1
|
Zheng Q, Wang D, Lin R, Xu W. Pyroptosis, ferroptosis, and autophagy in spinal cord injury: regulatory mechanisms and therapeutic targets. Neural Regen Res 2025; 20:2787-2806. [PMID: 39101602 PMCID: PMC11826477 DOI: 10.4103/nrr.nrr-d-24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 04/24/2024] [Accepted: 06/07/2024] [Indexed: 08/06/2024] Open
Abstract
Regulated cell death is a form of cell death that is actively controlled by biomolecules. Several studies have shown that regulated cell death plays a key role after spinal cord injury. Pyroptosis and ferroptosis are newly discovered types of regulated cell deaths that have been shown to exacerbate inflammation and lead to cell death in damaged spinal cords. Autophagy, a complex form of cell death that is interconnected with various regulated cell death mechanisms, has garnered significant attention in the study of spinal cord injury. This injury triggers not only cell death but also cellular survival responses. Multiple signaling pathways play pivotal roles in influencing the processes of both deterioration and repair in spinal cord injury by regulating pyroptosis, ferroptosis, and autophagy. Therefore, this review aims to comprehensively examine the mechanisms underlying regulated cell deaths, the signaling pathways that modulate these mechanisms, and the potential therapeutic targets for spinal cord injury. Our analysis suggests that targeting the common regulatory signaling pathways of different regulated cell deaths could be a promising strategy to promote cell survival and enhance the repair of spinal cord injury. Moreover, a holistic approach that incorporates multiple regulated cell deaths and their regulatory pathways presents a promising multi-target therapeutic strategy for the management of spinal cord injury.
Collapse
Affiliation(s)
- Qingcong Zheng
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Du Wang
- Arthritis Clinical and Research Center, Peking University People’s Hospital, Beijing, China
| | - Rongjie Lin
- Department of Orthopedic Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
| | - Weihong Xu
- Department of Spinal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
2
|
Hai E, Bai X, Song Y, Li B, Zhang J, Zhang J. Correlation between PRDX6 levels and freezability properties of sheep semen. Theriogenology 2025; 243:117461. [PMID: 40318452 DOI: 10.1016/j.theriogenology.2025.117461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 03/24/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Individual variations in the freezability properties of sheep semen may be related to the antioxidant levels of sperm. PRDX6 is a key antioxidant protein in sperm, but its relationship with the freezability properties of sheep semen remains unclear. This study, using Hu sheep as the research subject, investigated the levels of PRDX6 in sperm with different freezability properties. A positive correlation was found between the levels of PRDX6 and sperm freezability. Furthermore, when different concentrations of PRDX6 recombinant protein were added to high-freezing-resistant (HF) and low-freezing-resistant (LF) sperm, it was revealed that 20 μg/ml of PRDX6 recombinant protein significantly improved various sperm quality parameters in both HF and LF sperm and reduced the differences in sperm freezability properties. Finally, adding a PRDX6 peroxidase activity inhibitor (Ethacrynic acid) or a calcium - independent phospholipase A2 (iPLA2) activity inhibitor (MJ33) to the diluent significantly reduced the freezability parameters of sperm. This indicates that PRDX6 protects sperm from freezing damage through two pathways. In summary, this study reveals the crucial role of PRDX6 in the freezability properties of sheep semen. It suggests that increasing the levels of PRDX6 in sperm may be an effective strategy to improve the quality of cryopreserved semen. Future research can further explore the molecular mechanisms regulating the levels of PRDX6, providing a scientific basis for the optimization of sheep semen cryopreservation techniques.
Collapse
Affiliation(s)
- Erhan Hai
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Xue Bai
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China; Hinggan League Agriculture and Animal Husbandry Science Institute, Ulanhot, 137400, Inner Mongolia, China
| | - Yukun Song
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Boyuan Li
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Jian Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China
| | - Jiaxin Zhang
- Inner Mongolia Key Laboratory of Sheep & Goat Genetics Breeding and Reproduction, College of Animal Science, Inner Mongolia Agricultural University, Hohhot, 010018, Inner Mongolia, China.
| |
Collapse
|
3
|
Zhu Z, Li W, Ding K, Bastawy EM, Kamel AM, Kou X, Min L. Ellagic acid maintains post-thaw goat sperm quality via protecting mitochondrial function from ROS damage. Cryobiology 2025; 119:105231. [PMID: 40132303 DOI: 10.1016/j.cryobiol.2025.105231] [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: 03/02/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025]
Abstract
This study aimed to investigate the effects of ellagic acid (EA), an antioxidant, on goat sperm quality after freezing and thawing. Goat semen was frozen using Tris-citric acid-glucose (TCG) extender containing 0, 1.25, 2.5, 5, and 10 μM of EA. Egg yolk represented 20 % (v/v) and glycerol represented 5 % (v/v) of the extender's final concentration. Goat sperm post-thaw motility, acrosome integrity, plasma membrane integrity, mitochondrial activity, ATP content, NADH/NAD+ levels, and NADH-CoQ activity were evaluated. Moreover, to elucidate how EA enhanced the goat sperm characteristics, the post-thaw sperm mitochondrial reactive oxygen species (ROS) level, malondialdehyde (MDA) level, oxidative DNA damage, apoptosis, levels of NADH dehydrogenase 1 (MT-ND1) and NADH dehydrogenase 6 (MT-ND6) proteins, and the 4-hydroxynonenal (4-HNE) level were also measured after thawing. The results demonstrated that motility, plasma membrane integrity, and acrosome integrity rates were enhanced in the group treated with 5 μM of EA compared to the other concentrations (0 μM, 1.25 μM, 2.5 μM, 5, and 10 μM). Moreover, mitochondrial activity and ATP content were notably superior in the 5 μM EA group compared to all other treatment groups, along with a considerable decrease in ROS and MDA levels. The 4-HNE level and oxidative DNA damage in sperm were also reduced by EA supplementation. Additionally, it was found that EA (5 μM) significantly (p < 0.05) decreased sperm apoptosis levels. Furthermore, the addition of 5 μM EA maintained the post-thaw sperm MT-ND1 and MT-ND6 levels and reduced the negative impact of ROS on MT-ND1 and MT-ND6, thereby sustaining mitochondrial function for ATP generation. These results suggest that ellagic acid supplementation could maintain goat post-thaw sperm quality by reducing ROS damage and maintaining mitochondrial function for ATP generation. Antioxidant treatments, such as ellagic acid are a useful tool for maintaining frozen-thawed sperm quality.
Collapse
Affiliation(s)
- Zhendong Zhu
- College of Animal Science and Technology, Qingdao Agricultural University, No.700 Changcheng Road, Qingdao, 266109, China
| | - Wenjia Li
- College of Animal Science and Technology, Qingdao Agricultural University, No.700 Changcheng Road, Qingdao, 266109, China
| | - Kexin Ding
- College of Animal Science and Technology, Qingdao Agricultural University, No.700 Changcheng Road, Qingdao, 266109, China
| | - Eslam M Bastawy
- Faculty of Science, Ain Shams University, Cairo, Egypt; Faculty of Health, School of Medicine, Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong 3216, Australia
| | | | - Xin Kou
- Hongde Livestock Farm, Yingli Town, Weifang, 261000, China
| | - Lingjiang Min
- College of Animal Science and Technology, Qingdao Agricultural University, No.700 Changcheng Road, Qingdao, 266109, China.
| |
Collapse
|
4
|
Menail HA, Robichaud S, Cormier R, Blanchard A, Hunter-Manseau F, Léger A, Lamarre SG, Pichaud N. Can Ahiflower® (Buglossoides arvensis) seed-oil supplementation help overcome the adverse effects of imidacloprid in honey bees? Comp Biochem Physiol C Toxicol Pharmacol 2025; 296:110238. [PMID: 40436292 DOI: 10.1016/j.cbpc.2025.110238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 05/18/2025] [Accepted: 05/24/2025] [Indexed: 06/02/2025]
Abstract
In this study, we investigated the effects of nutritional supplementation as a strategy to mitigate the impacts of imidacloprid (neonicotinoid) on honey bees by using Ahiflower® (Buglossoides arvensis) seed-oil. This oil is rich in stearidonic-acid (SDA, 18:4n3), which is a precursor to eicosapentaenoic-acid (EPA) and docosahexaenoic-acid (DHA) that are known for their beneficial and protective effects. Specifically, we chronically fed newly emerged worker bees with sucrose syrup and pollen patties (control) that we supplemented with (i) imidacloprid (0.375 ng·μl-1), (ii) Ahiflower® oil (5 %) + imidacloprid (0.375 ng·μl-1), and (iii) Ahiflower® oil (5 %). Survival was recorded, and after 21 days, worker bees were sampled to measure mitochondrial respiration, ATP5A1 content, adenylate energy charge, lipid peroxidation in thorax as well as fatty acid composition and peroxidation index in whole bees. Our results indicate that (i) imidacloprid mostly hampers mitochondria, increases saturated fatty acids and decreases survival, (ii) oxidation of alternative substrates allows full recovery of mitochondrial respiration in the imidacloprid-treated group demonstrating mitochondrial flexibility, (iii) Ahiflower® oil in combination with imidacloprid partially restores mitochondrial respiration at the level of complexes I and II, restores fatty acid composition but fails to restore survival. These findings confirm the deleterious effects of imidacloprid on mitochondria while highlighting, for the first time, the potential benefits of Ahiflower® oil in mitochondrial function, though not on honey bee survival. In addition, this study highlights the importance of mitochondrial flexibility when organisms are exposed to toxicants at environmentally relevant levels.
Collapse
Affiliation(s)
- Hichem A Menail
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada.
| | - Samuel Robichaud
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Robert Cormier
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Arianne Blanchard
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Florence Hunter-Manseau
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Adèle Léger
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Simon G Lamarre
- Department of Biology, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Nicolas Pichaud
- New Brunswick Centre for Precision Medicine, Moncton, NB E1C8X3, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
5
|
Mao M, Zhang Y, Liu H, Jiang X, Zhao Y, Liu Q, Niu Z, Zhang X. Identification of a panel of volatile organic compounds in urine for early detection of for bladder cancer. Sci Rep 2025; 15:17383. [PMID: 40389471 PMCID: PMC12089490 DOI: 10.1038/s41598-025-01324-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 05/05/2025] [Indexed: 05/21/2025] Open
Abstract
The absence of specific markers makes early detection of bladder cancer (BC) challenging. Recent studies have reported the reliable diagnostic significance of volatile organic compounds (VOCs) in several cancers. This study aimed to investigate whether urinary VOCs could serve as potential biomarkers for BC. A total of 89 BC patients and 67 healthy individuals were recruited for this study. VOCs in urine samples were detected using gas chromatography-ion mobility spectrometry (GC-IMS). Machine learning algorithms were used to establish diagnostic models for predicting BC based on differential expressed VOCs. Compared with healthy individuals, A total of 20 differentially expressed VOCs were identified, including 17 upregulated and 3 downregulated. Among five machine-learning algorithms, the Random Forests (RF) provided the highest accuracy. Based on RF analysis, eight important VOCs (2-Undecenal, 2-propanone, 1-octanal, Ethyl 2-hydroxybenzoate, 2-ethyl hexanol-D, 2-Pentanone, 1-Propanol,3-(methylthio), 1-nonanal) were identified and used to constructed a diagnostic model, showing the highest diagnostic accuracy and area under the curve (AUC) of 91.5% and 0.958, respectively. Our results suggest a VOCs panel might be used as novel biomarker for early detection of BC.
Collapse
Affiliation(s)
- Mai Mao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yanli Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Haibo Liu
- Yidu Central Hospital of Weifang, Weifang, 262500, China
| | - Xiumei Jiang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yuxiao Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Qi Liu
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, 250031, China
| | - Zhongfang Niu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
6
|
Yang P, Su W, Wang L, Xu F, Kong Y, Long J. From aldehyde metabolism to delay aging: targeting ALDH2 as a novel strategy. Free Radic Biol Med 2025; 236:70-86. [PMID: 40349798 DOI: 10.1016/j.freeradbiomed.2025.05.389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/19/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Aldehydes are molecules that are commonly found in both human physiology and the environment. The accumulation of these substances can lead to the cross-linking of intracellular DNA and proteins, thereby disrupting cellular function and contributing to the processes of premature aging and age-related diseases. Aldehyde dehydrogenase 2 (ALDH2), the key member of ALDH family, is an enzyme responsible for aldehyde metabolism, composed of four identical subunits located within the mitochondrial matrix. Its primary role is to catalyze the oxidation of aldehydes, resulting in the formation of their corresponding acid metabolites. This paper presents a succinct overview of the sources and metabolic pathways of key aldehydes within the human body, compares the various primary enzymes involved in aldehyde metabolism, and explores the structural and functional characteristics of ALDH2. Furthermore, ALDH2 is proposed as a potential therapeutic target for addressing aging and associated diseases. The discussion also includes prospective research avenues, particularly focusing on ALDH2 agonists and aldehyde scavengers designed to enhance the clearance of reactive aldehydes and safeguard cellular functions, thereby mitigating aldehyde-induced cellular damage and potentially delaying the aging process.
Collapse
Affiliation(s)
- Peng Yang
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Wu Su
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Lizhuo Wang
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Fanding Xu
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Yu Kong
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China
| | - Jiangang Long
- Xi'an Key Laboratory of Aging Biology, Institude of Mitochondrial Biology and Medicine, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710116, China.
| |
Collapse
|
7
|
Dai JZ, Hsu WJ, Lin MH, Shueng PW, Lee CC, Yang CC, Lin CW. YAP-mediated DDX3X confers resistance to ferroptosis in breast cancer cells by reducing lipid peroxidation. Free Radic Biol Med 2025; 232:330-339. [PMID: 40089076 DOI: 10.1016/j.freeradbiomed.2025.03.019] [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: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
Metabolic shifts in cancer cells were found to participate in tumorigenesis, especially driving chemotherapeutic resistance. Ferroptosis is a newly discovered form of cell death induced by excessive accumulations of iron and lipid peroxidation. Susceptibility to ferroptosis can be intrinsically regulated by various cellular metabolic pathways. Therefore, inducing ferroptosis might be a promising anticancer therapeutic strategy. DEAD-box helicase 3 X-linked (DDX3X), a critical modulator of RNA metabolism, was identified as an oncogene in breast cancer and also participates in cancer metabolism and chemotherapeutic resistance. However, the molecular regulation of the association between DDX3X and ferroptosis is largely unknown. Herein, we investigated the correlation between resistance to ferroptosis and DDX3X expression in breast cancer cells. We found that elevation of DDX3X was associated with increased resistance to a ferroptosis inducer in breast cancer cells, and manipulating DDX3X expression regulated the sensitivity to the ferroptosis inducer. Importantly, DDX3X upregulated expression of the anti-ferroptotic enzyme glutathione peroxidase 4 (GPX4) gene to confer ferroptosis resistance in breast cancer cells. Moreover, DDX3X was transcriptionally upregulated by the yes-associated protein (YAP). Knockdown of YAP downregulated DDX3X mRNA expression and facilitated lipid peroxidation, but that were restored in the presence of DDX3X. Clinically, coexpression of DDX3X and YAP was found in a variety of malignancy, and their elevation conferred poor survival prognosis in patients with breast cancer. Together, our findings reveal the crucial role of DDX3X in sensitivity to ferroptosis and underscore its potential as a diagnostic marker and therapeutic target. DDX3X renders resistance to ferroptosis and plays a role in mitigating lipid peroxidation, paving the way for therapeutic vulnerability via targeting cancer metabolism.
