1
|
Xiao Y, He M, Zhang X, Yang M, Yuan Z, Yao S, Qin Y. Research progress on the mechanism of tumor cell ferroptosis regulation by epigenetics. Epigenetics 2025; 20:2500949. [PMID: 40327848 PMCID: PMC12064064 DOI: 10.1080/15592294.2025.2500949] [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: 02/14/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer remains a significant barrier to human longevity and a leading cause of mortality worldwide. Despite advancements in cancer therapies, challenges such as cellular toxicity and drug resistance to chemotherapy persist. Regulated cell death (RCD), once regarded as a passive process, is now recognized as a programmed mechanism with distinct biochemical and morphological characteristics, thereby presenting new therapeutic opportunities. Ferroptosis, a novel form of RCD characterized by iron-dependent lipid peroxidation and unique mitochondrial damage, differs from apoptosis, autophagy, and necroptosis. It is driven by reactive oxygen species (ROS)-induced lipid peroxidation and is implicated in tumorigenesis, anti-tumor immunity, and resistance, particularly in tumors undergoing epithelial-mesenchymal transition. Moreover, ferroptosis is associated with ischemic organ damage, degenerative diseases, and aging, regulated by various cellular metabolic processes, including redox balance, iron metabolism, and amino acid, lipid, and glucose metabolism. This review focuses on the role of epigenetic factors in tumor ferroptosis, exploring their mechanisms and potential applications in cancer therapy. It synthesizes current knowledge to provide a comprehensive understanding of epigenetic regulation in tumor cell ferroptosis, offering insights for future research and clinical applications.
Collapse
Affiliation(s)
- Yuyang Xiao
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Mengyang He
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xupeng Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Meng Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Zhangchi Yuan
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Shanhu Yao
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Medical Information Research, Central South University, Changsha, Hunan, China
| | - Yuexiang Qin
- Department of Health Management Medical, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Otolaryngology, Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
2
|
Danev N, Harman RM, Sipka AS, Oliveira L, Huntimer L, Van de Walle GR. The secretomes of bovine mammary epithelial cell subpopulations differentially modulate macrophage function. Vet Q 2025; 45:1-14. [PMID: 39921381 PMCID: PMC11809179 DOI: 10.1080/01652176.2025.2463338] [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: 10/19/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Bovine mammosphere-derived epithelial cell (MDEC) cultures are heterogeneous and enriched for stem and progenitor cells. We previously reported that the bovine MDEC secretome, comprised of all bioactive factors secreted by the cells, displays regenerative properties, exerts antimicrobial effects, and modulates neutrophil activity, positioning it as a promising non-antibiotic biologic therapy for infectious diseases important to the dairy industry, like mastitis. Mastitis is defined as inflammation of the udder, and it is typically caused by bacterial infection. The effect of the MDEC secretome on macrophages, a first line of defense against bacterial infections in the udder, is unknown and could impact the utility of the secretome as a therapy for mastitis. To address this, we isolated bovine monocytes from peripheral blood and maintained them as an unpolarized (M0) population or polarized them into M1 or M2 phenotypes. Macrophages cultured with the secretome of bovine MDECs were assessed for their ability to phagocytose labeled bacterial particles and accumulate reactive oxygen species (ROS). We used single-cell RNA sequencing (scRNA-seq) and fluorescence-activated cell sorting (FACS) to isolate a subpopulation of MDECs that exert enhanced effects on macrophages. We found that the secretome of MDECs that do not express cluster of differentiation (CD) 73, a cell surface enzyme used as a marker for mesenchymal stromal cells, most strongly increased macrophage phagocytosis and ROS accumulation. These findings will help optimize the generation of the bovine MDEC secretome as a suitable treatment option for mastitis.
Collapse
Affiliation(s)
- Nikola Danev
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Rebecca M. Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Anja S. Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | | | | | - Gerlinde R. Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- The Royal (Dick) School of Veterinary Studies and the Roslin Institute, University of Edinburgh, Midlothian, Scotland
| |
Collapse
|
3
|
Achouri H, Derguini A, Idres T, Selamoglu Z, Hamadi NB, Jalouli M, Elfalleh W, Bendif H, Badraoui R, Boufahja F, Dellali M. Impact of climate change on the toxicity of bisphenol A in Mytilus galloprovincialis and assessment of phycoremediation using Nannochloropsis salina via a multi-biomarker strategy and modeling. MARINE POLLUTION BULLETIN 2025; 216:118010. [PMID: 40253969 DOI: 10.1016/j.marpolbul.2025.118010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
In the current study, the mussels Mytilus galloprovincialis, exposed to four varying temperatures (17, 20, 23, and 26 °C), were contaminated with 50 μg/L of bisphenol A both with and without Nannochloropsis salina. The toxicity evaluation is determined by quantifying various biomarkers related to oxidative stress, neurotoxicity, and cellular damage. The key findings indicate that the toxicity of bisphenol A is heightened by rising temperature. The impact of bisphenol A is most evident at 26 °C, leading to excessive production of reactive oxygen species, depletion of non-enzymatic antioxidants, and activation of antioxidant enzymes (catalase and glutathione-S-transferase). The rise in malondialdehyde levels confirms lipid peroxidation caused by bisphenol A and intensified by thermal stress. These findings have been supported by strong molecular interactions between bisphenol A and lectin mytilec apo-form and proximal thread matrix protein 1 from M. galloprovincialis following the computational modeling assay. The incorporation of N. salina as a food additive helped, firstly, to mitigate the stress effects and, secondly, resulted in a noticeable enhancement of oxidative balance and filtration ability, along with decreased lipid peroxidation.
Collapse
Affiliation(s)
- Haifa Achouri
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Environment Biomonitoring, Coastal Ecology and Ecotoxicology Unit, 7021 Zarzouna, Tunisia
| | - Assia Derguini
- Microbial Ecology Laboratory, FSNV, Abderrahmane MIRA University, 06017 Bejaïa, Algeria.
| | - Takfarinas Idres
- Laboratory for Livestock Animal Production and Health Research, Rabie Bouchama National Veterinary School of Algiers, Issad ABBAS Street, BP 161 Oued Semar, Algiers, Algeria.
| | - Zeliha Selamoglu
- Department of Medical Biology, Medicine Faculty, Nigde Omer Halisdemir University, Nigde, Turkey.
| | - Naoufel Ben Hamadi
- Chemistry Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Box 5701, Riyadh 11432, Saudi Arabia.
| | - Maroua Jalouli
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia.
| | - Walid Elfalleh
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia.
| | - Hamdi Bendif
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia.
| | - Riadh Badraoui
- Department of Biology, University of Ha'il, Ha'il 45851, Saudi Arabia.
| | - Fehmi Boufahja
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia.
| | - Mohamed Dellali
- University of Carthage, Faculty of Sciences of Bizerte, Laboratory of Environment Biomonitoring, Coastal Ecology and Ecotoxicology Unit, 7021 Zarzouna, Tunisia.
| |
Collapse
|
4
|
Zhou J, Shou Y, Shi Q, Ye J, Li X, Zhu Z, Wang X. Fibroblast growth factor 18 attenuates renal fibrosis via AMPK/NOX4 pathway in mice. Biochem Biophys Res Commun 2025; 766:151913. [PMID: 40311293 DOI: 10.1016/j.bbrc.2025.151913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 04/15/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
Renal fibrosis, particularly tubulointerstitial fibrosis, is a prevalent pathological process contributing to the progression of chronic kidney disease (CKD). A growing body of evidence indicates that fibroblast growth factors (FGFs) play critical roles in kidney pathophysiology. However, the role of FGF18 in the pathogenesis of kidney fibrosis and the underlying mechanisms remain unclear. In this study, we discovered a significant upregulation of FGF18 in a folic acid (FA)-induced renal fibrosis model, as well as in transforming growth factor β (TGF-β) stimulated human proximal tubular cells (HK-2 cells). Furthermore, overexpression of FGF18 in the kidney significantly alleviated FA-induced fibrosis and diminished oxidative stress. Mechanistically, FGF18 upregulated AMP-activated protein kinase (AMPK) phosphorylation via its receptor FGFR3, leading to decreased NOX4-ROS levels, reduced oxidative stress, and ultimately inhibited the expression of fibrosis-related proteins. In conclusion, our findings suggest that FGF18 attenuates FA-induced renal fibrosis, at least in partly via AMPK/NOX4 pathway.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Yanni Shou
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Qiaoyan Shi
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Junbo Ye
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Xianzhe Li
- Life Science of Pharmacy, College of Pharmacy, The Catholic University of Korea, 43, Jibong-ro, Wonmi-gu, Bucheon-si, Gyeonggi-do, 14662, South Korea
| | - Zhongxin Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China.
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
5
|
Gu W, Pan T, Wang X, Kang L, Liu N, Piao M, Feng C. Sevoflurane exposure triggers ferroptosis of neuronal cells initiated by the activation of ATM/p53 in the neonatal mouse brain via JNK/p38 MAPK-mediated oxidative DNA damage. Int Immunopharmacol 2025; 158:114866. [PMID: 40378436 DOI: 10.1016/j.intimp.2025.114866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/27/2025] [Accepted: 05/09/2025] [Indexed: 05/18/2025]
Abstract
Neuronal death has long been regarded as a pivotal pathological factor in the developmental neurotoxicity caused by the volatile anesthetic sevoflurane in the neonatal brain, but the detailed mechanism remains controversial. Ferroptosis is a novel type of regulated cell death driven by excess lipid peroxidation secondary to intracellular iron overload, and it is implicated in the pathogenesis of various neurological disorders. Acting as a death messenger, p53 is primarily activated by ATM during DNA damage and mediates various forms of cell death, including apoptosis, autophagy, and ferroptosis. JNK/p38 MAPK are important stress-responsive pathways that can exacerbate intracellular ROS production, thereby linking DNA damage to many pathological conditions such as neurodegeneration and ischemic injury. In our present study, we demonstrated that sevoflurane exposure-induced neuronal death was correlated with intracellular iron overload and lipid peroxidation in HT22 cells, primary hippocampal neurons, and the hippocampi of neonatal mice, consistent with the hallmarks of ferroptosis. Furthermore, we found that sevoflurane-induced neuronal ferroptosis was associated with ATM/p53 activation in response to DNA damage. Additionally, sevoflurane exposure caused JNK/p38 MAPK activation followed by intracellular ROS accumulation, ultimately leading to DNA damage. Mechanistically, ATM/p53 contributed to ferroptosis caused by sevoflurane via two pathways: (1) enhancing iron uptake (upregulating TFR and downregulating FPN) and (2) promoting lipid peroxidation through NOX4, ALOX12, ALOX15 activation and SLC7A11 suppression. Collectively, these findings demonstrated that sevoflurane exposure induced ferroptosis of neuronal cells in the neonatal brain, triggered by ATM/p53 activation via JNK/p38 MAPK-mediated ROS accumulation and subsequent DNA damage.
Collapse
Affiliation(s)
- Wanping Gu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Tingting Pan
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Xuedong Wang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Liheng Kang
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Nan Liu
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China.
| | - Chunsheng Feng
- Department of Anesthesiology, The First Hospital of Jilin University, No. 1 Xinmin St., Changchun 130021, China.
| |
Collapse
|
6
|
Vlad ML, Mares RG, Jakobsson G, Manea SA, Lazar AG, Preda MB, Popa MA, Simionescu M, Schiopu A, Manea A. Therapeutic S100A8/A9 inhibition reduces NADPH oxidase expression, reactive oxygen species production and NLRP3 inflammasome priming in the ischemic myocardium. Eur J Pharmacol 2025; 996:177575. [PMID: 40180274 DOI: 10.1016/j.ejphar.2025.177575] [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: 10/09/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Oxidative stress and alterations in redox signalling have been implicated in the pathophysiology of myocardial infarction (MI). NADPH oxidase (Nox) is an important source of reactive oxygen species (ROS) in the infarcted myocardium. Alarmin S100A8/A9 amplifies acute myocardial inflammation in MI and has been shown to be a promising therapeutic target to improve cardiac function post-MI. We aimed to elucidate the underlying mechanisms linking S100A8/A9, oxidative stress and the inflammatory response in MI. MI was induced by permanent left coronary artery ligation in C57BL/6J mice, followed by treatment with the S100A8/A9 inhibitor ABR-238901 (30 mg/kg) or PBS for 3 days. The in-vivo experiments were complemented with mechanistic studies on cultured macrophages (Mac), important cellular effectors in MI. Compared to sham-operated animals, we detected significant increases in the Nox1, Nox2, Nox4 catalytic subunits at mRNA and protein levels, and NADPH-dependent ROS production in the left ventricle of MI mice. S100A8/A9 blockade prevented the up-regulation of Nox1/2/4 expression, reduced ROS formation, suppressed NF-kB activation and prevented NLRP3 inflammasome priming and activation, leading to reduced levels of active IL-1β. In-vitro, S100A8/A9 induced gene expression of Nox catalytic subtypes and NLRP3 in Mac in a TLR4-dependent and dose-dependent manner. These effects were counteracted by pharmacological inhibition of S100A8/9, TLR4, Nox1/4 and Nox2. In conclusion, we show that Nox upregulation and ROS formation triggered by S100A8/A9 contributes to NLRP3 inflammasome priming and increased IL-1β production in the infarcted myocardium. These mechanisms can be therapeutically targeted to prevent inflammatory and oxidant myocardial damage in acute MI.
