1
|
Gao Y, Han R, Guo Z, Zhuang Z, Fu J, Hou Y, Yu J, Tang K. Multi-pathway copper metabolisms regulation based on an engineered copper/ferrous nanoplatform for enhanced tumor cuproptosis therapy. Colloids Surf B Biointerfaces 2025; 252:114682. [PMID: 40215641 DOI: 10.1016/j.colsurfb.2025.114682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 05/18/2025]
Abstract
Cuproptosis is a currently discovered programmed cell death modality driven by copper (Cu) ions, which shows potential application prospects in overcoming apoptotic resistance in cancer therapy due to its unique mechanism. Nevertheless, the efficiency of cuproptosis is restricted by strict Cu metabolism regulation. Herein, elesclomol (ES) and glucose oxidase (GOx) co-loaded CuFe2O4 (CF) nanoplatform (termed as CFEG) was elaborately engineered to boost cuproptosis through multi-pathway copper metabolisms regulation. After triggered by tumor-overexpressed glutathione (GSH), the released ES continuously chelated and targeted transport Cu ions through a shuttle mechanism to mitochondria where cuproptosis was initiated, which dramatically improved the influx efficiency of Cu. Additionally, GOx-mediated glucose oxidation reaction together with Cu and Fe ions stimulated Fenton reaction simultaneously amplified intracellular oxidative pressure by hydrogen peroxide (H2O2) self-supply and subsequent hydroxyl radical (•OH) generation, which down-regulated Cu exporter ATP7A expression and inhibited Cu ions efflux, thereby exacerbating cuproptosis. Furthermore, the consumption of GSH simultaneously reduced the chelation of GSH with Cu and promote the •OH generation, further potentiating the occurrence of cuproptosis. Collectively, such the multi-pathway copper metabolisms regulation including improved Cu influx, inhibited Cu efflux and GSH depletion significantly boosted cuproptosis, which synergized with photothermal effect of CF to efficiently repressed the growth of tumor in mice without causing systemic toxicity. This work provides a multivariate mode for enhanced tumor cuproptosis therapy, and may also inspire the design of advanced cuproptosis-related nanomedicine system.
Collapse
Affiliation(s)
- Yun Gao
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Renlu Han
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China; Zhenhai Institute of Mass Spectrometry, Ningbo 315211, PR China.
| | - Zhen Guo
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Zekai Zhuang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Jie Fu
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China
| | - Yafei Hou
- School of Physical Science and Technology, Ningbo University, Ningbo 315211, PR China.
| | - Jiancheng Yu
- Faculty of Electrical Engineering and Computer Science, Ningbo University, Ningbo 315211, PR China
| | - Keqi Tang
- Zhejiang Engineering Research Center of Advanced Mass Spectrometry and Clinical Application, Institute of Mass Spectrometry, School of MaterialsScience & Chemical Engineering, Ningbo University, Ningbo 315211, PR China; Zhenhai Institute of Mass Spectrometry, Ningbo 315211, PR China.
| |
Collapse
|
2
|
Zhi DD, He XY, Yang LF, Xue YF, Liu YQ, Yue D, Feng YN, Dong K, Tian YK. Royal jelly acid alleviates diet-induced hyperlipidemia through regulation of oxidative stress and tryptophan metabolism. Eur J Pharmacol 2025; 998:177500. [PMID: 40086581 DOI: 10.1016/j.ejphar.2025.177500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 02/24/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Royal jelly acid is a unique unsaturated fatty acid isolated from royal jelly. Recently, royal jelly acid was proposed to have potential therapeutic effects on hyperlipidemia. However, its effect on hyperlipidemia and the underlying molecular mechanism remains unclear. Therefore, in this study, we analyzed the mechanism of anti-hyperlipidemia of royal jelly acid through animal experiments and plasma metabolomics in conjunction with human network pharmacology and molecular docking analyses. We found that royal jelly acid could significantly decrease the serum lipid levels, ameliorate hepatic pathological injury, and reduce the level of oxidative stress in the experimental rats. A total of 41 key metabolites and 10 hub targets played key roles in the exertion of anti-hyperlipidemic effects, including tumor necrosis factor (TNF), insulin (INS) and epidermal growth factor receptor (EGFR). A total of 24 pathways, including tryptophan, citrate cycle, and arachidonic acid metabolisms, were identified as the key pathways involved in royal jelly acid-alleviated hyperlipidemia. The present findings provide new insights into the pathogenesis, diagnosis, and treatment targets of hyperlipidemia as well as contribute to the development and utilization of royal jelly acid related products.
Collapse
Affiliation(s)
- Dan-Dan Zhi
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Xi-Ying He
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Lin-Fu Yang
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yun-Fei Xue
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yi-Qiu Liu
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Dan Yue
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yi-Nan Feng
- College of Food Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Kun Dong
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
| | - Ya-Kai Tian
- Yunnan Provincial Engineering and Research Center for Sustainable Utilization of Honey Bee Resources, Eastern Bee Research Institute, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
3
|
Sharma P, Mohanty S, Ahmad Y. Decoding Proteomic cross-talk between hypobaric and normobaric hypoxia: Integrative analysis of oxidative stress, cytoskeleton remodeling, and inflammatory pathways. Life Sci 2025; 371:123611. [PMID: 40187642 DOI: 10.1016/j.lfs.2025.123611] [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: 12/26/2024] [Revised: 03/22/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
AIMS To investigate the differential regulation of proteomic landscapes elicited by hypobaric hypoxia (HH) and normobaric hypoxia (NH) and to shed light on the molecular cross-talk underlying pre-acclimatization strategies. MATERIALS AND METHODS Label-free LCMS-MS quantitative proteomics was employed to evaluate the lung tissues of SD rats (n = 6) subjected to 6 h of acute HH at 25,000 ft associated with reduced barometric pressure, 282 mmHg, and NH at 8 % FiO2. KEY FINDINGS Our findings indicate that NH facilitated the minimal downregulation of proteins involved in maintaining pulmonary cytoskeleton integrity, including calpain 2, vitronectin, and beta-arrestin 1, whereas HH leads to severe downregulation of these proteins, causing a greater cytoskeleton disruption. Proteins contributing to redox homeostasis such as iNOS and SOD, were upregulated in both hypoxic conditions. However, SIRT1-mediated ROS-triggered proteins, including FOXO1 and FOXO4, exhibited upregulation in HH and downregulation in NH. Other proteins, HIF-1α and IDH, were upregulated in HH compared to NH. Additionally, Hemopexin was severely downregulated in HH relative to NH. SIGNIFICANCE For the first time, this study uncovers the comparative proteomic analysis of two distinct pre-acclimatization interventions by employing varied hypoxia modeling strategies highlighting the key molecular mechanism involved in HH acclimatization induced by differential hypoxia simulating technique.
Collapse
Affiliation(s)
- Poornima Sharma
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Swaraj Mohanty
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India
| | - Yasmin Ahmad
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence R&D Organization (DRDO), Timarpur, New Delhi 110054, India.
| |
Collapse
|
4
|
Luo L, Liu Q, Zhang Y, Yu X, Wang L, Sun W, Li T, Xu B, Zhang K, Yu Y, Cui C, Li C, Mei L. Precisely edited gut microbiota by tungsten-doped Prussian blue nanoparticles for the treatment of inflammatory bowel disease. J Control Release 2025; 382:113755. [PMID: 40258476 DOI: 10.1016/j.jconrel.2025.113755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/16/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025]
Abstract
Inflammatory bowel disease (IBD) is characterized by recurring gastrointestinal inflammation, accompanied by a significant rise in global prevalence and disease severity. The overaccumulation of reactive oxygen and nitrogen species (RONS) in the intestinal environment disrupts redox homeostasis and drives pathological overgrowth of Escherichia coli, which are central to IBD pathogenesis. Herein, we designed a multifunctional nanozyme (W-PB) to enable sustained and targeted regulation of intestinal homeostasis through dual mechanisms: specific inhibition of E. coli overgrowth during colitis and efficient RONS clearance. To ensure colon-specific delivery, W-PB was encapsulated in an electrostatically crosslinked hydrogel composed of alginate and chitosan. This formulation protects W-PB from degradation in harsh gastrointestinal conditions and releases the nanoparticles selectively under weakly alkaline intestinal pH. The released tungsten ions suppress E. coli growth via competitive displacement of molybdenum in the molybdopterin cofactor, while W-PB simultaneously neutralizes excess RONS to shield intestinal cells from oxidative damage. In DSS-induced colitis models, the W-PB gel demonstrated significant therapeutic efficacy, achieved through intestinal microbiota remodeling and oxidative stress mitigation.
Collapse
Affiliation(s)
- Lingpeng Luo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Qingyun Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Yushi Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Xuya Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Ling Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, PR China
| | - Weiting Sun
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Tingxuan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Bin Xu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China
| | - Kai Zhang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yongkang Yu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| | - Chunhui Cui
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, PR China.
| | - Chen Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Tianjin Institute of Health Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 301600, PR China; Furong Laboratory, Central South University, Changsha 410008, PR China.
| |
Collapse
|
5
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 PMCID: PMC12146052 DOI: 10.1016/j.mcn.2025.104003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
6
|
Mukherjee U, Ghosh S, Maitra S. Multi-mechanistic effects of bisphenol A on testicular dysfunction and endocrine disruption in adult male Labeo bata: oxidative stress, inflammation, and dysregulated energy sensors. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 374:126231. [PMID: 40221117 DOI: 10.1016/j.envpol.2025.126231] [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: 12/22/2024] [Revised: 03/24/2025] [Accepted: 04/10/2025] [Indexed: 04/14/2025]
Abstract
Leaching of BPA, a potent endocrine disruptor, from microplastics in aquatic environments has garnered renewed interest in its impact on male reproduction. However, the mechanisms underlying BPA modulation of testicular metabolic, endocrine, and autocrine-paracrine axes in teleost remained relatively unexplored, prompting us to examine BPA-induced testicular dysfunction in adult male Labeo bata. Present results demonstrate that congruent with reduced gonadosomatic index (GSI), chronic BPA treatment at environmentally relevant concentrations enhanced ROS synthesis, oxidative stress, and testicular histopathology characterized by reduced diameter of seminiferous tubules, a sharp decline in spermatozoa, and elevated fibrosis during the spawning season. Elevated NO levels, pro-inflammatory cytokines and NLRP3-inflammasome activation correlated with cleaved caspase -8, -9, -3 activation, altered Bax/Bcl-2 ratio, heightened caspase -3 immunolocalization and TUNEL staining, suggesting DNA damage and apoptosis in BPA-treated testis. Besides, BPA attenuation of cyclin B synthesis and p-p34cdc2 (Thr161) phosphorylation (activation), markers associated with altered meiotic cell cycle progression, corresponded with heightened apoptosis and loss of spermatogenic cells. Importantly, disrupted estrogen and membrane progestin receptor (ERα, ERβ, mPRα-PGRMC1) homeostasis, alteration in gonadotropin receptor (LHCGR) and steroidogenic markers and elevated P450 aromatase immunolocalization indicate endocrine disruption and heightened estrogenic influence of BPA, inducing testicular dysfunction. Congruently, BPA modulation of cellular energy sensors (SIRT1/p-AMPKα/PGC-1α), factors influencing testicular endocrine, paracrine and prostaglandin signalling, and MAPK modulation might indicate pleiotropic regulation by BPA triggering reproductive toxicity. While Pearson's correlation and elevated multi-biomarker IBR indices support testicular dysfunction, present findings highlight the need for further research initiatives and strict regimens to combat ecological risks posed by BPA contamination.
Collapse
Affiliation(s)
- Urmi Mukherjee
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Soumyajyoti Ghosh
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India
| | - Sudipta Maitra
- Molecular and Cellular Endocrinology Laboratory, Department of Zoology, Visva-Bharati University, Santiniketan, 731235, India.
| |
Collapse
|
7
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Elkin AM, Robbins S, Barros CS, Bossing T. The Critical Balance Between Quiescence and Reactivation of Neural Stem Cells. Biomolecules 2025; 15:672. [PMID: 40427564 PMCID: PMC12108614 DOI: 10.3390/biom15050672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Neural stem cells (NSC) are multipotent, self-renewing cells that give rise to all neural cell types within the central nervous system. During adulthood, most NSCs exist in a quiescent state which can be reactivated in response to metabolic and signalling changes, allowing for long-term continuous neurogenesis and response to injury. Ensuring a critical balance between quiescence and reactivation is required to maintain the limited NSC reservoir and neural replenishment throughout lifetime. The precise mechanisms and signalling pathways behind this balance are at the focus of current research. In this review, we highlight and discuss recent studies using Drosophila, mammalian and zebrafish models contributing to the understanding of molecular mechanisms underlying quiescence and reactivation of NSCs.
