1
|
Yu T, Gao S, Jin F, Yan B, Wang W, Wang Z. Characteristics of the vaginal microbiota and vaginal metabolites in women with cervical dysplasia. Front Cell Infect Microbiol 2024; 14:1457216. [PMID: 39450338 PMCID: PMC11499233 DOI: 10.3389/fcimb.2024.1457216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/13/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Emerging evidence suggests that the vaginal microbiota is closely associated with cervical cancer. However, little is known about the relationships among the vaginal microbiota, vaginal metabolites, and cervical lesion progression in women undergoing cervical dysplasia. Methods In this study, to understand vaginal microbiota signatures and vaginal metabolite changes in women with cervical lesions of different grades and cancer, individuals with normal or cervical dysplasia were recruited and divided into healthy controls (HC) group, low-grade squamous intraepithelial lesions (LSIL) group, high-grade squamous intraepithelial lesions (HSIL) group, and cervical cancer (CC) group. Vaginal secretion samples were collected for 16S rRNA gene sequencing, liquid chromatography coupled with mass spectrometry (LC-MS)-based metabolomics, and integrated analysis. Results The results demonstrated that bacterial richness and diversity were greater in the CC group than the other three groups. Additionally, Lactobacillus was found to be negatively associated with bacterial diversity and bacterial metabolic functions, which increased with the degree of cervical lesions and cancer. Metabolomic analysis revealed that distinct metabolites were enriched in these metabolite pathways, including tryptophan metabolism, retinol metabolism, glutathione metabolism, alanine, aspartate, and glutamate metabolism, as well as citrate cycle (TCA cycle). Correlation analysis revealed positive associations between CC group-decreased Lactobacillus abundance and CC group-decreased metabolites. Lactobacillus iners was both negative to nadB and kynU genes, the predicted abundance of which was significantly higher in the CC group. The linear regression model showed that the combination of the vaginal microbiota and vaginal metabolites has good diagnostic performance for cervical cancer. Discussion Our results indicated a clear difference in the vaginal microbiota and vaginal metabolites of women with cervical dysplasia. Specifically altered bacteria and metabolites were closely associated with the degree of cervical lesions and cancer, indicating the potential of the vaginal microbiota and vaginal metabolites as modifiable factors and therapeutic targets for preventing cervical cancer.
Collapse
Affiliation(s)
- Tiantian Yu
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Shan Gao
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Fen Jin
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Bingbing Yan
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Women and Children’s Medical Group, Dalian, China
| | - Wendong Wang
- Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
| | - Zhongmin Wang
- Female Pelvic Floor Urinary Reconstructive Center, Dalian Women and Children’s Medical Group, Dalian, China
| |
Collapse
|
2
|
Zhong T, Zhang Y, Yang H, Luo F, Xu Y, Fan Y, Liu J. Phenylpropanoid-rich maize root extract serves as a natural antidepressant. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155893. [PMID: 39111191 DOI: 10.1016/j.phymed.2024.155893] [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: 02/26/2024] [Revised: 06/27/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND Depression is a serious and complex mental disease that has attracted worldwide attention because of its high incidence rate, high disability rate and high mortality. Excitotoxicity is one of the most important mechanisms involved in the pathophysiological process of depression. In our previous studies, n-butanol extract from maize roots was found to have good neuroprotective effects due to its antioxidative activity. However, the antidepressive effective constituents, efficacy in vivo and mechanism of action of maize root extracts have not been determined. PURPOSE This study aimed to determine the main active neuroprotective compound in maize root extract and investigate its antidepressant effects and possible underlying mechanism in vitro and in vivo. METHODS Sixteen extracts were isolated and purified from maize roots. The active components of the most active extracts of maize roots (hereafter referred to as EM 2) were identified using UF-HPLC-QTOF/MS. In vitro cell models of NMDA-induced excitotoxicity in SH-SY5Y cells were used to analyze the anti-excitatory activity of the extracts. The MTT assay and Annexin V-FITC/PI Apoptosis Detection were used to evaluate cell viability. Several network pharmacological strategies have been employed to investigate the potential mechanism of action of EM 2. The effects of EM 2 on depressive-like behaviors were evaluated in CUMS mice. Changes in the levels of related proteins were detected via western blotting. RESULTS Among the 16 extracts extracted by n-butanol, EM 2 was determined to be the most active extract against NMDA-induced excitotoxicity by n-butanol extraction. Meanwhile, seventeen compounds were further identified as the main active components of EM 2. Mechanistically, EM 2 inhibited NMDA-induced excitatory injury in SH-SY5Y cells and alleviated the depressive-like behaviors of CUMS mice by suppressing NR2B and subsequently mediating the downstream CREB/TRKB/BDNF, PI3K/Akt and MAPK pathways, as well as the Nrf2/HO-1 antioxidant signaling pathway. CONCLUSION The study indicated that EM 2 could potentially be developed as a potential therapeutic candidate to cure depression in NMDA-induced excitatory damage.
Collapse
Affiliation(s)
- Ting Zhong
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Yunqiang Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Huiyin Yang
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Fang Luo
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China
| | - Yongnan Xu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| | - Yanhua Fan
- State Key Laboratory for Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, PR China; Natural Products Research Center of Guizhou Province, Guiyang 550014, PR China.
| | - Jianyu Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
3
|
Iriawati I, Vitasasti S, Rahmadian FNA, Barlian A. Isolation and characterization of plant-derived exosome-like nanoparticles from Carica papaya L. fruit and their potential as anti-inflammatory agent. PLoS One 2024; 19:e0304335. [PMID: 38959219 PMCID: PMC11221653 DOI: 10.1371/journal.pone.0304335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 05/10/2024] [Indexed: 07/05/2024] Open
Abstract
Inflammation is an immune system response that identifies and eliminates foreign material. However, excessive and persistent inflammation could disrupt the healing process. Plant-derived exosome-like nanoparticles (PDENs) are a promising candidate for therapeutic application because they are safe, biodegradable and biocompatible. In this study, papaya PDENs were isolated by a PEG6000-based method and characterized by dynamic light scattering (DLS), transmission Electron Microscopy (TEM), bicinchoninic acid (BCA) assay method, GC-MS analysis, total phenolic content (TPC) analysis, and 2,2-diphenyl-1-picrylhydrazyl (DPPH) assay. For the in vitro test, we conducted internalization analysis, toxicity assessment, determination of nitrite concentration, and assessed the expression of inflammatory cytokine genes using qRT-PCR in RAW 264.7 cells. For the in vivo test, inflammation was induced by caudal fin amputation followed by analysis of macrophage and neutrophil migration in zebrafish (Danio rerio) larvae. The result showed that papaya PDENs can be well isolated using the optimized differential centrifugation method with the addition of 30 ppm pectolyase, 15% PEG, and 0.2 M NaCl, which exhibited cup-shaped and spherical morphological structure with an average diameter of 168.8±9.62 nm. The papaya PDENs storage is stable in aquabidest and 25 mM trehalose solution at -20˚C until the fourth week. TPC estimation of all papaya PDENs ages did not show a significant change, while the DPPH test exhibited a significant change in the second week. The major compounds contained in Papaya PDENs is 2,3-dihydro-3,5-dihydroxy-6-methyl-4H-pyran-4-one (DDMP). Papaya PDENs can be internalized and is non-cytotoxic to RAW 264.7 cells. Moreover, LPS-induced RAW 264.7 cells treated with papaya PDENs showed a decrease in NO production and downregulation mRNA expression of pro-inflammatory cytokine genes (IL-1B and IL-6) and an upregulation in mRNA expression of anti-inflammatory cytokine gene (IL-10). In addition, in vivo tests conducted on zebrafish treated with PDENs papaya showed inhibition of macrophage and neutrophil cell migration. These findings suggest that PDENs papaya possesses anti-inflammatory properties.
