1
|
Dong Q, Li X, Cheng K. Hsp90 and HIF-1α regulate mitophagy by promoting BNIP3 expression in renal ischemia-reperfusion injury. Transpl Immunol 2025:102177. [PMID: 39892761 DOI: 10.1016/j.trim.2025.102177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 01/09/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is a major cause of acute kidney injury (AKI). Studies have shown that mitochondrial damage is involved in the pathogenesis of AKI, and that inhibition of Hsp90 expression can improve IR-induced AKI. However, the mechanisms by which Hsp90 improves IR-induced AKI and whether it is involved in mitochondrial autophagy remain unclear. METHODS An IR-induced AKI mouse model was established, and the degree of renal injury was analyzed using hematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining. The expression of Hsp90, HIF-1α, BNIP3, and mitochondrial autophagy proteins was detected by western blotting in vivo and in vitro. HK2 cell viability, apoptosis, mitochondrial autophagy, reactive oxygen species (ROS), and inflammatory cytokines levels were detected using Cell Counting Kit 8 (CCK8) assays, Terminal·deoxynucleotidyl transferase-mediated dUTP nick end·labeling (TUNEL) labeling, immunofluorescence, and enzyme-linked immunosorbent (ELISA). RESULTS A murine IR-induced AKI model was successfully generated, and increased expression levels of Hsp90, HIF-1α, and inflammatory cytokines were observed, accompanied by a worsening of renal injury. After induction of IRI in HK2 cells, downregulation of Hsp90 or HIF-1α expression resulted in decreased downstream BNIP3 expression, an increase in HK2 cell viability, and a decrease in the level of mitochondrial autophagy. CONCLUSION Hsp90 upregulated the expression of HIF-1αand BNIP3, thereby enhancing mitochondrial autophagy in IR-induced AKI.
Collapse
Affiliation(s)
- Qi Dong
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Xia Li
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China
| | - Ke Cheng
- Transplantation Center, Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, China.
| |
Collapse
|
2
|
Zhang Y, Li R, Chen X, Li Y, Zhang Q, Yang L, Wang L, Sun Y, Mao F, Zhuo CJ. Clozapine Induces Agranulocytosis via Inflammatory and Hematopoietic Cytokine Induction of the JAK-STAT Signaling Pathway: Evidence From Network Pharmacology and Molecular Docking. CNS Neurosci Ther 2025; 31:e70206. [PMID: 39776289 PMCID: PMC11707432 DOI: 10.1111/cns.70206] [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: 09/12/2024] [Revised: 11/09/2024] [Accepted: 12/01/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Clozapine exhibits significant therapeutic efficacy in schizophrenia, especially treatment-resistant schizophrenia. However, clozapine can cause agranulocytosis, a fatal adverse effect, and the aim of this study is to explore this mechanism based on network pharmacology and molecular docking. METHOD Six and two databases were used to identify targets associated with clozapine and agranulocytosis, respectively. The bioinformatics online platform was used to identify overlaps between the drug and disease targets. The protein-protein interaction (PPI) network was characterized using Cystoscope 3.10.1 and STRING. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) were analyzed using the DAVID online platform. A drug-target-pathway-disease network was constructed utilizing Cystoscope 3.10.1. The Auto Dock Vina and PyMOL software were used to verify the molecular docking of clozapine and core targets. RESULTS The analysis revealed 188 overlapping targets. The PPI and KEGG enrichment pathway analyses demonstrated that clozapine induces agranulocytosis by modulating the hematopoietic cell lineage and JAK-STAT signaling pathways via interleukin-3 (IL3), IL6, IL2 receptor subunit alpha (IL2RA), and granulocyte colony-stimulating factor. Binding energies between clozapine and core targets were favorable (< -7.0 kcal/mol). CONCLUSION Clozapine-induced agranulocytosis may be linked to the JAK-STAT inflammatory signaling pathway through inflammatory and hematopoietic-related cytokines. Our findings enhance our comprehension of the potential mechanisms underlying clozapine-induced agranulocytosis.