Collapse
Affiliation(s)
| | | | - Claudia S. Barros
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth PL6 8BU, UK; (A.M.E.); (S.R.)
| | - Torsten Bossing
- Peninsula Medical School, Faculty of Health, University of Plymouth, Plymouth PL6 8BU, UK; (A.M.E.); (S.R.)
| |
Collapse
|
9
|
Pappert FA, Wüst VA, Fontanes Eguiguren C, Roth O. Surviving on Limited Resources: Effects of Caloric Restriction on Growth, Gene Expression and Gut Microbiota in a Species With Male Pregnancy (Hippocampus erectus). Mol Ecol 2025; 34:e17754. [PMID: 40192444 PMCID: PMC12010458 DOI: 10.1111/mec.17754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/18/2025] [Accepted: 03/25/2025] [Indexed: 04/22/2025]
Abstract
Caloric restriction (CR) studies have traditionally focused on species with conventional reproductive roles, emphasising female's greater investment in costly gametes and parental care. While the divergent impact of CR on males and females is evident across species, the factors driving this variation, that is, resource allocation to reproductive elements as part of distinct life history strategies, remain unclear. To address this, we investigated the effects of CR on development, gene expression and intestinal microbiota in the lined seahorse Hippocampus erectus, a species with male pregnancy, where fathers invest in offspring through gestation. Juvenile seahorses were subjected to ad libitum (AL) or CR feeding for 5 months. CR stunted male growth and brood pouch development, reflecting the energy demands of this crucial parental care trait. However, condition index declined in CR females but not males, while ovarian weight remained unchanged. Transcriptome analysis demonstrated organ- and sex-specific responses to CR with distinct lipid and energy-related pathways activated in male and female livers, indicative of survival enhancement strategies. CR had minimal impact on genes associated with spermatogenesis, but downregulated lipid metabolic and inflammatory genes in ovaries, emphasising the importance of pre-copulatory resource allocation in female gametes. CR strongly shaped gut microbial composition, creating distinct communities from AL seahorses while also driving sex-specific taxonomic differences. Our research indicates that nutrient limitation's impact on males and females is influenced by their allocation of resources to reproduction and parental investment. We underscore the significance of studying species with diverse reproductive strategies, sex roles and life-history strategies.
Collapse
Affiliation(s)
- Freya Adele Pappert
- Marine Evolutionary Biology, Zoological InstituteChristian‐Albrechts‐Universität KielKielGermany
- Evolutionary Ecology of Marine FishesHelmholtz‐Centre for Ocean Research Kiel (GEOMAR)KielGermany
| | - Vincent Alexander Wüst
- Marine Evolutionary Biology, Zoological InstituteChristian‐Albrechts‐Universität KielKielGermany
| | | | - Olivia Roth
- Marine Evolutionary Biology, Zoological InstituteChristian‐Albrechts‐Universität KielKielGermany
- Evolutionary Ecology of Marine FishesHelmholtz‐Centre for Ocean Research Kiel (GEOMAR)KielGermany
| |
Collapse
|
10
|
Wang Y, Gu C, Zhao H, Li Z, Thirupathi A. Redox signaling‑mediated muscle atrophy in ACL injury: Role of physical exercise (Review). Mol Med Rep 2025; 31:119. [PMID: 40052558 PMCID: PMC11904765 DOI: 10.3892/mmr.2025.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/03/2025] [Indexed: 03/15/2025] Open
Abstract
Muscle atrophy frequently occurs in patients with anterior cruciate ligament (ACL) injury, despite active participation in muscle strengthening programs. Without appropriate countermeasures such as exercise and pharmacological interventions, the atrophy may worsen. At the cellular and molecular levels, various protein synthesis‑related pathways and redox‑dependent molecules regulate processes associated with atrophy by activating or deactivating key signaling pathways. Muscle atrophy and the associated dysfunction can be reversed by physical exercise, which increases protein synthesis, thereby improving muscle strength and function around the ACL. However, the influence of different features of exercise protocols, including exercise type, intensity and duration, as well as the individual capacity of the patient, on the activity of the aforementioned pathways requires further investigation. Additionally, the mechanism by which redox‑sensitive molecules attenuate atrophy in ACL injury remains to be fully understood. The present review discusses exercise, signaling pathways and muscle atrophy in ACL injury, and highlights potential therapeutic strategies. These findings may also have implications for other joint diseases associated with ACL‑related injury.
Collapse
Affiliation(s)
- Yucong Wang
- Department of Joint Surgery, Ningbo No. 9 Hospital, Ningbo, Zhejiang 315020, P.R. China
| | - Chunxiao Gu
- Department of Joint Surgery, Ningbo No. 9 Hospital, Ningbo, Zhejiang 315020, P.R. China
| | - Hui Zhao
- Department of Joint Surgery, Ningbo No. 9 Hospital, Ningbo, Zhejiang 315020, P.R. China
| | - Zhongzheng Li
- Department of Joint Surgery, Ningbo No. 9 Hospital, Ningbo, Zhejiang 315020, P.R. China
| | - Anand Thirupathi
- Faculty of Sports Science, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
11
|
López-López M, Regueiro U, Bravo S, Pena C, Pastoriza Y, Conde-Amboage M, Hervella P, Lema I. Tear Proteomic Analysis From Offspring of Keratoconus Patients: New Insights Into Corneal Biomechanical Weakness and Disease Risk Stages. Invest Ophthalmol Vis Sci 2025; 66:41. [PMID: 40434347 DOI: 10.1167/iovs.66.5.41] [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: 05/29/2025] Open
Abstract
Purpose To analyze the tear proteome of keratoconus offspring (O-KC) and assess the molecular drivers underlying corneal biomechanical weakening at KC-risk stages. Methods This cross-sectional study included 80 O-KC young participants and 42 controls without a KC family history. Based on the corneal biomechanical behavior, O-KC eyes were classified as low, moderate, and high-risk of KC development (O-KC-LR, O-KC-MR, O-KC-HR). Tear fluid was extracted using Schirmer strips, and the proteomic profile was mapped using LC-MS/MS. Bioinformatic tools were used to determine the dysregulated protein's biological implications. The sensitivity-specificity of each biomarker for differentiating between controls and O-KC groups was determined. Logistic regression analysis (LRA) identified the optimal subset of predictors for modeling each biomechanical condition's probability. Results Twenty-nine percent of O-KC eyes showed moderate/high alterations in corneal biomechanical behavior. Fifteen proteins were dysregulated in the tear samples of O-KC groups compared to controls (P < 0.05). Dysregulated proteins were associated with oxidative stress, cell adhesion, cytoskeleton organization, and mechanotransduction paths such as RhoA, mTOR, or E-cadherin/N-cadherin signaling. LRA determined three protein panels with high sensitivity-specificity for discriminating between the control and O-KC groups with different biomechanical risks. Conclusions This study revealed promising new biomarkers for early detection of KC risk. Oxidative stress and cellular structural alterations seem to begin long before clinical signs and even before the biomechanical alterations can be detected with current clinical tools. Understanding how an initial imbalance of oxidative stress affects cellular mechanobiology is critical to developing new therapeutic strategies for the early treatment of KC.
Collapse
Affiliation(s)
- Maite López-López
- Corneal Neurodegeneration Group (RENOIR), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Departamento de Ciruxía e Especialidades Médico-Cirúrxicas, Facultade de Óptica e Optometría, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Uxía Regueiro
- Corneal Neurodegeneration Group (RENOIR), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Departamento de Ciruxía e Especialidades Médico-Cirúrxicas, Facultade de Óptica e Optometría, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Susana Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Carmen Pena
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Yaiza Pastoriza
- Corneal Neurodegeneration Group (RENOIR), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Departamento de Ciruxía e Especialidades Médico-Cirúrxicas, Facultade de Óptica e Optometría, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mercedes Conde-Amboage
- Department of Statistics, Mathematical Analysis and Optimization, Universidade de Santiago de Compostela (USC), Spain
- CITMAga, Santiago de Compostela, Spain
| | - Pablo Hervella
- Neuroimaging and Biotechnology Group (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Isabel Lema
- Corneal Neurodegeneration Group (RENOIR), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Departamento de Ciruxía e Especialidades Médico-Cirúrxicas, Facultade de Óptica e Optometría, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Instituto Galego de Oftalmoloxía (INGO), Complexo Hospitalario Universitario de Santiago (CHUS), Santiago de Compostela, Spain
| |
Collapse
|
12
|
Zhang D, Liang Q, Jiang J, Liu W, Chu Y, Chen Z, Li B, Chen T, Tsao JR, Hu K. SIRT3 mitigates dry eye disease through the activation of autophagy by deacetylation of FOXO1. Exp Eye Res 2025; 254:110328. [PMID: 40064414 DOI: 10.1016/j.exer.2025.110328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/20/2025]
Abstract
Dry eye disease (DED) is a complex ocular condition characterized by oxidative stress, inflammation, and apoptosis. An increasing number of studies suggest that Sirtuin3 (SIRT3), a mitochondrial deacetylase, may offer protection against related pathologies. Despite these indications, the precise function and underlying mechanisms of SIRT3 in the context of DED have not been fully elucidated. Here, we observed a decline in SIRT3 expression in human corneal epithelial cells (HCE-Ts) and the corneal conjunctiva of mice as the disease advanced. Overexpression of SIRT3 in HCE-Ts reduced the accumulation of reactive oxygen species (ROS), inflammatory cytokines, and the rate of apoptosis, while its inhibition had the opposite effect. Importantly, the function of SIRT3 was exerted through the enhancement of autophagic flux. Further studies have shown that chloroquine-induced inhibition of autophagy neutralized the beneficial effects of SIRT3. In our in vivo experiments, the application of eye drops containing a SIRT3 agonist ameliorated the symptoms of DED and increased corneal autophagy in mice. Mechanistically, our study identified that the deacetylation and nuclear translocation of FOXO1 (Forkhead box O1) are pivotal for the SIRT3-mediated enhancement of autophagic flux. These findings posit that SIRT3 as an encouraging therapeutic target for DED, offering new insights into the disease's underlying mechanisms.
Collapse
Affiliation(s)
- Di Zhang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Qi Liang
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, Zhejiang China
| | - Jiaxuan Jiang
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Wei Liu
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, China
| | - Yiran Chu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Zeying Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Boda Li
- Department of Ophthalmology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, 321 Zhongshan Road, Nanjing, China
| | - Taige Chen
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China; Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Jia-Ruei Tsao
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China
| | - Kai Hu
- Department of Ophthalmology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, China.
| |
Collapse
|
13
|
Kalliampakou KI, Athanasopoulou E, Spanou A, Flemetakis E, Tsironi T. In vitro cytotoxicity evaluation of a CMC-SA edible packaging film for migration and safety assessment. Sci Rep 2025; 15:13304. [PMID: 40246963 PMCID: PMC12006323 DOI: 10.1038/s41598-025-98163-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/09/2025] [Indexed: 04/19/2025] Open
Abstract
Edible raw materials have gained attention as sustainable food packaging films, which are often considered a priori safe for human consumption. However, cytotoxicity issues may arise due to the incorporation of additives or modifications of film functionality during the manufacturing process. This study introduces an integrated methodology for the evaluation of potential migration of cytotoxic substances from materials used for the development of conventional and biodegradable food packaging. Carboxymethyl cellulose (CMC) and sodium alginate (SA) were tested as raw materials of an edible (CMC-SA) film, while a low-density-polyethylene (LDPE) film was tested as a conventional material. The CMC-SA film exhibited higher water vapor transmission rate and water vapor permeability, and lower hydrophobicity compared to LDPE (WVTRCMC-SA=1457.87 vs. WVTRLDPE=3.43 g×m-2×day-1, WVPCMC-SA=43.24 vs. WVPLDPE=0.0048 g×m-2×mm×day-1×kPa-1 and CACMC-SA=52.05 vs. CALDPE=94.28°, respectively). An analytical protocol based on EU Regulation 10/2011 was introduced, to evaluate the potential migration of cytotoxic packaging substances into food simulants, using different human cells. Caco2 cells were used to simulate human intestine, whereas Huh7 and Immortalized Human Hepatocytes (IHH) cells simulated human liver. Cell viability assays and gene expression results indicated that substances migrating from the tested packaging materials neither produced cell cytotoxicity, nor induced oxidative stress to Caco2 cells.
