1
|
Ouyang W, Yan D, Hu J, Liu Z. Multifaceted mitochondrial as a novel therapeutic target in dry eye: insights and interventions. Cell Death Discov 2024; 10:398. [PMID: 39242592 PMCID: PMC11379830 DOI: 10.1038/s41420-024-02159-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/09/2024] Open
Abstract
Dry eye, recognized as the most prevalent ocular surface disorder, has risen to prominence as a significant public health issue, adversely impacting the quality of life for individuals across the globe. Despite decades of extensive research into the chronic inflammation that characterizes dry eye, the intricate mechanisms fueling this persistent inflammatory state remain incompletely understood. Among the various cellular components under investigation, mitochondria-essential for cellular energy production and homeostasis-have attracted increasing attention for their role in dry eye pathogenesis. This involvement points to mechanisms such as oxidative stress, apoptosis, and sustained inflammation, which are central to the progression of the disease. This review aims to provide a thorough exploration of mitochondrial dysfunction in dry eye, shedding light on the critical roles played by mitochondrial oxidative stress, apoptosis, and mitochondrial DNA damage. It delves into the mechanisms through which diverse pathogenic factors may trigger mitochondrial dysfunction, thereby contributing to the onset and exacerbation of dry eye. Furthermore, it lays the groundwork for an overview of current therapeutic strategies that specifically target mitochondrial dysfunction, underscoring their potential in managing this complex condition. By spotlighting this burgeoning area of research, our review seeks to catalyze the development of innovative drug discovery and therapeutic approaches. The ultimate goal is to unlock promising avenues for the future management of dry eye, potentially revolutionizing treatment paradigms and improving patient outcomes. Through this comprehensive examination, we endeavor to enrich the scientific community's understanding of dry eye and inspire novel interventions that address the underlying mitochondrial dysfunctions contributing to this widespread disorder.
Collapse
Affiliation(s)
- Weijie Ouyang
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Dan Yan
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Xiamen, Fujian, China
| | - Jiaoyue Hu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China.
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, Eye Institute of Xiamen University, School of Medicine of Xiamen University, Department of Ophthalmology of Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China; Department of Ophthalmology, the First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China.
| |
Collapse
|
2
|
Zhao D, Zhao H, He Y, Zhang M. BMSC Alleviates Dry Eye by Inhibiting the ROS-NLRP3-IL-1β Signaling Axis by Reducing Inflammation Levels. Curr Eye Res 2024; 49:698-707. [PMID: 38450655 DOI: 10.1080/02713683.2024.2324434] [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: 08/17/2023] [Accepted: 02/24/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Bone marrow mesenchymal stem cells (BMSC) have multiple biological functions and are widely involved in regulating inflammatory diseases, tissue repair and regeneration. However, the mechanism of their action in dry eye disease (DED) is currently unclear. The purpose of this study was to investigate the effect of BMSCs in the treatment of dry eye mice and to explore its specific therapeutic mechanism. METHODS Mouse corneal epithelial cells (MCECs) were treated with 500 mOsM sodium chloride hypertonic solution to induce a DED cell model. The dry eye animal model was constructed by adding 5 μL 0.2% benzalkonium chloride solution to mouse eyes. Western blotting was used to detect the expression of related proteins, and flow cytometry, enzyme-linked immunosorbent assay (ELISA), terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining, hematoxylin-eosin (HE) staining, and periodic acid schiff (PAS) staining were used to detect cell and eye tissue damage. RESULTS The experimental results showed that BMSCs can reduce the levels of reactive oxygen species (ROS) and inflammatory factors in MCECs, promote cell proliferation, inhibit cell apoptosis, improve the integrity of the corneal epithelial layer in vivo, promote an increase in the number of goblet cells, and alleviate DED. Further exploration of the molecular mechanism of BMSCs treatment revealed that BMSCs alleviate the progression of DED by inhibiting the ROS-NLRP3-IL-1β signaling pathway. CONCLUSION BMSCs inhibit ROS-NLRP3-IL-1β signaling axis, reducing inflammation levels and alleviating dry eye symptoms. These findings provide new ideas and a basis for the treatment of DED and provide an experimental basis for further research on the application value of BMSCs in alleviating DED.
