting ferroptosis after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinyu Liu
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Zixuan Guo
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xinxin Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
21
|
Yang Z, Zhao K, Li X, Yanzhang R, Zhang H, Yu Y, Yan M, Fang S, Li T, Li H, Chu X, Han S, Zhang Z, Teng J, Jin G, Guo Z. ZIP8 modulates ferroptosis to drive esophageal carcinoma progression. Cell Death Dis 2025; 16:366. [PMID: 40328750 PMCID: PMC12056185 DOI: 10.1038/s41419-025-07692-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: 08/18/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Ferroptosis, a regulated form of cell death characterized by iron-dependent phospholipid peroxidation, remains poorly understood in the context of esophageal cancer development and its regulatory mechanisms. Through comprehensive bioinformatic analyses, we identified ferroptosis-related pathways as crucial mediators in esophageal cancer progression, with ZIP8 emerging as a key regulatory element. We observed significant upregulation of ZIP8 in esophageal cancer specimens, which correlated with poor clinical outcomes. Functional studies demonstrated that ZIP8 depletion significantly attenuated cellular proliferation in vitro. Mechanistically, elevated ZIP8 expression enhanced zinc-dependent phosphorylation of CREB, leading to upregulation of the ferroptosis suppressor GPX4 and inhibition of this iron-dependent cell death modality. Significantly, we discovered that the natural compound Nobiletin targeted ZIP8, inhibiting Esophageal squamous cell carcinoma (ESCC) cell growth in vitro and in vivo. Our findings demonstrate ZIP8 as a potential therapeutic target in ESCC and suggest that promoting ferroptosis through ZIP8 inhibition may represent a novel anti-cancer strategy for ESCC therapy.
Collapse
Affiliation(s)
- Zhaojie Yang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Kexin Zhao
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangping Li
- Department of Clinical laboratory, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Ruoping Yanzhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Huijun Zhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Yin Yu
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Mingyang Yan
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shaobo Fang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Department of Medical Imaging, Zhengzhou University People's Hospital& Henan Provincial People's Hospital, Zhengzhou, China
| | - Tao Li
- Healthy Management Center, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Hao Li
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Xiao Chu
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Siyuan Han
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Ziliang Zhang
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China
| | - Junyan Teng
- Laboratory of Bone Tumor, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, China
| | - Guoguo Jin
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China.
- China-US (Henan) Hormel Cancer Institute, No.126, Zhengzhou, Henan, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease, Fuwai Central China Cardiovascular Hospital, Zhengzhou, China.
- Healthy Management Center, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China.
- Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
22
|
Ichikawa A, Toyama T, Taguchi H, Shiina S, Takashima H, Takahashi K, Ogra Y, Mizuno A, Arisawa K, Saito Y. The selenoprotein P/ApoER2 axis facilitates selenium accumulation in selenoprotein P-accepting cells and confers prolonged resistance to ferroptosis. Redox Biol 2025; 83:103664. [PMID: 40345072 DOI: 10.1016/j.redox.2025.103664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Accepted: 05/03/2025] [Indexed: 05/11/2025] Open
Abstract
The essential trace element selenium (Se) plays a significant role in redox homeostasis, while Se is very reactive and has a potent toxicity. Understanding the molecular machinery that supports Se metabolism is important for the both physiological and pathophysiological context. Incorporated Se is translated/transformed in the liver into selenoprotein P (SeP; encoded by Selenop), an extracellular Se carrier protein that effectively transports Se to the cells via the binding to its receptor apolipoprotein E receptor 2 (ApoER2), which is taken up by cells. The present study shows that SeP is a source of Se that accumulates intracellularly and can be utilized for prolonged periods under Se-deficient conditions. In cultured cells (RD and SH-SY5Y), glutathione peroxidase (GPX) expression induced by Se supply via the SeP/ApoER2 pathway was maintained longer during Se deficiency than inorganic Se, which was promoted by ApoER2 overexpression. SeP-deficient mice showed a faster decline in brain Se levels when fed a Se-deficient diet. Preserved GPX expression induced by this SeP/ApoER2 axis contributed to oxidative stress and ferroptosis resistance, suggesting that this redundant Se metabolism contributes to prolonged Se utilization and cytoprotection.
Collapse
Affiliation(s)
- Atsuya Ichikawa
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Takashi Toyama
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
| | - Hiroki Taguchi
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan; Japan Society for the Promotion of Science (JSPS) Postdoctoral Fellow, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Satoru Shiina
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Hayato Takashima
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Kazuaki Takahashi
- Graduate School of Horticulture, Chiba University, 1-33 Yayoi, Inage-ku, Chiba, 263-8522, Japan
| | - Yasumitsu Ogra
- Laboratory of Toxicology and Environmental Health, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1, Inohana, Chuo-ku, Chiba, 260-8675, Japan
| | - Ayako Mizuno
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Kotoko Arisawa
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
| |
Collapse
|
23
|
Wang X, Liu Z, Lin C. Metal ions-induced programmed cell death: how does oxidative stress regulate cell death? Life Sci 2025:123688. [PMID: 40328371 DOI: 10.1016/j.lfs.2025.123688] [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: 11/17/2024] [Revised: 04/20/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
In recent years, the mechanisms of ferroptosis and cuproptosis, a novel mode of cell death, have been elucidated and have attracted much attention. Ferroptosis is dependent on the metabolic disruption of iron ions and lipid peroxidation, whereas cuproptosis is closely related to intracellular accumulation of copper ions, aggregation of lipoylated proteins and damage to FeS cluster proteins. In particular, oxidative stress plays an important role in both types of cell death. During ferroptosis, the central role of oxidative stress is reflected in the overproduction of reactive oxygen species (ROS) and lipid peroxidation of the cell membrane. Recent studies have revealed that ROS can propagate over long distances across cells in the form of trigger waves, triggering large-scale ferroptosis. In embryonic development, different regional redox states can limit the long-distance propagation of ferroptosis waves, which is critical for muscle remodeling and tissue formation during development. In cuproptosis, processes such as copper ion accumulation, tricarboxylic acid (TCA) cycle blockade, and reduced levels of FeS cluster proteins are closely associated with oxidative stress. In addition, there is a close link between oxidative stress and death induced by other metal ions (Ca2+, Zn2+, etc.). In this paper, we review the role of oxidative stress in ferroptosis and cuproptosis and the related research progress to provide new ideas for understanding the mechanism of cell death and the occurrence and treatment of related diseases.