Collapse
Affiliation(s)
- Katerina I Kalliampakou
- Laboratory of Food Process Engineering, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece
- Laboratory of Molecular Biology, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece
| | - Evmorfia Athanasopoulou
- Laboratory of Food Process Engineering, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece
| | - Aikaterini Spanou
- Laboratory of Food Process Engineering, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece
| | - Emmanouil Flemetakis
- Laboratory of Molecular Biology, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece
| | - Theofania Tsironi
- Laboratory of Food Process Engineering, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, Athens, 11855, Greece.
| |
Collapse
|
14
|
Li P, Meng X, Lu T, Sun C, Song G. Synergistic Effect of ROS and p38 MAPK in Apoptosis of TM4 Cells Induced by Titanium Dioxide Nanoparticles. J Appl Toxicol 2025. [PMID: 40229128 DOI: 10.1002/jat.4789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 03/31/2025] [Accepted: 04/06/2025] [Indexed: 04/16/2025]
Abstract
The adverse effects of titanium dioxide nanoparticles (TiO2 NPs) on the integrity of the blood-testis barrier (BTB) are widely recognized. However, the underlying mechanisms remain incompletely understood. The integrity of the BTB is imperative for the preservation of male reproductive health. TM4 cells, which are major component of the BTB, play a critical role in its integrity. The apoptosis of TM4 cells is closely associated with the disruption of the BTB. Therefore, we selected TM4 cells as experimental models to investigate the apoptosis induced by TiO2 NPs and the underlying mechanisms. Cell viability, excessive production of reactive oxygen species (ROS), activation of p38 mitogen-activated protein kinase (MAPK) pathway, and apoptosis-related protein expression levels were determined under various concentrations (50, 100, 150, and 200 μg/mL) of TiO2 NPs exposure. The results indicate that TiO2 NPs induced the overproduction of ROS and activated the p38 MAPK signaling pathway, which subsequently led to apoptosis. The ROS scavenger N-acetylcysteine (NAC) was able to suppress the activation of p38 MAPK pathway induced by TiO2 NPs, while the p38 MAPK inhibitor SB203580 mitigated TiO2 NPs-induced ROS overproduction and subsequent apoptosis, suggesting an interplay between ROS overproduction and p38 MAPK pathway activation. In summary, TiO2 NPs induced mitochondrial apoptosis via the ROS-p38 MAPK axis. A positive feedback regulatory mechanism exists between the two processes, promoting apoptosis in TM4 cells through a synergistic effect.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Xiaojia Meng
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
- Medical Research Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tianjiao Lu
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
- Beidaihe Rest and Recuperation Center of PLA, Qinhuangdao, China
| | - Chenhao Sun
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| | - Guanling Song
- Department of Preventive Medicine/the Key Laboratory for Prevention and Control of Emerging Infectious Diseases and Public Health Security, the Xinjiang Production and Construction Corps, School of Medicine, Shihezi University, Shihezi, China
| |
Collapse
|
15
|
Ma X, Lin Y, Zhang L, Huang Z, Zhang Y, Fu X, Li P. The dual missions of FoxO3a in inflammatory diseases: Regulation of antioxidant enzymes and involvement in programmed cell death. Int Immunopharmacol 2025; 151:114369. [PMID: 40031428 DOI: 10.1016/j.intimp.2025.114369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
The transcription factor FoxO3a plays a crucial role in the process of cells adapting to various stress conditions. Multiple post - translational modifications and epigenetic mechanisms work together to precisely regulate the activity of FoxO3a, influencing its subcellular localization, stability, interactions with other proteins, DNA - binding affinity, and transcriptional regulatory capacity. Under different chemical signal stimuli and subcellular environments, the activation of FoxO3a triggered by oxidative stress can initiate diverse transcriptional programs, which are essential for the body to resist oxidative damage. In the development and progression of inflammatory diseases, FoxO3a exerts an important function by regulating the expression levels of antioxidant enzymes and participating in key physiological processes such as programmed cell death. This article comprehensively reviews the structural characteristics, mechanism of action of FoxO3a, as well as its functions in regulating antioxidant enzymes and programmed cell death. The aim is to deeply explore the potential of FoxO3a as a potential therapeutic target for preventing and treating damages such as inflammatory diseases caused by cellular stress.
Collapse
Affiliation(s)
- Xiangli Ma
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yujie Lin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
16
|
Liang Y, Tian Y, Liu J, Lei P, Sun X, Zhang H, Lei Y. Smart Bioorthogonal Catalytic Factory for Glaucoma Therapy. NANO LETTERS 2025; 25:5502-5511. [PMID: 40102044 DOI: 10.1021/acs.nanolett.5c00607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Glaucoma is characterized by high intraocular pressure (IOP), oxidative stress, and distinct optic nerve damage. Natural enzymes have unparalleled advantages in the treatment of glaucoma due to their high efficiency, specificity, and selectivity. However, their poor stability and recoverability have constrained their application. The selection of good immobilization carriers is an effective strategy to protect natural enzymes. Here, we employ a mild room-temperature aqueous-phase enzyme immobilization technique to immobilize superoxide dismutase and catalase. Then, l-arginine is added to the pores and further modified with DSPE-mPEG to construct a bioorthogonal catalytic factory (SC@COF-L-D) with excellent biocompatibility. This strategy greatly protects the natural enzyme from inactivation and improves the operational stability. SC@COF-L-D can scavenge a large amount of reactive oxygen species to reduce oxidative/nitrative damage and activate the soluble guanylate cyclase pathway, thereby lowering the IOP for effective treatment of glaucoma. This work provides a paradigm for the design of materials for glaucoma therapy.
Collapse
Affiliation(s)
- Yuan Liang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, Jiangxi 341000, China
| | - Yi Tian
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Jiamin Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Pengpeng Lei
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Ganzhou, Jiangxi 341000, China
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yuan Lei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai 200031, China
| |
Collapse
|
17
|
Jacobsen V, Kunisch E, Merle C, Xue B, Zheng K, Renkawitz T, Boccaccini AR, Westhauser F. Cerium-doped mesoporous bioactive glass nanoparticles reduce oxidative stress and adipogenic differentiation in human bone marrow-derived mesenchymal stromal cells. J Trace Elem Med Biol 2025; 88:127617. [PMID: 39952087 DOI: 10.1016/j.jtemb.2025.127617] [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: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025]
Abstract
Increased levels of reactive oxygen species (ROS) favor adipogenic over osteogenic differentiation in human bone-marrow derived mesenchymal stromal cells (BMSCs). Therefore, biomaterials containing ROS-suppressing elements such as Cerium (Ce) have been introduced to cell-based bone-tissue-engineering (BTE) approaches. This study was conducted to assess the efficacy of Ce-doped mesoporous bioactive glass nanoparticles (MBGNs) in reducing ROS levels and subsequently inhibiting the adipogenic differentiation of BMSCs. To this end, BMSCs were cultivated in adipogenesis inducing medium (AIM) and exposed to ions released from Ce-free MBGNs (composition in mol%: 86SiO2-14CaO), Low-Ce-MBGNs (composition in mol%: 86.6SiO2-12.1CaO-1.3CeO2) and High-Ce-MBGNs (composition in mol%: 86.0SiO2-11.8CaO-2.2CeO2). The influence of the different MBGNs on the expression of adipogenic and ROS-scavenging genes was assessed as well as their influence on lipid formation and the physical presence of ROS. Ce-MBGNs significantly reduced lipid production and the expression of adipogenic marker genes when compared to BMSCs cultivated in the presence of MBGNs or AIM alone. Furthermore, ROS levels were decreased by Ce-MBGNs alongside an upregulation of the expression of genes encoding for ROS-scavenging enzymes. Ce-MBGNs have proven their antioxidative potential. Mediated by the reduction of ROS, the undesired differentiation of BMSCs towards adipogenic lineage within BTE applications has been effectively suppressed. Ce-MBGNs target differentiation pathways in BMSCs precisely and therefore constitute an attractive biomaterial in the field of ion-based BTE.
Collapse
Affiliation(s)
- V Jacobsen
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany
| | - E Kunisch
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany
| | - C Merle
- Joint Replacement Centre, Orthopedic Surgery Paulinenhilfe, Diakonie-Klinikum Stuttgart, Rosenbergstraße 38, Stuttgart 70176, Germany
| | - B Xue
- Translational Medicine Research Center, Children's Hospital of Nanjing Medical University, 136 Hanzhong Rd., Nanjing 210029, China
| | - K Zheng
- Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, 136 Hanzhong Rd., Nanjing 210029, China
| | - T Renkawitz
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany; Department of Orthopaedics, Regensburg University, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, Bad Abbach 93077, Germany
| | - A R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nuremberg, Cauerstrasse 6, 91058 Erlangen, Germany.
| | - F Westhauser
- Department of Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstraße 200a, Heidelberg 69118, Germany; Department of Orthopaedics, Regensburg University, Asklepios Klinikum Bad Abbach, Kaiser-Karl V.-Allee 3, Bad Abbach 93077, Germany.
| |
Collapse
|
18
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
19
|
Lučić I, Mlinarić M, Čipak Gašparović A, Milković L. The Influence of AQP5 on the Response to Hydrogen Peroxide in Breast Cancer Cell Lines. Int J Mol Sci 2025; 26:3243. [PMID: 40244097 PMCID: PMC11989815 DOI: 10.3390/ijms26073243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Breast cancer is a heterogeneous disease with varying responses to therapies. While targeted treatments have advanced, conventional therapies inducing oxidative stress remain widely used. H2O2 has emerged as a therapeutic candidate due to its role in signaling and cell-function regulation. Its transport is tightly regulated through peroxiporins such as AQP5, expression of which is linked to poor prognosis and metastatic spread, and its role in therapy resistance remains underexplored. This study examined AQP5's role in the acute oxidative stress response. We overexpressed AQP5 in breast cancer cell lines with low basal levels-HR+ (MCF7), HER2+ (SkBr-3), and TNBC (SUM 159)-and exposed them to H2O2 for 24 h. We assessed cell viability, intracellular ROS, changes in AQP3 and AQP5, and key antioxidative and cancer-related pathways (NRF2, PI3K/AKT, FOXOs). AQP5 overexpression elicited a cell-type-specific response. H2O2 treatment reduced viability in SkBr-3-AQP5 and MCF7-AQP5 cells, increased ROS levels in MCF7-AQP5, and decreased ROS in SUM 159-AQP5. It also increased AQP3 in MCF7-AQP5 and differentially affected NRF2, FOXOs, and PI3K/AKT signaling, notably activating NRF2/AKR1B10 axis in MCF7-AQP5 and decreasing FOXO1 in SUM 159-AQP5. These findings highlight the need for further research into AQP5's role in the oxidative stress response in breast cancer cells.
Collapse
Affiliation(s)
| | | | - Ana Čipak Gašparović
- Laboratory for Membrane Transport and Signaling, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia; (I.L.); (M.M.)
| | - Lidija Milković
- Laboratory for Membrane Transport and Signaling, Division of Molecular Medicine, Ruđer Bošković Institute, Bijenička Cesta 54, 10000 Zagreb, Croatia; (I.L.); (M.M.)
| |
Collapse
|
20
|
Li D, Meng K, Liu G, Wen Z, Han Y, Liu W, Xu X, Song L, Cai H, Yang P. Lactiplantibacillus plantarum FRT4 protects against fatty liver hemorrhage syndrome: regulating gut microbiota and FoxO/TLR-4/NF-κB signaling pathway in laying hens. MICROBIOME 2025; 13:88. [PMID: 40158133 PMCID: PMC11954192 DOI: 10.1186/s40168-025-02083-0] [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] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 03/08/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Fatty liver hemorrhage syndrome (FLHS) has become one of the major factors leading to the death of laying hen in caged egg production. FLHS is commonly associated with lipid peroxidation, hepatocyte injury, decreased antioxidant capacity, and inflammation. However, there are limited evidences regarding the preventive effect of Lactiplantibacillus plantarum on FLHS in laying hens and its mechanisms. Our previous results showed that Lp. plantarum FRT4 alleviated FLHS by regulating lipid metabolism, but did not focus on its antioxidant and anti-inflammatory functions and mechanisms. Therefore, this study aimed to investigate the preventive mechanisms of Lp. plantarum FRT4 in alleviating FLHS, with a focus on its role in antioxidant activity and inflammation regulation. RESULTS Supplementation with Lp. plantarum FRT4 enhanced the levels of T-AOC, T-SOD, and GSH-Px, while reducing the levels of TNF-α, IL-1β, IL-8, and NLRP3 in the liver and ovary of laying hens. Additionally, Lp. plantarum FRT4 upregulated the mRNA expressions of SOD1, SOD2, CAT, and GPX1, downregulated the mRNA expressions of pro-inflammatory factors IL-1β, IL-6, and NLRP3, and upregulated the mRNA expressions of anti-inflammatory factors IL-4 and IL-10. Lp. plantarum FRT4 improved the structure and metabolic functions of gut microbiota, and regulated the relative abundances of dominant phyla (Bacteroidetes, Firmicute, and Proteobacteria) and genera (Prevotella and Alistipes). Additionally, it influenced key KEGG pathways, including tryptophan metabolism, amino sugar and nucleotide sugar metabolism, insulin signaling pathway, FoxO signaling pathway. Spearman analysis revealed that the abundance of microbiota at different taxonomic levels was closely related to antioxidant enzymes and inflammatory factors. Furthermore, Lp. plantarum FRT4 modulated the mRNA expressions of related factors in the FoxO/TLR-4/NF-κB signaling pathway by regulating gut microbiota. Moreover, the levels of E2, FSH, and VTG were significantly increased in the ovary after Lp. plantarum FRT4 intervention. CONCLUSIONS Lp. plantarum FRT4 effectively ameliorates FLHS in laying hens. This efficacy is attributed to its antioxidant and anti-inflammatory properties, which are mediated by modulating the structure and function of gut microbiota, and further intervening in the FoxO/TLR-4/NF-κB signaling pathway. These actions enhance hepatic and ovarian function and increase estrogen levels. Video Abstract.
