1
|
Zhang M, Zhou S, Zhang T, Li J, Xue L, Liang B, Xing D. Shark skin and mussel-inspired polyurethane hydrogel sponge for wounds with infection and exudate. J Colloid Interface Sci 2025; 693:137658. [PMID: 40279845 DOI: 10.1016/j.jcis.2025.137658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Inspired by the antifouling properties of shark skin and the bioadhesion of mussels, our study presents a three-layer biomimetic wound dressing with hierarchical wettability and rapid exudate drainage capabilities. The shark skin-inspired hydrophobic modified polyurethane (PU) sponge provides antifouling properties and serves as a bacterial barrier. The mussel-inspired dopamine-functionalized carboxymethyl chitosan hydrogel (CMCS-DOP) absorbs exudates and forms an in situ hydrogel, effectively capturing and eliminating bacteria. The porous sponge layer in direct contact with the wound facilitates rapid exudate drainage, preventing excessive wound hydration. This hierarchical structure coordinates exudate transport and bacterial removal. The fabricated PCD hydrogel sponge dressing (PCD dressing) exhibits a wettability transition (contact angle: 3°-35°-101°) and a water vapor transmission rate of 1021-797-691 g/m2. It demonstrates potent bactericidal effects against Staphylococcus aureus and Escherichia coli, with survival rates of only 13 % and 14 %, respectively, and bacterial-blocking efficiencies of 89 % and 94 %. In a chronic bacterial infection wound model, the PCD dressing outperforms conventional clinical dressings, increasing the wound healing rate by 25.8 %, reducing inflammation, and enhancing angiogenesis and collagen deposition. Notably, the PCD mitigates oxidative stress at the wound site by regulating the polarization of anti-inflammatory macrophages. This exudate-draining and responsive dressing offers a promising strategy for promoting the healing of wounds with high exudate levels.
Collapse
Affiliation(s)
- Miao Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Sha Zhou
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Basic Medicine, Qingdao University, Qingdao, Shandong 266000, China
| | - Tingting Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Jiyixuan Li
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Basic Medicine, Qingdao University, Qingdao, Shandong 266000, China
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; School of Pharmacy, Qingdao University, Qingdao, Shandong 266000, China
| | - Bing Liang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
2
|
Huang Y, Hu T, Li S, Zhou W. Ferrihydrite/B, N co-doped biochar composites enhancing tetracycline degradation: The crucial role of boron incorporation in Fe(III) reduction and oxygen activation. J Environ Sci (China) 2025; 154:252-263. [PMID: 40049871 DOI: 10.1016/j.jes.2024.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 05/13/2025]
Abstract
Harnessing the redox potential of biochar to activate airborne O2 for contaminant removal is challenging. In this study, ferrihydrite (Fh) modified the boron (B), nitrogen (N) co-doped biochars (BCs) composites (Fh/B(n)NC) were developed for enhancing the degradation of a model pollutant, tetracycline (TC), merely by airborne O2. Fh/B(3)NC showed excellent O2 activation activity for efficient TC degradation with a apparent TC degradation rate of 5.54, 6.88, and 22.15 times that of B(3)NC, Fh, and raw BCs, respectively, where 1O2 and H2O2 were identified as the dominant ROS for TC degradation. The B incorporation into the carbon lattice of Fh/B(3)NC promoted the generation of electron donors, sp2 C and the reductive B species, hence boosting Fe(III) reduction and 1O2 generation. O2 adsorption was enhanced due to the positively charged adsorption sites (C-B+and NC+). And 1O2 was generated via Fe(II) catalyzed low-efficient successive one-electron transfer (O2 → O2·- → 1O2, H2O2), as well as biochar catalyzed high-efficient two-electron transfer (O2 → H2O2 → 1O2) that does not involve .O2- as the intermediate. Moreover, Fh/B, N co-doped biochar showed a wide pH range, remarkable anti-interference capabilities, and effective detoxification. These findings shed new light on the development of environmentally benign BCs materials capable of degradading organic pollutants.
Collapse
Affiliation(s)
- Yujiang Huang
- Department of Environmental Science, Zhejiang University, Hangzhou 310058, China
| | - Tong Hu
- Department of Environmental Science, Zhejiang University, Hangzhou 310058, China
| | - Sichen Li
- Department of Environmental Science, Zhejiang University, Hangzhou 310058, China
| | - Wenjun Zhou
- Department of Environmental Science, Zhejiang University, Hangzhou 310058, China; The Key Laboratory of Organic Pollution Process and Control, Zhejiang Province, Hangzhou 310058, China; Zhejiang Ecological Civilization Academy, Anji 313300, China.
| |
Collapse
|
3
|
Wang L, Yang M, Gao R, Pang Y, Zhao X, Zhou G, Gao S, Ge K, Zhang J. Thermal responsive nanobombs generating reactive oxygen species for synergistic anticancer therapy. J Colloid Interface Sci 2025; 687:607-616. [PMID: 39978266 DOI: 10.1016/j.jcis.2025.02.091] [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: 10/07/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
The nano-based therapeutics to induce cellular oxidative damage is considered promising in cancer treatment. Photodynamic therapy (PDT) is a primary antitumor oxidative damage treatment method. However, the hypoxic environment of tumor tissues and the short lifetime of singlet oxygen significantly hampers PDT efficacy. Fortunately, nitric oxide (NO), as a form of gas therapy, can generate more toxic oxidative peroxynitrite ions (ONOO-) with hydrogen peroxide (H2O2), which significantly enhance the efficacy of PDT. In this context, we fabricated a thermally controlled reactive oxygen nanobombs CaO2@LA-ICG@TD (CAI@TD), which can release many reactive oxygen species (ROS) to enhance the synergistic anticancer efficiency under a. The cellular studies revealed that CAI@TD could produce oxygen and H2O2 to heighten the efficacy of PDT and NO and induce necrotic-apoptosis of MDA-MB-231 cells by mitochondria damage, lipid peroxidation, and DNA fragments. Moreover, CAI@TD with 808 nm laser irradiation achieved a significant inhibition on the xenograft tumor growth. This work provides an efficient strategy to produce a high amount of ROS for synergistic anticancer therapy, offering a ray of hope in the fight against cancer.
Collapse
Affiliation(s)
- Li Wang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Mengzhen Yang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Ruijing Gao
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Yu Pang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Xiaoshu Zhao
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China
| | - Guoqiang Zhou
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China; College of Basic Medical Science, Hebei University, Baoding 071000, China
| | - Shutao Gao
- College of Science, Hebei Agricultural University, Baoding 071002, China.
| | - Kun Ge
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China.
| | - Jinchao Zhang
- College of Chemistry & Materials Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, State Key Laboratory of New Pharmaceutical Preparations and Excipients, Chemical Biology Key Laboratory of Hebei Province, Hebei University, Baoding 071002, China.
| |
Collapse
|
4
|
Rohaun SK, Chakraborti PK. Oxidation of active site cysteine leads to inactivation of peptide deformylase from Salmonella enterica. Biochem Biophys Res Commun 2025; 759:151675. [PMID: 40147353 DOI: 10.1016/j.bbrc.2025.151675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
Peptide deformylase (PDF) is an essential bacterial enzyme involved in first step of N-methionine excision (NME) pathway during bacterial protein synthesis. In first step of NME, N-formyl group of nascent polypeptide chains is removed by PDF. PDF is a metallo-protease where metal cofactor is co-ordinated to a Cys and two His residues. We cloned and expressed this iron containing metallo-protease from Salmonella typhimurium (sPDF). We characterized the sPDF which is a mononuclear iron containing enzyme that displayed optimal in vitro activity in the presence of oxidation preventing agent like catalase. To have an insight into the role of metal ion in catalase dependent enzyme activity, we generated surrogate sPDF-Ni2+ and sPDF-Co2+. Interestingly, these proteins also showed catalase requirement for optimum enzyme activity. Thus our results argue the presence of the target (amino acid) of oxidation in the protein itself that might be crucial for the activity of the enzyme. To ascertain this aspect, we examined the oxidation status of active site cysteine of sPDF by mass spectrometry. Our results indicated that the direct oxidation/over-oxidation of active site cysteine is responsible for inactivation of sPDF protein. Furthermore, a comparison of PDF sequences from Gram-negative bacteria revealed the presence of this cysteine throughout the lineage. Thus, our results per se are indicative of a similar behaviour of all these Gram-negative peptide deformylase proteins.
Collapse
|
5
|
Yan J, Bao L, Liang H, Zhao L, Liu M, Kong L, Fan X, Liang C, Liu T, Han X, Wang K, Shen C, Sun W, Zhou X, Chu B, McGlinchey MJ, Xu X, Qiu X, Wang Y. A Druglike Ferrostatin-1 Analogue as a Ferroptosis Inhibitor and Photoluminescent Indicator. Angew Chem Int Ed Engl 2025; 64:e202502195. [PMID: 39984310 DOI: 10.1002/anie.202502195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 02/23/2025]
Abstract
Redox-based diagnostic and therapeutic applications have long suffered from a shortage of suitable drugs and probes of high specificity. In the context of anti-ferroptosis research for neurological diseases, the inaccessibility of a blood-brain barrier (BBB) permeable small molecular ferroptosis inhibitor, and the lack of specific ferroptosis probes, seriously impeded a deeper understanding of the mechanism of ferroptosis and the development of clinically applicable drugs. We report herein a novel 1,3,4-thiadiazole-functionalized druglike ferrostatin analogue entitled Ferfluor-1 with superior anti-ferroptosis potency, favorable BBB permeability and in vivo activity against stroke and Parkinson's disease. Moreover, the exclusive pseudo-excited-state intramolecular proton-transfer (ESIPT) property of Ferfluor-1 via a long-distance hydrogen-bonding network makes it the first sensitive ratiometric photoluminescent probe to detect phospholipid hydroperoxides and a specific indicator for the fluctuation of ferroptosis. These unprecedented advantages not only make Ferfluor-1 a potential tool for ferroptosis-related diagnostic and therapeutic applications in the central nervous system, but also pave the way to developing new theragnostic agents for precision redox detection and regulation.
Collapse
Affiliation(s)
- Jiangkun Yan
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Luo Bao
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Huicong Liang
- Marine Biomedical Research Institute of Qingdao, School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Chinese Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, P. R. China
| | - Li Zhao
- College of Science, China University of Petroleum (East China), Qingdao, 266580, Shandong, P. R. China
| | - Ming Liu
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Lingxiu Kong
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Xuejing Fan
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Chunhui Liang
- School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, P. R. China
| | - Tongtong Liu
- Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, P. R. China
| | - Xinyu Han
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Kangnan Wang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, P. R. China
| | - Chuanbin Shen
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Wanyang Sun
- Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research, Jinan University, Guangzhou, 510632, P. R. China
| | - Xin Zhou
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266003, P. R. China
| | - Bo Chu
- School of Basic Medical Science, Shandong University, Jinan, Shandong, 250012, P. R. China
| | | | - Ximing Xu
- Marine Biomedical Research Institute of Qingdao, School of Medicine and Pharmacy, Key Laboratory of Marine Drugs, Chinese Ministry of Education, Ocean University of China, Qingdao, 266003, Shandong, P. R. China
| | - Xue Qiu
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Yong Wang
- Key Laboratory of Marine Drug, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| |
Collapse
|
6
|
Liu M, Liu H, Yang Y, Xiong X, Zou T. Subcellular Photocatalysis Enables Tumor-Targeted Inhibition of Thioredoxin Reductase I by Organogold(I) Complexes. J Am Chem Soc 2025; 147:15719-15731. [PMID: 40272019 DOI: 10.1021/jacs.5c03186] [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: 04/25/2025]
Abstract
Selective inhibition of TrxR1 over TrxR2 is a highly sought-after goal, because the two enzymes play distinct roles in cancer progression. However, achieving targeted inhibition is challenging due to their high homology and identical active site sequence. Herein we report a new subcellular photocatalysis approach for targeted inhibition by controllably activating organogold(I) prodrugs within the cytosol, the exclusive location of TrxR1. The NHC-Au(I)-alkynyl complexes are stable and evenly distributed in the cell; they can meanwhile be efficiently transformed into active NHC-Au(I)-L species (L = labile ligands) via a radical mechanism by photocatalysts released into the cytosol (from endosome/lysosome) upon light irradiation, leading to selective inhibition of TrxR1 without affecting TrxR2. This results in strong cytotoxicity to cancer cells with much higher selectivity than auranofin, a pan TrxR inhibitor that cannot discriminate TrxR1/2, along with potent antitumor activities in multiple zebrafish and mouse models. This subcellular prodrug activation may thus suggest a novel approach to precision targeting using the remarkable spatial control of photocatalysis.