Collapse
Affiliation(s)
- Mihaela-Loredana Vlad
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Razvan Gheorghita Mares
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania; Department of Cardiology II, Emergency Clinical County Hospital, Targu Mures, Romania.
| | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden.
| | - Simona-Adriana Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Alexandra-Gela Lazar
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Mihai Bogdan Preda
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Mirel Adrian Popa
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| | - Alexandru Schiopu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania; Department of Translational Medicine, Lund University, Malmö, Sweden; Department of Internal Medicine, Skåne University Hospital, Lund, Sweden.
| | - Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, Bucharest, Romania.
| |
Collapse
|
7
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [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: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
8
|
Li N, Chen Y, Xia X, Mao C, Wan M. Progress of nanomaterials in the treatment of ischemic heart disease. J Mater Chem B 2025; 13:6021-6043. [PMID: 40331910 DOI: 10.1039/d5tb00471c] [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: 05/08/2025]
Abstract
Medical or surgical interventions are commonly used to alleviate the clinical symptoms of individuals suffering from ischemic heart disease (IHD), but global morbidity and mortality remain high. This is due to the complexity of disease progression and the pathological basis of IHD, which primarily includes myocardial infarction (MI), myocardial ischemia-reperfusion injury (IRI), and heart failure (HF), as well as underlying mechanisms, such as mitochondrial damage, inflammation, oxidative stress, and cardiomyocyte death. However, many drugs have limitations, such as poor stability and low bioavailability, and surgical strategies are often ineffective in preventing disease recurrence. To overcome these problems, it is necessary to develop effective drug delivery systems and technologies. Due to their advantages in enhancing drug utilization, nanomaterials are being used to control drug biodistribution and achieve targeted accumulation, addressing the therapeutic needs of IHD. In this work, we first described the clinical aspects of MI, IRI, and HF in the context of IHD as well as their shared pathological origins. Next, clinical interventional procedures for IHD are summarized. Finally, recent developments in the use of nanomaterials for the treatment of MI, IRI, and HF are highlighted, along with potential directions for future research.
Collapse
Affiliation(s)
- Nan Li
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
- State Key Laboratory of Transvascular Implantation Devices, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310027, P. R. China
- Transvascular Implantation Devices Research Institute, Hangzhou, 310053, China
| | - Yu Chen
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Xue Xia
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
9
|
Zhang J, Wang H, Xue X, Wu X, Li W, Lv Z, Su Y, Zhang M, Zhao K, Zhang X, Jia C, Zhu F. Human endogenous retrovirus W family envelope protein (ERVWE1) regulates macroautophagy activation and micromitophagy inhibition via NOXA1 in schizophrenia. Virol Sin 2025:S1995-820X(25)00065-3. [PMID: 40419114 DOI: 10.1016/j.virs.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025] Open
Abstract
The human endogenous retrovirus type W envelope glycoprotein (ERVWE1), located at chromosome 7q21-22, has been implicated in the pathophysiology of schizophrenia. Our previous studies have shown elevated ERVWE1 expression in schizophrenia patients. Growing evidence suggests that autophagy dysfunction contributes to schizophrenia, yet the relationship between ERVWE1 and autophagy remains unclear. In this study, bioinformatics analysis of the human prefrontal cortex RNA microarray dataset (GSE53987) revealed that differentially expressed genes were predominantly enriched in autophagy-related pathways. Clinical data further demonstrated that serum levels of microtubule-associated protein 1 light chain 3β (LC3B), a key marker of macroautophagy, were significantly elevated in schizophrenia patients compared to controls, and positively correlated with ERVWE1 expression. Cellular and molecular experiments suggested that ERVWE1 promoted macroautophagy by increasing the LC3B II/I ratio, enhancing autophagosome formation, and reducing sequestosome 1 (SQSTM1) expression via upregulation of NADPH oxidase activator 1 (NOXA1). Concurrently, NOXA1 downregulated the expression of key micromitophagy-related genes, including PTEN-induced kinase 1 (PINK1), Parkin RBR E3 ubiquitin-protein ligase (Parkin), and the pyruvate dehydrogenase E1 subunit α 1 (PDHA1). As a result, ERVWE1, via NOXA1, inhibited micromitophagy by suppressing the expression of PINK1, Parkin, and PDHA1, thereby leading to impaired production of mitochondrial-derived vesicles (MDVs). Mechanistically, ERVWE1 enhanced NOXA1 transcription by upregulating upstream transcription factor 2 (USF2). In conclusion, ERVWE1 promotes macroautophagy and inhibits micromitophagy through USF2-NOXA1 axis, providing novel mechanistic insight into the role autophagy dysregulation in schizophrenia. These findings suggest that targeting autophagy pathways may offer novel therapeutic strategies for schizophrenia treatment.
Collapse
Affiliation(s)
- Jiahang Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Huiling Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Jiefang Road No.238, Wuhan, 430060, China
| | - Xing Xue
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xiulin Wu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Wenshi Li
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Zhao Lv
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Yaru Su
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Mengqi Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Kexin Zhao
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Xu Zhang
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Chen Jia
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China
| | - Fan Zhu
- State Key Laboratory of Virology and Biosafety, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China; Hubei Province Key Laboratory of Allergy and Immunology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, Hubei Province, China.
| |
Collapse
|
10
|
Zhao X, Gu C, Wang M, Huang S, Gong X, Kang M, Zhang T, Shen J, Yang X, Xi Y, Pan J. Human Neural Progenitor Cell-Derived Exosomes Deliver miR- 100 - 5p Targeting NOX4 mRNA to Alleviate Oxidative Stress in Acute Ischemia Injury. Mol Neurobiol 2025:10.1007/s12035-025-04952-z. [PMID: 40402409 DOI: 10.1007/s12035-025-04952-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/13/2025] [Indexed: 05/23/2025]
Abstract
The prevention and treatment of acute ischemic stroke have been longstanding challenges. Therapies targeting angiogenesis hold promising potential for ischemic injury repair. Reactive oxygen species (ROS) production, induced by the overexpression of NADPH oxidase 4 (NOX4), is a key factor that inhibits angiogenesis during the acute phase of ischemia/reperfusion. Therefore, targeting NOX4 gene expression can reduce ROS production and promote angiogenesis. In this study, human neural progenitor cell (hNPC)-derived-exosomal miRNAs were identified using high-throughput sequencing and online database, with miR-100-5p showing potential to suppress NOX4 expression. We then incorporated exogenous miR-100-5p into hNPC-derived exosomes through electroporation. These miR-100-5p-loaded exosomes were then applied to in vitro models of mouse brain microvascular endothelial cells (bEND.3) subjected to oxygen glucose deprivation and reperfusion (OGD/R). The results showed that miR-100-5p could significantly reduce NOX4 expression and ROS levels induced by OGD/R in bEND.3 cells. Similarly, in vivo analysis showed that mice models of middle cerebral artery occlusion (MCAO) injected with hNPC-derived exosomes loaded with miR-100-5p exhibited smaller brain infarct size, reduced apoptosis, and improved neurological performance compared to MCAO mice receiving PBS. Our findings demonstrate the successful delivery of miR-100-5p via hNPC-derived exosomes and its protective effect on brain microvascular endothelial cells following ischemia injury.
Collapse
Affiliation(s)
- Xianlei Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Min Wang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Sicong Huang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xinghan Gong
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Muxue Kang
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Tiesong Zhang
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jian Shen
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaohang Yang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yongmei Xi
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China.
- Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, 310000, China.
- Center for Genetic Medicine, Zhejiang University Lnternational Institute of Medicine, Yiwu, 322000, China.
- Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, 310000, China.
- The Women's Hospital and Institute of Genetics, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
11
|
Cui Y, Wu Y, Shi P, Ni Y, Zeng H, Zhang Z, Zhao C, Sun W, Yi Q. Mitigating microplastic-induced organ Damage: Mechanistic insights from the microplastic-macrophage axes. Redox Biol 2025; 84:103688. [PMID: 40412021 DOI: 10.1016/j.redox.2025.103688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 05/10/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025] Open
Abstract
We live in a world increasingly dominated by plastic, leading to the generation of microplastic particles that pose significant global health concerns. Microplastics can enter the body via ingestion, inhalation, and direct contact, accumulating in various tissues and potentially causing harm. Despite this, the specific cellular mechanisms and signaling pathways involved remain poorly understood. Macrophages are essential in absorbing, distributing, and eliminating microplastics, playing a key role in the body's defense mechanisms. Recent evidence highlights oxidative stress signaling as a key pathway in microplastic-induced macrophage dysfunction. The accumulation of microplastics generates reactive oxygen species (ROS), disrupting normal macrophage functions and exacerbating inflammation and organ damage. This review serves as the first comprehensive examination of the interplay between microplastics, macrophages, and oxidative stress. It discusses how oxidative stress mediates macrophage responses to microplastics and explores the interactions with gut microbiota. Additionally, it reviews the organ damage resulting from alterations in macrophage function mediated by microplastics and offers a novel perspective on the defense, assessment, and treatment of microplastic-induced harm from the viewpoint of macrophages.
Collapse
Affiliation(s)
- Yinxing Cui
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China; Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Yuqi Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Pan Shi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Yan Ni
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Huaying Zeng
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China
| | - Zhao Zhang
- Department of General Surgery, Dongguan Huangjiang Hospital, Dongguan, 523061, Guangdong, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| | - Weichao Sun
- Department of Orthopedics, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, 518035, China.
| | - Qian Yi
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646099, China.
| |
Collapse
|
12
|
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
|
13
|
Gu Y, Xia J, Guo Y, Tao L, Zhang G, Xu J. Leukemia cells remodel bone marrow stromal cells to generate a protumoral microenvironment via the S100A8-NOX2-ROS signaling pathway. Sci Rep 2025; 15:17179. [PMID: 40382430 PMCID: PMC12085644 DOI: 10.1038/s41598-025-01711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
The bone marrow microenvironment (BMM) plays a crucial role in the pathogenesis and progression of acute myeloid leukemia (AML). AML cells can modify the BMM to establish a more favorable environment for their survival. However, the mechanism about the complex regulatory interplay between the BMM and AML cells remains unclear. In this study, we used proteomic analysis to elucidate the potential mechanisms underlying the interaction between bone marrow stromal cells (BMSCs) and AML cells. We found that the co-culture of AML cells and BMSCs facilitated the proliferation of AML cells, suppressed the proliferation of BMSCs and triggered their senescence. Furthermore, we show the aberrant expression of S100A8 that plays a crucial role in the communication between AML cells and BMSCs. In the co-culture system, overexpression of S100A8 in AML cells activated NOX2 and induced the production of reactive oxygen species (ROS) in the supernatant, thereby suppressing the proliferation of BMSCs and facilitating the senescence of BMSCs. Subsequently, aging BMSCs secreted a variety of cytokines, including IL-6, CXCL5, MIP-1b, etc. as shown by Cytokine Array and qPCR analysis, which had stimulatory effects on the progression of AML. In conclusion, the present study reveals the crucial involvement of the S100A8-NOX2-ROS signaling pathway in mediating communication between AML cells and BMSCs, suggesting that targeting S100A8 may constitute an efficient strategy for AML therapy.
Collapse
Affiliation(s)
- Yangyang Gu
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Jingyi Xia
- Department of Blood Transfusion, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yuhong Guo
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Linfen Tao
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China
| | - Guanbin Zhang
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China.
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China.
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Jianping Xu
- Department of Laboratory Medicine, School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China.
- Key Laboratory of Clinical Laboratory Technology for Precision Medicine (Fujian Medical University), Fujian Province University, Fuzhou, Fujian, China.
| |
Collapse
|
14
|
Park S, Kim C, Heo S, Kang D. Endosomal H 2O 2 Molecules Act as Signaling Mediators in Akt/PKB Activation. Antioxidants (Basel) 2025; 14:594. [PMID: 40427476 DOI: 10.3390/antiox14050594] [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: 04/11/2025] [Revised: 05/14/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Receptor-mediated endocytosis (RME) is a commonly recognized receptor internalization process of receptor degradation or recycling. However, recent studies have supported that RME is closely related to signal propagation and amplification from the plasma membrane to the cytosol. Few studies have elucidated the role of H2O2, a mild oxidant among reactive oxygen species (ROS) in RME and second messenger of signal propagation. In the present study, we investigated the regulatory function of H2O2 in early endosomes during signaling throughout receptor-mediated endocytosis. In mammalian cells with a physiological amount of H2O2 generated during epidermal growth factor (EGF) activation, fluorescence imaging showed that the levels of two activating phosphorylations on Ser473 and Thr308 of Akt were transiently increased in the plasma membrane, but the predominant p-Akt on Ser473 appeared in early endosomes. To examine the role of endosomal H2O2 molecules as signaling mediators of Akt activation in endosomes, we modulated endosomal H2O2 through the ectopic expression of an endosomal-targeting catalase (Cat-Endo). The forced removal of endosomal H2O2 inhibited the Akt phosphorylation on Ser473 but not on Thr308. The levels of mSIN and rictor, two components of mTORC2 that work as a kinase in Akt phosphorylation on Ser473, were also selectively diminished in the early endosomes of Cat-Endo-expressing cells. We also observed a decrease in the endosomal level of the adaptor protein containing the PH domain, the PTB domain, and the Leucine zipper motif 1 (APPL1) protein, which is an effector of Rab5 and key player in the assembly of signaling complexes regulating the Akt pathway in Cat-Endo-expressing cells compared with those in normal cells. Therefore, the H2O2-dependent recruitment of the APPL1 adaptor protein into endosomes was required for full Akt activation. We proposed that endosomal H2O2 is a promoter of Akt signaling.
