1
|
Yan F, Chen B, Ma Z, Chen Q, Jin Z, Wang Y, Qu F, Meng Q. Exploring molecular mechanisms of postoperative delirium through multi-omics strategies in plasma exosomes. Sci Rep 2024; 14:29466. [PMID: 39604493 PMCID: PMC11603267 DOI: 10.1038/s41598-024-80865-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Currently, the diagnosis of delirium is solely based on clinical observation, lacking objective diagnostic tools, and the regulatory networks and pathological mechanisms behind it are not yet fully understood. Exosomes have garnered considerable interest as potential biomarkers for a variety of illnesses. This research aimed to delineate both the proteomic and metabolomic landscapes inherent to exosomes, assessing their diagnostic utility in postoperative delirium (POD) and understanding the underlying pathophysiological frameworks. Integrated analyses of proteomics and metabolomics were conducted on exosomes derived from plasma of individuals from both the non-postoperative delirium (NPOD) control group and the POD group. Subsequently, the study utilized the Connectivity Map (CMap) methodology for the identification of promising small-molecule drugs and carried out molecular docking assessments to explore the binding affinities with the enzyme MMP9 of these identified molecules. We identified significant differences in exosomal metabolites and proteins between the POD and control groups, highlighting pathways related to neuroinflammation and blood-brain barrier (BBB) integrity. Our CMap analysis identified potential small-molecule therapeutics, and molecular docking studies revealed two compounds with high affinity to MMP9, suggesting a new therapeutic avenue for POD. This study highlights MMP9, TLR2, ICAM1, S100B, and glutamate as key biomarkers in the pathophysiology of POD, emphasizing the roles of neuroinflammation and BBB integrity. Notably, molecular docking suggests mirin and orantinib as potential inhibitors targeting MMP9, providing new therapeutic avenues. The findings broaden our understanding of POD mechanisms and suggest targeted strategies for its management, reinforcing the importance of multidimensional biomarker analysis and molecular targeting in POD intervention.
Collapse
Affiliation(s)
- Fuhui Yan
- School of Clinical Medicine, Jining Medical University, Jining, China
| | - Bowang Chen
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China
| | - Zhen Ma
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China
| | - Qirong Chen
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China
| | - Zhi Jin
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China
| | - Yujie Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Feng Qu
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China.
| | - Qiang Meng
- Department of Intensive Care Unit, Affiliated Jining First People's Hospital of Shandong First Medical University, Jining, Shandong, China.
| |
Collapse
|
2
|
Souza MA, Zanella ML, Vanin GN, Dallepiane FG, Pizzi CYM, Ferreira ER, Fuhr MCS, Piccolo NM, Palhano HS, da Silva Koch J, Souza KR, da Costa UM, Dos Santos VV, Steier L, Bertol CD, Figueiredo JAPD. Antimicrobial action and cytotoxicity of hypochlorous acid obtained from an innovative electrolytic device - An in vitro study. Arch Oral Biol 2024; 163:105966. [PMID: 38657440 DOI: 10.1016/j.archoralbio.2024.105966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024]
Abstract
OBJECTIVE This study evaluated the antimicrobial effect and cytotoxicity of hypochlorous acid(HClO) obtained from an innovative electrolytic device. DESIGN The root canals of fifty extracted human teeth were inoculated with Enterococcus faecalis and divided into 5 groups (n = 10): DW (control); 2% chlorhexidine gel(CHX); 2.5% sodium hypochlorite(NaOCl); 250 ppm HClO and 500 ppm HClO. The counting of colony forming units evaluated the decontamination potential of each group. Cytotoxicity was evaluated after inoculation of tested protocols in fibroblastic cells for 3 min, calculating the cell viability. Specific statistical analysis was performed (α = 5%). RESULTS The highest bacterial reduction was observed in experimental groups, with no statistical differences from each other (p > 0.05). The highest number of viable cells was observed in control group, followed by 250 ppm HClO and 500 ppm HClO groups, with statistical differences from each other (p < 0.05). CONCLUSIONS It could be concluded that HClO presented high antimicrobial activity and low cytotoxicity at both tested concentrations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jordana da Silva Koch
- School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Kellyn Rocca Souza
- School of Dentistry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | - Liviu Steier
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA; Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | | | | |
Collapse
|
3
|
Cao Y, Wu B, Xu Y, Wang M, Wu X, Liang X, Lin J, Li Z, Lin H, Luo C, Chen S. Discovery of GPX4 inhibitors through FP-based high-throughput screening. Eur J Med Chem 2024; 265:116044. [PMID: 38145603 DOI: 10.1016/j.ejmech.2023.116044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/03/2023] [Accepted: 12/09/2023] [Indexed: 12/27/2023]
