1
|
Li J, Lv L, Hu M, Liu Z, Zhou S. Inhibition of N6-methyladenosine methylation of ASC by berberine ameliorates pyroptosis of renal tubular epithelial cells in acute kidney injury. Cell Signal 2025; 131:111732. [PMID: 40074191 DOI: 10.1016/j.cellsig.2025.111732] [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: 12/15/2024] [Revised: 03/04/2025] [Accepted: 03/09/2025] [Indexed: 03/14/2025]
Abstract
Acute kidney injury (AKI) lacks a definitive therapeutic approach beyond supportive care. One significant pathological mechanism involves the regulated death of tubular epithelial cells; however, the regulatory mechanisms underlying this cell death pathway require further investigation. The N6-methyladenosine (m6A) modification, recognized as the most prevalent modification in eukaryotes, plays a critical role in the regulatory processes associated with AKI. Here, this study investigates the association between methyltransferase-like 3 (METTL3) and pyroptosis in mice with folic acid (FA)-induced AKI. Both in vitro and in vivo experiments have confirmed that METTL3 plays a role in AKI progression, correlating with renal epithelial cell pyroptosis and inflammation. Moreover, RNA immunoprecipitation quantitative PCR (RIP-qPCR) analysis demonstrated that METTL3-mediated m6A methylation occurred in the mRNA of Apoptosis-associated speck-like protein containing a CARD (ASC) in H2O2-induced renal tubular epithelial (TCMK-1) cells. Notably, METTL3 knockdown resulted in reduced ASC protein expression, decreased release of inflammatory factors, and reduced pyroptosis. In addition, we verified the inhibitory effect of berberine hydrochloride, a monomer used in traditional Chinese medicine, on METTL3 expression. We also demonstrated that berberine ameliorated FA-induced AKI and H2O2-induced pyroptosis in TCMK-1 cells by inhibiting METTL3 and modulating the ASC/caspase-1/Gasdermin D axis. These findings provide insights into targeted therapies and drug development for AKI.
Collapse
Affiliation(s)
- Jiacheng Li
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Linxiao Lv
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Mingyang Hu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China
| | - Zhangsuo Liu
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China.
| | - Sijie Zhou
- Traditional Chinese Medicine Integrated Department of Nephrology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China; Research Institute of Nephrology, Zhengzhou University, Zhengzhou 450052, PR China; Henan Province Research Center for Kidney Disease, Zhengzhou 450052, PR China; Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou 450052, PR China; Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University Zhengzhou, Henan, China; Innovation Center of Basic Research for Metabolic-Associated Fatty Liver Disease, Ministry of Education of, China.
| |
Collapse
|
2
|
Lin T, Li Z, Yuan J, Ren T, Pang W, Xu S. Design, synthesis and evaluation of diphenyl ether-based kaiso inhibitors with enhanced potency. Bioorg Med Chem Lett 2025; 121:130158. [PMID: 40049243 DOI: 10.1016/j.bmcl.2025.130158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/10/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025]
Abstract
Kaiso, a potential target for the treatment of lung cancer. Our research focuses on Kaiso inhibitros. Through virtual screening and molecular dynamic simulations, we discovered a promising Kaiso inhibitor called compound 5 (ZINC20577650). By modifying the structure of compound 5, a series of novel Kaiso inhibitors that contain a diphenyl ether ring were synthesized. Among them, compound 20 exhibited the strongest inhibitory activity against A549 cells (IC50 = 0.34 μM). Notably, its inhibitory activity surpassed that of the positive control MIRA-1 (IC50 = 654.065 μM). Molecular docking and dynamic studies revealed that the binding of the compound's amino and ester moieties to the active site of kaiso protein, as well as the extension of the benzene ring towards the Asn561 position in the cavity, contributed significantly to its potency. These findings provide valuable insights for the development of new Kaiso inhibitors.
Collapse
Affiliation(s)
- Taofeng Lin
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Zhongqi Li
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China
| | - Juanchan Yuan
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Tinfeng Ren
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China
| | - Wan Pang
- College of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, China.
| | - Songhui Xu
- Department of Urology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University. Nanchang, China; Key Laboratory of Urinary System Diseases of Jiangxi Province, Nanchang, China.
| |
Collapse
|
3
|
Wang MY, Zhao HL, Fan YN, Zhang Y, Wang YX, Li B, Yang H, Li P. Network Pharmacology-Guided Identification of Bioactive Equivalent Combinatorial Components in Lanqin Oral Liquid for Acute Pharyngitis Treatment. JOURNAL OF ETHNOPHARMACOLOGY 2025:120038. [PMID: 40419204 DOI: 10.1016/j.jep.2025.120038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/23/2025] [Accepted: 05/24/2025] [Indexed: 05/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Lanqin Oral Liquid (LQL) is a pure herbal preparation with a good therapeutic effect on acute pharyngitis in clinic. However, its active ingredients and action mechanisms need further research. AIMS OF THE STUDY This work employed network pharmacology to uncover the bioactive equivalent combinatorial components (BECCs) of LQL and investigate their mechanisms in treating acute pharyngitis. MATERIALS AND METHODS In this study, a component-disease-target (C-D-T) network related to LQL and pharyngitis was constructed through network pharmacology and candidate BECCs were found based on the degree values of each node in the C-D-T network. The anti-acute pharyngitis effects between BECCs and LQL were evaluated by animal and cell experiments, and the vital signaling pathways predicted by network pharmacology were verified. Additionally, the synergistic effect of berberine (BBR) and geniposide (GE), the core active ingredients of BECCs, was further explored by the combination index (CI) method. RESULTS Network pharmacology indicated that 25 potential active ingredients in LQL and 35 targets were involved in the treatment of acute pharyngitis, covering 20 signaling pathways. Based on network pharmacology degree values, 13 compounds were identified as BECCs. In vitro and in vivo studies revealed that BECCs exhibit comparable therapeutic efficacy to LQL in treating acute pharyngitis. BECCs could treat acute pharyngitis by reducing the release of NO, secretion of inflammatory factors (IL-6, IL-1β, and TNF-α), and expression of related proteins (TLR4 and p-p65). Notably, BBR and GE emerged as BECCs' most effective components, demonstrating significant anti-inflammatory synergy. CONCLUSION BECCs of LQL demonstrate comparable efficacy against acute pharyngitis to the original formula, which may exert anti-inflammatory effects by regulating the TLR4/NF-κB signaling pathway. The diverse components in BECCs potentially exhibit synergistic pharmacological effects. These findings provide a solid scientific foundation for the clinical application of LQL.
Collapse
Affiliation(s)
- Meng-Yuan Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Heng-Li Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yu-Nuo Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Ye Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China
| | - Yong-Xiang Wang
- Nanjing Hailing Traditional Chinese Medicine Pharmaceutical Technology Research Co., Ltd., Nanjing 210049, China
| | - Bin Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
4
|
Emami L, Baziyar L, Al-Dies AAM, Sadeghian S, Mirjalili BBF, Faghih Z, Khorasani S, Zamani L, Khabnadideh S. Fe 3O 4@SiO 2-SnCl 4-promoted synthesis, cytotoxic evaluation, molecular docking, and MD simulation of some indenopyrido[2,3-d]pyrimidine derivatives. BMC Chem 2025; 19:131. [PMID: 40380230 PMCID: PMC12085073 DOI: 10.1186/s13065-025-01489-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/23/2025] [Indexed: 05/19/2025] Open
Abstract
In this study, an efficient and environmentally friendly method for the one-pot synthesis of indenopyrido[2,3-d]pyrimidine derivatives was developed using Fe3O4@SiO2-SnCl4 nanoparticles as a catalyst. Indenopyrido[2,3-d]pyrimidines (4a-4j) were synthesized via three-component couplings of 6-amino-2-(methylthio)pyrimidin-4(3H)-one, 1,3-indanedione, and aldehydes in water as the solvent. In this reaction, Fe3O4@SiO2-SnCl4 demonstrated a highly catalytic nature, an easy handling procedure, short reaction times, recyclability exploitation, and excellent yields. The cytotoxic activities of the synthesized indenopyrido[2,3-d] pyrimidines analogues were evaluated against three cancer cell lines; MCF-7 (breast carcinoma), A549 (lung non-small cell carcinoma), and SKOV3 (ovarian carcinoma) using MTT assay. Additionally, molecular docking studies and molecular dynamics (MD) simulation of the investigated compounds was performed to verify their binding modes toward EGFR kinase receptor as the possible targets. This analysis aimed to predict the antitumor mechanisms of the synthesized compounds. The binding free energy values of the compounds showed a satisfactory correlation with their cytotoxic activities.
Collapse
Affiliation(s)
- Leila Emami
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ladan Baziyar
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, P.O. Box: 71345-1798, Shiraz, Iran
| | - Al-Anood Mohammad Al-Dies
- Department of Chemistry, Al Qunfudah University College, UMM Al-Qura University, Mecca, Saudi Arabia
| | - Sara Sadeghian
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, P.O. Box: 71345-1798, Shiraz, Iran
| | | | - Zeinab Faghih
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sajad Khorasani
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Leila Zamani
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soghra Khabnadideh
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Shiraz University of Medical Sciences, P.O. Box: 71345-1798, Shiraz, Iran.
| |
Collapse
|
5
|
Jin C, Zhang Y, Chen L, Chen B, Chen C, Zhang H, Guo J, Chen W, Shi Y, Wen C. Lung Epithelial Cell Membrane-Camouflaged ROS-Activatable Berberine Nanoparticles for Targeted Treatment in Acute Lung Injury. Int J Nanomedicine 2025; 20:6163-6183. [PMID: 40391301 PMCID: PMC12087599 DOI: 10.2147/ijn.s514611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Introduction Acute lung injury (ALI) seriously threatens human health and is induced by multiple factors. When ALI occurs, lung lesions affect gas exchange and may trigger respiratory failure. Current clinical treatments are limited, and traditional drug delivery has drawbacks. Berberine, a natural drug with anti-inflammatory effects, has difficulty in effectively exerting its efficacy. Methods The study designed a nano-micelle. Hydrophobic berberine was encapsulated with diselenide bonds as the linker. Then, lung epithelial cell membranes were extracted to encapsulate and disguise the nano-micelle. These nanoparticles were injected intravenously. Thanks to the cell membrane's specificity, they could bind to lung tissue, achieving targeted lung delivery. In the inflamed area of acute lung injury, the significantly increased reactive oxygen species level was used to break the diselenide bonds, enabling precise berberine release at the lung injury site. Results The nano-drug (MM-NPs) was successfully prepared, with the encapsulation efficiency of berberine in the micelles reaching 68.2%. In a ROS environment, the nano-micelles could quickly release over 80% of berberine. In inflammatory MLE-12 cells, MM-NPs responded well to ROS, and cellular inflammatory factor levels were significantly improved after treatment. In a lipopolysaccharide (LPS)-induced pneumonia mouse model, MM-NPs achieved lung targeting. Further studies showed that MM-NPs administration significantly alleviated LPS-induced lung injury in mice. Additionally, evaluation indicated MM-NPs had good in-vivo safety with no obvious adverse reactions. Conclusion This study successfully developed a novel delivery system, MM-NPs, overcoming berberine's low bioavailability problem in treating acute lung injury. The system has excellent physicochemical properties, biocompatibility, and metabolic safety. In vitro and animal experiments verified it can significantly enhance the therapeutic effect, offering new ideas and hopes for acute lung injury treatment. In the future, clinical trials can be advanced, and new lung targeting strategies explored for more therapeutic breakthroughs.
