1
|
Zhao J, Gong Z, Bao W, Liu X, Yu Z, Dong YQ, Mao W, Liu B, Zhang S. Matrine alleviates Staphylococcus aureus-induced acute lung injury in mice by inhibiting MLKL and NLRP3-mediated inflammatory activity. Eur J Pharmacol 2025; 993:177385. [PMID: 39956265 DOI: 10.1016/j.ejphar.2025.177385] [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: 10/11/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
Acute lung injury (ALI) is a serious clinical condition with high incidence and mortality. The inflammatory response induced by gram-positive bacteria, especially Staphylococcus aureus (S. aureus), is a key factor contributing to ALI progression and other infectious diseases. Matrine, known for its diverse biological and pharmacological properties, has not been fully explored for its potential to prevent or treat S. aureus-induced ALI. Our study demonstrated that matrine exerts a protective effect against lung injury in mice infected with S. aureus. Specifically, matrine reduced pulmonary edema and decreased neutrophil infiltration in the infected lungs. Furthermore, matrine significantly reduced the expression of high-mobility group box protein 1 (HMGB1) and hyaluronic acid-binding protein 2 (HABP2) in the lungs of infected mice. Additionally, matrine modulated the production of inflammatory mediators, including tumor necrosis factor-α (TNF-α); interleukin-1β (IL-1β); regulated upon activation normal T cell expressed and secreted (RANTES) and Interleukin 10 (IL-10), in both infected lungs and macrophages, suggesting a protective role against tissue damage. Moreover, matrine influenced the secretion of proinflammatory cytokines and regulated the activation of the mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-κB) signaling pathways, along with the activation of Nod-like receptor pyrin domain-containing protein 3 (NLRP3) and Mixed-lineage kinase domain-like protein (MLKL) in macrophages. Notably, when MLKL, but not NLRP3, is deleted, the ability of matrine to regulate damage-associated proteins and prevent tissue injury is diminished. These findings suggest that matrine may be a promising therapeutic agent for the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Jiamin Zhao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhiguo Gong
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Wenhui Bao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Xinyu Liu
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Zhuoya Yu
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Yan Qin Dong
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Wei Mao
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China
| | - Bo Liu
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China.
| | - Shuangyi Zhang
- Key Laboratory of Clinical Diagnosis and Treatment Techniques for Animal Disease, Ministry of Agriculture, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China; Laboratory of Veterinary Clinical Pharmacology, College of Veterinary Medicine, Inner Mongolia Agricultural University, No. 29, Erdosdong Road, Saihan District, 010011, Hohhot, China.
| |
Collapse
|
2
|
Malik A, Huda NU, Tahir SS, Warsi Z, Arif R, Khan MA, Rasheed S. Identification of new 1,2,3-Triazole analogues of sulfanilamide as inhibitors of the carbonic anhydrase II enzyme: Comprehensive in-vitro and in-vivo analyses. Int J Biol Macromol 2025; 303:140426. [PMID: 39894100 DOI: 10.1016/j.ijbiomac.2025.140426] [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: 10/11/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Carbonic anhydrases (CAs) play a vital role in various physiological processes by catalyzing the reversible hydration of CO2 into HCO3-, hence maintaining the fluid and pH balance. Overexpression of carbonic anhydrases II (CA II) is associated with diseases, such as glaucoma, and epilepsy; therefore, it is considered as an important clinical target. Therapeutically used CA inhibitors exhibit several undesirable effects; therefore, there is an urgent need to identify new, safe, and effective inhibitors of the CAs. Keeping in view the importance of CA II inhibition, a library of new 1,3-disubstituted-1,2,3-triazole analogues of sulfanilamide is synthesized via Click chemistry, starting from sulfanilamide azide and different substituted propargyl ethers, incorporating benzyl and heteroarylmethyl moieties. The new derivatives showed significant CA II inhibitory activity (IC50 ranging between 0.19 0.66 μM) when compared with the standard inhibitor, acetazolamide (0.13 ± 0.01 μM). Among all, compounds 16 and 17 showed the most potent activity (IC50 = 0.19 μM) followed by compounds 23, and 18 (IC50 = 0.24 ± 0.014 and 0.26 ± 0.04 μM, respectively). Kinetics studies showed that all compounds are competitive inhibitors of bCA II enzyme (Ki ranging between 0.14-0.68 μM). Additionally, molecular docking studies revealed that all compounds formed network of interactions with the active site residues of the bCA II enzyme. All compounds were found to be non-toxic against BJ Human fibroblast cells. From in-vivo studies, we found that CA activity was significantly inhibited by the intraperitoneal administration of compounds 16 and 17 for up to 5 h. In conclusion, new 1,2,3-triazole analogues of sulfanilamide were identified as good CA II inhibitors.
Collapse
Affiliation(s)
- Aqsa Malik
- Dr. Panjwani Center of Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Noor Ul Huda
- Dr. Panjwani Center of Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Syeda Sarah Tahir
- Dr. Panjwani Center of Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zoha Warsi
- Dr. Panjwani Center of Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rida Arif
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Maria Aqeel Khan
- H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Third World Center for Science and Technology, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Saima Rasheed
- Dr. Panjwani Center of Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
3
|
Xu F, Qiu J, Liu N, Wei H, Gao Y, Fei Y, Xi J, Yu Z, Fan X, Chen L, Xia Y, Dou X. Therapeutic Potential of Raspberry Extract in High-Fat Diet-Induced Liver Injury via Apoptosis and AMPK/PPARα Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40168586 DOI: 10.1021/acs.jafc.4c09593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
This study aimed to explore the efficacy and mechanisms of raspberry (Rubus idaeus L. fruit) aqueous extract (RE) in alleviating high-fat diet (HFD)-induced metabolic-associated fatty liver disease (MAFLD). The MAFLD mouse model was established to examine the effects of RE through liver transcriptome and metabolomics analysis. In this study, RE supplementation significantly alleviated HFD-induced liver injury, hepatosteatosis, inflammation, and insulin resistance. Liver transcriptome analysis demonstrated that RE supplementation favorably regulated signaling pathways involved in fatty acid metabolism and inflammation, including the AMPK signaling pathway, PPAR signaling pathway, apoptosis, etc. Furthermore, the injection of compound C, an antagonist of AMPK, notably reversed the hepatoprotective effects of RE, evidenced by increased lipid profile levels, accelerated fatty acid-related gene disorder, and increased positive tunnel staining area. Furthermore, liver metabolomics analysis demonstrated that RE treatment led to substantial enrichment of the liver tissue metabolite umbelliferone (UMB), which has the potential to ameliorate lipid accumulation and hepatocyte injury through the AMPK signaling pathway. In summary, RE intervention mitigated HFD-induced liver dysfunction in mice, with UMB likely being the primary component responsible for its therapeutic efficacy in the liver. In addition, this study provided new insights, suggesting that RE could be used as a promising therapeutic approach for modulating MAFLD via apoptosis and the AMPK/PPARα signaling pathway.