Collapse
Affiliation(s)
- Sujin Park
- Department of Life Science, Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Chaewon Kim
- Department of Life Science, Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Sukyeong Heo
- Department of Life Science, Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul 03760, Republic of Korea
- Department of Biomedical Engineering, Dongguk University, Seoul 10326, Republic of Korea
| | - Dongmin Kang
- Department of Life Science, Fluorescence Core Imaging Center and Bioimaging Data Curation Center, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
15
|
Bhatt KS, Singh A, Marwaha GS, Ravendranathan N, Sandhu IS, Kim K, Singh E, Frisbee JC, Singh KK. Different Mechanisms in Doxorubicin-Induced Neurotoxicity: Impact of BRCA Mutations. Int J Mol Sci 2025; 26:4736. [PMID: 40429877 DOI: 10.3390/ijms26104736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/30/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The genotoxic drug doxorubicin (Dox) remains one of the most powerful chemotherapeutic options available for a wide range of cancers including breast, ovarian, and other cancers. However, emerging evidence links Dox treatment with chemotherapy-induced cognitive impairment, a condition that is popularly referred to as Dox-induced neurotoxicity or "chemobrain", which limits the use of the drug. There are no specific treatments for Dox-induced neurotoxicity, only interventions to mitigate the neurotoxic effects of the drug. Accumulating evidence indicates that DNA damage, oxidative stress, dysregulation of autophagy and neurogenesis, inflammation, and apoptosis play central roles in Dox-induced neurotoxicity. Additionally, germline mutations in the tumour suppressor genes breast cancer susceptibility genes 1 and 2 (BRCA1 and BRCA2) increase the risk of breast, ovarian, and related cancers. BRCA1 and BRCA2 are distinct proteins that play crucial, unique roles in homologous recombination-mediated double-stranded break repair. Furthermore, BRCA1 and 2 mitigate oxidative stress in both neural cells and brain microvascular endothelial cells, which suggests that they have a critical role as regulators of pathways central to the development of Dox-induced neurotoxicity. Despite research on the effects of Dox on cognitive function, there is a gap in knowledge about the role of BRCA1 and BRCA2 in Dox-induced neurotoxicity. In this review, we discuss existing findings about the role of different mechanisms and the role of BRCA1 and BRCA2 in Dox-induced neurotoxicity, along with future perspectives.
Collapse
Affiliation(s)
- Kriti S Bhatt
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Aman Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Gursharan S Marwaha
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Naresh Ravendranathan
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Inderbir S Sandhu
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Kristen Kim
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Eesha Singh
- London Central Secondary School, London, ON N6B 2P8, Canada
| | - Jefferson C Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Krishna K Singh
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada
| |
Collapse
|
16
|
Meister M, He X, Noël A, Park JA, Crotty Alexander L, Zelikoff J, Christiani D, Hess J, Shannahan J, Wright C. Beyond the puff: health consequences of vaping. Inhal Toxicol 2025:1-14. [PMID: 40367291 DOI: 10.1080/08958378.2025.2500646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/26/2025] [Indexed: 05/16/2025]
Abstract
Electronic nicotine delivery systems (ENDS) arrived on the U.S. market in 2007 and rapidly grew in popularity as a harm reduction tool for traditional cigarette users. While initially marketed as a healthier alternative to combustible cigarettes, the unique mixture of chemical constituents in ENDS products and their emissions have led to rising concern about their safety and the long-term health implications. Given the lack of long-term, epidemiological research on the health effects of these products, recent research has sought to understand the impacts on cellular components and gain understanding of acute effects to inform potential chronic health implications. Studies have demonstrated the deleterious effects the use of ENDS has on the oral cavity, respiratory, and cardiovascular systems. ENDS use has been linked to gingival inflammation and alterations in the oral microbiome contributing to periodontal disease. Further, the presence of heavy metals and other constituents in ENDS emissions contribute to aberrant oxidative stress and inflammation within the lung, contributing to alterations in functional lung capacity and respiratory symptoms in ENDS users. In addition, harmful components of ENDS emissions make their way to the circulatory system, leading to detrimental impacts in cardiovascular functioning such as a rise in blood pressure, impaired vascular functioning, and increased heart rate, all of which are known to underscore long-term cardiovascular ailments. This review will provide an in-depth discussion of the current literature available on the consequences of ENDS use on the oral cavity, respiratory, and cardiovascular systems as well as provide insight into long-term implications that may result.
Collapse
Affiliation(s)
- Maureen Meister
- Chemical Insights Research Institute, UL Research Institutes, Marietta, GA, USA
| | - Xiaojia He
- Chemical Insights Research Institute, UL Research Institutes, Marietta, GA, USA
| | - Alexandra Noël
- Comparative Biomedical Sciences, Louisiana State University, LA, USA
| | - Jin-Ah Park
- T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Judith Zelikoff
- Langone Medical Center, New York University, New York City, NY, USA
| | - David Christiani
- T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Joseph Hess
- Chemical Insights Research Institute, UL Research Institutes, Marietta, GA, USA
| | | | - Christa Wright
- Chemical Insights Research Institute, UL Research Institutes, Marietta, GA, USA
| |
Collapse
|
17
|
Liu W, Wang YR, Wu H, Cui W, Xu X. The role of myeloperoxidase in the pathogenesis of stroke. Brain Res 2025; 1861:149705. [PMID: 40379076 DOI: 10.1016/j.brainres.2025.149705] [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: 03/18/2025] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/19/2025]
Abstract
Stroke is the leading cause of mortality and morbidity worldwide, significantly impacting human welfare and overall health. Myeloperoxidase (MPO), a heme peroxidase secreted by neutrophils, plays a crucial role in the body's defense mechanisms, exhibiting pro-inflammatory and pro-oxidative properties. Additionally, MPO compromises the structural integrity and functional capacity of blood vessels, potentially leading to the formation and dislodgement of atherosclerotic plaques, vascular stenosis, thrombosis, and ultimately contributing to stroke occurrence. Following a stroke, a significant influx of neutrophils infiltrates the cerebral tissue, leading to an excessive release of MPO-derived oxidants and the subsequent promotion of various inflammatory mediators, thereby exacerbating cerebral tissue damage. Numerous studies have consistently demonstrated the pivotal role of MPO in the pathogenesis and progression of stroke, establishing it as a reliable prognostic indicator. Exploring the association between MPO and stroke enhances our understanding of the pathological mechanisms underlying stroke and aids in the development of therapeutic interventions. This review provides a comprehensive analysis of the molecular structure and cellular localization of MPO, elucidating its critical role in mediating vascular injury, the formation of Neutrophil Extracellular Traps (NETs), oxidative stress, neuroinflammation, disruption of the blood-brain barrier (BBB), and neuronal apoptosis during stroke pathogenesis. Additionally, we discuss recent advancements in MPO-targeted drugs and Traditional Chinese Medicine compounds as potential therapeutic strategies for stroke treatment.
Collapse
Affiliation(s)
- Wei Liu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yi-Ran Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Hongyun Wu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Wenqiang Cui
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| | - Xiangqing Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China; Department of Neurology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China.
| |
Collapse
|
18
|
Bezdíček J, Sekaninová J, Janků M, Makarevič A, Luhová L, Dujíčková L, Petřivalský M. Reactive oxygen and nitrogen species: multifaceted regulators of ovarian activity†. Biol Reprod 2025; 112:789-806. [PMID: 39936599 PMCID: PMC12078082 DOI: 10.1093/biolre/ioaf032] [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: 05/02/2024] [Revised: 09/10/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025] Open
Abstract
Reactive oxygen and nitrogen species are essential components of diverse intracellular signaling pathways. In addition to their involvement in apoptosis, reactive oxygen and nitrogen species are crucial in the regulation of multiple developmental and physiological processes. This review aims to summarize their role in the regulation of key ovarian stages: ovulation, maturation and postovulatory ageing of the oocyte, and the formation and regression of the corpus luteum. At the cellular level, a mild increase in reactive oxygen and nitrogen species is associated with the initiation of a number of regulatory mechanisms, which might be suppressed by increased activity of the antioxidant system. Moreover, a mild increase in reactive oxygen and nitrogen species has been linked to the control of mitochondrial biogenesis and abundance in response to increased cellular energy demands. Thus, reactive oxygen and nitrogen species should also be perceived in terms of their positive role in cellular signaling. On the other hand, an uncontrolled increase in reactive oxygen species production or strong down-regulation of the antioxidant system results in oxidative stress and damage of cellular components associated with ovarian pathologies and ageing. Similarly, the disturbance of signaling functions of reactive nitrogen species caused by dysregulation of nitric oxide production by nitric oxide synthases in ovarian tissues interferes with the proper regulation of physiological processes in the ovary.
Collapse
Affiliation(s)
- Jiří Bezdíček
- Department of Zoology, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Jana Sekaninová
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Martina Janků
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Alexander Makarevič
- National Agricultural and Food Centre, Research Institute for Animal Production Nitra, Lužianky-near-Nitra, Slovak Republic
| | - Lenka Luhová
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| | - Linda Dujíčková
- National Agricultural and Food Centre, Research Institute for Animal Production Nitra, Lužianky-near-Nitra, Slovak Republic
| | - Marek Petřivalský
- Department of Biochemistry, Faculty of Science, Palacký University, Olomouc, Czech Republic
| |
Collapse
|
19
|
Franklin ME, Grant JL, Lee GM, Alvarez-Ciara A, Bennett C, Mattis S, Gallardo N, Corrales N, Cui XT, Capadona JR, Streit WJ, Olivier JH, Keane RW, Dietrich WD, de Rivero Vaccari JP, Prasad A. Effects of iron accumulation and its chelation on oxidative stress in intracortical implants. Acta Biomater 2025:S1742-7061(25)00349-6. [PMID: 40355018 DOI: 10.1016/j.actbio.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
Long-term reliability of microelectrodes implanted in the cortex is hindered due to the foreign body response that occurs at the electrode-tissue interface. Following implantation, there is disruption of the blood-brain-barrier and vasculature, resulting in activation of immune cells and release of erythrocytes. As a result of hemolysis, erythrocytes degrade to heme and then to free iron. Excess free iron can participate in the Fenton Reaction, producing reactive oxygen species (ROS). Iron-mediated ROS production can contribute to oxidation of lipids, proteins, and DNA, facilitating a hostile environment of oxidative stress leading to oxidative cellular damage, cytotoxicity, and cell death. The objective of this study was to show the iron accumulation and the downstream effects of oxidative stress at the injury site. A 16-channel microelectrode array (MEA) was implanted in the rat somatosensory cortex. Our results indicated significant elevation of NOX complex subunits across timepoints, suggesting sustained oxidative stress. In a separate group of animals, we administered an iron chelator, deferoxamine mesylate (DFX), to evaluate the effects of chelation on iron accumulation, oxidative stress and damage, and neuronal survival. Results indicate that animals with iron chelation showed reduced ferric iron and markers of oxidative stress and damage corresponding with increased expression of neuronal cell bodies and electrophysiological functional performance. In summary, the study reveals the role of iron in mediating oxidative stress and the effects of modulating iron levels using iron chelation at the electrode-tissue interface. STATEMENT OF SIGNIFICANCE: Iron accumulation has been observed in central nervous system injuries and in neurodegenerative diseases such as Alzheimer's and Parkinson's disease. While the role of iron is studied in various neurodegenerative diseases and traumatic brain injury, iron accumulation and its effect on oxidative stress is not known for intracortical implants where there is a persistent injury due to the presence of a foreign device in the brain tissue. The study seeks to understand the effects of iron accumulation on oxidative stress and damage at the electrode-tissue interface in intracortical implants by using iron chelation as a method of modulating iron levels at the interface.