Collapse
Affiliation(s)
- Chengkang Jin
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Yingjie Zhang
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, People’s Republic of China
| | - Lin Chen
- School of Life sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Biological Macromolecules Development Department, Hangzhou Zhongmeihuadong Pharmaceutical Co., Ltd, Hangzhou, 310011, People’s Republic of China
| | - Bingqing Chen
- Yue Yang Hospital of Traditional Chinese & Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, People’s Republic of China
| | - Changjiang Chen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Hairui Zhang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Junping Guo
- Rainbowfish Rehabilitation and Nursing School, Hangzhou Vocational & Technical College, Hangzhou, 310018, People’s Republic of China
| | - Wei Chen
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, People’s Republic of China
| | - Chengping Wen
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| |
Collapse
|
6
|
Song D, Khan A, Dong MH, Lei CW, Feng TT, Zhou Y, Wei X. Anti-Inflammatory Mechanism Prediction of Sinomenine Based on Network Pharmacology and Its Biological Activity Verification. BIOLOGY 2025; 14:543. [PMID: 40427732 PMCID: PMC12109165 DOI: 10.3390/biology14050543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025]
Abstract
Inflammation is a widespread physiological response that occurs when the body is stimulated by pathogens or endogenous signals [...].
Collapse
Affiliation(s)
- Da Song
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
- Guizhou Key Laboratory of Modern Traditional Chinese Medicine Creation, Guiyang 550025, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan;
| | - Ming-Hong Dong
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
| | - Chuan-Wen Lei
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
- Guizhou Key Laboratory of Modern Traditional Chinese Medicine Creation, Guiyang 550025, China
| | - Ting-Ting Feng
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
- Guizhou Key Laboratory of Modern Traditional Chinese Medicine Creation, Guiyang 550025, China
| | - Ying Zhou
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
- Guizhou Key Laboratory of Modern Traditional Chinese Medicine Creation, Guiyang 550025, China
| | - Xin Wei
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China; (D.S.); (M.-H.D.); (C.-W.L.); (T.-T.F.)
- Guizhou Key Laboratory of Modern Traditional Chinese Medicine Creation, Guiyang 550025, China
| |
Collapse
|
7
|
Gujral P, Orozco-Alonso E, Saliba J, Yan X, Blank V. The NFE2L2 (NRF2) transcription factor controls genes involved in the oxidative stress response and inflammation in myometrial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119985. [PMID: 40360022 DOI: 10.1016/j.bbamcr.2025.119985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/15/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
The myometrium is the smooth muscle layer of the uterus, which mediates uterine contractions during labor. We treated PHM1-31 myometrial cells with the proinflammatory cytokine interleukin-1 beta (IL1B) and measured a significant increase in reactive oxygen species (ROS). We found that IL1B induces NFE2L2 (NRF2) transcription factor levels. We further showed that siRNA mediated knockdown of NFE2L2 results in a significant increase in ROS. Downregulation of NFE2L2 leads to a decrease of heme oxygenase-1 (HMOX1) and aldo-keto reductase family 1 member B (AKR1B) at the transcript and protein level both in the absence and presence of IL1B. NFE2L2 knockdown also results in reduced ferritin heavy chain 1 (FTH1) mRNA expression, but only upon IL1B exposure, while FTH1 protein is downregulated both under basal and IL1B treatment conditions. We confirmed that NFE2L2 directly binds to the regulatory regions of these targets. Previous reports have linked HMOX1 and FTH1 to the oxidative stress response, and AKR1B1 to prostaglandin synthesis. Our data demonstrate that NFE2L2 functions as a key regulator of inflammatory and oxidative stress signaling through the regulation of HMOX1, FTH1, and AKR1B1 expression in myometrial cells. While HMOX1 and FTH1 have established roles in oxidative stress responses, our findings identify AKR1B1 as a novel target of NFE2L2 in myometrial cells, suggesting a role for the transcription factor in prostaglandin metabolism. Thus, NFE2L2 links inflammation and the oxidative stress response to critical pathways that control myometrial cell function and parturition, highlighting their potential as therapeutic targets for treating infection-induced preterm labor.
Collapse
Affiliation(s)
- Palak Gujral
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada
| | - Eduardo Orozco-Alonso
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada
| | - James Saliba
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Xingyue Yan
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Volker Blank
- Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada; Department of Medicine, McGill University, Montreal H4A 3J1, Canada; Department of Physiology, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
8
|
da Silva Joaquim L, da Rosa LR, Strickert Y, Machado RS, Lanzzarin E, Bernardes G, de Souza Ramos S, de Novais LR, Steiner B, Farias B, Mathias K, Martins HM, Lins EMF, Chaves JS, Camilo D, da Silva LE, de Oliveira MP, da Silva MR, Barcelos PMP, Santos FP, Bobinski F, Rezin GT, Yonamine M, Inserra A, Petronilho F, de Bitencourt RM. Ayahuasca reverses ischemic stroke-induced neuroinflammation and oxidative stress. Behav Brain Res 2025; 485:115521. [PMID: 40043852 DOI: 10.1016/j.bbr.2025.115521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/21/2025] [Accepted: 03/01/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Ischemic stroke is a leading cause of death and disability worldwide. Survivors face disability and psychiatric sequelae resulting from ischemia-induced cell death and associated neuroinflammation, and oxidative stress. Herbal medicines have been shown to elicit neuroprotective effects following stroke due to their anti-inflammatory and antioxidant effects. Preliminary evidence suggests that Ayahuasca (AYA), a decoction made from the vine Banisteriopsis caapi containing β-carbolines and the shrub Psychotria viridis containing N, N-Dimethyltryptamine, might attenuate ischemia-induced neuroinflammation and oxidative stress. Therefore, in this study we investigated the putative protective effects of AYA in the middle cerebral artery occlusion (MCAO) model of ischemic stroke. METHODS Wistar rats were subjected to the MCAO stroke model or sham surgery on day 0. After 24-h, rats were treated for three days with AYA (2 and 4 mL/kg, gavage) or saline. Neurological score was assessed for 72-h post-stroke. Rats were tested in the elevated plus maze, open field, and novel object recognition tests to assess locomotion, anxiety-like behavior, and recognition memory. Interleukin (IL)-6, IL-10 myeloperoxidase (MPO) activity, and the nitrite/nitrate (N/N) concentrations were determined in the prefrontal cortex (PFC), hippocampus (HPC), hypothalamus (HYP) and cortex. as markers of inflammation. Oxidative stress was quantified in the same brain areas as measured by the levels of thiobarbituric acid reactive species (TBARS), protein carbonylation, and superoxide dismutase (SOD), and catalase (CAT) activity. Mitochondrial metabolism was assessed quantifying the activity of complex 1(CI), CII, citrate synthase (CS), succinate dehydrogenase (SDH), and creatine kinase (CK). RESULTS No differences were observed regarding neurological deficits, locomotion, anxiety-like behavior, and recognition memory. However, AYA reversed the stroke-induced increase in IL-6 levels in the PFC and the HPC, IL-10 in the PFC, HPC, and HYP, MPO activity in the PFC, and N/N concentration and CAT activity in the HYP. Moreover, AYA decreased TBARS levels in the PFC and HPC and brain-derived neurotrophic factor (BDNF) in the PFC, and increased SOD activity in the cortex. Lastly, AYA increased CI activity in the HPC and cortex and decreased SDH and CK activity in the HPC. CONCLUSION AYA administration following ischemic stroke modulates oxidative stress and neuroinflammation in the PFC, HPC, and HYP. Despite no significant improvements in neurological or behavioral scores, these molecular changes suggest a neuroprotective role of AYA. Future studies should explore the timing of AYA treatment and putative long-term effects on functional recovery, as well as its potential in other brain regions critical for cognitive and motor functions.