Collapse
Affiliation(s)
- Dandan Zhao
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Hongxia Zhao
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Yang He
- Ophthalmology Department, Yan'An Hospital of Kunming City, Kunming, China
| | - Meixia Zhang
- Ophthalmology Department, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Bu J, Liu Y, Zhang R, Lin S, Zhuang J, Sun L, Zhang L, He H, Zong R, Wu Y, Li W. Potential New Target for Dry Eye Disease-Oxidative Stress. Antioxidants (Basel) 2024; 13:422. [PMID: 38671870 PMCID: PMC11047456 DOI: 10.3390/antiox13040422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Dry eye disease (DED) is a multifactorial condition affecting the ocular surface. It is characterized by loss of tear film homeostasis and accompanied by ocular symptoms that may potentially result in damage to the ocular surface and even vision loss. Unmodifiable risk factors for DED mainly include aging, hormonal changes, and lifestyle issues such as reduced sleep duration, increased screen exposure, smoking, and ethanol consumption. As its prevalence continues to rise, DED has garnered considerable attention, prompting the exploration of potential new therapeutic targets. Recent studies have found that when the production of ROS exceeds the capacity of the antioxidant defense system on the ocular surface, oxidative stress ensues, leading to cellular apoptosis and further oxidative damage. These events can exacerbate inflammation and cellular stress responses, further increasing ROS levels and promoting a vicious cycle of oxidative stress in DED. Therefore, given the central role of reactive oxygen species in the vicious cycle of inflammation in DED, strategies involving antioxidants have emerged as a novel approach for its treatment. This review aims to enhance our understanding of the intricate relationship between oxidative stress and DED, thereby providing directions to explore innovative therapeutic approaches for this complex ocular disorder.
Collapse
Affiliation(s)
- Jinghua Bu
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yanbo Liu
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Rongrong Zhang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Sijie Lin
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Jingbin Zhuang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Le Sun
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Lingyu Zhang
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Hui He
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Rongrong Zong
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
| | - Yang Wu
- Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China
| | - Wei Li
- Department of Ophthalmology, Xiang’an Hospital of Xiamen University, Eye Institute of Xiamen University, Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Fujian Engineering and Research Center of Eye Regenerative Medicine, School of Medicine, Xiamen University, Xiamen 361005, China
- Xiamen University Affiliated Xiamen Eye Center, Xiamen 361102, China
| |
Collapse
|
4
|
Dong Y, Ding YY, Gao WP. Puerarin alleviates hyperosmotic stress-induced oxidative stress, inflammation, apoptosis and barrier damage of human corneal epithelial cells by targeting SIRT1/NLRP3 signaling. Toxicol In Vitro 2024; 94:105722. [PMID: 37865300 DOI: 10.1016/j.tiv.2023.105722] [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: 07/28/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
The increase of tear osmolarity caused by excessive evaporation of tear phase is the main pathological mechanism of dry eye disease (DED). Puerarin, the major bioactive ingredient isolated from the root of the Pueraria lobata (Willd.) Ohwi, has been reported to improve ophthalmic diseases in clinic. However, the effect and the potential regulatory mechanism related to silent information regulator sirtuin 1 (SIRT1)/NOD-like receptor family pyrin domain containing 3 (NLRP3) signaling of puerarin in DED has not been evaluated. In this study, we aimed to explore the effect and mechanism of hyperosmotic stress (Hyp)-induced human corneal epithelial cell line (HCE-2). The viability of HCE-2 cells induced by Hyp with or without puerarin treatment was assessed by a CCK-8 assay. Results indicated that puerarin treatment enhanced cell viability, reduced reactive oxygen species (ROS) content, increased CAT and SOD activities, and elevated the ratio of GSH/GSSG in HCE-2 cells exposed to Hyp. Besides, TNF-α, IL-1β and IL-6 contents were decreased by puerarin. Additionally, puerarin inhibited Hyp-induced apoptosis and barrier disruption of HCE-2 cells. Moreover, molecular docking method suggested that puerarin bound to SIRT1, and upregulated SIRT1 and downregulated NLRP3 inflammasome proteins after puerarin treatment was observed. Furthermore, SIRT1 silencing alleviated the protective effects of puerarin on Hyp-induced HCE-2 cell damage. Collectively, puerarin attenuates Hyp-induced injury of HCE-2 cells by targeting regulating SIRT1/NLRP3 signaling.
Collapse
Affiliation(s)
- Yue Dong
- Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province 210046, China; Department of Ophthalmology, Yangzhou Hospital affiliated to Nanjing University of Chinese Medicine, Yangzhou City, Jiangsu Province 225009, China
| | - Yin-Yin Ding
- Department of Ophthalmology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province 210022, China
| | - Wei-Ping Gao
- Department of Ophthalmology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province 210029, China.