Collapse
Affiliation(s)
- Fangying Xu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Nian Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Huaxin Wei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yanyan Gao
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yang Fei
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Jiale Xi
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 852, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Lin Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Yongliang Xia
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, China
| |
Collapse
|
4
|
Huang X, Li Q, Yan S, Wang C, Ren X, Wang J, Cheng J, Sun Z. The antibacterial efficacy of nitroxoline against multidrug resistant Escherichia coli associated with copper binding. Eur J Pharmacol 2025:177576. [PMID: 40180272 DOI: 10.1016/j.ejphar.2025.177576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
Nitroxoline (NIT) has been approved for the treatment of uncomplicated urinary tract infections (UTIs) for more than half a century, yet its antimicrobial properties remain incompletely understood. Here, we determined the intricate connections between NIT's metal-chelating capabilities and its antibacterial activity. Metal ion binding characteristics were measured by Ultraviolet-visible (UV-vis) spectroscopy. Biochemical assays and molecular docking studies were performed to elucidate the underlying mechanism. We found that NIT could interact with a diverse array of metal ions, including Cu2+, Fe2+, Zn2+ and Mn2+. While, the addition of Cu2+ significantly decreased NIT's antibacterial effect against uropathogenic Escherichia coli (UPEC) strain J96 and multidrug resistant E coli B2, with the minimum inhibitory concentration (MIC) increased from 8 mg/L to 64 mg/L. Mechanically, NIT significantly decreased the intracellular copper ion levels and reduced bacterial transmembrane electrical potential. Furthermore, NIT promoted production of nitric oxide, peroxynitrite (ONOO-), and reactive oxygen species (ROS). However, the interaction of Cu2+ with NIT suppressed the induced generation of ROS but not the generation of ONOO- in E coli, suggesting that the antibacterial activity of NIT arose from multiple functional groups within its molecular structure. Moreover, NIT triggered intracellular acidification concomitant with enhanced glucose uptake, yet paradoxically suppressed ATP generation, suggesting a potential uncoupling between glycolytic flux and oxidative phosphorylation. Finally, the action of NIT was predicted to bind to the CuB-metal redox centers of cytochrome bo(3) ubiquinol oxidase through molecular docking analyses. Collectively, these data illuminate the antibacterial activity of NIT as a potent copper-related metalloantibiotic against UPEC.
Collapse
Affiliation(s)
- Xiaoyong Huang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| | - Qianqian Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Shiqi Yan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Chenli Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Xiaomin Ren
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Jianzhong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Jia Cheng
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China; Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China.
| |
Collapse
|
5
|
Jiang Z, Kan J, Wang D, Lv Y, Kong C, Wu L, Chen Y, Yang M, Gu Y, Chen S. Inhibition of GRK2 reduced doxorubicin-induced oxidative stress and apoptosis through upregulating ADH1. Toxicol Appl Pharmacol 2025; 497:117261. [PMID: 39914624 DOI: 10.1016/j.taap.2025.117261] [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: 12/07/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025]
Abstract
OBJECTIVE Patients undergoing anti-cancer therapy with doxorubicin (DOX) face the risk of cumulative, irreversible cardiotoxicity. In failing hearts, the overexpressed and activated G protein-coupled receptor kinase 2 (GRK2) initiates pathological signaling, leading to cardiomyocyte death. This study aimed to investigate the potential role of GRK2 in DOX-induced cardiotoxicity (DIC). METHODS Mice were administered intraperitoneal injections of DOX (5 mg/kg) weekly for four weeks to induce DIC. Small interfering RNAs (siRNAs) targeting GRK2, ADH1, and PABPC1 were employed in H9c2 cells. Oxidative stress and cell apoptosis were assessed using Reactive Oxygen Species (ROS) staining and TUNEL staining, respectively. Co-immunoprecipitation (Co-IP) was utilized to detect the interaction between GRK2 and PABPC1. RNA immunoprecipitation (RIP) assay was employed to evaluate the binding between PABPC1 and ADH1 mRNA. RESULTS GRK2 was found to be upregulated in DOX-treated mouse hearts and H9c2 cells. Cardiomyocyte-specific GRK2 knockout partially mitigated oxidative stress, apoptosis, and cardiac dysfunction. Additionally, GRK2 knockdown attenuated DOX-induced oxidative damage and apoptosis both in vivo and in H9c2 cells. Furthermore, a reduction in ADH1 expression was observed in DOX-treated hearts and cardiomyocytes, with a pronounced increase following GRK2 knockdown. Notably, the beneficial effects of GRK2 knockdown in H9c2 cells were abolished after ADH1 knockdown. Mechanistically, GRK2 knockdown promoted the binding of PABPC1 to ADH1 mRNA, thereby inhibiting the degradation of ADH1 mRNA. Increased ADH1 expression alleviated DOX-induced oxidative stress and apoptosis in cardiomyocytes. CONCLUSION In conclusion, our study demonstrates that targeting GRK2 may represent a promising therapeutic strategy for mitigating DOX-associated cardiotoxicity.
Collapse
Affiliation(s)
- Zihao Jiang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Junyan Kan
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Dongchen Wang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yifei Lv
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Chaohua Kong
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Lida Wu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yunwei Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Meng Yang
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yue Gu
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| | - ShaoLiang Chen
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| |
Collapse
|
6
|
Bhati R, Zadeng H, Singh E, Kumar A, Jain M, Senthil Kumaran J, Singh AK, Muthukumaran J. Molecular dynamics simulations assisted investigation of phytochemicals as potential lead candidates against anti-apoptotic Bcl-B protein. J Biomol Struct Dyn 2025; 43:3049-3063. [PMID: 38111145 DOI: 10.1080/07391102.2023.2295385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/11/2023] [Indexed: 12/20/2023]
Abstract
Due to the multifarious nature of cancer, finding a single definitive cure for this dreadful disease remains an elusive challenge. The dysregulation of the apoptotic pathway or programmed cell death, governed by the Bcl-2 family of proteins plays a crucial role in cancer development and progression. Bcl-B stands out as a unique anti-apoptotic protein from the Bcl-2 family that selectively binds to Bax which inhibits its pro-apoptotic function. Although several inhibitors are reported for Bcl-2 family proteins, no specific inhibitors are available against the anti-apoptotic Bcl-B protein. This study aims to address this research gap by using virtual screening of an in-house library of phytochemicals from seven anti-cancer medicinal plants to identify lead molecules against Bcl-B protein. Through pharmacokinetic analysis and molecular docking studies, we identified three lead candidates (Enterolactone, Piperine, and Protopine) based on appreciable drug-likeliness, ADME properties, and binding affinity values. The identified molecules also exhibited specific interactions with critical amino acid residues of the binding cleft, highlighting their potential as lead candidates. Finally, molecular dynamics simulations and MM/PBSA based binding free energy analysis revealed that Enterolactone (CID_114739) and Piperine (CID_638024) molecules were on par with Obatoclax (CID_11404337), which is a known inhibitor of the Bcl-2 family proteins.