Collapse
Affiliation(s)
- Jia-Zih Dai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Jing Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Mei-Hsiang Lin
- Graduate Institute of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chi-Ching Lee
- Istanbul Sabahattin Zaim University, Faculty of Engineering and Natural Sciences, Department of Food Engineering, Istanbul, Turkey; Department of Food Technology and Nutrition, Faculty of Technologies, Klaipėda State University of Applied Sciences, Lithuania
| | - Ching-Chieh Yang
- Department of Radiation Oncology, Chi Mei Medical Center, Tainan, Taiwan; Department of Pharmacy, Chia-Nan University of Pharmacy and Science, Tainan, Taiwan; School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan.
| | - Cheng-Wei Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
8
|
Uti DE, Atangwho IJ, Alum EU, Ntaobeten E, Obeten UN, Bawa I, Agada SA, Ukam CIO, Egbung GE. Antioxidants in cancer therapy mitigating lipid peroxidation without compromising treatment through nanotechnology. DISCOVER NANO 2025; 20:70. [PMID: 40272665 PMCID: PMC12021792 DOI: 10.1186/s11671-025-04248-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/03/2025] [Indexed: 04/27/2025]
Abstract
BACKGROUND Cancer treatments often exploit oxidative stress to selectively kill tumour cells by disrupting their lipid peroxidation membranes and inhibiting antioxidant enzymes. However, lipid peroxidation plays a dual role in cancer progression, acting as both a tumour promoter and a suppressor. Balancing oxidative stress through antioxidant therapy remains a challenge, as excessive antioxidant activity may compromise the efficacy of chemotherapy and radiotherapy. AIM This review explores the role of antioxidants in mitigating lipid peroxidation in cancer therapy while maintaining treatment efficacy. It highlights recent advancements in nanotechnology-based targeted antioxidant delivery to optimize therapeutic outcomes. METHODS A comprehensive literature review was conducted using reputable databases, including PubMed, Scopus, Web of Science, and ScienceDirect. The search focused on publications from the past five years (2020-2025), supplemented by relevant studies from earlier years. Keywords such as "antioxidants," "lipid peroxidation," "nanotechnology in cancer therapy," and "oxidative stress" were utilized. Relevant articles were critically analysed, and graphical illustrations were created. RESULTS Emerging evidence suggests that nanoparticles, including liposomes, polymeric nanoparticles, metal-organic frameworks, and others, can effectively encapsulate and control the release of antioxidants in tumour cells while minimizing systemic toxicity. Stimuli-responsive carriers with tumour-specific targeting mechanisms further enhance antioxidant delivery. Studies indicate that these strategies help preserve normal cells, mitigate oxidative stress-related damage, and improve treatment efficacy. However, challenges such as bioavailability, stability, and potential interactions with standard therapies remain. CONCLUSION Integrating nanotechnology with antioxidant-based interventions presents a promising approach for optimizing cancer therapy. Future research should focus on refining lipid peroxidation modulation strategies, assessing oxidative stress profiles during treatment, and employing biomarkers to determine optimal antioxidant dosing. A balanced approach to antioxidant use may enhance therapeutic efficacy while minimizing adverse effects.
Collapse
Affiliation(s)
- Daniel Ejim Uti
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda.
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria.
| | - Item Justin Atangwho
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Esther Ugo Alum
- Department of Biochemistry, Research and Publications, Kampala International University, P.O. Box 20000, Kampala, Uganda
| | - Emmanuella Ntaobeten
- Department of Cancer and Haematology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Uket Nta Obeten
- Department of Chemistry/Biochemistry and Molecular Biology, Alex Ekwueme Federal University, Ndufu-Alike Ikwo, PMB 1010, Abakaliki, Ebonyi State, Nigeria
| | - Inalegwu Bawa
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | - Samuel A Agada
- Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Otukpo, Benue State, Nigeria
| | | | - Godwin Eneji Egbung
- Department of Biochemistry, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| |
Collapse
|
9
|
Shi Y, Zhao Y, Sun SJ, Lan XT, Wu WB, Zhang Z, Chen YX, Yan YY, Xu YP, Li DJ, Fu H, Shen FM. Targeting GPX4 alleviates ferroptosis and retards abdominal aortic aneurysm formation. Biochem Pharmacol 2025; 234:116800. [PMID: 39952331 DOI: 10.1016/j.bcp.2025.116800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/16/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Abdominal aortic aneurysm (AAA) is a potentially fatal cardiovascular disease, closely related to inflammation and loss of vascular smooth muscle cells (VSMCs). Ferroptosis is an iron-dependent cell death associated with peroxidation of lipids. However, the direct role of glutathione peroxidase 4 (GPX4) itself determined ferroptosis in the course of AAA pathogenesis remains unknown. Here, we reported that ferroptosis was triggered in human AAA, elastase- and angiotensin II (Ang II)-induced mouse AAA, and Ang II-incubated VSMCs. Inhibition of ferroptosis via global genetic overexpression of GPX4, a critical anti-ferroptosis molecule, markedly prevented both vascular remodeling and inflammatory response. Mechanistically, GPX4 changed the migration and activation of macrophages/monocytes in AAA tissues in mice. Experiments in vitro demonstrated that overexpression of GPX4 prevented the JAK1/STAT3 signaling activation in VSMCs induced by IL-6, production of pro-inflammatory macrophages. Finally, the role of ferroptosis was confirmed on an Ang II-induced mice AAA model. These results emphasized the significance of ferroptosis in AAA, and provided novel insights that therapy focusing on GPX4 might be a promising strategy for treatment of AAA in the clinic.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/physiology
- Animals
- Aortic Aneurysm, Abdominal/prevention & control
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Phospholipid Hydroperoxide Glutathione Peroxidase/genetics
- Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
- Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors
- Phospholipid Hydroperoxide Glutathione Peroxidase/biosynthesis
- Humans
- Mice
- Mice, Inbred C57BL
- Male
- Angiotensin II/toxicity
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cells, Cultured
Collapse
Affiliation(s)
- Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Zhao
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Si-Jia Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiu-Ting Lan
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wen-Bin Wu
- Department of Pharmacology, School of Pharmacy, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Xin Chen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu-Ying Yan
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yu-Ping Xu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Hui Fu
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Kunishige R, Noguchi Y, Okamoto N, Li L, Ono A, Murata M, Kano F. Protein covariation networks for elucidating ferroptosis inducer mechanisms and potential synergistic drug targets. Commun Biol 2025; 8:480. [PMID: 40164758 PMCID: PMC11958834 DOI: 10.1038/s42003-025-07886-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
In drug development, systematically characterizing a compound's mechanism of action (MoA), including its direct targets and effector proteins, is crucial yet challenging. Network-based approaches, unlike those focused solely on direct targets, effectively detect a wide range of cellular responses elicited by compounds. This study applied protein covariation network analysis, leveraging quantitative, morphological, and localization features from immunostained microscopic images, to elucidate the MoA of AX-53802, a novel ferroptosis inducer. From the candidate targets extracted through network analysis, GPX4 was verified as the direct target by validation experiments. Additionally, aggregates involving GPX4, TfR1, and F-actin were observed alongside iron reduction, suggesting a ferroptosis defense mechanism. Furthermore, combination therapies targeting GPX4 and FAK/Src were found to enhance cancer cell death, and MDM2, ezrin, and cortactin were identified as potential ferroptosis inhibitor targets. These findings highlight the effectiveness of network-based approaches in uncovering a compound's MoA and developing combination therapies for cancer.
Collapse
Affiliation(s)
- Rina Kunishige
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Yoshiyuki Noguchi
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
- International Research Center for Neurointelligence, Institutes for Advanced Study, The University of Tokyo, Tokyo, Japan
| | | | - Lei Li
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Akito Ono
- Axcelead Drug Discovery Partners, Inc., Fujisawa, Kanagawa, Japan
| | - Masayuki Murata
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan
| | - Fumi Kano
- Multimodal Cell Analysis Collaborative Research Cluster, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
- Cellshoot Therapeutics, Inc., Koto-ku, Tokyo, Japan.
- Cell Biology Center, Institute of Integrated Research, Institute of Science Tokyo, Yokohama-shi, Kanagawa, Japan.
| |
Collapse
|
11
|
Li Q, Yang X, Li T. Natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in central nervous system diseases: current preclinical evidence and future perspectives. Front Pharmacol 2025; 16:1570069. [PMID: 40196367 PMCID: PMC11973303 DOI: 10.3389/fphar.2025.1570069] [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: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/09/2025] Open
Abstract
Flavonoids are a class of important polyphenolic compounds, renowned for their antioxidant properties. However, recent studies have uncovered an additional function of these natural flavonoids: their ability to inhibit ferroptosis. Ferroptosis is a key mechanism driving cell death in central nervous system (CNS) diseases, including both acute injuries and chronic neurodegenerative disorders, characterized by iron overload-induced lipid peroxidation and dysfunction of the antioxidant defense system. This review discusses the therapeutic potential of natural flavonoids from herbs and nutraceuticals as ferroptosis inhibitors in CNS diseases, focusing on their molecular mechanisms, summarizing findings from preclinical animal models, and providing insights for clinical translation. We specifically highlight natural flavonoids such as Baicalin, Baicalein, Chrysin, Vitexin, Galangin, Quercetin, Isoquercetin, Eriodictyol, Proanthocyanidin, (-)-epigallocatechin-3-gallate, Dihydromyricetin, Soybean Isoflavones, Calycosin, Icariside II, and Safflower Yellow, which have shown promising results in animal models of acute CNS injuries, including ischemic stroke, cerebral ischemia-reperfusion injury, intracerebral hemorrhage, subarachnoid hemorrhage, traumatic brain injury, and spinal cord injury. Among these, Baicalin and its precursor Baicalein stand out due to extensive research and favorable outcomes in acute injury models. Mechanistically, these flavonoids not only regulate the Nrf2/ARE pathway and activate GPX4/GSH-related antioxidant pathways but also modulate iron metabolism proteins, thereby alleviating iron overload and inhibiting ferroptosis. While flavonoids show promise as ferroptosis inhibitors for CNS diseases, especially in acute injury settings, further studies are needed to evaluate their efficacy, safety, pharmacokinetics, and blood-brain barrier penetration for clinical application.
Collapse
Affiliation(s)
- Qiuhe Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Tiegang Li
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Smith MR, Jarrell ZR, Liu KH, Lee CM, Morgan ET, Go YM, Jones DP. Redox Metabolomics of Menthol in Children's Plasma with Second-Hand Cigarette and Electronic Cigarette Exposures. ADVANCES IN REDOX RESEARCH 2025; 14:100122. [PMID: 40357186 PMCID: PMC12068848 DOI: 10.1016/j.arres.2025.100122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Background Cigarettes and electronic cigarettes generate many redox-active materials which could impact children's health through second-hand exposures. High-resolution metabolomics methods enable use of non-targeted mass spectrometry of plasma to test for redox consequences of second-hand exposures. Objectives Our objectives were to test for oxidative stress metabolites and altered metabolic pathways associated with second-hand exposure to redox-active flavorants and flavorant metabolites in plasma of infants and children. Methods Untargeted plasma metabolomics data for infants and children in a population known to include individuals with second-hand exposures to cigarettes and electronic cigarettes were analyzed for cotinine and metabolites of flavorants. A metabolome-wide association study (MWAS) was performed separately for cotinine and menthol glucuronide, derived from the redox-active flavorant, menthol. Pathway enrichment analysis was used to identify metabolic pathways, and xMWAS was used to detect metabolic communities associated with flavorant metabolites. Results Menthol glucuronide was one of several flavorant metabolites positively correlated with cotinine. MWAS and pathway enrichment analysis revealed that some pathways associated with both menthol glucuronide and cotinine, while others only associated with menthol glucuronide, including sphingolipid, glycerophospholipid, antioxidant, N-glycan and mitochondrial energy metabolism. 4-hydroxynonenal and other oxidized lipids positively correlated with menthol glucuronide. Discussion The results show that flavorants from second-hand electronic cigarette and cigarette exposures in infants and children are associated with changes in redox metabolism which are known to associate with human lung diseases.