Collapse
Affiliation(s)
- Daojie Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Kun Meng
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Guohua Liu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhiguo Wen
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yunsheng Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Weiwei Liu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Xin Xu
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Liye Song
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Hongying Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Peilong Yang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
21
|
Zhang W, Li W, Du J, Yang C, Yu L, Yang P, Zhang H, Wu Z, Ge G, Yang H, Geng D. Dnmt3a-mediated hypermethylation of FoxO3 promotes redox imbalance during osteoclastogenesis. Proc Natl Acad Sci U S A 2025; 122:e2418023122. [PMID: 40106360 PMCID: PMC11962505 DOI: 10.1073/pnas.2418023122] [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: 09/03/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Redox imbalance contributes to aberrant osteoclastogenesis and osteoporotic bone loss. In this study, we observed lower Forkhead box protein O3 (FoxO3), a transcription factor associated with cellular oxidative stress, enhanced osteoclastogenesis in osteoporosis (OP). Single-cell RNA sequencing (scRNA-seq) analysis on the human femoral head indicated that FoxO3 is widely expressed in macrophages. Furthermore, Lysm-Cre;FoxO3f/f OVX mice showed increased reactive oxygen species (ROS), enhanced osteoclastogenesis, and more bone loss than normal OVX mice. Mechanistically, we identified FoxO3 promoter methylation as a crucial factor contributing to decreased FoxO3, thereby influencing osteoclastogenesis and OC function. Intriguingly, we observed that Dnmt3a, highly expressed during osteoclastogenesis, played a pivotal role in regulating the methylation of the FoxO3 promoter. Knockdown of Dnmt3a promoted FoxO3 expression, inhibiting osteoclastogenesis and mitigating OP. Interestingly, we observed that Dnmt3a alleviated osteoclastogenesis by suppressing ROS via upregulating FoxO3 rather than inducing the dissociation of RANK and TRAF6. Collectively, this study elucidates the role and mechanism of FoxO3 in osteoclastogenesis and OP, providing a epigenetic target for the treatment of OP.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Jun Du
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Chen Yang
- Department of Orthopaedics, Huaian Hospital Affiliated to Yangzhou University, Huaian, Jiangsu223300, China
| | - Lei Yu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Peng Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Haifeng Zhang
- Department of Orthopaedic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200080, China
| | - Zebin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou215006, Jiangsu, China
| |
Collapse
|
22
|
Li M, Chen P, Xue M, Wang J, Wang H, Liang X. AKT-FoxO1-PCK/ChREBP signaling pathway regulates metabolic liver disease induced by high glucose in largemouth bass. Int J Biol Macromol 2025; 295:139703. [PMID: 39793804 DOI: 10.1016/j.ijbiomac.2025.139703] [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: 05/23/2024] [Revised: 12/18/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Starch is widely used in aquaculture because of its low price and the advantages for processing expanded feed. Largemouth bass are naturally type 2 diabetic and intolerant to dietary carbohydrates. In this study, we found that the phosphorylation of AKT and FoxO1 were down-regulated in the fish suffering from metabolic liver disease (MLD). High glucose (25 mM) stimulation in hepatocytes significantly reduced AKT and FoxO1 phosphorylation level, while enhancing glycolysis and gluconeogenesis enzyme activities, leading to acute glucose metabolism disorder. However, after treatment of insulin or FoxO1 inhibitor, the related parameters returned to control level. The mRNA levels of ChREBP and lipid synthesis genes were increased after high glucose stimulation, and then decreased after adding FoxO1 inhibitor, accompanied by a reduction of TG content. Furtherly, plasmid transfection, dual-luciferase reporter assay experiments and EMSA proved that AKT positively regulated the phosphorylation of FoxO1 and FoxO1 positively regulated the promoter activities of PCK and ChREBP, and the transcription factor binding sites were found. In summary, these results support a critical role of AKT-FoxO1-PCK/ChREBP signaling pathway in regulating the occurrence of MLD in largemouth bass. Moreover, we identified a novel FoxO1-mediated gene regulation mechanism, revealing a previously unrecognized cross-talk between FoxO1 and ChREBP.
Collapse
Affiliation(s)
- Min Li
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Pei Chen
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China; Hubei Key Laboratory of Three Gorges Project for Conservation of Fishes, Chinese Sturgeon Research Institute, China Three Gorges Corporation, Yichang, Hubei 443100, China
| | - Min Xue
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jie Wang
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Hao Wang
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiaofang Liang
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
23
|
Gupta S, Afzal M, Agrawal N, Almalki WH, Rana M, Gangola S, Chinni SV, Kumar K B, Ali H, Singh SK, Jha SK, Gupta G. Harnessing the FOXO-SIRT1 axis: insights into cellular stress, metabolism, and aging. Biogerontology 2025; 26:65. [PMID: 40011269 DOI: 10.1007/s10522-025-10207-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 02/15/2025] [Indexed: 02/28/2025]
Abstract
Aging and metabolic disorders share intricate molecular pathways, with the Forkhead box O (FOXO)- Sirtuin 1 (SIRT1) axis emerging as a pivotal regulator of cellular stress adaptation, metabolic homeostasis, and longevity. This axis integrates nutrient signaling with oxidative stress defence, modulating glucose and lipid metabolism, mitochondrial function, and autophagy to maintain cellular stability. FOXO transcription factors, regulated by SIRT1 deacetylation, enhance antioxidant defence mechanisms, activating genes such as superoxide dismutase (SOD) and catalase, thereby counteracting oxidative stress and metabolic dysregulation. Recent evidence highlights the dynamic role of reactive oxygen species (ROS) as secondary messengers in redox signaling, influencing FOXO-SIRT1 activity in metabolic adaptation. Additionally, key redox-sensitive regulators such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) interact with this pathway, orchestrating mitochondrial biogenesis and adaptive stress responses. Pharmacological interventions, including alpha-lipoic acid (ALA), resveratrol, curcumin and NAD+ precursors, exhibit therapeutic potential by enhancing insulin sensitivity, reducing oxidative burden, and restoring metabolic balance. This review synthesizes current advancements in FOXO-SIRT1 regulation, its emerging role in redox homeostasis, and its therapeutic relevance, offering insights into future strategies for combating metabolic dysfunction and aging-related diseases.
Collapse
Affiliation(s)
- Saurabh Gupta
- Department of Pharmacology, Chameli Devi Institute of Pharmacy, Khandwa Road, Village Umrikheda, Near Tollbooth, Indore, Madhya Pradesh, 452020, India
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Saurabh Gangola
- Department of Microbiology, Graphic Era Deemed to be University, Dehradun, 248002, India
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, 42610, Jenjarom, Selangor, Malaysia
| | - Benod Kumar K
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
- Sunway Biofunctional Molecules Discovery Centre (SBMDC), School of Medical and Life Sciences, Sunway University, Sunway, Malaysia
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, New Delhi, India
- Centre for Himalayan Studies, University of Delhi, Delhi, 110007, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| |
Collapse
|
24
|
Omotoso MO, Est-Witte SE, Shannon SR, Li S, Nair NM, Neshat SY, Kang SS, Tzeng SY, Green JJ, Schneck JP. Alginate-based artificial antigen presenting cells expand functional CD8 + T cells with memory characteristics for adoptive cell therapy. Biomaterials 2025; 313:122773. [PMID: 39217794 PMCID: PMC11423771 DOI: 10.1016/j.biomaterials.2024.122773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 07/23/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
The development of artificial Antigen Presenting Cells (aAPCs) has led to improvements in adoptive T cell therapy (ACT), an immunotherapy, for cancer treatment. aAPCs help to streamline the consistent production and expansion of T cells, thus reducing the time and costs associated with ACT. However, several issues still exist with ACT, such as insufficient T cell potency, which diminishes the translational potential for ACT. While aAPCs have been used primarily to increase production efficiency of T cells for ACT, the intrinsic properties of a biomaterial-based aAPC may affect T cell phenotype and function. In CD8+ T cells, reactive oxygen species (ROS) and oxidative stress accumulation can activate Forkhead box protein O1 (FOXO1) to transcribe antioxidants which reduce ROS and improve memory formation. Alginate, a biocompatible and antioxidant rich biomaterial, is promising for incorporation into an aAPC formulation to modulate T cell phenotype. To investigate its utility, a novel alginate-based aAPC platform was developed that preferentially expanded CD8+ T cells with memory related features. Alginate-based aAPCs allowed for greater control of CD8+ T cell qualities, including, significantly improved in vivo persistence and augmented in vivo anti-tumor T cell responses.
Collapse
Affiliation(s)
- Mary O Omotoso
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA
| | - Savannah E Est-Witte
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Sydney R Shannon
- Department of Biomedical Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Shuyi Li
- Department of Pathology, School of Medicine, USA; Institute for NanoBioTechnology, USA
| | - Nina M Nair
- Department of Biomedical Engineering, Whiting School of Engineering, USA
| | - Sarah Y Neshat
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA
| | - Si-Sim Kang
- Department of Pathology, School of Medicine, USA
| | - Stephany Y Tzeng
- Translational Tissue Engineering Center, USA; Department of Biomedical Engineering, Whiting School of Engineering, USA; Johns Hopkins Translational ImmunoEngineering Center, USA
| | - Jordan J Green
- Department of Biomedical Engineering, School of Medicine, USA; Translational Tissue Engineering Center, USA; Institute for NanoBioTechnology, USA; Johns Hopkins Translational ImmunoEngineering Center, USA.
| | - Jonathan P Schneck
- Department of Biomedical Engineering, School of Medicine, USA; Institute for Cell Engineering, School of Medicine, USA; Department of Pathology, School of Medicine, USA; Johns Hopkins Translational ImmunoEngineering Center, USA; Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
25
|
Chen C, He J, Huang W, Xu D, Li Z, Yang A. PLK3 weakens antioxidant defense and inhibits proliferation of porcine Leydig cells under oxidative stress. Sci Rep 2025; 15:2612. [PMID: 39837970 PMCID: PMC11751325 DOI: 10.1038/s41598-025-86867-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025] Open
Abstract
Aging is characterized by cellular degeneration and impaired physiological functions, leading to a decline in male sexual desire and reproductive capacity. Oxidative stress (OS) lead to testicular aging by impairing the male reproductive system, but the potential mechanisms remain unclear. In the present study, the functional status of testicular tissues from young and aged boars was compared, and the transcriptional responses of Leydig cells (LCs) to hydrogen peroxide (H2O2)-induced senescence were explored, revealing the role of OS in promoting aging of the male reproductive system. 601 differentially expressed genes (DEGs) associated with OS, cell cycle regulation, and intracellular processes were identified. These DEGs were significantly enriched in critical aging pathways, including the p53 signaling pathway, autophagy, and cellular senescence. Protein-protein interaction (PPI) network analysis unveiled 15 key genes related to cell cycle and DNA replication, with polo-like kinase 3 (PLK3) exhibiting increased expression under OS. In vitro, PLK3 knockdown significantly enhanced the viability and antioxidant capacity of LCs under OS. This study deepens our understanding of how LCs respond to OS and provides new therapeutic targets for enhancing cellular resistance to oxidative damage and promoting tissue health.