Collapse
Affiliation(s)
- Larissa da Silva Joaquim
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil; Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Lara Rodrigues da Rosa
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Yasmin Strickert
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Richard Simon Machado
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil; Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Everton Lanzzarin
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Gabriela Bernardes
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Suelen de Souza Ramos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Linério Ribeiro de Novais
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Beatriz Steiner
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Brenno Farias
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Khiany Mathias
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Helena Mafra Martins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Elisa Mitkus Flores Lins
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Jéssica Schaefer Chaves
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Douglas Camilo
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Larissa Espindola da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariana Pacheco de Oliveira
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Mariella Reinol da Silva
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Pablo Michel Pereira Barcelos
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabiana Pereira Santos
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Franciane Bobinski
- Experimental Neuroscience Laboratory (LaNEx), Postgraduate Program in Health Sciences, University of Southern Santa Catarina (UNISUL), Palhoça, Brazil
| | - Gislaine Tezza Rezin
- Laboratory of Neurobiology of Inflammatory and Metabolic Processes, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Maurício Yonamine
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Antonio Inserra
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil
| | - Fabricia Petronilho
- Laboratory of Experimental Neurology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, Santa Catarina, Brazil
| | - Rafael Mariano de Bitencourt
- Behavioral Neuroscience Laboratory, Postgraduate Program in Health Sciences, University of South Santa Catarina (UNISUL), Tubarão, Brazil.
| |
Collapse
|
9
|
Zhang L, Hu Y, Zhang J, Cai M, Lan L, Sun G. Application of chemical pattern recognition and similarity evaluation in electrochemical and HPLC-DAD fingerprints for quality consistency study of herbal medicines. Anal Chim Acta 2025; 1349:343830. [PMID: 40074459 DOI: 10.1016/j.aca.2025.343830] [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: 12/12/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Herbal medicines and their preparations play a significant role in healthcare systems, yet concerns remain about their quality consistency. Chemical fingerprinting and multi-component quantitative analysis are the commonly used analytical methods and are widely applied in the quality analysis of herbal medicines. The study uses Gegen Qinlian tablets (GQTs) as a case to propose a comprehensive quality consistency evaluation system. RESULTS Initially, the evaluation system is based on three quality components and three mixtures representing RPL, RS, and RC, categorizing all samples into eight levels. Subsequently, a four-wavelength fusion HPLC profiling (FWFP) method was established, yielding a relative standard deviation (RSD) of 0.43 % for mean relative retention times (RRA) and 21.12 % for relative retention area using the normalized fingerprint method (NFM). The systematically quantified fingerprint method (SQFM) was employed, resulting in qualitative similarity (Sm) and quantitative similarity (Pm) ranges of 0.878-0.978 and 74.9%-120.4 %, respectively. Concurrently, the Electrochemical Fingerprint method (ECFM) was applied for joint evaluation with FWFP, producing SE and projection quantitative similarity (CE) ranges of 0.962-1.000 and 70.6-155.2 %, respectively. Ultimately, the series spectra from FWFP and ECFM were used to comprehensively assess sample quality, with SFW-EC and CEW-EC ranges of 0.891-0.979 and 87.5-120.9 %, leading to the classification of the 22 GQT batches into five grades. SIGNIFICANCE The study first proposes using characteristic parameters of the ECFM combined with SE and CE to evaluate the similarity of electrochemical fingerprints. It also comprehensively describes and uses SQFM to evaluate the quality of herbal medicines, including methodological validation, qualitative similarity (Sm), quantitative similarity (Pm), and reliability assessment. These methods may provide new insights for the similarity evaluation of different types of fingerprints, which can be applied in the quality consistency study of herbal medicines.
Collapse
Affiliation(s)
- Liuwei Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Yanlei Hu
- China National Pharmaceutical Industry Corporation Ltd., Beijing, 101301, China
| | - Jianglei Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Ming Cai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Lili Lan
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Guoxiang Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
10
|
Zhou P, Chao Q, Li C, Wang N, Guo S, Wang P, Ge P, Li C, Guo R, Yang N, Lu Z, Tang Z, Zhang Q, Fu T, Xiao Q, Zhu H. Microglia-targeting nanosystems that cooperatively deliver Chinese herbal ingredients alleviate behavioral and cognitive deficits in Alzheimer's disease model mice. J Nanobiotechnology 2025; 23:313. [PMID: 40275381 PMCID: PMC12020378 DOI: 10.1186/s12951-025-03385-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025] Open
Abstract
The effective treatment of Alzheimer's disease (AD) is challenging because of its complex and controversial pathological mechanisms. Moreover, multiple barriers, such as the blood-brain barrier (BBB), reduce drug delivery efficiency. Microglia-related neuroinflammation has recently attracted increasing attention as a possible cause of AD and has become a novel therapeutic target. Therefore, overcoming the BBB and targeted delivery of anti-inflammatory agents to microglia seem to be effective practical strategies for treating AD. A large proportion of natural active extracts possess exceptional immunomodulating capabilities. In this study, the cooperative delivery of berberine (Ber) and palmatine (Pal) by transferrin-decorated extracellular vesicles (Tf-hEVs-Ber/Pal), which can cross the BBB and precisely target microglia, was performed. This nanosystem effectively cleared amyloid β-protein (Aβ) aggregates, significantly regulated the neuroinflammatory environment both in vitro and in vivo and markedly altered the behavior and improved the cognitive and learning abilities of AD model mice. The efficacy of a microglia-targeting combined therapeutic approach for AD was demonstrated, which broadens the potential application of Chinese herbal ingredients.
Collapse
Affiliation(s)
- Peng Zhou
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Quan Chao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ningjing Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Siqi Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingping Wang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pingyuan Ge
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Caihong Li
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Guo
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Nianyun Yang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhangdi Lu
- Polymer Science and Materials Chemistry, Exponent Ltd, Hong Kong, 999077, China
| | - Zhishu Tang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources, Shannxi University of Chinese Medicine, Xianyang, 712038, China
- China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qichun Zhang
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tingming Fu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qingqing Xiao
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Huaxu Zhu
- Jiangsu Botanical Medicine Refinement Engineering Research Center, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
11
|
Li P, Liao H, Lv Y, Liang S, Zhong J, Su W. Ca 2+ crosslinked gelatin-sodium alginate film loaded with berberine hydrochloride that can effectively promote wound healing of MRSA infection. Int J Biol Macromol 2025; 310:143273. [PMID: 40250645 DOI: 10.1016/j.ijbiomac.2025.143273] [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: 11/20/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Wound infections are a primary cause of delayed wound healing, and film dressings have garnered significant attention in wound management. However, the development of biologically sourced and green-synthesized film dressings with rapid antimicrobial functionality remains an urgent technological breakthrough. In this study, we successfully developed gelatin sodium alginate/sea sheath molecularly modified berberine hydrochloride film (GS/CB). The incorporation of sea sheath powder significantly enhances the mechanical properties of the gelatin‑sodium alginate film.The calcium ions (Ca2+) provided by the sea sheath create a cross-linking effect within the sodium alginate-gelatin network, resulting in tensile properties of the GS/CB films that are 2.6 times greater than those of the control films. This enhancement allows the films to meet the requirements for wound applications. In in vitro antimicrobial experiments, the films incorporated with sodium oleate-modified berberine hydrochloride showed good antimicrobial effects, with an inhibition rate of 97.79 % against Staphylococcus aureus (S. aureus) and 98.57 % against methicillin-resistant Staphylococcus aureus (MRSA). In in vivo wound healing experiments, the film effectively eliminated MRSA from the wound, achieving a bacterial clearance rate of 98.5 %. Compared to transparent film dressings, the GS/CB film demonstrated superior wound care efficacy and significantly accelerated wound healing. Furthermore, both in vivo and in vitro tests indicated that the film possesses excellent biocompatibility. Therefore, the GS/CB film holds great potential and application value as a new wound dressing for the treatment of infectious wounds.
Collapse
Affiliation(s)
- Peiyuan Li
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China.
| | - Huan Liao
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yingbin Lv
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Siyan Liang
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Jingwen Zhong
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Wei Su
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, Nanning Normal University, Nanning 530001, China.
| |
Collapse
|
12
|
Efati M, Sahebkar A, Tavallaei S, Alidadi S, Hosseini H, Hamidi-Alamdari D. Protective effect of Leuco-methylene blue against acetaminophen-induced liver injury: an experimental study. Drug Chem Toxicol 2025:1-13. [PMID: 40207489 DOI: 10.1080/01480545.2025.2485347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/21/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
Acetaminophen is a commonly used drug for mild to moderate pain relief; however, acetaminophen toxicity due to the formation of toxic metabolites is a major cause of drug-induced liver injury. Methylene blue is an FDA-approved drug for the treatment of methemoglobinemia and has potential applications in the treatment of carbon monoxide and cyanide poisoning. Leuco-methylene blue, a colorless form of methylene blue, is more effective in entering cells and counteracting oxidative stress, making it a valuable option in regulating mitochondrial function and ATP production. In this study, we aimed to evaluate the effect of LMB on liver damage caused by acetaminophen toxicity. Thirty-six rats were divided into six groups: control, APAP, NAC, LMB, MB, and NAC+LMB. All groups except the control received acetaminophen (1500 mg/kg), followed by treatments with NAC (100 mg/kg), LMB (5 mg/kg), MB (5 mg/kg), and NAC+LMB after 3 hours. The rats were sacrificed 24 hours post-acetaminophen administration. LMB significantly reduced serum levels of liver enzymes (ALT, AST, and ALP) and increased the expression of genes involved in mitochondrial biogenesis and antioxidant defense (PGC-1, Nrf2, and Tfam). Additionally, LMB significantly increased total antioxidant capacity and glutathione reductase levels, decreased the prooxidant-antioxidant balance (PAB), and reduced the expression of inflammatory cytokines (IL-6 and TNF-α) in the liver tissue. LMB effectively reduced the severity of acetaminophen-induced liver damage through antioxidant and anti-inflammatory effects. LMB can effectively ameliorate APAP-induced toxicity in rats, with comparable efficacy to N-acetylcysteine with respect to most complications of acetaminophen-induced toxicity in rats.
Collapse
Affiliation(s)
- Majid Efati
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Tavallaei
- Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodeh Alidadi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daryoush Hamidi-Alamdari
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
13
|
Ullah N, Karim A, Iqbal M, Malekshah RE, Ali S, Haribabu J, Hsu SCN. Molecular docking of Cu(II) and Zn(II) complexes for tyrosinase inhibition and drug loading on boron nitride nanotube scaffolds using Monte Carlo simulations. J Mol Model 2025; 31:135. [PMID: 40192849 DOI: 10.1007/s00894-025-06355-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/22/2025] [Indexed: 05/09/2025]
Abstract
CONTEXT Recent studies on drug delivery systems incorporating boron nitride nanostructures (BNNTs) highlight their excellent chemical stability and non-cytotoxic properties, positioning them as a promising platform for drug release in biomedical applications. This study aimed to optimize the mono-nuclear structures of Cu(II) and Zn(II) complexes and to functionalize zigzag (13, 13) boron nitride nanotubes with glutamic acid (GABNNTs). Based on Monte Carlo, the results revealed that complexes 6 and 19 exhibited stronger interactions with GABNNTs, attributed to π-π stacking between bipyridine/phenanthroline ligands and GABNNTs. This interaction suggests a greater challenge in their release compared to other compounds. The interaction energy analysis further revealed that complexes 1, 4, and 12/GABNNTs exhibited the lowest stability, indicating weaker binding interactions between these complexes and the GABNNT surface. The adsorption of all complexes on GABNNTs was primarily found to be physisorption. Molecular docking with mushroom tyrosinase (2Y9X) identified complexes 5, 10, 11, 15, and 20 as having the strongest interactions, a trend that is partially supported by chemical hardness analysis. However, DFT-D results indicated that complexes 5, 11, and 20 exhibited the lowest chemical stability, suggesting a trade-off between strong interactions and lower stability in these complexes. METHODS The energies of these systems were estimated using dispersion-corrected density functional theory (DFT-D) calculations performed in Materials Studio 2017. To evaluate the drug delivery potential of GABNNTs for Cu(II) and Zn(II) complexes, the Monte Carlo (MC) method was employed. The structural and electronic properties, as well as the relationship between biological activities and ΔEg, were analyzed by calculating the HOMO-LUMO energy gap using the dispersion-corrected density functional theory (DFT-D) method. Molecular docking was used to interact with mushroom tyrosinase (2Y9X).
