1
|
Yang Y, Ma K, Li S, Xiong T. Multifaceted role of nitric oxide in vascular dementia. Med Gas Res 2025; 15:496-506. [PMID: 40300885 DOI: 10.4103/mgr.medgasres-d-24-00158] [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: 12/30/2024] [Accepted: 02/24/2025] [Indexed: 05/01/2025] Open
Abstract
Vascular dementia is a highly heterogeneous neurodegenerative disorder induced by a variety of factors. Currently, there are no definitive treatments for the cognitive dysfunction associated with vascular dementia. However, early detection and preventive measures have proven effective in reducing the risk of onset and improving patient prognosis. Nitric oxide plays an integral role in various physiological and pathological processes within the central nervous system. In recent years, nitric oxide has been implicated in the regulation of synaptic plasticity and has emerged as a crucial factor in the pathophysiology of vascular dementia. At different stages of vascular dementia, nitric oxide levels and bioavailability undergo dynamic alterations, with a marked reduction in the later stages, which significantly contributes to the cognitive deficits associated with the disease. This review provides a comprehensive review of the emerging role of nitric oxide in the physiological and pathological processes underlying vascular dementia, focusing on its effects on synaptic dysfunction, neuroinflammation, oxidative stress, and blood‒brain barrier integrity. Furthermore, we suggest that targeting the nitric oxide soluble guanylate cyclase-cyclic guanosine monophosphate pathway through specific therapeutic strategies may offer a novel approach for treating vascular dementia, potentially improving both cognitive function and patient prognosis. The review contributes to a better understanding of the multifaceted role of nitric oxide in vascular dementia and to offering insights into future therapeutic interventions.
Collapse
Affiliation(s)
- Yi Yang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Kangrong Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Shun Li
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Key Laboratory of the Jiangsu Higher Education Institutions for Integrated Traditional Chinese and Western Medicine in Senile Diseases Control (Yangzhou University), Yangzhou, Jiangsu Province, China
| |
Collapse
|
2
|
Liu Y, Song W, Dong W, Gong X, Dong C, Zhao J, Wang R, Song S, Shuang S. Preparation of mitochondrial targeted near-infrared ratio fluorescent probe and its dual response detection for viscosity and ONOO - and cell imaging. Talanta 2025; 292:127909. [PMID: 40081248 DOI: 10.1016/j.talanta.2025.127909] [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: 11/16/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/15/2025]
Abstract
The changes in viscosity and the concentration of ONOO- in mitochondria can effectively reflect the physiological and pathological status of cells. Therefore, the development of effective fluorescent probes for the sensing of viscosity and the concentration of ONOO- in mitochondria has great significance. In this article, a mitochondrial targeted fluorescent probe named Mito-RP was synthesized for the dual responsive sensing of viscosity and ONOO- by introducing pyridine ring and phenylboronic acid ester structure into 4-dimethylamino-cinnamaldehyde with long conjugated chain structure as the parent material. Mito-RP exhibits 600 folds fluorescence enhancement of viscosity in the red-light channel at 700 nm, with pyridine cation as the mitochondrial anchoring group. Simultaneously, Mito-RP appears excellent selectivity towards ONOO- using boronic acid esters as response sites. A new ratio fluorescence analysis method was constructed based on the linear correlation between the emission intensity ratio of Mito-RP at 616 nm/700 nm and the concentration of ONOO-. The linear range is 0.05-33 μM and the detection limit is 9.2 nM. Meanwhile, Mito-RP successfully monitored the changes in viscosity during lipopolysaccharide induced inflammation and rapamycin induced mitochondrial autophagy in HeLa cells. In addition, Mito-RP has also achieved visual imaging of intracellular exogenous/endogenous ONOO-. These studies provide a novel method for in-depth investigation of mitochondrial function and its role in diseases.
Collapse
Affiliation(s)
- Yang Liu
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenqiang Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Wenjuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Xiaojuan Gong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Chuan Dong
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Jie Zhao
- Shanxi Provincial Key Laboratory of Classical Prescription Strengthening Yang, Shanxi Provincial Integrated TCM and WM Hospital, Taiyuan, 030013, China
| | - Ruibing Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau
| | - Shengmei Song
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, and Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| |
Collapse
|
3
|
Jiang W, Liu L, Li W, Liu H, Yang J, Wang P. A lysosomal-targeted switchable fluorescent probe for the detection of peroxynitrite in living tumor cells and in vivo. Talanta 2025; 291:127866. [PMID: 40037163 DOI: 10.1016/j.talanta.2025.127866] [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: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Peroxynitrite (ONOO-) is a reactive nitrogen species whose abnormal accumulation in the body can lead to various diseases, including those related to oxidative stress. Accurate detection of ONOO- levels is essential for the diagnosis and treatment of these diseases. To address this need, we developed a lysosome-targeted fluorescent probe Lyso-PE for detecting ONOO- in tumors. In the presence of ONOO-, probe Lyso-PE showed a large Stokes shift of 100 nm. The probe exhibited high sensitivity, selectivity, and rapid response toward ONOO-. Additionally, Lyso-PE displayed excellent lysosomal targeting and was successfully employed in imaging the exogenous peroxynitrite in tumor cells. In the 4T1 subcutaneous graft tumor model, the probe could effectively distinguish tumors and normal tissues with the help of fluorescence imaging in vivo. Moreover, Lyso-PE could be used for tumor resection guided by fluorescent signals in vivo. These results suggested that Lyso-PE could enhance our understanding of lysosomal function in disease, identify new therapeutic targets, and aid in developing new diagnostic and therapeutic strategies with significant clinical implications.
Collapse
Affiliation(s)
- Wen Jiang
- Department of Pathology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Li Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Wenqing Li
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Huijia Liu
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China
| | - Jing Yang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| | - Peng Wang
- Department of Biomedical Engineering, School of Engineering, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
4
|
Costa-Beber LC, Dantas RM, Peres AM, Obelar Ramos JM, Farias HR, Santos Silva Bast RK, Custódio de Souza IC, Gioda A, de Oliveira J, Costa Rodrigues Guma FT. The effects of direct and macrophage-mediated exposure to aqueous fine particulate matter on vascular endothelial dysfunction. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 376:126407. [PMID: 40348271 DOI: 10.1016/j.envpol.2025.126407] [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: 12/13/2024] [Revised: 04/14/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Fine particulate matter (PM2.5) is an independent risk factor for vascular diseases. In this context, activated macrophages release inflammatory molecules that can contribute to endothelial dysfunction. While the effects of PM2.5's solid fraction on vascular endothelial cells are well-documented, the effect of its polar compounds circulating in the bloodstream remains unclear. In this study, we examined the effects of direct and indirect (macrophage-mediated) exposure to aqueous PM2.5 on the endothelium. CF-1 mice received intranasal instillations of PM2.5 (30 μg in 10 μL) or saline, 5 days per week for two weeks. These animals exhibited considerable endothelial dysfunction linked to oxidative stress. Similarly, macrophages (RAW264.7 lineage) exposed to aqueous PM2.5 (10-fold dilution) exhibited oxidative stress and inflammation, indicating that their reactive phenotype may contribute to the outcomes observed in vivo. Interestingly, their conditioned medium (10 % v/v) enhanced endothelial cell function (EOMA lineage) by reducing reactive oxygen species (ROS) production and promoting an endothelial nitric oxide synthase (eNOS)-dependent increase in nitrite levels, with the exact opposite effect observed in cells directly exposed to aqueous PM2.5. These findings suggest that the macrophage secretome, rather than residual metals, may be responsible for these effects. Consistent with these findings, incubation with the animals' plasma (1 % v/v) also stimulated nitrite production. Additionally, caveolin-1, a key mediator of vesicle uptake, was overexpressed in endothelial cells exposed to conditioned medium, suggesting its involvement in monocyte-endothelium crosstalk. Finally, our results indicated that the macrophage secretome might serve as a mild stimulus, activating protective mechanisms in endothelial cells, whereas direct exposure to aqueous PM2.5 induces dysfunction.
Collapse
Affiliation(s)
- Lílian Corrêa Costa-Beber
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Ricardo Maia Dantas
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil
| | - Ariadni Mesquita Peres
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jéssica Marques Obelar Ramos
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil
| | - Hémelin Resende Farias
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil
| | | | | | - Adriana Gioda
- Pontifícia Universidade Católica do Rio de Janeiro (PUC-Rio), Department of Chemistry, Rio de Janeiro, RJ, Brazil
| | - Jade de Oliveira
- Universidade Federal do Rio Grande do Sul (UFRGS), Department of Biochemistry, Porto Alegre, Rio Grande do Sul, Brazil
| | | |
Collapse
|
5
|
Geng S, Zhou Y, Ng G, Fan Q, Cheong S, Mazur F, Boyer C, Chandrawati R. Selenium nanoparticles as catalysts for nitric oxide generation. Colloids Surf B Biointerfaces 2025; 251:114592. [PMID: 40024109 DOI: 10.1016/j.colsurfb.2025.114592] [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: 12/04/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
The critical role of nitric oxide (NO), a potent signalling molecule, in various physiological processes has driven the development of NO delivery strategies for numerous therapeutic applications. However, NO's short half-life poses a significant challenge for its effective delivery. Glutathione peroxidase, a selenium-containing antioxidant enzyme, can catalyse the decomposition of S-nitrosothiols (endogenous NO prodrugs) to produce NO in situ. Inspired by this, we explored selenium nanoparticles (SeNPs) for their enzyme-mimicking NO-generating activity. Stabilised with polyvinyl alcohol (PVA) or chitosan (CTS), SeNPs demonstrated tuneable NO generation when exposed to varying concentrations of NO prodrug, nanoparticles, and glutathione (GSH). In the presence of GSH, a naturally occurring antioxidant in the human body, 0.1 µg mL-1 of SeNPs could catalytically generate 7.5 µM of NO under physiological conditions within 30 min. We investigated the effects of nanoparticle crystallinity and NO prodrug type on NO generation, as well as the stability and sustained NO generation of the catalytic nanoparticles. PVA-stabilised SeNPs were non-toxic to NIH 3T3 cells and effectively dispersed Pseudomonas aeruginosa biofilms upon NO generation. This study broadens the repertoire of nanomaterials for NO generation and highlights SeNPs as a non-toxic alternative for therapeutic NO delivery.
Collapse
Affiliation(s)
- Shu Geng
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Yingzhu Zhou
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Gervase Ng
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia; Cluster for Advanced Macromolecular Design (CAMD), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Qingqing Fan
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Soshan Cheong
- Electron Microscope Unit, Mark Wainwright Analytical Centre, The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Federico Mazur
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Cyrille Boyer
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia; Cluster for Advanced Macromolecular Design (CAMD), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia
| | - Rona Chandrawati
- School of Chemical Engineering and Australian Centre for Nanomedicine (ACN), The University of New South Wales (UNSW Sydney), Sydney, NSW 2052, Australia.
| |
Collapse
|
6
|
Aksu M, Kaschke K, Podojil JR, Chiang M, Steckler I, Bruce K, Cogswell AC, Schulz G, Kelly J, Wiseman RL, Miller S, Popko B, Chen Y. AA147 Alleviates Symptoms in a Mouse Model of Multiple Sclerosis by Reducing Oligodendrocyte Loss. Glia 2025; 73:1241-1257. [PMID: 39928347 PMCID: PMC12014361 DOI: 10.1002/glia.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Inflammation-induced oligodendrocyte death and CNS demyelination are key features of multiple sclerosis (MS). Inflammation-triggered endoplasmic reticulum (ER) stress and oxidative stress promote tissue damage in MS and in its preclinical animal model, experimental autoimmune encephalitis (EAE). Compound AA147 is a potent activator of the ATF6 signaling arm of the unfolded protein response (UPR) that can also induce antioxidant signaling through activation of the NRF2 pathway in neuronal cells. Previous work showed that AA147 protects multiple tissues against ischemia/reperfusion damage through ATF6 and/or NRF2 activation; however, its therapeutic potential in neuroinflammatory disorders remains unexplored. Here, we demonstrate that AA147 ameliorated the clinical symptoms of EAE and reduced ER stress, oligodendrocyte loss, and demyelination. Additionally, AA147 suppressed T cells in the CNS without altering the peripheral immune response. Importantly, AA147 significantly increased the expressions of Grp78, an ATF6 target gene, in oligodendrocytes, while enhancing levels of Grp78 as well as Ho-1, an NRF2 target gene, in microglia. In cultured oligodendrocytes, AA147 promoted nuclear translocation of ATF6, but not NRF2. Intriguingly, AA147 altered the microglia activation profile, possibly by triggering the NRF2 pathway. AA147 was not therapeutically beneficial during the acute EAE stage in mice lacking ATF6 in oligodendrocytes, indicating that protection primarily involves ATF6 activation in these cells. Overall, our results suggest AA147 as a potential therapeutic opportunity for MS by promoting oligodendrocyte survival and regulating microglia status through distinct mechanisms.