Collapse
Affiliation(s)
- Rittik Bhati
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Hazel Zadeng
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Ekampreet Singh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Ankit Kumar
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Monika Jain
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - J Senthil Kumaran
- Department of Chemistry, DLR Arts and Science College, Arcot, India
- Department of Science and Humanities, Er. Perumal Manimekalai College of Engineering, Hosur, India
| | - Amit Kumar Singh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| |
Collapse
|
7
|
Yasmeen N, Ahmad Chaudhary A, K Niraj RR, Lakhawat SS, Sharma PK, Kumar V. Screening of phytochemicals from Clerodendrum inerme (L.) Gaertn as potential anti-breast cancer compounds targeting EGFR: an in-silico approach. J Biomol Struct Dyn 2025; 43:2781-2823. [PMID: 38141177 DOI: 10.1080/07391102.2023.2294379] [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: 07/25/2023] [Accepted: 12/04/2023] [Indexed: 12/25/2023]
Abstract
Breast cancer (BC) is the most prevalent malignancy among women around the world. The epidermal growth factor receptor (EGFR) is a tyrosine kinase receptor (RTK) of the ErbB/HER family. It is essential for triggering the cellular signaling cascades that control cell growth and survival. However, perturbations in EGFR signaling lead to cancer development and progression. Hence, EGFR is regarded as a prominent therapeutic target for breast cancer. Therefore, in the current investigation, EGFR was targeted with phytochemicals from Clerodendrum inerme (L.) Gaertn (C. inerme). A total of 121 phytochemicals identified by gas chromatography-mass spectrometry (GC-MS) analysis were screened against EGFR through molecular docking, ADMET analyses (Absorption, Distribution, Metabolism, Excretion, and Toxicity), PASS predictions, and molecular dynamics simulation, which revealed three potential hit compounds with CIDs 10586 [i.e. alpha-bisabolol (-6.4 kcal/mol)], 550281 [i.e. 2,(4,4-Trimethyl-3-hydroxymethyl-5a-(3-methyl-but-2-enyl)-cyclohexene) (-6.5 kcal/mol)], and 161271 [i.e. salvigenin (-7.4 kcal/mol)]. The FDA-approved drug gefitinib was used to compare the inhibitory effects of the phytochemicals. The top selected compounds exhibited good ADMET properties and obeyed Lipinski's rule of five (ROF). The molecular docking analysis showed that salvigenin was the best among the three compounds and formed bonds with the key residue Met 793. Furthermore, the molecular mechanics generalized born surface area (MMGBSA) calculations, molecular dynamics simulation, and normal mode analysis validated the binding affinity of the compounds and also revealed the strong stability and compactness of phytochemicals at the docked site. Additionally, DFT and DOS analyses were done to study the reactivity of the compounds and to further validate the selected phytochemicals. These results suggest that the identified phytochemicals possess high inhibitory potential against the target EGFR and can treat breast cancer. However, further in vitro and in vivo investigations are warranted towards the development of these constituents into novel anti-cancer drugs.
Collapse
Affiliation(s)
- Nusrath Yasmeen
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | | | | | | | - Vikram Kumar
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
- Amity Institute of Pharmacy, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
8
|
Akash S, Mahmood S, Ahamed R, Bayıl I, Dev Bairagi R, Islam MR, Hosen ME, de Lima Menezes G, S Almaary K, Nafidi HA, Bourhia M, Ouahmane L. Novel computational and drug design strategies for the inhibition of human T-cell leukemia virus 1-associated lymphoma by Astilbin derivatives. J Biomol Struct Dyn 2025; 43:2746-2761. [PMID: 38131136 DOI: 10.1080/07391102.2023.2294376] [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: 09/13/2023] [Accepted: 12/01/2023] [Indexed: 12/23/2023]
Abstract
Human T-cell leukemia virus 1 (HTLV-1) associated lymphoma is a devastating malignancy triggered by HTLV-1 infections. We employeda comprehensive drug design and computational strategy in this work to explore the inhibitory activitiesof Astilbin derivatives against HTLV-1-associated lymphoma. We evaluated the stability, binding affinities, and various computational analysis of Astilbin derivatives against target proteins, such as HTLV-1 main protease and HTLV-1 capsid protein. The root mean square deviation (RMSD), root mean square fluctuation, radius of gyration, hydrogen bond analysis, principal component analysis (PCA) and dynamic cross-correlation matrix (DCCM) were applied to characterize these protein-ligand interactions further. Ligand-03 and ligand-04 exhibited notable binding affinity to HTLV-1 capsid protein, while ligand-05 displayed high binding affinity to HTLV-1 protease. MD simulation analysis revealed that ligand-03, bound to HTLV-1 capsid protein, demonstrated enhanced stability with lower RMSD values and fewer conformational changes, suggesting a promising binding orientation. Ligand-04, despite stable binding, exhibited increased structural deviations, making it less suitable. Ligand-05 demonstrated stable binding to HTLV-1 protease throughout the simulation period at 100 nanoseconds. Hydrogen bond analysis indicated that ligand-05 formed persistent hydrogen bonds with significantresidues, contributing to its stability. PCA highlighted ligand-03's more remarkable conformational changes, while DCCM showed ligand-05's distinct dynamics, indicating its different behavior in the complex. Furthermore, binding free energy calculations supported the favorable interactions of ligand-03 and ligand-04 with HTLV-1 capsid protein, while ligand-05 showed weaker interactions with HTLV-1 protease. Molecular electrostatic potential and frontier molecular orbital analyses provided insights into these compounds' charge distribution and stability. In conclusion, this research found Astilbin derivatives as potential inhibitors of HTLV-1-associated lymphoma. Future attempts at drug development will benefit from the steady interaction landscape provided by Ligand-03, Ligand-04 and Ligand-05, which showed the most attractive binding profile with the target protein. These results open up new opportunities for innovative drug development, and more experimental testing should be done between Astilbin derivatives and HTLV-1-associated lymphoma.