Collapse
Affiliation(s)
- Najeeb Ullah
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Chemistry, Bacha Khan University, KPK, Charsadda, 24420, Pakistan
| | - Amir Karim
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Chemistry, Bacha Khan University, KPK, Charsadda, 24420, Pakistan
| | - Muhammad Iqbal
- Department of Chemistry, Bacha Khan University, KPK, Charsadda, 24420, Pakistan.
| | - Rahime Eshaghi Malekshah
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Chemistry, Semnan University, Semnan, Iran.
| | - Saqib Ali
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45320, Pakistan
| | - Jebiti Haribabu
- Facultad de Medicina, Universidad de Atacama, Los Carreras 1579, 1532502, Copiapo, Chile
- Chennai Institute of Technology, Chennai, 600069, India
| | - Sodio C N Hsu
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
14
|
Liang J, Liu W, Zhang T, Guo D, Gong J, Yang Z. Utilization of natural products in diverse pathogeneses of diseases associated with single or double DNA strand damage repair. Chin Med 2025; 20:46. [PMID: 40197523 PMCID: PMC11974029 DOI: 10.1186/s13020-025-01089-y] [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: 11/24/2024] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
The appearance of DNA damage often involves the participation of related enzymes, which can affect the onset and development of various diseases. Several natural active compounds have been found to efficiently adjust the activity of crucial enzymes associated with single or double-strand DNA damage, thus demonstrating their promise in treating diseases. This paper provides an in-depth examination and summary of these modulation mechanisms, leading to a thorough review of the subject. The connection between natural active compounds and disease development is explored through an analysis of the structural characteristics of these compounds. By reviewing how different scholarly sources describe identical structures using varied terminology, this study also delves into their effects on enzyme regulation. This review offers an in-depth examination of how natural active compounds can potentially be used therapeutically to influence key enzyme activities or expression levels, which in turn can affect the process of DNA damage repair (DDR). These natural compounds have been shown to not only reduce the occurrence of DNA damage but also boost the efficiency of repair processes, presenting new therapeutic opportunities for conditions such as cancer and other disease pathologies. Future research should focus on clarifying the exact mechanisms of these compounds to maximize their clinical utility and support the creation of novel approaches for disease prevention and treatment.
Collapse
Affiliation(s)
- Jiali Liang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wanqing Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dean Guo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Jiyu Gong
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Zizhao Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Laboratory Animal Service and Experiments, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| |
Collapse
|
15
|
Lee KH, Kim MH, Nam HJ. Anti-Inflammatory Effects of Hyeonggaeyeongyo-tang: Evidence from In Vitro and In Vivo Studies. Life (Basel) 2025; 15:587. [PMID: 40283142 PMCID: PMC12028476 DOI: 10.3390/life15040587] [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: 02/25/2025] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Hyeonggaeyeongyo-tang (HGYGT), a traditional herbal formula, is used to treat inflammatory otorhinolaryngological diseases such as otitis media and sinusitis. In this study, we investigated the anti-inflammatory effects of HGYGT in LPS-stimulated RAW 264.7 cells (in vitro) and a carrageenan (CA)-induced rat paw edema model (in vivo). In LPS-stimulated RAW 264.7 cells, treatment with HGYGT (100 and 300 μg/mL) significantly reduced nitric oxide (NO) production by 24.5% and 51.3%, respectively (p < 0.05, p < 0.01). It also significantly suppressed the production of PGE2 (49.8%), IL-1β (42.7%), IL-6 (45.6%), and TNF-α (47.2%) at 300 μg/mL (p < 0.01). A Western blot analysis confirmed that HGYGT (300 μg/mL) significantly downregulated iNOS and COX-2 expression by 58.4% and 53.1%, respectively, while COX-1 remained unaffected. And HGYGT treatment at 300 μg/mL markedly inhibited NF-κB activation by 44.9% (p < 0.01). Furthermore, HGYGT selectively inhibited JNK phosphorylation by 46.7% (p < 0.01), without significantly affecting ERK1/2 or p38 MAPKs. In the CA-induced rat paw edema model, oral administration of HGYGT (1.0 g/kg) reduced paw swelling by 31.5% at 4 h post-injection (p < 0.01) and significantly decreased iNOS expression in inflamed paw tissues by 43.2% (p < 0.01). A histological analysis revealed that HGYGT (1.0 g/kg) reduced inflammatory cell infiltration by 39.6% in the affected tissue (p < 0.05), demonstrating its anti-inflammatory potential. Our findings demonstrate that HGYGT exerts anti-inflammatory effects by suppressing the JNK and NF-κB signaling pathways in LPS-stimulated RAW 264.7 cells, reducing the production of inflammatory mediators. Notably, HGYGT selectively inhibits COX-2 without affecting COX-1 and preferentially suppresses the JNK pathway. Moreover, its in vivo anti-inflammatory effects were confirmed through iNOS inhibition and histopathological analysis. These findings provide robust scientific evidence supporting the traditional use of HGYGT and its anti-inflammatory properties.
Collapse
Affiliation(s)
- Keun Hee Lee
- Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
- Kapsan Oriental Medicine Clinic, Gyeongju 38203, Republic of Korea
| | - Min Hee Kim
- Department of Ophthalmology, Otolaryngology, and Dermatology, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Hae Jeong Nam
- Department of Ophthalmology, Otolaryngology, and Dermatology, Kyung Hee University College of Korean Medicine, Kyung Hee University Medical Center, Seoul 02447, Republic of Korea
| |
Collapse
|
16
|
Van Eygen G, Gilles A, Garcia-Chirino J, Baylan N, Buekenhoudt A, Van der Bruggen B, Luis P. Effects of silica nanoparticle addition and PDMS coating on membrane performance and stability in the extraction of aromatic amines. J Colloid Interface Sci 2025; 683:68-78. [PMID: 39671901 DOI: 10.1016/j.jcis.2024.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/15/2024]
Abstract
This study investigates novel strategies to improve membrane performance and stability in the extraction of aromatic amines for chiral amine production. The effects of silica nanoparticle addition and polydimethylsiloxane (PDMS) coating were explored, with a focus on the selective extraction of α-methylbenzylamine (MBA) and 1-methyl-3-phenylpropylamine (MPPA) from isopropyl amine (IPA). This work introduces a comparative analysis between open and tight membrane extraction (ME) systems, with and without the ionic liquid (IL) [P6,6,6,14][N(Tf)2]. The results reveal that PDMS creates a uniform and dense coating, particularly on PTFE and PVDF supports, while silica nanoparticle coatings were less stable, retaining only 50 % of nanoparticles after ME testing. A PDMS-coated PTFE membrane achieved significantly higher solute fluxes of 1.12 ± 0.01, 1.66 ± 0.02, and 0.36 ± 0.08 g/(m2h) for MBA, MPPA, and IPA, respectively, compared to an IL-wetted PTFE membrane, which was found to have fluxes of 0.60 ± 0.06, 1.01 ± 0.04, and 0.33 ± 0.10 g/(m2h) for the same solutes. A reduction in the pore size of the PTFE support further increased the fluxes to 1.74 ± 0.28, 2.75 ± 0.25, and 0.45 ± 0.08 g/(m2h) for MBA, MPPA, and IPA, respectively, achieving selectivity values of 3.83 ± 0.65 for MBA/IPA and 6.24 ± 0.88 for MPPA/IPA. Although IL impregnation marginally improved selectivity, it caused a significant reduction in solute fluxes. The PDMS coating retained 92.1 % of its mass after 24 h, while the IL retained 87.2 % over the same period. Compared to the tested IL, which presents safety concerns due to its flammability and corrosiveness, PDMS coatings provide a safer and more environmentally friendly alternative, as PDMS is non-toxic and does not bioaccumulate. These findings underscore the superior performance and environmental benefits of novel PDMS-coated membranes in tight ME setups compared to IL-based open ME systems.
Collapse
Affiliation(s)
- Gilles Van Eygen
- Unit Separation and Conversion Technology, Vlaamse Instelling voor Technologisch Onderzoek (VITO NV), Boeretang 200, 2400 Mol, Belgium; Materials & Process Engineering (IMAP), UCLouvain, Place Sainte Barbe 2, B-1348 Louvain-la-Neuve, Belgium; Process Engineering for Sustainable Systems (ProcESS), KU Leuven, Celestijnenlaan 200f, 3001 Leuven, Belgium.