Collapse
Affiliation(s)
- Melissa E Franklin
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Jordan L Grant
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Grant M Lee
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | | | - Cassie Bennett
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Serene Mattis
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Nicolas Gallardo
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Natalie Corrales
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
| | - Xinyan Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeffrey R Capadona
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Wolfgang J Streit
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | | | - Robert W Keane
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - W Dalton Dietrich
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Cellular Physiology and Molecular Biophysics, University of Miami Miller School of Medicine, Miami, FL, USA; Center for Cognitive Neuroscience and Aging University of Miami Miller School of Medicine, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, University of Miami, Miami, FL, USA; Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
20
|
Zhang R, Vooijs MA, Keulers TG. Key Mechanisms in Lysosome Stability, Degradation and Repair. Mol Cell Biol 2025; 45:212-224. [PMID: 40340648 DOI: 10.1080/10985549.2025.2494762] [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: 11/21/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Lysosomes are organelles that play pivotal roles in macromolecule digestion, signal transduction, autophagy, and cellular homeostasis. Lysosome instability, including the inhibition of lysosomal intracellular activity and the leakage of their contents, is associated with various pathologies, including cancer, neurodegenerative diseases, inflammatory diseases and infections. These lysosomal-related pathologies highlight the significance of factors contributing to lysosomal dysfunction. The vulnerability of the lysosomal membrane and its components to internal and external stimuli make lysosomes particularly susceptible to damage. Cells are equipped with mechanisms to repair or degrade damaged lysosomes to prevent cell death. Understanding the factors influencing lysosome stabilization and damage repair is essential for developing effective therapeutic interventions for diseases. This review explores the factors affecting lysosome acidification, membrane integrity, and functional homeostasis and examines the underlying mechanisms of lysosomal damage repair. In addition, we summarize how various risk factors impact lysosomal activity and cell fate.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Marc A Vooijs
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Tom Gh Keulers
- Department of Radiation Oncology (MAASTRO)/GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
21
|
Zhang X, Gao X, Chi Z. Metabolite Monomethyl Phthalate (MMP) Induces Oxidative Damage in Rat Erythrocytes: Role of Vitamins C and E. TOXICS 2025; 13:379. [PMID: 40423458 DOI: 10.3390/toxics13050379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/28/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025]
Abstract
Dimethyl phthalate (DMP) can enter the human body and be absorbed into the bloodstream to produce monomethyl phthalate (MMP). MMP in the environment can also enter the bloodstream. However, little is known about the toxicity of the phthalate metabolite MMP in most organisms. In this study, the erythrocyte toxicity of MMP and a preventive approach were investigated using Sprague-Dawley (SD) rats as the model animal under MMP concentrations of 5-250 mg/kg (sub-chronic exposure in vivo) and 1.25-100 μg/mL (acute exposure in vitro). The experimental results indicate that the interaction of MMP with erythrocytes caused oxidative damage, which decreased the number of red blood cells and the hemoglobin content and increased the content of methemoglobin and the iron release of hemoglobin in rat blood. However, the above results were not observed when MMP directly interacted with hemoglobin. The antioxidants vitamin C and vitamin E improved the above blood indicators in rats. The results of this study provide certain theoretical guidance for the evaluation of the potential risks of phthalate metabolites.
Collapse
Affiliation(s)
- Xuxin Zhang
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| | - Xu Gao
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
| | - Zhenxing Chi
- School of Marine Science and Technology, Harbin Institute of Technology, Weihai 264209, China
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Hangzhou 310015, China
| |
Collapse
|
22
|
Xiong Z, Liao Y, Zhang Z, Wan Z, Liang S, Guo J. Molecular Insights into Oxidative-Stress-Mediated Cardiomyopathy and Potential Therapeutic Strategies. Biomolecules 2025; 15:670. [PMID: 40427563 DOI: 10.3390/biom15050670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiomyopathies comprise a heterogeneous group of cardiac disorders characterized by structural and functional abnormalities in the absence of significant coronary artery disease, hypertension, valvular disease, or congenital defects. Major subtypes include hypertrophic, dilated, arrhythmogenic, and stress-induced cardiomyopathies. Oxidative stress (OS), resulting from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has emerged as a key contributor to the pathogenesis of these conditions. ROS-mediated injury drives inflammation, protease activation, mitochondrial dysfunction, and cardiomyocyte damage, thereby promoting cardiac remodeling and functional decline. Although numerous studies implicate OS in cardiomyopathy progression, the precise molecular mechanisms remain incompletely defined. This review provides an updated synthesis of current findings on OS-related signaling pathways across cardiomyopathy subtypes, emphasizing emerging therapeutic targets within redox-regulatory networks. A deeper understanding of these mechanisms may guide the development of targeted antioxidant strategies to improve clinical outcomes in affected patients.
Collapse
Affiliation(s)
- Zhenyu Xiong
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yuanpeng Liao
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhaoshan Zhang
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Zhengdong Wan
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiawei Guo
- Department of Vascular and Endovascular Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou 434000, China
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
23
|
Mondragon RR, Wang S, Stevenson MD, Lozhkin A, Vendrov AE, Isom LL, Runge MS, Madamanchi NR. NOX4-driven mitochondrial oxidative stress in aging promotes myocardial remodeling and increases susceptibility to ventricular tachyarrhythmia. Free Radic Biol Med 2025; 235:294-305. [PMID: 40320218 DOI: 10.1016/j.freeradbiomed.2025.04.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/16/2025]
Abstract
Aging-associated mitochondrial oxidative stress has been implicated in ventricular tachyarrhythmias (VT), but the specific proarrhythmic mechanisms of mitochondrial pro-oxidative systems remain unclear. NADPH oxidase 4 (NOX4) expression in the heart increases with age, leading to mitochondrial oxidative stress, dysfunction, and adverse myocardial remodeling. This study investigated the susceptibility to aging-associated ventricular arrhythmia and the associated triggers and substrates using transgenic mice with mitochondria-targeted Nox4 overexpression (Nox4TG mice). Nox4TG mice showed a significantly higher incidence of pacing-induced VT, associated with shorter action potential duration (APD) due to increased transient outward potassium currents. Fractional sarcoplasmic reticulum (SR) Ca2+ release and Ca2+ leak remained intact despite these changes. However, the frequency of Ca2+ sparks was reduced, and ryanodine receptor 2 (RyR2) oxidation was observed. Compensatory upregulation of SERCA expression in response to RyR2 inhibition accelerated SR Ca2+ reuptake and improved cardiomyocyte mechanical relaxation. Nox4TG mice exhibited extensive ventricular fibrosis and marked pro-inflammatory macrophage infiltration, with elevated TNF, TGF-β, and MKI67 expression. Treatment with Setanaxib, a NOX1/NOX4 inhibitor, or co-expression of mitochondrial catalase in Nox4TG (Nox4TG + mCAT) mice, mitigated fibrosis, reduced inflammation, and protected against VT. These findings suggest that mitochondrial NOX4 overexpression promotes VT through electrical remodeling and pro-arrhythmogenic structural changes despite RyR2 oxidation and dysfunction. In conclusion, aging-related NOX4-driven mitochondrial oxidative stress increases the risk of VT by promoting changes in the electrical and structural properties of the myocardium, highlighting potential therapeutic strategies that target NOX4 in cardiac pathologies associated with aging.
Collapse
Affiliation(s)
- Roberto Ramos Mondragon
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Dr., 2301 Medical Science Research Building III, Ann Arbor, MI, 48109, USA
| | - Shuyun Wang
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Dr., 2301 Medical Science Research Building III, Ann Arbor, MI, 48109, USA
| | - Mark D Stevenson
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Aleksandr E Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Lori L Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Dr., 2301 Medical Science Research Building III, Ann Arbor, MI, 48109, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Marschall S Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Nageswara R Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Zhao Z, Wang R, Ge H, Hou L, Hatano T, Hattori N, Su H, Wang Q, Zhao J. ECHS1-NOX4 interaction suppresses rotenone-induced dopaminergic neurotoxicity through inhibition of mitochondrial ROS production. Free Radic Biol Med 2025; 232:56-71. [PMID: 40032032 DOI: 10.1016/j.freeradbiomed.2025.02.048] [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: 12/30/2024] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Parkinson's disease (PD) is the most common neurodegenerative movement disorder with uncleared mechanisms. Short-chain enoyl-CoA hydratase 1 (ECHS1) is a mitochondrial enzyme critical for the β-oxidation of fatty acids and ATP production. This study aims to explore the roles of ECHS1 in PD by using rotenone-induced experimental PD models. METHODS To evaluate the role of ECHS1 in rotenone-induced dopaminergic neurodegeneration, adeno-associated virus (AAV)-ECHS1 was stereotactically injected into the substantia nigra region of mice to overexpress ECHS1. Motor function of mice among groups was detected by rotarod test and gait analysis. Neurodegeneration, mitochondrial dysfunction and apoptosis were determined by immunohistochemistry, immunofluorescence staining, Western blot or kits, respectively. RESULTS The expression and activity of ECHS1 were decreased in PD mice and positive correlations between ECHS1 reduction and dopaminergic neurodegeneration were observed. Overexpression of ECHS1 by AAV delivery attenuated loss of dopaminergic neuron and motor deficits in PD mice. Mechanistically, ECHS1 attenuated rotenone-induced mitochondrial swelling and loss of cristae as well as decrease of ATP production, mitochondrial membrane potential, complex I/IV activities and oxygen consumption rate (OCR). Mitochondrial ROS (mtROS)-targeted antioxidant mito-TEMPO prevented ECHS1 silence-mediated mitochondrial dysfunction. Furthermore, we found that ECHS1 interacted with NADPH oxidase 4 (NOX4), resulting in decrease of NOX4 activation and subsequent reduction of mtROS production and mitochondrial dysfunction. Finally, inhibition of NOX4 by GLX351322 or mtROS production by mito-TEMPO greatly reduced ECHS1 silence-mediated apoptosis in rotenone-treated SH-SY5Y cells. CONCLUSIONS ECHS1 counteracted dopaminergic neurodegeneration through inhibition of mtROS and restoration of mitochondrial function via interaction with NOX4. Given the central role of mitochondrial dysfunction in PD pathogenesis, elucidating the role of ECHS1 holds great promise for uncovering novel therapeutic targets.
Collapse
Affiliation(s)
- Zirui Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Ruonan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Haitao Ge
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Liyan Hou
- Dalian Medical University Library, Dalian Medical University, No. 9 W. Lvshun South Road, Dalian, Liaoning Province, 116044, China
| | - Taku Hatano
- Department of Neurology, Juntendo University Faculty of Medicine. 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo, Tokyo, 113-8421, Japan; Neurodegenerative Disorders Collaborative Laboratory, RIKEN Center for Brain Science 2-1 Hirosawa, Wako-city, Saitama, 351-0198, Japan
| | - Hong Su
- School of Health-Preservation and Wellness, Dalian Medical University, Dalian, Liaoning Province, 116044, China
| | - Qingshan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Public Health, Dalian Medical University, Dalian, Liaoning Province, 116044, China.
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning Province, 116044, China; School of Integrated Chinese and Western Medicine, Dalian Medical University, Dalian, Liaoning Province, 116044, China.
| |
Collapse
|
25
|
Gong G, Wan W, Zhang X, Chen X, Yin J. Management of ROS and Regulatory Cell Death in Myocardial Ischemia-Reperfusion Injury. Mol Biotechnol 2025; 67:1765-1783. [PMID: 38852121 DOI: 10.1007/s12033-024-01173-y] [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: 12/13/2023] [Accepted: 04/02/2024] [Indexed: 06/10/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is fatal to patients, leading to cardiomyocyte death and myocardial remodeling. Reactive oxygen species (ROS) and oxidative stress play important roles in MIRI. There is a complex crosstalk between ROS and regulatory cell deaths (RCD) in cardiomyocytes, such as apoptosis, pyroptosis, autophagy, and ferroptosis. ROS is a double-edged sword. A reasonable level of ROS maintains the normal physiological activity of myocardial cells. However, during myocardial ischemia-reperfusion, excessive ROS generation accelerates myocardial damage through a variety of biological pathways. ROS regulates cardiomyocyte RCD through various molecular mechanisms. Targeting the removal of excess ROS has been considered an effective way to reverse myocardial damage. Many studies have applied antioxidant drugs or new advanced materials to reduce ROS levels to alleviate MIRI. Although the road from laboratory to clinic has been difficult, many scholars still persevere. This article reviews the molecular mechanisms of ROS inhibition to regulate cardiomyocyte RCD, with a view to providing new insights into prevention and treatment strategies for MIRI.
Collapse
Affiliation(s)
- Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xinghu Zhang
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xiangxuan Chen
- Department of Cardiology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Jiangsu Medical Vocational College, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Nanjing Medical University Kangda College, Nanjing, 211100, China.
| |
Collapse
|
26
|
Goossen CJ, Kufner A, Dustin CM, Al Ghouleh I, Yuan S, Straub AC, Sembrat J, Baust JJ, Gomez D, Kračun D, Pagano PJ. Redox regulation of lung endothelial PERK, unfolded protein response (UPR) and proliferation via NOX1: Targeted inhibition as a potential therapy for PAH. Redox Biol 2025; 82:103554. [PMID: 40154102 PMCID: PMC11986987 DOI: 10.1016/j.redox.2025.103554] [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: 12/17/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 04/01/2025] Open
Abstract
AIMS Reactive oxygen species (ROS) play an important role in the pathogenesis of pulmonary arterial hypertension (PAH) and NADPH oxidases (NOXs) as sources of ROS are implicated in the development of the disease. We previously showed that NOX isozyme 1 (NOX1)-derived ROS contributes to pulmonary vascular endothelial cell (EC) proliferation in response to PAH triggers in vitro. However, whether and how NOX1 is involved in PAH in vivo have not been explored nor has NOX1 been examined as a viable and effective therapeutic disease target. METHODS AND RESULTS Herein, infusion of mice exposed to Sugen/hypoxia (10 % O2) with a specific NOX1 inhibitor, NOXA1ds, delivered via osmotic minipumps (i.p.), significantly suppressed pathological changes in hemodynamic parameters characteristic of PAH. Furthermore, lungs of human patients with idiopathic PAH (iPAH) and exploratory RNA-seq analysis of hypoxic human pulmonary ECs, in which NOX1 was suppressed, were probed. The findings showed a clear indication of NOX1 in the promotion of both protein disulfide isomerase (PDI) and the unfolded protein response (UPR; in particular, the PERK arm of the pathway including eIF2α and ATF4) leading to proliferation. In aggregate, these results are consistent with a causal role for NOX1 in the development of mouse and human PAH and reveal a novel and mechanistic pathway by which NOX1 activates the UPR response during EC proliferation. CONCLUSION NOX1 promotes phenotypic changes in ECs that are pivotal to proliferation and PAH through activation of the UPR. Taken together, our results are consistent with selective inhibition of NOX1 as a novel modality for attenuating PAH.