Collapse
Affiliation(s)
- Moyi Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Haitao Liu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Yan Yang
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Xiaolin Xiong
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| | - Taotao Zou
- State Key Laboratory of Anti-Infective Drug Discovery and Development, Guangdong Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P. R. China
| |
Collapse
|
7
|
Zhang K, Xiao Q, Jia J, Gao Y, Zhou Y, Zhou XX, Yan B. Mechanistic Insights into Cadmium Sulfide Nanoparticles-Induced Digestive Gland Damage in Corbicula fluminea: Comparison with Cadmium Ions. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:8451-8460. [PMID: 40165425 DOI: 10.1021/acs.est.5c01150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The extensive use of cadmium sulfide nanoparticles (CdS-NPs), along with their natural formation through the complex biogeochemical transformation of anthropogenic cadmium ions (Cd2+), poses substantial risks to ecosystems and human health. Despite this, the mechanisms underlying the toxicity of CdS-NPs remain unclear. A key question is whether their toxicity arises from the nanoparticulate form of cadmium (Cd) or from the release of Cd2+. To explore this, we exposed freshwater clams (Corbicula fluminea) to environmentally relevant concentrations (0.01-1 mg/L) of CdS-NPs or Cd2+ for 10 days. Hematoxylin and eosin (HE) staining revealed significant damage to the digestive gland in both cases. Although CdS-NPs released some Cd2+ (≤10.4%), transcriptomic and quantitative reverse transcription polymerase chain reaction (qRT-PCR) analyses indicated different toxicity mechanisms. CdS-NPs primarily induce ferroptosis, triggered by lysosomal dysfunction that releases Fe2+ into the cytoplasm, disrupting the cellular iron metabolism. In contrast, Cd2+ primarily induces an autophagic response, as evidenced by the upregulation of autophagy-related markers and activation of apoptosis pathways linked to mitochondrial membrane permeabilization. Overall, our findings suggest that the toxicity of CdS-NPs is not solely derived from Cd2+, highlighting the need to evaluate the risks posed by metal sulfide nanoparticles to benthic ecosystems.
Collapse
Affiliation(s)
- Kena Zhang
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Quanzhi Xiao
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Jianbo Jia
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yan Gao
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Yanfei Zhou
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Xiao-Xia Zhou
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Bing Yan
- School of Environmental Science and Engineering, Shandong University, Qingdao 266237, China
- Institute of Environmental Research at the Greater Bay Area, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| |
Collapse
|
8
|
Xie S, Wang Y, Zhao C, Cui Y, Gu C, Wu W. Putrescine promotes maturation of oocytes from reproductively old mice via mitochondrial autophagy. Reprod Biomed Online 2025; 50:104495. [PMID: 40068351 DOI: 10.1016/j.rbmo.2024.104495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/25/2024] [Accepted: 10/14/2024] [Indexed: 05/12/2025]
Abstract
RESEARCH QUESTION Does putrescine (PUT) improve oocytes from reproductively old mice by promoting mitochondrial autophagy? DESIGN Germinal vesicle stage cumulus-oocyte complexes (COCs) were obtained from 9-month old female C57BL/6N mice and divided into control, PUT and difluoromethylornithine, inhibitor (DFMO) groups. These germinal vesicle COCs underwent mouse in-vitro maturation (IVM) culture to observe the extrusion of the first polar body in each group. Using JC-1, dichloro-dihydro-fluorescein diacetate fluorescent probes and a confocal microscope, the mitochondrial membrane potential integrity and reactive oxygen species levels were measured in metaphase II stage oocytes. The expression and cellular localization of the p53 protein were examined by immunofluorescence. Reverse transcription quantitative polymerase chain reaction was used to detect the activation of mitochondrial autophagy pathways. The potential mechanisms through which PUT improves oocytes from reproductively old mice were explored by single-cell transcriptomic analysis. Autophagosomes, autolysosomes and mitochondria in different groups were directly observed using transmission electron microscopy. RESULTS The addition of exogenous PUT can promote IVM of oocytes from reproductively old mice. It reduces oxidative stress by promoting the autophagy of damaged mitochondria, decreasing the levels of reactive oxygen species and increasing mitochondrial membrane potential. It affects the expression and subcellular localization of the p53 protein, and increases the expression of transcription factor EB, which may be the potential mechanism behind its promotion of autophagy. CONCLUSION The target and regulatory pathway of PUT in oocytes was clarified. Putrescine is an effective small molecule compound with significant potential for non-invasively improving the fertility of elderly women.
Collapse
Affiliation(s)
- Siyuan Xie
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China.; School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuyi Wang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chenxi Zhao
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Yugui Cui
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China
| | - Chunyan Gu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Clinical Center of Reproductive Medicine, First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu Province, China..
| |
Collapse
|
9
|
Wang C, Jin H, Wang C, Wu J, Meng Q, Zhong M, Sun H, Wei Y, Gao G, Kaku T, Huo X, Liu K. Molecular pharmacokinetic mechanism of JBP485 against aristolochic acid I (AAI) -induced nephrotoxicity. Front Pharmacol 2025; 16:1577942. [PMID: 40371353 PMCID: PMC12074917 DOI: 10.3389/fphar.2025.1577942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction In this study, we investigated the protective effect of JBP485 against aristolochic acid I (AAI)-induced nephrotoxicity and explored the pharmacokinetic mechanisms. The effects of JBP485 on AAI-induced cytotoxicity and nephrotoxicity were evaluated in vitro and in vivo, respectively. Methods To ascertain the protective effect of JBP485 against AAI-induced nephrotoxicity, we measured levels of urea nitrogen (BUN), creatinine (CRE), and indoxol sulfate in blood and urine; determined kidney weight-to-body weight ratio; and performed hematoxylin and eosin (H&E) staining. Cell viability and Western blotting assays, along with determination of malondialdehyde (MDA), superoxide dismutase (SOD), and intracellular reactive oxygen species (ROS) contents, were carried out to explore mechanisms underlying the protective effects of JBP485 against AAI-induced nephrotoxicity. Results JBP485 treatment attenuated AAI-induced injuries in rat kidney while decreasing the levels of indoxyl sulfate, CRE, and BUN in plasma and increasing those of indoxyl sulfate in urine compared to that in AAI alone-treated group. The co-administration of JBP485 with AAI significantly increased the concentration and AUC of AAI in plasma, while decreasing its cumulative urinary excretion and renal clearance. Moreover, JBP485 reduced the uptake of AAI in kidney slices and human organic anion transporter 1/3 (hOAT1/3)-transfected human embryonic kidney 293 (HEK293) cells, suggesting that JBP485 ameliorated AAI-induced nephrotoxicity by reducing renal exposure to AAI via OAT inhibition. Meanwhile, JBP485 modulated the abnormal expressions of Oat1, Oat3, organic cation transporter 2 (Oct2), P-glycoprotein (P-gp), multidrug resistance-associated protein 2 (Mrp2) and multidrug and toxin extrusion proteins 1 (Mate 1) in rat kidney, suggesting that JBP485 improved tubular secretion in AAI-treated rats. Moreover, JBP485 reversed the AAI-induced changes in the expression of heme oxygenase 1 (HO-1), NAD(P) H: quinone oxidoreductase-1 (NQO1), B-cell lymphoma-2 (Bcl-2) protein expressions and Bcl-2-like protein 4 (Bax) induced by AAI in rat kidney. JBP485 increased cell viability and reduced intracellular levels of ROS in NRK-52E cells treated with AAI. Discussion These results suggested that JBP485 protected against AAI-induced renal oxidative stress. All results indicated that JBP485 protected against AAI-induced nephrotoxicity by reducing renal exposure to AAI and alleviating oxidative stress. Our findings suggested that JBP485 has potential as a renoprotective agent for the prevention of AAI-induced nephrotoxicity.
Collapse
Affiliation(s)
- Chong Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Huan Jin
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Ming Zhong
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Yuheng Wei
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Ge Gao
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| | - Taiichi Kaku
- Japan Bioproducts Industry Co. Ltd., Tomigaya, Shibuya-ku, Tokyo, Japan
| | - Xiaokui Huo
- Pharmaceutical Research Center, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
- Provincial Key Laboratory for Pharmacokinetics and Transport, Liaoning Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
10
|
Lee JD, Nguyen A, Gibbs CE, Jin ZR, Wang Y, Moghadasi A, Wait SJ, Choi H, Evitts KM, Asencio A, Bremner SB, Zuniga S, Chavan V, Pranoto IKA, Williams CA, Smith A, Moussavi-Harami F, Regnier M, Baker D, Young JE, Mack DL, Nance E, Boyle PM, Berndt A. Monitoring in real time and far-red imaging of H 2O 2 dynamics with subcellular resolution. Nat Chem Biol 2025:10.1038/s41589-025-01891-7. [PMID: 40295764 DOI: 10.1038/s41589-025-01891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
Monitoring H2O2 dynamics in conjunction with key biological interactants is critical for elucidating the physiological outcome of cellular redox regulation. Optogenetic hydrogen peroxide sensor with HaloTag with JF635 (oROS-HT635) allows fast and sensitive chemigenetic far-red H2O2 imaging while overcoming drawbacks of existing red fluorescent H2O2 indicators, including oxygen dependency, high pH sensitivity, photoartifacts and intracellular aggregation. The compatibility of oROS-HT635 with blue-green-shifted optical tools allows versatile optogenetic dissection of redox biology. In addition, targeted expression of oROS-HT635 and multiplexed H2O2 imaging enables spatially resolved imaging of H2O2 targeting the plasma membrane and neighboring cells. Here we present multiplexed use cases of oROS-HT635 with other green fluorescence reporters by capturing acute and real-time changes in H2O2 with intracellular redox potential and Ca2+ levels in response to auranofin, an inhibitor of antioxidative enzymes, via dual-color imaging. oROS-HT635 enables detailed insights into intricate intracellular and intercellular H2O2 dynamics, along with their interactants, through spatially resolved, far-red H2O2 imaging in real time.
Collapse
Affiliation(s)
- Justin Daho Lee
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Amanda Nguyen
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chelsea E Gibbs
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Zheyu Ruby Jin
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Yuxuan Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Aida Moghadasi
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Sarah J Wait
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Hojun Choi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kira M Evitts
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Anthony Asencio
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Samantha B Bremner
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Shani Zuniga
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Vedant Chavan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Inez K A Pranoto
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - C Andrew Williams
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Annette Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Jessica E Young
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - David L Mack
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Elizabeth Nance
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Andre Berndt
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA.
| |
Collapse
|
11
|
Shen S, Yuan Y, Song J, Zhu Y, Wang Y, Yue C, Du M, Wei J, Feng F, Tian M. A phenothiazine-based ratiometric fluorescent probe for detecting hypochlorite (ClO -) and its application in foods and water samples. Food Chem 2025; 485:144547. [PMID: 40311560 DOI: 10.1016/j.foodchem.2025.144547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/09/2025] [Accepted: 04/25/2025] [Indexed: 05/03/2025]
Abstract
As a significant reactive oxygen species (ROS), ClO- plays versatile roles in daily life and many biological events. However, its abnormal levels are responsible for serious harm to human health. Therefore, it is of crucial interest to develop effective methods for detecting ClO- in foods and living organisms. In this work, a novel fluorescent probe ethyl 2-(3-formyl-2-methoxy-10H-phenothiazin-10-yl)acetate (PEA) for detecting ClO- was presented. It shows the merits of rapid ratiometric response (within 10 s), high selectivity and very large Stokes shift (190 nm). The detection limit of PEA for ClO- was determined to be 0.47 μM. We not only successfully prepared paper test strips for efficient qualitative naked eye ClO- detection, but also demonstrated its potential for the valid detection of ClO- in natural water samples, beverages and foods. Furthermore, the probe PEA was also applied to the fluorescence imaging of ClO- in onion epidermal cells.
Collapse
Affiliation(s)
- Siyi Shen
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Yuehua Yuan
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China.
| | - Jinping Song
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Yongjun Zhu
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Yuzhen Wang
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Chaoyi Yue
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Mengqing Du
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Jiyuan Wei
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China
| | - Feng Feng
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China.
| | - Maozhong Tian
- College of Chemistry and Chemical Engineering, Shanxi Provincial Key Laboratory of Chemical Biosensing, Shanxi Datong University, Datong 037009, PR China.
| |
Collapse
|
12
|
Yang J. Towards site-specific manipulation in cysteine-mediated redox signaling. Chem Sci 2025:d5sc02016f. [PMID: 40321179 PMCID: PMC12046419 DOI: 10.1039/d5sc02016f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Cysteine sulfenic acid (SOH) modifications are pivotal in redox signaling, yet establishing their causal biological roles remains challenging due to methodological limitations. Traditional approaches often lack precision or disrupt non-redox cysteine functions. This perspective highlights two innovative chemical biology strategies to address these challenges: (1) integrating bioorthogonal cleavage chemistry with genetic code expansion for site-specific SOH incorporation in proteins of interest, enabling controlled activation of redox events, and (2) developing redox-targeted covalent inhibitors (TCIs) to selectively block SOH modifications. By bridging technological innovation with mechanistic inquiry, these strategies not only help elucidate SOH-mediated signaling networks for a better understanding of redox biology, but also hold therapeutic promise for precise redox medicine.