Collapse
Affiliation(s)
- Ying Zhang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Department of Psychiatry and Psychology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Ranli Li
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Ximing Chen
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Yachen Li
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Qiuyu Zhang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Lei Yang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Lina Wang
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Yun Sun
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| | - Fuqiang Mao
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Department of Psychiatry and Psychology, School of Basic Medical SciencesTianjin Medical UniversityTianjinChina
| | - Chuan Jun Zhuo
- Computational Biology Center, Tianjin Anding HospitalNankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
- Laboratory of Psychiatric‐Neuroimaging‐Genetic and Co‐Morbidity (PGNP_Lab)Nankai University Affiliated Tianjin Anding Hospital, Tianjin Mental Health Center of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
3
|
Zhang Y, Li WW, Wang Y, Fan YW, Wang QY, Liu C, Jiang S, Shang EX, Duan JA. Investigation of the material basis and mechanism of Lizhong decoction in ameliorating ulcerative colitis based on spectrum-effect relationship and network pharmacology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117666. [PMID: 38159822 DOI: 10.1016/j.jep.2023.117666] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/11/2023] [Accepted: 12/25/2023] [Indexed: 01/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lizhong decoction (LZD), a classical herbal prescription recorded by Zhang Zhongjing in Treatise on Febrile and Miscellaneous Diseases, has been extensively used to treat ulcerative colitis (UC) in clinical practice for thousands of years. However, its material basis and underlying mechanism are not yet clear. AIM OF THE STUDY This study aims to explore the material basis and potential mechanism of LZD against UC based on the spectrum-effect relationship and network pharmacology. MATERIALS AND METHODS First, LZD was extracted by a systematic solvent extraction method into four parts. Ultra-high performance liquid chromatography tandem quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS/MS) technique was used to identify the compounds from different polar parts, and dextran sulfate sodium (DSS)-induced colitis model was used to evaluate the efficacy of each fraction. Then, the spectrum-effect analyses of compounds and efficacy indicators were established via grey relational analysis (GRA), bivariate correlation analysis (BCA) and partial least squares regression (PLSR). Finally, the potential mechanism of LZD for UC therapy was explored by network pharmacology, and the results were further verified by molecular docking and reverse transcription quantitative polymerase chain reaction (RT-qPCR). RESULTS 66 chemical components of LZD were identified by UPLC-Q-TOF-MS/MS technology. The pharmacodynamic results showed that extraction parts of LZD had different therapeutic effects on UC, among which ethyl acetate and n-butanol extracts had significant anti-colitis effects, which might be the main effective fractions of LZD. Furthermore, the spectrum-effect analyses indicated that 21 active ingredients such as liquiritin apioside, neolicuroside, formononetin, ginsenoside Rg1, 6-gingesulfonic acid, licoricesaponin A3, liquiritin, glycyrrhizic acid were the main material basis for LZD improving UC. Based on the above results, network pharmacology suggested that the amelioration of LZD on UC might be closely related to the PI3K-Akt signaling pathway. Additionally, molecular docking technology and RT-qPCR further verified that LZD could markedly inhibit the PI3K-Akt signaling pathway. CONCLUSION Overall, our study first identified the chemical compositions of LZD by using UPLC-Q-TOF-MS/MS. Furthermore, the material basis and potential mechanism of LZD in improving UC were comprehensively elucidated via spectrum-effect relationships, network pharmacology, molecular docking and experimental verification. The proposed strategy provided a systematic approach for exploring how herbal medicines worked. More importantly, it laid the solid foundation for further clinical application and rational development of LZD.