Collapse
Affiliation(s)
- Xingsheng Wang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Zuohao Liu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
| | - Changjun Lin
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
24
|
Xiao W, Yike W, Gongwen L, Youjia X. Ferroptosis-mediated immune responses in osteoporosis. J Orthop Translat 2025; 52:116-125. [PMID: 40271049 PMCID: PMC12017889 DOI: 10.1016/j.jot.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Osteoporosis is a common systemic metabolic disease, characterized by decreased bone mass and susceptibility to fragility fractures, often associated with aging, menopause, genetics, and immunity. Ferroptosis plays an underestimated yet crucial role in the further impact of immune function changes on osteoporosis. Cell ferroptosis can induce alterations in immune function, subsequently influencing bone metabolism. In this context, this review summarizes several mechanisms of ferroptosis and introduces the latest insights on how ferroptosis regulates immune responses, exploring the interactions between ferroptosis and other mechanisms such as oxidative stress, inflammation, etc. This review elucidates potential treatment strategies for osteoporosis, emphasizing the promising potential of ferroptosis as an emerging target in the treatment of osteoporosis. In conclusion, preparations related to ferroptosis exhibit substantial clinical promise for enhancing bone mass restoration. The translational potential of this article: This review elucidates a nuanced conversation between the immune system and osteoporosis, with ferroptosis serving as the connecting link. These findings underscore the potential of ferroptosis inhibition as a therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Wang Xiao
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wang Yike
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Liu Gongwen
- Department of Orthopaedics, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Xu Youjia
- Department of Orthopaedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
25
|
Cao S, Pang Y, Wei Y, Wang D, Xiong A, Yan J, Zeng H. Bibliometric and graphical analysis of ferroptosis and aging research: Trends, gaps, and future directions. Pathol Res Pract 2025; 269:155949. [PMID: 40174280 DOI: 10.1016/j.prp.2025.155949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/11/2025] [Accepted: 03/26/2025] [Indexed: 04/04/2025]
Abstract
Over the past 12 years, a significant body of evidence derived from extensive research has underscored the pivotal involvement of ferroptosis in the mechanisms underlying aging. Despite the growing body of literature on this topic, there remains a paucity of analytical and descriptive studies that explore its trajectory, key research directions, current trends, primary focal points, and future outlooks. This research endeavors to provide an exhaustive overview of the advancements in understanding the relationship between ferroptosis and aging over the past 12 years. The dataset utilized in this study was extracted from the Web of Science, encompassing records from January 1, 2012, through June 19, 2024. We conducted comprehensive bibliometric and visual analyses using advanced analytical tools. The results highlight China's dominant contribution, which accounts for 48.52 % of total publications, positioning it as a key player in this research area. Leading institutions, including Columbia University, Southern Medical University, and the Salk Institute for Biological Studies, demonstrate high research productivity. Pamela Maher and Gu Wei are identified as the most prolific researchers in this field. Free Radical Biology and Medicine is the leading journal, publishing the most articles in this field. This study identifies mitochondrial diseases, arrhythmias, Parkinson's disease, hepatocellular carcinoma, and iron-refractory iron deficiency anemia as the key diseases investigated in this field. This bibliometric evaluation offers critical perspectives for both experienced scholars and early-career researchers, enabling the identification of novel ideas and advancements within this domain.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Yingchen Pang
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Pulmonary and Critical Care Medicine, Shenzhen Xinhua Hospital, Shenzhen, Guangdong, PR China
| | - Yihao Wei
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong; Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, PR China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong, PR China
| | - Deli Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China
| | - Ao Xiong
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China.
| | - Jun Yan
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China.
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, PR China; Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, PR China.
| |
Collapse
|
26
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [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: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
27
|
Yang JY, Luo CH, Wang KB, Tu XY, Xiao YY, Ou YT, Xie YX, Guan CX, Zhong WJ. Unraveling the mechanisms of NINJ1-mediated plasma membrane rupture in lytic cell death and related diseases. Int J Biol Macromol 2025; 309:143165. [PMID: 40239793 DOI: 10.1016/j.ijbiomac.2025.143165] [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: 01/29/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Plasma membrane rupture (PMR), the ultimate event during lytic cell death, releases damage-associated molecular patterns (DAMPs) that trigger inflammation and immune responses in the development of various diseases. Recent years have witnessed significant advances in understanding the PMR mediated by ninjurin1 (NINJ1) in different lytic cell death processes. NINJ1 oligomerizes and ruptures the membrane in pyroptosis and other lytic cell death, participating in the pathogenesis of multiple diseases. Although the membrane-permeabilizing function of NINJ1 is well recognized, the role of NINJ1 in different types of lytic cell death and its impact on multiple disease processes have yet to be fully elucidated. This review summarizes the latest advances in the mechanisms of NINJ1-mediated PMR, discusses the membrane-inducing activity of NINJ1 in different lytic cell death, explains the implications of NINJ1 in lytic cell death-related diseases, and lists the inhibitory strategies for NINJ1. We expect to provide new insights into targeting NINJ1 to suppress lytic cell death for therapeutic benefit, which may become a new strategy to control inflammatory cell lysis-related diseases.
Collapse
Affiliation(s)
- Ji-Yan Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Chen-Hua Luo
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Kun-Bo Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xin-Yu Tu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yun-Ying Xiao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ye-Tong Ou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yan-Xin Xie
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China.
| |
Collapse
|
28
|
Xie L, He Z, Shen S, Chen L, Wang T, Li X. Fabrication of thermotolerant ovalbumin and carboxymethyl starch sodium complexed nanoparticles and high-internal-phase Pickering emulsion for food chemical hazards inhibition. Int J Biol Macromol 2025; 309:143072. [PMID: 40220834 DOI: 10.1016/j.ijbiomac.2025.143072] [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: 12/29/2024] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Protein/polysaccharide nanoparticle-stabilized Pickering high-internal-phase emulsions (HIPEs) can be customized to reduce foodborne hazards and enhance food quality. However, preparing thermostable HIPEs remains challenging. This study developed thermally stable OCCNPs-stabilized HIPEs by modulating the interactions between ovalbumin (OVA) and sodium carboxymethyl starch (CMSNa) to fabricate nanoparticles (OCCNPs). The results showed OCCNPs with a particle size of 378.6 nm were successfully prepared with a three-phase contact angle of 88.9° and a phase transition temperature of 104.6-111.9 °C. Confocal laser scanning microscopy revealed Na+ mainly distributed at the oil-water interface of HIPEs through electrostatic load of CMS, accompanied by a few in water phase, while functional oil rich in unsaturated fatty acids was encapsulated in dispersion phase of HIPEs. This spatial distribution and ordered structure (fractal dimension is 1.54) of OCCNPs at the interface enhanced the thermal stability of HIPEs, helping to inhibit hazardous compounds and reduce salt addition. Evaluation of HIPEs-based biscuits demonstrated a remarkable reduction in lipid hydroperoxide (46.5 %), malondialdehyde (27.4 %), and 5-hydroxymethylfurfural (75.7 %). Additionally, OCCNPs-stabilized HIPEs improved saltiness perception, allowing for a 52 % reduction in salt content while achieving higher sensory scores. The thermostable and multifunctional HIPEs we have developed are conducive to the development of the food safety field.
Collapse
Affiliation(s)
- Lei Xie
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Zhongchao He
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Shaodan Shen
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Ling Chen
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Tianxing Wang
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China
| | - Xiaoxi Li
- School of Food Science and Engineering, Ministry of Education Engineering Research Center of Starch and Protein Processing, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
29
|
Lu Q, Shao N, Fang Z, Ouyang Z, Shen Y, Yang R, Liu H, Cai B, Wei T. The anti-Alzheimer's disease effects of ganoderic acid A by inhibiting ferroptosis-lipid peroxidation via activation of the NRF2/SLC7A11/GPX4 signaling pathway. Chem Biol Interact 2025; 412:111459. [PMID: 40054827 DOI: 10.1016/j.cbi.2025.111459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 03/28/2025]
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system, characterized by a gradual decline in cognitive and memory abilities, social disorders, and behavioral abnormalities. Ferroptosis, an iron-dependent type of programmed cell death, is closely associated with the pathogenesis of AD. Ferroptosis is characterized by the accumulation of iron within cells, leading to increased oxidative stress, and ultimately lipid peroxidation and cell death. Ganoderic acid A (GAA), one of the major pharmacologically active components in Ganoderma lucidum, exhibits an excellent neuroprotective effect against AD. However, it is unclear whether GAA improves the symptoms of AD by inhibiting ferroptosis. This study investigated the anti-AD effects of GAA through both in vivo and in vitro experiments, and determined its molecular mechanism from the perspective of ferroptosis modulation. The results showed that GAA administration attenuated hippocampal neuronal loss, improved mitochondrial ultrastructure, and enhanced the memory and learning ability of the AD mice. In vitro assays suggested that GAA effectively protected HT22 AD cells against ferroptosis-related morphological damage, enhanced their antioxidant capacity, maintained their iron metabolism, and reduced mitochondrial dysfunction. Moreover, the immunofluorescence and western blotting results showed that the levels of NFE2 like bZIP transcription factor 2 (NRF2), glutathione peroxidase 4 (GPX4), and solute carrier family 7 member 11 (SLC7A11) both in the hippocampus of APP/PS1 mice and amyloid beta (Aβ)25-35-induced HT22 AD cells were markedly enhanced after GAA administration. In summary, these results revealed that GAA improves AD by activating on the NRF2/SLC7A11/GPX4 axis to inhibit ferroptosis-lipid peroxidation.