Collapse
Affiliation(s)
- Jing Yang
- Guangzhou National Laboratory, Guangzhou International Bio-Island Guangzhou China
- School of Pharmaceutical Sciences, Guangzhou Medical University Guangzhou China
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences - Beijing, Beijing Institute of Lifeomics Beijing China
| |
Collapse
|
13
|
Liu F, Guo L, Luo Y, Li J, Zhou Y, Wang J, Huang X, Tan X, Fu M, Yu B, Gao Y, Liu R, Takaya N, Zhou S. Peroxiredoxin PrxA and thioredoxin TrxA mediate the redox signal to the transcription factor NapA in the fungus Aspergillus nidulans. Int J Biol Macromol 2025; 310:143434. [PMID: 40274147 DOI: 10.1016/j.ijbiomac.2025.143434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/08/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Ap-1-like transcription factors play a crucial role in regulating antioxidant gene expression and protecting cells from oxidative stress. Extensive research on redox regulation in Saccharomyces cerevisiae and Schizosaccharomyces pombe has revealed notable differences in their mechanisms. However, it remains unclear whether filamentous fungi share similarities with either yeast system or employ a distinct signaling strategy. To address this, we investigated the redox signal relay in Aspergillus nidulans, focusing on how peroxiredoxin PrxA and thioredoxin TrxA modulate the activity of the Ap-1-like transcription factor NapA. We demonstrate that PrxA is essential for NapA activation, transmitting the H₂O₂ signal through a disulfide bond between its peroxidatic and resolving cysteines to NapA's Cys558, which subsequently forms an intramolecular disulfide bond with Cys404. Furthermore, we reveal that TrxA, rather than Txl1, is responsible for reducing and inactivating NapA by direct interaction in both the cytoplasm and nucleus, utilizing its own catalytic cysteine residues. These findings establish a mechanistic framework for NapA activation and reduction, providing new insights into oxidative stress responses in filamentous fungi and their divergence from yeast systems.
Collapse
Affiliation(s)
- Feiyun Liu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Lingyan Guo
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yiqing Luo
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Jingyi Li
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yao Zhou
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Jing Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaofei Huang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Xinyu Tan
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Mingxin Fu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Bingzi Yu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Yan Gao
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Renning Liu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China
| | - Naoki Takaya
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Shengmin Zhou
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.
| |
Collapse
|
14
|
Chang J, Zhang Z, Qu C, Han Q, Xu L. Organic Molecules as a Bridge Connecting Photoelectrochemistry and Fluorescence for Dual-Signal Assay. Anal Chem 2025; 97:7842-7850. [PMID: 40177944 DOI: 10.1021/acs.analchem.4c06431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
We report a strategy based on pyridyl-anchored organic small-molecule fluorescent probes to develop a dual-signal sensing platform. The strategy accomplishes an intelligent integration of fluorescence analysis with photoelectrochemical (PEC) sensing, thereby enabling rapid and precise detection of hypochlorite. In this work, the natural dye chromone was selected as the fluorophore for generating fluorescent signals. Meanwhile, by using phenothiazine (PTZ) as the specific recognition group and pyridine as the anchoring moiety, we designed and synthesized a novel organic small-molecule fluorescent probe. The obtained probe was used as a photosensitive material anchored to the TiO2 surface via N → Ti bonds, to form an FTO/TiO2/FPTZ-1 heterostructure-based dual-signal sensing platform for the detection of hypochlorite. This sensing platform has the characteristics of high specificity, sensitivity, and ease of preparation, enabling rapid qualitative fluorescence readout and quantitative photoelectrochemical readout of hypochlorite, with a limit of detection of 0.288 μM for fluorescence and 1.37 nM for PEC.
Collapse
Affiliation(s)
- Jiaxing Chang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Zhinan Zhang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Chulin Qu
- State Key Laboratory of Coordination Chemistry, Collaborative Innovation Center of Advanced Microstructures, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Qingzhi Han
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Li Xu
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| |
Collapse
|
15
|
Hamashima K, Fan L, Miyagawa R, Hara N, Nishida K, Saitoh H. Examining interactions of animal cells with chloroplasts and their light-induced responses in in vitro cell culture systems. Biochem Biophys Res Commun 2025; 758:151622. [PMID: 40117974 DOI: 10.1016/j.bbrc.2025.151622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Chloroplasts are organelles that convert light energy into chemical energy in plants. The potential to integrate chloroplasts into animal cells presents an exciting frontier in synthetic biology, allowing for photo-controllable biochemical processes within these cells. However, the lack of well-established in vitro experimental systems to study chloroplast-animal cell interactions remains a significant challenge. This study investigates the behavior of human cervical cancer HeLa cells and mouse macrophage-like J774.1 cells, along with the light-induced responses of these cells, when introduced into culture media containing spinach-derived chloroplasts. Additionally, we examine isolated cells from Elysia marginata, a sacoglossan sea slug known for its unique ability to acquire and retain functional chloroplasts through a process known as kleptoplasty. Our results show that HeLa cells primarily adhere to chloroplasts with minimal intracellular uptake, while J774.1 cells actively engulf them. Co-incubation with chloroplasts increases the rate of cell death upon light irradiation. In contrast, naturally chloroplast-containing cells from E. marginata exhibit minimal light-induced damage. Excessive reactive oxygen species (ROS) production is observed in HeLa and J774.1 cells co-incubated with chloroplasts under light exposure, suggesting that photoinduced ROS generation contributes to cytotoxicity. These findings highlight three different patterns of interactions between animal cells and chloroplasts and underscore the importance of considering ROS generation induced by light exposure when analyzing chloroplast-animal cell interactions in vitro experimental systems.
Collapse
Affiliation(s)
- Kyota Hamashima
- Department of Biological Science, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Lilingman Fan
- Department of Biological Science, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Reika Miyagawa
- Department of Biological Science, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Natsuki Hara
- Department of Biological Science, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan
| | | | - Hisato Saitoh
- Department of Biological Science, Graduate School of Science and Technology, Kumamoto University, Kumamoto, Japan; Faculty of Advanced Science and Technology (FAST), Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
16
|
Moura JP, Oliveira PJ, Urbano AM. Mitochondrial classic metabolism and its often-underappreciated facets. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167839. [PMID: 40220877 DOI: 10.1016/j.bbadis.2025.167839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
For many decades, mitochondria were essentially regarded as the main providers of the adenosine triphosphate (ATP) required to maintain the viability and function of eukaryotic cells, thus the widely popular metaphor "powerhouses of the cell". Besides ATP generation - via intermediary metabolism - these intracellular organelles have also traditionally been known, albeit to a lesser degree, for their notable role in biosynthesis, both as generators of biosynthetic intermediates and/or as the sites of biosynthesis. From the 1990s onwards, the concept of mitochondria as passive organelles providing the rest of the cell, from which they were otherwise isolated, with ATP and biomolecules on an on-demand basis has been challenged by a series of paradigm-shifting discoveries. Namely, it was shown that mitochondria act as signaling effectors to upregulate ATP generation in response to growth-promoting stimuli and are actively engaged, through signaling and epigenetics, in the regulation of a plethora of cellular processes, ultimately deciding cell function and fate. With the focus of mitochondrial research increasingly placed in these "non-classical" functions, the centrality of mitochondrial intermediary metabolism to other mitochondrial functions tends to be overlooked. In this article, we revisit mitochondrial intermediary metabolism and illustrate how its intermediates, by-products and molecular machinery underpin other mitochondrial functions. A certain emphasis is given to frequently overlooked mitochondrial functions, namely the biosynthesis of iron-sulfur (Fe-S) clusters, the only known function shared by all mitochondria and mitochondrion-related organelles. The generation of reactive oxygen species (ROS) and their putative role in signaling is also discussed in detail.
Collapse
Affiliation(s)
- João P Moura
- Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| | - Paulo J Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Portugal.
| | - Ana M Urbano
- Molecular Physical-Chemistry R&D Unit, Centre for Investigation in Environment, Genetics and Oncobiology (CIMAGO), Department of Life Sciences, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
17
|
Mizrachi A, Sadeh M, Ben-Dor S, Dym O, Ku C, Feldmesser E, Zarfin A, Brunson JK, Allen AE, Jinkerson RE, Schatz D, Vardi A. Cathepsin X is a conserved cell death protein involved in algal response to environmental stress. Curr Biol 2025:S0960-9822(25)00361-6. [PMID: 40233752 DOI: 10.1016/j.cub.2025.03.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/12/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Phytoplankton are responsible for half of the global photosynthesis and form vast blooms in aquatic ecosystems. Bloom demise fuels marine microbial life and is suggested to be mediated by programmed cell death (PCD) induced by diverse environmental stressors. Despite its importance, the molecular basis for algal PCD remains elusive. Here, we reveal novel PCD genes conserved across distant algal lineages using cell-to-cell heterogeneity in the response of the diatom Phaeodactylum tricornutum to oxidative stress. Comparative transcriptomics of sorted sensitive and resilient subpopulations following oxidative stress revealed genes directly linked to their contrasting fates of cell death and survival. Comparing these genes with those found in a large-scale mutant screen in the green alga Chlamydomonas reinhardtii identified functionally relevant conserved PCD gene candidates, including the cysteine protease cathepsin X/Z (CPX). CPX mutants in P. tricornutum CPX1 and C. reinhardtii CYSTEINE ENDOPEPTIDASE 12 (CEP12) exhibited resilience to oxidative stress and infochemicals that induce PCD, supporting a conserved function of these genes in algal PCD. Phylogenetic and predictive structural analyses show that CPX is highly conserved in eukaryotes, and algae exhibit strong structural similarity to human Cathepsin X/Z (CTSZ), a protein linked to various diseases. CPX is expressed by diverse algae across the oceans and correlates with upcoming demise events during toxic Pseudo-nitzschia blooms, providing support for its ecological significance. Elucidating PCD components in algae sheds light on the evolutionary origin of PCD in unicellular organisms and on the cellular strategies employed by the population to cope with stressful conditions.
Collapse
Affiliation(s)
- Avia Mizrachi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mai Sadeh
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Shifra Ben-Dor
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Orly Dym
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Chuan Ku
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ester Feldmesser
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Amichai Zarfin
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - John K Brunson
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Department of Environment and Sustainability, J. Craig Venter Institute, La Jolla, San Diego, CA 92037, USA
| | - Andrew E Allen
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, San Diego, CA 92093, USA; Department of Environment and Sustainability, J. Craig Venter Institute, La Jolla, San Diego, CA 92037, USA
| | - Robert E Jinkerson
- Department of Chemical and Environmental Engineering, University of California, Riverside, CA 92521, USA
| | - Daniella Schatz
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Assaf Vardi
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
18
|
Nie G, Liang W, Wang J, Du Z, Xiao F, Liu M, Chen D, Wang H. Rational design of hypochlorous acid-activatable fluorescent probe for diagnostic imaging and therapeutic evaluation in breast cancer recurrence. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 330:125743. [PMID: 39826172 DOI: 10.1016/j.saa.2025.125743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
The recurrent breast cancer (BC) has elicited significant concern due to its rising recurrence rates and associated mortality. However, there is currently no effective detection method to mitigate the deterioration of BC recurrence. The imbalance of HClO content could lead to oxidative stress in the body, which damaging host tissues. Additional, improper regulation of HClO may exacerbate the progression of BC and promote the metastasis of BC cells. Accurately diagnosing and monitoring the HClO levels is crucial for treating BC recurrence. Traditional fluorescent probes for HClO exhibit several limitations, including poor selectivity, susceptibility to photobleaching, a small Stokes shift, and vulnerability to disturbances from excitation and fluorescence self-absorption, which undermine the precise detection of target analytes and restrict their biological applications. Herein, rational designed hypochlorous acid-activatable fluorescent probe (QPIO) was synthesized based on phenothiazine (PZ), quinoline malononitrile (QM), and hemicyanine, which demonstrated high anti-interference capability and a significant Stokes shift for HClO detection. Under various stimuli, QPIO was able to monitor HClO levels in RAW 264.7 and 4T1 cells in the red channel. Furthermore, it elucidated the correlation between HClO concentration and the progression of BC recurrence. Consequently, QPIO was utilized to diagnose recurrent BC, track therapeutic progress, and monitor the recurrence status of breast tumors in mouse models through in vivo HClO fluorescence imaging. It was demonstrated that a close relationship exists between the dynamic changes in HClO levels and BC recurrence, potentially advancing the understanding of the early diagnosis and development of therapeutic agents for recurrent BC.