Collapse
Affiliation(s)
- Chujie Chen
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- College of Life Sciences and Resource Environment, Yichun university, Yichun, Jiangxi, China
| | - Jinyan He
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Weixian Huang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Dong Xu
- Department of Biological and Environmental Engineering, Yueyang Vocational Technical College, Yueyang, Hunan, China
| | - Zhaohui Li
- Xiangtan Livestock Breeding Station, Xiangtan, Hunan, China
| | - Anqi Yang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
26
|
Jiang ML, Liu L, Wang Z, Yang X, Lin Z, Jiang R, Zhang CJ, Wang W. Kanglaite alleviates lung squamous cell carcinoma through ferroptosis. Int Immunopharmacol 2025; 144:113616. [PMID: 39579539 DOI: 10.1016/j.intimp.2024.113616] [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: 09/19/2024] [Revised: 10/27/2024] [Accepted: 11/08/2024] [Indexed: 11/25/2024]
Abstract
Kanglaite, a compound predominantly composed of polyunsaturated fatty acids (PUFAs), has been employed in the clinical treatment of adenocarcinoma non-small cell lung cancer (NSCLC) in China for decades. However, its therapeutic efficacy and specific mechanism in the treatment of squamous NSCLC remains unexplored. In this study, we demonstrate that the co-treatment with ferric ion significantly enhances the cytotoxic effects of kanglaite by inducing ferroptosis in NCL-H1703, a cell line of human lung squamous cell carcinoma. Mechanistic investigations reveal that kanglaite induces mitochondrial dysfunction resulting in reactive oxygen species (ROS) excessive production, which is critical for the induction of ferroptosis. Further analysis shows that kanglaite suppresses the PI3K/AKT signaling pathway, leading to increased IP3 generation. IP3 subsequently binds to and activates IP3R, an endoplasmic reticulum (ER) calcium channel, exacerbating the excessive calcium transfer from the ER to mitochondria. The overloaded mitochondrial calcium contributes to its dysfunction and elevates ROS production. To optimize the synergistic effects of ferric ion and kanglaite, we develop a mesoporous silica-based nanodrug delivery system co-loaded with Kanglaite and Fe3O4, which offers several notable advantages, including reduced drug dosage and a faster therapeutic onset. Finally, in an NCL-H1703 xenograft model, the DMSN/Fe3O4-Kanglaite nanodrug significantly inhibited tumor growth. In conclusion, we identified the function and mechanism of kanglaite in treatment of squamous NSCLC and have developed a DMSN/Fe3O4-Kanglaite nanodrug, providing a superior therapeutic approach for the treatment of squamous NSCLC.
Collapse
Affiliation(s)
- Mei-Ling Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210000, China
| | - Li Liu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zilin Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu 611731, China
| | - Xue Yang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Zhiyong Lin
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Runqiu Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210000, China.
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China.
| | - Weiyan Wang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China.
| |
Collapse
|
27
|
Zeng C, Guo M, Xiao K, Li C. Autophagy mediated by ROS-AKT-FoxO pathway is required for intestinal regeneration in echinoderms. Cell Commun Signal 2025; 23:8. [PMID: 39762855 PMCID: PMC11705696 DOI: 10.1186/s12964-024-01993-0] [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: 05/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Autophagy is essential for maintaining material balance and energy circulation and plays a critical role as a regulatory mechanism in tissue regeneration. However, current studies primarily describe this phenotype, with limited exploration of its molecular mechanisms. In this study, we provided the first evidence that autophagy is required for intestinal regeneration in Apostichopus japonicus and identified a previously unrecognized regulatory mechanism involved in this process. We observed that autophagy activation was significantly associated with enhanced regeneration, and its upregulation was shown to be regulated by reactive oxygen species (ROS) bursts. Mechanistically, ROS induced the dephosphorylation of Forkhead box protein O (FoxO) through AjAKT dephosphorylation. The dephosphorylated AjFoxO translocated to the nucleus, where it bound to the promoters of AjLC3 and AjATG4, inducing their transcription. This study highlights the ROS-AjAKT-AjFoxO-AjATG4/AjLC3 pathway as a novel regulatory mechanism underlying autophagy-mediated intestinal regeneration in echinoderms, providing a reference for studying regenerative processes and cytological mechanisms in economically important echinoderms.
Collapse
Affiliation(s)
- Chuili Zeng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Ke Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
- Ningbo University, Zhejiang Province, Ningbo, 315211, P. R. China.
| |
Collapse
|
28
|
Bourgeois B, Madl T. Methods to Study Structure and Dynamics of FOXO Proteins. Methods Mol Biol 2025; 2871:85-98. [PMID: 39565580 DOI: 10.1007/978-1-0716-4217-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
FOXOs are highly dynamic transcription factors consisting of one conserved DNA-binding domain (forkhead domain) as well as intrinsically disordered regions (IDRs) at the N- and C-termini. These IDRs are essential and regulate transcriptional activity of FOXOs by serving as interaction platform for cofactors. Furthermore, the IDRs are involved in intra- and intermolecular homeotypic and heterotypic interactions between FOXOs and in turn mediate FOXO auto-inhibition and condensate formation. Here, we describe generalizable methods to study the structure and dynamics of FOXO proteins in vitro using a combination of biophysical techniques, in particular nuclear magnetic resonance spectroscopy. These methods can serve as a blueprint for the investigation of other IDR-containing transcription factors.
Collapse
Affiliation(s)
- Benjamin Bourgeois
- Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Tobias Madl
- Medicinal Chemistry, Otto Loewi Research Center, Medical University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
29
|
Guan H, Zhang W, Liu H, Jiang Y, Li F, Wang D, Liu Y, He F, Wu M, Ivan Neil Waterhouse G, Sun-Waterhouse D, Li D. Simultaneous binding of quercetin and catechin to FOXO3 enhances IKKα transcription inhibition and suppression of oxidative stress-induced acute alcoholic liver injury in rats. J Adv Res 2025; 67:71-92. [PMID: 38286301 PMCID: PMC11725110 DOI: 10.1016/j.jare.2024.01.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/11/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024] Open
Abstract
INTRODUCTION Oxidative stress is one of the major contributors to acute alcoholic liver injury (AALI), which is a common alcoholic liver disease. Quercetin and catechin are flavonoid antioxidants present in plant foods and possess chemopreventive and chemotherapeutic activities. Quercetin and catechin are often included in the same meal and ingested together. While they show cooperative actions against oxidative damage, the underlying mechanisms behind their counteracting effects against oxidative stress-induced AALI remain poorly understood. OBJECTIVES The aim of this study was to understand the mechanism underlying the enhanced antioxidant effect of quercetin-catechin combination to alleviate AALI in rats. METHODS The ethanol (EtOH)-treated rats and H2O2-treated liver cells were used to demonstrate the enhanced antioxidant effect of quercetin and catechin. Then we used RNA-sequencing to compare quercetin alone, catechin alone and quercetin-catechin combination and then identified the critical role of IKKα combining with gene silencing and overexpression techniques. Its transcription factor, FOXO3 was found through yeast one-hybrid assay, luciferase reporter assay, EMSA and ChIP assay. Finally, the interaction between quercetin, catechin and FOXO3 was verified through molecular docking, UV-Vis absorption spectroscopy, fluorescence spectroscopy, and CD spectroscopy. RESULTS The study demonstrated the enhanced antioxidant effect of a quercetin-catechin combination in EtOH-treated rats and in H2O2-treated liver cells. Quercetin and catechin cooperatively inhibited IKKα/p53 pathway and activated Nrf2 signaling pathway. IKKα was a critical negative regulator in their joint action. FOXO3 bound to IKKα promoter to regulate IKKα transcription. Quercetin and catechin influenced FOXO3-IKKα binding through attaching directly to FOXO3 at different sites and altering FOXO3's secondary structures. CONCLUSION Our study revealed the mechanism of quercetin and catechin against oxidative stress-induced AALI through jointly interacting with transcription factor. This research opens new vistas for examining the joint effect of therapeutics towards functional proteins and confirms the chemopreventive effects of multiple flavonoids via co-regulation.
Collapse
Affiliation(s)
- Hui Guan
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Wenyuan Zhang
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Hui Liu
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Yang Jiang
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Feng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Dan Wang
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Yang Liu
- College of Life Sciences, Shandong Agricultural University, State Key Laboratory of Crop Biology, Shandong Key Laboratory of Crop Biology, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China
| | - Fatao He
- Jinan Fruit Research Institute of All China Federation of Supply & Marketing Cooperatives, 16001 East Road Jingshi, Jinan 250220, Shandong, People's Republic of China
| | - Maoyu Wu
- Jinan Fruit Research Institute of All China Federation of Supply & Marketing Cooperatives, 16001 East Road Jingshi, Jinan 250220, Shandong, People's Republic of China
| | | | - Dongxiao Sun-Waterhouse
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China; School of Chemical Sciences, The University of Auckland, Auckland, New Zealand.
| | - Dapeng Li
- College of Food Science and Engineering, Shandong Agricultural University, Key Laboratory of Food Processing Technology and Quality Control of Shandong Higher Education Institutes, 61 Dai Zong Street, Tai'an 271018, Shandong, People's Republic of China.
| |
Collapse
|
30
|
Strilbytska OM, Semaniuk U, Yurkevych I, Berezovskyi V, Glovyak A, Gospodaryov DV, Bayliak MM, Lushchak O. 2,4-Dinitrophenol is toxic on a low caloric diet but extends lifespan of Drosophila melanogaster on nutrient-rich diets without an impact on metabolism. Biogerontology 2024; 26:27. [PMID: 39702849 DOI: 10.1007/s10522-024-10169-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Uncouplers of mitochondrial electron transport chain, such as 2,4-dinitrophehol (DNP), can mimic calorie restriction by decreasing efficiency of adenosine triphosphate (ATP) synthesis. However, DNP is also a toxic substance, whose overdosage can be lethal. In the fruit fly, Drosophila melanogaster model, we have found that DNP in concentrations of 0.05-0.2 g/L, led to a drastic decrease in fruit fly survival on a low caloric diet (1% sucrose and 1% yeast; 1S-1Y). On the 5S-5Y diet, DNP decreased lifespan of flies reared only in concentration 0.2 g/L, whilst on the diet 15S-15Y DNP either did not significantly shortened fruit fly lifespan or extended it. The lifespan extension on the high caloric 15S-15Y diet with DNP was accompanied by lower activity of lactate dehydrogenase and a decrease in activities of mitochondrial respiratory chain complexes I, II, and V, determined by blue native electrophoresis followed by in-gel activity assays. The exposure to DNP also did not affect key glycolytic enzymes, antioxidant and related enzymes, and markers of oxidative stress, such as aconitase activity and amount protein carbonyls. Consumption of DNP-supplemented diet did not affect flies' resistance to heat stress, though made male flies slightly more resistant to starvation compared with males reared on the control food. We also did not observe substantial changes in the contents of metabolic stores, triacylglycerols and glycogen, in the DNP-treated flies. All this suggest that a nutrient-rich diets provide effective protection against DNP, providing a mild uncoupling of the respiratory chain that allows lifespan extension without considerable changes in metabolism.
Collapse
Affiliation(s)
- Olha M Strilbytska
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Ihor Yurkevych
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Vladyslav Berezovskyi
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Andriy Glovyak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Dmytro V Gospodaryov
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Maria M Bayliak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka Str., Ivano-Frankivsk, 76018, Ukraine.
- Research and Development University, 13a Shota Rustaveli Str., Ivano-Frankivsk, 76018, Ukraine.
| |
Collapse
|
31
|
Zhang Y, Li B, Fu Y, Cai H, Zheng Y. Txnip promotes autophagic apoptosis in diabetic cardiomyopathy by upregulating FoxO1 and its acetylation. Cell Signal 2024; 124:111469. [PMID: 39396562 DOI: 10.1016/j.cellsig.2024.111469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/23/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Autophagy dysfunction and apoptosis exacerbate the risk of heart failure in patients with diabetic cardiomyopathy (DCM). However, the interactions between autophagy and apoptosis in DCM and their underlying mechanisms remain poorly understood. This study induced type 1 DCM in C57BL/6 mice via streptozotocin injection and exposed H9C2 cells to high glucose to investigate these mechanisms. The study revealed a significant elevation in autophagic vesicles and compromised autophagic flux, accompanied by pronounced myocardial cell apoptosis in the myocardium of diabetic mice. Long-term exposure to high glucose in H9C2 cells led to enhanced autophagosome formation and impaired autophagic flux, while inhibition of autophagy with 3-MA reduced cell apoptosis. Additionally, we observed an increase in Txnip expression in the myocardium of diabetic mice and in high glucose-treated H9C2 cells, which regulates autophagic apoptosis in high glucose-treated H9C2 cells. Furthermore, Txnip regulates autophagic apoptosis through the modulation of forkhead box-1 (FoxO1) expression and acetylation. Prolonged high glucose exposure resulted in increased levels of phosphorylated sirtuin 1 (SIRT1) and reduced SIRT1/FoxO1 interaction, changes that were ameliorated by Txnip knockdown. Txnip overexpression elevated FoxO1 levels, which could be suppressed by NAC and GSH. These findings revealed that Txnip mediates autophagic apoptosis in DCM by upregulating FoxO1 via ROS and enhancing FoxO1 acetylation through the suppression of SIRT1 activity. The discovery of this new mechanism provides new perspectives and potential therapeutic targets for understanding and treating DCM.