Collapse
Affiliation(s)
- Iriawati Iriawati
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, Indonesia
| | - Safira Vitasasti
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, Indonesia
| | | | - Anggraini Barlian
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung, Indonesia
| |
Collapse
|
4
|
Ahmad N, Lesa KN, Ujiantari NSO, Sudarmanto A, Fakhrudin N, Ikawati Z. Development of White Cabbage, Coffee, and Red Onion Extracts as Natural Phosphodiesterase-4B (PDE4B) Inhibitors for Cognitive Dysfunction: In Vitro and In Silico Studies. Adv Pharmacol Pharm Sci 2024; 2024:1230239. [PMID: 38808119 PMCID: PMC11132833 DOI: 10.1155/2024/1230239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 04/15/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Human cognition fundamentally depends on memory. Alzheimer's disease exhibits a strong correlation with a decline in this factor. Phosphodiesterase-4 B (PDE4B) plays a crucial role in neurodegenerative disorders, and its inhibition is one of the promising approaches for memory enhancement. This study aimed to identify secondary metabolites in white cabbage, coffee, and red onion extracts and identify their molecular interaction with PDE4B by in silico and in vitro experiments. Crushed white cabbage and red onion were macerated separately with ethanol to yield respective extracts, and ground coffee was boiled with water to produce aqueous extract. Thin layer chromatography (TLC)-densitometry was used to examine the phytochemicals present in white cabbage, coffee, and red onion extracts. Molecular docking studies were performed to know the interaction of test compounds with PDE4B. TLC-densitometry analysis showed that chlorogenic acid and quercetin were detected as major compounds in coffee and red onion extracts, respectively. In silico studies revealed that alpha-tocopherol (binding free energy (∆Gbind) = -38.00 kcal/mol) has the strongest interaction with PDE4B whereas chlorogenic acid (∆Gbind = -21.50 kcal/mol) and quercetin (∆Gbind = -17.25 kcal/mol) exhibited moderate interaction. In vitro assay showed that the combination extracts (cabbage, coffee, and red onion) had a stronger activity (half-maximal inhibitory concentration (IC50) = 0.12 ± 0.03 µM) than combination standards (sinigrin, chlorogenic acid, and quercetin) (IC50 = 0.17 ± 0.03 µM) and rolipram (IC50 = 0.15 ± 0.008 µM). Thus, the combination extracts are a promising cognitive enhancer by blocking PDE4B activity.
Collapse
Affiliation(s)
- Nazir Ahmad
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
| | - Kaisun Nesa Lesa
- Department of Food and Nutritional Science, Khulna City Corporation Women's College, Affiliated to Khulna University, Khulna, Bangladesh
- Department of Food and Agricultural Product Technology, Faculty of Agricultural Technology, Universitas Gadjah Mada, Yogyakarta 55281, Indonesia
- Department of Pediatrics, Nihon University Hospital, Tokyo, Japan
- Department of Nutrition and Food Technology, Jessore University of Science and Technology, Jessore, Bangladesh
| | - Navista Sri Octa Ujiantari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
| | - Ari Sudarmanto
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
| | - Nanang Fakhrudin
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
- Medicinal Plants and Natural Products Research Center, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Sleman 55281, Yogyakarta, Indonesia
| | - Zullies Ikawati
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia
| |
Collapse
|
5
|
Basharat Z, Meshal A. Pan-genome mediated therapeutic target mining in Kingella kingae and inhibition assessment using traditional Chinese medicinal compounds: an informatics approach. J Biomol Struct Dyn 2024; 42:2872-2885. [PMID: 37144759 DOI: 10.1080/07391102.2023.2208221] [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: 02/21/2023] [Accepted: 04/23/2023] [Indexed: 05/06/2023]
Abstract
Kingella kingae causes bacteremia, endocarditis, osteomyelitis, septic arthritis, meningitis, spondylodiscitis, and lower respiratory tract infections in pediatric patients. Usually it demonstrates disease after inflammation of mouth, lips or infections of the upper respiratory tract. To date, therapeutic targets in this bacterium remain unexplored. We have utilized a battery of bioinformatics tools to mine these targets in this study. Core genes were initially inferred from 55 genomes of K. kingae and 39 therapeutic targets were mined using an in-house pipeline. We selected aroG product (KDPG aldolase) involved in chorismate pathway, for inhibition analysis of this bacterium using lead-like metabolites from traditional Chinese medicinal plants. Pharmacophore generation was done using control ZINC36444158 (1,16-bis[(dihydroxyphosphinyl)oxy]hexadecane), followed by molecular docking of top hits from a library of 36,000 compounds. Top prioritized compounds were ZINC95914016, ZINC33833283 and ZINC95914219. ADME profiling and simulation of compound dosing (100 mg tablet) was done to infer compartmental pharmacokinetics in a population of 300 individuals in fasting state. PkCSM based toxicity analysis revealed the compounds ZINC95914016 and ZINC95914219 as safe and with almost similar bioavailability. However, ZINC95914016 takes less time to reach maximum concentration in the plasma and shows several optimal parameters compared to other leads. In light of obtained data, we recommend this compound for further testing and induction in experimental drug design pipeline.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
| | - Alotaibi Meshal
- Department of Pharmacy Practice, College of Pharmacy, University of Hafr Albatin, Saudi Arabia
| |
Collapse
|
6
|
Bian X, Chen L, Bian X, Li L, Liu D, Liu S, Xu L, Huo X, Yang X. Protective effect of Tibetan medicine Qiwei Tiexie pills on liver injury induced by acetaminophen overdose: An integrated strategy of network pharmacology, metabolomics and transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155221. [PMID: 38039903 DOI: 10.1016/j.phymed.2023.155221] [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: 07/24/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND Drug-induced liver injury, particularly from acetaminophen (APAP), has emerged as a significant public health concern. Unfortunately, there is currently no effective treatment strategy available. Qiwei Tiexie pills (QWTX), a traditional Tibetan medicine, have demonstrated considerable clinical efficacy in treating various liver diseases. Nevertheless, the protective effect of QWTX against drug-induced liver injury and its underlying mechanism remains poorly understood. PURPOSE This study aimed to assess the therapeutic potential of QWTX, a Tibetan medicine, in an animal model of APAP-induced liver injury. Additionally, we sought to investigate the molecular mechanism through which QWTX exerts its effects. METHODS We employed LC-MS and network pharmacology to predict the potential targets of QWTX in drug-induced liver injury. Subsequently, we employed HE staining, transcriptomics, metabolomics, and qRT-PCR to analyze the mechanism underlying QWTX treatment in drug-induced liver injury. RESULTS Network pharmacology analysis revealed that the active components of QWTX are involved in inflammatory and drug metabolism-related pathways. In mouse models, pretreatment with QWTX effectively mitigated the elevated levels of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and inflammatory factors (IL-1β, IL-6, and TNF-α) induced by APAP overdose. Moreover, APAP inhibited 1459 differentially expressed genes (DEGs) and 874 differential accumulation metabolites (DAMs), while QWTX promoted their expression. Conversely, APAP promoted 874 genes and 119 metabolites, which were inhibited by QWTX. Further analysis demonstrated that QWTX ameliorated the metabolic disorders induced by APAP overdose and potentially exerted a protective effect by inhibiting the expression of critical genes in crucial inflammatory pathways. QWTX also up-regulated antioxidant enzymes, thereby mitigating the oxidative stress resulting from APAP overdose. CONCLUSION QWTX treatment effectively protects against APAP-induced liver damage in mice. Transcriptomic and metabolomic analyses further revealed that QWTX ameliorated hepatic metabolic disorders induced by APAP overdose while significantly suppressing the inflammatory response and oxidative stress associated with drug-induced liver injury. This study provides a new insight into the treatment of drug-induced liver injury by the TCM system and provides a basis for the development of new therapies for drug-induced liver injury by QWTX and its active ingredients.
Collapse
Affiliation(s)
- Xingbo Bian
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Lizhu Chen
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Xuefeng Bian
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Lele Li
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Dan Liu
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Siying Liu
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Lu Xu
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Xuyang Huo
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China
| | - Xiaohang Yang
- College of Pharmacy, Jilin Medical University, Jilin 132013, Jilin, China.