Collapse
Affiliation(s)
- Matthew Ryan Smith
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
- Atlanta VA Healthcare System, Decatur, GA, 30033, USA
| | - Zachery R. Jarrell
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ken H Liu
- Department of Chemistry, Emory University, 1515 Dickey Drive NE, Atlanta, Georgia, 30322, USA
| | - Choon-Myung Lee
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, 30322, USA
| | - Edward T Morgan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, 30322, USA
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Dean P. Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
13
|
Berlingerio SP, Bondue T, Tassinari S, Siegerist F, Ferrulli A, Lismont C, Cairoli S, Goffredo BM, Ghesquière B, Fransen M, Endlich N, Oliveira Arcolino F, Bussolati B, van den Heuvel L, Levtchenko E. Targeting oxidative stress-induced lipid peroxidation enhances podocyte function in cystinosis. J Transl Med 2025; 23:206. [PMID: 39980044 PMCID: PMC11844038 DOI: 10.1186/s12967-024-05996-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: 05/30/2024] [Accepted: 12/15/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Cystinosis is a rare, incurable lysosomal storage disease caused by mutations in the CTNS gene encoding the cystine transporter cystinosin, which leads to lysosomal cystine accumulation in all cells of the body. Patients with cystinosis display signs of podocyte damage characterized by extensive loss of podocytes into the urine at early disease stages, glomerular proteinuria, and the development of focal segmental glomerulosclerosis (FSGS) lesions. Although standard treatment with cysteamine decreases cellular cystine levels, it neither reverses glomerular injury nor prevents the loss of podocytes. Thus, pathogenic mechanisms other than cystine accumulation are involved in podocyte dysfunction in cystinosis. METHODS We used immortalized patient-derived cystinosis, healthy, and CTNS knockdown podocytes to investigate podocyte dysfunction in cystinosis. The results were validated in our newly in-house developed fluorescent ctns-/-[Tg(fabp10a:gc-EGFP)] zebrafish larvae model. To understand impaired podocyte functionality, static and dynamic permeability assays, tracer-metabolomic analysis, flow cytometry, western blot, and chemical and dynamic redox-sensing fluorescent probes were used. RESULTS In the current study, we discovered that cystinosis podocytes demonstrate increased ferroptotic cell death caused by mitochondrial reactive oxygen species (ROS)-driven membrane lipid peroxidation. Moreover, cystinosis cells present a fragmented mitochondrial network with impaired tricarboxylic acid cycle (TCA) cycle and energy metabolism. Targeting mitochondrial ROS and lipid peroxidation improved podocyte function in vitro and rescued proteinuria in vivo in cystinosis zebrafish larvae. CONCLUSIONS Mitochondrial ROS contribute to podocyte injury in cystinosis by driving lipid peroxidation and ferroptosis, which in turn lead to podocyte detachment. This finding adds cystinosis to the list of podocytopathies associated with mitochondrial dysfunction. The identified mechanisms reveal new therapeutic targets and highlight lipid peroxidation as an exploitable vulnerability of cystinosis podocytes.
Collapse
Affiliation(s)
- Sante Princiero Berlingerio
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Tjessa Bondue
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Sarah Tassinari
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Florian Siegerist
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Angela Ferrulli
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Bari, Italy
| | - Celien Lismont
- Laboratory of Peroxisome Biology and Intracellular Communication, KU Leuven, Leuven, Belgium
| | - Sara Cairoli
- Laboratory of Metabolic Biochemistry, Department of Pediatric Medicine, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Bianca Maria Goffredo
- Laboratory of Metabolic Biochemistry, Department of Pediatric Medicine, Bambino Gesù Children's Hospital, IRCSS, Rome, Italy
| | - Bart Ghesquière
- Metabolomics Expertise Center, Department of Cellular and Molecular Medicine, VIB-KU Leuven, Leuven, Belgium
| | - Marc Fransen
- Laboratory of Peroxisome Biology and Intracellular Communication, KU Leuven, Leuven, Belgium
| | - Nicole Endlich
- Institute of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald, Germany
| | - Fanny Oliveira Arcolino
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
- Emma Center for Personalized Medicine, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - Benedetta Bussolati
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin, Italy
| | - Lambertus van den Heuvel
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Department of Pediatric Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Elena Levtchenko
- Laboratory of Pediatric Nephrology, Department of Development and Regeneration, KU Leuven, Leuven, Belgium.
- Department of Pediatric Nephrology, Emma Children's Hospital, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Thorwald MA, Godoy-Lugo JA, Kerstiens E, Garcia G, Kim M, Shemtov SJ, Silva J, Durra S, O'Day PA, Mack WJ, Hiniker A, Vermulst M, Benayoun BA, Higuchi-Sanabria R, Forman HJ, Head E, Finch CE. Down syndrome with Alzheimer's disease brains have increased iron and associated lipid peroxidation consistent with ferroptosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636731. [PMID: 39975068 PMCID: PMC11839036 DOI: 10.1101/2025.02.05.636731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
INTRODUCTION Cerebral microbleeds (MB) are associated with sporadic Alzheimer's Disease (AD) and Down Syndrome with AD (DSAD). Higher MB iron may cause iron mediated lipid peroxidation. We hypothesize that amyloid deposition is linked to MB iron and that amyloid precursor protein (APP) triplication increases iron load and lipid peroxidation. METHODS Prefrontal cortex and cerebellum of cognitively normal (CTL), AD and DSAD ApoE3,3 carriers were examined for proteins that mediated iron metabolism, antioxidant response, and amyloid processing in lipid rafts. RESULTS Iron was 2-fold higher in DSAD than CTL and AD. Iron storage proteins and lipid peroxidation were increased in prefrontal cortex, but not in the cerebellum. The glutathione synthesis protein GCLM was decreased by 50% in both AD and DSAD. Activity of lipid raft GPx4, responsible for membrane repair, was decreased by at least 30% in AD and DSAD. DISCUSSION DSAD shows greater lipid peroxidation than AD consistent with greater MBs and iron load.
Collapse
Affiliation(s)
- Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Jose A Godoy-Lugo
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Elizabeth Kerstiens
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Gilberto Garcia
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Minhoo Kim
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Sarah J Shemtov
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Justine Silva
- Department of Pathology and Laboratory Medicine, University of California, Medical Sciences, Irvine, CA 92617
| | - Salma Durra
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Peggy A O'Day
- Life and Environmental Sciences Department, University of California, 5200 N. Lake Rd., Merced, CA 95343
| | - Wendy J Mack
- Population and Public Health Sciences, Keck School of Medicine of the University of Southern California, 1975 Zonal Ave Los Angeles, CA 90033
| | - Annie Hiniker
- Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA; 1975 Zonal Ave, Los Angeles, CA 90033
| | - Marc Vermulst
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Bérénice A Benayoun
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Ryo Higuchi-Sanabria
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
| | - Henry Jay Forman
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
- School of Natural Sciences, University of California Merced, 5200 N. Lake Rd., Merced, CA 95343
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Medical Sciences, Irvine, CA 92617
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave, Los Angeles, CA 90089
- Dornsife College, University of Southern California, 3551 Trousdale Pkwy, Los Angeles, CA 90089
| |
Collapse
|
15
|
O'Flaherty C. Redox signaling regulation in human spermatozoa: a primary role of peroxiredoxins. Asian J Androl 2025:00129336-990000000-00281. [PMID: 39902615 DOI: 10.4103/aja2024126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
ABSTRACT Reactive oxygen species (ROS) play a dual role in mammalian spermatozoa. At high levels, they are detrimental to sperm function since they can promote oxidative stress that produces oxidation of protein, lipids, and sperm DNA. This oxidative damage is associated with male infertility. On the other hand, when ROS are produced at low levels, they participate in the redox signaling necessary for sperm capacitation. Capacitation-associated ROS are produced by the sperm oxidase, whose identity is still elusive, located in the plasma membrane of the spermatozoon. ROS, such as superoxide anion, hydrogen peroxide, nitric oxide, and peroxynitrite, activate protein kinases and inactivate protein phosphatases with the net increase of specific phosphorylation events. Peroxiredoxins (PRDXs), antioxidant enzymes that fight against oxidative stress, regulate redox signaling during capacitation. Among them, PRDX6, which possesses peroxidase and calcium-independent phospholipase A2 (iPLA2) activities, is the primary regulator of redox signaling and the antioxidant response in human spermatozoa. The lysophosphatidic acid signaling is essential to maintain sperm viability by activating the phosphatidylinositol 3-kinase/protein kinase (PI3K/AKT) pathway, and it is regulated by PRDX6 iPLA2, protein kinase C (PKC), and receptor-type protein tyrosine kinase. The understanding of redox signaling is crucial to pave the way for novel diagnostic tools and treatments of male infertility.
Collapse
Affiliation(s)
- Cristian O'Flaherty
- Department of Surgery (Urology Division), Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H4A 3J1, Canada
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3G 1Y6, Canada
- The Research Institute, McGill University Health Centre, Montréal, QC H4A 3J1, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada
| |
Collapse
|
16
|
Wang P, Ouyang J, Zhou K, Hu D, Zhang S, Zhang A, Yang Y. Olesoxime protects against cisplatin-induced acute kidney injury by attenuating mitochondrial dysfunction. Biomed J 2025; 48:100730. [PMID: 38643825 PMCID: PMC11751417 DOI: 10.1016/j.bj.2024.100730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/22/2024] [Accepted: 04/16/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Mitochondrial dysfunction is a critical factor in the pathogenesis of acute kidney injury (AKI). Agents that ameliorate mitochondrial dysfunction hold potential for AKI treatment. The objective of this study was to investigate the impact of olesoxime, a novel mitochondrial-targeted agent, on cisplatin-induced AKI. METHODS In vivo, a cisplatin-induced AKI mouse model was established by administering a single intraperitoneal dose of cisplatin (25 mg/kg) to male C57BL/6 mice for 72 hours, followed by gavage of either olesoxime or a control solution. In vitro, human proximal tubular HK2 cells were cultured and subjected to treatments with cisplatin, either in the presence or absence of olesoxime. RESULTS In vivo, our findings demonstrated that olesoxime administration significantly mitigated the nephrotoxic effects of cisplatin in mice, as evidenced by reduced blood urea nitrogen (BUN) and serum creatinine (SCr) levels, improved renal histopathology, and decreased expression of renal tubular injury markers such as kidney injury molecule 1 (KIM-1) and neutrophil gelatinase-associated lipocalin (NGAL). Furthermore, olesoxime administration markedly reduced cisplatin-induced apoptosis, inflammation, and oxidative stress in the kidneys of AKI mice. Additionally, olesoxime treatment effectively restored mitochondrial function in the kidneys of AKI mice. In vitro, our results indicated that olesoxime treatment protected against cisplatin-induced apoptosis and mitochondrial dysfunction in cultured HK2 cells. Notably, cisplatin's anticancer effects were unaffected by olesoxime treatment in human cancer cells. CONCLUSION The results of this study suggest that olesoxime is a viable and efficient therapeutic agent in the treatment of cisplatin-induced acute kidney injury presumably by alleviating mitochondrial dysfunction.
Collapse
Affiliation(s)
- Peipei Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Jing Ouyang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Kaiqian Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Dandan Hu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Shengnan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Yunwen Yang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Children's Hospital of Nanjing Medical University, China; Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
17
|
Liu X, Hage T, Chen L, Wang EHC, Liao I, Goldberg J, Gosto S, Cziryak P, Senna M, Chen Y, Zheng Q. Revealing the Therapeutic Potential: Investigating the Impact of a Novel Witch Hazel Formula on Anti-Inflammation and Antioxidation. J Cosmet Dermatol 2025; 24:e16662. [PMID: 39575475 PMCID: PMC11845955 DOI: 10.1111/jocd.16662] [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: 05/14/2024] [Revised: 10/04/2024] [Accepted: 10/18/2024] [Indexed: 02/23/2025]
Abstract
BACKGROUND Skin barrier health is crucial for preventive and corrective skincare across all skin types. Witch hazel (Hamamelis virginiana) extracts show potential in addressing skin issues, but their efficacy in treating chronic inflammation, improving skin barrier function, and combating UV-induced oxidation requires further investigation. AIMS To evaluate the efficacy of a novel formula containing witch hazel extracts in treating chronic inflammation, improving skin barrier function, and combating UV-induced oxidation. METHODS We employed a novel ex vivo chronic inflammation model to assess anti-inflammatory effects, measuring key pro-inflammatory cytokines. Barrier function markers, such as loricrin and transglutaminase-1, were analyzed. An ex vivo model with UV-induced Reactive Oxygen Species (ROS) elevation was used to evaluate antioxidant properties, measuring specific ROS markers like 4-Hydroxynonenal and carbonylated protein. RESULTS The novel witch hazel formula significantly reduced pro-inflammatory cytokines in both 2D and ex vivo models, including IL-6 and IL-8, demonstrating potent anti-inflammatory effects. Barrier function markers showed notable improvements compared to the inflamed condition. In the UV-induced ROS model, the formula remarkably decreased ROS levels, specifically 4-Hydroxynonenal and carbonylated protein, indicating strong antioxidant properties. CONCLUSIONS Our findings demonstrate that the novel witch hazel formula exhibits potent anti-inflammatory and antioxidant properties while enhancing skin barrier function. This natural, well-tolerated ingredient offers a promising treatment option for improving overall skin health, presenting new opportunities in skincare formulation and treatment strategies.
Collapse
Affiliation(s)
- Xue Liu
- L'Oreal Research and InnovationClarkNew JerseyUSA
| | | | - Li‐Chi Chen
- Harvard Medical School, Boston & Beth Israel Lahey HealthBurlingtonMassachusettsUSA
| | | | - I‐Chien Liao
- L'Oreal Research and InnovationClarkNew JerseyUSA
| | | | - Sabina Gosto
- L'Oreal Research and InnovationClarkNew JerseyUSA
| | | | - Maryanne Senna
- Harvard Medical School, Boston & Beth Israel Lahey HealthBurlingtonMassachusettsUSA
| | - Ying Chen
- L'Oreal Research and InnovationClarkNew JerseyUSA
| | - Qian Zheng
- L'Oreal Research and InnovationClarkNew JerseyUSA
| |
Collapse
|
18
|
Korbelik M, Heger M, Girotti AW. Participation of lipids in the tumor response to photodynamic therapy and its exploitation for therapeutic gain. J Lipid Res 2025; 66:100729. [PMID: 39675508 PMCID: PMC11911859 DOI: 10.1016/j.jlr.2024.100729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/19/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024] Open
Abstract
Hydroperoxides of unsaturated membrane lipids (LOOHs) are the most abundant non-radical intermediates generated by photodynamic therapy (PDT) of soft tissues such as tumors and have far longer average lifetimes than singlet oxygen or oxygen radicals formed during initial photodynamic action. LOOH-initiated post-irradiation damage to remaining membrane lipids (chain peroxidation) or to membrane-associated proteins remains largely unrecognized. Such after-light processes could occur during clinical oncological PDT, but this is not well-perceived by practitioners of this therapy. In general, the pivotal influence of lipids in tumor responses to PDT needs to be better appreciated. Of related importance is the fact that most malignant tumors have dramatically different lipid metabolism compared with healthy tissues, and this too is often ignored. The response of tumors to PDT appears especially vulnerable to manipulations within the tumor lipid microenvironment. This can be exploited for therapeutic gain with PDT, as exemplified here by the combined treatment with the antitumor lipid edelfosine.