Collapse
Affiliation(s)
- Metin Aksu
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Kevin Kaschke
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Joseph R. Podojil
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - MingYi Chiang
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Ian Steckler
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Kody Bruce
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Andrew C. Cogswell
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Gwen Schulz
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| | - Jeffery Kelly
- The Scripps Research Institute, Department of Chemistry, La Jolla, CA 92037, USA
| | - R. Luke Wiseman
- The Scripps Research Institute, Department of Molecular and Cellular Biology, La Jolla, CA 92037, USA
| | - Stephen Miller
- Northwestern University, Department of Microbiology-Immunology, Chicago, IL 60611, USA
| | - Brian Popko
- Northwestern University, Department of Neurology, Chicago, IL 60611, USA
| | - Yanan Chen
- Loyola University Chicago, Department of Biology, Chicago, IL 60660, USA
| |
Collapse
|
7
|
Zhao Y, Xu T, Wu Z, Li N, Liang Q. Rebalancing redox homeostasis: A pivotal regulator of the cGAS-STING pathway in autoimmune diseases. Autoimmun Rev 2025; 24:103823. [PMID: 40286888 DOI: 10.1016/j.autrev.2025.103823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Autoimmune diseases (ADs) arise from the breakdown of immune tolerance to self-antigens, leading to pathological tissue damage. Proinflammatory cytokine overproduction disrupts redox homeostasis across diverse cell populations, generating oxidative stress that induces DNA damage through multiple mechanisms. Oxidative stress-induced alterations in membrane permeability and DNA damage can lead to the recognition of double-stranded DNA (dsDNA), mitochondrial DNA (mtDNA) and micronuclei-DNA (MN-DNA) by DNA sensors, thereby initiating activation of the cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) pathway. While previous reviews have characterized cGAS-STING activation in autoimmunity, the reciprocal regulation between redox homeostasis and cGAS-STING activation remains insufficiently defined. This narrative review examines oxidative stress-mediated DNA damage as a critical driver of pathological cGAS-STING signaling and delineates molecular mechanisms linking redox homeostasis to autoimmune pathogenesis. Furthermore, we propose therapeutic strategies that combine redox restoration with the attenuation of aberrant cGAS-STING activation, thereby establishing a mechanistic foundation for precision interventions in autoimmune disorders. METHODS: The manuscript is formatted as a narrative review. We conducted a comprehensive search strategy using electronic databases such as PubMed, Google Scholar and Web of Science. Various keywords were used, such as "cGAS-STING," "Redox homeostasis," "Oxidative stress," "pentose phosphate pathway," "Ferroptosis," "mtDNA," "dsDNA," "DNA damage," "Micronuclei," "Reactive oxygen species," "Reactive nitrogen species," "Nanomaterial," "Autoimmune disease," "Systemic lupus erythematosus," "Type 1 diabetes," "Rheumatoid arthritis," "Multiple sclerosis," "Experimental autoimmune encephalomyelitis," "Psoriasis," etc. The titles and abstracts were reviewed for inclusion into this review. After removing duplicates and irrelevant studies, 174 articles met inclusion criteria (original research, English language).
Collapse
Affiliation(s)
- Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China
| | - Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, China; Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
8
|
Abolaji AO, Adedara AO, Madu JC, Owalude OT, Ogunyemi OM, Omoboyowa DA, Omage FB, Whitworth AJ, Aschner M. Experimental and computational insights into the therapeutic mechanisms of resveratrol in a Drosophila α-synuclein model of Parkinson's disease. Sci Rep 2025; 15:17769. [PMID: 40404673 PMCID: PMC12098997 DOI: 10.1038/s41598-025-00698-9] [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: 04/05/2024] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder driven by genetic predisposition and environmental exposure. Given its well-documented antioxidative and neuroprotective properties, resveratrol is increasingly being considered for its potential to counteract the neuronal damage characteristic of Parkinson's disease. Here, we investigated the therapeutic action of resveratrol in a transgenic Drosophila melanogaster model expressing human α-synuclein (SNCA, PD flies), in combination with network pharmacology and molecular docking analyses. The PD flies were fed diet supplemented with resveratrol (15, 30, and 60 mg/kg diet, approximately 6.57, 13.14 and 26.28 mM, respectively), to evaluate lifespan. This was followed by a 21-day treatment of PD flies with similar concentrations of resveratrol in the diet to evaluate cognitive function, oxidative stress, and antioxidant biomarkers, using Levodopa (0.1 mM) as positive control. The results showed that resveratrol supplementation in the diet significantly improved lifespan, locomotor activity, acetylcholinesterase and catalase activities, and thiol content compared to untreated PD flies. Furthermore, resveratrol reduced nitric oxide (nitrite/nitrate), malondialdehyde, and total hydroperoxide levels, and enhanced cellular metabolic activity and upregulated Sod1 mRNA expression (p < 0.05). The network pharmacology and molecular docking analyses identified key molecular targets that may account for the therapeutic action of resveratrol, including B-Cell Lymphoma 2, Monoamine Oxidase (MAO); in flies, MAO-Like, Dopa Decarboxylase, Protein Kinase A and Glycogen Synthase Kinase-3 (GSK-3). Among these, MAO and GSK-3 emerged as top targets as indicated by network prominence and strong binding interactions. Additionally, the binding interaction of resveratrol to SNCA at specific sites suggests a potential role in inhibiting its aggregation, which is a hallmark of PD pathology. Quantum mechanics calculations revealed that resveratrol functions as both a proton donor and acceptor, contributing to its strong target binding interactions and antioxidant potential. Overall, resveratrol supplementation in the diet may be beneficial for PD management by modulating dopamine metabolism, apoptosis, oxidative stress, and cell survival. The study provides valuable experimental and computational insights into the underlying therapeutic mechanisms of action of resveratrol and supports its potential use in PD management.
Collapse
Affiliation(s)
- Amos Olalekan Abolaji
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria.
- Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria.
| | - Adeola Oluwatosin Adedara
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
- Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria
| | - Judith Chizoba Madu
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
- Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria
| | - Oluwabunmi Tomilola Owalude
- Drosophila Laboratory, Molecular Drug Metabolism and Toxicology Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
- Drosophila Research and Training Centre, Basorun, Ibadan, Nigeria
| | - Oludare Michael Ogunyemi
- Structural and Computational Biology Group, Nutritional and Industrial Biochemistry Research Unit, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Damilola A Omoboyowa
- Department of Biochemistry, Adekunle Ajasin University, Akungba-Akoko, Ondo State, Nigeria
| | - Folorunsho Bright Omage
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria, Brazil
| | - Alexander J Whitworth
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY, UK.
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York, USA.
| |
Collapse
|
9
|
Farias HR, Costa-Beber LC, Costa Rodrigues Guma FT, de Oliveira J. Hypercholesterolemia, oxidative stress, and low-grade inflammation: a potentially dangerous scenario to blood-brain barrier. Metab Brain Dis 2025; 40:205. [PMID: 40380979 DOI: 10.1007/s11011-025-01620-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/23/2025] [Indexed: 05/19/2025]
Abstract
For more than a century, hypercholesterolemia has been linked to atherosclerotic cardiovascular disease. Notably, this metabolic condition has also been pointed out as a risk factor for neurodegenerative diseases, such as Alzheimer's disease (AD). Oxidative stress seems to be the connective factor between hypercholesterolemia and cardio and neurological disorders. By disturbing redox homeostasis, hypercholesterolemia impairs nitric oxide (NO) availability, an essential vasoprotective element, and jeopardizes endothelial function and selective permeability. The central nervous system (CNS) is partially protected from peripheral insults due to an arrangement between endothelial cells, astrocytes, microglia, and pericytes that form the blood-brain barrier (BBB). The endothelial dysfunction related to hypercholesterolemia increases the risk of developing cardiovascular diseases and also initiates BBB breakdown, which is a cause of brain damage characterized by neuroinflammation, oxidative stress, mitochondrial dysfunction, and, ultimately, neuronal and synaptic impairment. In this regard, we reviewed the mechanisms by which hypercholesterolemia-induced oxidative stress affects peripheral vessels, BBB, and leads to memory deficits. Finally, we suggest oxidative stress as the missing link between hypercholesterolemia and dementia.
Collapse
Affiliation(s)
- Hémelin Resende Farias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Lílian Corrêa Costa-Beber
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Fátima Theresinha Costa Rodrigues Guma
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil
| | - Jade de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
10
|
Shi X, Zou Y, Li Y, Jiang J. S-Nitrosylated Au@COF nanohybrids for synergistic light-nitric oxide killing of bacteria. Chem Commun (Camb) 2025. [PMID: 40370298 DOI: 10.1039/d5cc01785h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Nanohybrids integrating plasmonic Au and NO donors have been synthesized by a COF-mediated strategy, which effectively eradicate bacteria and biofilms, via mild phototherapy synergized with controlled NO release and ONOO- generation.
Collapse
Affiliation(s)
- Xiaobei Shi
- School of Materials Science & Engineering, Shanghai University, Shanghai 200444, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yu Zou
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yunbo Li
- School of Materials Science & Engineering, Shanghai University, Shanghai 200444, China
| | - Jiang Jiang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
11
|
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
|
12
|
Biancani B, Carosi M, Capasso M, Rossi G, Tafuri S, Ciani F, Cotignoli C, Zinno F, Venturelli E, Galliani M, Spani F. Assessment of Oxidative Stress and Biometric Data in a Captive Colony of Hamadryas Baboons ( Papio hamadryas Linnaeus, 1758) at the Ravenna Zoo Safari (Italy). Vet Sci 2025; 12:466. [PMID: 40431559 DOI: 10.3390/vetsci12050466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
This study evaluates the health of a captive colony of Hamadryas baboons at Ravenna Zoo Safari (Italy), focusing on oxidative stress markers and biometric data. Forty-eight individuals were assessed during routine veterinary procedures: males underwent vasectomy, and females were checked for pregnancy. Biometric data collected included body weight, body length, and genital measurements in males, while females were evaluated for reproductive status. Oxidative stress was measured using two tests that assess both harmful pro-oxidant levels and the body's antioxidant defenses. Results showed no significant differences in oxidative stress levels between sexes, although males and females differed in body weight. Pregnant and postpartum females exhibited higher oxidative stress, likely due to the metabolic and hormonal demands of reproduction. This supports the idea that reproductive activity increases the production of reactive oxygen species, requiring stronger antioxidant responses. In males, correlations between body weight and genital measurements suggest these could help estimate age in the absence of birth records. No link was found between oxidative stress and body weight, indicating limited age-related effects on these markers. Overall, the study highlights the importance of monitoring oxidative stress in captive primates to better understand the effects of reproduction and aging, and to improve welfare and management practices.
Collapse
Affiliation(s)
- Barbara Biancani
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80138 Napoli, Italy
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Monica Carosi
- Department of Science, Roma Tre University, 00146 Rome, Italy
| | - Michele Capasso
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80138 Napoli, Italy
| | - Giacomo Rossi
- School of Biosciences and Veterinary Medicine, University of Camerino, 62024 Matelica, Italy
| | - Simona Tafuri
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80138 Napoli, Italy
| | - Francesca Ciani
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80138 Napoli, Italy
| | | | - Francesco Zinno
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80138 Napoli, Italy
| | | | | | - Federica Spani
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
13
|
Ghosh R, Mazumdar R, Samanta B, Saha S, Mondal B. Reaction of a non-heme iron-nitrosyl with dioxygen: decomposition of the ligand through NOD-like activity. Dalton Trans 2025; 54:7793-7800. [PMID: 40260957 DOI: 10.1039/d5dt00009b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
A high-spin iron(II) nitrosyl, [(TPz)Fe(NO)](ClO4)2, 2 (TPz = Tris(3,5-dimethylpyrazol-1-ylmethyl)amine) with an {Fe(NO)}7 configuration was synthesized and characterized structurally. The dioxygen reactivity of complex 2 in acetonitrile solution results in the oxidation of the ligand. Chemical evidence suggests the involvement of a peroxynitrite intermediate in this reaction. A trapping experiment shows the formation of NO2 during the reaction which supports the proposition of the involvement of the peroxynitrite intermediate. This study gives an insight into an alternate possibility from the dioxygen reactivity of metal-nitrosyl leading to nitric oxide dioxygenase (NOD) activity.
Collapse
Affiliation(s)
- Riya Ghosh
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Rakesh Mazumdar
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Bapan Samanta
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Shankhadeep Saha
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| | - Biplab Mondal
- Department of Chemistry, Indian Institute of Technology Guwahati, Assam - 781039, India.
| |
Collapse
|
14
|
Xie J, Wu M, Li L, Zhu L, Hu L, Li Y, Li W. Integrated bioinformatics and experimental verification to dissect the mechanisms and bioactive ingredients of Radix Rehmanniae in treating multiple sclerosis. Biochem Biophys Res Commun 2025; 763:151790. [PMID: 40233432 DOI: 10.1016/j.bbrc.2025.151790] [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: 12/15/2024] [Revised: 03/22/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025]
Abstract
Multiple sclerosis (MS), as a primary cause of nontraumatic disability in young adults, has no effective treatment yet. Radix Rehmanniae (RR), a typical Traditional Chinese Medicine (TCM), is commonly used in MS patients as a most frequent herbal item in TCM formulas. Our recent study demonstrated that RR alleviated neurological deficits in an experimental MS model. However, direct evidence regarding the holistic mechanisms and bioactive components of RR for MS remains unclear. In this study, we employed an integrative strategy combining bioinformatics and experimental validation to profile the holistic mechanisms of RR, identify its bioactive components, and investigate their potential targets in MS. First, a network pharmacology approach was used to construct a "compound-target-pathway" network, indicating the action of RR on MS in a multicomponent-multitarget mode, and predicting Echinacoside and Acteoside as the primary bioactive ingredients. Bioinformatics analyses of transcriptomics and single-cell RNA sequencing based on GSE datasets indicated that oxidative stress and inflammatory/immune regulation in microglia might serve as crucial mechanisms of Echinacoside and Acteoside in MS pathology. Then, in vitro assays validated that Echinacoside and Acteoside possessed anti-inflammatory and antioxidant properties by scavenging ONOO- and H2O2 directly, and suppressing microglia-derived ONOO- production through inhibition of NF-κB-mediated iNOS and NADPH oxidase. In addition, molecular docking showed strong affinities between Acteoside and inflammation-related targets TGF-β and SMAD2. These findings provide the scientific evidence for clinical application of RR and bring novel insights into MS drug development.