Collapse
Affiliation(s)
- Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International, University, Dhaka, Bangladesh
| | - Sajjat Mahmood
- Department of Microbiology, Jagannath University, Dhaka, Bangladesh
| | - Rashel Ahamed
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Imren Bayıl
- Department of Bioinformatics and Computational Biology, Gaziantep University, Turkey
| | - Rahul Dev Bairagi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, Bangladesh
| | - Md Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International, University, Dhaka, Bangladesh
| | - Md Eram Hosen
- Department of Genetic Engineering and Biotechnology, University of Rajshahi, Rajshahi, Bangladesh
| | - Gabriela de Lima Menezes
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Khalid S Almaary
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hiba-Allah Nafidi
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, Quebec City, QC, Canada
| | - Mohammed Bourhia
- Department of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Ibn Zohr University, Laayoune, Morocco
| | - Lahcen Ouahmane
- Laboratory of Microbial Biotechnologies, Agrosciences and Environment (BioMAgE), Labeled Research Unit-CNRSTN°4, Cadi Ayyad University, Marrakesh, Morocco
| |
Collapse
|
9
|
Liu J, Li Z, Liu W, Jiang Z, Zhang X, Yuan Y, Shen Y. Quercetin down-regulates MCP-1 expression in autoimmune myocarditis via ERK1/2-C/EBPβ pathway: An integrative approach using network pharmacology and experimental models. Int Immunopharmacol 2025; 154:114559. [PMID: 40158430 DOI: 10.1016/j.intimp.2025.114559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/10/2025] [Accepted: 03/23/2025] [Indexed: 04/02/2025]
Abstract
Myocarditis is one of the common causes of sudden death in adolescents, and autoimmune response and inflammation play an essential role in the development of myocarditis. Quercetin is a natural flavonoid compound with anti-inflammatory and cardioprotective effects. However, the mechanism of quercetin in autoimmune myocarditis remains unclear. This study observed that quercetin significantly improved cardiac function, inflammation and fibrosis in mice with experimental autoimmune myocarditis (EAM). In addition, Network pharmacology predicts the key target C/EBPβ and signalling pathway MAPK for quercetin treatment of autoimmune myocarditis. CESTA and DARTS experiments verified that quercetin and C/EBPβ have strong binding ability. It is shown that quercetin down-regulates MCP-1 expression in H9C2 cells by dephosphorylation of ERK1/2 and C/EBPβ. Specifically, quercetin reduced the binding of C/EBPβ to the MCP-1 promoter, resulting in decreased expression of MCP-1, which was associated with decreased ERK1/2 dependent phosphorylation at the C/EBPβ threonine 188 site. This inhibitory effect of quercetin could be further enhanced by the ERK1/2 inhibitor PD98059. The biological relevance of this regulatory network is demonstrated in EAM mice. In conclusion, these results illustrate the protective effect of quercetin against autoimmune myocarditis. A novel regulatory mechanism was revealed, namely the down-regulation of MCP-1 through the ERK1/2-C/EBPβ axis. This provides a new therapeutic strategy for autoimmune myocarditis.
Collapse
Affiliation(s)
- Jinlin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Zhuolun Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Wei Liu
- Department of Liver Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510000, China
| | - Zuli Jiang
- Department of Blood Transfusion, Henan Provincial Children's Hospital, Zhengzhou 450052, China
| | - Xin Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yizhe Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yan Shen
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
10
|
Zheng H, Wang H, Zhang D, Gan Y, Wu Y, Xiang W, Fu P. Identification of therapeutic targets and immune landscape in glioblastoma through crosstalk with glioma-associated mesenchymal stem cells. Int Immunopharmacol 2025; 150:114228. [PMID: 39946771 DOI: 10.1016/j.intimp.2025.114228] [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: 11/16/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Glioma-associated mesenchymal stem cells (GA-MSCs) are one of the key factors limiting the effectiveness of glioblastoma (GBM) treatment and contributing to poor patient prognosis, making them a potential therapeutic target for GBM. In-depth research into the complex crosstalk between GA-MSCs and GBM cells not only aids in understanding the mechanisms of GBM progression but also provides valuable insights for developing new potential drugs. METHODS We conducted a comprehensive bioinformatics analysis aimed at identifying shared dysregulated genes between GBM and GA-MSCs. Through hub gene enrichment and immune infiltration analyses, we explored key molecular pathways and the immune landscape. Additionally, Cox regression analysis was employed to identify key factors influencing overall survival in GBM. The expression patterns and functional roles of hub genes were validated across various cancer types and datasets. Finally, dynamic simulations were used to assess the binding affinity of potential drugs to the targets, further supporting their potential as therapeutic candidates. RESULTS We identified 32 candidate genes primarily involved in the 1-kappa-B kinase/NF-kappa-B and MAPK signaling pathways, both of which played critical roles in tumor survival, proliferation, and invasion. Notable hub genes included DUSP1, FYN, FLNC, FN1, G3BP1, MYO1B, and WLS, each contributing uniquely to GBM progression. Among them, FLNC was highlighted as a key regulatory factor in GBM progression. Molecular dynamics simulations further revealed its potential as a therapeutic target, particularly demonstrating a high binding affinity with staurosporine. Additionally, a high proportion of dendritic cells contributed to the formation of the GBM immune microenvironment. CONCLUSIONS This study revealed the co-expression patterns and metabolic pathways between GA-MSCs and GBM, providing new insights into the molecular mechanisms of GBM progression. Targeting FLNC with staurosporine presents a promising therapeutic strategy for GBM. Aditionally, targeting the shared pathways of both may offer a valuable approach for treating malignant brain tumors.
Collapse
Affiliation(s)
- Haoyang Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Duo Zhang
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 China
| | - Yong Gan
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030 China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| |
Collapse
|
11
|
Zhang Y, Guo X, Sun K, Wang L, Huang S, Gan Y, Qin J, Liu Q, Li Y, Jin Z, Zhu L, Wei X. Exploring the classification and treatment of osteoporosis from the perspectives of natural medicines, molecular targets, and symptom clusters. Sci Rep 2025; 15:10218. [PMID: 40133588 PMCID: PMC11937308 DOI: 10.1038/s41598-025-95304-3] [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: 10/20/2024] [Accepted: 03/20/2025] [Indexed: 03/27/2025] Open
Abstract
Osteoporosis (OP) is a metabolic bone disease characterized by reduced bone density and fragility, impairing quality of life. Traditional treatments often overlook symptoms like back and joint pain, increasing burden. This study aims to map relationships between natural medicines, targets, and symptom clusters, demonstrating their effectiveness in personalized OP treatment to enhance clinical strategies and self-assessment. We used compounds and targets, applying Summary data-based Mendelian Randomisation (SMR) analysis for biological process and molecular function enrichment. Additionally, we employed Phenome-Wide Association Studies (PheWAS) to select two natural drugs-Rhizoma Drynariae (RD) and Lycii Fructus (LF)-for case analysis. The study found that RD primarily improves symptoms such as indigestion, constipation, fatigue, polyuria, and depression, while LF significantly ameliorates symptoms related to the nervous and muscular systems, such as hoarseness, dizziness, vertigo, and fever symptoms. This analysis successfully differentiated two groups of symptoms and precisely constructed a logical chain among "natural Medicines-molecular tArGets-Illness-symptom Clusters" (MAGIC chain) achieving a refined classification of OP. The results of this study support the effectiveness of implementing personalized medical strategies in the treatment of OP, providing a scientific basis for the clinical application of natural medicines and patient self-management.