Collapse
Affiliation(s)
- Qingyang Lu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Nan Shao
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ziyi Fang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Zhaorong Ouyang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Yiran Shen
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Ruiling Yang
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Houli Liu
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Biao Cai
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Tao Wei
- College of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, China; Medical Basic Research Innovation Center for Integrated Chinese and Western Medicine in the Prevention and Treatment of Neurodegenerative Diseases, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
30
|
Dai Z, Yu Y, Chen R, Zhu H, Fong H, Kuang J, Jiang Y, Chen Y, Niu Y, Chen T, Shi L. Selenium promotes neural development through the regulation of GPX4 and SEPP1 in an iPSC-derived neuronal model. Biomaterials 2025; 316:123011. [PMID: 39708777 DOI: 10.1016/j.biomaterials.2024.123011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Selenium (Se) is incorporated into selenoproteins in the form of selenocysteine, which has biological functions associated with neural development. Unfortunately, the specific roles and mechanisms of selenoproteins at different stages of neuronal development are still unclear. Therefore, in this study, we successfully established a neuronal model derived from induced pluripotent stem cells (iPSC-iNeuron) and used Se nanoparticles (SeNPs@LNT) with high bioavailability to intervene at different stages of neural development in iPSC-iNeuron model. Interestingly, our results showed that SeNPs@LNT could not only accelerate the proliferation of neural progenitor cells (NPCs) by upregulating glutathione peroxidase 4 (GPX4) during the NPC stage, but also can promote neuronal differentiation by increasing selenoprotein P (SEPP1) during the neuronal stage, resulting in efficient and rapid neural development. In addition, further mechanistic studies showed that SeNPs@LNT can regulate selenoproteins by activating the PI3K/Akt/Nrf2 signaling pathway, thereby affecting neuronal development. Notably, Further analysis of ASD patients in National Center for Biotechnology Information single-cell RNA-seq datasets also revealed significantly lower GPX4 expression within NRGN-expressing neurons in ASD patients, and GO enrichment analysis of genes in NRGN-expressing neurons from ASD patients showed that the downregulation of selenoproteins due to aberrant selenoprotein synthesis may be closely associated with decreased ATP synthesis resulting from abnormal mitochondrial and respiratory chain signaling pathways. Taken together, this study provides evidence that SeNPs@LNT exerts a beneficial effect on early neural development through the regulation of selenoproteins.
Collapse
Affiliation(s)
- Zhenzhu Dai
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yanzi Yu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Ruhai Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Hongyao Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Hin Fong
- Faculty of Medicine, International School, Jinan University, Guangzhou, 510632, China
| | - Junxin Kuang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China
| | - Yunbo Jiang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yalan Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Yimei Niu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China
| | - Tianfeng Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China; Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China.
| | - Lingling Shi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-Human Primate Research, GHM Institute of CNS Regeneration, Department of Chemistry, Jinan University, Guangzhou, 510632, China; Department of Psychiatry, The First Affiliated Hospital, Jinan University, Guangzhou, 510632, China; Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China; Department of Neurology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, 570100, China.
| |
Collapse
|
31
|
Li C, Hu G, Huang J, Lu C, Zhang K, Shen Y, Zou Y, Zhang L, Zheng X, Chang Y. Skin mucus proteomic provides insights into the alkaline tolerance of grass carp (Ctenopharyngodon idella). COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101523. [PMID: 40327979 DOI: 10.1016/j.cbd.2025.101523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025]
Abstract
Fish skin mucus is a dynamic mucosal layer located between the epidermis and the environment, serving as the first line of defense against external environmental stresses. Studying changes in fish skin mucus under high alkaline conditions can help identify potential biomarkers of alkaline tolerance, thereby providing a basis for developing non-invasive detection methods. In this study, grass carp (Ctenopharyngodon idella) were subjected to a 7-day stress test at three different alkalinity levels: low alkalinity (10 mmol/L sodium bicarbonate), medium alkalinity (30 mmol/L sodium bicarbonate), and high alkalinity (50 mmol/L sodium bicarbonate). Fish skin mucus was collected and analyzed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) in data-independent acquisition (DIA) mode. A total of 9656 proteins were identified across 12 samples from four groups. The low and medium alkalinity groups shared more common functional enrichments, while the high alkalinity group showed significant enrichments in pathways related to oxygen binding, the immune system, and myocarditis. More nodes and pairwise interactions were founded in the high alkalinity group. Multiple proteins related to osmoregulation, including Prostaglandin G/H Synthase 1 (PTGS1) and Group XV Phospholipase A2 (PLA2G15), are highly expressed in the high-alkalinity treatment group. These results suggest that high-alkalinity stress-induced changes in the skin mucus of grass carp reflect the continuous activation of osmoregulatory mechanisms, and the high expression of PTGS1 and PLA2G15 in the skin mucus may serve as signatures of the survival status of grass carp in alkaline conditions.
Collapse
Affiliation(s)
- Chenghao Li
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Guo Hu
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Jing Huang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Cuiyun Lu
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Kexin Zhang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yubang Shen
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Yuting Zou
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, China
| | - Limin Zhang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Xianhu Zheng
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China
| | - Yumei Chang
- National and Local Joint Engineering Laboratory for Freshwater Fish Breeding, Key Laboratory of Freshwater Aquatic Biotechnology and Genetic Breeding, Heilongjiang Province's Key Laboratory of Fish Stress Resistance Breeding and Germplasm Characteristics on Special Habitats, Heilongjiang River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Harbin 150070, China.
| |
Collapse
|
32
|
Hu J, Ma F, Chen X, Lu X, Lu Y, Jiang Y. F-box only protein 10 protects against kidney tubulointerstitial fibrosis by inhibiting ACSL4-mediated lipid peroxidation and ferroptosis. Cell Signal 2025:111841. [PMID: 40311989 DOI: 10.1016/j.cellsig.2025.111841] [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: 02/19/2025] [Revised: 04/02/2025] [Accepted: 04/28/2025] [Indexed: 05/03/2025]
Abstract
Renal tubular epithelial cell ferroptosis is significantly linked to kidney tubulointerstitial fibrosis, a critical pathological condition in chronic kidney disease. F-box only protein 10 (FBXO10), a newly identified ferroptosis-regulating gene, plays a role in various pathological contexts; however, its involvement in kidney tubulointerstitial fibrosis is not yet fully understood. This research sought to investigate whether FBXO10 regulates ferroptosis in kidney tubular epithelial cells and its relationship with kidney tubulointerstitial fibrosis in both animal and cellular models. We observed a significant decrease in FBXO10 levels in mice with unilateral ureteral obstruction (UUO) and HK-2 cells exposed to TGF-β1. FBXO10 overexpression inhibited the EMT process and counteracted the typical ferroptosis features evoked by TGF-β1 or erastin in HK-2 cells. Compared with those in wild-type (WT) mice, kidney injury, inflammation, and fibrosis are exacerbated in FBXO10-knockout (KO) mice, with elevated ferroptosis levels. Conversely, FBXO10 overexpression reversed these symptoms, alleviating kidney fibrosis and ferroptosis in both WT and FBXO10 KO mice with UUO. Mechanistically, FBXO10 directly interacted with ACSL4 and promoted its ubiquitination and degradation. Overexpression of ACSL4 reversed the inhibitory effect of FBXO10 overexpression on TGF-β1-induced ferroptotic death and fibrosis in HK-2 cells. In summary, FBXO10 mitigates ferroptosis in renal tubular epithelial cells by inhibiting ACSL4-mediated lipid peroxidation, thereby hindering the progression of kidney tubulointerstitial fibrosis. FBXO10 is proposed as a promising target for treating kidney disorders related to tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Jinping Hu
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Feng Ma
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Xinping Chen
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Xue Lu
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Yingying Lu
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China
| | - Yali Jiang
- Department of Nephrology, Honghui Hospital, Xi'an Jiaotong University, Xi'an 710054, Shaanxi Province, China.