Collapse
Affiliation(s)
- Jing Xie
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China; The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Meiling Wu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Li Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Lixia Zhu
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Liang Hu
- School of Integrative Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yuzhen Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| | - Wenting Li
- Department of Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
15
|
Han HH, Ge PX, Li WJ, Hu XL, He XP. Recent Advancement in Fluorescent Probes for Peroxynitrite (ONOO -). SENSORS (BASEL, SWITZERLAND) 2025; 25:3018. [PMID: 40431815 DOI: 10.3390/s25103018] [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: 03/27/2025] [Revised: 04/27/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025]
Abstract
Peroxynitrite (ONOO-) is a reactive nitrogen species (RNS) that plays pivotal roles in various physiological and pathological processes. The recent literature has seen significant progress in the development of highly sensitive and selective fluorescent probes applicable for monitoring ONOO- dynamics in live cells and a variety of animal models of human diseases. However, the clinical applications of those probes remain much less explored. This review delves into the biological roles of ONOO- and summarizes the design strategies, sensing mechanisms, and bioimaging applications of near-infrared (NIR), long-wavelength, two-photon, and ratiometric fluorescent probes modified with a diverse range of functional groups responsive to ONOO-. Furthermore, we will discuss the remaining problems that prevent the currently developed ONOO- probes from translating into clinical practice.
Collapse
Affiliation(s)
- Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Laboratory of Precision Chemistry and Molecular Engineering, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- Molecular Imaging Center, National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Pan-Xin Ge
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230031, China
| | - Wen-Jia Li
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- Molecular Imaging Center, National Center for Drug Screening, Stake Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xi-Le Hu
- Key Laboratory for Advanced Materials and Joint International Research, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Laboratory of Precision Chemistry and Molecular Engineering, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Laboratory of Precision Chemistry and Molecular Engineering, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- Frontiers Center for Materiobiology and Dynamic Chemistry, School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai 200237, China
- The International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Eastern Hepatobiliary Surgery Hospital, Shanghai 200438, China
| |
Collapse
|
16
|
Zhang J, Lin X, Huang Q, Fu Z, Huang Y, Chen Z, Li N, Lin X. The overexpression of miR-146a in hippocampal microglia via IRAK1/TRAF6/NF-κB pathway improves cognitive function in diabetic mice. Exp Neurol 2025; 391:115291. [PMID: 40349816 DOI: 10.1016/j.expneurol.2025.115291] [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: 02/13/2025] [Revised: 04/17/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND OBJECTIVE Diabetic encephalopathy (DEP), a central nervous system complication of diabetes, is primarily characterized by cognitive dysfunction. Despite its high prevalence and significant risks, the pathogenesis remains poorly understood. This study investigates the effects and mechanisms of miR-146a on cognitive function in DEP mice. METHODS Type 2 diabetic mice models were established by feeding a high-fat diet and administering a low-dose of streptozotocin. And the Morris water maze test was conducted to assess the learning and memory. The adeno-associated virus was delivered into hippocampus by stereotactic injection to overexpress miR-146a in microglia. The mRNA and protein expression levels were determined by quantitative real-time polymerase chain reaction, immunofluorescence, Western blot, and enzyme-linked immunosorbent assay. RESULTS DEP mice exhibited significantly reduced miR-146a expression in hippocampal microglia. This reduction was associated with elevated IRAK1, TRAF6, and NF-κB expression, increased markers of pro-inflammatory microglial phenotypes (CD86 and iNOS), and decreased markers of anti-inflammatory phenotypes (Arg-1 and CD206). Pro-inflammatory cytokines TNF-α and IL-6 were elevated, while anti-inflammatory IL-10 was reduced. Eventually, neuronal apoptosis and cognitive dysfunction were evident. Overexpression of miR-146a in hippocampal microglia reversed these molecular and phenotypic abnormalities, decreased neuronal apoptosis, and significantly improved cognitive performance in diabetic mice. CONCLUSION Downregulation of miR-146a in hippocampal microglia disrupts immune homeostasis through the IRAK1/TRAF6/NF-κB pathway, contributing to DEP. Targeted overexpression of miR-146a restores immune homeostasis, reduces neuronal apoptosis, and ameliorates cognitive impairment.
Collapse
Affiliation(s)
- Jingyu Zhang
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiaoyun Lin
- Department of Clinical Nutrition, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing Huang
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhang Fu
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yihuan Huang
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhiqing Chen
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ningning Li
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| | - Xiahong Lin
- Department of Geriatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China; Shenzhen Key Laboratory of Bone Tissue Repair and Translational Research, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China.
| |
Collapse
|
17
|
Guo Y, Huang H, Zhang Q, Wang H, Liu M, Lin W. A novel dual-channel fluorescent probe for the detection of peroxynitrite anions and lipid droplets in epileptic disease. Anal Chim Acta 2025; 1350:343863. [PMID: 40155169 DOI: 10.1016/j.aca.2025.343863] [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: 12/20/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 04/01/2025]
Abstract
Peroxynitrite (ONOO-) and lipid droplets (LDs) are crucial substances essential for maintaining normal physiological functions in biological systems. They play pivotal roles as biomarkers in the initiation and progression of various diseases, such as epilepsy. Therefore, the simultaneous detection of ONOO- and LDs in epilepsy disorders is of great importance. Here, we discovered that the fluorescence probe composed of trifluoromesulfonate and fluorophore can not only be used as the recognition site of ONOO-, but also has the property of LDs targeting. Therefore, we reasonable designed and synthesized a dual-channel fluorescent probe CBT, capable of simultaneously monitoring ONOO- and LDs. CBT exhibited exceptional dual-response properties: firstly, upon specific reaction with ONOO-, the resulting product BHD emitted a robust red fluorescent signal in the near-infrared region (749 nm); secondly, CBT selectively targeted and labeled LDs, emitting green fluorescence at 482 nm for effective LDs tracking. The signals from these two detection channels did not overlap, which significantly enhanced the accuracy and reliability of detection. Based on these characteristics, CBT has been successfully utilized in real-time imaging of ONOO- and LDs in epilepsy models of cells induced by various drugs. Notably, in a pentylenetetrazole (PTZ)-induced chronic epileptic mice model, CBT exhibited excellent efficacy in ONOO- imaging, further confirming its considerable potential for practical applications. In summary, this study validated CBT as an efficient tool capable of simultaneous detection and differentiation of ONOO- and LDs, presenting a novel and promising strategy for the early diagnosis and treatment of diseases such as epilepsy.
Collapse
Affiliation(s)
- Yingxin Guo
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Huawei Huang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Qian Zhang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Hongjian Wang
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Miaomiao Liu
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China
| | - Weiying Lin
- Institute of Optical Materials and Chemical Biology, Guangxi Key Laboratory of Electrochemical Energy Materials, School of Chemistry and Chemical Engineering, Guangxi University, Nanning, Guangxi, 530004, PR China.
| |
Collapse
|
18
|
Alemu BK, Tommasi S, Hulin JA, Meyers J, Mangoni AA. Current knowledge on the mechanisms underpinning vasculogenic mimicry in triple negative breast cancer and the emerging role of nitric oxide. Biomed Pharmacother 2025; 186:118013. [PMID: 40147105 DOI: 10.1016/j.biopha.2025.118013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Vasculogenic mimicry (VM) is the process by which cancer cells form vascular-like channels to support their growth and dissemination. These channels lack endothelial cells and are instead lined by the tumour cells themselves. VM was first reported in uveal melanomas but has since been associated with other aggressive solid tumours, such as triple-negative breast cancer (TNBC). In TNBC patients, VM is associated with tumour aggressiveness, drug resistance, metastatic burden, and poor prognosis. The lack of effective targeted therapies for TNBC has stimulated research on the mechanisms underpinning VM in order to identify novel druggable targets. In recent years, studies have highlighted the role of nitric oxide (NO), the NO synthesis inhibitor, asymmetric dimethylarginine (ADMA), and dimethylarginine dimethylaminohydrolase 1 (DDAH1), the key enzyme responsible for ADMA metabolism, in regulating VM. Specifically, NO inhibition through downregulation of DDAH1 and consequent accumulation of ADMA appears to be a promising strategy to suppress VM in TNBC. This review discusses the current knowledge regarding the molecular pathways underpinning VM in TNBC, anti-VM therapies under investigation, and the emerging role of NO regulation in VM.
Collapse
Affiliation(s)
- Belete Kassa Alemu
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Injibara University, College of Medicine and Health Sciences, Department of Pharmacy, Injibara, Ethiopia
| | - Sara Tommasi
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia
| | - Julie-Ann Hulin
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Jai Meyers
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Arduino A Mangoni
- Flinders University, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Adelaide, South Australia, Australia; Department of Clinical Pharmacology, Flinders Medical Centre, Southern Adelaide Local Health Network, Adelaide, Australia.
| |
Collapse
|
19
|
Tabtimmai L, Phonchan T, Thongprik N, Kaennakam S, Yodsin N, Choowongkomon K, Sonklin C, Jadsadajerm S, Wisetsai A. New oxepin and dihydrobenzofuran derivatives from Bauhinia saccocalyx roots and their anti-inflammatory, cytotoxic, and antioxidant activities. J Nat Med 2025; 79:543-555. [PMID: 40085400 DOI: 10.1007/s11418-025-01888-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/16/2025]
Abstract
Four new oxepin and dihydrobenzofuran derivatives, saccoxepins A-C (1-3) and saccobenzofurin A (4), along with one known compound, bauhinoxepin A (5), were isolated from the roots of Bauhinia saccocalyx. The structures were elucidated by extensive analysis of spectroscopic data in combination with ECD analysis. The EtOAc extract exhibited significant NO inhibition (94.4 ± 0.35%, 50 μg/mL), and saccoxepin A and bauhinoxepin A demonstrated strong NO suppression, with IC50 values of 49.35 µM and 30.28 µM, respectively, alongside notable antioxidant activity. Saccoxepin A and bauhinoxepin A selectively reduced interleukin-6 (IL-6) levels, while bauhinoxepin A slightly lowered tumor necrosis factor-alpha (TNF-α) at a low dose. Furthermore, bauhinoxepin A exhibited cytotoxicity against HCT-116 cells, with an IC50 of 8.88 µM. These findings suggest that the roots of B. saccocalyx possess potent antioxidant, anti-inflammatory, and anticancer activities, supporting its traditional medicinal applications and highlighting its potential as a source of therapeutic agents.
Collapse
Affiliation(s)
- Lueacha Tabtimmai
- Department of Biotechnology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
- Food and Agro-Industrial Research Center, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Thanyathon Phonchan
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Natrinee Thongprik
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Sutin Kaennakam
- Department of Agro-Industrial, Food, and Environmental Technology, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok (KMUTNB), Bangkok, 10800, Thailand
| | - Nuttapon Yodsin
- Department of Chemistry, Faculty of Science, Silpakorn University, Nakhon Pathom, 73000, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, 10900, Thailand
| | - Chanikan Sonklin
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Supachai Jadsadajerm
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand
| | - Awat Wisetsai
- Department of Industrial Chemistry, Faculty of Applied Science, King Mongkut's University of Technology North Bangkok, Bangkok, 10800, Thailand.
| |
Collapse
|
20
|
Gulcin İ. Antioxidants: a comprehensive review. Arch Toxicol 2025; 99:1893-1997. [PMID: 40232392 DOI: 10.1007/s00204-025-03997-2] [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: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 04/16/2025]
Abstract
Antioxidants had a growing interest owing to their protective roles in food and pharmaceutical products against oxidative deterioration and in the body and against oxidative stress-mediated pathological processes. Screening of antioxidant properties of plants and plant derived compounds requires appropriate methods, which address the mechanism of antioxidant activity and focus on the kinetics of the reactions including the antioxidants. Many studies have been conducted with evaluating antioxidant activity of various samples of research interest using by different methods in food and human health. These methods were classified methods described and discussed in this review. Methods based on inhibited autoxidation are the most suited for termination-enhancing antioxidants and, for chain-breaking antioxidants while different specific studies are needed for preventive antioxidants. For this purpose, the most commonly methods used in vitro determination of antioxidant capacity of food and pharmaceutical constituents are examined and also a selection of chemical testing methods is critically reviewed and highlighting. In addition, their advantages, disadvantages, limitations and usefulness were discussed and investigated for pure molecules and raw plant extracts. The effect and influence of the reaction medium on performance of antioxidants is also addressed. Hence, this overview provides a basis and rationale for developing standardized antioxidant capacity methods for the food, nutraceuticals, and dietary supplement industries. Also, the most important advantages and shortcomings of each method were detected and highlighted. The underlying chemical principles of these methods have been explained and thoroughly analyzed. The chemical principles of methods of 1,1-diphenyl-2-picrylhydrazyl (DPPH•) radical scavenging, 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulphonate) radical (ABTS·+) scavenging, ferric ions (Fe3+) reducing assay, ferric reducing antioxidant power (FRAP) assay, cupric ions (Cu2+) reducing power assay (Cuprac), Folin-Ciocalteu reducing capacity (FCR assay), superoxide radical anion (O2·-), hydroxyl radical (OH·) scavenging, peroxyl radical (ROO·) removing, hydrogen peroxide (H2O2) decomposing, singlet oxygen (1O2) quenching assay, nitric oxide radical (NO·) scavenging assay and chemiluminescence assay are overviewed and critically discussed. Also, the general antioxidant aspects of the main food and pharmaceutical components were discussed through several methods currently used for detecting antioxidant properties of these components. This review consists of two main sections. The first section is devoted to the main components in food and their pharmaceutical applications. The second general section includes definitions of the main antioxidant methods commonly used for determining the antioxidant activity of components. In addition, some chemical, mechanistic, and kinetic properties, as well as technical details of the above mentioned methods, are provided. The general antioxidant aspects of main food components have been discussed through various methods currently used to detect the antioxidant properties of these components.