Collapse
Affiliation(s)
- Christian J Goossen
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Alex Kufner
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Christopher M Dustin
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Imad Al Ghouleh
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Shuai Yuan
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Adam C Straub
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - John Sembrat
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Medicine, Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Jeffrey J Baust
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Delphine Gomez
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Damir Kračun
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Balgrist University Hospital, University of Zurich, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| | - Patrick J Pagano
- Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
27
|
Soto LJ, Del Tufiño C, Macias-Pérez ME, Castro-García S, Jiménez-Cruz E, Bobadilla-Lugo RA. Epicatechin prevents preeclampsia-associated hypertension and oxidative stress. J Obstet Gynaecol Res 2025; 51:e16290. [PMID: 40355116 DOI: 10.1111/jog.16290] [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: 11/15/2024] [Accepted: 03/28/2025] [Indexed: 05/14/2025]
Abstract
Preeclampsia (PE) is a frequent and dangerous multisystemic pregnancy complication, associated with blood pressure control. Some antioxidants, including chocolate-derived epicatechin, can effectively attenuate hypertensive disorders. AIM This study aimed to assess whether epicatechin or dark chocolate (DC) could revert vascular increased reactivity and oxidative stress, both features of an experimental PE model. METHODS Rats from healthy pregnant or PE groups received vehicle, epicatechin (10 mg/kg/day) po, or DC (1 g) po, administered on days 1-14 (early) or days 7-21(late) of pregnancy. Blood pressure was measured by the tail-cuff plethysmography method. Aorta contractility was evaluated using a conventional isolated organ bath, and oxidative stress was determined by nicotine adenine dinucleotide phosphate reduced (NADPH) serum activity. RESULTS Epicatechin and DC significantly reduced hypertension, decreased abdominal aorta contractility, and decreased NADPH activity of the PE animals. The effects were more evident when administered during the last 2 weeks of pregnancy. CONCLUSIONS Results suggest that epicatechin has a significant antihypertensive effect in PE mediated by an antioxidant activity that improves vascular contractility.
Collapse
Affiliation(s)
- Luis J Soto
- Escuela Superior de Medicina IPN, Mexico City, Mexico
| | | | | | | | | | | |
Collapse
|
28
|
Xiong B, Zhang Y, Liu S, Liao S, Zhou Z, He Q, Zhou Y. NOX Family: Regulators of Reactive Oxygen Species Balance in Tumor Cells. FASEB J 2025; 39:e70565. [PMID: 40266050 PMCID: PMC12017260 DOI: 10.1096/fj.202500238rrr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/24/2025]
Abstract
Cancer cells are capable of surviving, proliferating, and invading or migrating within hypoxic environments by regulating various adaptive mechanisms. Due to the activation of oncogenes and the inactivation of tumor suppressor genes, and relative deficiencies in oxygen and nutrients, cancer cells demonstrate elevated production of reactive oxygen species (ROS), primarily sourced from NADPH oxidases (NOX family). A key aspect of the reorientation of tumor cell metabolism is the combating of cellular oxidative stress through the promotion of antioxidant molecule synthesis to counteract ROS production. Given that most cancers experience hypoxia and that NOX is closely linked to numerous redox-dependent signaling pathways, the expression and function of NOX are altered in various malignancies. Therefore, this review summarizes the characteristics of NOX family members, their influence on tumor proliferation, invasion, and migration, the role of NOX in promoting tumor angiogenesis, the impact of NOX on the function of immune cells within the tumor microenvironment, and the potential of targeting NOX in tumor therapy. This aims to offer a fresh viewpoint on a comprehensive understanding of the functions of NOX family members.
Collapse
Affiliation(s)
- Bin Xiong
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Yang Zhang
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Siyi Liu
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| | - Shan Liao
- Department of PathologyThe Third Xiangya Hospital of Central South UniversityChangshaHunanChina
| | - Zihua Zhou
- Department of OncologyLoudi Central HospitalLoudiHunanChina
| | - Qian He
- Department of Radiation OncologyThe Affiliated Cancer Hospital of Xiangya School of Medicine Central South University/Hunan Cancer HospitalChangshaHunanChina
| | - Yanhong Zhou
- Cancer Research Institute, Basic School of MedicineCentral South UniversityChangshaHunanChina
| |
Collapse
|
29
|
Xiang G, Gong L, Wang K, Sun X, Liu Z, Cai Q. Suppression of NOX2-Derived Reactive Oxygen Species (ROS) Reduces Epithelial-to-MesEnchymal Transition Through Blocking SiO 2-Regulated JNK Activation. TOXICS 2025; 13:365. [PMID: 40423444 DOI: 10.3390/toxics13050365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025]
Abstract
(1) Background: Silicosis, a chronic lung fibrosis disorder triggered by the accumulation of silica dust in the deep lung regions, is characterized by intricate molecular mechanisms. Among these, the NOX2 (NADPH oxidase 2) and JNK (C-Jun N-terminal kinase) signaling pathways play pivotal roles in the progression of pulmonary fibrosis. Despite their significance, the precise mechanisms underlying the crosstalk between these pathways remain largely unexplored. (2) Methods: To unravel these interactions, we examined the interplay between JNK and NOX2 in human epithelial cells subjected to silica dust exposure through in vivo assays, followed by validation using single-cell sequencing. Our findings consistently revealed elevated expression levels of key components from both the JNK signaling pathway and NOX2 in the lungs of silicosis-induced mice and silica-treated human epithelial cells. (3) Results: Notably, the activation of these pathways was linked to increased ROS (reactive oxygen species) production, elevated levels of profibrogenic factors, and diminished cell proliferation in silica-exposed human lung epithelial cells. Further mechanistic analyses demonstrated that JNK signaling amplifies NOX2 expression and ROS production induced by silica exposure, while treatment with the JNK inhibitor SP600125 mitigates these effects. Conversely, overexpression of NOX2 enhanced silica-induced JNK activation and the expression of epithelial-mesenchymal transition (EMT)-related factors, whereas NOX2 knockdown exerted the opposite effect. These results suggest a positive feedback loop between JNK and NOX2 signaling, which may drive EMT in lung epithelial cells following silica exposure. (4) Conclusions: This reciprocal interaction appears to play a critical role in lung epithelial cell damage and the pathogenesis of silicosis, shedding light on the molecular mechanisms underlying profibrogenic disease and offering potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Guanhan Xiang
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| | - Liang Gong
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| | - Kai Wang
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| | - Xiaobo Sun
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| | - Zhihong Liu
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| | - Qian Cai
- School of Public Health, Ningxia Medical University, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750101, China
| |
Collapse
|
30
|
Magesh K, Wu SP, Velmathi S. A near-infrared julolidine probe for visualization of mitochondrial peroxynitrite in living cells. Org Biomol Chem 2025; 23:4142-4151. [PMID: 40168033 DOI: 10.1039/d5ob00036j] [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: 04/02/2025]
Abstract
The overproduction of peroxynitrite (ONOO-) in mitochondria has been associated with various pathophysiological conditions and disorders. However, the use of fluorescent probes to visualize mitochondrial ONOO- in biological systems is limited due to their low emission wavelengths and small Stokes shifts, which present significant challenges. In this study, we designed and synthesized julolidine-based near-infrared (NIR) fluorescent probes, named JQMe and JCN, specifically to monitor mitochondrial ONOO-. Comparative photophysical studies revealed that JQMe exhibits superior properties for sensing ONOO- compared to JCN. Initially, JQMe emitted fluorescence emission at 706 nm via an intramolecular charge transfer (ICT) mechanism. Upon the addition of ONOO-, the NIR fluorescence emission of JQMe at 706 nm was suppressed, resulting in a rapid on-off fluorescence response within 5 minutes. JQMe exhibited high specific selectivity towards ONOO- over other competing interferents, accompanied by a colorimetric change from deep blue to colorless. Additionally, JQMe exhibited a significant Stokes shift of 106 nm and a low detection limit of 6.5 nM. The proposed sensing mechanism was validated through ESI mass spectrometry and DFT studies. Furthermore, JQMe was successfully employed to monitor both endogenous and exogenous ONOO- in living cells using inducer and inhibitor tests. Remarkably, time-dependent colocalization experiments revealed that JQMe effectively targets and reacts with mitochondrial ONOO-.
Collapse
Affiliation(s)
- Kuppan Magesh
- Organic and Polymer Synthesis Laboratory, Department of Chemistry, National Institute of Technology, Tiruchirappalli - 620 015, India.
| | - Shu Pao Wu
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan, China
| | - Sivan Velmathi
- Organic and Polymer Synthesis Laboratory, Department of Chemistry, National Institute of Technology, Tiruchirappalli - 620 015, India.
| |
Collapse
|
31
|
Ponce-Lopez T. Peripheral Inflammation and Insulin Resistance: Their Impact on Blood-Brain Barrier Integrity and Glia Activation in Alzheimer's Disease. Int J Mol Sci 2025; 26:4209. [PMID: 40362446 PMCID: PMC12072112 DOI: 10.3390/ijms26094209] [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: 03/09/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory impairment, and synaptic dysfunction. The accumulation of amyloid beta (Aβ) plaques and hyperphosphorylated tau protein leads to neuronal dysfunction, neuroinflammation, and glial cell activation. Emerging evidence suggests that peripheral insulin resistance and chronic inflammation, often associated with type 2 diabetes (T2D) and obesity, promote increased proinflammatory cytokines, oxidative stress, and immune cell infiltration. These conditions further damage the blood-brain barrier (BBB) integrity and promote neurotoxicity and chronic glial cell activation. This induces neuroinflammation and impaired neuronal insulin signaling, reducing glucose metabolism and exacerbating Aβ accumulation and tau hyperphosphorylation. Indeed, epidemiological studies have linked T2D and obesity with an increased risk of developing AD, reinforcing the connection between metabolic disorders and neurodegeneration. This review explores the relationships between peripheral insulin resistance, inflammation, and BBB dysfunction, highlighting their role in glial activation and the exacerbation of AD pathology.
Collapse
Affiliation(s)
- Teresa Ponce-Lopez
- Centro de Investigación en Ciencias de la Salud (CICSA), Facultad de Ciencias de la Salud, Universidad Anáhuac México Campus Norte, Huixquilucan 52786, Mexico
| |
Collapse
|
32
|
Barbouti A, Varvarousis DN, Kanavaros P. The Role of Oxidative Stress-Induced Senescence in the Pathogenesis of Preeclampsia. Antioxidants (Basel) 2025; 14:529. [PMID: 40427411 DOI: 10.3390/antiox14050529] [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: 03/07/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Preeclampsia is a hypertension condition of human pregnancy that poses a significant risk to pregnant women and their fetus. It complicates about 2-8% of human pregnancies worldwide and displays multifactorial pathogenesis, including increased placental oxidative stress because of disturbed utero-placental blood flow. Recent evidence suggests that increased oxidative stress promotes acceleration of the placental senescence which is implicated in the pathogenesis of preeclampsia. This review focuses on the mechanisms that lead to oxidative stress in preeclamptic patients and examines the role of oxidative stress-induced placental senescence in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios N Varvarousis
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
33
|
Halimeh H. Red light induced seed germination and seedling growth by modulating antioxidant defense system, Rubisco, and NADPH oxidase activities in Capsicum frutescens. BMC PLANT BIOLOGY 2025; 25:519. [PMID: 40275137 PMCID: PMC12020111 DOI: 10.1186/s12870-025-06540-8] [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: 12/28/2024] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
In this study, the impact of light-emitting diodes (LEDs) in different spectrums was investigated on the seed germination and post-germinative performance of Capsicum frutescens seedlings. The seeds were exposed to different LED lights (full spectrum, white, red, blue, and red-blue) for 0, 1, 2, and 4 h (h). The seeds were placed for a week in darkness to investigate germination, and then the growth mechanisms were studied in four-week-old seedlings. Results indicated that germination percentage was promoted markedly under 2 h red and full lights and also in 1 h blue, which was accompanied by the regulation of H2O2 level and NADPH oxidase (NOX) activity. Sprout growth and height were more heightened under 2 h red light, but their contents decreased considerably under blue light with a rising incubation time. Red light induced more biomass yield, chlorophyll (Chl) pigments, Chl a/b ratio and florescence in four-week-old seedlings. Blue light also increased Chl pigments, but decreased biomass yield by enhancing malondialdehyde (MDA) level. Increased growth in seedlings treated to red light was associated with upregulating Rubisco gene expressions (rbcL and rbcS) and its activity. Red and red-blue lights promoted the activity of superoxide dismutase, glutathione reductase, and ascorbate peroxidase enzymes to increase ascorbic acid (ASA) production in the ascorbate-glutathione cycle. Total phenolic (0.22 mg DAG g- 1 DW), ASA (89.58 mg 100 g- 1 FW) and capsaicinoids (2.73 mg g- 1 DW) contents were heightened under red light, while carotenoid (11.78 µg g- 1 FW) content was more accumulated under blue light. The findings of this study suggest red light modulates NOX activity and H2O2 level for inducing seed germination and seedling quality in C. frutescens, which can create important implications for the production of antioxidant metabolites and increase the cultivation area of this plant.