Collapse
Affiliation(s)
- Yili Zhang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Xiangyun Guo
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Kai Sun
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Liang Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Siyuan Huang
- Department of Biochemistry, University of Oxford, South Parks Rd, Oxford, OX1 3QU, UK
| | - Yiwen Gan
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jinran Qin
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Qingqing Liu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Yan Li
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Zikai Jin
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Liguo Zhu
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China
| | - Xu Wei
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China.
| |
Collapse
|
12
|
Hu Y, Lu S, Xue C, Hu Z, Wang Y, Zhang W, Wang D, Wang J, Ding G, Yu J, Hu Y, Liu Y. Exploring the protective effect of metformin against sarcopenia: insights from cohort studies and genetics. J Transl Med 2025; 23:356. [PMID: 40119457 PMCID: PMC11927167 DOI: 10.1186/s12967-025-06357-x] [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: 11/12/2024] [Accepted: 03/08/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND The impact of metformin on sarcopenia remains uncertain. This study aimed to investigate whether metformin influences sarcopenia risk and evaluate the effects of potential drug targets on sarcopenia traits. METHODS We analyzed data from the National Health and Nutrition Examination Survey (NHANES) (n = 3549) to assess the association between metformin use and sarcopenia risk in elderly patients with type 2 diabetes. Mendelian randomization (MR) analysis using genome-wide association studies (GWAS) from UK Biobank (n = 1,366,167) and FinnGen (n = 218,007), with expression quantitative trait loci (eQTL) as instrumental variables, examined the causal effect of metformin-related targets on sarcopenia traits, while molecular docking explored the interaction between metformin and its drug targets. RESULTS Metformin use was associated with increased grip strength (OR = 2.46; 95% CI 1.49-2.38) and skeletal muscle mass (OR = 1.24; 95% CI 0.20-2.28), as well as reduced mortality (HR = 0.62; 95% CI 0.54-0.71). MR analysis suggested a possible link between GDF15 gene expression and sarcopenia traits, with no evidence of genetic confounding. Molecular docking indicated stable binding between metformin and GDF15. CONCLUSION This study suggests that metformin may lower sarcopenia risk, particularly in elderly patients with type 2 diabetes, with GDF15 identified as a promising target for sarcopenia treatment.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shan Lu
- Women and Children Department of the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Cheng Xue
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhaonian Hu
- Department of Psychology, School of Social and Behavioral Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yifei Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wensong Zhang
- Department of the Core Facility, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dan Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jizheng Wang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoxian Ding
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing Yu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yifang Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of the Clinical Medical Research, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yining, Xinjiang, China.
| | - Yun Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
13
|
Wu X, Pan X, Kang J, Huang Y, Ren J, Pan J, Yu K, Li Y. Ferulic acid inhibits ox-LDL-induced ferroptosis and apoptosis in RAW 264.7 cells via the HIF-1 signaling pathway. Front Pharmacol 2025; 16:1524736. [PMID: 40170728 PMCID: PMC11958962 DOI: 10.3389/fphar.2025.1524736] [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: 11/19/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
Objective Ferulic acid (FA) has shown potential in treating atherosclerosis (AS) by improving lipid metabolism and exerting anti-hypoxic effects. This study aimed to validate the mechanism of FA in AS through in vitro experiments. Methods Network analysis was employed to predict the mechanisms underlying the therapeutic effects of FA on AS. An in vitro foam cell model was established using RAW 264.7 cells treated with ox-LDL. Cellular lipid accumulation was detected using Oil Red O staining; cell viability was assessed by cell counting kit-8; mitochondrial morphology and function were evaluated by transmission electron microscopy and JC-1 staining; apoptosis levels were detected by TUNEL and DAPI staining; mitochondrial Fe2+ content was measured by Mito-FerroGreen; and Western blot was performed to determine the protein expression levels of HIF-1α, Bax, Bcl2, GPX4, and EGFR. Results Network analysis suggested that FA may exert its therapeutic effects on AS through the HIF-1 signaling pathway and is closely associated with the regulation of ferroptosis and apoptosis. FA upregulated the expression of ALOX5, BCL2, ERN1, GPX4, NOS3, and SLC2A1 mRNA and downregulated the expression of BAX, CYCS, EGFR, FLT1, HIF1A, NFKB1, NOS2, PARP1, and STAT3 mRNA. In vitro experiments demonstrated that FA reduces lipid accumulation, increases cell viability, improves mitochondrial function, and decreases reactive oxygen species content. Additionally, FA inhibited ferroptosis and apoptosis by suppressing the HIF-1 signaling pathway, up-regulating the expression of GPX4 and Bcl2, and down-regulating the expression of HIF-1α and Bax protein. HIF-1 agonists reversed these effects by activating the HIF-1 signaling pathway. Conclusion FA improves mitochondrial function and suppresses ferroptosis and apoptosis by inhibiting the HIF-1 signaling pathway, thereby treating AS.
Collapse
Affiliation(s)
- Xize Wu
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Xue Pan
- College of Traditional Chinese Medicine, Dazhou Vocational College of Chinese Medicine, Dazhou, Sichuan, China
| | - Jian Kang
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuxi Huang
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaqi Ren
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Jiaxiang Pan
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Kaifeng Yu
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yue Li
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
14
|
Yang X, Zhang J, Ma J, Huang J, Wang P, Wang F, Tang X. Integrative investigation on the mechanisms of modified Zuojin pill (SQQT) in ameliorating gastric metaplasia. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119643. [PMID: 40113112 DOI: 10.1016/j.jep.2025.119643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zuojin pill is a well-known traditional Chinese medicine (TCM) for treating gastric disorders. The modified Zuojin pill (SQQT) has been used in the treatment of gastric metaplasia (GM) in China for decades. However, the mechanisms of SQQT treat GM remain unclear. AIM OF THE STUDY Our goals are to evaluate the effect of SQQT on GM and to investigate its potential mechanisms. METHODS An animal model of metaplasia was established to study the mechanism of SQQT. RNA-seq was employed to analyze the pathogenesis of GM. Network pharmacological approaches and molecular docking were used to elucidate the mechanisms of SQQT. Common targets of the SQQT and GM mechanism pathways are defined as the key mechanisms of SQQT's treatment in GM. The key mechanisms were validated through in vivo and in vitro experiments. RESULTS RNA-seq analysis of GM animals and network pharmacology of SQQT indicated that SQQT might treat GM via 20 pathways, including the PPAR pathway. Among the 3 core targets of the PPAR pathway, only PPARG is related to GM progression. Besides, the core components of SQQT have a lower affinity for binding to PPARG. The main mechanism of SQQT ameliorated GM is related to PPARG. In animal experiments, SQQT ameliorated GM through ROS decreasing, mitochondrial damage repairing, and protein marker rectification. In cell experiments, SQQT notably decreased the levels of ferroptosis and metaplasia markers including GPX4, PPARG, MUC6, and ACSL4. CONCLUSION SQQT ameliorated gastric metaplasia by inhibiting the PPARG/ferroptosis pathway.