| |
Collapse
|
33
|
Qiu Y, Hüther JA, Wank B, Rath A, Tykwe R, Aldrovandi M, Henkelmann B, Mergner J, Nakamura T, Laschat S, Conrad M, Stöhr D, Rehm M. Interplay of ferroptotic and apoptotic cell death and its modulation by BH3-mimetics. Cell Death Differ 2025:10.1038/s41418-025-01514-7. [PMID: 40301648 DOI: 10.1038/s41418-025-01514-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/28/2025] [Accepted: 04/07/2025] [Indexed: 05/01/2025] Open
Abstract
Ferroptosis and apoptosis are widely considered to be independent cell death modalities. Ferroptotic cell death is a consequence of insufficient radical detoxification and progressive lipid peroxidation, which is counteracted by glutathione peroxidase-4 (GPX4). Apoptotic cell death can be triggered by a wide variety of stresses, including oxygen radicals, and can be suppressed by anti-apoptotic members of the BCL-2 protein family. Mitochondria are the main interaction site of BCL-2 family members and likewise a major source of oxygen radical stress. We therefore studied if ferroptosis and apoptosis might intersect and possibly interfere with one another. Indeed, cells dying from impaired GPX4 activity displayed hallmarks of both ferroptotic and apoptotic cell death, with the latter including (transient) membrane blebbing, submaximal cytochrome-c release and caspase activation. Targeting BCL-2, MCL-1 or BCL-XL with BH3-mimetics under conditions of moderate ferroptotic stress in many cases synergistically enhanced overall cell death and frequently skewed primarily ferroptotic into apoptotic outcomes. Surprisingly though, in other cases BH3-mimetics, most notably the BCL-XL inhibitor WEHI-539, counter-intuitively suppressed cell death and promoted cell survival following GPX4 inhibition. Further studies revealed that most BH3-mimetics possess previously undescribed antioxidant activities that counteract ferroptotic cell death at commonly employed concentration ranges. Our results therefore show that ferroptosis and apoptosis can intersect. We also show that combining ferroptotic stress with BH3-mimetics, context-dependently can either enhance and convert cell death outcomes between ferroptosis and apoptosis or can also suppress cell death by intrinsic antioxidant activities.
Collapse
Affiliation(s)
- Yun Qiu
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Juliana A Hüther
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Bianca Wank
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Antonia Rath
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - René Tykwe
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Maceler Aldrovandi
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Bernhard Henkelmann
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at MRI, TUM, Munich, Germany
| | - Toshitaka Nakamura
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Sabine Laschat
- Institute of Organic Chemistry, University of Stuttgart, Stuttgart, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Daniela Stöhr
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
34
|
Song L, Gao F, Man J. Ferroptosis: the potential key roles in idiopathic pulmonary fibrosis. Eur J Med Res 2025; 30:341. [PMID: 40296070 PMCID: PMC12036158 DOI: 10.1186/s40001-025-02623-2] [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: 12/06/2024] [Accepted: 04/21/2025] [Indexed: 04/30/2025] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive interstitial lung disease characterized by recurrent injury to alveolar epithelial cells, epithelial-mesenchymal transition, and fibroblast activation, which leads to excessive deposition of extracellular matrix (ECM) proteins. However, effective preventative and therapeutic interventions are currently lacking. Ferroptosis, a unique form of iron-dependent lipid peroxidation-induced cell death, exhibits distinct morphological, physiological, and biochemical features compared to traditional programmed cell death. Recent studies have revealed a close relationship between iron homeostasis and the pathogenesis of pulmonary interstitial fibrosis. Ferroptosis exacerbates tissue damage and plays a crucial role in regulating tissue repair and the pathological processes involved. It leads to recurrent epithelial injury, where dysregulated epithelial cells undergo epithelial-mesenchymal transition via multiple signaling pathways, resulting in the excessive release of cytokines and growth factors. This dysregulated environment promotes the activation of pulmonary fibroblasts, ultimately culminating in pulmonary fibrosis. This review summarizes the latest advancements in ferroptosis research and its role in the pathogenesis and treatment of IPF, highlighting the significant potential of targeting ferroptosis for IPF management. Importantly, despite the rapid developments in this emerging research field, ferroptosis studies continue to face several challenges and issues. This review also aims to propose solutions to these challenges and discusses key concepts and pressing questions for the future exploration of ferroptosis.
Collapse
Affiliation(s)
- Longfei Song
- Department of Rehabilitation Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428 Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Fusheng Gao
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China
| | - Jun Man
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Shandong Second Medical University, No. 2428, Yuhe Road, Kuiwen District, Weifang City, 261041, Shandong Province, China.
- Clinical Research Center, Affiliated Hospital of Shandong Second Medical University, No. 4948, Shengli East Street, Kuiwen District, Weifang City, 261041, Shandong Province, China.
| |
Collapse
|
35
|
Zheng J, Zhang W, Ito J, Henkelmann B, Xu C, Mishima E, Conrad M. N-acetyl-l-cysteine averts ferroptosis by fostering glutathione peroxidase 4. Cell Chem Biol 2025:S2451-9456(25)00100-X. [PMID: 40311609 DOI: 10.1016/j.chembiol.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/08/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
N-acetyl-l-cysteine (NAC) is a medication and a widely used antioxidant in cell death research. Despite its somewhat obscure mechanism of action, its role in inhibiting ferroptosis is gaining increasing recognition. In this study, we demonstrate that NAC treatment rapidly replenishes the intracellular cysteine pool, reinforcing its function as a prodrug for cysteine. Interestingly, its enantiomer, N-acetyl-d-cysteine (d-NAC), which cannot be converted into cysteine, also exhibits a strong anti-ferroptotic effect. We further clarify that NAC, d-NAC, and cysteine all act as direct reducing substrates for GPX4, counteracting lipid peroxidation. Consequently, only GPX4-rather than system xc-, glutathione biosynthesis, or ferroptosis suppressor protein 1-is necessary for NAC and d-NAC to prevent ferroptosis. Additionally, we identify a broad range of reducing substrates for GPX4 in vitro, including β-mercaptoethanol. These findings provide new insights into the mechanisms underlying the protective effects of NAC and other potential GPX4-reducing substrates against ferroptosis.
Collapse
Affiliation(s)
- Jiashuo Zheng
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Weijia Zhang
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Junya Ito
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Laboratory of Food Function Analysis, Tohoku University Graduate School of Agricultural Science, Sendai, Miyagi 980-8572, Japan
| | - Bernhard Henkelmann
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Chenxi Xu
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany
| | - Eikan Mishima
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8574, Japan
| | - Marcus Conrad
- Helmholtz Zentrum München, Institute of Metabolism and Cell Death, Molecular Targets and Therapeutics Center, 85764 Neuherberg, Germany; Translational Redox Biology, Technical University of Munich (TUM), TUM Natural School of Sciences, 85748 Garching, Germany.
| |
Collapse
|
36
|
Manavi MA, Mohammad Jafari R, Shafaroodi H, Dehpour AR. The Keap1/Nrf2/ARE/HO-1 axis in epilepsy: Crosstalk between oxidative stress and neuroinflammation. Int Immunopharmacol 2025; 153:114304. [PMID: 40117806 DOI: 10.1016/j.intimp.2025.114304] [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: 10/08/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Epilepsy is a complex neurological disorder characterized by recurrent seizures, which are driven by multifaceted pathophysiological mechanisms, including oxidative stress and neuroinflammation. Despite advancements in anti-seizure medications (ASMs), a significant proportion of patients remain resistant to treatment, highlighting the need for novel therapeutic strategies. This review focuses on the Kelch-like ECH-associated protein 1 (Keap1) / Nuclear factor erythroid 2-related factor 2 (Nrf2) / Antioxidant Response Element (ARE) / Heme Oxygenase-1 (HO-1) axis as a promising target for neuroprotection in epilepsy. We explored the intricate interactions between Keap1 and Nrf2 under homeostatic conditions and how oxidative stress disrupts this balance, triggering Nrf2 activation. This review details the subsequent process of Nrf2 nuclear translocation, its binding to AREs, and the induction of cytoprotective gene expression, which collectively orchestrate a robust cellular defense response. Special emphasis is placed on HO-1, a key effector of Nrf2-mediated neuroprotection, highlighting its enzymatic function and protective mechanisms, including antioxidant, anti-inflammatory, and anti-apoptotic effects. Additionally, the review examines HO-1's role in mitigating seizure-induced neuronal damage. However, challenges remain, including variability in therapeutic responses, gaps in long-term clinical validation, and the need for standardized protocols. Future research should focus on biomarkers for personalized treatment, advanced imaging, and genetic tools to explore the Keap1/Nrf2/ARE/HO-1 axis in greater depth. Future studies should focus on overcoming the challenges of translating preclinical findings into clinical applications and exploring the long-term effects of targeting this pathway in epilepsy treatment.