| |
Collapse
|
5
|
Zhao D, Zhao H, He Y, Zhang M. BMSC reduces ROS and inflammation levels by inhibiting TLR4/MYD88/NF-κB signaling axis to alleviate dry eye. RESEARCH SQUARE 2023:rs.3.rs-2739871. [PMID: 37131693 PMCID: PMC10153363 DOI: 10.21203/rs.3.rs-2739871/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Objective To investigate the therapeutic effect of Bone marrow mesenchymal stem cells (BMSCs) on dry eye mice, and to investigate the mechanism of TLR4/MYD88/NF-κB signaling pathway on corneal injury repair in dry eye mice. Methods To establish a hypertonic dry eye cell model. Western blot for measureing the protein expressions of caspase-1, IL-1β,NLRP3 and ASC,and Rt-qpcr for mRNA expression. Flow cytometry for detecting the ROS content and apoptosis rate. CCK-8 for detecting the proliferation activity of cells, and ELISA for the levels of inflammation-related factors.The levels of inflammation-related factors were detected by ELISA. The dry eye mouse model of benzalkonium chloride was established. Three clinical parameters used to evaluate ocular surface damage, namely tear secretion, tear film rupture time and corneal sodium fluorescein staining, were measured with phenol cotton thread. Flow cytometry and TUNEL staining are both for he apoptosis rate. Western blot also for detecting the protein expressions of TLR4, MYD88, NF-κB, inflammation-related factors and apoptosis-related factors . The pathological changes were evaluated by HE and PAS staining. Results In vitro, BMSCs and inhibitors of TLR4, MYD88 and NF-κB showed decreased ROS content, decreased inflammatory factor protein level, decreased apoptotic protein level and increased mRNA expression compared with NaCl group. BMSCS partially reversed cell apoptosis induced by NaCl and improved cell proliferation. In vivo, it reduces corneal epithelial defects, goblet cell loss and inflammatory cytokine production, and increases tear production. In vitro, BMSC and inhibitors of TLR4, MYD88 and NF-κB could protect mice from apoptosis induced by hypertonic stress. In terms of mechanism, NACL-induced NLRP3 inflammasome formation, caspase-1 activation and IL-1β maturation can be inhibited. Conclusion BMSCs treatment can reduce ROS and inflammation levels and alleviate dry eye by inhibiting TLR4/MYD88/NF-κBsignaling pathway.
Collapse
|
6
|
Shen J, Liang Y, Bi Z, Yin X, Chen C, Zhao X, Liu S, Li Y. Cyclosporin A improves the hyperosmotic response in an experimental dry eye model by inhibiting the HMGB1/TLR4/NF-κB signaling pathway. Exp Eye Res 2023; 229:109418. [PMID: 36806672 DOI: 10.1016/j.exer.2023.109418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/29/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Hyperosmolarity is closely related to dry eye disease (DED), which induces corneal epithelial cell structure and dysfunction leading to ocular surface inflammation. Cyclosporine A (CSA) is a cyclopeptide consisting of 11 deduced amino acids. It has an immunosuppressive effect and shows a vital function in inhibiting the inflammatory response. The mechanism of CSA in DED is still not entirely clear. This experiment aimed to investigate the possible mechanism of CSA in the hyperosmotic DED model. This study found that CSA can inhibit the transcript levels of DED high mobility group protein 1 (HMGB1), Toll-like receptor 4 (TLR4) and nuclear transcription factor κB (NF-κB) in signaling pathways. In addition, the study also found that 550 mOsm/L can induce the formation of DED models in vivo or in vitro. Furthermore, different concentrations of CSA have different effects on the expression of HMGB1 in human corneal epithelial cells under hyperosmotic stimulation, and high concentrations of CSA may increase the expression of HMGB1. In addition, CSA effectively reduced the corneal fluorescence staining score of the DE group and increased the tear volume of mice. Therefore, this experimental investigation might supply new evidence for the mechanism of CSA in DED, provide a potential new therapy for treating DED, and provide a theoretical basis for CSA treatment of DED.
Collapse
Affiliation(s)
- Jiachao Shen
- Department of Ophthalmology, Binzhou Medical College, Yantai, 264000, China; Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Yan Liang
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Zhaojing Bi
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xin Yin
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Chen Chen
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Xinmei Zhao
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China
| | - Shujun Liu
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China.
| | - Yuanbin Li
- Department of Ophthalmology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, China.