Collapse
Affiliation(s)
- Mladen Korbelik
- Department of Integrative Oncology, BC Cancer, Vancouver, BC, Canada
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, P. R. China; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Membrane Biochemistry and Biophysics, Department of Chemistry, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| | - Albert W Girotti
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
19
|
Wu SC, Chen YJ, Su SH, Fang PH, Liu RW, Tsai HY, Chang YJ, Li HH, Li JC, Chen CH. Dysfunctional BCAA degradation triggers neuronal damage through disrupted AMPK-mitochondrial axis due to enhanced PP2Ac interaction. Commun Biol 2025; 8:105. [PMID: 39838082 PMCID: PMC11751115 DOI: 10.1038/s42003-025-07457-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
Metabolic and neurological disorders commonly display dysfunctional branched-chain amino acid (BCAA) metabolism, though it is poorly understood how this leads to neurological damage. We investigated this by generating Drosophila mutants lacking BCAA-catabolic activity, resulting in elevated BCAA levels and neurological dysfunction, mimicking disease-relevant symptoms. Our findings reveal a reduction in neuronal AMP-activated protein kinase (AMPK) activity, which disrupts autophagy in mutant brain tissues, linking BCAA imbalance to brain dysfunction. Mechanistically, we show that excess BCAA-induced mitochondrial reactive oxygen species (ROS) triggered the binding of protein phosphatase 2 A catalytic subunit (PP2Ac) to AMPK, suppressing AMPK activity. This initiated a dysregulated feedback loop of AMPK-mitochondrial interactions, exacerbating mitochondrial dysfunction and oxidative neuronal damage. Our study identifies BCAA imbalance as a critical driver of neuronal damage through AMPK suppression and autophagy dysfunction, offering insights into metabolic-neuronal interactions in neurological diseases and potential therapeutic targets for BCAA-related neurological conditions.
Collapse
Affiliation(s)
- Shih-Cheng Wu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 10048, Taiwan.
- Department of Laboratory Medicine, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 10021, Taiwan.
| | - Yan-Jhen Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, 10617, Taiwan
| | - Shih-Han Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 10048, Taiwan
| | - Pai-Hsiang Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
| | - Rei-Wen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, 300093, Taiwan
| | - Hui-Ying Tsai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
| | - Yen-Jui Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, 10048, Taiwan
| | - Hsing-Han Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
- Division of Biological Sciences, Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Jian-Chiuan Li
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, 350401, Taiwan.
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Zhunan, 350401, Taiwan.
| |
Collapse
|
20
|
Huang B, Wang H, Liu S, Hao M, Luo D, Zhou Y, Huang Y, Nian Y, Zhang L, Chu B, Yin C. Palmitoylation-dependent regulation of GPX4 suppresses ferroptosis. Nat Commun 2025; 16:867. [PMID: 39833225 PMCID: PMC11746948 DOI: 10.1038/s41467-025-56344-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
S-palmitoylation is a reversible and widespread post-translational modification, but its role in the regulation of ferroptosis has been poorly understood. Here, we elucidate that GPX4, an essential regulator of ferroptosis, is reversibly palmitoylated on cysteine 66. The acyltransferase ZDHHC20 palmitoylates GPX4 and increases its protein stability. ZDHHC20 depletion or inhibition of protein palmitoylation by 2-BP sensitizes cancer cells to ferroptosis. Moreover, we identify APT2 as the depalmitoylase of GPX4. Genetic silencing or pharmacological inhibition of APT2 with ML349 increases GPX4 palmitoylation, thereby stabilizing the protein and conferring resistance to ferroptosis. Notably, disrupting GPX4 palmitoylation markedly potentiates ferroptosis in xenografted and orthotopically implanted tumor models, and inhibits tumor metastasis through blood vessels. In the chemically induced colorectal cancer model, knockout of APT2 significantly aggravates cancer progression. Furthermore, pharmacologically modulating GPX4 palmitoylation impacts liver ischemia-reperfusion injury. Overall, our findings uncover the intricate network regulating GPX4 palmitoylation, highlighting its pivotal role in modulating ferroptosis sensitivity.
Collapse
Affiliation(s)
- Bin Huang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Hui Wang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China
| | - Shuo Liu
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meng Hao
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Dan Luo
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yi Zhou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Ying Huang
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Yong Nian
- College of Pharmacy, Nanjing Drum Tower Hospital, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lei Zhang
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Peking University, Shenzhen, Guangdong, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China
| | - Bo Chu
- Department of Cell Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Chengqian Yin
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, China.
- Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong, China.
| |
Collapse
|
21
|
Zhang QY, Gong HB, Jiang MY, Jin F, Wang G, Yan CY, Luo X, Sun WY, Ouyang SH, Wu YP, Duan WJ, Liang L, Cao YF, Sun XX, Liu M, Jiao GL, Wang HJ, Hiroshi K, Wang X, He RR, Li YF. Regulation of enzymatic lipid peroxidation in osteoblasts protects against postmenopausal osteoporosis. Nat Commun 2025; 16:758. [PMID: 39824794 PMCID: PMC11742680 DOI: 10.1038/s41467-025-55929-4] [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: 10/10/2023] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
Oxidative stress plays a critical role in postmenopausal osteoporosis, yet its impact on osteoblasts remains underexplored, limiting therapeutic advances. Our study identifies phospholipid peroxidation in osteoblasts as a key feature of postmenopausal osteoporosis. Estrogen regulates the transcription of glutathione peroxidase 4 (GPX4), an enzyme crucial for reducing phospholipid peroxides in osteoblasts. The deficiency of estrogen reduces GPX4 expression and increases phospholipid peroxidation in osteoblasts. Inhibition or knockout of GPX4 impairs osteoblastogenesis, while the elimination of phospholipid peroxides rescues bone formation and mitigates osteoporosis. Mechanistically, 4-hydroxynonenal, an end-product of phospholipid peroxidation, binds to integrin-linked kinase and triggers its protein degradation, disrupting RUNX2 signaling and inhibiting osteoblastogenesis. Importantly, we identified two natural allosteric activators of GPX4, 6- and 8-Gingerols, which promote osteoblastogenesis and demonstrate anti-osteoporotic effects. Our findings highlight the detrimental role of phospholipid peroxidation in osteoblastogenesis and underscore GPX4 as a promising therapeutic target for osteoporosis treatment.
Collapse
Grants
- 82125038, T2341004, 82174054, 82321004, 82274123, 82350003 National Natural Science Foundation of China (National Science Foundation of China)
- 2021B1515120023, 2023B1515040016, 2023B0303000026, 2020A1515110596 Natural Science Foundation of Guangdong Province (Guangdong Natural Science Foundation)
- the Local Innovative and Research Teams Project of Guangdong Pearl River Talents Program (2017BT01Y036 to RRH), Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine (2023LSYS002), and Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility (2024A03J090) to RRH, Science and Technology Program of Guangzhou (202102010116) to YFL.
Collapse
Affiliation(s)
- Qiong-Yi Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Hai-Biao Gong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Man-Ya Jiang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Fujun Jin
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
| | - Guan Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chang-Yu Yan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Xiang Luo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wan-Yang Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Shu-Hua Ouyang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Yan-Ping Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wen-Jun Duan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Lei Liang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Yun-Feng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, NHC Key Laboratory of Reproduction Regulation, Shanghai, 200032, China
| | - Xin-Xin Sun
- Jiujiang Maternal and Child Health Hospital, Jiujiang, 332000, China
| | - Meijing Liu
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China
| | - Gen-Long Jiao
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Hua-Jun Wang
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China
| | - Kurihara Hiroshi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Xiaogang Wang
- Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, PR China.
| | - Rong-Rong He
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China.
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
- The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
- Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
| | - Yi-Fang Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, 510632, China.
- Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility, Jinan University, Guangzhou, 510632, China.
- International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), Jinan University, Guangzhou, 510632, China.
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, China.
- The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou, 510632, China.
- Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
22
|
Shah V, Lam HY, Leong CHM, Sakaizawa R, Shah JS, Kumar AP. Epigenetic Control of Redox Pathways in Cancer Progression. Antioxid Redox Signal 2025. [PMID: 39815993 DOI: 10.1089/ars.2023.0465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Significance: Growing evidence indicates the importance of redox reactions homeostasis, mediated predominantly by reactive oxygen species (ROS) in influencing the development, differentiation, progression, metastasis, programmed cell death, tumor microenvironment, and therapeutic resistance of cancer. Therefore, reviewing the ROS-linked epigenetic changes in cancer is fundamental to understanding the progression and prevention of cancer. Recent Advances: We review in depth the molecular mechanisms involved in ROS-mediated epigenetic changes that lead to alteration of gene expression by altering DNA, modifying histones, and remodeling chromatin and noncoding RNA. Critical Issues: In cancerous cells, alterations of the gene-expression regulatory elements could be generated by the virtue of imbalance in tumor microenvironment. Various oxidizing agents and mitochondrial electron transport chain are the major pathways that generate ROS. ROS plays a key role in carcinogenesis by activating pro-inflammatory signaling pathways and DNA damage. This loss of ROS-mediated epigenetic regulation of the signaling pathways may promote tumorigenesis. We address all such aspects in this review. Future Directions: Developments in this growing field of epigenetics are expected to contribute to further our understanding of human health and diseases such as cancer and to test the clinical applications of redox-based therapy. Recent studies of the cancer-epigenetic landscape have revealed pervasive deregulation of the epigenetic factors in cancer. Thus, the study of interaction between ROS and epigenetic factors in cancer holds a great promise in the development of effective and targeted treatment modalities. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Hoi-Mun Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigna S Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Su C, Xue Y, Fan S, Sun X, Si Q, Gu Z, Wang J, Deng R. Ferroptosis and its relationship with cancer. Front Cell Dev Biol 2025; 12:1423869. [PMID: 39877159 PMCID: PMC11772186 DOI: 10.3389/fcell.2024.1423869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025] Open
Abstract
Marked by iron buildup and lipid peroxidation, ferroptosis is a relatively new regulatory cell death (RCD) pathway. Many diseases like cancer, myocardial ischemia-reperfusion injury (MIRI), neurological disorders and acute renal failure (AKI) are corelated with ferroptosis. The main molecular processes of ferroptosis discovered yet will be presented here, along with the approaches in which it interacts with tumour-associated signaling pathways and its uses in systemic therapy, radiation therapy, and immunotherapy managing tumors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Runzhi Deng
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Institute of Stomatology, Nanjing University, Nanjing, China
| |
Collapse
|
24
|
Ueda H, Honda A, Miyazaki T, Morishita Y, Hirayama T, Iwamoto J, Ikegami T. High-fat/high-sucrose diet results in a high rate of MASH with HCC in a mouse model of human-like bile acid composition. Hepatol Commun 2025; 9:e0606. [PMID: 39670881 PMCID: PMC11637755 DOI: 10.1097/hc9.0000000000000606] [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: 07/03/2024] [Accepted: 10/21/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND Wild-type (WT) mice fed a conventional high-fat/high-sucrose diet (HFHSD) rarely develop metabolic dysfunction-associated steatohepatitis (MASH) with HCC. Because mouse bile acid (BA) is highly hydrophilic, we hypothesized that making it hydrophobic would lead to MASH with HCC. METHODS Eleven-week-old WT and Cyp2a12/Cyp2c70 double knockout (DKO) mice were divided into two groups, including one which was fed a normal chow diet, and one which was fed an HFHSD. Samples were collected after 15, 30, 47, and 58 weeks for histological, biochemical, and immunological analyses. RESULTS In the HFHSD group, body weight gain did not differ in WT versus DKO mice, although HFHSD-fed DKO mice exhibited markedly accelerated liver inflammation, fibrosis, and carcinogenesis. HFHSD upregulated lipogenesis and downregulated fatty acid oxidation in both WT and DKO mice, which increased liver lipid accumulation and lipotoxicity. However, the increase in reactive oxygen species production and carcinogenesis observed in DKO mice could not be explained by abnormal lipid metabolism alone. Regarding BA metabolism, DKO mice had a higher hydrophobicity index. They exhibited an age-associated increase in chenodeoxycholic acid (CDCA) levels because of CYP8B1 activity inhibition due to the farnesoid X receptor activation. HFHSD further downregulated CYP8B1, presumably by activating the Liver X receptor. Liver CDCA accumulation was associated with increased inflammation, reactive oxygen species production, and hepatocyte FGF15 induction. Moreover, in noncancerous liver tissues, HFHSD appeared to activate STAT3, an oncogenic transcription factor, which was enhanced by a CDCA-rich environment. CONCLUSIONS Here, we developed a new model of MASH with HCC using mice with human-like BA composition and found that HFHSD and elevated hepatic CDCA synergistically increased the risk of MASH with HCC.
Collapse
Affiliation(s)
- Hajime Ueda
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Akira Honda
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Teruo Miyazaki
- Joint Research Center, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Yukio Morishita
- Diagnostic Pathology Division, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Takeshi Hirayama
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Junichi Iwamoto
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| | - Tadashi Ikegami
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Tokyo Medical University Ibaraki Medical Center, Ami, Ibaraki, Japan
| |
Collapse
|
25
|
Lu F, Cheng X, Qi X, Li D, Hu L. Metabolic landscaping of extracellular vesicles from body fluids by phosphatidylserine imprinted polymer enrichment and mass spectrometry analysis. Talanta 2025; 282:126940. [PMID: 39341064 DOI: 10.1016/j.talanta.2024.126940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 09/30/2024]
Abstract
Extracellular vesicles (EVs) are emerging as new source of biomarkers discovery in liquid biopsy due to their stabilization in body fluids, protected by phospholipid bilayers. However, the metabolomics study of EVs is very little reported due to the lack of efficient and high-throughput isolation methods for clinical samples. In this study, phosphatidylserine imprinted polymers were employed for rapid and efficient EVs isolation from five human body fluids, including plasma, urine, amniotic fluid, cerebrospinal fluid, and saliva. The isolated EVs were subsequently analyzed for metabolomic studies by high-resolution mass spectrometry. Metabolic landscaping was conducted between the body fluids and their EVs, indicating EVs contain a large number of metabolites that are completely specific to the body fluid source. Finally, quantitative metabolomic analysis of EVs was carried out with plasma samples of hepatocellular carcinoma. Several differentially expressed exosomal metabolites were revealed including the upregulation of sphingosine (d18:1), taurochenodeoxycholic acid (TCDCA), pipecolic acid (PA), and 4-hydroxynonenal (4-HNE) and down-regulation of piperine, caffeine, and indole. We believe the proposed methodology will provide a deeper understanding of the molecular composition and functions of EVs as an alternative source for biomarker discovery.