Collapse
Affiliation(s)
- Hassanpour Halimeh
- Aerospace Research Institute, Ministry of Science Research and Technology, Tehran, 14665‑834, Iran.
| |
Collapse
|
34
|
Wang F, Zhang G, Zhai Q. Role and mechanism of molecular hydrogen in the treatment of Parkinson's diseases. Front Neurosci 2025; 19:1576773. [PMID: 40336538 PMCID: PMC12055789 DOI: 10.3389/fnins.2025.1576773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/08/2025] [Indexed: 05/09/2025] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disorder characterized by a pathology that includes the aggregation of alpha-synuclein (α-syn), oxidative stress, and neuroinflammation. While existing treatments can alleviate motor symptoms, they have limited efficacy in slowing disease progression and improving non-motor symptoms. In recent years, molecular hydrogen has been recognized for its potential neuroprotective effects, attributed to its selective antioxidant and anti-inflammatory properties. While preclinical studies demonstrate promising results, clinical trials conducted thus far have yielded mixed outcomes, with some trials reporting limited or no therapeutic benefit. This review systematically analyzes the mechanisms of action of molecular hydrogen in PD and related neurodegenerative disorders, emphasizing its antioxidant, anti-inflammatory, and anti-apoptotic properties. By evaluating evidence from both preclinical and clinical studies, this paper explores the potential of molecular hydrogen to attenuate oxidative stress, modulate inflammatory responses, and inhibit apoptosis in neuronal cells, while also identifying key gaps in current research. As a novel neuroprotective agent, molecular hydrogen holds potential in PD and other neurodegenerative diseases, but further well-designed clinical trials are needed to validate its efficacy. Future studies should focus on elucidating the mechanisms through which hydrogen exerts its neuroprotective effects, particularly concerning α-syn aggregation and its clearance pathways, as well as Nrf2-mediated immunomodulation. Furthermore, large-scale, multicenter clinical trials are necessary to establish efficacy benchmarks and personalized delivery protocols.
Collapse
Affiliation(s)
- Fengjiao Wang
- School of Public Health, Shandong Second Medical University, Weifang, China
| | - Guangjie Zhang
- Department of Medical Technology and Nursing, Laiwu Vocational and Technical College, Jinan, China
| | - Qingfeng Zhai
- School of Public Health, Shandong Second Medical University, Weifang, China
| |
Collapse
|
35
|
Paál Á, Dora D, Takács Á, Rivard C, Pickard SL, Hirsch FR, Roskó B, Kiraly P, Ferdinandy P, Varga ZV, Lohinai Z, Görbe A. Roles of Annexin A1 Expression in Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1407. [PMID: 40361334 PMCID: PMC12070913 DOI: 10.3390/cancers17091407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Small cell lung cancer (SCLC) is one of the malignancies with the worst prognosis, and there have been no major breakthroughs in its treatment for a long time. The majority of patients are diagnosed at the extensive stage, where the only option is chemotherapy, and even the addition of immune checkpoint inhibitors results in only modest benefits. The characterization of the molecular mechanisms behind therapy resistance has relevance in finding novel therapeutic approaches. Previous studies showed the possibility of annexin A1's (ANXA1) involvement in the immunosuppressive tumor microenvironment in SCLC, and there are studies showing the direct effects of ANXA1 modulation on cancer cell aggressiveness. METHODS We aimed to characterize the roles of ANXA1 expression using publicly available transcriptomic data, the RNA-seq-based predictive algorithms EPIC and ESTIMATE, and immunohistochemistry on patient samples. For the in vitro studies, we silenced ANXA1 expression with short hairpin RNA in three SCLC cell lines, measured the growth rate with the trypan blue exclusion assay, assessed the chemosensitivity to cisplatin and etoposide with the Presto BlueTM viability assay, and performed Western blots to assess changes in the levels of metabolic and mesenchymal markers and transcriptional drivers. RESULTS ANXA1-high tumors are associated with significantly increased immune infiltrates, stromality, and tumor-associated macrophages (TAMs). The ANXA1 protein is expressed on tumor cells and TAMs at the tissue level. ANXA1 silencing in H841 cells did not affect the growth rate; in SW1271 cells, shANXA1 cells grew significantly slower than shCTRL cells. Meanwhile, in H1048 cells, proliferation was significantly faster. Despite the different growth rates of the tested cell lines, ANXA1 silencing decreased the chemosensitivity to both cisplatin and etoposide in all three cell lines. Gene expression changes in mesenchymal markers, metabolic markers, dominant transcriptional drivers, and immune-relevant molecules were also characterized. CONCLUSIONS This is the first comprehensive characterization of ANXA1 in SCLC to reveal its role in the tumor's cell biology and the TME, aiming to boost further research in the field.
Collapse
Affiliation(s)
- Ágnes Paál
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Ákos Takács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
| | - Christopher Rivard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Shivaun Lueke Pickard
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
| | - Fred R. Hirsch
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (C.R.); (S.L.P.); (F.R.H.)
- Tisch Cancer Institute, Center for Thoracic Oncology, Mount Sinai Health System, New York, NY 10029, USA
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, 1085 Budapest, Hungary; (D.D.); (B.R.)
| | - Peter Kiraly
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, 1085 Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, 1089 Budapest, Hungary
| | - Zoltan Lohinai
- Translational Medicine Institute, Semmelweis University, 1085 Budapest, Hungary;
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (Á.P.); (Á.T.); (P.F.); (Z.V.V.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 1085 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
36
|
Johnson HE, Umutesi HG, Heo J. The small GTPase Rap1A expedites the NOX2 oxidative burst by facilitating Rac and NOX2 autoactivations. FEBS J 2025. [PMID: 40259664 DOI: 10.1111/febs.70107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/21/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025]
Abstract
Rac and Rap1A are small GTPases with the redox-sensitive GX4GK(S/T)C/ECS and NKCD motif. Of the known NADPH oxidase (NOX) isoforms, NOX1 and NOX2 function with the redox-sensitive Rac. Both exhibit an oxidative burst in which superoxide production is initially lagged but then accelerated. This burst is a reflection of NOX1 and NOX2 autoactivations occurring alongside the redox-dependent Rac autoactivation. NOX2 also contains the redox-sensitive Rap1A. However, its role in NOX2 function was unknown. In this study, we show that Rap1A is also autoactivated by its redox response, which is coupled to Rac and NOX2 autoactivations. This coupling is found to be mediated through the Rap1A-dependent recruitment of the Rac GEF P-REX1 to the NOX2 system. We further show that the initiation threshold and propagation rate of Rap1A autoactivation are lower and slower, respectively, than those of Rac and NOX2. The low-threshold Rap1A autoactivation recruits P-REX1 to the NOX2 system, resulting in the production of active Rac, thereby aiding the high-threshold initiation and propagation of Rac and NOX2 autoactivations. This results in the rapid completion of the NOX2 oxidative burst, which is specific to NOX2 because NOX1 lacks Rap1A. The redox response differences between the Rap1A NKCD motif and the Rac GX4GK(S/T)C/ECS motif appear to be the basis for the feature differences between Rap1A autoactivation and those of Rac and NOX2 autoactivations. The GX4GK(S/T)C/ECS and NKCD motifs are found in many redox-sensitive Rho/Rab and Ras family GTPases, respectively. Findings here shed light on previously unknown redox-dependent functional distinctions between these small GTPases.
Collapse
Affiliation(s)
- Hope Elizabeth Johnson
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Hope Gloria Umutesi
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| | - Jongyun Heo
- Department of Chemistry and Biochemistry, The University of Texas at Arlington, Arlington, TX, USA
| |
Collapse
|
37
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [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: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
38
|
Chu T, Xiao Z, Xun C, Yang C, Lu M, Wang Y, Chen H, Chen P. Peptidomic profiling of mesenchymal stem cell-derived extracellular vesicles and anti-inflammatory activity of degraded peptides. Int Immunopharmacol 2025; 152:114452. [PMID: 40096816 DOI: 10.1016/j.intimp.2025.114452] [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: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
Mesenchymal stem cell derived extracellular vesicles (MSC-EVs) are key paracrine mediators involved in various autoimmune diseases. While current research on EVs predominantly focuses on their protein and nucleic acid components, small peptides received less attention. In this study, we found IFN-γ-treated MSC-EVs, as engineered EVs, exhibit better anti-inflammatory effects both in vitro and in vivo. Through LC-MS/MS and bioinformatics analysis, we identified four peptides-C3-1, C3-2, B2M-1, and IFIT3-1-that are highly expressed in IFN-γ-treated MSCs-EVs. These peptides significantly mitigate the proliferation inhibition of HUVEC cells induced by H₂O₂ and enhance their migratory capacity. Furthermore, they downregulate the expression of inflammatory cytokines TNF-α and IL-6 in H₂O₂-induced oxidative stress models of HUVEC and LPS-induced inflammatory models of RAW264.7 macrophages. The peptides also upregulate p-AKT and HIF-1α, with C3-1 demonstrating superior anti-inflammatory efficacy in both cell models. Consistent with the in vitro findings, in vivo experiments revealed that C3-1 reduces LPS-induced inflammatory cell infiltration in liver tissue and restores hepatocyte structural integrity, as evidenced by HE-stained liver sections. Western blot analysis further confirmed that C3-1 upregulates p-AKT expression and suppresses inflammatory cytokines. Collectively, these findings suggest that C3-1 exerts its anti-inflammatory effects via activation of the AKT signaling pathway and regulation of TNF-α and IL-6. This study not only highlights the anti-inflammatory potential of IFN-γ-treated MSC-derived EVs but also identifies C3-1 as a promising candidate for anti-inflammatory drug development. Notably, this is the first study to identify degraded peptides within EVs, providing a foundation for future research in this area.
Collapse
Affiliation(s)
- Tianqi Chu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China
| | - Zixuan Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Chengfeng Xun
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Hunan Academy of Forestry, Changsha 410081, China
| | - Chunyan Yang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Mengqi Lu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuqiu Wang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Haiyan Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; East China Institute of Digital Medical Engineering, Shangrao 334000, China.
| | - Ping Chen
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha 410081, China; Peptide and Small Molecule Drug R&D Platform, Furong Laboratory, Hunan Normal University, Changsha 410081, Hunan, China; Institute of Interdisciplinary Studies, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
39
|
Aryal B, Kwakye J, Ariyo OW, Ghareeb AFA, Milfort MC, Fuller AL, Khatiwada S, Rekaya R, Aggrey SE. Major Oxidative and Antioxidant Mechanisms During Heat Stress-Induced Oxidative Stress in Chickens. Antioxidants (Basel) 2025; 14:471. [PMID: 40298812 PMCID: PMC12023971 DOI: 10.3390/antiox14040471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Heat stress (HS) is one of the most important stressors in chickens, and its adverse effects are primarily caused by disturbing the redox homeostasis. An increase in electron leakage from the mitochondrial electron transport chain is the major source of free radical production under HS, which triggers other enzymatic systems to generate more radicals. As a defense mechanism, cells have enzymatic and non-enzymatic antioxidant systems that work cooperatively against free radicals. The generation of free radicals, particularly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), under HS condition outweighs the cellular antioxidant capacity, resulting in oxidative damage to macromolecules, including lipids, carbohydrates, proteins, and DNA. Understanding these detrimental oxidative processes and protective defense mechanisms is important in developing mitigation strategies against HS. This review summarizes the current understanding of major oxidative and antioxidant systems and their molecular mechanisms in generating or neutralizing the ROS/RNS. Importantly, this review explores the potential mechanisms that lead to the development of oxidative stress in heat-stressed chickens, highlighting their unique behavioral and physiological responses against thermal stress. Further, we summarize the major findings associated with these oxidative and antioxidant mechanisms in chickens.
Collapse
Affiliation(s)
- Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Oluwatomide W. Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Ahmed F. A. Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Boehringer Ingelheim Animal Health (BIAH), Gainesville, GA 30501, USA
| | - Marie C. Milfort
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Alberta L. Fuller
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Saroj Khatiwada
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, The University of Georgia, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| |
Collapse
|
40
|
Lacerda-Abreu MA, Meyer-Fernandes JR. Hyperphosphataemia and NADPH Oxidase Regulation in Pathophysiological Processes: Implications for Oxidative Stress and Disease Progression. Antioxidants (Basel) 2025; 14:461. [PMID: 40298783 PMCID: PMC12024410 DOI: 10.3390/antiox14040461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Hyperphosphataemia is a key contributor to oxidative stress (OS) and cellular dysfunction across various pathological conditions. While numerous studies have associated phosphate overload with redox imbalances, the role of NADPH oxidase (NOX) in this process has received limited attention. NOX enzymes are major enzymatic sources of reactive oxygen species (ROS), and their activation has been implicated in the progression of chronic kidney disease, vascular calcification, metabolic disorders, and cancer development. Under hyperphosphataemic conditions, excessive ROS production exacerbates endothelial dysfunction, promotes vascular smooth muscle cell transdifferentiation, induces chronic inflammation, and facilitates tumour progression. Despite increasing evidence linking phosphate metabolism to NOX activation, the underlying molecular mechanisms remain poorly characterised. This review critically examines the relationship between hyperphosphataemia and NADPH oxidase-mediated OS and explores its impact on disease pathophysiology. By providing an integrated analysis of the current findings, this work aims to highlight the pathological consequences of phosphate-induced OS and identify potential therapeutic strategies to mitigate its effects.