Collapse
Affiliation(s)
- Yun Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Wen-Wen Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Yu Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Yu-Wen Fan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Qu-Yi Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Chen Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Shu Jiang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| | - Er-Xin Shang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, PR China.
| |
Collapse
|
4
|
Mohamed ME, El-Shafae AM, Fikry E, Elbaramawi SS, Elbatreek MH, Tawfeek N. Casuarina glauca branchlets' extract as a potential treatment for ulcerative colitis: chemical composition, in silico and in vivo studies. Front Pharmacol 2023; 14:1322181. [PMID: 38196993 PMCID: PMC10774231 DOI: 10.3389/fphar.2023.1322181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/20/2023] [Indexed: 01/11/2024] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease that is often resistant to current treatment options, leading to a need for alternative therapies. Herbal products have shown promise in managing various conditions, including UC. However, the potential of Casuarina glauca branchlets ethanolic extract (CGBRE) in treating UC has not been explored. This study aimed to analyze the chemical composition of CGBRE and evaluate its efficacy in UC treatment through in silico and in vivo experiments. LC-ESI-MS/MS was used to identify 86 compounds in CGBRE, with 21 potential bioactive compounds determined through pharmacokinetic analysis. Network pharmacology analysis revealed 171 potential UC targets for the bioactive compounds, including EGFR, LRRK2, and HSP90 as top targets, which were found to bind to key CGBRE compounds through molecular docking. Molecular docking findings suggested that CGBRE may be effective in the prevention or treatment of ulcerative colitis mediated by these proteins, where key CGBRE compounds exhibited good binding affinities through formation of numerous interactions. In vivo studies in rats with acetic acid-induced UC demonstrated that oral administration of 300 mg/kg CGBRE for 6 days reduced UC symptoms and colonic expression of EGFR, LRRK2, and HSP90. These findings supported the therapeutic potential of CGBRE in UC and suggested the need for further preclinical and clinical investigation.
Collapse
Affiliation(s)
- Maged E. Mohamed
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Azza M. El-Shafae
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Eman Fikry
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Samar S. Elbaramawi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mahmoud H. Elbatreek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Nora Tawfeek
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| |
Collapse
|
5
|
Fikry E, Orfali R, El-Sayed SS, Perveen S, Ghafar S, El-Shafae AM, El-Domiaty MM, Tawfeek N. Potential Hepatoprotective Effects of Chamaecyparis lawsoniana against Methotrexate-Induced Liver Injury: Integrated Phytochemical Profiling, Target Network Analysis, and Experimental Validation. Antioxidants (Basel) 2023; 12:2118. [PMID: 38136237 PMCID: PMC10740566 DOI: 10.3390/antiox12122118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Methotrexate (MTX) therapy encounters significant limitations due to the significant concern of drug-induced liver injury (DILI), which poses a significant challenge to its usage. To mitigate the deleterious effects of MTX on hepatic function, researchers have explored plant sources to discover potential hepatoprotective agents. This study investigated the hepatoprotective effects of the ethanolic extract derived from the aerial parts of Chamaecyparis lawsoniana (CLAE) against DILI, specifically focusing on MTX-induced hepatotoxicity. UPLC-ESI-MS/MS was used to identify 61 compounds in CLAE, with 31 potential bioactive compounds determined through pharmacokinetic analysis. Network pharmacology analysis revealed 195 potential DILI targets for the bioactive compounds, including TP53, IL6, TNF, HSP90AA1, EGFR, IL1B, BCL2, and CASP3 as top targets. In vivo experiments conducted on rats with acute MTX-hepatotoxicity revealed that administering CLAE orally at 200 and 400 mg/kg/day for ten days dose-dependently improved liver function, attenuated hepatic oxidative stress, inflammation, and apoptosis, and reversed the disarrayed hepatic histological features induced by MTX. In general, the findings of the present study provide evidence in favor of the hepatoprotective capabilities of CLAE in DILI, thereby justifying the need for additional preclinical and clinical investigations.