| |
Collapse
|
7
|
Hao L, Zhong X, Yu R, Chen J, Li W, Chen Y, Lu W, Wu J, Wang P. Integrating Network Pharmacology and Experimental Validation to Decipher the Anti-Inflammatory Effects of Magnolol on LPS-induced RAW264.7 Cells. Comb Chem High Throughput Screen 2024; 27:462-478. [PMID: 37818577 DOI: 10.2174/0113862073255964230927105959] [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: 04/14/2023] [Revised: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Magnolol is beneficial against inflammation-mediated damage. However, the underlying mechanisms by which magnolol exerts anti-inflammatory effects on macrophages remain unclear. OBJECTIVE In this study, network pharmacology and experimental validation were used to assess the effect of magnolol on inflammation caused by lipopolysaccharide (LPS) in RAW264.7 cells. MATERIALS AND METHODS Genes related to magnolol were identified in the PubChem and Swiss Target Prediction databases, and gene information about macrophage polarization was retrieved from the GeneCards, OMIM, and PharmGKB databases. Analysis of protein-protein interactions was performed with STRING, and Cytoscape was used to construct a component-target-disease network. GO and KEGG enrichment analyses were performed to ascertain significant molecular biological processes and signaling pathways. LPS was used to construct the inflammatory cell model. ELISA and qRT.PCR were used to examine the expression levels of inflammationassociated factors, immunofluorescence was used to examine macrophage markers (CD86 and CD206), and western blotting was used to examine protein expression levels. RESULTS The hub target genes of magnolol that act on macrophage polarization were MDM2, MMP9, IL-6, TNF, EGFR, AKT1, and ERBB2. The experimental validation results showed that magnolol treatment decreased the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6). Moreover, the levels of anti-inflammatory factors (IL-10 and IL-4) were increased. In addition, magnolol upregulated the expression of M2 markers (Agr-1, Fizzl, and CD206) and downregulated M1 markers (CD86). The cell experiment results supported the network pharmacological results and demonstrated that magnolol alleviated inflammation by modulating the PI3k-Akt and P62/keap1/Nrf2 signaling pathways. CONCLUSION According to network pharmacology and experimental validation, magnolol attenuated inflammation in LPS-induced RAW264.7 cells mainly by inhibiting M1 polarization and enhancing M2 polarization by activating the PI3K/Akt and P62/keap1/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Lei Hao
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runjia Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuzhong Chen
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqi Lu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianyu Wu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peizong Wang
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P.R. China
| |
Collapse
|
8
|
Xing Z, Yang C, Feng Y, He J, Peng C, Li D. Understanding aconite's anti-fibrotic effects in cardiac fibrosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 122:155112. [PMID: 37924690 DOI: 10.1016/j.phymed.2023.155112] [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: 04/17/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND The prevalence of cardiac fibrosis, intricately linked to various cardiovascular diseases, continues to rise. Aconite, a traditional Chinese herb renowned for its cardiovascular benefits, holds promise in treating heart ailments. However, the mechanisms underlying its anti-fibrotic effects, particularly in cardiac fibrosis, remain elusive. HYPOTHESIS/PURPOSE This study aims to shed light on aconite's potential as an anti-fibrotic agent and elucidate its mechanisms in a rat model of isoproterenol (ISO)-induced cardiac fibrosis. METHODS By inducing cardiac fibrosis through ISO injection, the study investigates the role of decoction of white aconite (DWA) in mitigating fibrotic processes. Techniques including metabolomics, RT-qPCR, western blot, and immunofluorescence were employed to unveil the molecular changes induced by DWA. RESULTS DWA exhibited a remarkable reduction in echocardiographic parameters, cardiac weight increase, myocardial infarction extent, inflammatory cell infiltration, collagen deposition in heart tissue, and serum CK-MB, cTnT, cTnI levels post ISO injection. Metabolomic analysis unveiled DWA's modulation of 27 metabolites, especially in galactose metabolism, addressing metabolic disturbances in cardiac fibrosis. Additionally, DWA suppressed mRNA expression of fibrosis markers (Collagen I, CTGF, TGF-β), inhibited protein levels of MMP-9, α-SMA, and Galectin-3, while elevating TIMP1 expression. CONCLUSION DWA demonstrated potent anti-fibrotic effects by curbing collagen deposition and alleviating metabolic disruptions in cardiac fibrosis via the galactose metabolism pathway, possibly mediated by the Gal-3/TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Ziwei Xing
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, China
| | - Chao Yang
- National Engineering Research Center for Marine Aquaculture, Institute of Innovation & Application, Zhejiang Ocean University, Zhoushan, China
| | - Yaqian Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, China
| | - Junyao He
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, China.
| | - Dan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, 1166 Liutai Avenue, Wenjiang District, Chengdu, China.
| |
Collapse
|
9
|
Liu F, Li J, Zhou B, Shen Y, Tang J, Han J, Chen C, Shao K, Chen H, Yuan L. The Role of Emodin in the Treatment of Bladder Cancer Based on Network Pharmacology and Experimental Verification. Comb Chem High Throughput Screen 2024; 27:1661-1675. [PMID: 38504574 DOI: 10.2174/0113862073294990240122140121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND AND PURPOSE Emodin, a compound derived from rhubarb and various traditional Chinese medicines, exhibits a range of pharmacological actions, including antiinflammatory, antiviral, and anticancer properties. Nevertheless, its pharmacological impact on bladder cancer (BLCA) and the underlying mechanism are still unclear. This research aimed to analyze the pharmacological mechanisms of Emodin against BLCA using network pharmacology analysis and experimental verification. METHODS Initially, network pharmacology was employed to identify core targets and associated pathways affected by Emodin in bladder cancer. Subsequently, the expression of key targets in normal bladder tissues and BLCA tissues was assessed by searching the GEPIA and HPA databases. The binding energy between Emodin and key targets was predicted using molecular docking. Furthermore, in vitro experiments were carried out to confirm the predictions made with network pharmacology. RESULTS Our analysis identified 148 common genes targeted by Emodin and BLCA, with the top ten target genes including TP53, HSP90AA1, EGFR, MYC, CASP3, CDK1, PTPN11, EGF, ESR1, and TNF. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses indicated a significant correlation between Emodin and the PI3KAKT pathway in the context of BLCA. Molecular docking investigations revealed a strong affinity between Emodin and critical target proteins. In vitro experiments demonstrated that Emodin inhibits T24 proliferation, migration, and invasion while inducing cell apoptosis. The findings also indicated that Emodin reduces both PI3K and AKT protein and mRNA expression, suggesting that Emodin may mitigate BLCA by modulating the PI3K-AKT signaling pathway. CONCLUSION This study integrates network pharmacology with in vitro experimentation to elucidate the potential mechanisms underlying the action of Emodin against BLCA. The results of this research enhance our understanding of the pharmacological mechanisms by which Emodin may be employed in treating BLCA.
Collapse
Affiliation(s)
- Fule Liu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Jianghao Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Boruo Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Yang Shen
- Department of Urology, The Second Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Second Chinese Medicine Hospital, Jiangsu, 210017, China
| | - Jingyuan Tang
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Jie Han
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Changpeng Chen
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Kang Shao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Haojie Chen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| | - Lin Yuan
- Department of Urology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210004, China
| |
Collapse
|
10
|
Li Z, Lee JE, Cho N, Yoo HM. Anti-viral effect of usenamine a using SARS-CoV-2 pseudo-typed viruses. Heliyon 2023; 9:e21742. [PMID: 38027904 PMCID: PMC10656252 DOI: 10.1016/j.heliyon.2023.e21742] [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: 06/18/2023] [Revised: 09/09/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
The escalating pandemic brought about by the novel SARS-CoV-2 virus is threatening global health, and thus, it is necessary to develop effective antiviral drugs. Usenamine A is a dibenzo-furan derivative separated from lichen Usnea diffracta showing broad-spectrum activity against different viruses. We evaluate that usenamine A has antiviral effects against novel SARS-CoV-2 Delta variant pseudotyped viruses (PVs) in A549 cells. In addition, usenamine A significantly suppresses SARS-CoV-2 PV-induced mitochondrial depolarization, elevated reactive oxygen species (ROS) levels, apoptosis, and inflammation. Usenamine A also causes the SARS-CoV-2 spike protein to become less stable. Thus, usenamine A shows potential as an antiviral drug that can provide protection against COVID-19.
Collapse
Affiliation(s)
- Zijun Li
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, South Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Joo-Eun Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, South Korea
| | - Hee Min Yoo
- Biometrology Group, Korea Research Institute of Standards and Science (KRISS), Daejeon 34113, South Korea
- Department of Precision Measurement, University of Science and Technology (UST), Daejeon 34113, South Korea
| |
Collapse
|
11
|
Lê HG, Kang JM, Võ TC, Na BK. Kaempferol induces programmed cell death in Naegleria fowleri. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154994. [PMID: 37597363 DOI: 10.1016/j.phymed.2023.154994] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/21/2023] [Accepted: 07/22/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Naegleria fowleri is a brain-eating amoeba causing a fatal brain infection called primary amoebic meningoencephalitis (PAM). Despite its high mortality over 95%, effective therapeutic drug for PAM has not been developed yet. Therefore, development of an effective and safe therapeutic drug for PAM is urgently needed. In this study, we investigated anti-amoebic effect of kaempferol (KPF) against N. fowleri and its underlying anti-amoebic molecular mechanisms. METHODS Anti-amoebic activity of KPF against N. fowleri trophozoites, as well as cytotoxicity of KPF in C6 glial cells and CHO-K1 cells were investigated. The programmed cell death mechanisms in KPF-treated N. fowleri were also analyzed by apoptosis-necrosis assay, mitochondrial dysfunction assay, TUNEL assay, RT-qPCR, and CYTO-ID assay. RESULTS KPF showed anti-amoebic activity against N. fowleri trophozoites with an IC50 of 29.28 ± 0.63 μM. However, it showed no significant cytotoxicity to mammalian cells. KPF induced significant morphological alterations of the amoebae, resulting in death. Signals associated with apoptosis were detected in the amoebae upon treatment with KPF. KPF induced an increase of intracellular reactive oxygen species level, loss of mitochondrial membrane potential, increases of expression levels of genes associated with mitochondria dysfunction, and reduction of ATP levels in the amoebae. Autophagic vacuole accumulations with increased expression levels of autophagy-related genes were also detected in KPF-treated amoebae. CONCLUSION KPF induces programmed cell death in N. fowleri trophozoites via apoptosis-like pathway and autophagy pathway. KPF could be used as a candidate of anti-amoebic drug or supplement compound in the process of developing or optimizing therapeutic drug for PAM.