Collapse
Affiliation(s)
- Marco Antonio Lacerda-Abreu
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil
| | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, RJ, Brazil
| |
Collapse
|
41
|
Sebghatollahi Z, Yogesh R, Mahato N, Kumar V, Mohanta YK, Baek KH, Mishra AK. Signaling Pathways in Oxidative Stress-Induced Neurodegenerative Diseases: A Review of Phytochemical Therapeutic Interventions. Antioxidants (Basel) 2025; 14:457. [PMID: 40298834 PMCID: PMC12024045 DOI: 10.3390/antiox14040457] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Oxidative stress, a pivotal driver of neurodegenerative diseases, results from an imbalance between the generation of reactive oxygen species (ROS) and cellular antioxidant defenses. This review provides a comprehensive analysis of key oxidative stress sources, focusing on NADPH oxidase (NOX) hyperactivity and mitochondrial Uncoupling Protein (UCP) downregulation. Critically, we examine the therapeutic potential of phytochemicals in mitigating NOX-mediated ROS generation through direct enzyme inhibition, including impacts on NOX subunit assembly and gene expression. Furthermore, we explore the ability of phytochemicals to bolster cellular antioxidant defenses by activating the Kelch-like ECH-associated protein 1 (KEAP1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway, elucidating the upregulation of antioxidant genes, such as GPx, SOD, CAT, and HO-1. This review expands beyond confined overviews; emphasizes specific molecular interactions between phytochemicals and target proteins, including NOX isoforms; and provides an in-depth analysis of the specific antioxidant genes upregulated via Nrf2. This approach aims to pave the way for targeted and translatable therapeutic strategies in neurodegenerative diseases. Ultimately, this review illuminates the intricate molecular dynamics of oxidative stress in neurodegenerative diseases; underscores the potential of phytochemicals to restore redox homeostasis and reverse pathological conditions through precise modulation of key signaling pathways.
Collapse
Affiliation(s)
- Zahra Sebghatollahi
- Department of Plant Breeding and Biotechnology, Faculty of Agricultural Sciences and Food Industries, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran;
| | - Ruchika Yogesh
- MaTestLab Inc., 2093 Philadelphia Pike, Claymont, DE 19703, USA;
| | - Neelima Mahato
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Republic of Korea;
| | - Vijay Kumar
- Department of Orthopaedic Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Yugal Kishore Mohanta
- Nano-Biotechnology and Translational Knowledge Laboratory, Department of Applied Biology, School of Biological Sciences, University of Science and Technology Meghalaya, Techno City, 9th Mile, Baridua 793101, Meghalaya, India;
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam 603103, Tamil Nadu, India
| | - Kwang-Hyun Baek
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Republic of Korea
| | - Awdhesh Kumar Mishra
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Gyeongsangbuk-do, Republic of Korea
| |
Collapse
|
42
|
Michalak KP, Michalak AZ. Understanding chronic inflammation: couplings between cytokines, ROS, NO, Ca i 2+, HIF-1α, Nrf2 and autophagy. Front Immunol 2025; 16:1558263. [PMID: 40264757 PMCID: PMC12012389 DOI: 10.3389/fimmu.2025.1558263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/14/2025] [Indexed: 04/24/2025] Open
Abstract
Chronic inflammation is an important component of many diseases, including autoimmune diseases, intracellular infections, dysbiosis and degenerative diseases. An important element of this state is the mainly positive feedback between inflammatory cytokines, reactive oxygen species (ROS), nitric oxide (NO), increased intracellular calcium, hypoxia-inducible factor 1-alpha (HIF-1α) stabilisation and mitochondrial oxidative stress, which, under normal conditions, enhance the response against pathogens. Autophagy and the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant response are mainly negatively coupled with the above-mentioned elements to maintain the defence response at a level appropriate to the severity of the infection. The current review is the first attempt to build a multidimensional model of cellular self-regulation of chronic inflammation. It describes the feedbacks involved in the inflammatory response and explains the possible pathways by which inflammation becomes chronic. The multiplicity of positive feedbacks suggests that symptomatic treatment of chronic inflammation should focus on inhibiting multiple positive feedbacks to effectively suppress all dysregulated elements including inflammation, oxidative stress, calcium stress, mito-stress and other metabolic disturbances.
Collapse
Affiliation(s)
- Krzysztof Piotr Michalak
- Laboratory of Vision Science and Optometry, Physics and Astronomy Faculty, Adam Mickiewicz University in Poznań, Poznań, Poland
| | | |
Collapse
|
43
|
Morii A, Matsuo I, Suita K, Ohnuki Y, Ishikawa M, Ito A, Miyamoto G, Abe M, Mitsubayashi T, Mototani Y, Nariyama M, Matsubara R, Hayakawa Y, Amitani Y, Gomi K, Nagano T, Okumura S. Allopurinol attenuates development of Porphyromonas gingivalis LPS-induced cardiomyopathy in mice. PLoS One 2025; 20:e0318008. [PMID: 40179080 PMCID: PMC11967946 DOI: 10.1371/journal.pone.0318008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/08/2025] [Indexed: 04/05/2025] Open
Abstract
Oxidative stress is involved in the progression of periodontitis, independently of confounding factors such as smoking, and numerous studies suggest that periodontitis is associated with increased risk of cardiovascular disease. In this study, therefore, we examined the effects of the xanthine oxidase inhibitor allopurinol on cardiac dysfunction in mice treated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS) at a dose (0.8 mg/kg/day) equivalent to the circulating level in patients with periodontal disease. Mice were divided into four groups: 1) control, 2) PG-LPS, 3) allopurinol, and 4) PG-LPS + allopurinol. After1 week, we evaluated cardiac function by echocardiography. The left ventricular ejection fraction was significantly decreased in PG-LPS-treated mice compared to the control (from 68 ± 1.3 to 60 ± 2.7%), while allopurinol ameliorated the dysfunction (67 ± 1.1%). The area of cardiac fibrosis was significantly increased (approximately 3.6-fold) and the number of apoptotic myocytes was significantly increased (approximately 7.7-fold) in the heart of the PG-LPS-treated group versus the control, and these changes were suppressed by allopurinol. The impairment of cardiac function in PG-LPS-treated mice was associated with increased production of reactive oxygen species by xanthine oxidase and NADPH oxidase 4, leading to calmodulin kinase II activation with increased ryanodine receptor 2 phosphorylation. These changes were also suppressed by allopurinol. Our results suggest that oxidative stress plays an important role in the PG-LPS-promoted development of cardiac diseases, and further indicate that allopurinol ameliorates Porphyromonas gingivalis LPS-induced cardiac dysfunction.
Collapse
Affiliation(s)
- Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Oral and Maxillofacial Surgery, Ibaraki Medical Center Tokyo Medical University, Ibaraki, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Go Miyamoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Mariko Abe
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takao Mitsubayashi
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ren Matsubara
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takatoshi Nagano
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
44
|
Kračun D, Görlach A, Snedeker JG, Buschmann J. Reactive oxygen species in tendon injury and repair. Redox Biol 2025; 81:103568. [PMID: 40023978 PMCID: PMC11915165 DOI: 10.1016/j.redox.2025.103568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Reactive oxygen species (ROS) are chemical moieties that in physiological concentrations serve as fast-acting signaling molecules important for cellular homeostasis. However, their excess either due to overproduction or inability of the antioxidant system to inactivate them results in oxidative stress, contributing to cellular dysfunction and tissue damage. In tendons, which are hypovascular, hypocellular, and composed predominantly of extracellular matrix (ECM), particularly collagen I, ROS likely play a dual role: regulating cellular processes such as inflammation, proliferation, and ECM remodeling under physiological conditions, while contributing to tendinopathy and impaired healing when dysregulated. This review explores the sources of ROS in tendons, including NADPH oxidases and mitochondria, and their role in key processes such as tissue adaptation to mechanical load and injury repair, also in systemic conditions such as diabetes. In addition, we integrate the emerging perspective that calcium signaling-mediated by mechanically activated ion channels-plays a central role in tendon mechanotransduction under daily mechanical loads. We propose that mechanical overuse (overload) may lead to hyperactivation of calcium channels, resulting in chronically elevated intracellular calcium levels that amplify ROS production and oxidative stress. Although direct evidence linking calcium channel hyperactivity, intracellular calcium dysregulation, and ROS generation under overload conditions is currently circumstantial, this review aims to highlight these connections and identify them as critical avenues for future research. By framing ROS within the context of both adaptive and maladaptive responses to mechanical load, this review provides a comprehensive synthesis of redox biology in tendon injury and repair, paving the way for future work, including development of therapeutic strategies targeting ROS and calcium signaling to enhance tendon recovery and resilience.
Collapse
Affiliation(s)
- Damir Kračun
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland; University Clinic Balgrist, Orthopaedic Biomechanics, Forchstrasse 340, 8008, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Gloriastrasse 37/39, 8092, Zurich, Switzerland.
| | - Agnes Görlach
- Experimental and Molecular Paediatric Cardiology, German Heart Centre Munich, TUM University Hospital, Technical University of Munich, Munich, 80636, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Jess G Snedeker
- University Clinic Balgrist, Orthopaedic Biomechanics, Forchstrasse 340, 8008, Zurich, Switzerland; Institute for Biomechanics, ETH Zurich, Gloriastrasse 37/39, 8092, Zurich, Switzerland
| | - Johanna Buschmann
- Division of Plastic Surgery and Hand Surgery, University Hospital Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland.
| |
Collapse
|
45
|
Chen W, Huang Y, Li W, Fan G, Tang Y, Zhao W, Chen K, Chen Z, Zhou K, Li Z, Zhang H. The potential of pomegranate peel supplementation in Yellow-feathered broilers: effects on growth performance, serum biochemistry, antioxidant capacity, intestinal health, intestinal microbiota, and duodenal mucosal metabolites. Poult Sci 2025; 104:104983. [PMID: 40058007 PMCID: PMC11930591 DOI: 10.1016/j.psj.2025.104983] [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: 01/11/2025] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to investigate the effects of dietary supplementation with pomegranate peel powder (PP) on the growth performance, serum biochemistry, antioxidant capacity, intestinal microbiota, and duodenal mucosal metabolites of yellow-feathered broilers. A total of 360 yellow-feathered broilers were randomly divided into three groups, with their diets supplemented with different levels of PP (0, 1, and 4 g/kg) for 42 days. Dietary supplementation with PP significantly increased the average body weight and average daily gain of yellow-feathered broilers during the periods of 1-21 and 22-42 days, while reducing the feed conversion ratio (p < 0.05). It also decreased the serum levels of aspartate aminotransferase, alanine aminotransferase, creatinine, and uric acid, increased the activities of glutathione peroxidase and superoxide dismutase, and reduced malondialdehyde content in the serum, liver, and intestinal mucosa (p < 0.05). Furthermore, PP supplementation promoted the mRNA expression of farnesoid X receptor, peroxisome proliferator-activated receptor alpha, fatty acid-binding protein 4, epidermal growth factor/epidermal growth factor receptor, and B-cell lymphoma 2, while decreasing the mRNA expression of caspase-1 and interleukin-1 beta (p < 0.05). Regarding mucosal metabolites, PP supplementation increased the contents of polyunsaturated fatty acids (cis-11-eicosenoic acid, cis-13,16-docosadienoic acid, and cis-11,14-eicosadienoic acid), prostaglandin E2/G2, and secondary bile acids (apocholic, hyodeoxycholic, 7-ketodeoxycholic, and omega-muricholic acids) in the mucosa (p < 0.05). In terms of cecal microbiota, PP supplementation increased the β-diversity index (p < 0.05), elevated the relative abundances of Bacteroidota, Alistipes, Bacilli, and Actinobacteriota, and reduced the relative abundances of Clostridia and Gammaproteobacteria (p < 0.05). In conclusion, dietary supplementation of PP can improve intestinal health and growth performance of yellow-feathered broilers by regulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Wang Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Yurong Huang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Wenlong Li
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Gao Fan
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China.
| | - Yanfang Tang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Weiru Zhao
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Kexin Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Zifan Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Keyue Zhou
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China.
| | - Zhaoyao Li
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China; College of Veterinary Medicine, South China Agricultural University, No. 483, Wushan Road, Tianhe District, Guangzhou, Guangdong, 510642, China.