Collapse
Affiliation(s)
- Mohammad Amin Manavi
- Experimental Medicine Research Center, Tehran university of medical sciences, Tehran, Iran; Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Razieh Mohammad Jafari
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Hamed Shafaroodi
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran university of medical sciences, Tehran, Iran.
| |
Collapse
|
37
|
Ke A, Yang W, Zhang W, Chen Y, Meng X, Liu J, Dai D. The cardiac glycoside periplocymarin sensitizes gastric cancer to ferroptosis via the ATP1A1-Src-YAP/TAZ-TFRC axis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156804. [PMID: 40311597 DOI: 10.1016/j.phymed.2025.156804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Targeting ferroptosis vulnerabilities in tumors has become an increasingly promising therapeutic strategy. While the regulatory effects of natural products on ferroptosis are progressively being elucidated, the role of cardiac glycosides in modulating ferroptosis remains poorly understood. PURPOSE This study aims to investigate the ferroptosis-sensitizing effects of periplocymarin (PPM), a cardiac glycoside derived from the traditional plant Periploca sepium, and to elucidate the underlying molecular mechanisms. METHODS The effects of PPM on ferroptosis regulation were comprehensively assessed through functional assays, followed by sequencing analysis to identify associated signaling pathways. Subsequent mechanistic validation experiments were conducted to confirm the upstream and downstream regulatory components involved in this ferroptosis-modulating axis. RESULTS PPM induced slow and mild apoptosis in gastric cancer cells through the inhibition of glycolysis. However, when combined with ferroptosis inducers, it promoted rapid and robust ferroptosis. In vivo, PPM sensitized gastric cancer xenografts to cisplatin-induced ferroptosis with no observable cardiotoxicity or renal impairment. Mechanistically, PPM targeted the α1 subunit of the Na+/K+-ATPase (ATP1A1), leading to the activation of Src, which subsequently induced tyrosine phosphorylation of YAP/TAZ in a Hippo-independent manner, promoting their nuclear translocation. The YAP/TAZ-TEAD transcriptional complex directly bound to the TFRC promoter region between nucleotides 401-409 upstream of the transcription start site, thereby activating TFRC transcription. This resulted in increased iron influx, elevated lipid peroxidation, and heightened sensitivity to ferroptosis. Notably, ATP1A1 was essential for ferroptosis resistance, as its knockdown mimicked the sensitizing effect of PPM on ferroptosis. Moreover, the oncogenic Src-YAP/TAZ-TFRC axis may have represented a ferroptosis vulnerability and a potential biomarker in ferroptosis therapy for cancer. Importantly, other cardiac glycosides targeting Na+/K+-ATPase, such as digitoxin and bufalin, also enhanced ferroptosis sensitivity in gastric cancer cells through activation of YAP/TAZ signaling. CONCLUSION Our findings establish the cardiac glycoside PPM as a novel ferroptosis sensitizer that targets ATP1A1 to activate the Src-YAP/TAZ-TFRC axis, providing mechanistic insights for repurposing cardiac glycosides as ferroptosis modulators in precision combinatorial cancer therapy.
Collapse
Affiliation(s)
- Angting Ke
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weiguang Yang
- Department of Nephrology, the First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Wanchuan Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Yibin Chen
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Xiangyu Meng
- Department of Gastric Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang 110042, China
| | - Jie Liu
- Translational Research Experiment Department, Science Experiment Center, China Medical University, Shenyang 110122, China
| | - Dongqiu Dai
- Department of Surgical Oncology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China; Cancer Center, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
38
|
Devos L, Dubois A, Fieuws S, Vanden Berghe T, Pirenne J, Ceulemans LJ, Monbaliu D, Jochmans I. The Efficacy of Ferroptosis Inhibition on Ischemia-Reperfusion Injury of Abdominal Organs: A Systematic Review and Meta-analysis. Transplantation 2025:00007890-990000000-01071. [PMID: 40269342 DOI: 10.1097/tp.0000000000005405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Solid organ transplantation is hampered by complications that arise after ischemia-reperfusion injury (IRI), a detrimental type of injury for which no adequate treatment options are available. Ferroptosis, an iron-dependent form of regulated cell death, is a major driver of IRI. This systematic review and meta-analysis summarizes the effects of pharmacological ferroptosis inhibition in abdominal organs in the setting of IRI. PubMed, Embase, Web of Science and Cochrane were searched for concepts "ferroptosis" and "IRI" in August 2023. To allow for meta-analyses, inhibitors were divided into different intervention pathways: (I) lipophilic radical scavengers, (II) iron chelators, (III) antioxidants, (IV) lipid metabolism inhibitors, (V) combination treatments, and (VI) others. When available, organ function and injury effect sizes were extracted and used for random-effects meta-analyses. In total 79 articles were included, describing 59 unique inhibitors in kidney, liver, and intestinal IRI. No studies in pancreas were found. Overall bias and study quality was unclear and average to low, respectively. Apart from 1 clinical study, all inhibitors were tested in preclinical settings. The vast majority of the studies showed ferroptosis inhibition to be protective against IRI under various treatment conditions. In liver and kidney IRI, meta-analyses on standardized effect sizes from 43 articles showed a combined protective effect against IRI compared with a nontreated controls for all analyzed intervention pathways. In conclusion, ferroptosis inhibition protects against abdominal IRI in preclinical research. Important questions regarding optimal intervention pathway, bioavailability, optimal dosage, side effects etc. should be addressed before clinical introduction.
Collapse
Affiliation(s)
- Lene Devos
- Laboratory of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Antoine Dubois
- Laboratory of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Steffen Fieuws
- Department of Public Health, Interuniversity Centre for Biostatistics and Statistical Bioinformatics, KU Leuven, Leuven, Belgium
| | - Tom Vanden Berghe
- Cell Death Signaling Lab, Department of Biomedical Sciences, Inflamed Centre of Excellence, University of Antwerp, Antwerp, Belgium
| | - Jacques Pirenne
- Laboratory of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Laurens J Ceulemans
- Department of Chronic Diseases and Metabolism, BREATHE, KU Leuven, Leuven, Belgium
- Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Diethard Monbaliu
- Laboratory of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Ina Jochmans
- Laboratory of Abdominal Transplantation, Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
- Abdominal Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Fan Y, Hao Y, Ding Y, Wang X, Ge X. PARK7 Is Required to Protect Epithelia Against Damage in Oral Lichen Planus. Oral Dis 2025. [PMID: 40275576 DOI: 10.1111/odi.15361] [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: 02/19/2025] [Revised: 03/26/2025] [Accepted: 04/12/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE To investigate the roles of Parkinson disease protein 7 (PARK7) in the context of oral lichen planus (OLP). METHODS Real-time PCR and western blot were performed to detect the expression of PARK7. Chromatin Immunoprecipitation (ChIP) and luciferase reporter assays were conducted to confirm the binding of p65 and NF-κB element in the promoter of the human PARK7 gene. The Caspase 3 and NF-κB detection kits were used for Caspase 3 and NF-κB activities measurement. Co-Immunoprecipitation (Co-IP) was applied to test the protein-protein interactions in HOKs. RESULTS The levels of PARK7 were up-regulated in the diseased oral keratinocytes derived from OLP patients compared to those from healthy donors. Mechanistically, the activated NF-κB pathway could increase PARK7 gene transcription in human oral keratinocytes (HOKs). Moreover, PARK7 deletion facilitated cell apoptosis and NF-κB activation in HOKs under OLP conditions. Overexpression of PARK7 could suppress cell apoptosis and NF-κB activation in the OLP cell model. At the molecular level, PARK7 could interact with BAX and p65 to inhibit cell apoptosis and NF-κB activation in HOKs, respectively. CONCLUSION Our data indicate that PARK7 is required to protect epithelia against damage in oral lichen planus.