Collapse
Affiliation(s)
- İlhami Gulcin
- Faculty of Sciences, Department of Chemistry, Atatürk University, 25240, Erzurum, Türkiye.
| |
Collapse
|
21
|
Piknova B, Park JW, Schechter AN. Nitrate as Warden of Nitric Oxide Homeostasis in Mammals. Nutrients 2025; 17:1544. [PMID: 40362853 PMCID: PMC12073257 DOI: 10.3390/nu17091544] [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: 04/04/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Homeostasis is the self-regulating processes in cells and organisms designed to maintain stability of the internal environment while adjusting to external changes. To achieve this dynamic stability, internal conditions oscillate within tightly regulated physiological tolerance limits. In mammals, maintaining nitric oxide (NO) availability appears crucial to sustain relatively constant blood flow into all organs and tissues. We hypothesize that NO homeostasis is one of the most important vital processes for warm-blooded animals. It is impossible to conserve the stability of most other vital substances, such as O2, CO2, blood sugar, pH, and temperature, to name just few, without well-functioning tissue perfusion. NO in mammals is generated either from L-arginine by nitric oxide synthases (NOSs) or by the reduction of nitrate (NO3-) to nitrite (NO2-) and NO by several proteins. Here we first discuss the organization of these two NO metabolic pathways, emphasizing that both pathways "cross" and "funnel" unused NO into the overall nitrate-nitrite-NO pathway. This pathway is cyclic, which gives nitrate a unique place in metabolism and predisposes it as a reservoir for NO. Then, we discuss the role of NO homeostasis that, by maintaining organ and tissue perfusion, supports and preserves constancy of other blood-delivered substances. This "governing" role of NO makes even clearer that the existence of NO storage and precursor molecules is necessary, to avoid NO shortages in cases of the precursor's or storage molecule's temporary unavailability, to ensure uninterrupted tissue access to NO. We propose that the skeletomuscular system and skin act as nitrate reservoirs assuring NO bioavailability at various external and internal conditions.
Collapse
Affiliation(s)
- Barbora Piknova
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | | | | |
Collapse
|
22
|
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
|
23
|
Tortolani D, Decandia D, Giacovazzo G, Scipioni L, Panuccio A, Ciaramellano F, Eugelio F, Fanti F, Latagliata EC, La Barbera L, Cutuli D, Compagnone D, D’Amelio M, Coccurello R, Oddi S, Petrosini L, Maccarrone M. Chronic palmitoylethanolamide administration via slow-release subcutaneous pellets promotes neuroprotection and mitigates neuroinflammation in the Tg2576 mouse model of Alzheimer's disease. Front Cell Neurosci 2025; 19:1571428. [PMID: 40313591 PMCID: PMC12043567 DOI: 10.3389/fncel.2025.1571428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive and non-cognitive decline associated with neuropathological hallmarks, including neuroinflammation. Palmitoylethanolamide (PEA), an endogenous lipid with anti-inflammatory and neuroprotective properties, has emerged as a promising therapeutic agent in managing AD. This study investigated the therapeutic effects of chronic (6-months) PEA administration via subcutaneous pellet in Tg2576 mice, a validated model of AD. The impact of PEA on amyloid precursor protein (APP) processing, astrocytic activation, microglial reactivity and neuroinflammation, nitrosative stress, dendritic spine density in hippocampal CA1 pyramidal neurons, and cognitive performance was assessed. Chronic PEA treatment of Tg2576 mice increased the expression of the α-secretase ADAM9 and reduced astrogliosis. Furthermore, PEA attenuated microglia reactivity, downregulated pro-inflammatory (CXCL13, MCP-1, GCSF) and upregulated anti-inflammatory (CXC3CL1 and IL-9) cytokine expression. Chronic PEA administration also decreased protein nitrosylation, downregulated calcineurin expression, restored dendritic spine density, and improved cognitive functions. Chronic PEA administration offers a promising therapeutic approach for AD by mitigating neuroinflammation, oxidative stress, and synaptic dysfunction, ultimately leading to cognitive function restoration.
Collapse
Affiliation(s)
- Daniel Tortolani
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Davide Decandia
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Lucia Scipioni
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Anna Panuccio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | | | - Fabiola Eugelio
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Federico Fanti
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Livia La Barbera
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Debora Cutuli
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Dario Compagnone
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Marcello D’Amelio
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Roberto Coccurello
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Institute for Complex Systems (ISC), National Council of Research (CNR), Rome, Italy
| | - Sergio Oddi
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Laura Petrosini
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research, Fondazione Santa Lucia IRCCS, Rome, Italy
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| |
Collapse
|
24
|
Hu X, Dou Q, Jiang P, Zhang M, Wang J. Targeting matrix metalloproteinases activating and indoleamine 2,3-dioxygenase suppression for triple-negative breast cancer multimodal therapy. Int J Biol Macromol 2025; 310:143289. [PMID: 40253020 DOI: 10.1016/j.ijbiomac.2025.143289] [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: 02/20/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
The dense extracellular matrix (ECM) and immunosuppressive tumor microenvironment represent two major challenges in the treatment of triple-negative breast cancer (TNBC). To address these obstacles, this study has developed a polymer micelle (NTP) for ECM remodeling and mitigation the immune microenvironment, based on activating endogenous matrix metalloproteinases (MMP) and suppression indoleamine 2,3-dioxygenase (IDO). Through self-assembly technology, this micelle effectively incorporates chemotherapy drugs (camptothecin (CPT) and cinnamaldehyde (CA)), reactive oxygen species (ROS) stimulants, nitric oxide (NO) donor and IDO inhibitor (NLG919), where CPT and CA have been reported to help generating ROS mainly in the mitochondrion. The guanidine group of poly-L-arginine (PArg), as an NO donor, can react with ROS to generate NO. The micelles aim to achieve significant therapeutic outcomes through robust drug penetration and anti-tumor immunity in multimodal therapy. They exhibit remarkable tumor tissue penetration ability, facilitating precise targeting of mitochondria and ROS production stimulation. Building upon this therapeutic foundation, the micellar system achieves in situ NO release, which effectively degrades the ECM through the activation of MMPs, while simultaneously promoting tumor cells apoptosis. Furthermore, the encapsulated NLG919 can be released and effectively mitigating the immunosuppressive milieu and triggering anti-tumor immune responses. Experimental results demonstrate that the micelles exhibit significant anti-tumor effects both in vitro and in vivo, accompanied by favorable biocompatibility. This study provides new insights into the application of subcellular targeting drug delivery systems in TNBC treatment, potentially heralding a new breakthrough in TNBC therapy.
Collapse
Affiliation(s)
- Xiaoxiao Hu
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China; Pharmaceutical Department, Baoding Second Hospital, Baoding 071051, China
| | - Qingqing Dou
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Peixiao Jiang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China
| | - Mo Zhang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jing Wang
- School of Pharmacy, National Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
25
|
Ahmed HS. Neuropharmacological effects of calycosin: a translational review of molecular mechanisms and therapeutic applications. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04154-3. [PMID: 40237798 DOI: 10.1007/s00210-025-04154-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
Calycosin, a naturally occurring isoflavonoid found predominantly in Astragalus membranaceus, exhibits significant therapeutic potential in various neurological conditions. Its multifaceted bioactive properties-antioxidant, anti-inflammatory, and anti-apoptotic-position it as a promising candidate for neuroprotection and neuroregeneration. This review explores calycosin's mechanisms of action, including its modulation of key signaling pathways such as HMGB1/TLR4/NF-κB (high mobility group box 1/toll-like receptor 4/nuclear factor kappa B), phosphatidylinositol-3-kinase (PI3 K)/Akt, ERK1/2 (extracellular signal-regulated kinase 1/2), and Hsp90/Akt/p38. In cerebral ischemia/reperfusion injury, calycosin reduces oxidative stress markers like ROS (reactive oxygen species) and MDA (malondialdehyde), enhances antioxidant enzymes (SOD (superoxide dismutase) and GPX (glutathione peroxidase)), and downregulates pro-inflammatory cytokines (TNF-α, IL-1β) through the HMGB1/TLR4/NF-κB pathway. It also inhibits autophagy via the STAT3/FOXO3a pathway and apoptosis by modulating Bax and Bcl-2 expression. In neuro-oncology, calycosin inhibits glioblastoma cell migration and invasion by modulating the TGF-β-mediated mesenchymal properties and suppressing the c-Met and CXCL10 signaling pathways. Additionally, it enhances the efficacy of temozolomide in glioma treatment through apoptotic pathways involving caspase-3 and caspase-9. Calycosin shows promise in Alzheimer's disease by reducing β-amyloid production and tau hyperphosphorylation via the GSK-3β pathway and improving mitochondrial function through the peroxisome proliferator-activated receptor gamma coactivator 1-Alpha (PGC-1α)/mitochondrial transcription factor A (TFAM) signaling pathway. In Parkinson's disease, calycosin mitigates oxidative stress, prevents dopaminergic neuronal death, and reduces neuroinflammation by inhibiting the TLR/NF-κB and MAPK pathways. It has also shown therapeutic potential in meningitis and even neuroprotective effects against hyperbilirubinemia-induced nerve injury. Despite these promising findings, further research, including detailed mechanistic studies and clinical trials, is needed to fully understand calycosin's therapeutic mechanisms and validate its potential in human subjects. Developing advanced delivery systems and exploring synergistic therapeutic strategies could further enhance its clinical application and effectiveness.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
26
|
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
|
27
|
Grzelakowska A, Podsiadły R, Zielonka J. Phenyl Radical-Mediated Fluorogenic Cyclization for Specific Detection of Peroxynitrite. Anal Chem 2025; 97:7299-7306. [PMID: 40146989 PMCID: PMC11983361 DOI: 10.1021/acs.analchem.4c06983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/25/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Peroxynitrite (ONOO-), a biological oxidizing and nitrating species responsible for post-translational modification of cellular proteins, has been implicated in numerous pathologies carrying an inflammatory component. Specific detection of ONOO- in biological systems remains a challenge, and boronates are regarded as the most promising class of probes for the detection and quantitation of ONOO-. Oxidation of boronate probes by ONOO- results in the formation of minor ONOO--specific products via a pathway involving a phenyl radical-type intermediate, in addition to the major phenolic product. Here, we report fluorogenic cyclization of the phenyl-type radical formed during oxidation of a boronate probe by ONOO-, with the production of a fluorescent product, and we propose a new approach for the specific detection of ONOO- based on this observation. We characterized the kinetics and stoichiometry of the reaction of benzophenone-2-boronic acid with ONOO- and identified 2-hydroxybenzophenone as the major product and fluorenone (FLN) and 2-nitrobenzophenone as the minor ONOO--specific products. Hydrogen peroxide neither alone nor in the presence of myeloperoxidase and nitrite produces FLN or 2-nitrobenzophenone. FLN can be selectively detected using fluorescence spectroscopy, providing a chemical principle for the development of next-generation probes for ONOO-, with noninvasive, fluorescence-based detection of ONOO--specific products. Fluorescence-based monitoring of FLN was successfully applied for the detection of ONOO- generated in situ from the decomposition of SIN-1, a thermal source of the superoxide radical anion and nitric oxide.
Collapse
Affiliation(s)
- Aleksandra Grzelakowska
- Institute
of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, Lodz 90-537, Poland
- Department
of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| | - Radosław Podsiadły
- Institute
of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, Lodz 90-537, Poland
| | - Jacek Zielonka
- Department
of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, United States
| |
Collapse
|
28
|
Gong T, Chen HJ, Fan RYS, Zhang X, Yong KT, Kong KV. In Situ 3D SERS Imaging of CO 2 Reduction in Living Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410250. [PMID: 40042411 DOI: 10.1002/smll.202410250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/20/2025] [Indexed: 04/17/2025]
Abstract
The advancement of catalytic processes for therapeutic applications is pivotal to the development of next-generation medical technologies. One of the major challenges in this field lies in elucidating the intracellular generation of small molecules, such as carbon monoxide (CO), nitric oxide (NO), and others, which possess significant therapeutic potential. In this study, in situ surface-enhanced Raman spectroscopy (SERS) is employed to visualize and monitor the carbon dioxide (CO2) reduction process mediated by a rhenium coated gold nanoflower (Re@Au) catalyst within living cells. The findings provide direct spectroscopic evidence of CO2 reduction under intracellular conditions, demonstrating that CO can be catalytically generated from CO2 in the cellular environment. These results position SERS as an indispensable tool for investigating catalytic processes in biological systems, providing molecular-level insights through the analysis of molecular fingerprint spectra that are typically beyond the capabilities of conventional microscopy techniques.