| | - Amaury Gilles
- Materials & Process Engineering (IMAP), UCLouvain, Place Sainte Barbe 2, B-1348 Louvain-la-Neuve, Belgium
| | - Julieta Garcia-Chirino
- Process Engineering for Sustainable Systems (ProcESS), KU Leuven, Celestijnenlaan 200f, 3001 Leuven, Belgium
| | - Nilay Baylan
- Process Engineering for Sustainable Systems (ProcESS), KU Leuven, Celestijnenlaan 200f, 3001 Leuven, Belgium; Department of Chemical Engineering, Istanbul University-Cerrahpaşa, Avcılar, 34320 Istanbul, Turkey
| | - Anita Buekenhoudt
- Unit Separation and Conversion Technology, Vlaamse Instelling voor Technologisch Onderzoek (VITO NV), Boeretang 200, 2400 Mol, Belgium
| | - Bart Van der Bruggen
- Process Engineering for Sustainable Systems (ProcESS), KU Leuven, Celestijnenlaan 200f, 3001 Leuven, Belgium
| | - Patricia Luis
- Materials & Process Engineering (IMAP), UCLouvain, Place Sainte Barbe 2, B-1348 Louvain-la-Neuve, Belgium; Research & Innovation Centre for Process Engineering (ReCIPE), Place Sainte Barbe 2, bte L5.02.02, B-1348 Louvain-la-Neuve, Belgium
| |
Collapse
|
17
|
Saadh MJ, Ahmed HH, Kareem RA, Jain V, Ballal S, Singh A, Sharma GC, Devi A, Nasirov A, Sameer HN, Yaseen A, Athab ZH, Adil M. In Silico design and molecular dynamics analysis of imidazole derivatives as selective cyclooxygenase-2 inhibitors. Comput Biol Chem 2025; 115:108341. [PMID: 39808951 DOI: 10.1016/j.compbiolchem.2025.108341] [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: 12/08/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
Cyclooxygenase-2 (COX-2), a key enzyme in the inflammatory pathway, is the target for various nonsteroidal anti-inflammatory drugs (NSAIDs) and selective inhibitors known as coxibs. This study focuses on the development of novel imidazole derivatives as COX-2 inhibitors, utilizing a Structure-Activity Relationship (SAR) approach to enhance binding affinity and selectivity. Molecular docking was performed using Autodock Vina, revealing binding energies of -6.928, -7.187, and -7.244 kJ/mol for compounds 5b, 5d, and 5e, respectively. Molecular dynamics simulations using GROMACS provided insights into the stability and conformational changes of the protein-ligand complexes. Key metrics such as RMSD, RMSF, Rg, SASA, and hydrogen bond analysis were employed to assess the interactions. The binding free energy of the inhibitors was estimated using the MMPBSA method, highlighting compound 5b (N-[(3-benzyl-2-methylsulfonylimidazol-4-yl)methyl]-4-methoxyaniline) with the lowest binding energy of -162.014 kcal/mol. ADMET analysis revealed that compound 5b exhibited the most favorable pharmacokinetic properties and safety profile. Overall, this investigation underscores the potential of these novel imidazole derivatives as effective COX-2 inhibitors, with compound 5b emerging as the most promising candidate for further development.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | | | - Vicky Jain
- Marwadi University Research Center, Department of Chemistry, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab 140401, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Anita Devi
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - Abdulaziz Nasirov
- Department of Psychiatry, narcology and pediatric narcology, medical psychology and psychotherapy, Tashkent Pediatric Medical Institute, Bogishamol Street 223, Tashkent 100140, Uzbekistan
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
18
|
Enioutina EY, Job KM, Sherwin CM. The Special Issue "The 20th Anniversary of Pharmaceuticals-Multi-Targeted Natural Products as Therapeutics" Editorial-Multi-Targeted Therapeutics from Natural Sources: What Do We Know? Pharmaceuticals (Basel) 2025; 18:442. [PMID: 40283880 PMCID: PMC12030276 DOI: 10.3390/ph18040442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/14/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Herbal and marine products in the form of extracts, infusions, and decoctions have been used for centuries in folk and traditional medicine [...].
Collapse
Affiliation(s)
- Elena Y. Enioutina
- The Division of Clinical Pharmacology, Pediatrics, The Spencer Fox Eccles School of Medicine, Salt Lake City, UT 84108, USA;
| | - Katheleen M. Job
- The Division of Clinical Pharmacology, Pediatrics, The Spencer Fox Eccles School of Medicine, Salt Lake City, UT 84108, USA;
| | - Catherine M. Sherwin
- Internal Medicine, UWA Medical School, The University of Western Australia, Perth, WA 6009, Australia;
| |
Collapse
|
19
|
Zou W, Wang B, Feng X, Lai C, Tan Q. Ultra-High-Performance Liquid Chromatography-Tandem Mass Spectrometry Combined With Network Pharmacology to Elucidate the Bioactive Ingredients and Potential Mechanism of Wu-Teng Decoction for Treatment of Rheumatoid Arthritis. Chem Biodivers 2025:e202403454. [PMID: 40035706 DOI: 10.1002/cbdv.202403454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/06/2025]
Abstract
Wu-Teng Decoction (WTD) is a significant in-hospital preparation widely used in clinical practice to treat rheumatoid arthritis (RA) in China, however, its active substances and underlying mechanisms remain unclear. In this study, the chemical constituents of WTD were analyzed using ultra-high-performance liquid chromatography-tandem mass spectrometry, identifying a total of 120 compounds, including flavonoids, phenylpropanoids, phenolic acids, alkaloids, etc. Subsequently, network pharmacology analyses revealed that 29 compounds were potential active compounds in WTD for the treatment of RA, as well as 48 core anti-RA targets, including tumor necrosis factor-α, V-Akt murine thymoma viral oncogene homolog 1, and albumin. Further analysis suggested that WTD treats RA via the phosphoinositide 3-kinase-Akt, mitogen-activated protein kinase, and Ras signaling pathways. Molecular docking analysis of the top five pivotal targets with the core active ingredients demonstrated suitable binding interactions at the active site of target proteins. The significant reduction of nitric oxide levels in lipopolysaccharides-induced RAW264.7 cells validated the anti-inflammation activity of WTD.
Collapse
Affiliation(s)
- Wei Zou
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Bangyong Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Xiaoqing Feng
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
| | - Chunhua Lai
- Department of Traditional Chinese Medicine, Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, China
| | - Qingang Tan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, China
| |
Collapse
|
20
|
Zhang T, Liu M. Cytokines in age-related eye diseases: pathogenesis and potential targets for innovative therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03926-1. [PMID: 40021512 DOI: 10.1007/s00210-025-03926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Age-related eye diseases (AREDs), such as dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), are significant worldwide health concerns due to their rising prevalence and debilitating effects. Despite substantial research on the pathobiology of AREDs, the impact of immune-related alterations caused by aging is still not well understood. Tissue-resident cells and invading immune cells in the eye control innate responses in the event of damage or infection. However, as cells age, they gradually lose their ability to perform their protective duties and develop abnormal characteristics. Therefore, the disrupted regulation of immune responses in the eyes of older individuals enhances their vulnerability to and the intensity of eye disorders. Cytokines, immune system components, have a role in developing AREDs by contributing to inflammation. This paper examines the deficiencies in the pathogenic and therapeutic aspects of pro-inflammatory cytokines in AREDs that require further investigation in future studies.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China
| | - Ming Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China.
| |
Collapse
|
21
|
Karami E, Bazgir B, Shirvani H, Mohammadi MT, Khaledi M. Unraveling the bidirectional relationship between muscle inflammation and satellite cells activity: influencing factors and insights. J Muscle Res Cell Motil 2025; 46:35-51. [PMID: 39508952 DOI: 10.1007/s10974-024-09683-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
Inflammation stands as a vital and innate function of the immune system, essential for maintaining physiological homeostasis. Its role in skeletal muscle regeneration is pivotal, with the activation of satellite cells (SCs) driving the repair and generation of new myofibers. However, the relationship between inflammation and SCs is intricate, influenced by various factors. Muscle injury and repair prompt significant infiltration of immune cells, particularly macrophages, into the muscle tissue. The interplay of cytokines and chemokines from diverse cell types, including immune cells, fibroadipogenic progenitors, and SCs, further shapes the inflammation-SCs dynamic. While some studies suggest heightened inflammation associates with reduced SC activity and increased fibro- or adipogenesis, others indicate an inflammatory stimulus benefits SC function. Yet, the existing literature struggles to delineate clearly between the stimulatory and inhibitory effects of inflammation on SCs and muscle regeneration. This paper comprehensively reviews studies exploring the impact of pharmacological agents, dietary interventions, genetic factors, and exercise regimes on the interplay between inflammation and SC activity.
Collapse
Affiliation(s)
- Esmail Karami
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Bazgir
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mansoor Khaledi
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
22
|
Thakur GS, Gupta AK, Pal D, Vaishnav Y, Kumar N, Annadurai S, Jain SK. Designing novel cabozantinib analogues as p-glycoprotein inhibitors to target cancer cell resistance using molecular docking study, ADMET screening, bioisosteric approach, and molecular dynamics simulations. Front Chem 2025; 13:1543075. [PMID: 40084274 PMCID: PMC11903459 DOI: 10.3389/fchem.2025.1543075] [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: 12/10/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction One of the foremost contributors to mortality worldwide is cancer. Chemotherapy remains the principal strategy for cancer treatment. A significant factor leading to the failure of cancer chemotherapy is the phenomenon of multidrug resistance (MDR) in cancer cells. The primary instigator of MDR is the over expression of P-glycoprotein (P-gp), a protein that imparts resistance and facilitates the ATP-dependent efflux of various anticancer agents. Numerous efforts have been made to inhibit P-gp function with the aim of restoring the effectiveness of chemotherapy due to its broad specificity. The main objective has been to create compounds that either serve as direct P-gp inhibitors or interact with cancer therapies to modulate transport. Despite substantial in vitro achievements, there are currently no approved drugs available that can effectively "block" P-gp mediated resistance. Cabozantinib (CBZ), a multi-kinase inhibitor, is utilized in the treatment of various carcinomas. CBZ has been shown to inhibit P-gp efflux activity, thereby reversing P-gp mediated MDR. Consequently, P-gp has emerged as a critical target for research in anti-cancer therapies. Methods The purpose of this study was to computationally identify new andsafer analogues of CBZ using bioisosteric approach, focusing on improved pharmacokinetic properties andreduced toxicity. The physicochemical, medicinal, and ADMET profiles of generated analogues were computed using the ADMETLab 3.0 server. We also predicted the drug likeness (DL) and drug score (DS) of analogues. The molecular docking studies of screened analogues against the protein (PDB ID: 3G5U) were conducted using AutoDock Vina flowing by BIOVIA Discovery Studio for visualizing interactions.Molecular dynamics (MD) simulation of docked ligands was done using Schrödinger suite. Results and Discussion The docking scores for the ligands CBZ01, CBZ06, CBZ11, CBZ13, CBZ25, CBZ34, and CBZ38 ranged from -8.0 to -6.4 kcal/mol against the protein (PDB ID: 3G5U). A molecular dynamics (MD) simulation of CBZ01, CBZ13, and CBZ38 was conducted using the Schrödinger suite, revealing that these complexesmaintained stability throughout the 100 ns simulation. Conclusion An integrated computational approach combining bioisosteric approach, molecular docking, drug likeness calculations, and MD simulations highlights the promise of ligands CBZ01 and CBZ13 as candidates for the development of potential anticancer agents for the treatment of various cancers.