Collapse
Affiliation(s)
- Gang Nie
- Department of Pharmacy, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology 430016 Wuhan, PR China
| | - Wenjie Liang
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology 430205 Wuhan, PR China
| | - Jun Wang
- Department of Pharmacy, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology 430016 Wuhan, PR China
| | - Zhaosong Du
- Department of Pharmacy, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology 430016 Wuhan, PR China
| | - Fengping Xiao
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, Chemical Biology Center, College of Chemistry, and International Joint Research Center for Intelligent Biosensing Technology and Health, Central China Normal University 430079 Wuhan, PR China
| | - Maochang Liu
- Department of Pharmacy, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology 430016 Wuhan, PR China.
| | - Dugang Chen
- Key Laboratory for Green Chemical Process of Ministry of Education, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology 430205 Wuhan, PR China.
| | - Huiling Wang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, Chemical Biology Center, College of Chemistry, and International Joint Research Center for Intelligent Biosensing Technology and Health, Central China Normal University 430079 Wuhan, PR China.
| |
Collapse
|
19
|
Song H, Lee J, Lee Y, Kim S, Kang S. Reactive Oxygen Species as a Common Pathological Link Between Alcohol Use Disorder and Alzheimer's Disease with Therapeutic Implications. Int J Mol Sci 2025; 26:3272. [PMID: 40244088 PMCID: PMC11989502 DOI: 10.3390/ijms26073272] [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: 02/13/2025] [Revised: 03/29/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025] Open
Abstract
Chronic alcohol consumption leads to excessive production of reactive oxygen species (ROS), driving oxidative stress that contributes to both alcohol use disorder (AUD) and Alzheimer's disease (AD). This review explores how ROS-mediated mitochondrial dysfunction and neuroinflammation serve as shared pathological mechanisms linking these conditions. We highlight the role of alcohol-induced oxidative damage in exacerbating neurodegeneration and compare ROS-related pathways in AUD and AD. Finally, we discuss emerging therapeutic strategies, including mitochondrial antioxidants and inflammasome inhibitors, that target oxidative stress to mitigate neurodegeneration. Understanding these overlapping mechanisms may provide new insights for preventing and treating ROS-driven neurodegenerative disorders.
Collapse
Affiliation(s)
| | | | | | | | - Shinwoo Kang
- Department of Clinical Pharmacology, College of Medicine, Soonchunhyang University, 31, Soonchunhyang 6-gil, Dongnam-gu, Cheonan-si 31151, Chungcheongnam-do, Republic of Korea; (H.S.); (J.L.); (Y.L.); (S.K.)
| |
Collapse
|
20
|
Cobley JN, Chatzinikolaou PN, Schmidt CA. The nonlinear cysteine redox dynamics in the i-space: A proteoform-centric theory of redox regulation. Redox Biol 2025; 81:103523. [PMID: 39929052 PMCID: PMC11849597 DOI: 10.1016/j.redox.2025.103523] [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: 12/05/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The post-translational redox regulation of protein function by cysteine oxidation controls diverse biological processes, from cell division to death. However, most current site-centric paradigms fail to capture the nonlinear and emergent nature of redox regulation in proteins with multiple cysteines. Here, we present a proteoform-centric theory of redox regulation grounded in the i-space. The i-space encapsulates the theoretical landscape of all possible cysteine proteoforms. Using computational approaches, we quantify the vast size of the abstract i-space, revealing its scale-free architecture-elucidating the disproportionate influence of cysteine-rich proteins. We define mathematical rules governing cysteine proteoform dynamics. Their dynamics are inherently nonlinear, context-dependent, and fundamentally constrained by protein copy numbers. Monte Carlo simulations of the human protein PTP1B reveal extensive i-space sampling beyond site-centric models, supporting the "oxiform conjecture". This conjecture posits that highly oxidised proteoforms, molecules bearing multiple oxidised cysteines, are central to redox regulation. In support, even 90%-reduced proteomes can house vast numbers of unique, potentially functioanlly diverse, oxiforms. This framework offers a transformative lens for understanding the redox biology of proteoforms.
Collapse
|
21
|
Xu M, Li W, Xu R, Liu L, Wu Z, Li W, Ma C, Xue L. Gp93 safeguards tissue homeostasis by preventing ROS-JNK-mediated apoptosis. Redox Biol 2025; 81:103537. [PMID: 39965405 PMCID: PMC11875814 DOI: 10.1016/j.redox.2025.103537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
Reactive oxygen species (ROS) play a pivotal role in maintaining tissue homeostasis, yet their overabundance can impair normal cellular functions, induce cell death, and potentially lead to neurodegenerative disorders. This study identifies Drosophila Glycoprotein 93 (Gp93) as a crucial factor that safeguards tissue homeostasis and preserves normal neuronal functions by preventing ROS-induced, JNK-dependent apoptotic cell death. Firstly, loss of Gp93 induces JNK-dependent apoptosis primarily through the induction of ROS. Secondary, neuro-specific depletion of Gp93 results in ROS-JNK-mediated neurodegeneration. Thirdly, overexpression of Gp93 effectively curtails oxidative stress and neurodegeneration caused by paraquat exposure or the aging process. Furthermore, these functions of Gp93 can be substituted by its human ortholog, HSP90B1. Lastly, depletion of HSP90B1 in cultured human cells triggers ROS production, JNK activation, and apoptosis. Thus, this study not only unveils a novel physiological function of Gp93, but also provides valuable insights for understanding the physiological and pathological functions of human HSP90B1.
Collapse
Affiliation(s)
- Meng Xu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Wanzhen Li
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Ruihong Xu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lixia Liu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Zhihan Wu
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Wenzhe Li
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Chao Ma
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Lei Xue
- Department of Nuclear Medicine, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China; National Clinical Research Center for Interventional Medicine, Shanghai 10th People's Hospital, 200072, Shanghai, China.
| |
Collapse
|
22
|
Aravapally PSN, Chandrasekar N, Verma A, Shah RP. Strategic approaches to assess and quantify the oxidative stress biomarkers in complex biological systems. Bioanalysis 2025; 17:561-574. [PMID: 40183176 DOI: 10.1080/17576180.2025.2486929] [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: 11/14/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Oxidative stress (OS) is an emerging research area in clinical and biological sciences due to its association with various diseases and physiological processes. OS occurs when there is an imbalance between the production of reactive oxygen species (ROS) and the body's ability to neutralize or repair the damage caused. Chronic oxidative stress is linked to diseases like diabetes, cardiovascular diseases, cancer, and neurodegenerative disorders. Accurate monitoring of OS is crucial for diagnosing diseases, evaluating disease progression, and predicting clinical results. Despite challenges in measuring free radicals due to their short half-life and low concentrations, it can be indirectly assessed through biomarkers like lipid peroxidation, DNA damage, and protein oxidation. The most effective analytical techniques for assessing OS biomarkers in various biological fluids were developed. Furthermore, an in-depth exploration of these various analytical methodologies, underscoring their sensitivity, specificity, and reliability in detecting low concentrations of biomarkers across complex matrices is necessary. A comprehensive literature search was conducted using databases such as Google Scholar, PubMed and Reaxys to identify relevant studies on OS biomarkers. This review explores the evolution of these techniques, highlighting advancements in sample preparation procedures and the specifications of each technique, offering a thorough evaluation of biomarker analysis.
Collapse
Affiliation(s)
- Padmasri Sai Nandana Aravapally
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A), Opposite Air force Station Palaj, Gandhinagar, India
| | - Naveen Chandrasekar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A), Opposite Air force Station Palaj, Gandhinagar, India
| | - Arvind Verma
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A), Opposite Air force Station Palaj, Gandhinagar, India
| | - Ravi P Shah
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A), Opposite Air force Station Palaj, Gandhinagar, India
| |
Collapse
|
23
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
24
|
Xie X, Huang M, Ma S, Xin Q, Wang Y, Hu L, Zhao H, Li P, Liu M, Yuan R, Miao Y, Zhu Y, Cong W. The role of long non-coding RNAs in cardiovascular diseases: A comprehensive review. Noncoding RNA Res 2025; 11:158-187. [PMID: 39896344 PMCID: PMC11783329 DOI: 10.1016/j.ncrna.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide, posing significant challenges to healthcare systems. Despite advances in medical interventions, the molecular mechanisms underlying CVDs are not yet fully understood. For decades, protein-coding genes have been the focus of CVD research. However, recent advances in genomics have highlighted the importance of long non-coding RNAs (lncRNAs) in cardiovascular health and disease. Changes in lncRNA expression specific to tissues may result from various internal or external factors, leading to tissue damage, organ dysfunction, and disease. In this review, we provide a comprehensive discussion of the regulatory mechanisms underlying lncRNAs and their roles in the pathogenesis and progression of CVDs, such as coronary heart disease, atherosclerosis, heart failure, arrhythmias, cardiomyopathies, and diabetic cardiomyopathy, to explore their potential as therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Xuena Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Meiwen Huang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Shudong Ma
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Faculty of Chinese Medicine, Macau University of Science and Technology, 999078, China
| | - Qiqi Xin
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yuying Wang
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Lantian Hu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Pengqi Li
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Mei Liu
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rong Yuan
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yu Miao
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
| | - Weihong Cong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, 999078, China
- Laboratory of Cardiovascular Diseases, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| |
Collapse
|
25
|
Kervella M, Bertile F, Bouillaud F, Criscuolo F. The cell origin of reactive oxygen species and its implication for evolutionary trade-offs. Open Biol 2025; 15:240312. [PMID: 40237040 PMCID: PMC12001088 DOI: 10.1098/rsob.240312] [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: 10/21/2024] [Revised: 01/08/2025] [Accepted: 02/09/2025] [Indexed: 04/17/2025] Open
Abstract
The allocation of resources in animals is shaped by adaptive trade-offs aimed at maximizing fitness. At the heart of these trade-offs, lies metabolism and the conversion of food resources into energy, a process mostly occurring in mitochondria. Yet, the conversion of nutrients to utilizable energy molecules (adenosine triphosphate) inevitably leads to the by-production of reactive oxygen species (ROS) that may cause damage to important biomolecules such as proteins or lipids. The 'ROS theory of ageing' has thus proposed that the relationship between lifespan and metabolic rate may be mediated by ROS production. However, the relationship is not as straightforward as it may seem: not only are mitochondrial ROS crucial for various cellular functions, but mitochondria are also actually equipped with antioxidant systems, and many extra-mitochondrial sources also produce ROS. In this review, we discuss how viewing the mitochondrion as a regulator of cellular oxidative homeostasis, not merely a ROS producer, may provide new insights into the role of oxidative stress in the reproduction-survival trade-off. We suggest several avenues to test how mitochondrial oxidative buffering capacity might complement current bioenergetic and evolutionary studies.
Collapse
|
26
|
Zhang K, Jagannath C. Crosstalk between metabolism and epigenetics during macrophage polarization. Epigenetics Chromatin 2025; 18:16. [PMID: 40156046 PMCID: PMC11954343 DOI: 10.1186/s13072-025-00575-9] [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: 11/24/2024] [Accepted: 02/17/2025] [Indexed: 04/01/2025] Open
Abstract
Macrophage polarization is a dynamic process driven by a complex interplay of cytokine signaling, metabolism, and epigenetic modifications mediated by pathogens. Upon encountering specific environmental cues, monocytes differentiate into macrophages, adopting either a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype, depending on the cytokines present. M1 macrophages are induced by interferon-gamma (IFN-γ) and are characterized by their reliance on glycolysis and their role in host defense. In contrast, M2 macrophages, stimulated by interleukin-4 (IL-4) and interleukin-13 (IL-13), favor oxidative phosphorylation and participate in tissue repair and anti-inflammatory responses. Metabolism is tightly linked to epigenetic regulation, because key metabolic intermediates such as acetyl-coenzyme A (CoA), α-ketoglutarate (α-KG), S-adenosylmethionine (SAM), and nicotinamide adenine dinucleotide (NAD+) serve as cofactors for chromatin-modifying enzymes, which in turn, directly influences histone acetylation, methylation, RNA/DNA methylation, and protein arginine methylation. These epigenetic modifications control gene expression by regulating chromatin accessibility, thereby modulating macrophage function and polarization. Histone acetylation generally promotes a more open chromatin structure conducive to gene activation, while histone methylation can either activate or repress gene expression depending on the specific residue and its methylation state. Crosstalk between histone modifications, such as acetylation and methylation, further fine-tunes macrophage phenotypes by regulating transcriptional networks in response to metabolic cues. While arginine methylation primarily functions in epigenetics by regulating gene expression through protein modifications, the degradation of methylated proteins releases arginine derivatives like asymmetric dimethylarginine (ADMA), which contribute directly to arginine metabolism-a key factor in macrophage polarization. This review explores the intricate relationships between metabolism and epigenetic regulation during macrophage polarization. A better understanding of this crosstalk will likely generate novel therapeutic insights for manipulating macrophage phenotypes during infections like tuberculosis and inflammatory diseases such as diabetes.
Collapse
Affiliation(s)
- Kangling Zhang
- Department of Pharmacology and Toxicology, School of Medicine, University of Texas Medical Branch, Galveston, TX, USA.