Collapse
Affiliation(s)
- Tianxun Gong
- School of Integrated Circuit Science and Engineering (Exemplary School of Microelectronics), University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Hsin-Jou Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan (ROC)
| | - Ricky Yu-Syun Fan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan (ROC)
| | - Xiaosheng Zhang
- School of Integrated Circuit Science and Engineering (Exemplary School of Microelectronics), University of Electronic Science and Technology of China, Chengdu, 611731, P. R. China
| | - Ken-Tye Yong
- School of Biomedical Engineering; Faculty of Engineering, The University of Sydney, NSW, 2006, Australia
| | - Kien Voon Kong
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan (ROC)
- Center for Emerging Material and Advanced Devices, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan (ROC)
| |
Collapse
|
29
|
Muccilli SG, Schwarz B, Shue B, Jessop F, Shannon JG, Larson CL, Hage A, Hong SH, Bohrnsen E, Hsu T, Ashbrook AW, Sturdevant GL, Robertson SJ, Guarnieri JW, Lack J, Wallace DC, Bosio CM, MacDonald MR, Rice CM, Yewdell JW, Best SM. Mitochondrial hyperactivity and reactive oxygen species drive innate immunity to the yellow fever virus-17D live-attenuated vaccine. PLoS Pathog 2025; 21:e1012561. [PMID: 40258014 PMCID: PMC12052391 DOI: 10.1371/journal.ppat.1012561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 05/05/2025] [Accepted: 03/31/2025] [Indexed: 04/23/2025] Open
Abstract
The yellow fever virus 17D (YFV-17D) live attenuated vaccine is considered one of the most successful vaccines ever generated associated with high antiviral immunity, yet the signaling mechanisms that drive the response in infected cells are not understood. Here, we provide a molecular understanding of how metabolic stress and innate immune responses are linked to drive type I IFN expression in response to YFV-17D infection. Comparison of YFV-17D replication with its parental virus, YFV-Asibi, and a related dengue virus revealed that IFN expression requires RIG-I-Like Receptor signaling through MAVS, as expected. However, YFV-17D uniquely induces mitochondrial respiration and major metabolic perturbations, including hyperactivation of electron transport to fuel ATP synthase. Mitochondrial hyperactivity generates reactive oxygen species (ROS) including peroxynitrite, blocking of which abrogated MAVS oligomerization and IFN expression in non-immune cells without reducing YFV-17D replication. Scavenging ROS in YFV-17D-infected human dendritic cells increased cell viability yet globally prevented expression of IFN signaling pathways. Thus, adaptation of YFV-17D for high growth imparts mitochondrial hyperactivity to meet energy demands, resulting in generation of ROS as the critical messengers that convert a blunted IFN response into maximal activation of innate immunity essential for vaccine effectiveness.
Collapse
Affiliation(s)
- Samantha G. Muccilli
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Benjamin Schwarz
- Research Technologies Branch, NIAID, NIH, Hamilton, Montana, United States of America
| | - Byron Shue
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Forrest Jessop
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, Montana, United States of America
| | - Jeffrey G. Shannon
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Charles L. Larson
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Adam Hage
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Seon-Hui Hong
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Eric Bohrnsen
- Research Technologies Branch, NIAID, NIH, Hamilton, Montana, United States of America
| | - Thomas Hsu
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Alison W. Ashbrook
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Gail L. Sturdevant
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Shelly J. Robertson
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| | - Joseph W. Guarnieri
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Division on Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Catharine M. Bosio
- Immunity to Pulmonary Pathogens Section, Laboratory of Bacteriology, NIAID, NIH, Hamilton, Montana, United States of America
| | - Margaret R. MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Charles M. Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York, United States of America
| | - Jonathan W. Yewdell
- Cellular Biology Section, Laboratory of Viral Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Sonja M. Best
- Innate Immunity and Pathogenesis Section, Laboratory of Neurological Infections and Immunity, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, United States of America
| |
Collapse
|
30
|
Patel RB, Kumskova M, Kodali H, Budnik I, Kuznetsov V, Jain A, Jha A, Thedens D, Dhanesha N, Sutariya B, Nagarkatti KA, Lamb J, Kamat P, Shi Y, Avery B, Imai T, Jin X, Chauhan A, Boisserand LS, Khan MB, Dhandapani K, Sanganahalli BG, Sansing LH, Hess DC, Koehler RC, McCullough LD, Aronowski J, Ayata C, Diniz MA, Lyden PD, Planas AM, Chamorro A, Chauhan AK, Leira EC. Uric Acid Stroke Cerebroprotection Transcended Sex, Age, and Comorbidities in a Multicenter Preclinical Trial. Stroke 2025; 56:965-973. [PMID: 40091742 PMCID: PMC11932773 DOI: 10.1161/strokeaha.124.048748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 12/06/2024] [Accepted: 01/21/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Past failures in translating stroke cerebroprotection provoked calls for a more rigorous methodological approach, leading to the stroke preclinical assessment network SPAN (Stroke Preclinical Assessment Network), where uric acid (UA) treatment exceeded a prespecified efficacy boundary for the primary functional outcome. Still, successful translation to humans requires confirmation of the effect of UA across key biological variables relevant to patients with stroke. METHODS We measured the effects of intravenous UA treatment (16 mg/kg) versus intravenous saline in groups of animals enrolled in the SPAN network with diverse comorbidities, sex, and age. The masked study drug or placebo was administered during reperfusion in rodents undergoing a transient middle cerebral artery filament occlusion. The primary outcome was the modified corner test index at day 30 poststroke, and numerous secondary outcomes were collected. A modified intention-to-treat population was used in the analysis. We tested for any interactions with sex, age, and comorbidities (obesity-induced hyperglycemia and hypertension). RESULTS In total, 710 animals were randomized to receive either intravenous UA or saline. After accounting for procedural dropouts and exclusions from treatment, a total of 687 animals were qualified and analyzed, including 458 assigned to UA and 229 to intravenous saline control. UA-treated animals exhibited a better primary functional outcome at day 30 (probability, 0.56 [95% CI, 0.52-0.60]; P=0.006). UA-treated animals also had a better corner test index at day 7 (probability, 0.55 [95% CI, 0.5-0.59]; P=0.035) and a higher survival rate at day 30 (hazard ratio, 1.41 [95% CI, 1.08-1.83]; P=0.011). Brain morphometry at day 2 and 30 was comparable between the treatment groups. The improved functional outcome and survival in UA-treated animals were preserved across different species, sexes, ages, and comorbidities. CONCLUSIONS UA provides ischemic stroke cerebroprotection across key relevant biological variables, making it a promising intervention to be further tested in human clinical trials.
Collapse
Affiliation(s)
- Rakesh B. Patel
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mariia Kumskova
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Hanish Kodali
- Department of Population Health Science and Policy, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Ivan Budnik
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Aditi Jain
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Abhishek Jha
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Daniel Thedens
- Department of Radiology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nirav Dhanesha
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Brijesh Sutariya
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Karisma A. Nagarkatti
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jessica Lamb
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Pradip Kamat
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Yanrong Shi
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Brooklyn Avery
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Takahiko Imai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xuyan Jin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Anjali Chauhan
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | | | - Mohammad B. Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Krishnan Dhandapani
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | | | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - David C. Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Raymond C. Koehler
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Jaroslaw Aronowski
- Department of Neurology, McGovern Medical School, University of Texas HSC, Houston, TX, USA
| | - Cenk Ayata
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Márcio A. Diniz
- Department of Population Health Science and Policy, Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Patrick D. Lyden
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anna M. Planas
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Angel Chamorro
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Neurology, Hospital Clinic, University of Barcelona, Barcelona, Spain
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Anil K. Chauhan
- Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Enrique C. Leira
- Department of Neurology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Neurosurgery, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
31
|
Marian AJ. Causes and consequences of DNA double-stranded breaks in cardiovascular disease. Mol Cell Biochem 2025; 480:2043-2064. [PMID: 39404936 DOI: 10.1007/s11010-024-05131-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/29/2024] [Indexed: 04/02/2025]
Abstract
The genome, whose stability is essential for survival, is incessantly exposed to internal and external stressors, which introduce an estimated 104 to 105 lesions, such as oxidation, in the nuclear genome of each mammalian cell each day. A delicate homeostatic balance between the generation and repair of DNA lesions maintains genomic stability. To initiate transcription, DNA strands unwind to form a transcription bubble and provide a template for the RNA polymerase II (RNAPII) complex to synthesize nascent RNA. The process generates DNA supercoils and introduces torsional stress. To enable RNAPII processing, the supercoils are released by topoisomerases by introducing strand breaks, including double-stranded breaks (DSBs). Thus, DSBs are intrinsic genomic features of gene expression. The breaks are quickly repaired upon processing of the transcription. DNA lesions and damaged proteins involved in transcription could impede the integrity and efficiency of RNAPII processing. The impediment, which is referred to as transcription stress, not only could lead to the generation of aberrant RNA species but also the accumulation of DSBs. The latter is particularly the case when topoisomerase processing and/or the repair mechanisms are compromised. The DSBs activate the DNA damage response (DDR) pathways to repair the damaged DNA and/or impose cell cycle arrest and cell death. In addition, the release of DSBs into the cytosol activates the cytosolic DNA-sensing proteins (CDSPs), which along with the nuclear DDR pathways induce the expression of senescence-associated secretory phenotype (SASP), cell cycle arrest, senescence, cell death, inflammation, and aging. The primary stimulus in hereditary cardiomyopathies is a mutation(s) in genes encoding the protein constituents of cardiac myocytes; however, the phenotype is the consequence of intertwined complex interactions among numerous stressors and the causal mutation(s). Increased internal DNA stressors, such as oxidation, alkylation, and cross-linking, are expected to be common in pathological conditions, including in hereditary cardiomyopathies. In addition, dysregulation of gene expression also imposes transcriptional stress and collectively with other stressors provokes the generation of DSBs. In addition, the depletion of nicotinamide adenine dinucleotide (NAD), which occurs in pathological conditions, impairs the repair mechanism and further facilitates the accumulation of DSBs. Because DSBs activate the DDR pathways, they are expected to contribute to the pathogenesis of cardiomyopathies. Thus, interventions to reduce the generation of DSBs, enhance their repair, and block the deleterious DDR pathways would be expected to impart salubrious effects not only in pathological states, as in hereditary cardiomyopathies but also aging.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetic Studies, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX, 77030, USA.
| |
Collapse
|
32
|
Zhao HY, Zhan ZC, Ou HL, Wu TY, Zhu HH, Lin Q, Li YL, Wang JH, Zhou GX, Tang Q, Zhang YB, Wang GC. Dihydro-β-agarofuran sesquiterpenoids from the root bark of Tripterygium wilfordii and their anti-neuroinflammatory activities. Bioorg Chem 2025; 157:108236. [PMID: 39952061 DOI: 10.1016/j.bioorg.2025.108236] [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: 01/16/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
A phytochemical study of Tripterygium wilfordii root bark was conducted 25 novel dihydro-β-agarofuran sesquiterpenoids (1-25) and 20 known analogues (26-45). Structural analysis elucidated by comprehensive spectroscopic analysis, including X-ray crystallography and electronic circular dichroism (ECD). Anti-neuroinflammatory assessments in BV-2 cells revealed certain compounds effectively suppressed tumor necrosis factor-α (TNF-α) and interleukin 6 (IL-6). A preliminary structure-activity relationships analysis explored the relationship between compound structure and their inflammatory mediator inhibition. Notably, compound 7 modulated nuclear factor-κB (NF-κB) signaling by inhibiting IκBα and p65 phosphorylation. These findings offer novel perspectives on the bioactivity and anti-neuroinflammatory mechanisms of Tripterygium wilfordii derivatives.
Collapse
Affiliation(s)
- Hai-Yue Zhao
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Zhao-Chun Zhan
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Hui-Lin Ou
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Tian-Yuan Wu
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Hui-Hui Zhu
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Qiang Lin
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Yao-Lan Li
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Jing-Hao Wang
- The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, the First Affiliated Hospital, Jinan University, Guangzhou 510632 China
| | - Guang-Xiong Zhou
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China
| | - Qing Tang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China.
| | - Yu-Bo Zhang
- Guangdong Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, and Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou 510632 China; The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, the First Affiliated Hospital, Jinan University, Guangzhou 510632 China.
| | - Guo-Cai Wang
- Institute of Traditional Chinese Medicine & Natural Products, College of Pharmacy, and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, China; The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, the First Affiliated Hospital, Jinan University, Guangzhou 510632 China.
| |
Collapse
|
33
|
Menoni M, Alcoba P, Zuluaga MJ, Peluffo RD. Generation of cellular reactive oxygen and nitrogen species by exposure to ultraviolet radiation. Biophys Rev 2025; 17:547-560. [PMID: 40376412 PMCID: PMC12075055 DOI: 10.1007/s12551-025-01298-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 05/18/2025] Open
Abstract
Reactive oxygen and nitrogen species, such as superoxide and peroxynitrite anions, are produced in our body as a result of normal metabolic functions or under pathologic conditions (oxidative and nitro-oxidative stress). A well-documented battery of antioxidant enzymes and cofactors are in place to fight this stress and restore the redox balance of the cell. However, comprehensive information on the generation of these reactive species by exposing cell components to ultraviolet (UV) light, specifically UVA and UVB sunlight, is scarce or missing. In this short review, we attempt to cover several enzymes and cofactors that are targets of UV radiation as it relates to the production (or consumption) of these oxidants, and, when known, discuss the underlying mechanisms. Because of their key importance, UV light effects on DNA are briefly discussed.