Abstract
Ferroptosis is a form of non-apoptotic cell death, regulated by phospholipid hydroperoxide glutathione peroxidase 4 (GPX4), a selenoprotein with a selenocysteine residue (sec) in the active site. GPX4 is a promising target for cancer cells in therapy-resistant conditions via ferroptosis, which can reduce the level of lipid reactive oxygen species (ROS). So far, all existing GPX4 inhibitors covalently bind to GPX4 via a reactive alkyl chloride moiety or masked nitrile-oxide electrophiles with poor selectivity and pharmacokinetic properties and most were obtained by cell phenotype-based screening. Lacking of effective high-throughput screening methods for GPX4 protein limits the discovery of GPX4 inhibitors. Here, we report a fluorescence polarization (FP)-based high throughput screening (HTS) assay for GPX4-U46C-C10A-C66A in vitro, and found Metamizole sodium from our in-house compound library inhibits GPX4-U46C-C10A-C66A enzyme activity. Structure-activity relationships (SAR) demonstrated the importance of sulfonyl group on interaction between Metamizole sodium and GPX4-U46C-C10A-C66A. Our FP assay could be an effective tool for discovery of GPX4 inhibitors and Metamizole sodium was a potential inhibitor for GPX4 in vitro.
Collapse
Affiliation(s)
- Yu Cao
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Bin Wu
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Ying Xu
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; China Pharmaceutical University, Nanjing, 210009, China
| | - Mingchen Wang
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Life Science and Technology, ShanghaiTech University, 100 Haike Road, Shanghai, 201210, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Xinyu Wu
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiaochen Liang
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; China Pharmaceutical University, Nanjing, 210009, China
| | - Jin Lin
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Zhihai Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Hua Lin
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Key Laboratory of Microbial Pathogenesis and Interventions of Fujian Province University, The Key Laboratory of Innate Immune Biology of Fujian Province, Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China.
| | - Cheng Luo
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China; Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528437, China.
| | - Shijie Chen
- The Center for Chemical Biology, Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
4
|
Saliba ASMC, Quirino DJG, Favaro-Trindade CS, Sartori AGDO, Massarioli AP, Lazarini JG, de Souza Silva AP, Alencar SMD. Effects of simulated gastrointestinal digestion/epithelial transport on phenolics and bioactivities of particles of brewer's spent yeasts loaded with Brazilian red propolis. Food Res Int 2023; 173:113345. [PMID: 37803652 DOI: 10.1016/j.foodres.2023.113345] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/01/2023] [Accepted: 08/03/2023] [Indexed: 10/08/2023]
Abstract
Red propolis from northeast Brazil contains mainly isoflavonoids as bioactive compounds, and its consumption may counteract unregulated and exacerbated formation of reactive oxygen species and inflammatory cytokines/chemokines. Moreover, the production of particles using sustainable carriers have been studied to increase the use of propolis as a functional food ingredient. Hence, the objective of this work was to investigate the effects of simulated gastrointestinal digestion followed by a cell-based epithelial transport on phenolic profile, anti-inflammatory and antioxidant activities of particles of brewer's spent yeasts (BSY) loaded with ethanolic extract of Brazilian red propolis (EEP). As a result, the EEP phenolic diversity decreased throughout the simulated gastrointestinal system, and was modulated by the particle production, as detected by high-performance liquid chromatography - electrospray ionization - quadrupole-time-of-flight-mass spectrometry (HPLC-ESI-QTOF-MS). Concomitantly, the antioxidant activity, as assessed by the ability to scavenge peroxyl and superoxide radicals, hydrogen peroxide, and hypochlorous acid, generally decreased at a higher extent for the particles of EEP with BSY (EEP-BSY) throughout the experiments. Nonetheless, after epithelial transport through the Caco-2 cell monolayer, the basolateral fraction of both EEP-BSY and EEP decreased the activation of pro-inflammatory transcription factor NF-κB by 83% and 65%, respectively, as well as the release of TNF-α (up to 51% and 38%, respectively), and CXCL2/MIP-2 (up to 33% and 25%, respectively). Therefore, BSY may be an interesting carrier for EEP bioencapsulation, since it preserves its anti-inflammatory activity. Further studies should be encouraged to investigate the feasibility of adding it in formulations of functional foods, considering its effect on sensory attributes.