Collapse
Affiliation(s)
- Hương Giang Lê
- Department of Parasitology and Tropical Medicine, Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Jung-Mi Kang
- Department of Parasitology and Tropical Medicine, Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Tuấn Cường Võ
- Department of Parasitology and Tropical Medicine, Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Byoung-Kuk Na
- Department of Parasitology and Tropical Medicine, Institute of Health Science, Gyeongsang National University College of Medicine, Jinju 52727, Korea; Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea.
| |
Collapse
|
12
|
Quimque MTJ, Go AD, Lim JAK, Vidar WS, Macabeo APG. Mycobacterium tuberculosis Inhibitors Based on Arylated Quinoline Carboxylic Acid Backbones with Anti- Mtb Gyrase Activity. Int J Mol Sci 2023; 24:11632. [PMID: 37511390 PMCID: PMC10380224 DOI: 10.3390/ijms241411632] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/30/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
New antitubercular agents with either a novel mode of action or novel mode of inhibition are urgently needed to overcome the threat of drug-resistant tuberculosis (TB). The present study profiles new arylated quinoline carboxylic acids (QCAs) having activity against replicating and non-replicating Mycobacterium tuberculosis (Mtb), the causative agent of TB. Thus, the synthesis, characterization, and in vitro screening (MABA and LORA) of 48 QCAs modified with alkyl, aryl, alkoxy, halogens, and nitro groups in the quinoline ring led to the discovery of two QCA derivatives, 7i and 7m, adorned with C-2 2-(naphthalen-2-yl)/C-6 1-butyl and C-2 22-(phenanthren-3-yl)/C-6 isopropyl, respectively, as the best Mtb inhibitors. DNA gyrase inhibition was shown to be exhibited by both, with QCA 7m illustrating better activity up to a 1 μM test concentration. Finally, a docking model for both compounds with Mtb DNA gyrase was developed, and it showed a good correlation with in vitro results.
Collapse
Affiliation(s)
- Mark Tristan J Quimque
- The Graduate School, University of Santo Tomas, España Blvd., Manila 1015, Philippines
- Laboratory for Organic Reactivity, Discovery and Synthesis (Rm. 410), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Blvd., Manila 1015, Philippines
- Chemistry Department, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, Iligan City 9200, Philippines
| | - Adrian D Go
- Laboratory for Organic Reactivity, Discovery and Synthesis (Rm. 410), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Blvd., Manila 1015, Philippines
| | - Justin Allen K Lim
- Laboratory for Organic Reactivity, Discovery and Synthesis (Rm. 410), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Blvd., Manila 1015, Philippines
| | - Warren S Vidar
- Laboratory for Organic Reactivity, Discovery and Synthesis (Rm. 410), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Blvd., Manila 1015, Philippines
| | - Allan Patrick G Macabeo
- Laboratory for Organic Reactivity, Discovery and Synthesis (Rm. 410), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Blvd., Manila 1015, Philippines
| |
Collapse
|
13
|
Liu Y, Zhang X, Yang L, Zhou S, Li Y, Shen Y, Lu S, Zhou J, Liu Y. Proteomics and transcriptomics explore the effect of mixture of herbal extract on diabetic wound healing process. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154892. [PMID: 37267693 DOI: 10.1016/j.phymed.2023.154892] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/28/2023] [Accepted: 05/16/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND The annual incidence of diabetic foot ulcers (DFUs) has been reported to vary from 0.2% to 11% in diabetes-specific clinical settings and less than 0.1% to 8% in community- and population-based cohorts. According to the International Diabetes Foundation, approximately 40 million to 60 million people worldwide are affected by DFUs, and a recent meta-analysis indicates a global prevalence of 6.3% among adults with diabetes, or about 33 million individuals. The cost of diabetes care is significant, amounting to $273 billion in direct and $90 billion in indirect expenses annually, in America. Foot complications in diabetes care excess annual expenditures ranging from 50% to 200% above the baseline cost of diabetes-related care. The cost of advanced-stage ulcers can be more than $50,000 per wound episode, and the direct expenses of major amputation are even higher. DFUs can be treated using various methods, including wound dressings, antibiotics, pressure-off loading, skin substitutes, stem cells, debridement, topical oxygen therapy, gene therapy and growth factors. For severe DFUs patients are at risk of amputation if treatment is not timely or appropriate. Amputating limbs not only causes physical pain to patients, but also brings economic burden due to lost productivity, and decreased employment linked to DFUs. Currently, long-term use of local antibiotics in clinical practice is prone to induce drug resistance, while growth factors do not effectively inhibit bacterial growth and control inflammation in wounds. Stem cell and gene therapies are still in the experimental stage. The method of local debridement combined with negative pressure therapy is expensive. Therefore, we urgently need an affordable, non-surgical method to treat diabetic ulcers. Extracts of bark of Bauhinia purpurea, Paeoniae rubrae, Angelica dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav. (Hoffm.) Benth. & Hook.f. ex Franch. & Sav., Acorus calamus L, and Radix Angelicae biseratae have been used as traditional remedies to treat inflammation-related diseases and cutaneous wounds due to their anti-inflammatory properties and their ability to promote vascular renewal. However, there have been few studies on the mixture of these five herbal extracts on diabetic wound healing. PURPOSE This study was designed to assess the healing effect of a mixture of five aforementioned herbal extracts on diabetic ulcer wounds in rats, and to reveal the potential mechanisms behind any potential wound healing using transcriptomics and proteomics. STUDY DESIGN We designed the experiment to explore the effects of five herbal extracts on diabetic wound healing process through in vivo experiments and to investigate the underlying mechanisms through proteomics and transcriptomics. METHODS We used a mixture of five aforementioned herbal extract to treat rat model of diabetic established by intraperitoneal injection of streptozotocin, and a 2 × 2 cm round full-thickness skin defect was created on the back of the rat. Staphylococcus aureus (1 ml of 1.5 × 109 cfu/ml) was evenly applied to the wound. The wound was then observed for 72 h. The infected ulcer model of diabetic rats was considered to be successfully established if the wound was found to be infected with S. aureus. According to different medications, the rats were divided into three groups, namely mixture of herbal extract (MHE), Kangfuxin solution (KFS) and control (Ctrl). The effects of the medicine on wound healing were observed. HE staining and Masson staining were performed to evaluate the histopathological changes and collagen synthesis. IHC staining was used to assess the neovascularization, and M2 macrophage proliferation was determined by immunofluorescence staining. Proteomic and transcriptomic studies were performed to explore potential mechanism of five herbal extracts to promote wound healing. UHPLC-QE-MS was performed to identify the chemical composition of mixture of herbal extract. RESULTS The study show that the mixed herbal extract promotes angiogenesis, proliferation of M2 macrophages, and collagen synthesis. Transcriptomics showed that rno-miR-1298, rno-miR-144-5p, and rno-miR-92a-1-5p are vital miRNAs which also play a significant role in role in regulating wound healing. Proteomics results showed that the following proteins were important in wounds treated with MHE: Rack1, LOC100362366, Cops2, Cops6, Eif4e, Eif3c, Rpl12, Srp54, Rpl13 and Lsm7. Autophagy, PI3-Akt and mTOR signaling pathways were enriched after treatment with MHE compared to other groups. CONCLUSION Herein, we have shown that MHE containing extracts of bark of Bauhinia purpurea, P. rubrae, A. dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav., A. calamus L, and R. A. biseratae has significant wound healing effects in the diabetic ulcer wound rat model. These results suggest that local application of MHE in diabetic wounds can accelerate the wound healing process. Moreover, in vivo experiments revealed that the diabetic wound healing process was primarily mediated by angiogenesis and M2 macrophage transition. Therefore, this study may provide a promising and non-surgical therapeutic strategy to accelerate diabetic wound healing, thereby decreasing the number of limb amputations in diabetic patients.