Collapse
Affiliation(s)
- Eman Fikry
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Raha Orfali
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Shaimaa S. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt;
| | - Shagufta Perveen
- Department of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA;
| | - Safina Ghafar
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Azza M. El-Shafae
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Maher M. El-Domiaty
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| | - Nora Tawfeek
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt; (E.F.); (A.M.E.-S.); (N.T.)
| |
Collapse
|
6
|
Yang S, Wu H, Li Y, Li L, Xiang J, Kang L, Yang G, Liang Z. Inhibition of PFKP in renal tubular epithelial cell restrains TGF-β induced glycolysis and renal fibrosis. Cell Death Dis 2023; 14:816. [PMID: 38086793 PMCID: PMC10716164 DOI: 10.1038/s41419-023-06347-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023]
Abstract
Metabolic reprogramming to glycolysis is closely associated with the development of chronic kidney disease (CKD). Although it has been reported that phosphofructokinase 1 (PFK) is a rate-limiting enzyme in glycolysis, the role of the platelet isoform of PFK (PFKP) in kidney fibrosis initiation and progression is as yet poorly understood. Here, we investigated whether PFKP could mediate the progression of kidney interstitial fibrosis by regulating glycolysis in proximal tubular epithelial cells (PTECs). We induced PFKP overexpression or knockdown in renal tubules via an adeno-associated virus (AAV) vector in the kidneys of mice following unilateral ureteral occlusion. Our results show that the dilated tubules, the area of interstitial fibrosis, and renal glycolysis were promoted by proximal tubule-specific overexpression of PFKP, and repressed by knockdown of PFKP. Furthermore, knockdown of PFKP expression restrained, while PFKP overexpression promoted TGF-β1-induced glycolysis in the human PTECs line. Mechanistically, Chip-qPCR revealed that TGF-β1 recruited the small mothers against decapentaplegic (SMAD) family member 3-SP1 complex to the PFKP promoter to enhance its expression. Treatment of mice with isorhamnetin notably ameliorated PTEC-elevated glycolysis and kidney fibrosis. Hence, our results suggest that PFKP mediates the progression of kidney interstitial fibrosis by regulating glycolysis in PTECs.
Collapse
Affiliation(s)
- Shu Yang
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
| | - Han Wu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- Department of Endocrinology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Yanchun Li
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Lixin Li
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
| | - Lin Kang
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China
- The Biobank of National Innovation Center for Advanced Medical Devices, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Guangyan Yang
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
| | - Zhen Liang
- Department of Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, Guangdong, China.
| |
Collapse
|
7
|
Elbatreek MH, Fathi AM, Mahdi I, Abdelfattah MAO, Mahmoud MF, Sobeh M. Thymus satureioides Coss. combats oral ulcer via inhibition of inflammation, proteolysis, and apoptosis. Inflammopharmacology 2023; 31:2557-2570. [PMID: 37477794 DOI: 10.1007/s10787-023-01285-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023]
Abstract
Oral ulcer is a frequent condition that commonly affects the tongue and in which 75% of the patients experience pain, and 25% report taste changes. The available therapies are not sufficiently effective for rapid and complete healing of tongue ulcers. We previously annotated the metabolites of Thymus satureioides (TS) aerial parts and reported their antioxidant, dermacosmeceutical and hepatoprotective properties. In this study, we performed in silico analysis, by applying network pharmacology and molecular docking, followed by experimental validation of the effect of local application of T. satureioides (TS) gel at two different concentrations on the healing of acetic-acid-induced tongue ulcer in rats. Salvianolic acid A, phloretic acid caffeate, rosmarinic acid, apigenin, and luteolin were the top bioactive ingredients of TS extract. Network pharmacology showed that the most relevant targets of these active constituents were TLR4, COX-2, MMP-9, TNF-α, and Caspase-3. Molecular docking showed that rosmarinic acid and salvianolic acid had a relatively strong binding affinity, compared to the other compounds, toward all the target proteins. Experimental validation in tongue ulcer model in rats and immunohistochemistry experiments showed that application of a gel containing TS extract (5 and 10%) was effective in healing the tongue ulcer via downregulation of COX-2, TNF-α, MMP-9, and Caspase-3. This study suggests that T. satureioides extract could act as a topical treatment for tongue ulcers by combating inflammation, apoptosis, and proteolysis. The possible treatment potential of some constituents including rosmarinic acid and salvianolic acid in oral ulcerations awaits further investigations to confirm their potency.