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Research Institute, Weill-Cornell Medicine, Houston, TX, USA.
| |
Collapse
|
27
|
Pană IO, Ciorîță A, Boca S, Guțoiu S, Kacso I, Miclăuș MO, Grad O, Gherman AMR, Leostean C, Suciu M. Interaction of Manganese-Doped Copper Oxide Nano-Platelets with Cells: Biocompatibility and Anticancer Activity Assessment. Biomimetics (Basel) 2025; 10:203. [PMID: 40277602 PMCID: PMC12024727 DOI: 10.3390/biomimetics10040203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/26/2025] Open
Abstract
Understanding cellular interaction with nanomaterials represents a subject of great interest for the validation of new diagnostic and therapeutic tools. A full characterization of a designed product includes the evaluation of its impact on specific biological systems, including the study of cell behavior as a response to that particular interaction. Copper and copper-based nanoparticles (CuO NPs) have emerged as valuable building blocks for various biomedical applications such as antibacterial and disinfecting agents for infectious diseases, and the evaluation of the metabolism of food, including the iron required for proteins and enzymes or as drug delivery systems in cancer therapy. In this study, the biological impact of manganese-doped crystalline copper oxide (CuO:Mn) nano-platelets on human normal BJ fibroblasts and human A375 skin melanoma was assessed. The particles were synthesized at room temperature via the hydrothermal method. A complete physicochemical characterization of the materials was performed by employing various techniques including X-ray diffraction, electron microscopy, X-Ray photoelectron spectroscopy, and dynamic light scattering. Morphological investigations revealed a flat structure with nearly straight edges, with sizes spanning in the nanometer range. XRD analysis confirmed the formation of the CuO phase with good crystallinity, while XPS provided insights into the Mn doping. The findings indicate that nano-platelets interact with cells actively by mediating essential molecular processes. The exogenous manganese triggers increased MnSOD production in mitochondria, compensating ROS produced by external stress factors (Cu2+ ions), and mimics the endogenous SODs production, which compensates internal ROS production as it normally results from cell biochemistry. The effect is differentiated in normal cells compared to malignant cells and deserves investigation.
Collapse
Affiliation(s)
- Ioan-Ovidiu Pană
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Alexandra Ciorîță
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
- Electron Microscopy Center C. Craciun, Faculty of Biology and Geology, Babeș-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
| | - Sanda Boca
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
- Interdisciplinary Research Institute in Bio-Nano-Sciences, Babeș-Bolyai University, 42 Treboniu Laurian, 400271 Cluj-Napoca, Romania
| | - Simona Guțoiu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Irina Kacso
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Maria Olimpia Miclăuș
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Oana Grad
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Ana Maria Raluca Gherman
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Cristian Leostean
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
| | - Maria Suciu
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat Street, 400293 Cluj-Napoca, Romania; (I.-O.P.); (A.C.); (S.G.); (I.K.); (M.O.M.); (O.G.); (A.M.R.G.); (C.L.); (M.S.)
- Electron Microscopy Center C. Craciun, Faculty of Biology and Geology, Babeș-Bolyai University, 5-7 Clinicilor Street, 400006 Cluj-Napoca, Romania
| |
Collapse
|
28
|
Meng X, Qu H, Zhang X, Ji H, Sun M. Ginsenoside Rb3 Inhibits Oxidative Stress and Alleviates Experimental Periodontitis in Rats. Oral Dis 2025. [PMID: 40127169 DOI: 10.1111/odi.15318] [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: 01/03/2025] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 03/26/2025]
Abstract
OBJECTIVE This study investigated the anti-inflammatory and antioxidative effects of ginsenoside Rb3 on experimental periodontitis. METHODS Experimental periodontitis was established by ligating the maxillary first molars. After 21 days of Rb3 treatment, rats were sacrificed for micro-CT and H&E staining analysis and serum assay. qPCR detected the expression of inflammatory genes. IF detected MAPKs pathway activation in periodontal tissues. The effects were determined using LPS-induced RAW264.7 cells. Gene expression levels were determined by qPCR. Intracellular ROS generation was detected by DCFH-DA staining and flow cytometry. WB detected the activation of signaling pathways. RESULTS Rb3 inhibited oxidative stress in experimental periodontitis, showing reduced gingivitis and alveolar bone resorption on H&E staining and micro-CT, and reduced iNOS mRNA in rats. Colorimetric results showed that Rb3 increased serum SOD and GSH-Px activities and decreased MDA levels in rats. IF analysis showed that Rb3 inhibited P38, ERK, and JNK activation. Rb3 pretreatment significantly decreased iNOS and IL-6 mRNA expression and ROS in Raw264.7 cells. WB analysis demonstrated that Rb3 blocked the activation of P38 and ERK. CONCLUSION Rb3 inhibits the level of oxidative stress and attenuates gingivitis and alveolar bone resorption in rats via the MAPKs signaling pathway.
Collapse
Affiliation(s)
- Xin Meng
- School of Stomatology, Shandong Second Medical University, Weifang, China
| | - Huijuan Qu
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Xueying Zhang
- School of Stomatology, Shandong Second Medical University, Weifang, China
| | - Honghai Ji
- School of Stomatology, Shandong Second Medical University, Weifang, China
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Minmin Sun
- School of Stomatology, Shandong Second Medical University, Weifang, China
- Department of Stomatology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
29
|
Corcini CD, Varela Junior AS, Yeste M. Environmental contamination and male reproductive health: (ir) reversible effects in child- and adulthood. AN ACAD BRAS CIENC 2025; 97:e20240717. [PMID: 40136200 DOI: 10.1590/0001-3765202520240717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/02/2024] [Indexed: 03/27/2025] Open
Abstract
Infertility affects 10-15% of reproductive-age couples, with causes ranging from genetic factors to unidentified reasons. Environmental conditions, particularly pollutants, play a significant role in male fertility. Yet, public health policies often overlook reproductive health, despite mounting evidence of pollutants' detrimental repercussion. Understanding this impact is crucial to prevent the effects of dangerous exposure, especially given the high levels of environmental pollutants in today's world. Most of the previous research about the adverse effects from contaminants has been conducted in rodents, with limited human epidemiological research. This article reviews the evidence about the impact of various contaminants (air pollutants, water contaminants, pesticides, herbicides, radiation, heavy metals, and plastics) on male reproductive health, particularly sperm quality and fertility. The literature suggests that exposure to contaminants during fetal development and childhood has irreversible effects, while those of adult exposure are often reversible. These findings highlight the need to alert society about reproductive health threats from certain contaminants. Public authorities should consider this situation when designing health plans, and individuals envisaging fatherhood should be aware of these risks.
Collapse
Affiliation(s)
- Carine Dahl Corcini
- Universidade Federal de Pelotas, Faculdade de Medicina Veterinária, Departamento de Patologia Animal, Reprodução Animal Comparada, Campus Capão do Leão, s/n, 96160-000 Capão do Leão, RS, Brazil
| | - Antonio Sergio Varela Junior
- Universidade Federal do Rio Grande, Instituto de Ciências Biológicas, Reprodução Animal Comparada, Av. Itália, km 8, Carreiros, 96203-900 Rio Grande, RS, Brazil
| | - Marc Yeste
- University of Girona, Institute of Food and Agricultural Technology, Biotechnology of Animal and Human Reproduction (TechnoSperm), ES-17003 Girona, Spain
- University of Girona, Faculty of Sciences, Department of Biology, Unit of Cell Biology, ES-17003 Girona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), ES-08010 Barcelona, Spain
| |
Collapse
|
30
|
Ishida H, Sasaki Y, Shibata T, Sasaki H, Chhunchha B, Singh DP, Kubo E. Topical Instillation of N-Acetylcysteine and N-Acetylcysteine Amide Impedes Age-Related Lens Opacity in Mice. Biomolecules 2025; 15:442. [PMID: 40149978 PMCID: PMC11940285 DOI: 10.3390/biom15030442] [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: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Cataracts, the leading cause of blindness globally, are caused by oxidative stress and inflammation, which disrupt lens transparency due to increased accumulation of reactive oxygen species (ROS) as well as protein and DNA damage during aging. Using in vitro, ex vivo, and in vivo models, we determined the protective efficacy of N-acetylcysteine amide (NACA) against oxidative stress-induced and aging-induced cataractogenesis. We found that lens epithelial cells exposed to the oxidative stress inducers hydrogen peroxide (H2O2) or tert-butyl hydroperoxide showed significant ROS accumulation and reduced cellular viability. These effects were inhibited by NACA via the suppression of ROS and thioredoxin-interacting protein (Txnip) expression, a regulator of oxidative stress-related cellular damage and inflammation. In ex vivo lens experiments, NACA significantly reduced H2O2-induced lens opacity and preserved lens integrity. Similarly to NACA-treated lenses ex vivo, the integrity and opacity of aged mouse lenses, when topically instilled with NACA, were preserved and reduced, respectively, and are directly related to reduced Txnip and increased thioredoxin (Trx) expression levels. Overall, our findings demonstrated the protective ability of NACA to abate aberrant redox-active pathways, particularly the ROS/TRX/TXNIP axis, thereby preventing cataractogenesis and preserving eye lens integrity and ultimately impeding aging-related cataracts.
Collapse
Affiliation(s)
- Hidetoshi Ishida
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan; (H.I.); (Y.S.); (T.S.); (H.S.)
| | - Yu Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan; (H.I.); (Y.S.); (T.S.); (H.S.)
| | - Teppei Shibata
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan; (H.I.); (Y.S.); (T.S.); (H.S.)
| | - Hiroshi Sasaki
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan; (H.I.); (Y.S.); (T.S.); (H.S.)
| | - Bhavana Chhunchha
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (B.C.); (D.P.S.)
| | - Dhirendra P. Singh
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (B.C.); (D.P.S.)
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kanazawa 9200293, Japan; (H.I.); (Y.S.); (T.S.); (H.S.)
| |
Collapse
|
31
|
Aggarwal T, Wang L, Gutierrez B, Guven H, Erguven H, Cho S, Izgu EC. A Small-Molecule Approach Enables RNA Aptamers to Function as Sensors for Reactive Inorganic Targets. Angew Chem Int Ed Engl 2025; 64:e202421936. [PMID: 39666858 PMCID: PMC11914935 DOI: 10.1002/anie.202421936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 12/14/2024]
Abstract
Fluorescent light-up aptamer (FLAP) systems are promising (bio)sensing platforms that are genetically encodable. However, FLAP-mediated detection of each distinct target necessitates either in vitro selection or engineering of nucleic acid sequences. Furthermore, an aptamer that binds an inorganic target or a chemical species with a short lifetime is challenging to realize. Here, we describe a small-molecule approach that makes it possible for a single FLAP system to detect chemically unique, non-fluorogenic, and reactive inorganics. We developed functionalized pre-ligands of RNA aptamers that bind benzylidene imidazolinones (Baby Spinach, Broccolli, Squash). Reactive inorganics, hydrogen sulfide (H2S/HS-) and hydrogen peroxide (H2O2), can specifically convert these pre-ligands into native ligands that fluoresce with FLAPs. Adaptation of this platform to live cells opened an opportunity for constructing whole-cell sensors: Escherichia coli transformed with a Baby Spinach-encoding plasmid and incubated with pre-ligands generated fluorescence in response to exogenous H2S/HS- or H2O2. Leveraging the functional group reactivity of small molecules eliminates the requirement of in vitro selection of a new aptamer sequence or oligonucleotide scaffold engineering for distinct molecular targets. Our method allows for detecting inorganic, short-lived species, thereby advancing FLAP systems beyond their current capabilities.
Collapse
Affiliation(s)
- Tushar Aggarwal
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Liming Wang
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Bryan Gutierrez
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Hakan Guven
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Huseyin Erguven
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Sarah Cho
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
| | - Enver Cagri Izgu
- Department of Chemistry and Chemical BiologyRutgers UniversityNew BrunswickNJ-08854USA
- Cancer Institute of New JerseyRutgers UniversityNew BrunswickNJ-08901USA
- Rutgers Center for Lipid ResearchNew Jersey Institute for FoodNutritionand HealthRutgers UniversityNew BrunswickNJ-08901USA
| |
Collapse
|
32
|
Xie G, Chen G, Yuan M, Song Y, Xiao Y, Qu Y, Cui T, Ren Y. Mechanisms and potential applications of different stimulants enhancing benzo[a]pyrene degradation based on cellular characteristics and transcriptomic analysis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 369:125788. [PMID: 39914567 DOI: 10.1016/j.envpol.2025.125788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/24/2025]
Abstract
Benzo[a]pyrene (BaP) is a highly carcinogenic persistent organic pollutant, and biostimulation is an effective strategy to enhance its degradation. This study utilized Bacillus subtilis MSC4 as a BaP-degrading bacterium to investigate the effects of two different fermentation waste liquids as stimulants on BaP degradation. The mechanisms were analyzed and compared at both the cellular and molecular levels. The results showed that the stimulation percentages of yeast Yarrowia lipolytica extracellular metabolites (YEMs) and Lactobacillus plantarum fermentation waste solution (LPS) on the biodegradation of BaP reached 52.8% and 63.4%, respectively, compared to B treatment without biostimulant. Physiological analyses showed that both stimulants repaired cell morphology, more than doubled bacterial biomass, increased EPS secretion, enhanced bacterial activity, and significantly reduced oxidative stress by lowering ROS levels to 75-78% of those in the BaP-stressed group, allowing for repair of oxidative damage. Transcriptomic analysis indicated that both stimulants upregulated pathways related to central carbon metabolism, enhancing cell proliferation and energy supply. Additionally, YEMs promoted electron transport and BaP transmembrane transport and upregulated the synthesis of various monooxygenases, while LPS induced the upregulation of genes encoding quercetin dioxygenase and played a more active role in biofilm formation and enhancing BaP bioavailability. This study reveals the shared and distinct mechanisms by which YEMs and LPS enhance BaP biodegradation, providing theoretical guidance for the application of YEMs and LPS in the bioremediation of BaP-contaminated environments.