Collapse
Affiliation(s)
- Gajendra Singh Thakur
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ajay Kumar Gupta
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Dipti Pal
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Yogesh Vaishnav
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles’ College of Pharmacy, Udaipur, Rajasthan, India
| | - Sivakumar Annadurai
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Sanmati Kumar Jain
- Drug Discovery and Research Laboratory, Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
23
|
Rushendran R, Singh S A, Begum RF, Chitra V, Ali N, Prajapati BG. Bioinformatics Exploration of the Therapeutic Potential of Lotus Seed Compounds in Multiple Sclerosis: A Network Analysis of c-Jun Pathway. Drug Dev Res 2025; 86:e70038. [PMID: 39756059 DOI: 10.1002/ddr.70038] [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: 10/28/2024] [Revised: 12/04/2024] [Accepted: 12/08/2024] [Indexed: 01/07/2025]
Abstract
The central nervous system is affected by multiple sclerosis (MS), a chronic autoimmune illness characterized by axonal destruction, demyelination, and inflammation. This article summarizes the state of the field, highlighting its complexity and significant influence on people's quality of life. The research employs a network pharmacological approach, integrating systems biology, bioinformatics, and pharmacology to identify biomarkers associated with MS. Utilizing Nelumbo Nucifera (Lotus) seeds, the study involves toxicity assessments, biomolecule screening, and target prediction. Advanced computational methodologies are employed, including molecular docking and dynamic simulations, to assess potential therapeutic interactions. Biomolecule screening identifies eight active compounds from Lotus seeds, including Anonaine and Liriodenine. Target prediction reveals 264 common targets with MS-related genes. Protein-protein interaction analysis establishes a complex network, identifying central targets like SRC and AKT1. Bioinformatics enrichment analysis uncovers potential therapeutic candidates and pathways. A Biomolecule-Target-Pathway network diagram visualizes interactions, with Anonaine and Liriodenine exhibiting strong binding affinities in molecular docking studies. Molecular dynamics simulations provide insights into dynamic interactions. In conclusion, through advanced computational techniques, it unveils molecular interactions, potential therapies, and pathways, bridging predictions with practical applications. Anonaine and Liriodenine show promise in curbing MS biomarkers.
Collapse
Affiliation(s)
- Rapuru Rushendran
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College of Pharmacy, Chennai, India
| | - Ankul Singh S
- Department of Pharmacology, Dr. M.G.R Educational and Research Institute, Faculty of Pharmacy, Chennai, India
| | - Rukaiah Fatma Begum
- Department of Pharmacology, Institute of Pharmaceutical Research, GLA University, Mathura, India
| | - Vellapandian Chitra
- Department of Pharmacology, SRM Institute of Science and Technology, SRM College of Pharmacy, Chennai, India
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Bhupendra G Prajapati
- Department of Pharmaceutics, Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, India
- Department of Industrial Pharmacy, Faculty of Pharmacy, Sanam Chandra Palace Campus, Silpakorn University, Nakhon Pathom, Thailand
| |
Collapse
|
24
|
Dai ZQ, Guo ZQ, Zhang T, Chu YF, Yan Y, Gao F, Li SL, Gu YH, Jiao JY, Lin YX, Zhao SW, Xu B, Lei HM. Integrating network pharmacology and transcriptomics to study the potential mechanism of Jingzhi Niuhuang Jiedu tablet in rats with accumulation of heat in the lungs and stomach. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118890. [PMID: 39366495 DOI: 10.1016/j.jep.2024.118890] [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: 07/07/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Accumulation of heat in the lungs and stomach (AHLS) is an important syndrome within the realm of traditional Chinese medicine (TCM). It is the fundamental reason behind numerous illnesses, including mouth ulcers, dermatological conditions, acne, and pharyngitis. Jingzhi Niuhuang Jiedu tablet (JN) serves as the representative prescription for treatment of AHLS clinically. However, the effective components and mechanism of JN's impact on AHLS remain unexplored. AIM OF THE STUDY The objective of this research was to analyze the effective components of JN and investigate the therapeutic effect and potential mechanism of JN on AHLS. MATERIALS AND METHODS The effective compounds of JN extract were analyzed and identified using UHPLC-Q-Exactive/HRMS. Utilizing network pharmacology to investigate JN's multi-target, multi-pathway process in treating AHLS. Subsequently, anti-inflammatory activities of JN extract were evaluated in the RAW264.7 cells stimulated by lipopolysaccharide (LPS). In addition, a rat AHLS model induced by LPS and dried ginger was established. Pathological changes in rat lung and stomach tissues observed by HE staining and Masson's trichrome staining. Additionally, the expression of TNF-α, IL-6, and IL-1β in bronchoalveolar lavage fluid (BALF) was identified through the ELISA assay. For a deeper understanding of how JN might affect AHLS, transcriptomics was utilized to examine differential genes and their underlying mechanisms. Concurrently, techniques like quantitative polymerase chain reaction (q-PCR), immunofluorescence, and western blotting (WB) were employed to confirm various mRNA and protein expression, including Il17ra, Il17re, IL-17A, IL-1β, IL-6, PPARγ, PGC1-α and UCP1. RESULTS We identified 178 potential effective components in the JN extract. Network pharmacology analysis showed that the 144 components in JN act on 200 key targets for the treatment of AHLS by suppressing inflammation, regulating energy metabolism, and gastric function. In addition, JN suppressed the LPS-stimulated generation of NO, TNF-α, IL-1β, and IL-6 in RAW264.7 cells. And JN treatment effectively alleviated lung and stomach injury and reduced inflammation in rats. Analysis of RNA-seq from lung tissues revealed JN's substantial control over crucial genes in the IL-17 signaling pathway, including Il1b and Il17ra. Likewise, RNA sequencing of stomach tissues revealed that JN markedly decreased crucial genes in the Thermogenesis pathway, including Ppargc1a and Ppara. Additional experimental findings confirmed that treatment with JN significantly reduced the expression levels of mRNA (Il17ra, Il17re, Il1b, Ppargc1a and Ucp1), and the expression levels of protein (IL-17A, IL-1β, IL-6, PPARγ, PGC1-α and UCP1). CONCLUSION This study not only analyzes the effective components of JN but also reveals that JN could effectively ameliorate AHLS by inhibiting IL-17 signaling pathway and Thermogenesis pathway, which provides evidence for subsequent clinical studies and drug development.
Collapse
Affiliation(s)
- Zi-Qi Dai
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Zhuo-Qian Guo
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Tong Zhang
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Ya-Fen Chu
- Beijing Tongrentang Science and Technology Development Co. Technology Development Co., Ltd., Beijing, 100079, China
| | - Ying Yan
- Beijing Tongrentang Science and Technology Development Co. Technology Development Co., Ltd., Beijing, 100079, China
| | - Feng Gao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Shan-Lan Li
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yu-Hao Gu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Jing-Yi Jiao
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Yi-Xuan Lin
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China
| | - Shu-Wu Zhao
- Beijing Tongrentang Science and Technology Development Co. Technology Development Co., Ltd., Beijing, 100079, China.
| | - Bing Xu
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China.
| | - Hai-Min Lei
- School of Chinese Pharmacy, Beijing University of Chinese Medicine, Beijing, 100102, China.
| |
Collapse
|
25
|
Belal A, Abdou A, Miski SF, Ali MAM, Ghamry HI, Obaidullah AJ, Zaky MY, Hassan AHE, Roh EJ, Al-Karmalawy AA, Ibrahim MH. Exploring the potential of some natural indoles as antiviral agents: quantum chemical analysis, inverse molecular docking, and affinity calculations. Front Chem 2025; 12:1521298. [PMID: 39886558 PMCID: PMC11779707 DOI: 10.3389/fchem.2024.1521298] [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: 11/01/2024] [Accepted: 12/17/2024] [Indexed: 02/01/2025] Open
Abstract
Human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections represent critical global health challenges due to the high morbidity and mortality associated with co-infections. HIV, the causative agent of acquired immunodeficiency syndrome (AIDS), infects 4,000 people daily, potentially leading to 1.2 million new cases by 2025, while HCV chronically affects 58 million people, causing cirrhosis and hepatocellular carcinoma. Indole-based compounds play a crucial role in antiviral drug development due to their "privileged scaffold" structure. This study investigates the antiviral potential of natural indoles, gardflorine A-C, derived from Gardneria multiflora Makino, a plant traditionally used to treat various ailments. We employed molecular docking, ADMET analysis, and computational techniques [frontier molecular orbital (FMO), natural bond orbital (NBO), and density functional theory (DFT)] to evaluate these compounds" potential as multi-target antiviral agents against HIV and HCV proteins.
Collapse
Affiliation(s)
- Amany Belal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aly Abdou
- Chemistry Department, Faculty of science, Sohag university, Sohag, Egypt
| | - Samar F. Miski
- Pharmacology and Toxicology Department, College of Pharmacy, Taibah University, Madinah, Saudi Arabia
| | - Mohamed A. M. Ali
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Heba I. Ghamry
- Nutrition and Food Science, Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Ahmad J. Obaidullah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Drug Exploration and Development Chair (DEDC), Department of Pharmaceutical Chemistry, College of Pharmacy, King Saudi University, Riyadh, Saudi Arabia
| | - Mohamed Y. Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, BeniSuef, Egypt
| | - Ahmed H. E. Hassan
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Eun Joo Roh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon, Republic of Korea
| | - Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Chemistry, College of Pharmacy, The University of Mashreq, Baghdad, Iraq
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta, Egypt
| | - Mona H. Ibrahim
- Department of Pharmaceutical Medicinal Chemistry and Drug Design, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
26
|
Du M, Liu X, Ji X, Wang Y, Liu X, Zhao C, Jin E, Gu Y, Wang H, Zhang F. Berberine alleviates enterotoxigenic Escherichia coli-induced intestinal mucosal barrier function damage in a piglet model by modulation of the intestinal microbiome. Front Nutr 2025; 11:1494348. [PMID: 39877539 PMCID: PMC11772193 DOI: 10.3389/fnut.2024.1494348] [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: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Enterotoxic Escherichia coli (ETEC) is the main pathogen that causes diarrhea, especially in young children. This disease can lead to substantial morbidity and mortality and is a major global health concern. Managing ETEC infections is challenging owing to the increasing prevalence of antibiotic resistance. Berberine, categorized as a substance with similarities in "medicine and food," has been used in China for hundreds of years to treat gastrointestinal disorders and bacteria-induced diarrhea. This study investigated the preventive effect of dietary berberine on the intestinal mucosal barrier induced by ETEC and the microbial community within the intestines of weaned piglets. Methods Twenty-four piglets were randomly divided into four groups. Piglets were administered either a standard diet or a standard diet supplemented with berberine at concentrations of 0.05 and 0.1%. and orally administered ETEC or saline. Results Dietary supplementation with berberine reduced diamine oxidase, d-lactate, and endotoxin levels in piglets infected with ETEC (P < 0.05). Berberine increased jejunal villus height, villus/crypt ratio, mucosal thickness (P < 0.05), and goblet cell numbers in the villi and crypts (P < 0.05). Furthermore, berberine increased the optical density of mucin 2 and the mucin 2, P-glycoprotein, and CYP3A4 mRNA expression levels (P < 0.05). Berberine increased the expressions of zonula occludins-1 (ZO-1), zonula occludins-2 (ZO-2), Claudin-1, Occludin, and E-cadherin in the ileum (P < 0.05). Moreover, berberine increased the expression of BCL2, reduced intestinal epithelial cell apoptosis (P < 0.05) and decreased the expression of BAX and BAK in the duodenum and jejunum, as well as that of CASP3 and CASP9 in the duodenum and ileum (P < 0.05). Berberine decreased the expression of IL-1β, IL-6, IL-8, TNF-α, and IFN-γ (P < 0.05) and elevated total volatile fatty acids, acetic acid, propionic acid, valeric acid, and isovaleric acid concentrations (P < 0.05). Notably, berberine enhanced the abundance of beneficial bacteria including Enterococcus, Holdemanella, Weissella, Pediococcus, Muribaculum, Colidextribacter, Agathobacter, Roseburia, Clostridium, Fusicatenibacter, and Bifidobacterium. Simultaneously, the relative abundance of harmful and pathogenic bacteria, such as Prevotella, Paraprevotella, Corynebacterium, Catenisphaera, Streptococcus, Enterobacter, and Collinsella, decreased (P < 0.05). Discussion Berberine alleviated ETEC-induced intestinal mucosal barrier damage in weaned piglets models. This is associated with enhancement of the physical, chemical, and immune barrier functions of piglets by enhancing intestinal microbiota homeostasis.