Collapse
Affiliation(s)
- Mahmoud H Elbatreek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Ayman M Fathi
- Department of Pharmaceutics, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt
| | - Ismail Mahdi
- AgroBioSciences Department, College for Sustainable Agriculture and Environmental Science, Mohammed VI Polytechnic University, 43150, Ben Guerir, Morocco
| | - Mohamed A O Abdelfattah
- College of Engineering and Technology, American University of the Middle East, Egaila, 54200, Kuwait
| | - Mona F Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
| | - Mansour Sobeh
- AgroBioSciences Department, College for Sustainable Agriculture and Environmental Science, Mohammed VI Polytechnic University, 43150, Ben Guerir, Morocco.
| |
Collapse
|
8
|
Fikry E, Orfali R, Elbaramawi SS, Perveen S, El-Shafae AM, El-Domiaty MM, Tawfeek N. Chamaecyparis lawsoniana Leaf Essential Oil as a Potential Anticancer Agent: Experimental and Computational Studies. PLANTS (BASEL, SWITZERLAND) 2023; 12:2475. [PMID: 37447036 DOI: 10.3390/plants12132475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023]
Abstract
Cancer remains one of the leading causes of death worldwide, affected by several factors including oxidative stress; and although conventional synthetic medicines have been used to treat cancer, they often result in various side effects. Consequently, there is a growing need for newer, safer and more effective alternatives, such as natural plant products. Essential oils (EOs) are one such alternative, offering a wide range of bioactivities, including antibacterial, antiviral, antioxidant, and anticancer properties. Accordingly, the objective of the present study was to investigate the chemical composition, as well as the antioxidant and anticancer properties of the leaf essential oil of Chamaecyparis lawsoniana (CLLEO) belonging to the Cupressaceae family. Totally, 59 constituents were identified by gas chromatography-mass spectrometry (GC-MS) analysis. cis-Abienol, trans-ferruginol, α-cadinol, δ-muurolene and α-pinene were the major components. The in vitro cytotoxicity study against human breast (MCF-7), colon (HCT-116), lung (A-549), hepatocellular (HepG-2) carcinoma cells using MTT assay indicated a promising cytotoxic activity against all the tested cancer cells, particularly HepG-2, with significant selectivity indices. CLLEO exhibited weak antioxidant activity according to the DPPH, ABTS and FRAP assays. In silico docking of these constituents against the epidermal growth factor receptor (EGFR), the myeloid cell leukemia-1 (Mcl-1) and caspase-8 using Molecular Operating Environment (MOE) software demonstrated good binding affinities of the components with the active site of these targets. These findings suggested using CLLEO, or its individual components, as a potentially viable therapeutic option for managing cancerous conditions.