Collapse
Affiliation(s)
- Xuefei Yang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jiaqi Zhang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jing Ma
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jinke Huang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Ping Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Fengyun Wang
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xudong Tang
- Peking University Traditional Chinese Medicine Clinical Medical School (Xiyuan), Beijing, China; Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
15
|
Jin X, Zhang Y, Qin G, Fang X, Zhang X, Sun J, Zhou M, Tong X, Xue Y, Yang H, Tian W. Elucidating Molecule-Crosstalk of Neutrophil Extracellular Traps Between Cardiovascular Disease and Psoriasis: Insights Into Mendelian Randomization, Single-Cell RNA Analysis, Shared Targets and the Role of Resveratrol. J Inflamm Res 2025; 18:3913-3935. [PMID: 40125083 PMCID: PMC11929043 DOI: 10.2147/jir.s493416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
Background Left ventricular hypertrophy (LVH) associated with hypertension and psoriasis (PSO) are linked by poor prognosis. Neutrophil extracellular traps (NETs) are implicated in both conditions, but the mechanisms remain unclear. Methods We integrated bulk and single-cell RNA sequencing, mendelian randomization, immune microenvironment analysis, and molecular docking to explore the molecular interactions between LVH and PSO, focusing on NET-related pathways. Prediction models were also developed, and gene function was validated through in vivo and in vitro experiments. Results Our findings identified NETs-associated genes, AKT serine/threonine kinase 1 (AKT1), and receptor-interacting protein kinase 1 (RIPK1), regulating inflammation, fibroblast activation, and apoptosis in LVH and PSO. Functional enrichment analysis revealed mitochondrial metabolic disorders and extensive inflammation as the most prominent shared features of LVH and PSO diseases. Additionally, resveratrol exhibited a high binding affinity to the AKT1 and RIPK1 proteins. Functional validation showed that knockdown of AKT1 reduced LVH cell hypertrophy and PSO cell apoptosis. Conclusion This study highlights molecular pathways linking LVH and PSO, suggesting novel targets for treating cardiac involvement in PSO patients with LVH.
Collapse
Affiliation(s)
- Xiaojun Jin
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
- Department of Clinical Medicine, School of Medicine, Ningbo University, Ningbo, Zhejiang, People’s Republic of China
| | - Yun Zhang
- Department of Ophthalmology, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, 510000, People’s Republic of China
| | - Gaofeng Qin
- Department of Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning, 121001, People’s Republic of China
| | - Xinyan Fang
- School of Statistics, East China Normal University, Shanghai, 200062, People’s Republic of China
| | - Xinnan Zhang
- School of Teacher Education, Ningbo University, Ningbo, Zhejiang, 315000, People’s Republic of China
| | - Jialin Sun
- School of Statistics, East China Normal University, Shanghai, 200062, People’s Republic of China
| | - Min Zhou
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Xuecheng Tong
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Yuan Xue
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Hui Yang
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
| | - Weihong Tian
- Department of Clinical Technology Laboratory, Changzhou Clinical College of Xuzhou Medical University, Changzhou Third People’s Hospital, Changzhou, Jiangsu, 213000, People’s Republic of China
| |
Collapse
|
16
|
Pipaliya R, Basaiawmoit B, Sakure AA, Maurya R, Bishnoi M, Kondepudi KK, Tiwary BK, Mankad M, Patil GB, Gawai K, Sarkar P, Hati S. Peptidomics and molecular dynamics on bioactive peptides produced and characterized from the fermented whey of "Panchali" sheep of West India. Food Chem 2025; 468:142466. [PMID: 39689486 DOI: 10.1016/j.foodchem.2024.142466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 12/19/2024]
Abstract
The study assessed the peptide production by using potent Lactiplantibacillus plantarum KGL3A (MG722814) culture to ferment the sheep milk for evaluation of α-glucosidase inhibition, ACE inhibition, α-amylase inhibition, & inhibiting lipase activities. The maximal ACE inhibitory, α-amylase, α-glucosidase, & lipase inhibiting actions were 71.69 %, 71.32 %, 67.14 %, and 64.15 %, respectively, at 37 °C after 48 h. Proteolytic activity was tested at various incubation times & inoculation rates to maximise the conditions for growth and the greatest action (9.38 mg/mL) was reported at 2.5 % rate of inoculation after incubation of 48 h. The anti-diabetic as well as ACE inhibitory properties of less than 3 kDa were maximum in contrast to >3 kDa, <10 kDa, and > 10 kDa cut-off fractions. Further, when LPS stimulation is applied to RAW 267.4 macrophage cells, the overabundance generation of IL-6, IL-1β, NO, and TNF-α is greatly reduced by using KGL3A to ferment sheep milk. 2D gel electrophoresis & SDS-PAGE were utilized in relation to protein purifications. Maximum numbers of sheep milk's fermented protein bands were present, about 10 to 124 kDa by SDS-PAGE, and 38 spots of protein were discovered using 2D gel chromatography. Ultra-filtered fractions water soluble extracts (WSEs) were employed in RP-HPLC to differentiate between various peptide fractions. The peptide sequences produced were matched using the databases of AHTPDB and BIOPEP to match hypertensive peptides & antidiabetic peptides, respectively. Furthermore, the discovered peptide sequences from the fermenting sheep milk was studied due to their penchant for binding against the active locations of human bile salt activated lipase (hBAL); human maltase-glucoamylase (hMGA); human pancreatic alpha-amylase (hPAM); & human angiotensin-converting enzyme (hACE) through molecular docking.
Collapse
Affiliation(s)
- Rinkal Pipaliya
- Department of Dairy Microbiology, SMC College of Dairy Science, Kamdhenu University, Anand 388110, Gujarat, India
| | - Bethsheba Basaiawmoit
- Department of Rural Development and Agricultural Production, North-Eastern Hill University, Chasingre, Meghalaya, India
| | - Amar A Sakure
- Departmentof Agriculture Biotechnology, Anand Agricultural University, Anand 388110, Gujarat, India
| | - Ruchika Maurya
- Regional Center for Biotechnology, Faridabad, Haryana 121001, India; Healthy Gut Research Group, Food & Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute, Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India
| | - Mahendra Bishnoi
- Healthy Gut Research Group, Food & Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute, Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India
| | - Kanthi Kiran Kondepudi
- Healthy Gut Research Group, Food & Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute, Knowledge City, Sector 81, SAS Nagar, Punjab, 140306, India
| | - Bipransh Kumar Tiwary
- Department of Microbiology, North Bengal St. Xavier's College, North Bengal University, West Bengal, India
| | - Maunil Mankad
- Department of Tissue Culture, Anand Agricultural University, Anand 388110, Gujarat, India
| | - G B Patil
- Department of Tissue Culture, Anand Agricultural University, Anand 388110, Gujarat, India
| | - Kunal Gawai
- Department of Dairy Microbiology, College of Dairy Science, Kamdhenu University, Amreli, Gujarat, India
| | - Preetam Sarkar
- Department of Food Process Engineering, National Institute of Technology, Rourkela, India
| | - Subrota Hati
- Department of Dairy Microbiology, SMC College of Dairy Science, Kamdhenu University, Anand 388110, Gujarat, India.