Collapse
Affiliation(s)
- Macarena Menoni
- Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de La República, Rivera 1350, CP: 50000, Salto, Uruguay
| | - Pablo Alcoba
- Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de La República, Rivera 1350, CP: 50000, Salto, Uruguay
| | - María J. Zuluaga
- Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de La República, Rivera 1350, CP: 50000, Salto, Uruguay
| | - R. Daniel Peluffo
- Group of Biophysical Chemistry, Department of Biological Sciences, CENUR Litoral Norte, Universidad de La República, Rivera 1350, CP: 50000, Salto, Uruguay
| |
Collapse
|
34
|
Mollace R, Macrì R, Serra M, Ritorto G, Ussia S, Scarano F, Cardamone A, Musolino V, Coppoletta AR, Gliozzi M, Scipione G, Carresi C, Pozharova K, Muscoli C, Barillà F, Volterrani M, Mollace V. The Antioxidant Power of Bergamot Polyphenolic Fraction Gold Potentiates the Effects of L-Citrulline in Athlete Performance and Vasodilation in a Pilot Study. Nutrients 2025; 17:1106. [PMID: 40218864 PMCID: PMC11990273 DOI: 10.3390/nu17071106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background: The dietary supplement citrulline might increase nitric oxide levels, leading to vasodilation and improved blood flow, potentially benefiting athletes' aerobic exercise performance. However, rapid oxidative impairment of the L-arginine/nitric oxide (NO) pathway limits these effects. This is countered by Bergamot Polyphenolic Fraction Gold® (BPFG), a strong natural antioxidant. To investigate L-citrulline + BPFG supplementation's effects, we performed a randomized, double-blind, placebo-controlled pilot trial on athletic performance and blood flow in trained athletes (cyclists). Methods: Random assignment of 90 male athletes resulted in nine different groups: placebo for Group 1, BPFG at 500 and 1000 mg daily for Groups 2 and 3, L-citrulline at 1000 and 2000 mg/daily for Groups 4 and 5, and the combination product of BPFG plus citrulline (N.O. Max) for Groups 6-9. Baseline and 3-month pre- and post-exercise biochemical, reactive vasodilation (RHI), and maximal oxygen consumption measurements were taken for all subjects. Results: Three months of the combination of BPFG and L-citrulline (N.O. Max) produced a significant synergistic effect, markedly increasing NO (p < 0.001 vs. placebo) release and RHI (p < 0.001 vs. placebo). Cardiorespiratory fitness improved significantly with the BPFG and L-citrulline combination, resulting in substantially higher VO2 max, VT1, VT2, and peak power and a significantly lower heart rate (p < 0.01 vs. placebo). No harmful adverse effects were observed. Conclusions: N.O. Max supplementation, providing beneficial effects on the antioxidant state and preserving the vascular endothelium might be a supplementation strategy to improve athletic performance and potentiate results. Given the small sample size, this study serves as a pilot, and further research is needed to validate these findings on a larger scale.
Collapse
Affiliation(s)
- Rocco Mollace
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
- Department of Experimental Medicine, University “Tor Vergata” of Rome, 00133 Rome, Italy;
| | - Roberta Macrì
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Maria Serra
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Giovanna Ritorto
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Sara Ussia
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Federica Scarano
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Antonio Cardamone
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Vincenzo Musolino
- Laboratory of Pharmaceutical Biology, Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy;
| | - Anna Rita Coppoletta
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Micaela Gliozzi
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Giuseppe Scipione
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Cristina Carresi
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Kateryna Pozharova
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Carolina Muscoli
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
| | - Francesco Barillà
- Department of Experimental Medicine, University “Tor Vergata” of Rome, 00133 Rome, Italy;
| | | | - Vincenzo Mollace
- Institute of Research for Food Safety and Health (IRC-FSH), Department of Health Sciences, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (M.S.); (G.R.); (S.U.); (F.S.); (A.C.); (A.R.C.); (M.G.); (G.S.); (C.C.); (K.P.); (C.M.)
- Renato Dulbecco Institute, 88046 Lamezia Terme, Italy
| |
Collapse
|
35
|
Mukhopadhyay P, Yokus B, Paes-Leme B, Bátkai S, Ungvári Z, Haskó G, Pacher P. Chronic alcohol consumption accelerates cardiovascular aging and decreases cardiovascular reserve capacity. GeroScience 2025:10.1007/s11357-025-01613-w. [PMID: 40111699 DOI: 10.1007/s11357-025-01613-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
The pathology of cardiovascular aging is complex, involving mitochondrial dysfunction, oxidative and nitrative stress, oxidative DNA injury, impaired lipid metabolism, cell death, senescence, and chronic inflammation. These processes lead to remodeling and structural changes in the cardiovascular system, resulting in a progressive decline in cardiovascular reserve capacity and health, and an increased risk of diseases and mortality. Excessive alcohol consumption exacerbates these risks by promoting hypertension, stroke, arrhythmias, coronary artery disease, cardiomyopathy, and sudden cardiac death, yet the effects of chronic alcohol consumption on cardiovascular aging remain unclear. Herein, we explored the impact of a 6-month 5% Lieber-DeCarli alcohol diet in young (3 months old) and aging (24-26 months old) Fisher F344BNF1 rats. We assessed detailed hemodynamics, mitochondrial function, oxidative/nitrative stress, lipid metabolism, inflammation, cell death, senescence, and myocardial fibrosis using the pressure-volume system, isolated vascular rings, and various histological, biochemical, and molecular biology methods. Alcohol consumption in both young and aging rats impaired mitochondrial function, disrupted cholesterol and triglyceride metabolism, and increased oxidative/nitrative stress, inflammation, cell death, and senescence, leading to a decline in systolic contractile function. In aging rats, alcohol further exacerbated diastolic dysfunction and myocardial fibrosis. Alcohol also increased oxidative/nitrative stress, apoptosis, and senescence in the vasculature, contributing to endothelial dysfunction and increased total peripheral resistance. Additionally, alcohol exacerbated the aging-related ventriculo-arterial uncoupling and diminished cardiac efficiency, further reducing cardiovascular reserve capacity. In conclusion, chronic alcohol consumption promotes cardiovascular aging and further diminishes the already impaired cardiac and vascular reserve capacity associated with aging.
Collapse
Affiliation(s)
- Partha Mukhopadhyay
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Burhan Yokus
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Bruno Paes-Leme
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Sándor Bátkai
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Zoltán Ungvári
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
36
|
Patel TA, Zheng H, Patel KP. Sodium-Glucose Cotransporter 2 Inhibitors as Potential Antioxidant Therapeutic Agents in Cardiovascular and Renal Diseases. Antioxidants (Basel) 2025; 14:336. [PMID: 40227417 PMCID: PMC11939188 DOI: 10.3390/antiox14030336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Redox (reduction-oxidation) imbalance is a physiological feature regulated by a well-maintained equilibrium between reactive oxygen species (ROS) and oxidative stress (OS), the defense system of the body (antioxidant enzymes). The redox system comprises regulated levels of ROS in the cells, tissues and the overall organ system. The levels of ROS are synchronized by gradients of electrons that are generated due to sequential reduction and oxidation of various biomolecules by various enzymes. Such redox reactions are present in each cell, irrespective of any tissue or organ. Failure in such coordinated regulation of redox reactions leads to the production of excessive ROS and free radicals. Excessively produced free radicals and oxidative stress affect various cellular and molecular processes required for cell survival and growth, leading to pathophysiological conditions and, ultimately, organ failure. Overproduction of free radicals and oxidative stress are the key factors involved in the onset and progression of pathophysiological conditions associated with various cardiovascular and renal diseases. Sodium-glucose cotransporter 2 inhibitors (SGLT2is) are glucose-lowering drugs prescribed to diabetic patients. Interestingly, apart from their glucose-lowering effect, these drugs exhibit beneficial effects in non-diabetic patients suffering from various cardiovascular and chronic kidney diseases, perhaps due to their antioxidant properties. Recently, it has been demonstrated that SGLT2is exhibit strong antioxidant properties by reducing ROS and OS. Hence, in this review, we aim to present the novel antioxidant role of SGLT2is and their consequent beneficial effects in various cardiovascular and renal disease states.
Collapse
Affiliation(s)
- Tapan A. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| | - Hong Zheng
- Division of Basic Biomedical Sciences, Sanford School of Medicine of the University of South Dakota, Vermillion, SD 57069, USA
| | - Kaushik P. Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center (UNMC), Omaha, NE 68198, USA;
| |
Collapse
|
37
|
Sharma B, Agriantonis G, Twelker K, Ebelle D, Kiernan S, Siddiqui M, Soni A, Cheerasarn S, Simon W, Jiang W, Cardona A, Chapelet J, Agathis AZ, Gamboa A, Dave J, Mestre J, Bhatia ND, Shaefee Z, Whittington J. Gut Microbiota Serves as a Crucial Independent Biomarker in Inflammatory Bowel Disease (IBD). Int J Mol Sci 2025; 26:2503. [PMID: 40141145 PMCID: PMC11942158 DOI: 10.3390/ijms26062503] [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: 01/17/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease (CD), ulcerative colitis (UC), and IBD unclassified (IBD-U), is a complex intestinal disorder influenced by genetic, environmental, and microbial factors. Recent evidence highlights the gut microbiota as a pivotal biomarker and modulator in IBD pathogenesis. Dysbiosis, characterized by reduced microbial diversity and altered composition, is a hallmark of IBD. A consistent decrease in anti-inflammatory bacteria, such as Faecalibacterium prausnitzii, and an increase in pro-inflammatory species, including Escherichia coli, have been observed. Metabolomic studies reveal decreased short-chain fatty acids (SCFAs) and secondary bile acids, critical for gut homeostasis, alongside elevated pro-inflammatory metabolites. The gut microbiota interacts with host immune pathways, influencing morphogens, glycosylation, and podoplanin (PDPN) expression. The disruption of glycosylation impairs mucosal barriers, while aberrant PDPN activity exacerbates inflammation. Additionally, microbial alterations contribute to oxidative stress, further destabilizing intestinal barriers. These molecular and cellular disruptions underscore the role of the microbiome in IBD pathophysiology. Emerging therapeutic strategies, including probiotics, prebiotics, and dietary interventions, aim to restore microbial balance and mitigate inflammation. Advanced studies on microbiota-targeted therapies reveal their potential to reduce disease severity and improve patient outcomes. Nevertheless, further research is needed to elucidate the bidirectional interactions between the gut microbiome and host immune responses and to translate these insights into clinical applications. This review consolidates current findings on the gut microbiota's role in IBD, emphasizing its diagnostic and therapeutic implications, and advocates for the continued exploration of microbiome-based interventions to combat this debilitating disease.
Collapse
Affiliation(s)
- Bharti Sharma
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - George Agriantonis
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Kate Twelker
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Danielle Ebelle
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Samantha Kiernan
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Maham Siddiqui
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Aditi Soni
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Sittha Cheerasarn
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Whenzdjyny Simon
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Winston Jiang
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Angie Cardona
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
| | - Jessica Chapelet
- Department of Medicine, St. George’s University, Grenada FZ818, West Indies; (D.E.); (M.S.); (W.S.); (J.C.)
| | - Alexandra Z. Agathis
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Alejandro Gamboa
- Department of Medicine, Medical University of the Americas, Devens, MA 01434, USA;
| | - Jasmine Dave
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Juan Mestre
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Navin D. Bhatia
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Zahra Shaefee
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| | - Jennifer Whittington
- Department of Surgery, NYC Health and Hospitals—Elmhurst, New York, NY 11373, USA; (B.S.); (G.A.); (S.K.); (S.C.); (A.C.); (J.D.); (J.M.); (N.D.B.); (Z.S.)
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (W.J.); (A.Z.A.)
| |
Collapse
|
38
|
Oh YJ, Lee JM, Yeo JH, Kim SS, Yeo SG. Induction of Nitric Oxide and Its Role in Otitis Media. Antioxidants (Basel) 2025; 14:327. [PMID: 40227356 PMCID: PMC11939237 DOI: 10.3390/antiox14030327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/26/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025] Open
Abstract
Although many studies have investigated the expression and role of nitric oxide (NO) in various diseases, it remains unclear whether NO has a beneficial or detrimental impact on otitis media. This review examines the literature on the expression and role of NO in different forms of otitis media, including acute otitis media, otitis media with effusion, chronic otitis media, and cholesteatomatous otitis media. Of the 22 studies reviewed, 18 reported that NO induces or exacerbates otitis media, whereas two studies suggested that NO may aid in its treatment. Factors contributing to these conflicting results include the type of otitis media studied, the duration of the condition, the types of samples collected, and the specific type of NO synthase targeted. Comprehensive analysis indicates that NO expression may be higher in chronic otitis media than in acute forms and is more pronounced in patients with cholesteatoma than in those without it. Although these findings suggest that NO inhibitors could potentially aid in the treatment of otitis media, NO could also aid in its treatment by inhibiting bacterial infections. Despite the dual potential of NO, current evidence suggests a strong association between NO and the pathophysiology of otitis media.