Collapse
Affiliation(s)
- Yang Liu
- Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010107, China; Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Xi Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Liping Yang
- Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010107, China
| | - Shuai Zhou
- Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010107, China
| | - Yuewei Li
- Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010107, China
| | - Yiyu Shen
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Shengli Lu
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China
| | - Jianda Zhou
- Department of Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, China.
| | - Yu Liu
- Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, 010107, China; Hunan University of Chinese Medicine, Changsha, 410007, China.
| |
Collapse
|
14
|
Tang D, Wang C, Gu Z, Li J, Jin L, Li J, Wang Z, Jiang RW. Discovery of anti-allergic components in Guomingkang Formula using sensitive HEMT biochips coupled with in vitro and in vivo validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 115:154837. [PMID: 37126969 DOI: 10.1016/j.phymed.2023.154837] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 04/04/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Allergic rhinitis (AR) is a prevalent allergic disease, which seriously affects the sufferers' life quality and increases the socioeconomic burden. Guominkang (GMK), a well-known prescription for AR treatment, showed satisfactory effects; while its anti-allergic components remain to be disclosed. AlGaN/GaN HEMT biochip is more sensitive and cost-effective than other binding equipments, indicating its great potential for screening of active ingredients from herbal medicines. METHODS AR mouse models were first established to test the anti-allergic effect of GMK and discover the ingredients absorbed into blood by ultra-high performance liquid chromatography-mass spectra (UHPLC-MS). Then, novel Syk/Lyn/Fyn-functionalized high electron mobility transistor (HEMT) biochips with high sensitivity and specificity were constructed and applied to screen the active components. Finally, the results from HEMT biochips screening were validated via in silico (molecular docking and molecular dynamics simulation), in vitro (RBL-2H3 cells), and in vivo (PCA mice model) assays. RESULTS GMK showed a potent therapeutic effect on AR mice, and fifteen components were identified from the medicated plasma. Furthermore, hamaudol was firstly found to selectively inhibit the Syk and Lyn, and emodin was to selectively inhibit Lyn, which were further confirmed by isothermal titration calorimetry, molecular docking, and molecular dynamics simulation analyses. Suppression of the activation of FcεRI-MAPK signals might be the possible mechanism of the anti-allergic effect of hamaudol. CONCLUSIONS The targets of emodin and hamaudol were discovered by HEMT biochips for the first time. This study provided a novel and effective strategy to discover active components in a complex herbal formula by using AlGaN/GaN HEMT biochips.
Collapse
Affiliation(s)
- Ding Tang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 511436, PR China; Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Chen Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 511436, PR China
| | - Zhiqi Gu
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, PR China
| | - Jiadong Li
- Key Laboratory of Multifunctional Nanomaterials and Smart Systems, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215125, PR China
| | - Lu Jin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 511436, PR China
| | - Juan Li
- Key Laboratory of Ministry of Education on Traditional Chinese Medicine Resource and Compound Prescription, Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, Hubei 430065, China
| | - Zhixin Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 511436, PR China.
| | - Ren-Wang Jiang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research and International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education, College of Pharmacy, Jinan University, Guangzhou 511436, PR China.
| |
Collapse
|
15
|
Sabzian-Molaei F, Hosseini S, Alipour A, Ghaderi H, Fotouhi-Chahouki F, Hadi A, Shahsavarani H. Urtica dioica agglutinin (UDA) as a potential candidate for inhibition of SARS-CoV-2 Omicron variants: In silico prediction and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154648. [PMID: 36681052 PMCID: PMC9815882 DOI: 10.1016/j.phymed.2023.154648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND The high number of mutations and consequent structure modifications in a Receptor-Binding Domain (RBD) of the spike protein of the Omicron variant of SARS-CoV-2 increased concerns about evading neutralization by antibodies induced by previous infection or vaccination. Thus, developing novel drugs with potent inhibitory activity can be considered an alternative for treating this highly transmissible variant. Considering that Urtica dioica agglutinin (UDA) displays antiviral activity against SARS-CoV-2, the potency of this lectin to inhibit the Receptor Binding Domain of the Omicron variant (RBDOmic) was examined in this study. PURPOSE This study examines how UDA inhibits the Omicron variant of SARS-CoV-2 by blocking its RBD, using a combination of in silico and experimental methods. METHODS To investigate the interaction between UDA and RBDOmic, the CLUSPRO 2.0 web server was used to dock the RBDOmic-UDA complex, and molecular dynamics simulations were performed by the Gromacs 2020.2 software to confirm the stability of the selected docked complex. Finally, the binding affinity (ΔG) of the simulation was calculated using MM-PBSA. In addition, ELISA and Western blot tests were used to examine UDA's binding to RBDOmic. RESULTS Based on the docking results, UDA forms five hydrogen bonds with the RBDOmic active site, which contains mutated residues Tyr501, Arg498, Arg493, and His505. According to MD simulations, the UDA-RBDOmic complex is stable over 100 ns, and its average binding energy during the simulation is -87.201 kJ/mol. Also, the ELISA test showed that UDA significantly binds to RBDOmic, and by increasing the concentration of UDA protein, the attachment to RBDOmic became stronger. In Western blotting, RBDOmic was able to attach to and detect UDA. CONCLUSION This study indicates that UDA interaction with RBDOmic prevents virus attachment to Angiotensin-converting enzyme 2 (ACE2) and, therefore, its entry into the host cell. Altogether, UDA exhibited a significant suppression effect on the Omicron variant and can be considered a new candidate to improve protection against severe infection of this variant.
Collapse
Affiliation(s)
- Fatemeh Sabzian-Molaei
- Laboratory of Regenerative Medicine & Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | | | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran.
| | - Hajarossadat Ghaderi
- Laboratory of Regenerative Medicine & Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran
| | | | - Amin Hadi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Hosein Shahsavarani
- Laboratory of Regenerative Medicine & Biomedical Innovations, Pasteur Institute of Iran, Tehran, Iran; Department of Cell and Molecular Biology, Faculty of Life science and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
16
|
Notarte KI, Quimque MTJ, Macaranas IT, Khan A, Pastrana AM, Villaflores OB, Arturo HC, Pilapil IV DYH, Tan SMM, Wei DQ, Wenzel-Storjohann A, Tasdemir D, Yen CH, Ji SY, Kim GY, Choi YH, Macabeo APG. Attenuation of Lipopolysaccharide-Induced Inflammatory Responses through Inhibition of the NF-κB Pathway and the Increased NRF2 Level by a Flavonol-Enriched n-Butanol Fraction from Uvaria alba. ACS OMEGA 2023; 8:5377-5392. [PMID: 36816691 PMCID: PMC9933231 DOI: 10.1021/acsomega.2c06451] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/18/2023] [Indexed: 05/12/2023]
Abstract
Pathologic hyperreactive inflammatory responses occur when there is excessive activation of a proinflammatory NF-κB pathway and a reduced cytoprotective NRF2 cascade. The noncytotoxic, highly selective COX-2 inhibitory flavonol-enriched butanol fraction (UaB) from Uvaria alba (U. alba) was investigated for its inflammatory modulating potential by targeting NF-κB activation and NRF2 activity. Enzyme-linked immunosorbent assay was initially performed to measure levels of proinflammatory mediators [nitric oxide (NO), prostaglandin E2, and reactive oxygen species (ROS)] and cytokines [tumor necrosis factor-alpha (TNF-α), IL-1β, and IL-6], followed by reverse transcription-polymerase chain reaction and western blotting to determine mRNA and protein expression, respectively. Using immunofluorescence staining combined with western blot analysis, the activation of NF-κB was further investigated. NRF2 activity was also measured using a luciferase reporter assay. UaB abrogated protein and mRNA expressions of inducible nitric oxide synthase (iNOS), COX-2, TNF-α, IL-1β, and IL-6 in RAW 264.7 macrophages, thereby suppressing the production of proinflammatory mediators and cytokines. This was further validated when a concentration-dependent decrease in NO and ROS production was observed in zebrafish (Danio rerio) larvae. UaB also increased NRF2 activity in HaCaT/ARE cell line and attenuated NF-κB activation by inhibiting the nuclear translocation of transcription factor p65 in RAW 264.7 macrophages. Nontargeted LC-MS analysis of UaB revealed the presence of the flavonols quercitrin (1), quercetin (2), rutin (3), kaempferol (4), and kaempferol 3-O-rutinoside (5). Molecular docking indicates that major flavonol aglycones have high affinity toward COX-2 NSAID-binding sites, TNF-α, and TNF-α converting enzyme, while the glycosylated flavonoids showed strong binding toward iNOS and IKK-all possessing dynamic stability when performing molecular dynamics simulations at 140 ns. This is the first report to have elucidated the mechanistic anti-inflammatory potential of the Philippine endemic plant U. alba.