Collapse
Affiliation(s)
- Feng Lu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xianhui Cheng
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xiulei Qi
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Dejun Li
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Lianghai Hu
- Center for Supramolecular Chemical Biology, State Key Laboratory of Supramolecular Structure and Materials, School of Life Sciences, Jilin University, Changchun, 130012, China.
| |
Collapse
|
26
|
Kim JE, Lee DS, Wang SH, Kim TH, Kang TC. GPx1-ERK1/2-CREB pathway regulates the distinct vulnerability of hippocampal neurons to oxidative stress via modulating mitochondrial dynamics following status epilepticus. Neuropharmacology 2024; 260:110135. [PMID: 39214451 DOI: 10.1016/j.neuropharm.2024.110135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Glutathione peroxidase-1 (GPx1) and cAMP/Ca2+ responsive element (CRE)-binding protein (CREB) regulate neuronal viability by maintaining the redox homeostasis. Since GPx1 and CREB reciprocally regulate each other, it is likely that GPx1-CREB interaction may play a neuroprotective role against oxidative stress, which are largely unknown. Thus, we investigated the underlying mechanisms of the reciprocal regulation between GPx1 and CREB in the male rat hippocampus. Under physiological condition, L-buthionine sulfoximine (BSO)-induced oxidative stress increased GPx1 expression, extracellular signal-regulated kinase 1/2 (ERK1/2) activity and CREB serine (S) 133 phosphorylation in CA1 neurons, but not dentate granule cells (DGC), which were diminished by GPx1 siRNA, U0126 or CREB knockdown. GPx1 knockdown inhibited ERK1/2 and CREB activations induced by BSO. CREB knockdown also decreased the efficacy of BSO on ERK1/2 activation. BSO facilitated dynamin-related protein 1 (DRP1)-mediated mitochondrial fission in CA1 neurons, which abrogated by GPx1 knockdown and U0126. CREB knockdown blunted BSO-induced DRP1 upregulation without affecting DRP1 S616 phosphorylation ratio. Following status epilepticus (SE), GPx1 expression was reduced in CA1 neurons and DGC. SE also decreased CREB activity CA1 neurons, but not DGC. SE degenerated CA1 neurons, but not DGC, accompanied by mitochondrial elongation. These post-SE events were ameliorated by N-acetylcysteine (NAC, an antioxidant), but deteriorated by GPx1 knockdown. These findings indicate that a transient GPx1-ERK1/2-CREB activation may be a defense mechanism to protect hippocampal neurons against oxidative stress via maintenance of proper mitochondrial dynamics.
Collapse
Affiliation(s)
- Ji-Eun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| | - Duk-Shin Lee
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| | - Su Hyeon Wang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Cheon Kang
- Department of Anatomy and Neurobiology, Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
27
|
Maria Frare J, Rodrigues P, Andrighetto Ruviaro N, Trevisan G. Chronic post-ischemic pain (CPIP) a model of complex regional pain syndrome (CRPS-I): Role of oxidative stress and inflammation. Biochem Pharmacol 2024; 229:116506. [PMID: 39182734 DOI: 10.1016/j.bcp.2024.116506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Complex regional pain syndrome (CRPS) presents as a persistent and distressing pain condition often stemming from limb trauma or ischemia, manifesting as either CRPS-I (without initial nerve injury) or CRPS-II (accompanied by nerve injury). Despite its prevalence and significant impact on functionality and emotional well-being, standard treatments for CRPS remain elusive. The multifaceted nature of CRPS complicates the identification of its underlying mechanisms. In efforts to elucidate these mechanisms, researchers have turned to animal models such as chronic post-ischemic pain (CPIP), which mirrors the symptoms of CRPS-I. Various mechanisms have been proposed to underlie the acute and chronic pain experienced in CRPS-I, including oxidative stress and inflammation. Traditional treatment approaches often involve antidepressants, non-steroidal anti-inflammatory drugs (NSAIDs), and opioids. However, these methods frequently fall short of providing adequate relief. Accordingly, there is a growing interest in exploring alternative treatments, such as antioxidant supplementation, anti-inflammatory agents, and non-pharmacological interventions. Future research directions should focus on optimizing treatment strategies and addressing remaining gaps in knowledge to improve patient outcomes. This review aims to delve into the pathophysiological mechanisms implicated in the CPIP model, specifically focusing on oxidative stress and inflammation, with the ultimate goal of proposing innovative therapeutic strategies for alleviating the symptoms of CRPS-I.
Collapse
Affiliation(s)
- Julia Maria Frare
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil
| | - Patrícia Rodrigues
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil
| | - Náthaly Andrighetto Ruviaro
- Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil
| | - Gabriela Trevisan
- Graduated Program in Pharmacology, Federal University of Santa Maria (UFSM), 97105-900 Santa Maria, RS, Brazil; Graduated Program in Biological Sciences: Toxicological Biochemistry, Federal University of Santa Maria (UFSM), 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|
28
|
Cheng L, Zheng Q, Qiu K, Elmer Ker DF, Chen X, Yin Z. Mitochondrial destabilization in tendinopathy and potential therapeutic strategies. J Orthop Translat 2024; 49:49-61. [PMID: 39430132 PMCID: PMC11488423 DOI: 10.1016/j.jot.2024.09.003] [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: 04/20/2024] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 10/22/2024] Open
Abstract
Tendinopathy is a prevalent aging-related disorder characterized by pain, swelling, and impaired function, often resulting from micro-scarring and degeneration caused by overuse or trauma. Current interventions for tendinopathy have limited efficacy, highlighting the need for innovative therapies. Mitochondria play an underappreciated and yet crucial role in tenocytes function, including energy production, redox homeostasis, autophagy, and calcium regulation. Abnormalities in mitochondrial function may lead to cellular senescence. Within this context, this review provides an overview of the physiological functions of mitochondria in tendons and presents current insights into mitochondrial dysfunction in tendinopathy. It also proposes potential therapeutic strategies that focus on targeting mitochondrial health in tenocytes. These strategies include: (1) utilizing reactive oxygen species (ROS) scavengers to mitigate the detrimental effects of aberrant mitochondria, (2) employing mitochondria-protecting agents to reduce the production of dysfunctional mitochondria, and (3) supplementing with exogenous normal mitochondria. In conclusion, mitochondria-targeted therapies hold great promise for restoring mitochondrial function and improving outcomes in patients with tendinopathy. The translational potential of this article: Tendinopathy is challenging to treat effectively due to its poorly understood pathogenesis. This review thoroughly analyzes the role of mitochondria in tenocytes and proposes potential strategies for the mitochondrial treatment of tendinopathy. These findings establish a theoretical basis for future research and the clinical translation of mitochondrial therapy for tendinopathy.
Collapse
Affiliation(s)
- Linxiang Cheng
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Qiangqiang Zheng
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Kaijie Qiu
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, New Territories, Hong Kong
| | - Xiao Chen
- Department of Sports Medicine & Orthopedic Surgery, The Second Affiliated Hospital, And Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| | - Zi Yin
- Department of Orthopedic Surgery of Sir Run Run Shaw Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou City, Zhejiang Province, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
| |
Collapse
|
29
|
Yuan Z, Wang X, Qin B, Hu R, Miao R, Zhou Y, Wang L, Liu T. Targeting NQO1 induces ferroptosis and triggers anti-tumor immunity in immunotherapy-resistant KEAP1-deficient cancers. Drug Resist Updat 2024; 77:101160. [PMID: 39490240 DOI: 10.1016/j.drup.2024.101160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/27/2024] [Accepted: 10/13/2024] [Indexed: 11/05/2024]
Abstract
Immunotherapy has revolutionized cancer treatment, yet the efficacy of immunotherapeutic approaches remains limited. Resistance to ferroptosis is one of the reasons for the poor therapeutic outcomes in tumors with Kelch-like ECH-associated protein 1 (KEAP1) mutations. However, the specific mechanisms by which KEAP1-mutant tumors resist immunotherapy are not fully understood. In this study, we showed that the loss of function in KEAP1 results in resistance to ferroptosis. We identified NAD(P)H Quinone Dehydrogenase 1 (NQO1) as a transcriptional target of nuclear factor erythroid 2-related factor 2 (NRF2) and revealed that inducing NQO1-mediated ferroptosis in KEAP1-deficient tumors triggers an antitumor immune cascade. Additionally, it was found that NQO1 protein levels could serve as a candidate biomarker for predicting sensitivity to immunotherapy in clinical tumor patients. We validated these findings in several preclinical tumor models. Overall, KEAP1 mutations define a unique disease phenotype, and targeting its key downstream molecule NQO1 offers new hope for patients with resistance to immunotherapy.
Collapse
Affiliation(s)
- Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Boyu Qin
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rulong Hu
- Department of Otolaryngology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Rui Miao
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Yang Zhou
- Department of Respiratory Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lei Wang
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Tong Liu
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province 150001, China.
| |
Collapse
|
30
|
Wen C, Yu X, Zhu J, Zeng J, Kuang X, Zhang Y, Tang S, Zhang Q, Yan J, Shen H. Gastrodin ameliorates oxidative stress-induced RPE damage by facilitating autophagy and phagocytosis through PPARα-TFEB/CD36 signal pathway. Free Radic Biol Med 2024; 224:103-116. [PMID: 39173893 DOI: 10.1016/j.freeradbiomed.2024.08.023] [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: 07/10/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024]
Abstract
Age-related macular degeneration (AMD), the leading cause of irreversible blindness in the elderly, is primarily characterized by the degeneration of the retinal pigment epithelium (RPE). However, effective therapeutic options for dry AMD are currently lacking, necessitating further exploration into preventive and pharmaceutical interventions. This study aimed to investigate the protective effects of gastrodin on RPE cells exposed to oxidative stress. We constructed an in vitro oxidative stress model of 4-hydroxynonenal (4-HNE) and performed RNA-seq, and demonstrated the protective effect of gastrodin through mouse experiments. Our findings reveal that gastrodin can inhibit 4-HNE-induced oxidative stress, effectively improving the mitochondrial and lysosomal dysfunction of RPE cells. We further elucidated that gastrodin promotes autophagy and phagocytosis through activating the PPARα-TFEB/CD36 signaling pathway. Interestingly, these outcomes were corroborated in a mouse model, in which gastrodin maintained retinal integrity and reduced RPE disorganization and degeneration under oxidative stress. The accumulation of LC3B and SQSTM1 in mouse RPE-choroid was also reduced. Moreover, activating PPARα and downstream pathways to restore autophagy and phagocytosis, thereby countering RPE injury from oxidative stress. In conclusion, this study demonstrated that gastrodin maintains the normal function of RPE cells by reducing oxidative stress, enhancing their phagocytic function, and restoring the level of autophagic flow. These findings suggest that gastrodin is a novel formulation with potential applications in the development of AMD disease.
Collapse
Affiliation(s)
- Chaojuan Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xinyue Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jingya Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jingshu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Xielan Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Eye Biobank, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China
| | - Youao Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Shiyu Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qingjiong Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jianhua Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Eye Biobank, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China.
| |
Collapse
|
31
|
Banadaki MD, Rummel NG, Backus S, Butterfield DA, St Clair DK, Campbell JM, Zhong W, Mayer K, Berry SM, Chaiswing L. Extraction of redox extracellular vesicles using exclusion-based sample preparation. Anal Bioanal Chem 2024; 416:6317-6331. [PMID: 39243301 PMCID: PMC11807383 DOI: 10.1007/s00216-024-05518-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: 05/17/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Studying specific subpopulations of cancer-derived extracellular vesicles (EVs) could help reveal their role in cancer progression. In cancer, an increase in reactive oxygen species (ROS) happens which results in lipid peroxidation with a major product of 4-hydroxynonenal (HNE). Adduction by HNE causes alteration to the structure of proteins, leading to loss of function. Blebbing of EVs carrying these HNE-adducted proteins as a cargo or carrying HNE-adducted on EV membrane are methods for clearing these molecules by the cells. We have referred to these EVs as Redox EVs. Here, we utilize a surface tension-mediated extraction process, termed exclusion-based sample preparation (ESP), for the rapid and efficient isolation of intact Redox EVs, from a mixed population of EVs derived from human glioblastoma cell line LN18. After optimizing different parameters, two populations of EVs were analyzed, those isolated from the sample (Redox EVs) and those remaining in the original sample (Remaining EVs). Electron microscopic imaging was used to confirm the presence of HNE adducts on the outer leaflet of Redox EVs. Moreover, the population of HNE-adducted Redox EVs shows significantly different characteristics to those of Remaining EVs including smaller size EVs and a more negative zeta potential EVs. We further treated glioblastoma cells (LN18), radiation-resistant glioblastoma cells (RR-LN18), and normal human astrocytes (NHA) with both Remaining and Redox EV populations. Our results indicate that Redox EVs promote the growth of glioblastoma cells, likely through the production of H2O2, and cause injury to normal astrocytes. In contrast, Remaining EVs have minimal impact on the viability of both glioblastoma cells and NHA cells. Thus, isolating a subpopulation of EVs employing ESP-based immunoaffinity could pave the way for a deeper mechanistic understanding of how subtypes of EVs, such as those containing HNE-adducted proteins, induce biological changes in the cells that take up these EVs.
Collapse
Affiliation(s)
| | - Nicole G Rummel
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Spencer Backus
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA
| | - David Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Daret K St Clair
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - James M Campbell
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Weixiong Zhong
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Kristy Mayer
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Scott M Berry
- Department of Mechanical Engineering, University of Kentucky, Lexington, KY, 40506, USA.
| | - Luksana Chaiswing
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
32
|
Teranishi M, Ito M, Huang Z, Nishiyama Y, Masuda A, Mino H, Tachibana M, Inada T, Ohno K. Extremely Low-Frequency Electromagnetic Field (ELF-EMF) Increases Mitochondrial Electron Transport Chain Activities and Ameliorates Depressive Behaviors in Mice. Int J Mol Sci 2024; 25:11315. [PMID: 39457098 PMCID: PMC11508854 DOI: 10.3390/ijms252011315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Compromised mitochondrial electron transport chain (ETC) activities are associated with depression in humans and rodents. However, the effects of the enhancement of mitochondrial ETC activities on depression remain elusive. We recently reported that an extremely low-frequency electromagnetic field (ELF-EMF) of as low as 10 μT induced hormetic activation of mitochondrial ETC complexes in human/mouse cultured cells and mouse livers. Chronic social defeat stress (CSDS) for 10 consecutive days caused behavioral defects mimicking depression in mice, and using an ELF-EMF for two to six weeks ameliorated them. CSDS variably decreased the mitochondrial ETC proteins in the prefrontal cortex (PFC) in 10 days, which were increased by an ELF-EMF in six weeks. CSDS had no effect on the mitochondrial oxygen consumption rate in the PFC in 10 days, but using an ELF-EMF for six weeks enhanced it. CSDS inactivated SOD2 by enhancing its acetylation and increased lipid peroxidation in the PFC. In contrast, the ELF-EMF activated the Sirt3-FoxO3a-SOD2 pathway and suppressed lipid peroxidation. Furthermore, CSDS increased markers for mitophagy, which was suppressed by the ELF-EMF in six weeks. The ELF-EMF exerted beneficial hormetic effects on mitochondrial energy production, mitochondrial antioxidation, and mitochondrial dynamics in a mouse model of depression. We envisage that an ELF-EMF is a promising therapeutic option for depression.