Collapse
Affiliation(s)
- Yeon Ju Oh
- Department of Medicine, College of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea;
| | - Jae Min Lee
- Department of Otorhinolaryngology Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea;
| | - Joon Hyung Yeo
- Public Health Center, Danyang-gun 27010, Chungcheongbuk-do, Republic of Korea;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung Geun Yeo
- Department of Otorhinolaryngology Head and Neck Surgery, Kyung Hee University School of Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea;
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Convergence Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
39
|
Saputro RE, Chou CC, Lin YY, Tarumi T, Liao YH. Exercise-mediated modulation of autonomic nervous system and inflammatory response in sleep-deprived individuals: A narrative reviews of implications for cardiovascular health. Auton Neurosci 2025; 259:103256. [PMID: 40073691 DOI: 10.1016/j.autneu.2025.103256] [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: 09/02/2024] [Revised: 01/25/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Sleep deprivation is a growing concern in cardiovascular risk, causing physiological disruptions like autonomic dysregulation and inflammation. Recent research indicates that sleep deprivation increases sympathetic nervous activity while decreasing parasympathetic activity, leading to increased blood pressure, impaired endothelial function, and heightened inflammation. Exercise has emerged as a non-pharmacological approach to increase cardiovascular health. However, the impact of exercise on sleep deprivation-induced changes in autonomic activity and inflammation remains unclear. To explore this, we reviewed studies investigating the effects of acute exercise on autonomic regulation and inflammatory markers following sleep deprivation. We conducted a narrative review of the literature. PubMed/MEDLINE, Google Scholar, and Web of Science (WOS) searched the articles between May 2022 and April 2023. The papers had to: [1] focus on recent studies between 2000 and 2023; [2] consist of sleep deprivation participants; [3] be published in English. Acute moderate- to high-intensity exercise after sleep deprivation may reduce parasympathetic activity, trigger pro-inflammatory cytokines, and delay recovery to normal levels. In contrast, regular exercise routines may mitigate the adverse effects of sleep deprivation on autonomic regulation and reduce systemic inflammation. Sleep deprivation can lead to autonomic imbalance, increased blood pressure, and increased inflammatory responses, which are further amplified by acute exercise, increasing the cardiovascular burden. When sleep deprivation occurs, exercise intensity and timing should be carefully chosen to avoid adverse cardiovascular health risks.
Collapse
Affiliation(s)
- Riki Edo Saputro
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan; Department of Physical Education, Universitas Wahid Hasyim, Semarang 50224, Indonesia
| | - Chun-Chung Chou
- Physical Education Office, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Yi-Yuan Lin
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan.
| | - Takashi Tarumi
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yi-Hung Liao
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan.
| |
Collapse
|
40
|
Zand A, Macharia JM, Szabó I, Gerencsér G, Molnár Á, Raposa BL, Varjas T. The Impact of Tartrazine on DNA Methylation, Histone Deacetylation, and Genomic Stability in Human Cell Lines. Nutrients 2025; 17:913. [PMID: 40077783 PMCID: PMC11902176 DOI: 10.3390/nu17050913] [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: 02/01/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Tartrazine (TRZ), a synthetic red azo dye derived from coal tar, is widely used as a food colorant in various food products, pharmaceuticals, and cosmetics. This study aims to investigate the impact of TRZ on the expression levels of DNA methyltransferases (DNMT1, DNMT3a, and DNMT3b) and histone deacetylases (HDAC5 and HDAC6). Additionally, we evaluate genomic DNA stability using the alkaline comet assay in three human cell lines: immortalized human keratinocyte (HaCaT), human hepatocellular carcinoma (HepG2), and human lung adenocarcinoma (A549). The research question focuses on whether TRZ exposure alters epigenetic regulation and DNA integrity, potentially implicating its role in carcinogenesis. METHODS The selected human cell lines were exposed to different concentrations of TRZ (20 µM, 40 µM, and 80 µM), with DMBA serving as a positive control. After treatment, we quantified the expression levels of DNMT1, DNMT3a, DNMT3b, HDAC5, and HDAC6 using quantitative real-time PCR. Additionally, we assessed DNA fragmentation via the alkaline comet assay to determine the extent of DNA damage resulting from TRZ exposure. RESULTS Our findings indicate that TRZ significantly upregulates the expression of HDAC5, HDAC6, DNMT1, DNMT3a, and DNMT3b in comparison to the control group. Furthermore, TRZ exposure leads to a notable increase in DNA damage, as evidenced by elevated tail moments across all examined human cell lines. CONCLUSIONS These results suggest that TRZ may play a role in carcinogenesis and epigenetic modifications. The observed upregulation of DNMTs and HDACs, coupled with increased DNA damage, highlights the potential risks associated with TRZ exposure. Further research is necessary to explore these mechanisms and assess their implications for human health.
Collapse
Affiliation(s)
- Afshin Zand
- Department of Public Health Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (I.S.); (G.G.); (T.V.)
| | - John M. Macharia
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary;
| | - Istvan Szabó
- Department of Public Health Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (I.S.); (G.G.); (T.V.)
| | - Gellért Gerencsér
- Department of Public Health Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (I.S.); (G.G.); (T.V.)
- Preclinical Research Center, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Ádám Molnár
- Preclinical Research Center, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Bence L. Raposa
- Institute of Basics of Health Sciences, Midwifery and Health Visiting, Faculty of Health Sciences, University of Pécs, 7621 Pécs, Hungary;
| | - Timea Varjas
- Department of Public Health Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary; (I.S.); (G.G.); (T.V.)
| |
Collapse
|
41
|
Zhu H, Uno H, Matsuba K, Hamachi I. Profiling Proteins Involved in Peroxynitrite Homeostasis Using ROS/RNS Conditional Proteomics. J Am Chem Soc 2025; 147:7305-7316. [PMID: 39988859 DOI: 10.1021/jacs.4c14060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Peroxynitrite (ONOO-), the product of the diffusion-controlled reaction of superoxide (O2•-) with nitric oxide (NO•), plays a crucial role in oxidative and nitrative stress and modulates key physiological processes such as redox signaling. While biological ONOO- is conventionally analyzed using 3-nitrotyrosine antibodies and fluorescent sensors, such probes lack specificity and sensitivity, making high-throughput and comprehensive profiling of ONOO--associated proteins challenging. In this study, we used a conditional proteomics approach to investigate ONOO- homeostasis by identifying its protein neighbors in cells. We developed Peroxynitrite-responsive protein Labeling reagents (Porp-L) and, for the first time, discovered 2,6-dichlorophenol as an ideal moiety that can be selectively and rapidly activated by ONOO- for labeling of proximal proteins. The reaction of Porp-L with ONOO- generated several short-lived reactive intermediates that can modify Tyr, His, and Lys residues on the protein surface. We have demonstrated the Porp-L-based conditional proteomics in immune-stimulated macrophages, which indeed identified proteins known to be involved in the generation and modification of ONOO- and revealed the endoplasmic reticulum (ER) as a ONOO- hot spot. Moreover, we discovered a previously unknown role for Ero1a, an ER-resident protein, in the formation of ONOO-. Overall, Porp-L represent a promising research tool for advancing our understanding of the biological roles of ONOO-.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Hiroaki Uno
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Kyoichi Matsuba
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
- ERATO (Exploratory Research for Advanced Technology, JST), Sanbancho, Chiyodaku, Tokyo 102-0075, Japan
| |
Collapse
|
42
|
Shang J, Yang Y, Sun Y, Gao W, Ma K, Wang C, Yu X, Li L, Zheng J, Zhao N, Shu X, Zhang Y. Real-time monitoring of ONOO⁻ in cerebral ischemia-reperfusion injury mouse models using a hydrazine-based NIR fluorescent probe. Redox Biol 2025; 80:103494. [PMID: 39827589 PMCID: PMC11787443 DOI: 10.1016/j.redox.2025.103494] [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: 10/20/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Accurate and selective techniques for visualizing endogenous peroxynitrite (ONOO-) in cerebral ischemia-reperfusion injury (CIRI) models are essential for understanding its complex pathological processes. Here, we introduced a longwave fluorescent probe TJO for detecting ONOO- rapidly and sensitively, with a low detection limit of 91 nM. Furthermore, TJO exhibits excellent fluorescence imaging capabilities, enabling detailed visualization of ONOO⁻ in CIRI mice model. This highlights its potential for real-time monitoring of ONOO⁻-related pathological conditions.
Collapse
Affiliation(s)
- Jinting Shang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.
| | - Yan Yang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Yaojian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Wanxia Gao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Kang Ma
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Chen Wang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Xin Yu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liping Li
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Jiang Zheng
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - Na Zhao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China
| | - XiJi Shu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China.
| | - Yibin Zhang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan, 430056, China; College of Chemistry and Chemical Engineering, Yangtze Normal University, Chongqing, China.
| |
Collapse
|
43
|
Tao G, Wang X, Wang J, Ye Y, Zhang M, Lang Y, Ding S. Identifying Specificity Protein 2 as a key marker for diabetic encephalopathy in the context of predictive, preventive, and personalized medicine. EPMA J 2025; 16:67-93. [PMID: 39991102 PMCID: PMC11842694 DOI: 10.1007/s13167-024-00394-0] [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: 09/17/2024] [Accepted: 12/18/2024] [Indexed: 02/25/2025]
Abstract
Background Transcription factor specificity protein (SP2) regulates various cellular functions, including cell division, proliferation, invasion, metastasis, differentiation, and death; however, its role has not been studied in prominent medical conditions including diabetic encephalopathy (DE). Therefore, this study addressed its physiological function in the context of DE to also better characterize its possible use in the context of predictive, preventive, and personalized medicine (PPPM). Methods The anti-inflammatory and anti-DE actions of SP2 were investigated using three animal models (SP2-/- mice, streptozocin-treated mice, and db/db mice) and two cell lines (primary cultured hippocampal neurons and N2A cells). The db/db mice were a leptin deficiency model often used to study type 2 diabetes. An equal number of males and females (8-12 weeks of age) was selected. Behavioral changes in mice were determined using both morris water maze (MWM) test and Y-maze (YM) test. The alterations in oxidative stress and inflammation were examined via immunofluorescence assay, flow cytometry, co-immunoprecipitation, and immunoblotting. Results Mechanistically, SP2-knockout (SP2-/-) mice showed dysregulation of insulin/glucose homeostasis, neuroinflammation, and cognitive loss. Otherwise, in db/db DE mice and STZ-induced DE mice, neuroinflammation, neuroapoptosis, and cognitive decline were significantly attenuated when SP2 was overexpressed in the brain. On the other hand, SP2 overexpression activates the insulin signaling pathway and improves insulin resistance via targeting X-box binding protein 1 (XBP1) in neurons. Moreover, SP2 overexpression significantly reduces oxidative stress by interacting with XBP1 and nuclear factor erythroid 2-related factor 2 (NRF2) in neurons. Furthermore, SP2 enhances the suppression of inflammatory response triggered by nuclear factor kappa B (NFκB) through the recruitment of XBP1 and NRF2 and by the in vitro inactivation of IκB kinase (IKK) complex. Conclusions These findings highlight SP2 as key biological targets for DE and reveal the infammation-related potential molecular mechanism of DE, which is helpful for early risk prediction and targeted prevention of DE. In conclusion, our study provides a new perspective for developing a PPPM method for managing DE patients. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00394-0.
Collapse
Affiliation(s)
- Guorong Tao
- Laboratory Animal Center, Fudan University, Shanghai, 200032 China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xuebao Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Jian Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
- Huangshi Love & Health Hospital, Hubei Polytechnic University, Huangshi, 435000 China
| | - Yiru Ye
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
- School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325035 Zhejiang China
| | - Minxue Zhang
- Laboratory Animal Center, Fudan University, Shanghai, 200032 China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Yan Lang
- Laboratory Animal Center, Fudan University, Shanghai, 200032 China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| | - Saidan Ding
- Laboratory Animal Center, Fudan University, Shanghai, 200032 China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 China
- Central Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang China
| |
Collapse
|
44
|
Khan AA, Dewald HD. Nitric oxide and peroxynitrite as new biomarkers for early diagnosis of autism. Brain Res 2025; 1850:149438. [PMID: 39793916 DOI: 10.1016/j.brainres.2024.149438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025]
Abstract
Autism spectrum disorder, or autism, is a neurodevelopmental disorder of the developing child's brain with a genetic causality. It can be diagnosed at about three years after birth when it begins to present itself via a range of neuropsychiatric symptoms. Nitric oxide is a crucial small molecule of life synthesized within cells of our body systems, including cells of our brain. Peroxynitrite is the product of reaction between superoxide anion and nitric oxide. It normally isomerizes into harmless nitrates or nitrites. However, when excessive superoxide anion is present, the cellular concentration of peroxynitrite can increase to a toxic level. Autism has been suggested to cause oxidative damage to brain cells. Until now, it is impossible to sample tissue from a live brain. Instead, stem cells can be derived (from an autism patient's somatic cells) which can then be differentiated and chemically directed to grow into miniature 3-dimensional tissue masses resembling specific brain regions (e.g., the cortex) called brain organoids. This review discusses utilizing nitric oxide and peroxynitrite as biomarkers and comparing their relative concentrations in stem cells and stem cell derived brain organoids of healthy and autistic individuals to develop a bioanalytical process for early diagnosis of autism.
Collapse
Affiliation(s)
- Abdullah Asif Khan
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States
| | - Howard D Dewald
- Department of Chemistry and Biochemistry, Ohio University, Athens, OH, United States.
| |
Collapse
|
45
|
Chen M, Song L, Zhou Y, Xu T, Sun T, Liu Z, Xu Z, Zhao Y, Du P, Ma Y, Huang L, Chen X, Yang G, Jing J, Shi H. Promotion of triple negative breast cancer immunotherapy by combining bioactive radicals with immune checkpoint blockade. Acta Biomater 2025; 194:305-322. [PMID: 39805523 DOI: 10.1016/j.actbio.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025]
Abstract
Although immunotherapy has revolutionized clinical cancer treatment, the efficacy is limited due to the lack of tumor-associated antigens (TAAs) and the presence of compensatory immune checkpoints. To overcome the deficiency, a nano-system loaded with ozone and CD47 inhibitor RRx-001 is designed and synthesized. Upon irradiation, reactive oxygen species (ROS) generated from ozone reacts with nitric oxide (NO) metabolized from RRx-001 to form reactive nitrogen species (RNS), which presents a much stronger cell-killing ability than ROS. Molecular mechanism studies further reveal that RNS induce extensive immunogenic cell death (ICD). The released TAAs promote infiltration of cytotoxic T lymphocytes, which provides the basis for immune checkpoint blockade (ICB) therapy. Meanwhile, RRx-001 carried by the nanoparticles and the produced radicals repolarize M2-type tumor-associated macrophages (TAMs) into the anti-tumor M1-type, consequently reversing the immunosuppressive tumor microenvironment (TME). In a xenograft triple-negative breast cancer (TNBC) animal model, O3-001@lipo (liposome enwrapping O3 and RRx-001) plus irradiation shows a significant anti-tumor efficacy by improving cytotoxic lymphocyte infiltration and regulating immunosuppressive TME. In summary, the O3-001@lipo nano-system triggered by irradiation potently improves the efficacy of immunotherapy by introducing strong cytotoxic RNS, which not only enriches the toolbox of ICD inducer but also provides a strategy of treatment for immune deficient tumor. STATEMENT OF SIGNIFICANCE: This study introduces a nano-system that leverages ozone and RRx-001 in the presence of X-ray irradiation to generate reactive nitrogen species, enhancing immunogenic cell death and promoting T-lymphocyte infiltration in triple-negative breast cancer, addressing a significant unmet need in the field. The scientific contribution is the development of a clinically translatable nano-system that not only induces ICD but also reshapes the tumor microenvironment, which is expected to have a profound impact on the readership in pharmaceutics, material science, and nano-bio interaction, particularly for those interested in advanced immune therapy approaches.