Collapse
Affiliation(s)
- Kin Israel
R. Notarte
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore, Maryland21218, United
States
| | - Mark Tristan J. Quimque
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
- Chemistry
Department, College of Science and Mathematics, Mindanao State University - Iligan Institute of Technology, Tibanga, 9200Iligan City, Philippines
| | - Imee T. Macaranas
- Faculty
of Medicine and Surgery, University of Santo
Tomas, España, 1008Manila, Philippines
| | - Abbas Khan
- Department
of Bioinformatics and Biostatistics, State Key Laboratory of Microbial
Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road Shanghai, Minhang
District, Shanghai200240, China
| | - Adriel M. Pastrana
- Faculty
of Medicine and Surgery, University of Santo
Tomas, España, 1008Manila, Philippines
| | - Oliver B. Villaflores
- Laboratory
of Phytochemistry, Research Center for Natural and Applied Sciences, University of Santo Tomas, España, 1015Manila, Philippines
| | - Hans Christian
P. Arturo
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
| | - Delfin Yñigo H. Pilapil IV
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
| | - Sophia Morgan M. Tan
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
| | - Dong-Qing Wei
- Department
of Bioinformatics and Biostatistics, State Key Laboratory of Microbial
Metabolism, Shanghai Jiao Tong University, 800 Dongchuan Road Shanghai, Minhang
District, Shanghai200240, China
| | - Arlette Wenzel-Storjohann
- GEOMAR
Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine
Natural Product Chemistry, GEOMAR Helmholtz Centre for Ocean Research
Kiel, 24106Kiel, Germany
| | - Deniz Tasdemir
- GEOMAR
Centre for Marine Biotechnology (GEOMAR-Biotech), Research Unit Marine
Natural Product Chemistry, GEOMAR Helmholtz Centre for Ocean Research
Kiel, 24106Kiel, Germany
- Faculty
of Mathematics and Natural Sciences, Kiel
University, 24118Kiel, Germany
| | - Chia-Hung Yen
- National
Natural Product Libraries and High-Throughput Screening Core Facility, Kaohsiung Medical University, Kaohsiung80708, Taiwan
| | - Seon Yeong Ji
- Department
of Biochemistry, Dongeui University College
of Korean Medicine, 52-57,
Yangjeong-ro, Busanjin-gu, Busan47227Republic of Korea
| | - Gi-Young Kim
- Department
of Marine Life Science, Jeju National University, 102 Jejudaehak-ro, Jeju-si, Jeju Special Self-Governing Province63243, Republic of Korea
| | - Yung Hyun Choi
- Department
of Biochemistry, Dongeui University College
of Korean Medicine, 52-57,
Yangjeong-ro, Busanjin-gu, Busan47227Republic of Korea
| | - Allan Patrick G. Macabeo
- Laboratory
of Organic Reactivity, Discovery and Synthesis (LORDS), Research Center
for Natural and Applied Sciences, University
of Santo Tomas, España, 1015Manila, Philippines
- ;
| |
Collapse
|
17
|
Oxyresveratrol from mulberry branch extract protects HUVECs against oxidized Low-density Lipoprotein-induced oxidative injury via activation of the Nrf-2/HO-1 pathway. J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
18
|
Quimque MT, Magsipoc RJY, Llames LCJ, Flores AIG, Garcia KY, Ratzenböck A, Hussain H, Macabeo APG. Polyoxygenated Cyclohexenes from Uvaria grandiflora with Multi-Enzyme Targeting Properties Relevant in Type 2 Diabetes and Obesity. ACS OMEGA 2022; 7:36856-36864. [PMID: 36278100 PMCID: PMC9583304 DOI: 10.1021/acsomega.2c05544] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
Shikimic acid-derived polyoxygenated cyclohexene natural products commonly occurring in several species of the Uvaria represent natural products with promising biological activities. While a number of derivatives have been reported from Uvaria grandiflora (U. grandiflora), further studies are needed to discover additional bioactive congeners, particularly derivatives with multi-protein target inhibitory properties implicated in diseases such as diabetes and obesity. In this paper, isolation and identification of a new highly oxygenated cyclohexene, uvagrandol (1), along with the known compound (-)-zeylenone (2) from the DCM sub-extract of U. grandiflora following in vitro and in silico assessment of their enzyme inhibitory properties against α-glucosidase, dipeptidyl peptidase IV, porcine lipase, and human recombinant monoacylglycerol lipase are reported. The structure of 1 was elucidated using 1D and 2D NMR data analysis. The absolute configuration of 1 was established by quantum chemical calculations via the Gauge-Independent Atomic Orbital (GIAO) NMR method followed by TDDFT-Electronic Circular Dichroism (ECD) calculations. The structures of the eight possible stereoisomers were optimized by means of DFT calculations (B3LYP/6-31+G[d,p] in vacuum), and then their isotropic shielding tensors were obtained using the GIAO method at mPW1PW91/6-31G(d,p) in chloroform. Through DP4+, the isomer of configuration (1S,2S,3R,6R) for 1 was predicted with 96.3% probability. Compounds 1 and 2 significantly inhibited the four target enzymes in vitro. Binding studies through molecular docking simulations showed strong binding affinities for (-)-zeylenone (2), thus validating the in vitro results. Our findings suggest the potential of polyoxygenated cyclohexenes, in particular (-)-zeylenone (2), in anti-diabetic and anti-obesity drug discovery.
Collapse
Affiliation(s)
- Mark Tristan
J. Quimque
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
- The
Graduate School, University of Santo Tomas, Espana Street, Manila1015, Philippines
- Department
of Chemistry, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, Iligan City9200, Philippines
| | - Ryan Joseph Y. Magsipoc
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
| | - Lloyd Christian J. Llames
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
| | - Angeli Izza G. Flores
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
| | - Katherine Yasmin
M. Garcia
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
| | - Andreas Ratzenböck
- Institut
für Organische Chemie, Universität
Regensburg, Universitätstrasse
31, RegensburgD-93053, Germany
| | - Hidayat Hussain
- Leibniz-Institut
für Pflanzenbiochemie, Weinberg 3, HalleD-06120, Germany
| | - Allan Patrick G. Macabeo
- Laboratory
for Organic Reactivity, Discovery and Synthesis (LORDS), Research
Center for the Natural and Applied Sciences, University of Santo Tomas, Espana Street, Manila1015, Philippines
| |
Collapse
|
19
|
Yan T, Zhu X, Zhang X, Jia X, Liu J, Wang X, Xiao Y, Xiao Z, Liu T, Dong Y. The application of proteomics and metabolomics to reveal the molecular mechanism of Nutmeg-5 in ameliorating cardiac fibrosis following myocardial infarction. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154382. [PMID: 35963196 DOI: 10.1016/j.phymed.2022.154382] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Nutmeg-5, an ancient and classic formula in traditional Mongolian medicine comprising five kinds of traditional Chinese medicine, is widely used in the treatment of myocardial infarction (MI, called heart "Heyi" disease in Mongolian medicine). Cardiac fibrosis plays a critical role in the development and progression of heart failure after MI. However, the material basis and pharmacological mechanisms of the effect of Nutmeg-5 on cardiac fibrosis after MI remain unclear. OBJECTIVE The aim of this study was to first explore the potential material basis and molecular mechanism of action of Nutmeg-5 in improving cardiac fibrosis after MI via a multiomics approach. METHODS The constituents in Nutmeg-5 were identified by ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS). High-performance liquid chromatography (HPLC) and gas chromatography (GC)-based fingerprints of Nutmeg-5 were analysed, and characteristic peaks were identified by comparison to standard samples. A rat MI model was created by permanent ligation of the left anterior descending artery. The protective effect of Nutmeg-5 on cardiac fibrosis after MI was evaluated by tissue histology and measurement of the serum biomarkers of myocardial injury. Cardiac fibrosis levels were evaluated by Sirius red staining. Differentially expressed proteins in the myocardium and metabolites in the serum were explored by proteomic and untargeted metabolome analyses, respectively. Pearson correlation analysis was performed to explore the association between serum metabolites and myocardial proteins. RESULTS A total of 67 constituents were identified in Nutmeg-5 by UPLC-MS/MS. Sixteen components were identified in the fingerprint of Nutmeg-5 by comparison with a standard sample. Six lactones were isolated from Nutmeg-5 and quantified by HPLC and GC. MI was significantly alleviated in Nutmeg-5-treated rats compared to MI rats, as demonstrated by their decreased mortality, improved cardiac function, and attenuated cardiac fibrosis and myocardial injury. A total of 252 significant differential metabolites were identified in plasma between model and Nutmeg-5-treated rats by untargeted metabolome analysis. Among these, 36 critical metabolites were associated with Nutmeg-5 activity. Proteomic analysis identified 338 differentially expressed proteins in the rat myocardium between MI and Nutmeg-5-treated rats, including 204 upregulated and 134 downregulated proteins. Protein set enrichment analysis revealed that Nutmeg-5 treatment significantly inhibited the extracellular matrix (ECM)-receptor interaction pathway, which was activated in the myocardium of MI rats. A significant decrease in collagen and alpha smooth muscle actin expression levels was found in the myocardium of Nutmeg-5-treated rats compared to MI rats. These results illustrated that Nutmeg-5 had a significant protective effect on cardiac fibrosis after MI. A significant correlation was found between the ECM-receptor interaction pathway in the myocardium and critical metabolites in the serum. In addition, there were positive correlations between the levels of critical metabolites and the expression levels of transforming growth factor (TGF)-β1 and Smad2 in the rat myocardium. CONCLUSIONS Nutmeg-5 alleviated cardiac fibrosis after MI in rats by inhibiting the myocardial ECM-receptor interaction pathway and TGF-β1/Smad2 signalling, which was achieved by regulating plasma metabolites.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot 010110, PR China
| | - Xiaoling Zhu
- Inner Mongolian International Mongolian Hospital, University East Street, Hohhot 010065, PR China
| | - Xueni Zhang
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot 010110, PR China
| | - Xin Jia
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot 010110, PR China; Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, PR China
| | - Jing Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, PR China
| | - Xianjue Wang
- Clinical Medical Research Center, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, PR China
| | - Yunfeng Xiao
- Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, PR China
| | - Zhibin Xiao
- Department of Clinical Pharmacy, College of Pharmacy, Inner Mongolia Medical University, Hohhot 010110, PR China
| | - Tianlong Liu
- Department of Pharmacy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, PR China.