Collapse
Affiliation(s)
- Guanghong Xie
- School of Environment and Energy, South China University of Technology, Guangzhou, 510006, PR China
| | - Guotao Chen
- School of Environment and Energy, South China University of Technology, Guangzhou, 510006, PR China
| | - Meng Yuan
- School of Environment and Energy, South China University of Technology, Guangzhou, 510006, PR China
| | - Yuxin Song
- School of Environment and Energy, South China University of Technology, Guangzhou, 510006, PR China
| | - Yibo Xiao
- Protoga Biotechnology Co., Ltd., Shenzhen, 518000, PR China; Microalgae Biosynthesis R&D Center, Research Institute of Tsinghua University in Shenzhen, Shenzhen, 518057, PR China
| | - Yujiao Qu
- Protoga Biotechnology Co., Ltd., Shenzhen, 518000, PR China; Microalgae Biosynthesis R&D Center, Research Institute of Tsinghua University in Shenzhen, Shenzhen, 518057, PR China
| | - Tangbing Cui
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Yuan Ren
- School of Environment and Energy, South China University of Technology, Guangzhou, 510006, PR China; The Key Lab of Pollution Control and Ecosystem Restoration in Industry Clusters, Ministry of Education, Guangzhou, 510006, PR China; The Key Laboratory of Environmental Protection and Eco-Remediation of Guangdong Regular Higher Education Institutions, Guangzhou, 510006, PR China.
| |
Collapse
|
33
|
Wei W, Liu Z, Pan X, Yang T, An C, Wang Y, Li L, Liao W, Wang C. Effects of reactive oxygen species on fruit ripening and postharvest fruit quality. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 352:112391. [PMID: 39805341 DOI: 10.1016/j.plantsci.2025.112391] [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: 08/07/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Reactive oxygen species (ROS) serve as important signaling molecule, involved in numerous biological processes, particularly in the physiological changes associated with fruit ripening and postharvest handing. This review explores ROS key role in plant fruit ripening and postharvest quality. The mechanism of ROS production and degradation in maintaining ROS homeostasis are analyzed in detail. Fruit ripening is a complex and highly coordinated process involving physiological and biochemical changes. Studies have observed that the content of ROS, mainly hydrogen peroxide (H2O2), dynamically changes in various types of fruits during ripening. Furthermore, ROS have significant effects on fruit softening, color change, and other ripening processes. In addition, in the postharvest stage, the abnormal accumulation of ROS isclosely related to the decline in fruit quality and the occurrence of decay browning, which seriously affects the market value and shelf life of fruit. Overall, this review demonstrates the crucial role of ROS in regulating the ripening process and postharvest quality of fruit.
Collapse
Affiliation(s)
- Wenying Wei
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Zesheng Liu
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Xuejuan Pan
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Tingyue Yang
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Caiting An
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Yuanhui Wang
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Long Li
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Weibiao Liao
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China
| | - Chunlei Wang
- College of Horticulture, Gansu Agricultural University, Lanzhou 730070, China.
| |
Collapse
|
34
|
Chen Y, Li M, Wu Y. Heat shock protein 22: A new direction for cardiovascular disease (Review). Mol Med Rep 2025; 31:82. [PMID: 39886946 PMCID: PMC11800183 DOI: 10.3892/mmr.2025.13447] [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: 10/07/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Small heat shock proteins (sHSPs) are common molecular chaperone proteins that function in various biological processes, and serve indispensable roles in maintaining cellular protein homeostasis and regulating the hydrolysis of unfolded proteins. HSP22 is a member of the sHSP family that is primarily expressed in the heart and skeletal muscle, as well as in various types of cancer. There have been important findings concerning the role of HSP22 in cardiovascular diseases. The aim of the present study was to provide insights into the various molecular mechanisms by which HSP22 functions in the heart, including oxidative stress, autophagy, apoptosis, the subcellular distribution of proteins and the promoting effect of proteasomes. In addition, drugs and cytokines, including geranylgeranylacetone, can exert protective effects on the heart by regulating the expression of HSP22. Based on increasingly abundant research, HSP22 may be considered a potential therapeutic target in cardiovascular diseases.
Collapse
Affiliation(s)
- Yi Chen
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
35
|
Luo N, Zhang K, Li X, Hu Y, Guo L. Tanshinone IIA destabilizes SLC7A11 by regulating PIAS4-mediated SUMOylation of SLC7A11 through KDM1A, and promotes ferroptosis in breast cancer. J Adv Res 2025; 69:313-327. [PMID: 38615741 PMCID: PMC11954794 DOI: 10.1016/j.jare.2024.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/17/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024] Open
Abstract
INTRODUCTION Breast cancer (BC) is the most common malignancy in women with unfavorite prognosis. OBJECTIVES Tanshinone IIA (Tan IIA) inhibits BC progression, however, the underlying mechanism remains largely undefined. METHODS The cytotoxicity of Tan IIA was assessed by CCK-8 and LDH assays. Ferroptosis was monitored by the level of MDA, Fe2+, lipid ROS and GSH. IHC and western blot were employed to detect the localization and expression of SLC7A11, PIAS4, KDM1A and other key molecules. The SUMOylation of SLC7A11 was detected by Ni-beads pull-down assay and Co-IP. Luciferase and ChIP assays were employed to detect the direct association between KDM1A and PIAS4 promoter. The proliferative and metastatic properties of BC cells were assessed by colony formation, CCK-8 and Transwell assays, respectively. The in vitro findings were verified in xenograft and lung metastasis models. RESULTS Tan IIA promoted ferroptosis by suppressing SLC7A11 in BC cells. Silencing of PIAS4 or KDM1A inhibited cell growth and metastasis in BC. Mechanistically, PIAS4 facilitated the SUMOylation of SLC7A11 via direct binding to SLC7A11, and KDM1A acted as a transcriptional activator of PIAS4. Functional studies further revealed that Tan IIA decreased KDM1A expression, thus suppressing PIAS4 expression transcriptionally. The inhibition of PIAS4-dependent SUMOylation of SLC7A11 further induced ferroptosis, thereby inhibiting proliferation and metastasis in BC. CONCLUSION Tan IIA promoted ferroptosis and inhibited tumor growth and metastasis via suppressing KDM1A/PIAS4/SLC7A11 axis.
Collapse
Affiliation(s)
- Na Luo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center For Breast Cancer Control and Prevention in Hunan Province, China
| | - KeJing Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center For Breast Cancer Control and Prevention in Hunan Province, China
| | - Xin Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center For Breast Cancer Control and Prevention in Hunan Province, China
| | - Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center For Breast Cancer Control and Prevention in Hunan Province, China
| | - Lei Guo
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Clinical Research Center For Breast Cancer Control and Prevention in Hunan Province, China.
| |
Collapse
|
36
|
Xu X, Ge C, Wang S, Gao L, Wang C, Dai F, Wang Y, Xie S. Polyamine-modified naphthalimide derivative 9C inhibits colorectal cancer through ROS-mediated ER stress, migration and invasion. Toxicol In Vitro 2025; 103:105974. [PMID: 39586364 DOI: 10.1016/j.tiv.2024.105974] [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: 08/03/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Mounting evidence over the past decades has demonstrated the therapeutic potential of targeting endoplasmic reticulum (ER) stress signaling in cancer. Naphthalimdes exert their anti-cancer activities in a variety of ways. However, the effects of naphthalimides on ER stress are rarely reported. In this study, based on RNA-sequencing analysis, we observed that 9C, a naphthalimide derivative, could trigger ER stress to activate death receptor signaling and autophagy. Pretreatment of ER stress inhibitor, such as salubrinal, and autophagy inhibitor, such as 3-methyladenine (3-MA), partially reversed 9C-induced inhibition of cell growth. Furthermore, our results unveiled a reactive oxygen species (ROS)-dependent inhibitory effect of 9C. In addition, 9C inhibited colorectal cancer (CRC) cells migration and invasion. Removal of ROS using N-acetyl-L-cysteine (NAC) attenuated the expression of ATF4, CHOP, death receptors, E-cadherin, and the apoptosis and autophagy related proteins. Taken together, our results suggested that ROS-mediated ER stress, migration, and invasion is responsible for the therapeutic potential of naphthalimides including 9C in CRC.
Collapse
Affiliation(s)
- Xiaojuan Xu
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China
| | - Chaochao Ge
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China; School of Pharmacy, Heze University, Heze 274015, Shandong, China
| | - Senzhen Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China; School of Life Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Lei Gao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China
| | - Chaojie Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China
| | - Fujun Dai
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China.
| | - Yuxia Wang
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, Henan, China.
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
37
|
Tian Y, Wei Y, Zhang M, Wei X, Chen M, Wang J. Ratio type nanoprobe with boric acid as recognition unit for imaging intracellular H 2O 2 with SERS. Talanta 2025; 284:127224. [PMID: 39591861 DOI: 10.1016/j.talanta.2024.127224] [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: 07/01/2024] [Revised: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
Fluctuations of intracellular H2O2 level are intricately linked to diverse biological processes, e.g., cellular proliferation, biosynthesis, and metabolic activities. The dynamics of intracellular H2O2 levels holds immense significance as it represents a pivotal metabolite within the realm of free radicals. This study describes a novel internal standard assisted surface enhanced Raman scattering (SERS) nanoprobe based on H2O2 boronic acid oxide group for precise detection of H2O2 levels in vitro and within living cells. The nanoprobe consists of a core molecule shell Au nanostar (Au@MPBN@Au NPs) embedded with internal marker (4-mercaptobenzonitrile, 4-MPBN) as a high plasma active SERS substrate and a 4-mercaptophenylboronic acid (4-MPBA) molecule fixed on its surface as H2O2 recognition unit. The presence of H2O2 leads to a decrease of the band intensity for B-O group at 998 cm-1, while the absorption for the internal standard molecule 4-MPBN at 2223 cm-1 remains unaffected by the variation of external environment. H2O2 quantification may be achieved by analyzing the ratio of band intensities at I2223 cm-1/I998 cm-1, with a linear relationship between I2223cm-1/I998cm-1 and H2O2 concentration within the range of 5-100 μM, along with a limit of detection (LOD) of 1.97 μM. Meanwhile, due to the presence of internal marker molecules, measurement errors caused by environmental or instrumental fluctuations can be calibrated in real-time. The AuNPs/4-MPBN/4-MPBA nanoprobe provides an accurate tool for dynamic monitoring and quantitative characterization of intracellular H2O2 content during cell apoptosis or other cell growth processes.
Collapse
Affiliation(s)
- Yizhuo Tian
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Yujia Wei
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Mingyu Zhang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Xing Wei
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| | - Mingli Chen
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Jianhua Wang
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang, 110819, China.
| |
Collapse
|
38
|
Dev W, Sultana F, Li H, Hu D, Peng Z, He S, Zhang H, Waqas M, Geng X, Du X. Molecular mechanisms of cold stress response in cotton: Transcriptional reprogramming and genetic strategies for tolerance. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2025; 352:112390. [PMID: 39827949 DOI: 10.1016/j.plantsci.2025.112390] [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: 09/27/2024] [Revised: 01/07/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Cold stress has a huge impact on the growth and development of cotton, presenting a significant challenge to its productivity. Comprehending the complex molecular mechanisms that control the reaction to CS is necessary for developing tactics to improve cold tolerance in cotton. This review paper explores how cotton responds to cold stress by regulating gene expression, focusing on both activating and repressing specific genes. We investigate the essential roles that transcription factors and regulatory elements have in responding to cold stress and controlling gene expression to counteract the negative impacts of low temperatures. Through a comprehensive examination of new publications, we clarify the intricacies of transcriptional reprogramming induced by cold stress, emphasizing the connections between different regulatory elements and signaling pathways. Additionally, we investigate the consecutive effects of cold stress on cotton yield, highlighting the physiological and developmental disturbances resulting from extended periods of low temperatures. The knowledge obtained from this assessment allows for a more profound comprehension of the molecular mechanisms that regulate cold stress responses, suggesting potential paths for future research to enhance cold tolerance in cotton by utilizing targeted genetic modifications and biotechnological interventions.