Collapse
Affiliation(s)
- Meixu Chen
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Linlin Song
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China; Department of Ultrasound & Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yao Zhou
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Tianyue Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Ting Sun
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China; Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Zhihui Liu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Zihan Xu
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Yujie Zhao
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Peixin Du
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Yingying Ma
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Liwen Huang
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China
| | - Xiaoting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Guang Yang
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Jing
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China.
| | - Hubing Shi
- Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
46
|
Bojovic D, Nikolic M, Nedeljkovic N, Vesovic M, Zivanovic A, Karovic M. Medicinal Chemistry Insights in Neuronal Nitric Oxide Synthase Inhibitors Containing Nitrogen Heterocyclic Compounds: A Mini Review. Chem Biodivers 2025; 22:e202402637. [PMID: 39436922 DOI: 10.1002/cbdv.202402637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
Many scientific reports over the last two decades have focused on the discovery and development of novel nNOS inhibitors. The structural identity of isoforms, bioavailability, pharmacokinetic, and safety profile issues remain major obstacles in the discovery of more potent and selective nNOS inhibitors. This review aims to provide an in-depth overview of the molecular interaction patterns between nNOS active site and inhibitors containing structurally diverse nitrogen heterocyclic compounds and highlight the structural properties needed to develop selective nNOS inhibitors. Previously published data allowed the usage of the structure-driven approach in the designing of selective nNOS inhibitors, which relies on the specific structural features required to achieve isoform-selectivity towards nNOS. The incorporation of chiral pyrrolidine ring, two aminopyridine heads, or a specific amino tail group, along with the inhibitor's capacity to adopt the curled conformation in the nNOS environment significantly strengthens the molecular interaction between the inhibitor and nNOS residues by forming specific electrostatic interactions and non-bonded contacts that are vital for isoform selectivity. Additional structure-activity relationship investigations are necessary to elucidate more structural characteristics that will ultimately resolve the exact structural basis required for isoform-selective inhibition of nNOS.
Collapse
Affiliation(s)
- Dijana Bojovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Milos Nikolic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Nikola Nedeljkovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Marina Vesovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Ana Zivanovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Marko Karovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| |
Collapse
|
47
|
Zuhra K, Petrosino M, Janickova L, Petric J, Ascenção K, Vignane T, Khalaf M, Philipp TM, Ravani S, Anand A, Martins V, Santos S, Erdemir S, Malkondu S, Sitek B, Kelestemur T, Kieronska-Rudek A, Majtan T, Filgueira L, Maric D, Chlopicki S, Hoogewijs D, Haskó G, Papapetropoulos A, Logue BA, Boss GR, Filipovic MR, Szabo C. Regulation of mammalian cellular metabolism by endogenous cyanide production. Nat Metab 2025; 7:531-555. [PMID: 40033006 PMCID: PMC11946912 DOI: 10.1038/s42255-025-01225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
Small, gaseous molecules such as nitric oxide, carbon monoxide and hydrogen sulfide are produced as signalling molecules in mammalian cells. Here, we show that low concentrations of cyanide are generated endogenously in various mammalian tissues and cells. We detect cyanide in several cellular compartments of human cells and in various tissues and the blood of mice. Cyanide production is stimulated by glycine, occurs at the low pH of lysosomes and requires peroxidase activity. When generated at a specific rate, cyanide exerts stimulatory effects on mitochondrial bioenergetics, cell metabolism and cell proliferation, but impairs cellular bioenergetics at high concentrations. Cyanide can modify cysteine residues via protein S-cyanylation, which is detectable basally in cells and mice, and increases in response to glycine. Low-dose cyanide supplementation exhibits cytoprotective effects in hypoxia and reoxygenation models in vitro and in vivo. Conversely, pathologically elevated cyanide production in nonketotic hyperglycinaemia is detrimental to cells. Our findings indicate that cyanide should be considered part of the same group of endogenous mammalian regulatory gasotransmitters as nitric oxide, carbon monoxide and hydrogen sulfide.
Collapse
Affiliation(s)
- Karim Zuhra
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Maria Petrosino
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Lucia Janickova
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Jovan Petric
- Leibniz Institute for Analytical Sciences, Dortmund, Germany
| | - Kelly Ascenção
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Thibaut Vignane
- Leibniz Institute for Analytical Sciences, Dortmund, Germany
| | - Moustafa Khalaf
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA
| | - Thilo M Philipp
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stella Ravani
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Abhishek Anand
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Vanessa Martins
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Sidneia Santos
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Serkan Erdemir
- Selcuk University, Science Faculty, Department of Chemistry, Konya, Turkey
| | - Sait Malkondu
- Giresun University, Faculty of Engineering, Department of Environmental Engineering, Giresun, Turkey
| | - Barbara Sitek
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Taha Kelestemur
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Anna Kieronska-Rudek
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Tomas Majtan
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Luis Filgueira
- Section of Anatomy, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Darko Maric
- Department of Endocrinology, Metabolism and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - David Hoogewijs
- Department of Endocrinology, Metabolism and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Brian A Logue
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA
| | - Gerry R Boss
- Department of Medicine, University of California, San Diego, San Diego, CA, USA
| | - Milos R Filipovic
- Leibniz Institute for Analytical Sciences, Dortmund, Germany.
- School of Molecular Biosciences, University of Glasgow, Glasgow, UK.
| | - Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
48
|
Kanipe C, Putz EJ, Palmer MV. Differential expression of vascular endothelial growth factor A (VEGFA) and M1 macrophage marker nitric oxide synthase 2 (NOS2) in lymph node granulomas of BCG-vaccinated and non-vaccinated cattle infected with Mycobacterium bovis. Tuberculosis (Edinb) 2025; 151:102609. [PMID: 39862443 DOI: 10.1016/j.tube.2025.102609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Bovine tuberculosis is mainly caused by Mycobacterium bovis. Bacillus Calmette-Guérin (BCG) is an attenuated strain of M. bovis which provides variable disease protection. Lesions have been characterized in infected cattle, but little comparison has been done with lesions which form in BCG-vaccinates. Here, in situ hybridization examined differences in expression of M. bovis RNA, inducible nitric oxide synthase 2, and vascular endothelial growth factor A in relation to vaccination status and granuloma grade, using two different groups of cattle. Data found no differences between vaccination groups or granuloma grade in average copies of M. bovis mRNA per μm2 of total granuloma area or per μm2 of necrotic areas. Within a vaccination group high-grade granulomas had more NOS2 per cell, per μm2 and a higher percentage of cells expressing NOS2 than low-grade granulomas. Non-vaccinates had a higher percentage of cells producing NOS2 than vaccinates. Differences in NOS2 expression varied by group. Vaccination status and granuloma grade did not affect the average copies of VEGFA per cell or the percent of cells expressing RNA, however VEGFA copies per μm2 varied between groups. These findings suggest NOS2 and VEGFA are likely not mechanisms of BCG vaccination protection but may impact disease severity.
Collapse
Affiliation(s)
- C Kanipe
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA; Immunobiology Program, Iowa State University, Ames, IA, 50010, USA.
| | - E J Putz
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA
| | - M V Palmer
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, Iowa, USA
| |
Collapse
|
49
|
Calligaris M, Aleksova A, Fluca AL, Janjusevic M, Carpi G, Stefanizzi D, Carnevali S, Curcio F, Puca AA, Cattaneo M, Beltrami AP. Protective role of the longevity-associated BPIFB4 gene on cardiac microvascular cells and cardiac aging. Vascul Pharmacol 2025; 158:107470. [PMID: 39909151 DOI: 10.1016/j.vph.2025.107470] [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: 12/20/2024] [Revised: 01/31/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
In recent years, the role of the cardiac microvasculature in modulating the symptoms and disease progression of patients affected by cardiac pathology has been reconsidered. The term cardiac microvascular disease (CMD) describes the set of functional and/or structural alterations of the cardiac microvasculature that reduce the ability of the heart to adequately increase its coronary blood flow to keep up with increased metabolic demand. CMD is involved in the evolution of heart disease of both ischemic and non-ischemic origin as well as in cardiac aging. The primary actors involved in this process are the cells of the stromal compartment, whose nature and biology are now investigated to a new level of detail thanks to single-cell omics studies. Recent studies on the genetics of extreme longevity have identified a polymorphic haplotype variant of the BPIFB4 gene that confers prolonged life span and health span, atheroprotective advantages, and an improved immune response. The aim of this review was to focus on the beneficial effects of the longevity-associated variant (LAV) of BPIFB4 on cardiac microvascular cell biology, providing novel and exciting mechanisms of its action directed against the development or progression of many age-related cardiovascular diseases, thus emphasizing its translational therapeutic potential.
Collapse
Affiliation(s)
| | - Aneta Aleksova
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy; Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Alessandra Lucia Fluca
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy
| | - Milijana Janjusevic
- Department of Medical Surgical and Health Sciences of University of Trieste, Trieste, Italy
| | - Giada Carpi
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | | | | | - Francesco Curcio
- Department of Medicine (DMED), University of Udine, Udine, Italy; Laboratory Medicine Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy
| | - Annibale Alessandro Puca
- IRCCS MultiMedica, Milan, Italy; Department of Medicine, Surgery and Dentistry, "Scuola Medica Salernitana", Università degli Studi di Salerno, Salerno, Italy.
| | | | - Antonio Paolo Beltrami
- Department of Medicine (DMED), University of Udine, Udine, Italy; Laboratory Medicine Department, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy.
| |
Collapse
|
50
|
Abukhalil MH, Al-Alami Z, Alfwuaires MA, Imran MR, Aladaileh SH, Althunibat OY. Taxifolin Protects Against 5-Fluorouracil-Induced Cardiotoxicity in Mice Through Mitigating Oxidative Stress, Inflammation, and Apoptosis: Possible Involvement of Sirt1/Nrf2/HO-1 Signaling. Cardiovasc Toxicol 2025; 25:455-470. [PMID: 39827225 DOI: 10.1007/s12012-025-09962-w] [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: 10/09/2024] [Accepted: 01/12/2025] [Indexed: 01/22/2025]
Abstract
Although 5-fluorouracil (5-FU) is widely utilized in cancer treatment, its side effects, including cardiotoxicity, limit its use. Taxifolin (TAX) is a bioactive anti-inflammatory and antioxidant flavonoid. This study aimed to elucidate the protective effect of TAX against 5-FU-induced cardiac injury in male mice. Mice were treated with TAX (25 and 50 mg/kg, orally) for 10 days and a single dose of 150 mg/kg 5-FU at day 8. Mice intoxicated with 5-FU showed increased creatine kinase-MB and lactate dehydrogenase activities and troponin I levels, with multiple cardiac histopathological changes. They also showed a significant increase in cardiac malondialdehyde (MDA) and nitric oxide (NO) and decreases in myocardial reduced glutathione (GSH) content and superoxide dismutase (SOD) and catalase (CAT) activities (P < 0.001). Pretreatment of 5-FU-injected mice with TAX suppressed cardiac injury, decreased MDA and NO contents (P < 0.001), and boosted antioxidant defenses in the myocardium. Moreover, TAX attenuated cardiac inflammatory response, as evidenced by the decreased expression levels of cardiac NF-κB p65, inducible nitric oxide synthase (iNOS), and pro-inflammatory cytokines (P < 0.001). Largely, TAX ameliorated the decrease in Bcl-2 expression and the increase in BAX and caspase-3 in the heart. It also restored the cardiac Sirt1/Nrf2/HO-1 signaling pathway. In conclusion, TAX showed significant cardioprotective effects on 5-FU-induced cardiac injury and might represent a promising adjuvant in preventing cardiac injury associated with oxidative stress and inflammation.
Collapse
Affiliation(s)
- Mohammad H Abukhalil
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan.
- Department of Biology, College of Science, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan.
| | - Zina Al-Alami
- Department of Basic Medical Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Manal A Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, 31982, Al-Ahsa, Saudi Arabia
| | - Mohd Rasheeduddin Imran
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39553, Hafr Al Batin, Saudi Arabia
| | - Saleem H Aladaileh
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Al Batin, 39553, Hafr Al Batin, Saudi Arabia
| | - Osama Y Althunibat
- Department of Medical Analysis, Princess Aisha Bint Al-Hussein College of Nursing and Health Sciences, Al-Hussein Bin Talal University, Ma'an, 71111, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Jadara University, Irbid, 21110, Jordan
| |
Collapse
|