| | - Yu Dong
- Department of Natural Medicinal Chemistry, College of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot 010110, PR China; Engineering Technology Research Center of Pharmacodynamic Substance and Quality Control of Mongolian Medicine in Inner Mongolia, Hohhot 010110, PR China.
| |
Collapse
|
20
|
Ramalingam A, Kuppusamy M, Sambandam S, Medimagh M, Oyeneyin OE, Shanmugasundaram A, Issaoui N, Ojo ND. Synthesis, spectroscopic, topological, hirshfeld surface analysis, and anti-covid-19 molecular docking investigation of isopropyl 1-benzoyl-4-(benzoyloxy)-2,6-diphenyl-1,2,5,6-tetrahydropyridine-3-carboxylate. Heliyon 2022; 8:e10831. [PMID: 36211997 PMCID: PMC9526874 DOI: 10.1016/j.heliyon.2022.e10831] [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: 08/18/2022] [Revised: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 01/18/2023] Open
Abstract
Isopropyl 1-benzoyl-4-(benzoyloxy)-2,6-diphenyl-1,2,5,6-tetrahydropyridine-3-carboxylate (IDPC) was synthesized and characterized via spectroscopic (FT-IR and NMR) techniques. Hirshfeld surface and topological analyses were conducted to study structural and molecular properties. The energy gap (Eg), frontier orbital energies (EHOMO, ELUMO) and reactivity parameters (like chemical hardness and global hardness) were calculated using density functional theory with B3LYP/6-311++G (d,p) level of theory. Molecular docking of IDPC at the active sites of SARS-COVID receptors was investigated. IDPC molecule crystallized in the centrosymmetric triclinic ( P 1 ¯ ) space group. The topological and Hirshfeld surface analysis revealed that covalent, non-covalent and intermolecular H-bonding interactions, and electron delocalization exist in the molecular framework. Higher binding score (-6.966 kcal/mol) of IDPC at the active site of SARS-COVID main protease compared to other proteases suggests that IDPC has the potential of blocking polyprotein maturation. H-bonding and π-cationic and interactions of the phenyl ring and carbonyl oxygen of the ligand indicate the effective inhibiting potential of the compound against the virus.
Collapse
Affiliation(s)
- Arulraj Ramalingam
- Department of Electrical and Computer Engineering, National University of Singapore, 117583, Singapore
| | - Murugavel Kuppusamy
- PG & Research Department of Chemistry, Government Arts College, Chidambaram, Tamil Nadu, India
| | - Sivakumar Sambandam
- Research and Development Centre, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Mouna Medimagh
- University of Monastir, Faculty of Sciences, Laboratory of Quantum and Statistical Physics (LR18ES18), Monastir, 5079, Tunisia
| | - Oluwatoba Emmanuel Oyeneyin
- Theoretical and Computational Chemistry Unit, Department of Chemical Sciences Adekunle Ajasin University, Akungba Akoko, Ondo State, Nigeria
| | | | - Noureddine Issaoui
- University of Monastir, Faculty of Sciences, Laboratory of Quantum and Statistical Physics (LR18ES18), Monastir, 5079, Tunisia
| | | |
Collapse
|
21
|
Identification of Potential Allosteric Site Binders of Indoleamine 2,3-Dioxygenase 1 from Plants: A Virtual and Molecular Dynamics Investigation. Pharmaceuticals (Basel) 2022; 15:ph15091099. [PMID: 36145319 PMCID: PMC9502501 DOI: 10.3390/ph15091099] [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: 07/13/2022] [Revised: 08/27/2022] [Accepted: 08/31/2022] [Indexed: 12/02/2022] Open
Abstract
Ligand and structure-based computational screenings were carried out to identify flavonoids with potential anticancer activity. Kushenol E, a flavonoid with proven anticancer activity and, at the same time, an allosteric site binder of the enzyme indoleamine 2,3-dioxygenase-1 (IDO1), was used as the reference compound. Molecular docking and molecular dynamics simulations were performed for the screened flavonoids with known anticancer activity. The following two of these flavonoids were identified as potential inhibitors of IDO1: dichamanetin and isochamanetin. Molecular dynamics simulations were used to assess the conformational profile of IDO1-flavonoids complexes, as well as for calculating the bind-free energies.
Collapse
|
22
|
Elumalai E, Suresh Kumar M. Identification of neo-andrographolide compound targeting NS1 Lys14: an important residue in NS1 activity driving dengue pathogenesis. J Biomol Struct Dyn 2022:1-11. [DOI: 10.1080/07391102.2022.2068073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Elakkiya Elumalai
- Center for Bioinformatics, Pondicherry University, Pondicherry, India
| | - M. Suresh Kumar
- Center for Bioinformatics, Pondicherry University, Pondicherry, India
| |
Collapse
|
23
|
Investigation of glutathione as a natural antioxidant and multitarget inhibitor for Alzheimer’s disease: Insights from molecular simulations. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.117960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
24
|
COX Inhibitory and Cytotoxic Naphthoketal-Bearing Polyketides from Sparticola junci. Int J Mol Sci 2021; 22:ijms222212379. [PMID: 34830260 PMCID: PMC8619024 DOI: 10.3390/ijms222212379] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
Axenic fermentation on solid rice of the saprobic fungus Sparticola junci afforded two new highly oxidized naphthalenoid polyketide derivatives, sparticatechol A (1) and sparticolin H (2) along with sparticolin A (3). The structures of 1 and 2 were elucidated on the basis of their NMR and HR-ESIMS spectroscopic data. Assignment of absolute configurations was performed using electronic circular dichroism (ECD) experiments and Time-Dependent Density Functional Theory (TDDFT) calculations. Compounds 1-3 were evaluated for COX inhibitory, antiproliferative, cytotoxic and antimicrobial activities. Compounds 1 and 2 exhibited strong inhibitory activities against COX-1 and COX-2. Molecular docking analysis of 1 conferred favorable binding against COX-2. Sparticolin H (2) and A (3) showed a moderate antiproliferative effect against myelogenous leukemia K-562 cells and weak cytotoxicity against HeLa and mouse fibroblast cells.
Collapse
|
25
|
Santos J, Quimque MT, Liman RA, Agbay JC, Macabeo APG, Corpuz MJA, Wang YM, Lu TT, Lin CH, Villaflores OB. Computational and Experimental Assessments of Magnolol as a Neuroprotective Agent and Utilization of UiO-66(Zr) as Its Drug Delivery System. ACS OMEGA 2021; 6:24382-24396. [PMID: 34604621 PMCID: PMC8482410 DOI: 10.1021/acsomega.1c02555] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 05/26/2023]
Abstract
The phenolic natural product magnolol exhibits neuroprotective properties through β-amyloid toxicity in PC-12 cells and ameliorative effects against cognitive deficits in a TgCRND8 transgenic mice model. Its bioavailability and blood-brain barrier crossing ability have been significantly improved using the metal-organic framework (MOF) UiO-66(Zr) as a drug delivery system (DDS). To investigate the neuroprotective effects of the Zr-based DDS, magnolol and magnolol-loaded-UiO-66(Zr) (Mag@UiO-66(Zr)) were evaluated for inhibitory activity against β-secretase and AlCl3-induced neurotoxicity. Due to the moderate inhibition observed for magnolol in vitro, in silico binding studies were explored against β-secretase along with 11 enzymes known to affect Alzheimer's disease (AD). Favorable binding energies against CDK2, CKD5, MARK, and phosphodiesterase 3B (PDE3B) and dynamically stable complexes were noted through molecular docking and molecular dynamic simulation experiments, respectively. The magnolol-loaded DDS UiO-66(Zr) also showed enhanced neuroprotective activity against two pathological indices, namely, neutrophil infiltration and apoptotic neurons, in addition to damage reversal compared to magnolol. Thus, MOFs are promising drug delivery platforms for poorly bioavailable drugs.