| |
Collapse
|
17
|
Xu QH, Wang YL, Wang C, Jiang SS, Zhang BR, Tian J. Exploring the active ingredients and potential mechanisms of Pingchan granules in Parkinson's disease treatment through network pharmacology and transcriptomics. Sci Rep 2025; 15:7847. [PMID: 40050654 PMCID: PMC11885611 DOI: 10.1038/s41598-025-91344-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 02/19/2025] [Indexed: 03/09/2025] Open
Abstract
Parkinson's disease (PD), the second most prevalent neurodegenerative disorder, poses significant challenges to single-target therapeutic strategies due to its complex etiology. This has driven interest in multi-target approaches, particularly those leveraging natural compounds. Pingchan granules (PCG), a traditional Chinese medicine composed of plant- and animal-derived compounds, have shown efficacy in alleviating PD symptoms. Here, we identify 96 PCG-associated anti-PD targets, enriched in neuronal synaptic signaling and G protein-coupled receptor pathways. Through protein-protein interaction network analysis of anti-PD targets and random forest modeling of substantia nigra transcriptomic data from PD patients, SLC6A3 and SRC emerged as central hub targets, with Mendelian randomization further validating SRC as a potential therapeutic target. Molecular docking and single-cell sequencing reveal that dauricine, PCG's principal active compound, binds strongly to SLC6A3 and SRC, modulating glucose metabolism pathways in dopaminergic neurons. These findings illuminate the molecular basis of PCG's therapeutic effects, offer a foundation for future drug development, and underscore the potential of dauricine as a targeted treatment for PD.
Collapse
Affiliation(s)
- Qiu-Han Xu
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Yi-Ling Wang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Cheng Wang
- Department of Neurosurgey, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Si-Si Jiang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
| | - Bao-Rong Zhang
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| | - Jun Tian
- Department of Neurology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
18
|
Liu R, Dong J, Wang J, Xu Q, Dong Z, Wang L, Bao Y, Wang K, Han X, Shi X, Xiong Y, Lyu Q, Shan Q, Cao G. MCnebula analysis combined with alpha-glucosidase inhibitory screening reveals potential chemical contributors to efficacy enhancement of natural products after processing. Food Res Int 2025; 205:115985. [PMID: 40032476 DOI: 10.1016/j.foodres.2025.115985] [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: 12/09/2024] [Revised: 01/22/2025] [Accepted: 02/08/2025] [Indexed: 03/05/2025]
Abstract
Natural products often show enhanced therapeutic effects after processing, largely due to changes in their chemical profiles. However, identifying the key chemical classes and components responsible for these improvements remains a challenge. In this study, we present a novel workflow for mass spectrometry data analysis, based on our previously developed MCnebula, combined with in vivo and in vitro testing to identify contributors to the anti-diabetic effects of processed natural products. Cornus officinalis (CO), a traditional food and medicinal plant, showed strong α-glucosidase inhibition, particularly its steamed and wine-steamed products, outperforming acarbose in both in vitro and in vivo testing. MCnebula analysis revealed that flavonoids underwent the most significant changes during processing. Further validation of selected flavonoid compounds, such as quercetin and kaempferol, demonstrated their α-glucosidase inhibitory effects to be 207 and 263 times more potent than acarbose, respectively. The combined use of MCnebula analysis and bioactivity validation revealed the key compounds that contribute to the enhanced anti-glucose effects of CO after processing, offering insights into the chemical transformations with bioactive potential as anti-diabetic dietary supplements and agents.
Collapse
Affiliation(s)
- Ruina Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Jie Dong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Jiaping Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Qiongfang Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Zhixiang Dong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Lu Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Yini Bao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Kuilong Wang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Xin Han
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Xingyang Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Yu Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China
| | - Qiang Lyu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| | - Qiyuan Shan
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| | - Gang Cao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 311400, China.
| |
Collapse
|
19
|
Ghosh P, Singh R, Chatterjee C, Kumar A, Singh SK. Computational screening of coumarin derivatives as inhibitors of the NACHT domain of NLRP3 inflammasome for the treatment of Alzheimer's disease. J Biomol Struct Dyn 2025; 43:2187-2203. [PMID: 38116751 DOI: 10.1080/07391102.2023.2294173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR), leucine-rich-repeat (LRR), and pyrin domain containing 3 (NLRP3) is one of the key players in neuroinflammation, which is a major pathological hallmark of Alzheimer's Disease (AD). Activated NLRP3 causes release of pro-inflammatory molecules that aggravate neurodegeneration. Thus, pharmacologically inhibiting the NLRP3 inflammasome has the potential to alleviate the inflammatory injury to the neurons. Coumarin is a multifunctional nucleus with potent anti-inflammatory properties and can be utilized to develop novel drugs for the treatment and management of AD. In the present study, we have explored the NLRP3-inhibitory activities of a library of coumarin derivatives through a computational drug discovery approach. Drug-like, PAINS free, and potentially BBB permeable compounds were screened out and subjected to molecular docking and in silico ADMET studies, resulting in three virtual hits, i.e. MolPort-050-872-358, MolPort-050-884-068, and MolPort-051-135-630. The hits exhibited better NLRP3-binding affinity than MCC950, a selective inhibitor of NLRP3. Further, molecular dynamics (MD) simulations, post-MD simulation analyses, and binding free energy calculations of the hits established their potential as promising virtual leads with a common coumarin scaffold for the inhibition of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Powsali Ghosh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ravi Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Chayanika Chatterjee
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Ashok Kumar
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| | - Sushil Kumar Singh
- Pharmaceutical Chemistry Research Laboratory 1, Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, India
| |
Collapse
|
20
|
Hjouji K, Barnossi AE, Er-Rajy M, Atemni I, Grenha A, Yagoubi M, Ainane T, Taleb M, Rais Z. Inhibitory potency of active metabolites from different polarities of Datura Stramonium seed extracts: GC-MS analysis, biological evaluations, and molecular docking studies. J Mycol Med 2025; 35:101521. [PMID: 39705886 DOI: 10.1016/j.mycmed.2024.101521] [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: 03/28/2024] [Revised: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024]
Abstract
Datura Stramonium is a well-known and important medicinal plant that is widely used in various medical systems to treat conditions such as asthma, diabetes, and inflammatory diseases. The aim of this study was to prepare extracts of D. stramonium seeds in different solvent polarities for assessing phytochemical potential, in vitro biological activities, and molecular docking studies. Phytochemical screening was conducted to determine the phytochemical composition, while GC-MS analysis was used to identify secondary metabolites of D. Stramonium. The seed extracts were molecularly docked to evaluate bioactive and antioxidant activity. The minimum inhibitory concentration (MIC) of the extracts against bacteria such as S. aureus, E. coli, Bacillus subtilis, and Proteus mirabilis was determined. Antifungal activity was also tested against fungi including Aspergillus flavus, Fusarium oxysporum, Aspergillus niger, and Candida albicans. The results of phytochemical screening indicated that the primary plant constituents in all extracts of different polarities are alkaloids, flavonoids, tannins, mucilage, sterols, heterosides, triterpenoids, and cardiac glycosides. Most molecules in the hexane, ether, and chloroform extracts consist of fatty acids, sterols, glycosides, triterpenoids, alkaloids, and phenolic compounds, as revealed by GC-MS analysis. Conversely, extracts from polar solvents like methanol, ethanol, and water are abundant in alkaloids. In vitro antibacterial and antifungal activities demonstrated that ether, methanol, and ethanol extracts were more effective than inhibiting the four bacterial strains compared to the antibiotics Oxacillin and Cefuroxime. The ether and methanol extracts exhibited better zones of inhibition and significant MIC values against A. niger and C. albicans compared to the control and fluconazole. The free radical inhibition (DPPH) (24.92 ± 5.31 μg/mL) for the ethanol extract indicates Datura's antioxidant capacity. It is believed that the main phytochemicals are responsible for the enhanced antioxidant activity observed in other studies. The docking study revealed that the bioactive compounds linoleic acid and atropine formed better hydrogen bonding interactions with proteins than pi-alkyl and alkyl bonds.