Collapse
Affiliation(s)
- Washu Dev
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Fahmida Sultana
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Hongge Li
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China
| | - Daowu Hu
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China
| | - Zhen Peng
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Shoupu He
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Haobo Zhang
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Muhammad Waqas
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Xiaoli Geng
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China
| | - Xiongming Du
- National Key Laboratory of Cotton Bio-breeding and Integrated Utilization, Institute of Cotton Research of the Chinese Academy of Agricultural Sciences, Anyang 455000, China; National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya, Hainan 57202, China.
| |
Collapse
|
39
|
Li J, Liu Y, Geng K, Lu X, Shen X, Guo Q. ROS-Responsive Nanoparticles with Antioxidative Effect for the treatment of Diabetic Retinopathy. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:440-461. [PMID: 39316729 DOI: 10.1080/09205063.2024.2406628] [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: 02/19/2024] [Accepted: 04/29/2024] [Indexed: 09/26/2024]
Abstract
Diabetic retinopathy (DR) is a common microvascular complication of diabetes necessitating early intervention to impede progression, despite current clinical treatments focusing on advanced stages. Essential oils from Fructus Alpiniae zerumbet (EOFAZ) have demonstrated efficacy in protecting against high glucose (HG)-induced Müller cell activation and DR development. This study introduced a reactive oxidative species (ROS)-responsive drug delivery system (NPSPHE@EOFAZ) targeting early DR stages and oxidative stress. Our engineered nanoparticles effectively deliver EOFAZ into HG-exposed Müller cells by detecting and responding to elevated oxidative stress levels. The NPSPHE@EOFAZ significantly inhibited abnormal cell growth, reduced oxidative stress, and alleviated inflammation in vitro. In vivo experiments on diabetic mice with DR revealed that NPSPHE@EOFAZ mitigated early pathological changes by reducing oxidative stress and inflammation while also alleviating organ damage in the heart, liver, spleen, lung, and kidney. These findings underscore the potential of NPSPHE@EOFAZ as a promising antioxidant for early intervention in DR pathogenesis.
Collapse
Affiliation(s)
- Jinjin Li
- The Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yujia Liu
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Kedui Geng
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xin Lu
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xiangchun Shen
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Qianqian Guo
- The Department of Pharmacology of Materia Medica (the High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, the Key Laboratory of Optimal Utilization of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
- The Guizhou Provincial Scientific and Technologic Innovation Base ([2023]003), Guizhou Medical University, Guiyang, Guizhou Province, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| |
Collapse
|
40
|
Luo X, Zhang Y, Zeng Y, Yang D, Zhou Z, Zheng Z, Xiao P, Ding X, Li Q, Chen J, Deng Q, Zhong X, Qiu S, Yan W. Nanotherapies Based on ROS Regulation in Oral Diseases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409087. [PMID: 39887942 PMCID: PMC11884622 DOI: 10.1002/advs.202409087] [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: 08/03/2024] [Revised: 12/28/2024] [Indexed: 02/01/2025]
Abstract
Oral diseases rank among the most prevalent clinical conditions globally, typically involving detrimental factors such as infection, inflammation, and injury in their occurrence, development, and outcomes. The concentration of reactive oxygen species (ROS) within cells has been demonstrated as a pivotal player in modulating these intricate pathological processes, exerting significant roles in restoring oral functionality and maintaining tissue structural integrity. Due to their enzyme-like catalytic properties, unique composition, and intelligent design, ROS-based nanomaterials have garnered considerable attention in oral nanomedicine. Such nanomaterials have the capacity to influence the spatiotemporal dynamics of ROS within biological systems, guiding the evolution of intra-ROS to facilitate therapeutic interventions. This paper reviews the latest advancements in the design, functional customization, and oral medical applications of ROS-based nanomaterials. Through the analysis of the components and designs of various novel nanozymes and ROS-based nanoplatforms responsive to different stimuli dimensions, it elaborates on their impacts on the dynamic behavior of intra-ROS and their potential regulatory mechanisms within the body. Furthermore, it discusses the prospects and strategies of nanotherapies based on ROS scavenging and generation in oral diseases, offering alternative insights for the design and development of nanomaterials for treating ROS-related conditions.
Collapse
Affiliation(s)
- Xin Luo
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yanli Zhang
- Stomatological HospitalSchool of StomatologySouthern Medical UniversityGuangzhou510280China
| | - Yuting Zeng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Dehong Yang
- Department of Orthopedics Spinal SurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Zhiyan Zhou
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Ziting Zheng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Ping Xiao
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xian Ding
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qianlin Li
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Jiaping Chen
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qianwen Deng
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Xincen Zhong
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Sijie Qiu
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Wenjuan Yan
- Department of StomatologyNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
41
|
Falero C, Huanca W, Barrios-Arpi L, Lira-Mejía B, Ramos-Coaguila O, Torres E, Ramos E, Romero A, Ramos-Gonzalez M. Oxidative and Molecular-Structural Alterations of Spermatozoa in Swine and Ram Exposed to the Triazole Ipconazole. TOXICS 2025; 13:176. [PMID: 40137503 PMCID: PMC11945538 DOI: 10.3390/toxics13030176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Triazole pesticides are widely used throughout the world, but their abuse causes toxic effects in non-targeted organisms. In the present study, the cytotoxic effect of the triazole ipconazole was evaluated in porcine and ram spermatozoa. Ipconazole significantly reduced sperm viability, increased ROS levels, altered catalase and SOD enzyme activity, and caused alterations in the molecular mRNA expression of structural biomarkers (PRM1, ODF2, AKAP4, THEG, SPACA3 and CLGN) related to fertility in males, as well as the overexpression of BAX (cell death) and ROMO1 (oxidative stress) mRNA. Our results indicate that the fungicide triazole is involved in cellular, enzymatic and molecular alteration of porcine and ram spermatozoa, and is possibly a factor in the development of infertility in male mammals.
Collapse
Affiliation(s)
- Cristian Falero
- Zootecnia an Animal Production Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru; (C.F.); (O.R.-C.)
| | - Wilfredo Huanca
- Reproduction Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru;
| | - Luis Barrios-Arpi
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru; (L.B.-A.); (B.L.-M.)
| | - Boris Lira-Mejía
- Animal Physiology Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru; (L.B.-A.); (B.L.-M.)
| | - Olger Ramos-Coaguila
- Zootecnia an Animal Production Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru; (C.F.); (O.R.-C.)
| | - Edith Torres
- Reproduction Laboratory, School of Veterinary and Zootecnic Medicine, Jorge Basadre Grohmann University, Tacna 23001, Peru;
| | - Eva Ramos
- Department of Pharmacology and Toxicology, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary, Complutense University of Madrid, 28040 Madrid, Spain; (E.R.); (A.R.)
| | - Mariella Ramos-Gonzalez
- Zootecnia an Animal Production Laboratory, Faculty of Veterinary Medicine, Major National University of San Marcos, Lima 15021, Peru; (C.F.); (O.R.-C.)
| |
Collapse
|
42
|
Vecchio E, Gallo R, Mimmi S, Gentile D, Giordano C, Straface E, Marino R, Caiazza C, Pastore A, Ruocco MR, Arcucci A, Schiavone M, Palmieri C, Iaccino E, Stornaiuolo M, Quinto I, Mallardo M, Martini F, Tognon M, Fiume G. FABP5 is a key player in metabolic modulation and NF-κB dependent inflammation driving pleural mesothelioma. Commun Biol 2025; 8:324. [PMID: 40016284 PMCID: PMC11868402 DOI: 10.1038/s42003-025-07754-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 02/17/2025] [Indexed: 03/01/2025] Open
Abstract
Pleural mesothelioma (PM) poses a significant challenge in oncology due to its intricate molecular and metabolic landscape, chronic inflammation, and heightened oxidative stress, which contribute to its notorious resilience and clinical complexities. Despite advancements, the precise mechanisms driving PM carcinogenesis remain elusive, impeding therapeutic progress. Here, we explore the interplay between tumor growth dynamics, lipid metabolism, and NF-κB dysregulation in malignant pleural mesothelioma, shedding light on novel molecular mechanisms underlying its pathogenesis. Our study reveals distinctive growth dynamics in PM cells, characterized by heightened proliferation, altered cell cycle progression, and resistance to apoptosis. Intriguingly, PM cells exhibit increased intracellular accumulation of myristic, palmitic, and stearic acids, suggestive of augmented lipid uptake and altered biosynthesis. Notably, we identify FABP5 as a key player in driving metabolic alterations and inflammation through NF-κB dysregulation in mesothelioma cells, distinguishing them from normal mesothelial cells. Silencing of FABP5 leads to significant alterations in cell dynamics, metabolism, and NF-κB activity, highlighting its potential as a therapeutic target. Our findings unveil a reciprocal relationship between lipid metabolism and inflammation in PM, providing a foundation for targeted therapeutic strategies. Overall, this comprehensive investigation offers insights into the intricate molecular mechanisms driving PM pathogenesis and identifies potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Raffaella Gallo
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Selena Mimmi
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Debora Gentile
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Caterina Giordano
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Emilio Straface
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Rossana Marino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Carmen Caiazza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Arianna Pastore
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, Zebrafish Facility, University of Brescia, Brescia, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | | | - Ileana Quinto
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Naples, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University of Catanzaro "Magna Graecia", Catanzaro, Italy.
| |
Collapse
|
43
|
Chen Z, Fan J, Chen X, Yang K, Wang K. Oxidative Stress and Redox Signaling in Gastric Cancer: From Mechanisms to Therapeutic Implications. Antioxidants (Basel) 2025; 14:258. [PMID: 40227215 PMCID: PMC11939249 DOI: 10.3390/antiox14030258] [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: 01/27/2025] [Revised: 02/18/2025] [Accepted: 02/22/2025] [Indexed: 04/15/2025] Open
Abstract
Oxidative stress, which is characterized by an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, has critical roles in the initiation, progression, and treatment of gastric cancer. On the one hand, an excessive ROS accumulation induces oxidative damage and cancer cell death. On the other hand, moderate levels of ROS cause genetic mutations and dysregulation of signaling pathways to promote proliferation, inflammation, angiogenesis, and metastasis in gastric cancer. Notably, emerging evidence has revealed that ROS also mediate oxidative post-translational modifications (oxPTMs) of redox-sensitive proteins, which can directly affect protein functions and regulate redox signaling in cancer cells. Therefore, elucidating the regulatory mechanisms of oxidative stress and redox signaling in gastric cancer holds great promise to identify novel therapeutic targets or redox-targeting strategies. This review will summarize the mechanisms of oxidative stress in regulating the hallmarks of gastric cancer and highlight the roles of ROS-mediated oxPTMs in gastric cancer. In addition, we will discuss emerging strategies targeting oxidative stress for the treatment of gastric cancer, with an emphasis on the use of bioactive natural products and nanomaterials.