Collapse
Affiliation(s)
| | | | | | | | - Adna Prado Massarioli
- Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, CEP: 13418-900, Brazil
| | - Josy Goldoni Lazarini
- Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, CEP: 13418-900, Brazil
| | - Anna Paula de Souza Silva
- Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, CEP: 13418-900, Brazil
| | - Severino Matias de Alencar
- Centro de Energia Nuclear na Agricultura, Universidade de São Paulo, CEP: 13416-000, Piracicaba, Brazil; Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, CEP: 13418-900, Brazil.
| |
Collapse
|
5
|
Freires IA, Morelo DFC, Soares LFF, Costa IS, de Araújo LP, Breseghello I, Abdalla HB, Lazarini JG, Rosalen PL, Pigossi SC, Franchin M. Progress and promise of alternative animal and non-animal methods in biomedical research. Arch Toxicol 2023; 97:2329-2342. [PMID: 37394624 DOI: 10.1007/s00204-023-03532-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 05/24/2023] [Indexed: 07/04/2023]
Abstract
Cell culture and invertebrate animal models reflect a significant evolution in scientific research by providing reliable evidence on the physiopathology of diseases, screening for new drugs, and toxicological tests while reducing the need for mammals. In this review, we discuss the progress and promise of alternative animal and non-animal methods in biomedical research, with a special focus on drug toxicity.
Collapse
Affiliation(s)
- Irlan Almeida Freires
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil.
| | - David Fernando Colon Morelo
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | | | | | | | | | - Henrique Ballassini Abdalla
- Laboratory of Neuroimmune Interface of Pain Research, São Leopoldo Mandic Institute and Research Center, Campinas, SP, Brazil
| | - Josy Goldoni Lazarini
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
| | - Pedro Luiz Rosalen
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil
- Graduate Program in Biological Sciences, Federal University of Alfenas, Alfenas, Brazil
| | | | - Marcelo Franchin
- School of Dentistry, Federal University of Alfenas, Alfenas, Brazil
- Bioactivity and Applications Lab, Department of Biological Sciences, Faculty of Science and Engineering, School of Natural Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
6
|
Basappa B, Jung YY, Ravish A, Xi Z, Swamynayaka A, Madegowda M, Pandey V, Lobie PE, Sethi G, Ahn KS. Methyl-Thiol-Bridged Oxadiazole and Triazole Heterocycles as Inhibitors of NF-κB in Chronic Myelogenous Leukemia Cells. Biomedicines 2023; 11:1662. [PMID: 37371757 DOI: 10.3390/biomedicines11061662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/29/2023] Open
Abstract
Nuclear factor kappa beta (NF-κB) is a transcriptional factor that plays a crucial role in regulating cancer cell proliferation. Therefore, the inhibition of NF-κB activity by small molecules may be beneficial in cancer therapy. In this report, methyl-thiol-bridged oxadiazole and triazole heterocycles were synthesized via click chemistry and it was observed that the lead structure, 2-(((1-(3,4-dichlorophenyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5-(4-methoxybenzyl)-1,3,4-oxadiazole (4c), reduced the viability of MCF-7 cells with an IC50 value of 7.4 µM. Compound 4c also caused concentration-dependent loss of cell viability in chronic myelogenous leukemia (CML) cells. Furthermore, compound 4c inhibited the activation of NF-κB in human CML cells as observed by nuclear translocation and DNA binding assays. Functionally, compound 4c produced PARP cleavage and also suppressed expression of Bcl-2/xl, MMP-9, COX-2, survivin, as well as VEGF, resulting in apoptosis of CML cells. Moreover, ChIP assay showed that compound 4c decreased the binding of COX-2 to the p65 gene promoter. Detailed in silico analysis also indicated that compound 4c targeted NF-κB in CML cells. In conclusion, a novel structure bearing both triazole and oxadiazole moieties has been identified that can target NF-κB in CML cells and may constitute a potential novel drug candidate.
Collapse
Affiliation(s)
- Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India
| | - Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Akshay Ravish
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore 570006, India
| | - Zhang Xi
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Ananda Swamynayaka
- Department of Studies in Physics, University of Mysore, Mysore 570006, India
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Mysore 570006, India
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Peter E Lobie
- Shenzhen Bay Laboratory, Shenzhen 518055, China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|