Collapse
Affiliation(s)
- Masaki Teranishi
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Mikako Ito
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Zhizhou Huang
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Yuki Nishiyama
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Akio Masuda
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
| | - Hiroyuki Mino
- Division of Material Science (Physics), Graduate School of Science, Nagoya University, Nagoya 464-8602, Japan;
| | - Masako Tachibana
- Department of Psychiatry, Nagoya University Hospital, Nagoya 466-8560, Japan;
| | - Toshiya Inada
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan;
| | - Kinji Ohno
- Division of Neurogenetics, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; (M.T.); (Z.H.); (Y.N.); (A.M.)
- Graduate School of Nutritional Sciences, Nagoya University of Arts and Sciences, Nisshin 470-0196, Japan
| |
Collapse
|
33
|
Zheng X, Zhang Y, Zhang L, Yang T, Zhang F, Wang X, Zhu SJ, Cui N, Lv H, Zhang X, Li H, Liu W. Taurolithocholic acid protects against viral haemorrhagic fever via inhibition of ferroptosis. Nat Microbiol 2024; 9:2583-2599. [PMID: 39294459 DOI: 10.1038/s41564-024-01801-y] [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: 10/31/2023] [Accepted: 07/31/2024] [Indexed: 09/20/2024]
Abstract
Bile acids are microbial metabolites that can impact infection of enteric and hepatitis viruses, but their functions during systemic viral infection remain unclear. Here we show that elevated levels of the secondary bile acid taurolithocholic acid (TLCA) are associated with reduced fatality rates and suppressed viraemia in patients infected with severe fever with thrombocytopenia syndrome virus (SFTSV), an emerging tick-borne haemorrhagic fever virus. TLCA inhibits viral replication and mitigates host inflammation during SFTSV infection in vitro, and indirectly suppresses SFTSV-mediated induction of ferroptosis by upregulating fatty acid desaturase 2 via the TGR5-PI3K/AKT-SREBP2 axis. High iron and ferritin serum levels during early infection were correlated with decreased TLCA levels and fatal outcomes in SFTSV-infected patients, indicating potential biomarkers. Furthermore, treatment with either ferroptosis inhibitors or TLCA protected mice from lethal SFTSV infection. Our findings highlight the therapeutic potential of bile acids to treat haemorrhagic fever viral infection.
Collapse
Affiliation(s)
- Xiaojie Zheng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Yunfa Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Lingyu Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Tong Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Faxue Zhang
- School of Public Health, Wuhan University, Wuhan, People's Republic of China
| | - Xi Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
- Graduate School of Anhui Medical University, Hefei, People's Republic of China
| | - Shu Jeffrey Zhu
- Key Laboratory of Animal Virology of Ministry of Agriculture, Center for Veterinary Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Ning Cui
- The 154th Hospital, Xinyang, People's Republic of China
| | - Hongdi Lv
- The 154th Hospital, Xinyang, People's Republic of China
| | - Xiaoai Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China.
- School of Public Health, Wuhan University, Wuhan, People's Republic of China.
- Graduate School of Anhui Medical University, Hefei, People's Republic of China.
| | - Wei Liu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, People's Republic of China.
- School of Public Health, Wuhan University, Wuhan, People's Republic of China.
- Graduate School of Anhui Medical University, Hefei, People's Republic of China.
| |
Collapse
|
34
|
Taylor JL, Baudel MMA, Nieves-Cintron M, Navedo MF. Vascular Function and Ion Channels in Alzheimer's Disease. Microcirculation 2024; 31:e12881. [PMID: 39190776 PMCID: PMC11498901 DOI: 10.1111/micc.12881] [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: 07/04/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
This review paper explores the critical role of vascular ion channels in the regulation of cerebral artery function and examines the impact of Alzheimer's disease (AD) on these processes. Vascular ion channels are fundamental in controlling vascular tone, blood flow, and endothelial function in cerebral arteries. Dysfunction of these channels can lead to impaired cerebral autoregulation, contributing to cerebrovascular pathologies. AD, characterized by the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles, has been increasingly linked to vascular abnormalities, including altered vascular ion channel activity. Here, we briefly review the role of vascular ion channels in cerebral blood flow control and neurovascular coupling. We then examine the vascular defects in AD, the current understanding of how AD pathology affects vascular ion channel function, and how these changes may lead to compromised cerebral blood flow and neurodegenerative processes. Finally, we provide future perspectives and conclusions. Understanding this topic is important as ion channels may be potential therapeutic targets for improving cerebrovascular health and mitigating AD progression.
Collapse
Affiliation(s)
- Jade L. Taylor
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| | | | | | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| |
Collapse
|
35
|
Sridharan PS, Koh Y, Miller E, Hu D, Chakraborty S, Tripathi SJ, Kee TR, Chaubey K, Vázquez-Rosa E, Barker S, Liu H, León-Alvarado RA, Franke K, Cintrón-Pérez CJ, Dhar M, Shin MK, Flanagan ME, Castellani RJ, Gefen T, Bykova M, Dou L, Cheng F, Wilson BM, Fujioka H, Kang DE, Woo JAA, Paul BD, Qi X, Pieper AA. Acutely blocking excessive mitochondrial fission prevents chronic neurodegeneration after traumatic brain injury. Cell Rep Med 2024; 5:101715. [PMID: 39241772 PMCID: PMC11525032 DOI: 10.1016/j.xcrm.2024.101715] [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: 01/20/2024] [Revised: 06/03/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Progression of acute traumatic brain injury (TBI) into chronic neurodegeneration is a major health problem with no protective treatments. Here, we report that acutely elevated mitochondrial fission after TBI in mice triggers chronic neurodegeneration persisting 17 months later, equivalent to many human decades. We show that increased mitochondrial fission after mouse TBI is related to increased brain levels of mitochondrial fission 1 protein (Fis1) and that brain Fis1 is also elevated in human TBI. Pharmacologically preventing Fis1 from binding its mitochondrial partner, dynamin-related protein 1 (Drp1), for 2 weeks after TBI normalizes the balance of mitochondrial fission/fusion and prevents chronically impaired mitochondrial bioenergetics, oxidative damage, microglial activation and lipid droplet formation, blood-brain barrier deterioration, neurodegeneration, and cognitive impairment. Delaying treatment until 8 months after TBI offers no protection. Thus, time-sensitive inhibition of acutely elevated mitochondrial fission may represent a strategy to protect human TBI patients from chronic neurodegeneration.
Collapse
Affiliation(s)
- Preethy S Sridharan
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yeojung Koh
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Emiko Miller
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Di Hu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teresa R Kee
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Kalyani Chaubey
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sarah Barker
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hui Liu
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rose A León-Alvarado
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Earlham College Neuroscience Program, Richmond, IN, USA
| | - Kathryn Franke
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Coral J Cintrón-Pérez
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matasha Dhar
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Min-Kyoo Shin
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08226, Republic of Korea
| | - Margaret E Flanagan
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Glenn Bigg's Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rudolph J Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marina Bykova
- Department of Regulatory Biology, Cleveland State University, Cleveland, OH, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lijun Dou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brigid M Wilson
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David E Kang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA; Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Jung-A A Woo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
36
|
Reis PM, Vargas BS, Rebelo RA, Massafera MP, Prado FM, Oreliana H, de Oliveira HV, Freitas F, Ronsein GE, Miyamoto S, Di Mascio P, Medeiros MHG. Quantitative Analysis of Glutathione and Carnosine Adducts with 4-Hydroxy-2-nonenal in Muscle in a hSOD1 G93A ALS Rat Model. Chem Res Toxicol 2024; 37:1306-1314. [PMID: 39066735 PMCID: PMC11337210 DOI: 10.1021/acs.chemrestox.4c00052] [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: 02/07/2024] [Revised: 07/12/2024] [Accepted: 07/16/2024] [Indexed: 07/30/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the dysfunction and death of motor neurons through multifactorial mechanisms that remain unclear. ALS has been recognized as a multisystemic disease, and the potential role of skeletal muscle in disease progression has been investigated. Reactive aldehydes formed as secondary lipid peroxidation products in the redox processes react with biomolecules, such as DNA, proteins, and amino acids, resulting in cytotoxic effects. 4-Hydroxy-2-nonenal (HNE) levels are elevated in the spinal cord motor neurons of ALS patients, and HNE-modified proteins have been identified in the spinal cord tissue of an ALS transgenic mice model, suggesting that reactive aldehydes can contribute to motor neuron degeneration in ALS. One biological pathway of aldehyde detoxification involves conjugation with glutathione (GSH) or carnosine (Car). Here, the detection and quantification of Car, GSH, GSSG (glutathione disulfide), and the corresponding adducts with HNE, Car-HNE, and GS-HNE, were performed in muscle and liver tissues of a hSOD1G93A ALS rat model by reverse-phase high-performance liquid chromatography coupled to electrospray ion trap tandem mass spectrometry in the selected reaction monitoring mode. A significant increase in the levels of GS-HNE and Car-HNE was observed in the muscle tissue of the end-stage ALS animals. Therefore, analyzing variations in the levels of these adducts in ALS animal tissue is crucial from a toxicological perspective and can contribute to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo
V. M. Reis
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Bianca S. Vargas
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Rafael A. Rebelo
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Mariana P. Massafera
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Fernanda M. Prado
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Hector Oreliana
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Henrique V. de Oliveira
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Florêncio
P. Freitas
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Graziella E. Ronsein
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Paolo Di Mascio
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| | - Marisa H. G. Medeiros
- Departamento de Bioquímica,
Instituto de Química, Universidade
de São Paulo, São
Paulo, SP 05508-900, Brazil
| |
Collapse
|
37
|
Gonzalez-Gutierrez L, Motiño O, Barriuso D, de la Puente-Aldea J, Alvarez-Frutos L, Kroemer G, Palacios-Ramirez R, Senovilla L. Obesity-Associated Colorectal Cancer. Int J Mol Sci 2024; 25:8836. [PMID: 39201522 PMCID: PMC11354800 DOI: 10.3390/ijms25168836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Colorectal cancer (CRC) affects approximately 2 million people worldwide. Obesity is the major risk factor for CRC. In addition, obesity contributes to a chronic inflammatory stage that enhances tumor progression through the secretion of proinflammatory cytokines. In addition to an increased inflammatory response, obesity-associated cancer presents accrued molecular factors related to cancer characteristics, such as genome instability, sustained cell proliferation, telomere dysfunctions, angiogenesis, and microbial alteration, among others. Despite the evidence accumulated over the last few years, the treatments for obesity-associated CRC do not differ from the CRC treatments in normal-weight individuals. In this review, we summarize the current knowledge on obesity-associated cancer, including its epidemiology, risk factors, molecular factors, and current treatments. Finally, we enumerate possible new therapeutic targets that may improve the conditions of obese CRC patients. Obesity is key for the development of CRC, and treatments resulting in the reversal of obesity should be considered as a strategy for improving antineoplastic CRC therapies.
Collapse
Affiliation(s)
- Lucia Gonzalez-Gutierrez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Omar Motiño
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Daniel Barriuso
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Juan de la Puente-Aldea
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Lucia Alvarez-Frutos
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France;
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France
| | - Roberto Palacios-Ramirez
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
| | - Laura Senovilla
- Unidad de Excelencia Instituto de Biología y Genética Molecular (IBGM), Universidad de Valladolid–CSIC, 47003 Valladolid, Spain; (L.G.-G.); (O.M.); (D.B.); (J.d.l.P.-A.); (L.A.-F.); (R.P.-R.)
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université Paris Cité, Sorbonne Université, Inserm U1138, Institut Universitaire de France, 75006 Paris, France;
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, 94805 Villejuif, France
| |
Collapse
|
38
|
Wang S, Xu F, Liu H, Shen Y, Zhang J, Hu L, Zhu L. Suppressing Endoplasmic Reticulum Stress Alleviates LPS-Induced Acute Lung Injury via Inhibiting Inflammation and Ferroptosis. Inflammation 2024; 47:1067-1082. [PMID: 38308704 DOI: 10.1007/s10753-023-01962-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/29/2023] [Accepted: 12/26/2023] [Indexed: 02/05/2024]
Abstract
Acute lung injury (ALI) is a life-threatening clinical disorder with high mortality rate. Ferroptosis is a new type of programmed cell death with lipid peroxidation and iron ion overloading as the main characteristics. Endoplasmic reticulum (ER) stress and ferroptosis play pivotal roles in the pathogenesis of ALI. The study aimed to investigate the underlying relationship between ER stress and ferroptosis in ALI. The ER stress inhibitor 4-phenylbutyric acid (4-PBA) alleviated LPS-induced inflammation, and decreased IL-1β, IL-6, and TNF-α levels in BALF and lungs. The increased MDA and decreased GSH induced by LPS were partially reversed by 4-PBA, which also inhibited the expressions of ferroptosis-related protein ACSL4, COX-2, and FTH1. TEM further confirmed the ferroptosis within airway epithelia cells was ameliorated by 4-PBA. Moreover, 4-PBA reduced the production of ROS and lipid ROS in LPS-exposed BEAS-2B cells in a concentration-dependent way. Meanwhile, 4-PBA mitigated LPS-induced cell apoptosis in vivo and in vitro. Mechanistically, the MAPK signaling pathway activated by LPS was downregulated by 4-PBA. Collectively, these findings suggested that 4-PBA protected against ALI by inhibiting inflammation and ferroptosis through downregulating ER stress, thus providing a potential intervention for ALI and revealing the possible interaction between ER stress and ferroptosis in ALI.