Collapse
Affiliation(s)
- Yaoting Zhang
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Bing Li
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yu Fu
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - He Cai
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Yang Zheng
- Department of Cardiovascular Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
32
|
Lu C, Gao C, Wei J, Dong D, Sun M. SIRT1-FOXOs signaling pathway: A potential target for attenuating cardiomyopathy. Cell Signal 2024; 124:111409. [PMID: 39277092 DOI: 10.1016/j.cellsig.2024.111409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Cardiomyopathy constitutes a global health burden. It refers to myocardial injury that causes alterations in cardiac structure and function, ultimately leading to heart failure. Currently, there is no definitive treatment for cardiomyopathy. This is because existing treatments primarily focus on drug interventions to attenuate symptoms rather than addressing the underlying causes of the disease. Notably, the cardiomyocyte loss is one of the key risk factors for cardiomyopathy. This loss can occur through various mechanisms such as metabolic disturbances, cardiac stress (e.g., oxidative stress), apoptosis as well as cell death resulting from disorders in autophagic flux, etc. Sirtuins (SIRTs) are categorized as class III histone deacetylases, with their enzyme activity primarily reliant on the substrate nicotinamide adenine dinucleotide (NAD (+)). Among them, Sirtuin 1 (SIRT1) is the most intensively studied in the cardiovascular system. Forkhead O transcription factors (FOXOs) are the downstream effectors of SIRT1. Several reports have shown that SIRT1 can form a signaling pathway with FOXOs in myocardial tissue, and this pathway plays a key regulatory role in cell loss. Thus, this review describes the basic mechanism of SIRT1-FOXOs in inhibiting cardiomyocyte loss and its favorable role in cardiomyopathy. Additionally, we summarized the SIRT1-FOXOs related regulation factor and prospects the SIRT1-FOXOs potential clinical application, which provide reference for the development of cardiomyopathy treatment.
Collapse
Affiliation(s)
- Changxu Lu
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Can Gao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Jinwen Wei
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China.
| | - Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China.
| |
Collapse
|
33
|
Wang K, Hu X, Xie XL, Huang M, Wang D, Yu FL. Phytocosmetic potential of Blumea balsamifera oil in mitigating UV-induced photoaging: Evidence from cellular and mouse models. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118535. [PMID: 38972529 DOI: 10.1016/j.jep.2024.118535] [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: 04/21/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Blumea balsamifera (L.) DC. (BB), the source of Blumea balsamifera oil (BBO), is an aromatic medicinal plant, renowned for its pharmacological properties and its traditional use in Southeast Asian countries such as China, Thailand, Vietnam, Malaysia, and the Philippines for centuries. Traditionally, BB has been used as a raw herbal medicine for treating various skin conditions like eczema, dermatitis, athlete's foot, and wound healing for skin injuries. AIM OF THE STUDY This research aimed to explore the inhibitory effects of BBO on skin aging using two models: in vitro analysis with human dermal fibroblasts (HDF) under UVB-induced stress, and in vivo studies on UVA-induced dorsal skin aging in mice. The study sought to uncover the mechanisms behind BBO's anti-aging effects, specifically, its impact on cellular and tissue responses to UV-induced skin aging. MATERIALS AND METHODS We applied doses of 10-20 μL/mL of BBO to HDF cells that had been exposed to UVB radiation to simulate skin aging. We measured cell viability, and levels of reactive oxygen species (ROS), SA-β-gal, pro-inflammatory cytokines, and matrix metalloproteinases (MMPs). In addition, we investigated the involvement of mitogen-activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB) signaling pathways in mediating the anti-aging effects of BBO. Histopathological and biochemical analyses were conducted in a mouse model to examine the effects of BBO on UV-induced photoaging. RESULTS UV exposure accelerated aging, and caused cellular damage and inflammatory responses through ROS-mediated pathways. In HDF cells, BBO treatment countered the UVB-induced senescence, and the recovery of cell viability was correlated to notable reductions in SA-β-gal, ROS, pro-inflammatory cytokines, and MMPs. Mechanistically, the anti-aging effect of BBO was associated with the downregulation of the JNK/NF-κB signaling pathways. In the in vivo mouse model, BBO exhibited protective capabilities against UV-induced photoaging, which were manifested by the enhanced antioxidant enzyme activities and tissue remodeling. CONCLUSIONS BBO effectively protects fibroblasts from UV-induced photoaging through the JNK/NF-κB pathway. Recovery from photoaging involves an increase in dermal fibroblasts, alleviation of inflammation, accelerated synthesis of antioxidant enzymes, and slowed degradation of ECM proteins. Overall, BBO enhances the skin's defensive capabilities against oxidative stress, underscoring its potential as a therapeutic agent for oxidative stress-related skin aging.
Collapse
Affiliation(s)
- Kai Wang
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China
| | - Xuan Hu
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China
| | - Xiao-Li Xie
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China
| | - Mei Huang
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China
| | - Dan Wang
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China
| | - Fu-Lai Yu
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences/Key Laboratory of Biology and Cultivation of Herb Medicine (Haikou), Ministry of Agriculture and Rural Affairs, P.R. China/Hainan Provincial Engineering Research Center for Blumea Balsamifera, Haikou, Hainan, 571101, P.R. China.
| |
Collapse
|
34
|
Chen S, Sun D, Zhang S, Xu L, Wang N, Li H, Xu X, Wei F. TIN2 modulates FOXO1 mitochondrial shuttling to enhance oxidative stress-induced apoptosis in retinal pigment epithelium under hyperglycemia. Cell Death Differ 2024; 31:1487-1505. [PMID: 39080375 PMCID: PMC11519896 DOI: 10.1038/s41418-024-01349-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 10/30/2024] Open
Abstract
Progressive dysfunction of the retinal pigment epithelium (RPE) and the adjacent photoreceptor cells in the outer retina plays a pivotal role in the pathogenesis of diabetic retinopathy (DR). Here, we observed a marked increase in oxidative stress-induced apoptosis in parallel with higher expression of telomeric protein TIN2 in RPE cells under hyperglycemia in vivo and in vitro. Delving deeper, we confirm that high glucose-induced elevation of mitochondria-localized TIN2 compromises mitochondrial activity and weakens the intrinsic antioxidant defense, thereby leading to the activation of mitochondria-dependent apoptotic pathways. Mechanistically, mitochondrial TIN2 promotes the phosphorylation of FOXO1 and its relocation to the mitochondria. Such translocation of transcription factor FOXO1 not only promotes its binding to the D-loop region of mitochondrial DNA-resulting in the inhibition of mitochondrial respiration-but also hampers its availability to nuclear target DNA, thereby undermining the intrinsic antioxidant defense. Moreover, TIN2 knockdown effectively mitigates oxidative-induced apoptosis in diabetic mouse RPE by preserving mitochondrial homeostasis, which concurrently prevents secondary photoreceptor damage. Our study proposes the potential of TIN2 as a promising molecular target for therapeutic interventions for diabetic retinopathy, which emphasizes the potential significance of telomeric proteins in the regulation of metabolism and mitochondrial function. Created with BioRender ( https://www.biorender.com/ ).
Collapse
Affiliation(s)
- Shimei Chen
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Dandan Sun
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Shuchang Zhang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Li Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Ning Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China
| | - Huiming Li
- Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xun Xu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| | - Fang Wei
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Disease; Shanghai Engineering Center for Visual Science and Photo Medicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, 200080, China.
| |
Collapse
|
35
|
Grzeczka A, Graczyk S, Kordowitzki P. Pleiotropic Effects of Resveratrol on Aging-Related Cardiovascular Diseases-What Can We Learn from Research in Dogs? Cells 2024; 13:1732. [PMID: 39451250 PMCID: PMC11505706 DOI: 10.3390/cells13201732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Resveratrol (RES) is a polyphenol with natural anti-inflammatory and antioxidant properties. It is found in abundance in plants, i.e., grapes and mulberry fruit. In addition, synthetic forms of RES exist. Since the discovery of its specific biological properties, RES has emerged as a candidate substance not only with modeling effects on the immune response but also as an important factor in preventing the onset and progression of cardiovascular disease (CVD). Previous research provided strong evidence of the effects of RES on platelets, mitochondria, cardiomyocytes, and vascular endothelial function. In addition, RES positively affects the coagulation system and vasodilatory function and improves blood flow. Not only in humans but also in veterinary medicine, cardiovascular diseases have one of the highest incidence rates. Canine and human species co-evolved and share recent evolutionary selection processes, and interestingly, numerous pathologies of companion dogs have a human counterpart. Knowledge of the impact of RES on the cardiovascular system of dogs is becoming clearer in the literature. Dogs have long been recognized as valuable animal models for the study of various human diseases as they share many physiological and genetic similarities with humans. In this review, we aim to shed light on the pleiotropic effects of resveratrol on cardiovascular health in dogs as a translational model for human cardiovascular diseases.
Collapse
Affiliation(s)
| | | | - Pawel Kordowitzki
- Department for Basic and Preclinical Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, 87-100 Toruń, Poland; (A.G.)
| |
Collapse
|
36
|
Alanazi ST, Salama SA, Althobaiti MM, Alotaibi RA, AlAbdullatif AA, Musa A, Harisa GI. Alleviation of Copper-Induced Hepatotoxicity by Bergenin: Diminution of Oxidative Stress, Inflammation, and Apoptosis via Targeting SIRT1/FOXO3a/NF-κB Axes and p38 MAPK Signaling. Biol Trace Elem Res 2024:10.1007/s12011-024-04401-3. [PMID: 39347884 DOI: 10.1007/s12011-024-04401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Despite its biological importance, excess copper induces organ damage, especially to the liver. Disruption of critical signaling cascades that control redox status, inflammatory responses, and cellular apoptosis significantly contributes to the copper-induced hepatotoxicity. The present work explored the hepatoprotective ability of bergenin against the copper-induced hepatotoxicity using male Wistar rats as a mammalian model. The results revealed that bergenin suppressed the copper-evoked histopathological changes and hepatocellular necrosis as indicated by decreased activity of the liver enzymes ALT and AST in the sera of the copper-intoxicated rats. It decreased hepatic copper content and the copper-induced oxidative stress as revealed by reduced lipid peroxidation and improved activity of the antioxidant enzymes thioredoxin reductase, glutathione peroxidase, catalase, and superoxide dismutase. Bergenin downregulated the inflammatory cytokines TNF-α and IL-6, and the inflammatory cell infiltration to the liver tissues. Additionally, it inhibited the copper-induced apoptosis as indicated by significant reduction in caspase-3 activity. At the molecular level, bergenin activated the antioxidant transcription factor FOXO3a, inhibited the nuclear translocation of the inflammatory transcription factor NF-κB, and suppressed the inflammatory signaling molecules p38 MAPK and c-Fos. Interestingly, bergenin improved the expression of the anti-apoptotic protein Bcl2 and reduced the pro-apoptotic protein BAX. Bergenin markedly enhanced the expression of the histone deacetylase protein SIRT1 that regulates activity of NF-κB and FOXO3a. Collectively, these findings highlight the alleviating activity of bergenin against the copper-induced hepatotoxicity via controlling oxidative stress, inflammation, and apoptosis potentially through upregulation of SIRT1, activation of FOXO3a along with suppression of NF-κB and p38 MAPK signaling.