Collapse
Affiliation(s)
- Min Du
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xinran Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Yue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Hongyu Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| |
Collapse
|
27
|
Amin A, Nikdoust F, Khorram S, Marashi SM, Ghanavati P, Ameri F, Akbarzadeh A, Hasanvand A, Khodakarim N. Dengue virus infection: how platelet-leukocyte crosstalk shapes thrombotic events and inflammation. Mol Biol Rep 2025; 52:119. [PMID: 39804486 DOI: 10.1007/s11033-025-10222-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/02/2025] [Indexed: 05/02/2025]
Abstract
Dengue virus (DENV) poses a considerable threat to public health on a global scale, since about two-thirds of the world's population is currently at risk of contracting this arbovirus. Being transmitted by mosquitoes, this virus is associated with a range of illnesses and a small percentage of infected individuals might suffer from severe vascular leakage. This leakage leads to hypovolemic shock syndrome, generally known as dengue shock syndrome, organ failure, and bleeding complications. The severe form of this disease is believed to be, at least partially, associated with inflammatory and thrombotic states. These issues are significantly affected by the activation of platelets and leukocytes, as well as their interactions, which may influence its prognosis. The platelets present in a thrombus are able to attract leukocytes to the site of injury. The intricate process leads to the significant accumulation, activation, and migration of leukocytes, thereby promoting thrombotic events and triggering inflammatory responses. The occurrence of these events, combined with the direct viral infection of endothelial cells, leads to vascular endothelialitis, the disruption of cellular membranes, and the subsequent release of DAMPs. As a result, considerable damage occurs in the endothelium, which activates neutrophils and platelets; thisleads to their interaction and initiates the process of Netosis. Collectively, these processes exacerbate inflammatory and thrombotic conditions. In this respect, current research has focused on understanding whether effective anti-inflammatory protocols can prevent thrombotic events or, conversely, if efficient anticoagulant regimens may lead to a reduction in cytokine storms and tissue damage. This review article aims to illuminate the platelet leukocyte crosstalk, detailing the mechanisms through which platelets may play a role in the pathogenesis of DENV. The research outputs are particularly important in severe cases, in which case their interactions with leukocytes can exacerbate both inflammation and thrombosis in a mutually reinforcing manner.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Farahnaz Nikdoust
- Department of Cardiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, IR, Iran
| | | | | | - Pedram Ghanavati
- Department of Neurosurgery, Firouzgar Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Farshid Ameri
- Department of Infection, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ashkan Akbarzadeh
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Hasanvand
- Department of General Surgery, Lorestan University of Medical Science, Khorramabad, Iran
- Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Nastaran Khodakarim
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
- Department of medical oncology and hematology, Hazrat Rasoul Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Xue Y, Ye L, Huang C, Ye H. Lights and Shadows of Cytokines in Age-Related Eye Diseases: A Narrative Literature Review. J Biochem Mol Toxicol 2025; 39:e70121. [PMID: 39756061 DOI: 10.1002/jbt.70121] [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: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/21/2024] [Indexed: 01/07/2025]
Abstract
The eye is considered to be an immune-privileged region. However, several parts of the eye have distinct mechanisms for delivering immune cells to the injury sites or even in response to aging. Although these immune responses are intended to be protective, the visual acuity can be compromised by the release of pro-inflammatory cytokines by immune cells, which induce chronic inflammation and fibrosis. Age-related eye diseases (AREDs) are the primary cause of vision impairment (VI) in the elderly, with a poor comprehension of their pathophysiology. Age-related eye diseases affect both the anterior and posterior segments, resulting in diminished quality of life and risk of irreversible blindness. Immune system dysregulation and the upregulation of pro-inflammatory cytokines have been linked to AREDs, underscoring the need to comprehend inflammation's impact on ocular disorders to enhance patient symptom management. In this framework, the PubMed database was searched using the medical subject headings (MeSH) terms "Age-related eye diseases," "dry eye syndrome," "glaucoma," "cataract," "diabetic retinopathy," "inflammation," "interleukin," and "cytokine" with the aim of overview the role of cytokines in AREDs and discuss their potential therapeutic approaches.
Collapse
Affiliation(s)
- Yuyu Xue
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Lu Ye
- Department of Oncology, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, Sichuan, China
| | - Chan Huang
- School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan, China
| | - Hejiang Ye
- Department of Ophthalmology, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
29
|
Qadir S, Alshabrmi FM, Aba Alkhayl FF, Muzammil A, Kaur S, Rehman A. Advancing COVID-19 Treatment: The Role of Non-covalent Inhibitors Unveiled by Integrated Machine Learning and Network Pharmacology. Curr Pharm Des 2025; 31:1307-1326. [PMID: 39819536 DOI: 10.2174/0113816128342951241210175314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 01/19/2025]
Abstract
INTRODUCTION The COVID-19 pandemic has necessitated rapid advancements in therapeutic discovery. This study presents an integrated approach combining machine learning (ML) and network pharmacology to identify potential non-covalent inhibitors against pivotal proteins in COVID-19 pathogenesis, specifically B-cell lymphoma 2 (BCL2) and Epidermal Growth Factor Receptor (EGFR). METHODS Employing a dataset of 13,107 compounds, ML algorithms such as k-Nearest Neighbors (kNN), Support Vector Machine (SVM), Random Forest (RF), and Naïve Bayes (NB) were utilized for screening and predicting active inhibitors based on molecular features. Molecular docking and molecular dynamics simulations, conducted over a 100 nanosecond period, enhanced the ML-based screening by providing insights into the binding affinities and interaction dynamics with BCL2 and EGFR. Network pharmacology analysis identified these proteins as hub targets within the COVID-19 protein-protein interaction network, highlighting their roles in apoptosis regulation and cellular signaling. RESULTS The identified inhibitors exhibited strong binding affinities, suggesting potential efficacy in disrupting viral life cycles and impeding disease progression. Comparative analysis with existing literature affirmed the relevance of BCL2 and EGFR in COVID-19 therapy and underscored the novelty of integrating network pharmacology with ML. This multidisciplinary approach establishes a framework for emerging pathogen treatments and advocates for subsequent in vitro and in vivo validation, emphasizing a multi-targeted drug design strategy against viral adaptability. CONCLUSION This study's findings are crucial for the ongoing development of therapeutic agents against COVID-19, leveraging computational and network-based strategies.
Collapse
Affiliation(s)
- Saba Qadir
- Department of Biochemistry, College of Chemistry, Zhengzhou University, No. 100 Science Avenue, Zhengzhou, Henan, 450001, China
| | - Fahad M Alshabrmi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Faris F Aba Alkhayl
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, 51452 Buraydah, Saudi Arabia
| | - Aqsa Muzammil
- Department of Biology, College of Art and Science, New Mexico State University, 88001, Las Cruces, USA
| | - Snehpreet Kaur
- Department of Computer Sciences, College of Art and sciences. New Mexico State University, 88001, Las Cruces, USA
| | - Abdur Rehman
- Center of Bioinformatics, College of Life Sciences, Northwest Agriculture and Forestry University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
30
|
Qin Y, Zhang D, Shangguan C, Xu C, Li S, Li Y, Liu Y, Jiang S. Altingia chinensis petroleum ether extract suppresses NSCLC via induction of apoptosis, attenuation of EMT, and downregulation of PI3K/Akt pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156218. [PMID: 39566405 DOI: 10.1016/j.phymed.2024.156218] [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: 05/03/2024] [Revised: 10/12/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) is the primary type of lung cancer with the leading cause of fatalities from cancer, and the effective treatment is minimal. Altingia chinensis is a medicinal plant utilized as a traditional folk remedy to alleviate rheumatism, punch injury and paralysis. APE is the petroleum ether extract from A. Chinensis, whose antitumor effects are rarely studied. PURPOSE To explore the antitumor effects of APE on NSCLC and its molecular mechanism. METHODS LLC and H1299 cells were used to explore the anticancer effect of APE on NSCLC in vitro. MTT assay and colony formation were employed to evaluate cell viability. Flow cytometry was used to evaluate apoptosis. Wound healing and transwell were employed to evaluate cell migration and invasive capacity. Meanwhile, an LLC tumor-bearing C57BL/6J mice model was established for assessing the anticancer effect of APE on NSCLC in vivo. H&E staining was used to assess histopathology. TUNEL assay was performed to assess apoptosis in tumor tissue. Network pharmacology, CESTA, and kinase assay were employed to analyze potential molecular mechanisms. Western blots were performed to detect proteins involved in apoptosis, EMT, and the PI3K/Akt pathway. RESULTS This is the first investigation to identify APE's antitumor potential in both NSCLC cells and tumor-bearing mice models. Significantly, APE dose-dependently decreased cell viability and caused morphological changes both in LLC and H1299 cells. Furthermore, APE (31.25, 62.5, and 125 μg/ml) induced apoptosis in NSCLC cells, as demonstrated by increased Annexin V-FITC/PI-stained cells, the cleaved-caspase 3 levels, and the Bax/Bcl-2 ratio. Additionally, APE suppressed cell migration and invasion by the increase of E-cadherin expression and the downregulation of vimentin, implying that APE inhibited cell metastasis via attenuation of EMT. Importantly, intragastric administration of 100 mg/kg APE significantly inhibited tumor growth without apparent side effects. TUNEL assay confirmed the apoptosis in tumor tissue. Western blots validated the alteration of EMT and apoptotic markers in tumor tissue, which matched the in vitro findings. Moreover, APE directly bound to PI3Kα and inhibited its activity, leading to inhibition of the PI3K-Akt pathway. CONCLUSION Overall, APE exhibits anti-tumor effects on NSCLC via induction of apoptosis, attenuation of EMT, and its mechanism involves the suppression of the PI3K/Akt pathway. Our study offers new insights for the identification of novel drug development for NSCLC.