Collapse
Affiliation(s)
- Sijiao Wang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Fan Xu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Hanhan Liu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yue Shen
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jun Zhang
- Department of Pulmonary Medicine, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Lijuan Hu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
39
|
Kuang Z, Ge Y, Cao L, Wang X, Liu K, Wang J, Zhu X, Wu M, Li J. Precision Treatment of Anthracycline-Induced Cardiotoxicity: An Updated Review. Curr Treat Options Oncol 2024; 25:1038-1054. [PMID: 39066853 PMCID: PMC11329674 DOI: 10.1007/s11864-024-01238-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Anthracycline (ANT)-induced cardiotoxicity (AIC) is a particularly prominent form of cancer therapy-related cardiovascular toxicity leading to the limitations of ANTs in clinical practice. Even though AIC has drawn particular attention, the best way to treat it is remaining unclear. Updates to AIC therapy have been made possible by recent developments in research on the underlying processes of AIC. We review the current molecular pathways leading to AIC: 1) oxidative stress (OS) including enzymatic-induced and other mechanisms; 2) topoisomerase; 3) inflammatory response; 4) cardiac progenitor cell damage; 5) epigenetic changes; 6) renin-angiotensin-aldosterone system (RAAS) dysregulation. And we systematically discuss current prevention and treatment strategies and novel pathogenesis-based therapies for AIC: 1) dose reduction and change; 2) altering drug delivery methods; 3) antioxidants, dexrezosen, statina, RAAS inhibitors, and hypoglycemic drugs; 4) miRNA, natural phytochemicals, mesenchymal stem cells, and cardiac progenitor cells. We also offer a fresh perspective on the management of AIC by outlining the current dilemmas and challenges associated with its prevention and treatment.
Collapse
Affiliation(s)
- Ziyu Kuang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Yuansha Ge
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Luchang Cao
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xinmiao Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Kexin Liu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, 10029, China
| | - Jiaxi Wang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China
| | - Xiaojuan Zhu
- The 3rd affiliated hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China.
| | - Min Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| | - Jie Li
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 10053, China.
| |
Collapse
|
40
|
NAGINI SIDDAVARAM, KALLAMADI PRATHAPREDDY, TANAGALA KRANTHIKIRANKISHORE, REDDY GEEREDDYBHANUPRAKASH. Aldo-keto reductases: Role in cancer development and theranostics. Oncol Res 2024; 32:1287-1308. [PMID: 39055885 PMCID: PMC11267078 DOI: 10.32604/or.2024.049918] [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: 01/22/2024] [Accepted: 05/08/2024] [Indexed: 07/28/2024] Open
Abstract
Aldo-keto reductases (AKRs) are a superfamily of enzymes that play crucial roles in various cellular processes, including the metabolism of xenobiotics, steroids, and carbohydrates. A growing body of evidence has unveiled the involvement of AKRs in the development and progression of various cancers. AKRs are aberrantly expressed in a wide range of malignant tumors. Dysregulated expression of AKRs enables the acquisition of hallmark traits of cancer by activating oncogenic signaling pathways and contributing to chemoresistance. AKRs have emerged as promising oncotherapeutic targets given their pivotal role in cancer development and progression. Inhibition of aldose reductase (AR), either alone or in combination with chemotherapeutic drugs, has evolved as a pragmatic therapeutic option for cancer. Several classes of synthetic aldo-keto reductase (AKR) inhibitors have been developed as potential anticancer agents, some of which have shown promise in clinical trials. Many AKR inhibitors from natural sources also exhibit anticancer effects. Small molecule inhibitors targeting specific AKR isoforms have shown promise in preclinical studies. These inhibitors disrupt the activation of oncogenic signaling by modulating transcription factors and kinases and sensitizing cancer cells to chemotherapy. In this review, we discuss the physiological functions of human AKRs, the aberrant expression of AKRs in malignancies, the involvement of AKRs in the acquisition of cancer hallmarks, and the role of AKRs in oncogenic signaling, and drug resistance. Finally, the potential of aldose reductase inhibitors (ARIs) as anticancer drugs is summarized.
Collapse
|
41
|
Waltrick APF, Radulski DR, de Oliveira KM, Acco A, Verri WA, da Cunha JM, Zanoveli JM. Early evidence of beneficial and protective effects of Protectin DX treatment on behavior responses and type-1 diabetes mellitus related-parameters: A non-clinical approach. Prog Neuropsychopharmacol Biol Psychiatry 2024; 133:111028. [PMID: 38754696 DOI: 10.1016/j.pnpbp.2024.111028] [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: 01/02/2024] [Revised: 04/24/2024] [Accepted: 05/12/2024] [Indexed: 05/18/2024]
Abstract
Protectin DX (PDX), a specialized pro-resolving lipid mediator, presents potential therapeutic applications across various medical conditions due to its anti-inflammatory and antioxidant properties. Since type-1 diabetes mellitus (T1DM) is a disease with an inflammatory and oxidative profile, exploring the use of PDX in addressing T1DM and its associated comorbidities, including diabetic neuropathic pain, depression, and anxiety becomes urgent. Thus, in the current study, after 2 weeks of T1DM induction with streptozotocin (60 mg/kg) in Wistar rats, PDX (1, 3, and 10 ng/animal; i.p. injection of 200 μl/animal) was administered specifically on days 14, 15, 18, 21, 24, and 27 after T1DM induction. We investigated the PDX's effectiveness in alleviating neuropathic pain (mechanical allodynia; experiment 1), anxiety-like and depressive-like behaviors (experiment 2). Also, we studied whether the PDX treatment would induce antioxidant effects in the blood plasma, hippocampus, and prefrontal cortex (experiment 3), brain areas involved in the modulation of emotions. For evaluating mechanical allodynia, animals were repeatedly submitted to the Von Frey test; while for studying anxiety-like responses, animals were submitted to the elevated plus maze (day 26) and open field (day 28) tests. To analyze depressive-like behaviors, the animals were tested in the modified forced swimming test (day 28) immediately after the open field test. Our data demonstrated that PDX consistently increased the mechanical threshold throughout the study at the two highest doses, indicative of antinociceptive effect. Concerning depressive-like and anxiety-like behavior, all PDX doses effectively prevented these behaviors when compared to vehicle-treated T1DM rats. The PDX treatment significantly protected against the increased oxidative stress parameters in blood plasma and in hippocampus and prefrontal cortex. Interestingly, treated animals presented improvement on diabetes-related parameters by promoting weight gain and reducing hyperglycemia in T1DM rats. These findings suggest that PDX improved diabetic neuropathic pain, and induced antidepressant-like and anxiolytic-like effects, in addition to improving parameters related to the diabetic condition. It is worth noting that PDX also presented a protective action demonstrated by its antioxidant effects. To conclude, our findings suggest PDX treatment may be a promising candidate for improving the diabetic condition per se along with highly disabling comorbidities such as diabetic neuropathic pain and emotional disturbances associated with T1DM.
Collapse
Affiliation(s)
- Ana Paula Farias Waltrick
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil
| | - Débora Rasec Radulski
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil
| | - Kauê Marcel de Oliveira
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil
| | - Alexandra Acco
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil
| | | | - Joice Maria da Cunha
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil
| | - Janaina Menezes Zanoveli
- Department of Pharmacology, Biological Sciences Building, Federal University of Paraná, Street Coronel Francisco H dos Santos S/N, P.O. Box 19031, Curitiba, PR 81540-990, Brazil.
| |
Collapse
|
42
|
Gao T, Wang Q, Sun H, Liu Y, Li J, He Y. Physiological Adaptation of Fenneropenaeus chinensis in Response to Saline-Alkaline Stress Revealed by a Combined Proteomics and Metabolomics Method. BIOLOGY 2024; 13:488. [PMID: 39056683 PMCID: PMC11274245 DOI: 10.3390/biology13070488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024]
Abstract
The rapid development of the mariculture industry has been hindered by limited coastal aquaculture space. To utilize the abundant inland saline-alkaline water, we studied the physiological effects of high carbonate alkalinity stress and high pH stress on Fenneropenaeus chinensis. The study employed quantitative proteomics by tandem mass tag (TMT) and non-targeted metabolomics analysis using a liquid chromatograph mass spectrometer (LC-MS) to understand the physiological and biochemical adaptive mechanisms of the hepatopancreas of F. chinensis in response to saline-alkaline stress at the molecular level. We designed two stress groups as follows: a high carbonate alkalinity (CA) group and a combined high carbonate alkalinity and high pH (CP) group. The study found that the protein and metabolic profiles of the two stress groups were changed, and the CP group, which was exposed to dual stresses, incurred more severe damage to the hepatopancreas compared to that of the CA group. After exposure to CA and CP, the hepatopancreas of F. chinensis showed significant alterations in 455 proteins and 50 metabolites, and 1988 proteins and 272 metabolites, respectively. In addition, F. chinensis upregulated the level of energy metabolism in the hepatopancreas to defend against osmotic imbalance caused by CA or CP stress, which was demonstrated by the significant upregulation of important proteins and metabolites in glycolysis, pyruvate metabolism, TCA cycle, and fatty acid oxidation. Additionally, pattern recognition receptors, the phenol oxidase system, and various immune-related metabolic enzymes and metabolites were also affected. The immune homeostasis of F. chinensis was affected by the alteration of the antioxidant system following exposure to CA or CP. These findings provide valuable information for F. chinensis saline-alkaline water cultivation practices.
Collapse
Affiliation(s)
- Tian Gao
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; (T.G.); (H.S.); (Y.L.)
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China;
| | - Qiong Wang
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China;
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, China
| | - Huarui Sun
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; (T.G.); (H.S.); (Y.L.)
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China;
| | - Yang Liu
- College of Fisheries and Life Science, Dalian Ocean University, Dalian 116023, China; (T.G.); (H.S.); (Y.L.)
| | - Jitao Li
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China;
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, China
| | - Yuying He
- National Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China;
- Function Laboratory for Marine Fisheries Science and Food Production Processes, Pilot National Laboratory for Marine Science and Technology, Qingdao 266200, China
| |
Collapse
|
43
|
Mokhtarpour K, Razi S, Rezaei N. Ferroptosis as a promising targeted therapy for triple negative breast cancer. Breast Cancer Res Treat 2024:10.1007/s10549-024-07387-7. [PMID: 38874688 DOI: 10.1007/s10549-024-07387-7] [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: 12/06/2023] [Accepted: 05/22/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Triple negative breast cancer (TNBC) is a challenging subtype characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. Standard treatment options are limited, and approximately 45% of patients develop distant metastasis. Ferroptosis, a regulated form of cell death triggered by iron-dependent lipid peroxidation and oxidative stress, has emerged as a potential targeted therapy for TNBC. METHODS This study utilizes a multifaceted approach to investigate the induction of ferroptosis as a therapeutic strategy for TNBC. It explores metabolic alterations, redox imbalance, and oncogenic signaling pathways to understand their roles in inducing ferroptosis, characterized by lipid peroxidation, reactive oxygen species (ROS) generation, and altered cellular morphology. Critical pathways such as Xc-/GSH/GPX4, ACSL4/LPCAT3, and nuclear factor erythroid 2-related factor 2 (NRF2) are examined for their regulatory roles in ferroptosis and their potential dysregulation contributing to cancer cell survival and resistance. RESULTS Inducing ferroptosis has been shown to inhibit tumor growth, enhance the efficacy of conventional therapies, and overcome drug resistance in TNBC. Lipophilic antioxidants, GPX4 inhibitors, and inhibitors of the Xc- system have been demonstrated to be potential ferroptosis inducers. Additionally, targeting the NRF2 pathway and exploring other ferroptosis regulators, such as ferroptosis suppressor protein 1 (FSP1), and the PERK-eIF2α-ATF4-CHOP pathway, may offer novel therapeutic avenues. CONCLUSION Further research is needed to understand the mechanisms, optimize therapeutic strategies, and evaluate the safety and efficacy of ferroptosis-targeted therapies in TNBC treatment. Overall, targeting ferroptosis represents a promising approach to improving treatment outcomes and overcoming the challenges posed by TNBC.
Collapse
Affiliation(s)
- Kasra Mokhtarpour
- Faculty of Veterinary Medicine, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Imunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
| | - Nima Rezaei
- Research Center for Imunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
44
|
Thorwald M, Godoy-Lugo JA, Garcia G, Silva J, Kim M, Christensen A, Mack WJ, Head E, O'Day PA, Benayoun BA, Morgan TE, Pike CJ, Higuchi-Sanabria R, Forman HJ, Finch CE. Iron associated lipid peroxidation in Alzheimers disease is increased in lipid rafts with decreased ferroptosis suppressors, tested by chelation in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.28.534324. [PMID: 37034750 PMCID: PMC10081222 DOI: 10.1101/2023.03.28.534324] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Iron-mediated cell death (ferroptosis) is a proposed mechanism of Alzheimers disease (AD) pathology. While iron is essential for basic biological functions, its reactivity generates oxidants which contribute to cell damage and death. To further resolve mechanisms of iron-mediated toxicity in AD, we analyzed postmortem human brain and ApoEFAD mice. AD brains had decreased antioxidant enzymes, including those mediated by glutathione (GSH). Subcellular analyses of AD brains showed greater oxidative damage and lower antioxidant enzymes in lipid rafts, the site of amyloid processing, than in the non-raft membrane fraction. ApoE4 carriers had lower lipid raft yield with greater membrane oxidation. The hypothesized role of iron to AD pathology was tested in ApoEFAD mice by iron chelation with deferoxamine, which decreased fibrillar amyloid and lipid peroxidation, together with increased GSH-mediated antioxidants. These novel molecular pathways in iron mediated damage during AD.