| | - Huihua Zhang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| |
Collapse
|
46
|
Zhu J, Jiang C, Wang F, Tao MY, Wang HX, Sun Y, Hui HX. NOX4 Suppresses Ferroptosis Through Regulation of the Pentose Phosphate Pathway in Colorectal Cancer. Curr Med Sci 2025; 45:264-279. [PMID: 40029499 DOI: 10.1007/s11596-025-00013-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/23/2024] [Accepted: 11/28/2024] [Indexed: 03/05/2025]
Abstract
OBJECTIVE Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) are known as major sources of reactive oxygen species (ROS), yet their role in regulating cellular antioxidative metabolism and ferroptosis is unclear. This study assessed the expression and clinical relevance of NOXs across pan-cancer and investigated the role of NOX4 in colorectal cancer progression METHODS: We analyzed transcriptomic and survival data from The Cancer Genome Atlas (TCGA) for NOXs across 22 types of solid tumors. A CRISPR library targeting NOXs was developed for potential therapeutic target screening in colorectal cancer cells (CRCs). Techniques such as CRISPR-knockout cell lines, 1,2-13C-glucose tracing, PI staining, BrdU assays, and coimmunoprecipitation were employed to elucidate the function of NOX4 in CRCs. RESULTS NOX4 emerged as a key therapeutic target for colorectal cancer from TCGA data. CRISPR screening highlighted its essential role in CRC survival, with functional experiments confirming that NOX4 upregulation promotes cell survival and proliferation. The interaction of NOX4 with glucose‑6‑phosphate dehydrogenase (G6PD) was found to enhance the pentose phosphate pathway (PPP), facilitating ROS clearance and protecting CRCs against ferroptosis. CONCLUSIONS This study identified NOX4 as a novel ferroptosis suppressor and a therapeutic target for the treatment of colorectal cancer. The findings suggest that a coupling between NADPH oxidase enzyme NOX4 and the PPP regulates ferroptosis and reveal an accompanying metabolic vulnerability for therapeutic targeting in colorectal cancer.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Chao Jiang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Fan Wang
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Ming-Yue Tao
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hai-Xiao Wang
- Department of General Surgery, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Yuan Sun
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China
| | - Hong-Xia Hui
- Department of Medical Oncology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, 223300, China.
| |
Collapse
|
47
|
de la Rosa F, Piloni NE, De Troch M, Malanga G, Hernando M. Increasing temperature counteracts the negative effects of ultraviolet radiation on Microcystis aeruginosa under future climate scenarios in relation to physiological processes. Comp Biochem Physiol C Toxicol Pharmacol 2025; 290:110124. [PMID: 39793845 DOI: 10.1016/j.cbpc.2025.110124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/23/2024] [Accepted: 01/04/2025] [Indexed: 01/13/2025]
Abstract
Heat waves, are a major concern related to climate change, and are projected to increase in frequency and severity. This temperature rise causes thermal stratification, exposing surface-dwelling organisms to higher levels of ultraviolet radiation (UVR). This study aims to understand how the toxic bloom-forming cyanobacterium Microcystis aeruginosa adapts to changing climatic conditions. The effects of increased temperature and UVR were evaluated in terms of cell abundance, reactive oxygen and nitrogen species (ROS/RNS), the antioxidant activity of catalase (CAT), superoxide dismutase (SOD), glutathione S transferase (GST), fatty acid (FA) content, and lipid damage. Negative UVR effects on biomass, lipid damage, and polyunsaturated fatty acids (PUFAs) were more pronounced at 26 °C compared to 29 °C. However, antioxidant responses were higher at 29 °C. The relative abundance of ω6 FAs was less affected by UVA, while ω3 FAs were highly sensitive at 29 °C but unsaturated fatty acids (UFA) did not experience peroxidation. The differential response in FA to high temperature and UVR results in differences in lipid damage and antioxidants. Changes in membrane FA may suggest an adaptation strategy at high UVR conditions. The exposure to environmental changes can alter membrane fluidity, affecting cell physiology. Thus, to survive UVR exposure, M. aeruginosa maintains a balance between damage and stress adaptation, increasing the protection of selected PUFAs at high temperatures, allowing them to effectively cope with the harmful effects of elevated temperature and UVR.
Collapse
Affiliation(s)
- F de la Rosa
- Instituto de Ciencias Básicas y Experimentales, Universidad de Morón, Morón, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, CABA, Argentina
| | - N E Piloni
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CABA, Argentina; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina - Universidad de Buenos Aires, CABA, Argentina
| | - M De Troch
- Faculty of Sciences, Marine Biology, Ghent University, Krijgslaan 281-S8, Ghent, Belgium
| | - G Malanga
- Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, CABA, Argentina; Instituto de Bioquímica y Medicina Molecular, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina - Universidad de Buenos Aires, CABA, Argentina
| | - M Hernando
- Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, CABA, Argentina; Departamento de Radiobiología, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica, General San Martín, Argentina; Red de Investigación de estresores Marinos-Costeros en América Latina y el Caribe, Mar del Plata, Argentina.
| |
Collapse
|
48
|
Münzel T, Kuntic M, Lelieveld J, Aschner M, Nieuwenhuijsen MJ, Landrigan PJ, Daiber A. The links between soil and water pollution and cardiovascular disease. Atherosclerosis 2025; 403:119160. [PMID: 40074641 DOI: 10.1016/j.atherosclerosis.2025.119160] [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: 12/22/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025]
Abstract
Soil and water pollution represent significant threats to global health, ecosystems, and biodiversity. Healthy soils underpin terrestrial ecosystems, supporting food production, biodiversity, water retention, and carbon sequestration. However, soil degradation jeopardizes the health of 3.2 billion people, while over 2 billion live in water-stressed regions. Pollution of soil, air, and water is a leading environmental cause of disease, contributing to over 9 million premature deaths annually. Soil contamination stems from heavy metals, synthetic chemicals, pesticides, and plastics, driven by industrial activity, agriculture, and waste mismanagement. These pollutants induce oxidative stress, inflammation, and hormonal disruption, significantly increasing risks for non-communicable diseases (NCDs) such as cardiovascular disease (CVD). Emerging contaminants like micro- and nanoplastics amplify health risks through cellular damage, oxidative stress, and cardiovascular dysfunction. Urbanization and climate change exacerbate soil degradation through deforestation, overfertilization, and pollution, further threatening ecosystem sustainability and human health. Mitigation efforts, such as reducing chemical exposure, adopting sustainable land-use practices, and advancing urban planning, have shown promise in lowering pollution-related health impacts. Public health initiatives, stricter pollution controls, and lifestyle interventions, including antioxidant-rich diets, can also mitigate risks. Pollution remains preventable, as demonstrated by high-income nations implementing cost-effective solutions. Policies like the European Commission's Zero-Pollution Vision aim to reduce pollution to safe levels by 2050, promoting sustainable ecosystems and public health. Addressing soil pollution is critical to combating the global burden of NCDs, particularly CVDs, and fostering a healthier environment for future generations.
Collapse
Affiliation(s)
- Thomas Münzel
- University Medical Center Mainz, Department of Cardiology at the Johannes Gutenberg University, Germany; German Cardiovascular Research Center (DZHK), Partner Site Rhine Main, Mainz, Germany.
| | - Marin Kuntic
- University Medical Center Mainz, Department of Cardiology at the Johannes Gutenberg University, Germany
| | - Jos Lelieveld
- Atmospheric Chemistry Department, Max Planck Institute for Chemistry, Mainz, Germany
| | - Michael Aschner
- Molecular Pharmacology, Albert Einstein College of Medicine, United States
| | - Mark J Nieuwenhuijsen
- Institute for Global Health (ISGlobal), Barcelona, Spain; Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Philip J Landrigan
- Global Observatory on Planetary Health, Boston College, USA; Centre Scientifique de Monaco, MC, Monaco
| | - Andreas Daiber
- University Medical Center Mainz, Department of Cardiology at the Johannes Gutenberg University, Germany; German Cardiovascular Research Center (DZHK), Partner Site Rhine Main, Mainz, Germany
| |
Collapse
|
49
|
Wang M, Wang Y, Zhang P, Gu C, Zhao X, Gong X, Yang X, Pan J, Xi Y. Neutrophil-like cell membrane-coated metal-organic frameworks for siRNA delivery targeting NOX4 to alleviate oxidative stress in acute ischemic injury. Acta Biomater 2025; 196:487-505. [PMID: 40024424 DOI: 10.1016/j.actbio.2025.02.061] [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: 11/30/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Although reperfusion is the most effective treatment for acute ischemic stroke, it often results in serious secondary ischemia/reperfusion (I/R) injury due to oxidative stress. This oxidative stress primarily results from the overproduction of reactive oxygen species (ROS) during reperfusion which, in turn, is largely induced by high expression of NADPH oxidase 4 (NOX4). Inhibiting NOX4 gene expression has therefore been proposed as a direct approach to reduce ROS production and promote angiogenesis. Recognizing both the potential of siRNA-based therapies for selective gene silencing and the critical role of neutrophil-endothelial interactions during I/R injury, here we present a unique therapeutic approach where neutrophil-like cell membrane coated porous metal-organic framework nanoparticles are loaded with siNOX4 (M-MOF-siNOX4) and designed to target damaged brain microvascular tissue. These then mitigate oxidative stress by suppressing NOX4 expression. Using an in vitro oxygen-glucose deprivation/re-oxygenation model, we demonstrate that M-MOF-siNOX4 nanoparticles specifically bind to activated endothelial cells, effectively reducing NOX4 expression, decreasing both ROS production and cell apoptosis, and restoring cell viability. Use of an in vivo mouse model of middle cerebral artery occlusion further confirmed M-MOF-siNOX4 nanoparticles to substantially alleviate brain damage and protect neurological function following ischemic stroke. Taken together, our study presents an innovative and effective siRNA-based strategy for reducing oxidative stress in ischemic stroke therapy. STATEMENT OF SIGNIFICANCE: Ischemia/reperfusion (I/R) injury, a major complication of acute ischemic stroke, is primarily driven by oxidative stress due to the excessive production of reactive oxygen species (ROS). Current treatments targeting oxidative stress and cell death often lack specificity, leading to off-target effects. This study introduces an innovative nanoparticle-based therapy using neutrophil-like cell membrane-coated metal-organic frameworks (MOFs) to deliver siNOX4, an siRNA targeting NOX4, a key ROS-producing enzyme. This approach enhances targeted delivery, reduces ROS production and cell death, and significantly improves neurological recovery in stroke models. By overcoming the limitations of existing therapies, this strategy holds strong potential for revolutionizing ischemic stroke treatment and addressing other disorders related to oxidative stress.
Collapse
Affiliation(s)
- Min Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Yunbo Wang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Pengqi Zhang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Chenjie Gu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Xianlei Zhao
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xinghan Gong
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China
| | - Xiaohang Yang
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| | - Jianwei Pan
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China.
| | - Yongmei Xi
- Center for Genetic Medicine and Department of Neurosurgery, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, China; Institute of Genetics, Zhejiang University International School of Medicine, Hangzhou, Zhejiang 310058, China; Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Yiwu, Zhejiang 322000, China; Zheiiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
50
|
Sekiguchi A, Ishikawa M, Uchiyama A, Yokoyama Y, Ogino S, Torii R, Akai R, Iwawaki T, Motegi SI. Rapid thawing enhances tissue destruction in a mouse model of cutaneous cryoablation: Insights into oxidative stress and neutrophil activation. J Dermatol Sci 2025; 118:9-17. [PMID: 40000304 DOI: 10.1016/j.jdermsci.2025.02.003] [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: 10/16/2024] [Revised: 01/09/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND Cryoablation is an integral therapeutic approach in dermatology for eliminating viral warts and benign tumors by damaging tissue through freeze-thaw cycles. Rapid thawing of the frozen area by warming it with fingertips during cryoablation is a common technique in Japan; however, its efficacy has not been elucidated. OBJECTIVE This study aimed to evaluate the effect of rapid thawing on cryoablation-treated skin and clarify the underlying mechanisms using cryoablation model mice. METHODS Cryoablation was performed on the dorsal skin of mice using a liquid nitrogen-soaked cotton swab, followed by rapid thawing by warming with fingertips or natural thawing without treatment. The effects on skin ulcers, immune cell infiltration, and oxidative stress were assessed. RESULTS Rapid thawing enlarged cryoablation-induced skin ulcers. The numbers of cryoablation-induced CD3+ T cells, neutrophils, neutrophil extracellular traps (NETs), and TUNEL+ cells increased with rapid thawing. Visualization of oxidative stress in OKD48 transgenic mice showed that oxidative stress signals in the cryoablation-treated area were enhanced with rapid thawing. Real-time PCR analysis of mouse skin demonstrated that cryoblation-induced levels of NOX2 and HO-1 were significantly elevated with rapid thawing. In mouse melanoma tumors treated with cryoablation, rapid thawing significantly inhibited tumor growth and increased the infiltration of neutrophils, NETs, and TUNEL+ cells compared to the group without rapid thawing. CONCLUSION Rapid thawing during cryoablation enhances neutrophil and lymphocyte infiltration, increases oxidative stress, and induces cell death, leading to greater tissue destruction in mice. Dermatologists should consider employing rapid thawing techniques during cryoablation when higher therapeutic intensities are required.
Collapse
Affiliation(s)
- Akiko Sekiguchi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Mai Ishikawa
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Akihiko Uchiyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoko Yokoyama
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Sachiko Ogino
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Torii
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Ryoko Akai
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kanazawa, Japan
| | - Sei-Ichiro Motegi
- Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Japan.
| |
Collapse
|