Collapse
Affiliation(s)
- Eman Fikry
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Raha Orfali
- Department of Pharmacognosy, Collage of Pharmacy, King Saud University, Ryiadh 11451, Saudi Arabia
| | - Samar S Elbaramawi
- Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Shagufta Perveen
- Department of Chemistry, School of Computer, Mathematical and Natural Sciences, Morgan State University, Baltimore, MD 21251, USA
| | - Azza M El-Shafae
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Maher M El-Domiaty
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Nora Tawfeek
- Department of Pharmacognosy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
9
|
Younis NS. Myrrh Essential Oil Mitigates Renal Ischemia/Reperfusion-Induced Injury. Curr Issues Mol Biol 2023; 45:1183-1196. [PMID: 36826023 PMCID: PMC9955815 DOI: 10.3390/cimb45020078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Ischemia/reperfusion (I/R)-induced renal injury is a common reason for kidney injury in clinical settings; therefore, continuous investigation of novel nephroprotective agents is crucial. Myrrh, the oleoresin exudates generated by the genus Commiphora, display numerous pharmacological actions. This study tried to assess the preventive effects of myrrh essential oil against I/R-induced renal damage. METHODS Rats were randomized into five groups. In the sham group, the animals were subjected to bilateral renal artery separation with no occlusion. In the sham + myrrh group; the rats were administered myrrh essential oil and then treated similarly to the sham group. Renal I/R group: the animals were challenged with renal I/R. In the myrrh + renal I/R groups, rats were administered 50 or 100 mg/kg of myrrh essential oil orally for three weeks before being confronted with I/R. RESULTS Serum levels of renal function tests and renal injury biomarkers, including NGAL, KIM-1, and CysC, were amplified in the renal I/R group. Animals that experienced renal I/R exhibited elevated lipid peroxidation (MDA); declined SOD, CAT, and GPx activity; declined GSH content; augmented TLR4/NFκB gene expression; and subsequent enhancement of inflammatory mediators (TNF-α, IFN-γ, IL-1β, and IL-6). Myrrh reduced renal function tests and injury biomarkers and amended renal histological alterations. Pretreatment with myrrh reduced MDA, elevated the antioxidant enzymes' activities and GSH content, and reduced the TLR4 and NFκB gene expression, leading to subsequent inflammation and apoptosis alleviation. CONCLUSIONS The outcomes of the present investigation established the protective effect of myrrh essential oil against renal I/R via pointing out the antioxidant, anti-inflammatory, and anti-apoptotic effects of myrrh.
Collapse
Affiliation(s)
- Nancy S. Younis
- College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Zagazig University Hospitals, University of Zagazig, Zagazig 44519, Egypt
| |
Collapse
|
10
|
Ji W, Gu L, Zou X, Li Z, Xu X, Wu J, Zhang S, Deng H. Discovery, Validation, and Target Prediction of Antibacterial and Antidiabetic Components of Archidendron clypearia Based on a Combination of Multiple Analytical Methods. Molecules 2023; 28:molecules28031329. [PMID: 36770996 PMCID: PMC9919075 DOI: 10.3390/molecules28031329] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Archidendron clypearia (A. clypearia), a Fabaceae family member, is widely used as an anti-inflammatory herbal medicine; however, its antibacterial and antidiabetic properties have not been extensively investigated. This study aimed to systematically analyze the antibacterial and antidiabetic components of A. clypearia by utilizing a combination of analytical methods. First, ten different polarity extracts were analyzed through ultra-performance liquid chromatography (UPLC), and their antibacterial and antidiabetic activities were evaluated. Then the spectrum-effect relationship between the biological activity and UPLC chromatograms was analyzed by partial least squares regression and gray relational analysis, followed by corresponding validation using isolated components. Finally, network pharmacology and molecular docking were implemented to predict the main antibacterial target components of A. clypearia and the enzyme inhibition active sites of α-amylase and α-glucosidase. P15, P16, and P20 were found to be the antibacterial and antidiabetic active components. The inhibitory effect of 7-O-galloyltricetiflavan (P15) on six bacterial species may be mediated through the lipid and atherosclerosis pathway, prostate cancer, adherens junctions, and targets such as SRC, MAPK1, and AKT1. The molecular docking results revealed that 7-O-galloyltricetiflavan and 7,4'-di-O-galloyltricetiflavan (P16/P20) can bind to α-amylase and α-glucosidase pockets with binding energies lower than -6 kcal/mol. Our study provides guidance for the development of antibacterial and antidiabetic products based on A. clypearia and can be used as a reference for the evaluation of bioactivity of other herbs.
Collapse
Affiliation(s)
- Wenduo Ji
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lixia Gu
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xuezhe Zou
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhichao Li
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaohong Xu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Jialin Wu
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shu Zhang
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Department of Pharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hong Deng
- The Center for Drug Research and Development, Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, Department of Pharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Correspondence:
| |
Collapse
|