Collapse
Affiliation(s)
- Yufeng Fan
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
- Department of Endodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Yukai Hao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
- Department of Endodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Yan Ding
- Department of Dermatology, Hainan Provincial Hospital of Skin Disease, Haikou, Hainan, China
- Department of Dermatology, Affiliated Dermatology Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Xiangyu Wang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| | - Xuejun Ge
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
- Department of Endodontics, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, China
| |
Collapse
|
40
|
Yoldas A, Bahtiyar N, Aydemir B, Toplan S. Evaluation of selenoproteins and proinflammatory cytokines in L-thyroxine-induced hyperthyroid rats: effects of selenium supplementation. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2025; 69:e240444. [PMID: 40271987 PMCID: PMC12017630 DOI: 10.20945/2359-4292-2024-0444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/25/2025] [Indexed: 04/25/2025]
Abstract
OBJECTIVE This study investigated the effects of selenium, which is known for its antioxidant and immune-supporting properties, on serum levels of thyroid function markers, selenoproteins, and proinflammatory cytokines in a model of hyperthyroidism. MATERIALS AND METHODS A total of 48 Wistar albino rats were distributed into 6 groups: a control group; a hyperthyroid group (HT group); a group fed 0.5 mg/kg sodium selenite (Se 1 group); a group fed 1 mg/kg sodium selenite (Se 2 group); a hyperthyroid group fed 0.5 mg/kg sodium selenite (HT + Se 1 group); and a hyperthyroid group fed 1 mg/kg sodium selenite (HT + Se 2 group) added to standard fodder. Serum levels of interleukin (IL)-1β, IL-6, IL-18, tumour necrosis factor alpha (TNF-α), selenoprotein P (SelP), and glutathione peroxidase 1 (GPx1) were measured using ELISAs. RESULTS IL-Iβ, IL-6, IL-18, and TNF-α levels were increased, but selenium, GPx1, and SelP levels were decreased in the hyperthyroid group compared with those in the control group. Selenium and GPx1 levels were increased, but TNF-α levels were decreased in the HT + Se 1 group compared with those in the HT group. Selenium, SelP, and GPx1 levels were increased, but TNF-α, IL-6, and IL-18 levels were decreased in the HT + Se 2 group compared with those in the HT group. CONCLUSION Our results suggest that appropriate doses of selenium may be effective at preventing inflammation and providing protection against oxidative stress in hyperthyroid rats.
Collapse
Affiliation(s)
- Aysun Yoldas
- Department of Biophysics, Istanbul Yeniyuzyil University, Faculty
of Medicine, Istanbul, Turkey
| | - Nurten Bahtiyar
- Department of Biophysics, Istanbul University-Cerrahpaşa,
Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| | - Birsen Aydemir
- Department of Biophysics, Sakarya University, Faculty of Medicine,
Sakarya, Turkey
| | - Selmin Toplan
- Department of Biophysics, Istanbul University-Cerrahpaşa,
Cerrahpaşa Faculty of Medicine, Istanbul, Turkey
| |
Collapse
|
41
|
He Z, Long H, Ma H, Ran W, Song Y, Shang W, Wang J, Qin Y, Deng Z. 4, 9-dihydroxy-α-lapachone as a potent antiproliferation agent for triple-negative breast cancer via ferroptosis. Fitoterapia 2025; 183:106567. [PMID: 40280251 DOI: 10.1016/j.fitote.2025.106567] [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: 02/18/2025] [Revised: 04/07/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant breast cancer. Ferroptosis is an oxidative, iron-dependent form of regulated cell death. Ferroptosis-targeted therapies is a promising approach to improving treatment outcomes of TNBC. Combining death pathway inhibitors with relevant indices for ferroptosis and LipROS, this study uncovered that a natural product of 4, 9-dihydroxy-α-lapachone (DLN) from Catalpa bungei "jinsi" exhibited in vitro and in vivo inhibitory activity against TNBC via ferroptosis. The molecular mechanism is an activation of the FTH1 led to iron overload, and then inhibition of cysteine-glutamate antiporter (system Xc-) and GPX4, which further sensitized TNBC cells to ferroptosis. This study clarified the pathway of DLN-induced cell death in TNBC treatment and exhibited its potential as therapeutic agent for TNBC.
Collapse
Affiliation(s)
- Zhong He
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hui Long
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Hongyue Ma
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China
| | - Wanrong Ran
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China
| | - Yinhong Song
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; Institute of Infection and Inflammation, China Three Gorges University, Yichang 443002, China.
| | - Wenjuan Shang
- Medical College, Hubei Three Gorges Polytechnic, Yichang 443000, China
| | - Junhui Wang
- State Key Laboratory of Tree Genetics and Breeding, Key Laboratory of Tree Breeding and Cultivation of State Forestry Administration, Research Institute of Forestry, Chinese Academy of Forestry, Beijing 100091, China.
| | - Ye Qin
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443002, China; State Key Laboratory of Drug Research, Molecular Imaging Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China.
| | - Zhangshuang Deng
- Hubei Key Laboratory of Natural Products Research and Development, College of Biological and Pharmaceutical Sciences, China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
42
|
Moktip T, Salaipeth L, Cope AE, Taherzadeh MJ, Watanabe T, Phitsuwan P. Current Understanding of Feather Keratin and Keratinase and Their Applications in Biotechnology. Biochem Res Int 2025; 2025:6619273. [PMID: 40308531 PMCID: PMC12041636 DOI: 10.1155/bri/6619273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 01/29/2025] [Indexed: 05/02/2025] Open
Abstract
The food industry generates substantial keratin waste, particularly chicken feathers, which are rich in amino acids and essential nutrients. However, the insolubility of keratin presents a significant challenge to its conversion. Keratinase, an enzyme produced by certain fungi and bacteria, offers a promising solution by degrading feather keratin into amino acids and soluble proteins. Among these, bacterial keratinase is notable for its superior stability and activity, although its production remains constrained, necessitating continued research to identify efficient microbial strains. Keratin-derived hydrolyzates, recognized for their biological and immunological properties, have garnered significant research interest. This review examines the structural characteristics of chicken feather keratin, its resistance to conventional proteases, and advances in keratinase production and purification techniques. Additionally, the keratin degradation mechanism and the adoption of environmentally friendly technologies for managing feather waste are explored. Finally, the review highlights the potential applications of keratinase across diverse industries, including animal feed and cosmetics.
Collapse
Affiliation(s)
- Thanakorn Moktip
- LigniTech-Lignin Technology Research Group, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
- Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
| | - Lakha Salaipeth
- LigniTech-Lignin Technology Research Group, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
- Natural Resource Management and Sustainability, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
| | - Ana Eusebio Cope
- Future Genetic Resources Cluster, Rice Breeding Innovation Platform, IRRI, Los Banos, Philippines
| | | | - Takashi Watanabe
- Research Institute for Sustainable Humanosphere, Kyoto University, Kyoto 611-0011, Japan
| | - Paripok Phitsuwan
- LigniTech-Lignin Technology Research Group, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
- Division of Biochemical Technology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkuntien, Bangkok 10150, Thailand
| |
Collapse
|
43
|
Hirakawa T, Taniuchi M, Iguchi Y, Bogahawaththa S, Yoshitake K, Werellagama S, Uemura T, Tsujita T. NF-E2-related factor 1 suppresses the expression of a spermine oxidase and the production of highly reactive acrolein. Sci Rep 2025; 15:12405. [PMID: 40258928 PMCID: PMC12012012 DOI: 10.1038/s41598-025-96388-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/27/2025] [Indexed: 04/23/2025] Open
Abstract
Polyamines (putrescine, spermidine, and spermine) are among the most abundant intracellular small molecular metabolites, with concentrations at the mM level. The ratios of these three molecules remain constant under physiological conditions. Stress (i.e. polyamine overload, oxidative stress, aging, infection, etc.) triggers the catabolic conversion of spermine to spermidine, ultimately yielding acrolein and hydrogen peroxide. The potential of acrolein to induce DNA damage and protein denaturation is 1,000 times greater than that of reactive oxygen species. We have shown that these polyamine metabolic pathways also involve the nuclear factor erythroid-2-related factor 1 (NRF1) transcription factor. In our chemically-inducible, liver-specific Nrf1-knockout mice, the polyamine catabolic pathway dominated the anabolic pathway, producing free acrolein and accumulating acrolein-conjugated proteins in vivo. This metabolic feature implicates SMOX as an important causative enzyme. Chromatin immunoprecipitation and reporter assays confirmed that NRF1 directly suppressed Smox expression. This effect was also observed in vitro. Ectopic overexpression of SMOX increased the accumulation of free acrolein and acrolein-conjugated proteins. SMOX knockdown reversed the accumulation of free acrolein and acrolein-conjugated proteins. Our results show that NRF1 typically suppresses Smox expression when NRF1 is downregulated, SMOX is upregulated, and polyamine metabolic pathways are altered, producing low molecular weight polyamines and acrolein.