Collapse
Affiliation(s)
- Yiran Qin
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Ganzhou, 341000, China; School of Rehabilitation Medicine, Gannan Medical University, Ganzhou 341000, China
| | - De Zhang
- Department of Pathology, Ganzhou Cancer Hospital, Gannan Medical University, Ganzhou 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| | - Chenxin Shangguan
- First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China.
| | - Chunming Xu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Ganzhou, 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Shuqing Li
- School of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Yumei Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Ganzhou, 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yang Liu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; School of Pharmacy, Nanchang Medical College, Nanchang 330052, China.
| | - Shuping Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China; Jiangxi Provincial Key Laboratory of Tissue Engineering, Ganzhou, 341000, China; School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
31
|
Ouassaf M, Bourougaa L, Bahaz F, Alhatlani BY. Exploring the Antiviral Potential of Artemisia annua Through JAK-STAT Pathway Targeting: A Network Pharmacology Approach. Pharmaceuticals (Basel) 2024; 17:1539. [PMID: 39598448 PMCID: PMC11597232 DOI: 10.3390/ph17111539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Artemisia annua, a plant with antiviral potential, has shown promise against various viral infections, yet its mechanisms of action are not fully understood. This study explores A. annua's antiviral effects using network pharmacology and molecular docking, focusing on key active compounds and their interactions with viral protein targets, particularly within the JAK-STAT signaling pathway-a critical mediator of immune responses to viral infections. METHODS From the TCMSP database, we identified eight active compounds and 335 drug targets for A. annua, with 19 intersecting targets between A. annua compounds and viral proteins. A protein-protein interaction (PPI) network highlighted 10 key hub genes, analyzed further through Gene Ontology (GO) and KEGG pathways to understand their immune and antiviral roles. ADMET properties of the active compound Patuletin (MOL004112) were assessed, followed by 200 ns molecular dynamics simulations to examine its stability in complex with JAK2. RESULTS PPI analysis identified JAK2, MAPK3, MAPK1, JAK1, PTPN1, HSPA8, TYK2, RAF1, MAPT, and HMOX1 as key hub genes, with JAK2 emerging as a critical regulator of immune and antiviral pathways. ADMET analysis confirmed Patuletin's favorable pharmacokinetic properties, and molecular dynamics simulations showed a stable Patuletin-JAK2 complex, with FEL analysis indicating minimal disruption to JAK2's intrinsic flexibility. CONCLUSIONS These findings highlight JAK2 as a promising target in the antiviral activity of A. annua compounds, particularly Patuletin, supporting its potential as an antiviral agent and providing a foundation for further research on A. annua's therapeutic applications.
Collapse
Affiliation(s)
- Mebarka Ouassaf
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 07000, Algeria;
| | - Lotfi Bourougaa
- Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Biskra, BP 145, Biskra 07000, Algeria;
| | - Farial Bahaz
- Laboratory of Organic Materials and Heterochemistry, Echahid Cheikh Larbi Tebessi University, Tebessa 12000, Algeria;
| | - Bader Y. Alhatlani
- Unit of Scientific Research, Applied College, Qassim University, Buraydah 52571, Saudi Arabia
| |
Collapse
|
32
|
Di Serio BF, Muller JDAI, Silva MJD, Figueiredo FDF, Martins DTDO. Phytochemistry and Evaluation of the Anti-Inflammatory Activity of the Hydroethanolic Extract of Virola elongata (Benth.) Warb. Stem Bark. BIOLOGY 2024; 13:776. [PMID: 39452085 PMCID: PMC11505066 DOI: 10.3390/biology13100776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Previous studies of the hydroethanolic extract of Virola elongata inner stem bark (HEVe) have demonstrated its antioxidant, gastroprotective, and antiulcer properties, but have not evaluated its anti-inflammatory potential. METHODS HEVe was obtained by maceration and phytochemically analyzed. Its systemic anti-inflammatory activity was assessed by its effect on lipopolysaccharide (LPS)-induced peritonitis in mice. HEVe gel (HEgVe) was employed to evaluate topical anti-inflammatory activity by measuring the ear edema resulting from croton-oil-induced dermatitis in mice. A cell viability assay was conducted to determine the non-cytotoxic concentrations of the HEVe. RAW 264.7 cells were stimulated by LPS to determinate cytokine and nitric oxide production. RESULTS A phytochemical analysis of the HEVe revealed the presence of phenolic acids, neolignans, flavonoids, and monomeric catechins. The oral treatment of acute peritonitis with HEVe reduced the total leukocytes, neutrophils, TNF-α, and IL-1β and elevated IL-10 levels. The application of the HEgVe reduced local edema. The HEVe on the RAW 264.7 cells exhibited no cytotoxicity, and the cells with HEVe displayed reduced TNF-α, IL-1β, and NO levels and increased IL-13 levels. CONCLUSIONS HEVe demonstrated systemic and topical multitarget anti-inflammatory activity, likely due to the combined effects of secondary metabolites. HEVe emerges as a promising herbal remedy for inflammation with minimal cytotoxicity, emphasizing its potential therapeutic significance.
Collapse
Affiliation(s)
- Bruna Fioravante Di Serio
- Post-Graduate Program in Health Sciences, School of Medicine, Federal University of Mato Grosso (UFMT), Cuiabá 78060-900, Brazil; (J.d.A.I.M.); (F.d.F.F.)
| | - Jessica de Araujo Isaias Muller
- Post-Graduate Program in Health Sciences, School of Medicine, Federal University of Mato Grosso (UFMT), Cuiabá 78060-900, Brazil; (J.d.A.I.M.); (F.d.F.F.)
| | - Marcelo José Dias Silva
- Medicinal Plants and Phytotherapeutics Laboratory, Federal University of Alfenas (UNIFAL), Alfenas 37130-001, Brazil;
| | - Fabiana de Freitas Figueiredo
- Post-Graduate Program in Health Sciences, School of Medicine, Federal University of Mato Grosso (UFMT), Cuiabá 78060-900, Brazil; (J.d.A.I.M.); (F.d.F.F.)
| | | |
Collapse
|
33
|
Wang Y, Zhang Z, Du M, Ji X, Liu X, Zhao C, Pang X, Jin E, Wen A, Li S, Zhang F. Berberine alleviates ETEC-induced intestinal inflammation and oxidative stress damage by optimizing intestinal microbial composition in a weaned piglet model. Front Immunol 2024; 15:1460127. [PMID: 39351242 PMCID: PMC11440249 DOI: 10.3389/fimmu.2024.1460127] [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: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Enterotoxigenic Escherichia coli (ETEC) is the main diarrhea-causing pathogen in children and young animals and has become a global health concern. Berberine is a type of "medicine and food homology" and has a long history of use in China, particularly in treating gastrointestinal disorders and bacterial diarrhea. Methods In this study, we explored the effects of berberine on growth performance, intestinal inflammation, oxidative damage, and intestinal microbiota in a weaned piglet model of ETEC infection. Twenty-four piglets were randomly divided into four groups-a control group (fed a basal diet [BD] and infused with saline), a BD+ETEC group (fed a basal diet and infused with ETEC), a LB+ETEC group (fed a basal diet with 0.05% berberine and infused with ETEC infection), and a HB+ETEC group (fed a basal diet with 0.1% berberine and infused with ETEC). Results Berberine significantly improved the final body weight (BW), average daily gain (ADG), and average daily feed intake (ADFI) (P<0.05) of piglets, and effectively decreased the incidence of diarrhea among the animals (P<0.05). Additionally, berberine significantly downregulated the expression levels of the genes encoding TNF-α, IL-1β, IL-6, IL-8, TLR4, MyD88, NF-κB, IKKα, and IKKβ in the small intestine of piglets (P<0.05). ETEC infection significantly upregulated the expression of genes coding for Nrf2, CAT, SOD1, GPX1, GST, NQO1, HO-1, GCLC, and GCLM in the small intestine of the animals (P<0.05). Berberine significantly upregulated 12 functional COG categories and 7 KEGG signaling pathways. A correlation analysis showed that berberine significantly increased the relative abundance of beneficial bacteria (Gemmiger, Pediococcus, Levilactobacillus, Clostridium, Lactiplantibacillus, Weissella, Enterococcus, Blautia, and Butyricicoccus) and decreased that of pathogenic bacteria (Prevotella, Streptococcus, Parabacteroides, Flavonifractor, Alloprevotella) known to be closely related to intestinal inflammation and oxidative stress in piglets. In conclusion, ETEC infection disrupted the intestinal microbiota in weaned piglets, upregulating the TLR4/MyD88/NF-κB and Nrf2 signaling pathways, and consequently leading to intestinal inflammation and oxidative stress-induced damage. Discussion Our data indicated that berberine can optimize intestinal microbiota balance and modulate the TLR4/MyD88/NF-κB and Nrf2 signaling pathways, thus helping to alleviate intestinal inflammation and oxidative damage caused by ETEC infection in weaned piglets.
Collapse
Affiliation(s)
- Yue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Ziting Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Min Du
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Xunsheng Pang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Aiyou Wen
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| |
Collapse
|