| |
Collapse
|
7
|
Gange WS, Qiao JB, Park PJ, McDonnell JF, Tan Z, Perlman JI, Bu P. Protection of Retinal Function by Nucleoside Reverse Transcriptase Inhibitors Following Retinal Ischemia/Reperfusion Injury. J Ocul Pharmacol Ther 2021; 37:485-491. [PMID: 34448620 DOI: 10.1089/jop.2020.0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: Retinal ischemia/reperfusion (I/R) injury is a common cause of visual impairment and blindness for which there remain limited treatment options. Nucleoside reverse transcriptase inhibitors (NRTIs), such as zidovudine (AZT), have been shown to block the NLRP3 inflammasome and prevent retinal degeneration in a mouse model of age-related macular degeneration. The NLRP3 inflammasome has also been shown to be triggered in I/R injury. Therefore, we studied the neuroprotective effects of AZT using a pressure-induced retinal ischemia mouse model. Methods: C57BL/6J mice were randomly assigned to 1 of 2 treatment groups: vehicle-treated retinal I/R injury (n = 6) or AZT-treated retinal I/R injury (n = 6). Vehicle (1% dimethyl sulfoxide [DMSO] in phosphate-buffered saline [PBS]) or AZT 50 mg/kg in 1% DMSO in PBS were injected intraperitoneally twice daily for 5 days. On day 2 of treatment, retinal ischemia was induced by transient elevation of intraocular pressure for 45 min. Scotopic electroretinography (ERG) was used to quantify retinal function before and 1 week after retinal ischemic insult. Retinal morphology was examined 1 week after ischemic insult. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assays and caspase 1 immunostaining was performed 24 h after retinal I/R injury. Results: Following I/R injury, ERG a- and b-wave amplitudes were significantly reduced in the vehicle-treated mice. AZT treatment significantly attenuated I/R-induced loss of retinal function as compared with vehicle-treated mice. Additionally, AZT-treated mice experienced significantly less inner retinal thinning as compared with vehicle-treated mice. TUNEL-positive cells were prevalent in the vehicle-treated I/R injury mouse retinas compared with the AZT-treated I/R injury mouse retinas. More caspase-1 immunoreactivity was detected in ganglion cell layer and inner nuclear layer (INL) in vehicle-treated I/R injury group than in AZT-treated I/R injury group. Conclusion: AZT treatment resulted in relative preservation of retinal structure and function following ischemic insult as compared with controls. This suggests AZT may have therapeutic value in the management of retinal ischemic diseases.
Collapse
Affiliation(s)
- William S Gange
- Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - James B Qiao
- Health Sciences Division, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Paul J Park
- Health Sciences Division, Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - James F McDonnell
- Health Sciences Division, Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Zhiqun Tan
- Institute for Neurological Impairments and Neurological Disorders, University of California Irvine, Irvine, California, USA
| | - Jay I Perlman
- Health Sciences Division, Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA.,Surgery Service and Edward Hines Jr. VA Hospital, Hines, Illinois, USA
| | - Ping Bu
- Health Sciences Division, Department of Ophthalmology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA.,Research Service, Edward Hines Jr. VA Hospital, Hines, Illinois, USA
| |
Collapse
|
8
|
Wubulikasimu M, Muhammad T, Imerhasan M, Hudaberdi N, Yang W, Zhao J, Peng X. Synthesis of fluorescent drug molecules for competitive binding assay based on molecularly imprinted polymers. RSC Adv 2019; 9:6779-6784. [PMID: 35518505 PMCID: PMC9061092 DOI: 10.1039/c9ra00422j] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 02/13/2019] [Indexed: 11/26/2022] Open
Abstract
Fluorescent immunosorbent assay (FIA) is very promising for sensitive and selective analysis in bio-medical applications. Here, we proposed an assay, using fluorescent engineering of analytes and the corresponding molecularly imprinted polymers (MIPs) as a plastic antibody. Three drug molecules (metronidazole, zidovudine and lamivudine) were condensed with 9-aminoacridine, using succinic anhydride as a spacer. The target products were characterized with 1H-NMR, IR and mass spectrometry. UV-vis absorption and fluorescent properties of the fluorophore-labeled drug molecules were investigated. Feasibility of the fluorescent biomimetic immunosorbent assay based on MIPs was demonstrated in the solution. This work will provide sound foundation for the future application in real sample.
Collapse
Affiliation(s)
- Muyasier Wubulikasimu
- College of Chemistry & Chemical Engineering, Xinjiang University, Xinjiang Key Laboratory of Oil and Gas Fine Chemicals Urumqi 830046 P. R. China
| | - Turghun Muhammad
- College of Chemistry & Chemical Engineering, Xinjiang University, Xinjiang Key Laboratory of Oil and Gas Fine Chemicals Urumqi 830046 P. R. China
| | - Mukhtar Imerhasan
- College of Chemistry & Chemical Engineering, Xinjiang University, Xinjiang Key Laboratory of Oil and Gas Fine Chemicals Urumqi 830046 P. R. China
| | | | - Wenwu Yang
- College of Chemistry & Chemical Engineering, Xinjiang University, Xinjiang Key Laboratory of Oil and Gas Fine Chemicals Urumqi 830046 P. R. China
| | - Jianzhang Zhao
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 P. R. China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Dalian University of Technology Dalian 116024 P. R. China
| |
Collapse
|