Collapse
Affiliation(s)
- Zehua Chen
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiawu Fan
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
| | - Xiaolong Chen
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kun Yang
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kui Wang
- Department of General Surgery and Laboratory of Gastric Cancer, West China School of Basic Medical Sciences & Forensic Medicine, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.C.); (J.F.); (X.C.)
| |
Collapse
|
44
|
Lacerda-Abreu MA, Carvalho-Kelly LF, Meyer-Fernandes JR. Hypoxia Modulates Transmembrane Prostatic Acid Phosphatase (TM-PAP) in MCF-7 Breast Cancer Cells. Int J Mol Sci 2025; 26:1918. [PMID: 40076544 PMCID: PMC11900489 DOI: 10.3390/ijms26051918] [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: 01/16/2025] [Revised: 02/09/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
In MCF-7 breast cancer cells, transmembrane prostatic acid phosphatase (TM-PAP) plays a critical role in tumor progression, particularly under hypoxic conditions. In this study, the impact of hypoxia on ectophosphatase activity in MCF-7 cells was examined, and the underlying biological mechanisms that influence the breast cancer microenvironment were explored. Compared with normoxic cells, hypoxic cells presented significant reductions in ectophosphatase activity, indicating that hypoxia altered dephosphorylation processes critical for tumor growth and metastasis. Specific decreases in the hydrolysis of substrates, such as p-nitrophenylphosphate (pNPP) and adenosine monophosphate (AMP), were observed under hypoxic conditions, suggesting that hypoxia impaired TM-PAP activity. Further investigation revealed that hypoxia induced an increase in the concentration of reactive oxygen species (ROS), such as hydrogen peroxide (H2O2), which inhibited ectophosphatase activity. This effect was reversed by the introduction of ROS scavengers. Additionally, hypoxia activated protein kinase C (PKC), further modulating ectophosphatase activity in MCF-7 cells. Collectively, these findings enhanced the understanding of the mechanisms through which hypoxia could influence enzyme activity associated with cancer progression and provide valuable insights into the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
| | | | - José Roberto Meyer-Fernandes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-901, Brazil; (M.A.L.-A.); (L.F.C.-K.)
| |
Collapse
|
45
|
Hao Y, Wang N, Wang J, Shao S, Gao B, Tao Y, Huo L, Yan L, Wu J, Chen Z. Vacancy engineering enhanced photothermal-catalytic properties of Co 9S 8-x nanozymes for mild NIR-II hyperthermia-amplified nanocatalytic cancer therapy. J Mater Chem B 2025; 13:2480-2489. [PMID: 39829359 DOI: 10.1039/d4tb02032d] [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: 01/22/2025]
Abstract
While nanozymes are commonly employed in nanocatalytic therapy (NCT), the efficacy of NCT is hampered by the limited catalytic activity of nanozymes and the intricate tumor microenvironment (TME). In this work, we design a high-efficiency nanozyme with NIR-II photothermal property for the mild hyperthermia-augmented NCT. In order to endow a single-component nanomaterial the ability to simultaneously catalyze and exhibit NIR-II photothermal properties, a straightforward template method is utilized to fabricate sulfur vacancies (VS)-doped Co9S8-x nanocages. Introducing VS not only lowers the bandgap structure of Co9S8, enhancing its NIR-II photothermal properties, but also facilitates the control of the Co2+ and Co3+ ratio in Co9S8, leading to a boost in its catalytic activity. Furthermore, the catalytic efficiency of Co9S8-x nanocages was boosted by the mild hyperthermia. Moreover, the Co9S8-x nanocages exhibited high-efficiency GSH-px-mimic catalytic activity, facilitating the cascade amplification of ROS production. Through the integrated multifunctionality of Co9S8-x nanocages, we successfully enhanced the effectiveness of antitumor treatment with a single drug injection and a single 1064 nm laser irradiation for mild hyperthermia-augmented NCT. This work provides a distinct paradigm of endowing nanomaterials with catalytic activity and photothermal property for mild NIR-II PTT-amplified NCT through a vacancy engineering strategy.
Collapse
Affiliation(s)
- Yongyu Hao
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Nan Wang
- Department of Obstetrics and Gynecology, The First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
- Medicine School of Chinese PLA, Beijing 100853, China
| | - Jiaxu Wang
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Shuilin Shao
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Bo Gao
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Youping Tao
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Litao Huo
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Jigong Wu
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| | - Zhiming Chen
- Department of Spine Surgery, The Ninth Medical Center of PLA General Hospital, Beijing 100101, China.
| |
Collapse
|
46
|
You Y, Guo Z, Wolter T, Hu Q. Intracellular metal ion-based chemistry for programmed cell death. Chem Soc Rev 2025; 54:1552-1582. [PMID: 39744985 DOI: 10.1039/d4cs00930d] [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: 02/04/2025]
Abstract
Intracellular metal ions play essential roles in multiple physiological processes, including catalytic action, diverse cellular processes, intracellular signaling, and electron transfer. It is crucial to maintain intracellular metal ion homeostasis which is achieved by the subtle balance of storage and release of metal ions intracellularly along with the influx and efflux of metal ions at the interface of the cell membrane. Dysregulation of intracellular metal ions has been identified as a key mechanism in triggering programmed cell death (PCD). Despite the importance of metal ions in initiating PCD, the molecular mechanisms of intracellular metal ions within these processes are infrequently discussed. An in-depth understanding and review of the role of metal ions in triggering PCD may better uncover novel tools for cancer diagnosis and therapy. Specifically, the essential roles of calcium (Ca2+), iron (Fe2+/3+), copper (Cu+/2+), and zinc (Zn2+) ions in triggering PCD are primarily explored in this review, and other ions like manganese (Mn2+/3+/4+), cobalt (Co2+/3+) and magnesium ions (Mg2+) are briefly discussed. Further, this review elaborates on the underlying chemical mechanisms and summarizes these metal ions triggering PCD in cancer therapy. This review bridges chemistry, immunology, and biology to foster the rational regulation of metal ions to induce PCD for cancer therapy.
Collapse
Affiliation(s)
- Yawen You
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhaochen Guo
- Department of Biochemistry, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Tyler Wolter
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
- Institute for Clinical and Translational Research, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin Madison, Madison, WI 53705, USA.
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
47
|
Zhou Q, Guan Y, Zhao P, Chu H, Xi Y. Combined anti-leukemic effect of gilteritinib and GSK-J4 in FLT3-ITD + acute myeloid leukemia. Transl Oncol 2025; 52:102271. [PMID: 39813767 PMCID: PMC11783125 DOI: 10.1016/j.tranon.2025.102271] [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: 09/11/2024] [Revised: 12/25/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025] Open
Abstract
Gilteritinib treats acute myeloid leukemia (AML) with the FMS-like receptor tyrosine kinase-3 (FLT3) internal tandem duplication (ITD) mutation. Dysregulation of histone modification affects the genesis and progression of AML. Strategies targeting key histone regulators have not been applied to the treatment of AML. Lysine demethylase 6B (KDM6B) is dysregulated in a variety of cancers and regulates the expression of oncogenes, which has potential in anticancer therapy. We explored whether GSK-J4 (an inhibitor of the demethylase KDM6B) has an anti-leukemic effect in the gilteritinib treatment of FLT3-ITD+ AML and the effect of gilteritinib combined with GSK-J4 in leukemia. In our study, we evaluated the anti-leukemic effect of GSK-J4 in gilteritinib therapy through in vitro and in vivo experiments. The results revealed that the combined treatment of gilteritinib and GSK-J4 has greater anti-proliferation and pro-apoptosis effects than gilteritinib alone. Gilteritinib and GSK-J4 performed synergistically to arrest the cell cycle. Gilteritinib mainly induces cell cycle phase arrest at the S or G0/G1, and GSK-J4 inhibits the cell cycle progression in the S phase and reduces cell viability by reducing the expression of key regulatory factors from the G1 phase to the S phase. At the same time, GSK-J4 enhances the expression of apoptosis-related proteins (Bax and cleavage caspase-9). In addition, gilteritinib or GSK-J4 monotherapy increases reactive oxygen species (ROS) production, and the combination has a synergistic effect, accelerating leukemic cell death. Our study provides proof that the combined therapy of gilteritinib and GSK-J4 has a synergistic antileukemic effect on FLT3-ITD+ AML.
Collapse
Affiliation(s)
- Qi Zhou
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Yongyu Guan
- Clinical laboratory, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730000, China
| | - Pingping Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Huiyuan Chu
- School of Public Health, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Yaming Xi
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China; Department of Hematology, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
48
|
Pace PE, Fu L, Hampton MB, Winterbourn CC. Redox proteomic analysis of H 2O 2 -treated Jurkat cells and effects of bicarbonate and knockout of peroxiredoxins 1 and 2. Free Radic Biol Med 2025; 227:221-232. [PMID: 39489196 DOI: 10.1016/j.freeradbiomed.2024.10.314] [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: 08/01/2024] [Revised: 10/17/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
Oxidation of thiol proteins and redox signaling occur in cells exposed to H2O2 but mechanisms are unclear. We used redox proteomics to seek evidence of oxidation of specific proteins either by a mechanism involving reaction of H2O2 with CO2/bicarbonate to give the more reactive peroxymonocarbonate, or via a relay involving peroxiredoxins (Prdxs). Changes in oxidation state of specific Cys-SH residues on treating Jurkat T lymphoma cells with H2O2 were measured by isotopically labeling reduced thiols and analysis by mass spectrometry. The effects of bicarbonate and of knocking out either Prdx1 or Prdx2 were examined. Approximately 14,000 Cys-peptides were detected, of which ∼1 % underwent 2-10 fold loss in thiol content with H2O2. Those showing the most oxidation were not affected by the presence of bicarbonate or knockout of either Prdx. Consistent with previous evidence that bicarbonate potentiates inactivation of glyceraldehyde-3-phosphate dehydrogenase, the GAPDH active site Cys residues were significantly more sensitive to H2O2 when bicarbonate was present. Several other proteins were identified as promising candidates for further investigation. Although we identified some potential protein candidates for Prdx-dependent oxidation, most of the significant differences between KO and WT cells were seen in proteins for which H2O2 unexpectedly increased their CysSH content over untreated cells. We conclude that facilitation of protein oxidation by bicarbonate or Prdx-mediated relays is restricted to a small number of proteins and is insufficient to explain the majority of the oxidation of the cell thiols that occured in response to H2O2.
Collapse
Affiliation(s)
- Paul E Pace
- Mātai Hāora - Centre for Redox Biology & Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Mark B Hampton
- Mātai Hāora - Centre for Redox Biology & Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Christine C Winterbourn
- Mātai Hāora - Centre for Redox Biology & Medicine, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
49
|
Shi B, Chen J, Guo H, Shi X, Tai Q, Chen G, Yao H, Mi X, Zhong R, Lu Y, Zhao Y, Sun L, Zhou D, Yao Y, He S. ACOX1 activates autophagy via the ROS/mTOR pathway to suppress proliferation and migration of colorectal cancer. Sci Rep 2025; 15:2992. [PMID: 39849090 PMCID: PMC11757735 DOI: 10.1038/s41598-025-87728-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/21/2025] [Indexed: 01/25/2025] Open
Abstract
Acyl-CoA oxidase 1 (ACOX1), a member of the acyl-coenzyme A oxidase family, is considered a crucial regulator whose dysregulation is implicated in the occurrence and progression of various cancers. This study aims to elucidate the impact of ACOX1 in CRC, shedding light on its potential as a therapeutic target. Through analysis of the GEO dataset, it was found that ACOX1 is significantly downregulated in colorectal cancer (CRC), and this lower expression level is associated with a worse prognosis. Additionally, in vitro as well as in vivo, ACOX1 overexpression dramatically reduced the proliferation and metastasis of CRC cells. Mass spectrometry revealed the crucial role of ACOX1 in fatty acid β-oxidation, as its overexpression led to a substantial increase in reactive oxygen species (ROS) derived from fatty acid β-oxidation. Further experiments demonstrated that ACOX1 overexpression, through modulation of fatty acid metabolism, increased ROS levels, reduced the phosphorylation activation of the key autophagy regulator mTOR, enhanced autophagy, and ultimately suppressed the growth and metastasis of CRC. In conclusions, ACOX1 expression is decreased in CRC. ACOX1 may regulate autophagy by reprogramming lipid metabolism to modulate the ROS/mTOR signaling pathway, consequently inhibiting the proliferation and migration of CRC.
Collapse
Affiliation(s)
- Bo Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Junjie Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
- Department of General Surgery, Suzhou Ninth People's Hospital, Suzhou Ninth Hospital, Soochow University, Suzhou, 215200, Jiangsu, China
| | - Haoran Guo
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, P.R. China
| | - Xinyu Shi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Qingliang Tai
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Guoliang Chen
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Huihui Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Xiuwei Mi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Runze Zhong
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yang Lu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yiyuan Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Liang Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Diyuan Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
50
|
Pan Y, Matsunaga T, Zhang T, Akaike T. The Therapeutic Potential of Supersulfides in Oxidative Stress-Related Diseases. Biomolecules 2025; 15:172. [PMID: 40001475 PMCID: PMC11852411 DOI: 10.3390/biom15020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidation-reduction (redox) reactions are fundamental to sustaining life, with reactive oxygen and nitrogen species playing pivotal roles in cellular signaling and homeostasis. However, excessive oxidative stress disrupts redox balance, contributing to a wide range of diseases, including inflammatory and pulmonary disorders, neurodegeneration, and cancer. Although numerous antioxidant therapies have been developed and tested for oxidative stress-related diseases, their clinical efficacy remains limited. Here, we introduce the emerging concept of 'supersulfides', a class of redox molecule species with unique antioxidant and nucleophilic properties, which have recently been recognized as crucial regulators of cellular redox homeostasis. Unlike traditional antioxidants, supersulfides offer novel mechanisms of action that directly target the underlying processes of oxidative stress. This review summarizes current knowledge on supersulfides, highlighting their roles in oxidative stress and associated diseases, as well as the mechanisms underlying oxidative stress-related pathology. The therapeutic potential of synthetic supersulfides for treating oxidative stress-related diseases is also discussed. A comprehensive understanding of the molecular and cellular basis of redox biology can help to guide the development of innovative redox-based therapeutic strategies aimed at preventing and treating diseases associated with disturbed redox regulation.
Collapse
Grants
- 20348438 Japan Science and Technology Agency
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 21H05258 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 23K20040 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 24H00063 Ministry of Education, Culture, Sports, Science and Technology of Japan
- JP21zf0127001 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Yuexuan Pan
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| |
Collapse
|