Collapse
Affiliation(s)
- Samyah T Alanazi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, 11433, Riyadh, Saudi Arabia
| | - Samir A Salama
- Division of Biochemistry, Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia.
| | - Musaad M Althobaiti
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Rana A Alotaibi
- College of Pharmacy, Taif University, P.O. Box 11099, 21944, Taif, Saudi Arabia
| | - Ammar A AlAbdullatif
- Pharmaceutical Care Services, Ministry of the National Guard-Health Affairs, P.O. Box 4616, 31412, Dammam, Saudi Arabia
| | - Arafa Musa
- Department of Pharmacognosy, College of Pharmacy, Jouf University, 72341, Sakaka, Aljouf, Saudi Arabia
| | - Gamaleldin I Harisa
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
37
|
Tatone C, Di Emidio G, Battaglia R, Di Pietro C. Building a Human Ovarian Antioxidant ceRNA Network "OvAnOx": A Bioinformatic Perspective for Research on Redox-Related Ovarian Functions and Dysfunctions. Antioxidants (Basel) 2024; 13:1101. [PMID: 39334761 PMCID: PMC11428640 DOI: 10.3390/antiox13091101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
The ovary is a major determinant of female reproductive health. Ovarian functions are mainly related to the primordial follicle pool, which is gradually lost with aging. Ovarian aging and reproductive dysfunctions share oxidative stress as a common underlying mechanism. ROS signaling is essential for normal ovarian processes, yet it can contribute to various ovarian disorders when disrupted. Therefore, balance in the redox system is crucial for proper ovarian functions. In the present study, by focusing on mRNAs and ncRNAs described in the ovary and taking into account only validated ncRNA interactions, we built an ovarian antioxidant ceRNA network, named OvAnOx ceRNA, composed of 5 mRNAs (SOD1, SOD2, CAT, PRDX3, GR), 10 miRNAs and 5 lncRNAs (XIST, FGD5-AS1, MALAT1, NEAT1, SNHG1). Our bioinformatic analysis indicated that the components of OvAnOx ceRNA not only contribute to antioxidant defense but are also involved in other ovarian functions. Indeed, antioxidant enzymes encoded by mRNAs of OvAnOx ceRNA operate within a regulatory network that impacts ovarian reserve, follicular dynamics, and oocyte maturation in normal and pathological conditions. The OvAnOx ceRNA network represents a promising tool to unravel the complex dialog between redox potential and ovarian signaling pathways involved in reproductive health, aging, and diseases.
Collapse
Affiliation(s)
- Carla Tatone
- Department of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.T.); (G.D.E.)
| | - Giovanna Di Emidio
- Department of Life, Health and Experimental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (C.T.); (G.D.E.)
| | - Rosalia Battaglia
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics, University of Catania, 95123 Catania, Italy;
| | - Cinzia Di Pietro
- Department of Biomedical and Biotechnological Sciences, Section of Biology and Genetics, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
38
|
Wang W, Liu M, Fu X, Qi M, Zhu F, Fan F, Wang Y, Zhang K, Chu S. Hydroxysafflor yellow A ameliorates alcohol-induced liver injury through PI3K/Akt and STAT3/NF-κB signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155814. [PMID: 38878526 DOI: 10.1016/j.phymed.2024.155814] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/10/2024] [Accepted: 06/06/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND Alcohol-associated liver disease (ALD) is a prevalent liver ailment. It has escalated into a significant public health issue, imposing substantial burdens on medical, economic, and social domains. Currently, oxidative stress, inflammation, and apoptosis are recognized as crucial culprits in improving ALD. Consequently, mitigating these issues has emerged as a promising avenue for enhancing ALD. Hydroxysafflor yellow A (HSYA) is the main ingredient in safflower, showing excellent antioxidative stress, anti-inflammatory, and anti-apoptosis traits. However, there are limited investigations into the mechanisms by which HSYA ameliorates ALD PURPOSE: We investigated whether HSYA, a significant constituent of Asteraceae safflower, exerts antioxidant stress and attenuates inflammation and anti-apoptotic effects through PI3K/Akt and STAT3/NF-κB pathways, thereby ameliorating ALD METHODS: We established two experimental models: an ethanol-induced liver damage mouse model in vivo and a HepG2 cell alcohol injury model in vitro RESULTS: The results demonstrated that HSYA effectively ameliorated liver tissue damage, reduced levels of ALT, AST, LDL-C, TG, TC, and MDA, enhanced HDL-C levels, SOD and GSH activities, reduced ROS accumulation in cells, and activated the Nrf2 pathway, a transcription factor involved in antioxidant defense. By regulating the PI3K/Akt and STAT3/NF-κB pathways, HSYA exhibits notable antioxidative stress, anti-inflammatory, and anti-apoptotic effects, effectively impeding ALD's advancement. To further confirm the regulatory effect of HSYA on PI3K/Akt and downstream signaling pathways, the PI3K activator 740 Y-P was used and was found to reverse the downregulation of PI3K by HSYA CONCLUSION: This study supports the effectiveness of HSYA in reducing ALD by regulating the PI3K/Akt and STAT3/NF-κB pathways, indicating its potential medicinal value.
Collapse
Affiliation(s)
- Wenxuan Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Min Liu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Xianglei Fu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Man Qi
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Furong Zhu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Furong Fan
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Yuanchuang Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Kaiyue Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China
| | - Shenghui Chu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, North 4th Road 221, Shihezi, PR China.
| |
Collapse
|
39
|
Iliadis S, Papanikolaou NA. Reactive Oxygen Species Mechanisms that Regulate Protein-Protein Interactions in Cancer. Int J Mol Sci 2024; 25:9255. [PMID: 39273204 PMCID: PMC11395503 DOI: 10.3390/ijms25179255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Reactive oxygen species (ROS) are produced during cellular metabolism and in response to environmental stress. While low levels of ROS play essential physiological roles, excess ROS can damage cellular components, leading to cell death or transformation. ROS can also regulate protein interactions in cancer cells, thereby affecting processes such as cell growth, migration, and angiogenesis. Dysregulated interactions occur via various mechanisms, including amino acid modifications, conformational changes, and alterations in complex stability. Understanding ROS-mediated changes in protein interactions is crucial for targeted cancer therapies. In this review, we examine the role that ROS mechanisms in regulating pathways through protein-protein interactions.
Collapse
Affiliation(s)
- Stavros Iliadis
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| | - Nikolaos A Papanikolaou
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| |
Collapse
|
40
|
Song J, Li Z, Zhou L, Chen X, Sew WQG, Herranz H, Ye Z, Olsen JV, Li Y, Nygaard M, Christensen K, Tong X, Bohr VA, Rasmussen LJ, Dai F. FOXO-regulated OSER1 reduces oxidative stress and extends lifespan in multiple species. Nat Commun 2024; 15:7144. [PMID: 39164296 PMCID: PMC11336091 DOI: 10.1038/s41467-024-51542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 08/12/2024] [Indexed: 08/22/2024] Open
Abstract
FOXO transcription factors modulate aging-related pathways and influence longevity in multiple species, but the transcriptional targets that mediate these effects remain largely unknown. Here, we identify an evolutionarily conserved FOXO target gene, Oxidative stress-responsive serine-rich protein 1 (OSER1), whose overexpression extends lifespan in silkworms, nematodes, and flies, while its depletion correspondingly shortens lifespan. In flies, overexpression of OSER1 increases resistance to oxidative stress, starvation, and heat shock, while OSER1-depleted flies are more vulnerable to these stressors. In silkworms, hydrogen peroxide both induces and is scavenged by OSER1 in vitro and in vivo. Knockdown of OSER1 in Caenorhabditis elegans leads to increased ROS production and shorter lifespan, mitochondrial fragmentation, decreased ATP production, and altered transcription of mitochondrial genes. Human proteomic analysis suggests that OSER1 plays roles in oxidative stress response, cellular senescence, and reproduction, which is consistent with the data and suggests that OSER1 could play a role in fertility in silkworms and nematodes. Human studies demonstrate that polymorphic variants in OSER1 are associated with human longevity. In summary, OSER1 is an evolutionarily conserved FOXO-regulated protein that improves resistance to oxidative stress, maintains mitochondrial functional integrity, and increases lifespan in multiple species. Additional studies will clarify the role of OSER1 as a critical effector of healthy aging.
Collapse
Affiliation(s)
- Jiangbo Song
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Zhiquan Li
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Lei Zhou
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Xin Chen
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Wei Qi Guinevere Sew
- Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Zilu Ye
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
- Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Jesper Velgaard Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Yuan Li
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Marianne Nygaard
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Kaare Christensen
- Epidemiology, Biostatistics and Biodemography, Department of Public Health, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Department of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Xiaoling Tong
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Vilhelm A Bohr
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark
- Section on DNA repair, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lene Juel Rasmussen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Fangyin Dai
- State Key Laboratory of Resource Insects, Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
41
|
Tian L, Luo Y, Ren J, Zhao C. The Role of Oxidative Stress in Hypomagnetic Field Effects. Antioxidants (Basel) 2024; 13:1017. [PMID: 39199261 PMCID: PMC11352208 DOI: 10.3390/antiox13081017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/12/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
The geomagnetic field (GMF) is crucial for the survival and evolution of life on Earth. The weakening of the GMF, known as the hypomagnetic field (HMF), significantly affects various aspects of life on Earth. HMF has become a potential health risk for future deep space exploration. Oxidative stress is directly involved in the biological effects of HMF on animals or cells. Oxidative stress occurs when there is an imbalance favoring oxidants over antioxidants, resulting in cellular damage. Oxidative stress is a double-edged sword, depending on the degree of deviation from homeostasis. In this review, we summarize the important experimental findings from animal and cell studies on HMF exposure affecting intracellular reactive oxygen species (ROS), as well as the accompanying many physiological abnormalities, such as cognitive dysfunction, the imbalance of gut microbiota homeostasis, mood disorders, and osteoporosis. We discuss new insights into the molecular mechanisms underlying these HMF effects in the context of the signaling pathways related to ROS. Among them, mitochondria are considered to be the main organelles that respond to HMF-induced stress by regulating metabolism and ROS production in cells. In order to unravel the molecular mechanisms of HMF action, future studies need to consider the upstream and downstream pathways associated with ROS.
Collapse
Affiliation(s)
- Lanxiang Tian
- Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (J.R.); (C.Z.)
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Beijing National Observatory of Space Environment, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China
| | - Yukai Luo
- Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (J.R.); (C.Z.)
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Ren
- Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (J.R.); (C.Z.)
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenchen Zhao
- Key Laboratory of Earth and Planetary Physics, Institute of Geology and Geophysics, Chinese Academy of Sciences, Beijing 100029, China; (Y.L.); (J.R.); (C.Z.)
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
42
|
Zhang Y, Wu Y, Li B, Tian J. Phloretin prolongs lifespan of Caenorhabditis elegans via inhibition of NDUFS1 and NDUFS6 at mitochondrial complex Ⅰ. Free Radic Biol Med 2024; 221:283-295. [PMID: 38705496 DOI: 10.1016/j.freeradbiomed.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/02/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Phloretin has been widely perceived as an antioxidant. However, the bioavailability of phloretin in vivo is generally far too low to elicit a direct antioxidant effect by scavenging reactive oxygen species (ROS). Here we showed that administration of phloretin of apple polyphenols extended lifespan of Caenorhabditis elegans and promoted fitness. Specially phloretin enhanced the survival rates of nematodes under oxidants in an inverted U-shaped dose-response manner. The lifespan-extending effects of phloretin were mediated by ROS via mitochondrial complex I inhibition. The increase of ROS stimulated p38 MAPK/PMK-1 as well as transcription factors of NRF2/SKN-1 and FOXO/DAF-16. Consistent with the involvement of NRF2/SKN-1 and FOXO/DAF-16 in lifespan-extending effects, activities of superoxide dismutase (SOD) and catalase (CAT) were enhanced by phloretin. The exogenous application of antioxidants butylated hydroxyanisole and N-acetylcysteine abolished the increase of ROS, the enhancement of SOD and CAT activities, and the lifespan extending effects of phloretin. Meanwhile, with the inhibition of mitochondrial complex I, ATP was instantly decreased. Both energy sensors of AMPK/AAK-2 and SIRT1/SIR-2.1 were involved in the lifespan extension by phloretin. Transcriptomic, real-time qPCR and molecular docking analyses demonstrated that the binding of phloretin at complex I located at NDUFS1/NUO-5, NDUFS2/GAS-1, and NDUFS6/NDUF-6. The molecular dynamic simulation and binding free energy calculations showed that phloretin had high binding affinities towards NDUFS1 (-7.21 kcal/mol) and NDUFS6 (-7.02 kcal/mol). Collectively, our findings suggested phloretin had effects of life expectancy enhancement and fitness promotion via redox regulations in vivo. NDUFS1/NUO-5 and NDUFS6/NDUF-6 might be new targets in the lifespan and wellness regulations.