Collapse
|
45
|
Wu X, Yang SA, Kan Y, Li M, Dong J, Qiu T, Gu Y, Zhao Y, Liang D. Revealing Metabolic Dysregulation Induced by Polypropylene Nano- and Microplastics in Nile Tilapia via Noninvasive Probing Epidermal Mucus. Anal Chem 2024; 96:9416-9423. [PMID: 38809415 DOI: 10.1021/acs.analchem.4c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
A noninvasive sampling technology was conceived, employing a disposable acupuncture needle in conjunction with high-resolution mass spectrometry (termed as noninvasive direct sampling extractive electrospray ionization mass spectrometry, NIDS-EESI-MS) to scrutinize the epidermal mucus of Nile tilapia for insights into the metabolic dysregulation induced by polypropylene nano- and microplastics. This analytical method initiates with the dispensing of an extraction solvent onto the needles coated with the mucus sample, almost simultaneously applying a high voltage to generate analyte ions. This innovative strategy obliterates the necessitation for laborious sample preparation, thereby simplifying the sampling process. Employing this technique facilitated the delineation of a plethora of metabolites, encompassing, but not confined to, amino acids, peptides, carbohydrates, ketones, fatty acids, and their derivatives. Follow-up pathway enrichment analysis exposed notable alterations within key metabolic pathways, including the biosynthesis of phenylalanine, tyrosine, and tryptophan, lysine degradation, as well as the biosynthesis and metabolism of valine, leucine, and isoleucine pathways in Nile tilapia, consequent to increased concentrations of polypropylene nanoplastics. These metabolic alterations portend potential implications such as immune suppression, among other deleterious outcomes. This trailblazing application of this methodology not only spares aquatic life from sacrifice but also inaugurates an ethical paradigm for conducting longitudinal studies on the same organisms, facilitating detailed investigations into the long-term effects of environmental pollutants. This technique enhances the ability to observe and understand the subtle yet significant impacts of such contaminants over time.
Collapse
Affiliation(s)
- Xiaokang Wu
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Sheng-Ao Yang
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Ying Kan
- Division of Chemical Metrology and Analytical Science, National Institute of Metrology, Beijing 100029, PR China
| | - Ming Li
- Division of Chemical Metrology and Analytical Science, National Institute of Metrology, Beijing 100029, PR China
| | - Jiaxin Dong
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Tao Qiu
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Yu Gu
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Yuanxin Zhao
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| | - Dapeng Liang
- Key Laboratory of Groundwater Resources and Environment of Ministry of Education, College of New Energy and Environment, Jilin University, Changchun 130012, PR China
| |
Collapse
|
46
|
Yang Q, Xia Y, Chen K, Wang Y, Song D, Zhu J, Tong J, Shen Y. Blue light induced ferroptosis via STAT3/GPX4/SLC7A11/FTH1 in conjunctiva epithelium in vivo and in vitro. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112908. [PMID: 38663336 DOI: 10.1016/j.jphotobiol.2024.112908] [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: 12/04/2023] [Revised: 03/28/2024] [Accepted: 04/12/2024] [Indexed: 05/13/2024]
Abstract
The prevalence of Light-emitting diodes (LEDs) has exposed us to an excessive amount of blue light (BL) which causes various ophthalmic diseases. Previous studies have shown that conjunctiva is vulnerable to BL. In this study, we aimed to investigate the underlying mechanism of BL-induced injury in conjunctiva. We placed C57BL/6 mice and human conjunctival epithelial cell lines (HCECs) under BL (440 nm ± 15 nm, 0.2 mW/cm2) to establish a BL injury model in vivo and in vitro. Immunohistochemistry and MDA assay were used to identify lipid peroxidation (LPO) in vivo. HE staining was applied to detect morphological damage of conjunctival epithelium. DCFH-DA, C11-BODIPY 581/591, Calcein-AM, and FeRhoNox™-1 probes were performed to identify ferroptosis levels in vitro. Real-time qPCR and Western blotting techniques were employed to uncover signaling pathways of blue light-induced ferroptosis. Our findings demonstrated that BL affected tear film instability and induced conjunctival epithelium injury in vivo. Ferrostatin-1 significantly alleviated blue light-induced ferroptosis in vivo and in vitro. BL downregulates the levels of solute carrier family 7 member 11 (SLC7A11), Ferritin heavy chain (FTH1), and glutathione peroxidase (GPX4) by inhibiting the activation and translocation of the Signal transducer and activator of transcription 3 (STAT3) from inducing Fe2+ burst, ROS and LPO accumulation, ultimately resulting in ferroptosis. This study will offer new insight into BL-induced conjunctival injury and LED-induced dry eye.
Collapse
Affiliation(s)
- Qianjie Yang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yutong Xia
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Kuangqi Chen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Yinhao Wang
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Dongjie Song
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jiru Zhu
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Jianping Tong
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ye Shen
- Department of Ophthalmology, the First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, China.
| |
Collapse
|
47
|
Wang X, Zhou Y, Luo C, Zhao J, Ji Y, Wang Z, Zheng P, Li D, Shi Y, Nishiura A, Matsumoto N, Honda Y, Xu B, Huang F. Senolytics ameliorate the failure of bone regeneration through the cell senescence-related inflammatory signalling pathway. Biomed Pharmacother 2024; 175:116606. [PMID: 38670048 DOI: 10.1016/j.biopha.2024.116606] [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: 11/25/2023] [Revised: 04/02/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Stress-induced premature senescent (SIPS) cells induced by various stresses deteriorate cell functions. Dasatinib and quercetin senolytics (DQ) can alleviate several diseases by eliminating senescent cells. α-tricalcium phosphate (α-TCP) is a widely used therapeutic approach for bone restoration but induces bone formation for a comparatively long time. Furthermore, bone infection exacerbates the detrimental prognosis of bone formation during material implant surgery due to oral cavity bacteria and unintentional contamination. It is essential to mitigate the inhibitory effects on bone formation during surgical procedures. Little is known that DQ improves bone formation in Lipopolysaccharide (LPS)-contaminated implants and its intrinsic mechanisms in the study of maxillofacial bone defects. This study aims to investigate whether the administration of DQ ameliorates the impairments on bone repair inflammation and contamination by eliminating SIPS cells. α-TCP and LPS-contaminated α-TCP were implanted into Sprague-Dawley rat calvaria bone defects. Simultaneously, bone formation in the bone defects was investigated with or without the oral administration of DQ. Micro-computed tomography and hematoxylin-eosin staining showed that senolytics significantly enhanced bone formation at the defect site. Histology and immunofluorescence staining revealed that the levels of p21- and p16-positive senescent cells, inflammation, macrophages, reactive oxygen species, and tartrate-resistant acid phosphatase-positive cells declined after administering DQ. DQ could partially alleviate the production of senescent markers and senescence-associated secretory phenotypes in vitro. This study indicates that LPS-contaminated α-TCP-based biomaterials can induce cellular senescence and hamper bone regeneration. Senolytics have significant therapeutic potential in reducing the adverse osteogenic effects of biomaterial-related infections and improving bone formation capacity.
Collapse
Affiliation(s)
- Xinchen Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China; Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yue Zhou
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan; Department of Stomatological Research Center, Affiliated Hospital of Yunnan University, Kunming, Yunnan, China
| | - Chuyi Luo
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Jianxin Zhao
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yuna Ji
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zheng Wang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Pengchao Zheng
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dingji Li
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuhan Shi
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Aki Nishiura
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Naoyuki Matsumoto
- Department of Orthodontics, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan
| | - Yoshitomo Honda
- Department of Oral Anatomy, Osaka Dental University, 8-1 Kuzuhahanazonocho, Hirakata, Osaka 573-1121, Japan.
| | - Baoshan Xu
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Fang Huang
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Institute of Stomatological Research, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Yadav RB, Pathak DP, Varshney R, Arora R. Elucidation of the Role of TRPV1, VEGF-A, TXA2, Redox Homeostasis, and Inflammatory Cascades in Protection against Cold Injuries by Herbosomal-Loaded PEG-Poloxamer Topical Formulation. ACS APPLIED BIO MATERIALS 2024; 7:2836-2850. [PMID: 38717017 DOI: 10.1021/acsabm.3c01197] [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: 05/21/2024]
Abstract
High-altitude regions, cold deserts, permafrost regions, and the polar region have some of the severest cold conditions on earth and pose immense perils of cold injuries to exposed individuals. Accidental and unintended exposures to severe cold, either unintentionally or due to occupational risks, can greatly increase the risk of serious conditions including hypothermia, trench foot, and cold injuries like frostbite. Cold-induced vasoconstriction and intracellular/intravascular ice crystal formation lead to hypoxic conditions at the cellular level. The condition is exacerbated in individuals having inadequate and proper covering and layering, particularly when large area of the body are exposed to extremely cold environments. There is a paucity of preventive and therapeutic pharmacological modalities that have been explored for managing and treating cold injuries. Given this, an efficient modality that can potentiate the healing of frostbite was investigated by studying various complex pathophysiological changes that occur during severe cold injuries. In the current research, we report the effectiveness and healing properties of a standardized formulation, i.e., a herbosomal-loaded PEG-poloxamer topical formulation (n-HPTF), on frostbite. The intricate mechanistic pathways modulated by the novel formulation have been elucidated by studying the pathophysiological sequelae that occur following severe cold exposures leading to frostbite. The results indicate that n-HPTF ameliorates the outcome of frostbite, as it activates positive sensory nerves widely distributed in the epidermis transient receptor potential vanilloid 1 (TRPV1), significantly (p < 0.05) upregulates cytokeratin-14, promotes angiogenesis (VEGF-A), prominently represses the expression of thromboxane formation (TXA2), and significantly (p < 0.05) restores levels of enzymatic (glutathione reductase, superoxide dismutase, and catalase) and nonenzymatic antioxidants (glutathione). Additionally, n-HPTF attenuates oxidative stress and the expression of inflammatory proteins PGF-2α, NFκB-p65, TNF-α, IL-6, IL-1β, malondialdehyde (MDA), advanced oxidative protein products (AOPP), and protein carbonylation (PCO). Masson's Trichrome staining showed that n-HPTF stimulates cellular proliferation, and increases collagen fiber deposition, which significantly (p < 0.05) promotes the healing of frostbitten tissue, as compared to control. We conclude that protection against severe cold injuries by n-HPTF is mediated via modulation of pathways involving TRPV1, VEGF-A, TXA2, redox homeostasis, and inflammatory cascades. The study is likely to have widespread implications for the prophylaxis and management of moderate-to-severe frostbite conditions.
Collapse
Affiliation(s)
- Renu Bala Yadav
- Disruptive and Deterrence Technologies Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, New Delhi 110054, India
- Delhi Pharmaceutical Science and Research University, Pushp Vihar, New Delhi 110017, India
| | - Dharam Pal Pathak
- Delhi Pharmaceutical Science and Research University, Pushp Vihar, New Delhi 110017, India
| | - Rajeev Varshney
- Disruptive and Deterrence Technologies Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, New Delhi 110054, India
| | - Rajesh Arora
- Disruptive and Deterrence Technologies Division, Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, New Delhi 110054, India
| |
Collapse
|
49
|
Jin S, Wang H, Zhang X, Song M, Liu B, Sun W. Emerging regulatory mechanisms in cardiovascular disease: Ferroptosis. Biomed Pharmacother 2024; 174:116457. [PMID: 38518600 DOI: 10.1016/j.biopha.2024.116457] [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: 12/21/2023] [Revised: 03/03/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024] Open
Abstract
Ferroptosis, distinct from apoptosis, necrosis, autophagy, and other types of cell death, is a novel iron-dependent regulated cell death characterized by the accumulation of lipid peroxides and redox imbalance with distinct morphological, biochemical, and genetic features. Dysregulation of iron homeostasis, the disruption of antioxidative stress pathways and lipid peroxidation are crucial in ferroptosis. Ferroptosis is involved in the pathogenesis of several cardiovascular diseases, including atherosclerosis, cardiomyopathy, myocardial infarction, ischemia-reperfusion injury, abdominal aortic aneurysm, aortic dissection, and heart failure. Therefore, a comprehensive understanding of the mechanisms that regulate ferroptosis in cardiovascular diseases will enhance the prevention and treatment of these diseases. This review discusses the latest findings on the molecular mechanisms of ferroptosis and its regulation in cardiovascular diseases, the application of ferroptosis modulators in cardiovascular diseases, and the role of traditional Chinese medicines in ferroptosis regulation to provide a comprehensive understanding of the pathogenesis of cardiovascular diseases and identify new prevention and treatment options.
Collapse
Affiliation(s)
- Sijie Jin
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Xiaohao Zhang
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Mengyang Song
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 4026 YaTai Street, Changchun 130041, China.
| |
Collapse
|
50
|
Cox A, Brown KC, Valentovic MA. The E-liquid flavoring vanillin alters energy and autophagic pathways in human proximal tubule (HK-2) epithelial cells. Chem Biol Interact 2024; 394:111003. [PMID: 38608998 DOI: 10.1016/j.cbi.2024.111003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/04/2024] [Accepted: 04/07/2024] [Indexed: 04/14/2024]
Abstract
The use of flavored e-liquids in electronic nicotine delivery systems (ENDS) has become very popular in recent years, but effects of these products have not been well characterized outside the lung. In this study, acute exposure to the popular flavoring vanillin (VAN) was performed on human proximal tubule (HK-2) kidney cells. Cells were exposed to 0-1000 μM VAN for 24 or 48 h and cellular stress responses were determined. Mitochondrial viability using MTT assay showed a significant decrease between the control and 1000 μM group by 48 h. Seahorse XFp analysis showed significantly increased basal respiration, ATP production, and proton leak after 24 h exposure. By 48 h exposure, these parameters remained significantly increased in addition to non-mitochondrial respiration and maximal respiration. Glycolytic activity after 24 h exposure showed significant decreases in glycolysis, glycolytic capacity, glycolytic reserve, and non-glycolytic acidification. The autophagy markers microtubule-associated protein 1A/1B light chain 3 (LC3B-I and LC3B-II) were probed via western blotting. The ratio of LC3B-II/LC3B-I was significantly increased after 24 h exposure to VAN, but by 48 h this ratio significantly decreased. The mitophagy marker PINK1 showed an increasing trend at 24 h, and its downstream target Parkin was significantly increased between the control and 750 μM group only. Finally, the oxidative stress marker 4-HNE was significantly decreased after 48 h exposure to VAN. These results indicate that acute exposure to VAN in the kidney HK-2 model can induce energy and autophagic changes within the cell.
Collapse
Affiliation(s)
- Ashley Cox
- Department of Biomedical Sciences, Toxicology Research Cluster, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 25701, USA
| | - Kathleen C Brown
- Department of Biomedical Sciences, Toxicology Research Cluster, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 25701, USA
| | - Monica A Valentovic
- Department of Biomedical Sciences, Toxicology Research Cluster, Marshall University Joan C. Edwards School of Medicine, Huntington, WV, 25701, USA.
| |
Collapse
|