Collapse
Affiliation(s)
- Tomoaki Hirakawa
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Megumi Taniuchi
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Yoko Iguchi
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Sudarma Bogahawaththa
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Kiko Yoshitake
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Shanika Werellagama
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
| | - Takeshi Uemura
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Tadayuki Tsujita
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan.
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
44
|
Sun H, Xu C, Xiong Z, Liu M, Ning X, Zhuang Y. Therapeutic prospects and potential mechanisms of Prdx6: as a novel target in musculoskeletal disorders. Front Physiol 2025; 16:1524100. [PMID: 40313876 PMCID: PMC12043587 DOI: 10.3389/fphys.2025.1524100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
With the global population aging, musculoskeletal disorders (MSDs) have posed significant physical and psychological health challenges for patients as well as a substantial economic burden on society. The advancements in conservative and surgical interventions for MSDs have been remarkable in recent years; however, the current treatment modalities still fall short of meeting the optimal requirements of patients. Recently, peroxiredoxin 6 (Prdx6) has gained considerable attention from researchers due to its remarkable antioxidative, anti-inflammatory, and anti-apoptotic properties. It has been found that Prdx6 is involved in multiple system diseases, including MSDs; however, the exact role of Prdx6 in MSDs is still lacking. This study aimed to summarize the structure, regulatory mechanism, and potential function of Prdx6. These findings may demonstrate Prdx6 as a novel target for inhibiting the advancement of MSDs.
Collapse
Affiliation(s)
- Hong Sun
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Chao Xu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Zhilin Xiong
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xu Ning
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yong Zhuang
- Department of Orthopaedics, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
45
|
Urbano T, Wise LA, Fiore G, Vinceti M, Filippini T. Effects of Selenium Administration on Blood Lipids: A Systematic Review and Dose-Response Meta-Analysis of Experimental Human Studies. Nutr Rev 2025:nuaf049. [PMID: 40243093 DOI: 10.1093/nutrit/nuaf049] [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] [Indexed: 04/18/2025] Open
Abstract
CONTEXT Overexposure to the essential trace element selenium has been associated with adverse metabolic and cardiovascular outcomes, hypertension, and diabetes. However, dose-response meta-analyses analyzing the effects of selenium administration on the lipid profile in experimental human studies are lacking. OBJECTIVE Through a restricted cubic spline regression meta-analysis, the dose-response relation between the dose of selenium administered or blood selenium concentrations at the end of the trials and changes over time in blood lipids, ie, total, high-density lipoprotein (HDL) and low-density lipoprotein (LDL) cholesterol, and triglycerides was assessed. DATA SOURCES Searches were performed on PubMed, Web of Science, Embase, and the Cochrane Library from inception up to January 11, 2025 to identify randomized controlled trials (RCTs) investigating the impact of selenium supplementation on blood lipid profiles among adults. DATA EXTRACTION A total of 27 eligible RCTs that enrolled healthy individuals, pregnant individuals, and participants with specific health conditions were identified and the relevant data was extracted. DATA ANALYSIS Dose-response analysis indicated that selenium administration at and above 200 µg/day decreased HDL and LDL cholesterol and increased triglyceride levels. Blood selenium concentrations at the end of the trial above approximately 150 µg/L were positively associated with triglyceride and LDL cholesterol concentrations, and inversely associated with HDL cholesterol. Inorganic selenium supplementation showed stronger associations than organic selenium. At the lowest levels of baseline intake, selenium supplementation appeared instead to have beneficial effects on the lipid profile, with an overall indication of U-shaped curves, apart from HDL-cholesterol. The adverse effects of selenium were stronger in studies involving healthy participants as compared with unhealthy participants and pregnant females, in those having a longer duration of the intervention, particularly more than 3 months, and in European populations at selenium intake levels of above 300 µg/day. CONCLUSIONS In this dose-response meta-analysis of experimental human studies, an adverse effect of selenium administration on blood lipids at levels around or above the current upper level of intake was observed. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration No. CRD42022380432.
Collapse
Affiliation(s)
- Teresa Urbano
- Environmental, Genetic and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Medical School-University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Lauren A Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, United States
| | - Gianluca Fiore
- Environmental, Genetic and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Medical School-University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Marco Vinceti
- Environmental, Genetic and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Medical School-University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department of Epidemiology, Boston University School of Public Health, Boston, MA 02118, United States
| | - Tommaso Filippini
- Environmental, Genetic and Nutritional Epidemiology Research Center (CREAGEN), Department of Biomedical, Metabolic and Neural Sciences, Medical School-University of Modena and Reggio Emilia, 41125 Modena, Italy
- School of Public Health, University of California Berkeley, Berkeley, CA 94704, United States
| |
Collapse
|
46
|
Kobayashi N, Okazaki Y, Iwane A, Hara K, Horikoshi M, Awazawa M, Soeda K, Matsushita M, Sasako T, Yoshimura K, Itoh N, Kobayashi K, Seto Y, Yamauchi T, Aburatani H, Blüher M, Kadowaki T, Ueki K. Activin B improves glucose metabolism via induction of Fgf21 and hepatic glucagon resistance. Nat Commun 2025; 16:3678. [PMID: 40246973 PMCID: PMC12006358 DOI: 10.1038/s41467-025-58836-w] [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: 08/07/2024] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
Orchestrated hormonal interactions in response to feeding and fasting play a pivotal role in regulating glucose homeostasis. Here, we show that in obesity, the production of follistatin-like 3 (FSTL3), an endogenous inhibitor of Activin B, in adipose tissue is increased in both mice and humans. The knockdown of FSTL3 improves insulin sensitivity and glucose tolerance in diabetic obese db/db mice. Notably, the overexpression of Activin B, a member of the TGFβ superfamily that is induced in liver sinusoidal endothelial cells by fasting, exerts multiple metabolically beneficial effects, including improvement of insulin sensitivity, suppression of hepatic glucose production, and enhancement of glucose-stimulated insulin secretion, all of which are attenuated by the overexpression of FSTL3. Activin B increases insulin sensitivity and reduces fat by inducing fibroblast growth factor 21 (FGF21) while suppressing glucagon action in the liver by increasing phosphodiesterase 4 B (PDE4B), leading to hepatic glucagon resistance and resultant hyperglucagonemia. Activin B-induced hyperglucagonemia enhances glucose-stimulated insulin secretion by stimulating glucagon-like peptide-1 (GLP-1) receptor in pancreatic β-cells. Thus, enhancing the action of Activin B which improves multiple components of the pathogenesis of diabetes may be a promising strategy for diabetes treatment.
Collapse
Affiliation(s)
- Naoki Kobayashi
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukiko Okazaki
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aya Iwane
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kazuo Hara
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Momoko Horikoshi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Motoharu Awazawa
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kotaro Soeda
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Maya Matsushita
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takayoshi Sasako
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kotaro Yoshimura
- Department of Plastic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nobuyuki Itoh
- Department of Genetic Biochemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Aichi, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Research Center for Advanced Science and Technology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Kohjiro Ueki
- Department of Molecular Diabetic Medicine, Diabetes Research Center, National Center for Global Health and Medicine, Tokyo, Japan.