Collapse
Affiliation(s)
- Joshua Santos
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Mark Tristan Quimque
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Laboratory
of Organic Reactivity, Discovery, and Synthesis (LORDS), Research
Center for Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department
of Chemistry, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, 9200 Iligan City, Philippines
| | - Rhenz Alfred Liman
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Jay Carl Agbay
- Department
of Chemistry, College of Science and Mathematics, Mindanao State University-Iligan Institute of Technology, Tibanga, 9200 Iligan City, Philippines
- Philippine
Science High School-Central Mindanao Campus, 9217 Balo-i, Lanao del Norte, Philippines
| | - Allan Patrick G. Macabeo
- Laboratory
of Organic Reactivity, Discovery, and Synthesis (LORDS), Research
Center for Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| | - Mary Jho-Anne Corpuz
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Pharmacology
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department
of Pharmacy, Faculty of Pharmacy, University
of Santo Tomas, España
Blvd., 1015 Manila, Philippines
| | - Yun-Ming Wang
- Department
of Biological Science and Technology, Institute of Molecular Medicine
and Bioengineering, Center for Intelligent Drug Systems and Smart
Bio-devices (IDS2B), National Chiao Tung
University, 30010 Hsinchu, Taiwan
| | - Tsai-Te Lu
- Institute
of Biomedical Engineering, National Tsing
Hua University, 30013 Hsinchu, Taiwan
| | - Chia-Her Lin
- College
of Science, Chung Yuan Christian University, Zhongli District, 320 Taoyuan City, Taiwan
| | - Oliver B. Villaflores
- The
Graduate School, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Phytochemistry
Laboratory, Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
- Department of Biochemistry, Faculty of Pharmacy, University of Santo Tomas, España Blvd., 1015 Manila, Philippines
| |
Collapse
|
26
|
Fernandez RA, Quimque MT, Notarte KI, Manzano JA, Pilapil DY, de Leon VN, San Jose JJ, Villalobos O, Muralidharan NH, Gromiha MM, Brogi S, Macabeo APG. Myxobacterial depsipeptide chondramides interrupt SARS-CoV-2 entry by targeting its broad, cell tropic spike protein. J Biomol Struct Dyn 2021; 40:12209-12220. [PMID: 34463219 PMCID: PMC8436362 DOI: 10.1080/07391102.2021.1969281] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022]
Abstract
The severity of the COVID-19 pandemic has necessitated the search for drugs against SARS-CoV-2. In this study, we explored via in silico approaches myxobacterial secondary metabolites against various receptor-binding regions of SARS-CoV-2 spike which are responsible in recognition and attachment to host cell receptors mechanisms, namely ACE2, GRP78, and NRP1. In general, cyclic depsipeptide chondramides conferred high affinities toward the spike RBD, showing strong binding to the known viral hot spots Arg403, Gln493 and Gln498 and better selectivity compared to most host cell receptors studied. Among them, chondramide C3 (1) exhibited a binding energy which remained relatively constant when docked against most of the spike variants. Chondramide C (2) on the other hand exhibited strong affinity against spike variants identified in the United Kingdom (N501Y), South Africa (N501Y, E484K, K417N) and Brazil (N501Y, E484K, K417T). Chondramide C6 (9) showed highest BE towards GRP78 RBD. Molecular dynamics simulations were also performed for chondramides 1 and 2 against SARS-CoV-2 spike RBD of the Wuhan wild-type and the South African variant, respectively, where resulting complexes demonstrated dynamic stability within a 120-ns simulation time. Protein-protein binding experiments using HADDOCK illustrated weaker binding affinity for complexed chondramide ligands in the RBD against the studied host cell receptors. The chondramide derivatives in general possessed favorable pharmacokinetic properties, highlighting their potential as prototypic anti-COVID-19 drugs limiting viral attachment and possibly minimizing viral infection.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Rey Arturo Fernandez
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Mark Tristan Quimque
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
- The Graduate School, University of Santo Tomas, Manila, Philippines
- Chemistry Department, College of Science and Mathematics, Mindanao State University – Iligan Institute of Technology, Tibanga, Iligan City, Philippines
| | - Kin Israel Notarte
- Faculty of Medicine and Surgery, University of Santo Tomas, Manila, Philippines
| | - Joe Anthony Manzano
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
| | - Delfin Yñigo Pilapil
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
| | - Von Novi de Leon
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
| | - John Jeric San Jose
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Omar Villalobos
- Department of Pharmacy, Faculty of Pharmacy, University of Santo Tomas, Manila, Philippines
| | - Nisha Harur Muralidharan
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai, Tamil Nadu, India
| | - M. Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology (IIT) Madras, Chennai, Tamil Nadu, India
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | - Allan Patrick G. Macabeo
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| |
Collapse
|
27
|
de Leon VNO, Manzano JAH, Pilapil DYH, Fernandez RAT, Ching JKAR, Quimque MTJ, Agbay JCM, Notarte KIR, Macabeo APG. Anti-HIV reverse transcriptase plant polyphenolic natural products with in silico inhibitory properties on seven non-structural proteins vital in SARS-CoV-2 pathogenesis. J Genet Eng Biotechnol 2021; 19:104. [PMID: 34272647 PMCID: PMC8284420 DOI: 10.1186/s43141-021-00206-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Accessing COVID-19 vaccines is a challenge despite successful clinical trials. This burdens the COVID-19 treatment gap, thereby requiring accelerated discovery of anti-SARS-CoV-2 agents. This study explored the potential of anti-HIV reverse transcriptase (RT) phytochemicals as inhibitors of SARS-CoV-2 non-structural proteins (nsps) by targeting in silico key sites in the structures of SARS-CoV-2 nsps. One hundred four anti-HIV phytochemicals were subjected to molecular docking with nsp3, 5, 10, 12, 13, 15, and 16. Top compounds in complex with the nsps were investigated further through molecular dynamics. The drug-likeness and ADME (absorption, distribution, metabolism, and excretion) properties of the top compounds were also predicted using SwissADME. Their toxicity was likewise determined using OSIRIS Property Explorer. RESULTS Among the top-scoring compounds, the polyphenolic functionalized natural products comprised of biflavones 1, 4, 11, 13, 14, 15; ellagitannin 9; and bisisoquinoline alkaloid 19 were multi-targeting and exhibited strongest binding affinities to at least two nsps (binding energy = - 7.7 to - 10.8 kcal/mol). The top ligands were stable in complex with their target nsps as determined by molecular dynamics. Several top-binding compounds were computationally druggable, showed good gastrointestinal absorptive property, and were also predicted to be non-toxic. CONCLUSIONS Twenty anti-HIV RT phytochemicals showed multi-targeting inhibitory potential against SARS-CoV-2 non-structural proteins 3, 5, 10, 12, 13, 15, and 16. Our results highlight the importance of polyhydroxylated aromatic substructures for effective attachment in the binding/catalytic sites of nsps involved in post-translational mechanism pathways. As such with the nsps playing vital roles in viral pathogenesis, our findings provide inspiration for the design and discovery of novel anti-COVID-19 drug prototypes.
Collapse
Affiliation(s)
- Von Novi O de Leon
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - Joe Anthony H Manzano
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - Delfin Yñigo H Pilapil
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- Department of Biological Sciences, College of Science, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - Rey Arturo T Fernandez
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - James Kyle Anthony R Ching
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- Department of Chemistry, College of Science, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - Mark Tristan J Quimque
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- The Graduate School, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
- Chemistry Department, College of Science and Mathematics, Mindanao State University - Iligan Institute of Technology, Tibanga, 9200, Iligan City, Philippines
| | - Jay Carl M Agbay
- Chemistry Department, College of Science and Mathematics, Mindanao State University - Iligan Institute of Technology, Tibanga, 9200, Iligan City, Philippines
- Philippine Science High School - Central Mindanao Campus, 9217 Balo-I, Lanao del Norte, Philippines
| | - Kin Israel R Notarte
- Faculty of Medicine and Surgery, University of Santo Tomas, España Blvd., 1015, Manila, Philippines
| | - Allan Patrick G Macabeo
- Laboratory for Organic Reactivity, Discovery and Synthesis (LORDS), Research Center for the Natural and Applied Sciences, University of Santo Tomas, España Blvd., 1015, Manila, Philippines.
| |
Collapse
|