Collapse
Affiliation(s)
- Yu Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Yonglin Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Engineering & Technology Research Center of Hubei Province, China
| | - Jing Tian
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, China; Functional Food Engineering & Technology Research Center of Hubei Province, China.
| |
Collapse
|
43
|
Na J, Ryu HG, Park H, Park H, Lee E, Nam Y, Kim H, Jang SM, Kim DY, Kim S. FoxO1 Alleviates the Mitochondrial ROS Levels Induced by α-Synuclein Preformed Fibrils in BV-2 Microglial Cells. Inflammation 2024:10.1007/s10753-024-02119-x. [PMID: 39145787 DOI: 10.1007/s10753-024-02119-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/16/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder marked by the gradual deterioration of dopaminergic neurons, especially in the substantia nigra pars compacta (SNc). Dysregulation of the transcription factor FoxO1 is associated with various neurodegenerative conditions, including Alzheimer's disease and PD, though the specific mechanisms involved are not fully understood. This study explores the effects of α-Synuclein preformed fibrils (PFF) on BV-2 microglial cells, focusing on changes in molecular characteristics and their impact on neuronal degeneration. Our results demonstrate that PFF treatment significantly increases FoxO1 mRNA (p = 0.0443) and protein (p = 0.0216) levels, leading to its nuclear translocation (p = 0.0142) and enhanced expression of genes involved in the detoxification of reactive oxygen species (ROS), such as Catalase (Cat, p = 0.0249) and superoxide dismutase 2 (Sod2, p = 0.0313). Furthermore, we observed that PFF treatment elevates mitochondrial ROS levels. However, cells lacking FoxO1 or treated with FoxO1 inhibitors showed increased vulnerability to PFF-induced ROS, attributed to reduced expression of ROS detoxifying enzymes Cat and Sod2 (p < 0.0001). Besides enhancing ROS production, inhibiting FoxO1 also heightens neurotoxicity induced by PFF treatment in microglia-conditioned medium (p < 0.0001). Conversely, treatment with N-acetylcysteine or bacterial superoxide dismutase A mitigated the ROS increase induced by PFF (p < 0.0001). These findings suggest the essential role of FoxO1 in regulating ROS levels, which helps alleviate pathology in PFF-induced PD models. Our study provides insights into the genetic mechanisms of PD and suggests potential pathways for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Jiyeon Na
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Hye Guk Ryu
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Haeun Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Hyeonwoo Park
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Eunmin Lee
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Younwoo Nam
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Hyerynn Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Sang-Min Jang
- Department of Biochemistry, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea
| | - Do-Yeon Kim
- Department of Pharmacology, School of Dentistry, Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41940, Republic of Korea.
| | - Sangjune Kim
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, Chungbuk, 28644, Republic of Korea.
| |
Collapse
|
44
|
Shafaati T, Gopal K. Forkhead box O1 transcription factor; a therapeutic target for diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:13193. [PMID: 39206323 PMCID: PMC11349536 DOI: 10.3389/jpps.2024.13193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular disease including diabetic cardiomyopathy (DbCM) represents the leading cause of death in people with diabetes. DbCM is defined as ventricular dysfunction in the absence of underlying vascular diseases and/or hypertension. The known molecular mediators of DbCM are multifactorial, including but not limited to insulin resistance, altered energy metabolism, lipotoxicity, endothelial dysfunction, oxidative stress, apoptosis, and autophagy. FoxO1, a prominent member of forkhead box O transcription factors, is involved in regulating various cellular processes in different tissues. Altered FoxO1 expression and activity have been associated with cardiovascular diseases in diabetic subjects. Herein we provide an overview of the role of FoxO1 in various molecular mediators related to DbCM, such as altered energy metabolism, lipotoxicity, oxidative stress, and cell death. Furthermore, we provide valuable insights into its therapeutic potential by targeting these perturbations to alleviate cardiomyopathy in settings of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Tanin Shafaati
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
45
|
Ahmad S, Single S, Liu Y, Hough KP, Wang Y, Thannickal VJ, Athar M, Goliwas KF, Deshane JS. Heavy Metal Exposure-Mediated Dysregulation of Sphingolipid Metabolism. Antioxidants (Basel) 2024; 13:978. [PMID: 39199224 DOI: 10.3390/antiox13080978] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Exposure to heavy metals (HMs) is often associated with inflammation and cell death, exacerbating respiratory diseases including asthma. Most inhaled particulate HM exposures result in the deposition of HM-bound fine particulate matter, PM2.5, in pulmonary cell populations. While localized high concentrations of HMs may be a causative factor, existing studies have mostly evaluated the effects of systemic or low-dose chronic HM exposures. This report investigates the impact of local high concentrations of specific HMs (NaAsO2, MnCl2, and CdCl2) on sphingolipid homeostasis and oxidative stress, as both play a role in mediating responses to HM exposure and have been implicated in asthma. Utilizing an in vitro model system and three-dimensional ex vivo human tissue models, we evaluated the expression of enzymatic regulators of the salvage, recycling, and de novo synthesis pathways of sphingolipid metabolism, and observed differential modulation in these enzymes between HM exposures. Sphingolipidomic analyses of specific HM-exposed cells showed increased levels of anti-apoptotic sphingolipids and reduced pro-apoptotic sphingolipids, suggesting activation of the salvage and de novo synthesis pathways. Differential sphingolipid regulation was observed within HM-exposed lung tissues, with CdCl2 exposure and NaAsO2 exposure activating the salvage and de novo synthesis pathway, respectively. Additionally, using spatial transcriptomics and quantitative real-time PCR, we identified HM exposure-induced transcriptomic signatures of oxidative stress in epithelial cells and human lung tissues.
Collapse
Affiliation(s)
- Shaheer Ahmad
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Sierra Single
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yuelong Liu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Kenneth P Hough
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Yong Wang
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Victor J Thannickal
- John W. Deming Department of Medicine, Tulane University School of Medicine and Southeast Veterans Healthcare System, New Orleans, LA 70119-6535, USA
| | - Mohammad Athar
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kayla F Goliwas
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| | - Jessy S Deshane
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0006, USA
| |
Collapse
|
46
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
47
|
Rudnitsky E, Braiman A, Wolfson M, Muradian KK, Gorbunova V, Turgeman G, Fraifeld VE. Stem cell-derived extracellular vesicles as senotherapeutics. Ageing Res Rev 2024; 99:102391. [PMID: 38914266 DOI: 10.1016/j.arr.2024.102391] [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: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Cellular senescence (CS) is recognized as one of the hallmarks of aging, and an important player in a variety of age-related pathologies. Accumulation of senescent cells can promote a pro-inflammatory and pro-cancerogenic microenvironment. Among potential senotherapeutics are extracellular vesicles (EVs) (40-1000 nm), including exosomes (40-150 nm), that play an important role in cell-cell communications. Here, we review the most recent studies on the impact of EVs derived from stem cells (MSCs, ESCs, iPSCs) as well as non-stem cells of various types on CS and discuss potential mechanisms responsible for the senotherapeutic effects of EVs. The analysis revealed that (i) EVs derived from stem cells, pluripotent (ESCs, iPSCs) or multipotent (MSCs of various origin), can mitigate the cellular senescence phenotype both in vitro and in vivo; (ii) this effect is presumably senomorphic; (iii) EVs display cross-species activity, without apparent immunogenic responses. In summary, stem cell-derived EVs appear to be promising senotherapeutics, with a feasible application in humans.
Collapse
Affiliation(s)
- Ekaterina Rudnitsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Marina Wolfson
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Khachik K Muradian
- Department of Biology of Aging and Experimental Life Span Extension, State Institute of Gerontology of National Academy of Medical Sciences of Ukraine, Kiev 4114, Ukraine
| | - Vera Gorbunova
- Department of Biology, Rochester Aging Research Center, University of Rochester, Rochester, NY 14627, USA
| | - Gadi Turgeman
- Department of Molecular Biology, Faculty of Natural Sciences and Medical School, Ariel University, Ariel 40700, Israel.
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
48
|
Ahmadieh-Yazdi A, Karimi M, Afkhami E, Hajizadeh-Tafti F, Kuchakzadeh F, Yang P, Sheykhhasan M. Unveiling therapeutic potential: Adipose tissue-derived mesenchymal stem cells and their exosomes in the management of diabetes mellitus, wound healing, and chronic ulcers. Biochem Pharmacol 2024; 226:116399. [PMID: 38944396 DOI: 10.1016/j.bcp.2024.116399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Diabetes mellitus (DM) is a pervasive global health issue with substantial morbidity and mortality, often resulting in secondary complications, including diabetic wounds (DWs). These wounds, arising from hyperglycemia, diabetic neuropathy, anemia, and ischemia, afflict approximately 15% of diabetic patients, with a considerable 25% at risk of lower limb amputations. The conventional approaches for chronic and diabetic wounds management involves utilizing various therapeutic substances and techniques, encompassing growth factors, skin substitutes and wound dressings. In parallel, emerging cell therapy approaches, notably involving adipose tissue-derived mesenchymal stem cells (ADMSCs), have demonstrated significant promise in addressing diabetes mellitus and its complications. ADMSCs play a pivotal role in wound repair, and their derived exosomes have garnered attention for their therapeutic potential. This review aimed to unravel the potential mechanisms and provide an updated overview of the role of ADMSCs and their exosomes in diabetes mellitus and its associated complications, with a specific focus on wound healing.
Collapse
Affiliation(s)
- Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahdieh Karimi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Afkhami
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Hajizadeh-Tafti
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Kuchakzadeh
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| |
Collapse
|
49
|
Hao XD, Liu JX, Zhang JS. Longevity factor FOXO3a: A potential therapeutic target for age-related ocular diseases. Life Sci 2024; 350:122769. [PMID: 38848943 DOI: 10.1016/j.lfs.2024.122769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/22/2024] [Accepted: 06/02/2024] [Indexed: 06/09/2024]
Abstract
The forkhead box protein O3 (FOXO3a) belongs to the subgroup O of the forkhead transcription factor family and plays an important role in regulating the aging process by participating in the regulation of various life processes, including cell cycle arrest, apoptosis, autophagy, oxidative stress, and DNA repair. The eye is an organ that is affected by aging earlier. However, the functional role and potential clinical applications of FOXO3a in age-related eye diseases have not received widespread attention and lacked comprehensive and clear clarification. In this review, we demonstrated the relationship between FOXO3a and visual system health, summarized the functional roles of FOXO3a in various eye diseases, and potential ocular-related therapies and drugs targeting FOXO3a in visual system diseases through a review and summary of relevant literature. This review indicates that FOXO3a is an important factor in maintaining the normal function of various tissues in the eye, and is closely related to the occurrence and development of ophthalmic-related diseases. Based on its vital role in the normal function of the visual system, FOXO3a has potential clinical application value in related ophthalmic diseases. At present, multiple molecules and drugs targeting FOXO3a have been reported to have the potential for the treatment of related ophthalmic diseases, but further clinical trials are needed. In conclusion, this review can facilitate us to grasp the role of FOXO3a in the visual system and provide new views and bases for the treatment strategy research of age-related eye diseases.
Collapse
Affiliation(s)
- Xiao-Dan Hao
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China.
| | - Jin-Xiu Liu
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Jing-Sai Zhang
- Institute for Translational Medicine, the Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| |
Collapse
|
50
|
da Silva IV, Mlinarić M, Lourenço AR, Pérez-Garcia O, Čipak Gašparović A, Soveral G. Peroxiporins and Oxidative Stress: Promising Targets to Tackle Inflammation and Cancer. Int J Mol Sci 2024; 25:8381. [PMID: 39125952 PMCID: PMC11313477 DOI: 10.3390/ijms25158381] [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: 06/26/2024] [Revised: 07/25/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Peroxiporins are a specialized subset of aquaporins, which are integral membrane proteins primarily known for facilitating water transport across cell membranes. In addition to the classical water transport function, peroxiporins have the unique capability to transport hydrogen peroxide (H2O2), a reactive oxygen species involved in various cellular signaling pathways and regulation of oxidative stress responses. The regulation of H2O2 levels is crucial for maintaining cellular homeostasis, and peroxiporins play a significant role in this process by modulating its intracellular and extracellular concentrations. This ability to facilitate the passage of H2O2 positions peroxiporins as key players in redox biology and cellular signaling, with implications for understanding and treating various diseases linked to oxidative stress and inflammation. This review provides updated information on the physiological roles of peroxiporins and their implications in disease, emphasizing their potential as novel biomarkers and drug targets in conditions where they are dysregulated, such as inflammation and cancer.
Collapse
Affiliation(s)
- Inês V. da Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Monika Mlinarić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ana Rita Lourenço
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Olivia Pérez-Garcia
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | | | - Graça Soveral
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
| |
Collapse
|