- Department of Molecular Diabetology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
47
|
Lei J, Chen W, Gu Y, Lv X, Kang X, Jiang X. Ferroptosis regulation by traditional chinese medicine for ischemic stroke intervention based on network pharmacology and data mining. PLoS One 2025; 20:e0321751. [PMID: 40238820 PMCID: PMC12002469 DOI: 10.1371/journal.pone.0321751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/27/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVE The aim of this study is to use network pharmacology and data mining to explore the role of traditional Chinese medicine (TCM) in ischemic stroke (IS) intervention by ferroptosis regulation. The results will provide reference for related research on ferroptosis in IS. METHODS The ferroptosis-related targets were obtained from the GeneCards, GeneCLiP3, and FerrDdb databases, while the IS targets were sourced from the GeneCards and DisGeNET databases. Venny was used to identify IS targets associated with ferroptosis. A protein-protein interaction (PPI) analysis was then conducted, and machine learning screening was used to validate these potential targets. The potential targets that met specific criteria and their related compounds allowed us to select TCMs. A mechanistic analysis of the potential targets was conducted using the DAVID database. PPI network diagrams, target-compound network diagrams, and target-compound-TCM network diagrams were then constructed. Finally, molecular docking technology was used to verify the binding activities of the TCM compounds and core components with the identified targets. In addition, the properties, flavors, meridian tropism, and therapeutic effects of the candidate TCMs were analyzed and statistically evaluated. RESULTS A total of 706 targets associated with ferroptosis in IS were obtained, and 14 potential ferroptosis targets in IS were obtained using machine learning. Furthermore, 413 compounds and 301 TCMs were screened, and the binding activities of the targets to the TCM compounds and the core prescriptions were stable. The candidate TCMs primarily exhibited cold, warm, bitter taste, pungent taste, liver meridian, heat-cleaning medicinal, and tonify deficiency properties. CONCLUSIONS This study investigated ferroptosis regulation for IS intervention using TCM. We began by investigating the targets of IS and ferroptosis, and we also analyzed the relevant mechanism of ferroptosis in IS. The results of this study provide reference for related research on ferroptosis in IS.
Collapse
Affiliation(s)
- Jun Lei
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yaodong Gu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xueyan Lv
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingyu Kang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xicheng Jiang
- Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
48
|
Direksunthorn T, T Ahmed A, Pluetrattanabha N, Uthirapathy S, Ballal S, Singh A, Al-Hetty HRAK, Devi A, Sharma GC, Yumashev A. Ferroptosis in immune chaos: Unraveling its impact on disease and therapeutic potential. J Physiol Biochem 2025:10.1007/s13105-025-01078-7. [PMID: 40237936 DOI: 10.1007/s13105-025-01078-7] [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: 11/28/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025]
Abstract
Since its introduction in 2012, ferroptosis has garnered significant attention from researchers over the past decade. Unlike autophagy and apoptosis, ferroptosis is an atypical iron-dependent programmed cell death that falls under necrosis. It is regulated by various cellular metabolic and signaling processes, which encompass amino acid, lipid, iron, and mitochondrial metabolism. The initiation of ferroptosis occurs through iron-dependent phospholipid peroxidation. Notably, ferroptosis exhibits a dual effect and is associated with various diseases. A significant challenge lies in managing autoimmune disorders with unknown origins that stem from the reactivation of the immune system. Two contributing factors to autoimmunity are the aberrant stimulation of cell death and the inadequate clearance of dead cells, which can expose or release intracellular components that activate the immune response. Ferroptosis is distinct from other forms of cell death, such as apoptosis, necroptosis, autophagy, and pyroptosis, due to its unique morphological, biochemical, and genetic characteristics and specific relationship with cellular iron levels. Recent studies indicate that immune cells can both induce and undergo ferroptosis. To better understand how ferroptosis influences immune responses and its imbalance in disease, a molecular understanding of the relationship between ferroptosis and immunity is essential. Consequently, further research is needed to develop immunotherapeutics that target ferroptosis. This review primarily focuses on the role of ferroptosis in immune-related disorders.
Collapse
Affiliation(s)
| | | | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | | | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
49
|
Akagawa M, Sugasawa K, Ura K, Sassa A. Impact of an oxidative RNA lesion on in vitro replication catalyzed by SARS-CoV-2 RNA-dependent RNA polymerase. J Biol Chem 2025:108512. [PMID: 40250563 DOI: 10.1016/j.jbc.2025.108512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/02/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
The production of reactive oxygen species in response to RNA virus infection results in the oxidation of viral genomic RNA within infected cells. These oxidative RNA lesions undergo replication catalyzed by the viral replisome. G to U transversion mutations are frequently observed in the SARS-CoV-2 genome and may be linked to the replication process catalyzed by RNA-dependent RNA polymerase (RdRp) past the oxidative RNA lesion 7,8-dihydro-8-oxo-riboguanosine (8-oxo-rG). To better understand the mechanism of viral RNA mutagenesis, it is crucial to elucidate the role of RdRp in replicating across oxidative lesions. In this study, we investigated the RNA synthesis catalyzed by the reconstituted SARS-CoV-2 RdRp past a single 8-oxo-rG. The RdRp-mediated primer extension was significantly inhibited by 8-oxo-rG on the template RNA. A steady-state multiple-turnover reaction demonstrated that the turnover rate of RdRp was significantly slow when replication was blocked by 8-oxo-rG, reflecting low bypass efficiency even with prolonged reaction time. Once RdRp was able to bypass 8-oxo-rG, it preferentially incorporated rCMP, with a lesser amount of rAMP opposite 8-oxo-rG. In contrast, RdRp demonstrated greater activity in extending from the mutagenic rA:8-oxo-rG terminus compared to the lower efficiency of extension from the rC:8-oxo-rG pair. Based on steady-state kinetic analyses for the incorporation of rNMPs opposite 8-oxo-rG and chain extension from rC:8-oxo-rG or rA:8-oxo-rG, the relative bypass frequency for rA:8-oxo-rG was found to be seven-fold higher than that for rC:8-oxo-rG. Therefore, the properties of RdRp indicated in this study may contribute to the mechanism of mutagenesis of the SARS-CoV-2 genome.
Collapse
Affiliation(s)
- Masataka Akagawa
- Department of Biology, Graduate School of Science, Chiba University, Chiba, 263-8522, Japan
| | - Kaoru Sugasawa
- Biosignal Research Center, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, 657-8501, Japan
| | - Kiyoe Ura
- Department of Biology, Graduate School of Science, Chiba University, Chiba, 263-8522, Japan
| | - Akira Sassa
- Department of Biology, Graduate School of Science, Chiba University, Chiba, 263-8522, Japan.
| |
Collapse
|
50
|
Zhang T, Pan Y, Sawa T, Akaike T, Matsunaga T. Supersulfide donors and their therapeutic targets in inflammatory diseases. Front Immunol 2025; 16:1581385. [PMID: 40308575 PMCID: PMC12040673 DOI: 10.3389/fimmu.2025.1581385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Inflammation is one defense mechanism of the body that has multiple origins, ranging from physical agents to infectious agents including viruses and bacteria. The resolution of inflammation has emerged as a critical endogenous process that protects host tissues from prolonged or excessive inflammation, which can become chronic. Failure of the inflammation resolution is a key pathological mechanism that drives the progression of numerous inflammatory diseases. Owing to the various side effects of currently available drugs to control inflammation, novel therapeutic agents that can prevent or suppress inflammation are needed. Supersulfides are highly reactive and biologically potent molecules that function as antioxidants, redox regulators, and modulators of cell signaling. The catenation state of individual sulfur atoms endows supersulfides with unique biological activities. Great strides have recently been made in achieving a molecular understanding of these sulfur species, which participate in various physiological and pathological pathways. This review mainly focuses on the anti-inflammatory effects of supersulfides. The review starts with an overview of supersulfide biology and highlights the roles of supersulfides in both immune and inflammatory responses. The various donors used to generate supersulfides are assessed as research tools and potential therapeutic agents. Deeper understanding of the molecular and cellular bases of supersulfide-driven biology can help guide the development of innovative therapeutic strategies to prevent and treat diseases associated with various immune and inflammatory responses.
Collapse
Affiliation(s)
- Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
| | - Yuexuan Pan
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Akaike
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| |
Collapse
|