Collapse
Affiliation(s)
- K Hjouji
- Laboratory of Engineering, Electrochemistry, Modeling and Environment Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco.
| | - A El Barnossi
- Laboratory of Biological Engineering, Faculty of Sciences and Techniques, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - M Er-Rajy
- Laboratory of Systems Engineering, Modeling and Analysis Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - I Atemni
- Laboratory of Engineering, Electrochemistry, Modeling and Environment Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - A Grenha
- Centre for Marine Sciences (CCMAR), Faculty of Science and Technology, University of Algarve, 8005-139, Faro, Portugal
| | - M Yagoubi
- Laboratory of Microbiology, Clinical Medical Biology Department, Faculty of Medicine and Pharmacy of Rabat
| | - T Ainane
- Ecole Supérieure de Technologie, Université Sultan Moulay Slimane, PB 170, Khenifra, 54000, Morocco
| | - M Taleb
- Laboratory of Engineering, Electrochemistry, Modeling and Environment Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Z Rais
- Laboratory of Engineering, Electrochemistry, Modeling and Environment Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
21
|
Dhillon D, Jain M, Singh AK, Muthukumaran J. Withania somnifera-derived phytochemicals as Bcl-B inhibitors in cancer therapy: A computational approach from byte to bench to bedside. Biochem Biophys Res Commun 2025; 750:151383. [PMID: 39884007 DOI: 10.1016/j.bbrc.2025.151383] [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: 11/10/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Cancer is the second foremost cause of fatalities associated with non-communicable diseases across the globe, affecting multiple organs and often necessitating costly treatments with adverse side effects. Apoptosis, the body's natural cell death process, plays a crucial role in the prevention of cancer, but it's often disrupted in cancer cells, allowing uncontrolled proliferation. Restoring apoptosis in cancer cells is one of the promising therapeutic strategies to curb tumor growth and enhance clinical outcomes. Bcl-B, an anti-apoptotic protein within the Bcl-2 family, interacts with Bax to mitigate apoptosis, indicating it as a druggable target for cancer therapy. There's a critical need for natural, cost-effective alternatives with minimal adverse effects to reduce morbidity rates of cancer patients. Plant-based immunoprotective medications, particularly from sustainable sources like known medicinal plants, offer substantial potential for cancer treatment. This study involves comprehensive in silico approaches (byte) to evaluate the inhibition potential of the phytochemicals derived from Withania somnifera against the anti-apoptotic Bcl-B protein. Research into Bcl-B's binding affinity with 80 phytochemicals from this plant aims to identify interaction sites for promising anticancer agents. This study's focus on Bcl-B protein highlights its potential in modulating apoptotic pathways and exploring novel anti-cancer therapeutics. Through comprehensive screening based on drug-likeness and pharmacokinetic properties, combined with in-house virtual screening, molecular docking, molecular dynamics simulations, and MM/PBSA-based binding free energy analysis, three promising candidate inhibitors-Withanolide L (IMPHY009438), Withanolide M (IMPHY003143), and Withanolide A (IMPHY000090)-were identified and prioritized. These candidates showed favorable estimated binding free energy values, along with desirable drug-likeness and pharmacokinetic profiles. The results demonstrated that the selected and prioritized phytochemicals, Withanolide L, Withanolide M, and Withanolide A display comparable efficacy to Obatoclax (CID: 11404337) and other known synthetic, semi-synthetic, and natural inhibitors of Bcl-2 family proteins. These findings establish a strong bench foundation for further experimental validation and bedside application, potentially offering an alternative natural approach to cancer therapy.
Collapse
Affiliation(s)
- Deepika Dhillon
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Monika Jain
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Amit Kumar Singh
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Jayaraman Muthukumaran
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, India.
| |
Collapse
|
22
|
Zhang J, Hu D, Fang P, Qi M, Sun G. Deciphering key roles of B cells in prognostication and tailored therapeutic strategies for lung adenocarcinoma: a multi-omics and machine learning approach towards predictive, preventive, and personalized treatment strategies. EPMA J 2025; 16:127-163. [PMID: 39991096 PMCID: PMC11842682 DOI: 10.1007/s13167-024-00390-4] [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: 08/28/2024] [Accepted: 11/24/2024] [Indexed: 02/25/2025]
Abstract
Background Lung adenocarcinoma (LUAD) remains a significant global health challenge, with an urgent need for innovative predictive, preventive, and personalized medicine (PPPM) strategies to improve patient outcomes. This study leveraged multi-omics and machine learning approaches to uncover the prognostic roles of B cells in LUAD, thereby reinforcing the PPPM approach. Methods We integrated multi-omics data, including bulk RNA, ATAC-seq, single-cell RNA, and spatial transcriptomics sequencing, to characterize the B cell landscape in LUAD within the PPPM framework. Subsequently, we developed an integrative machine learning program that generated the Scissor+ related B cell score (SRBS). This score was validated in the training and validation sets, and its prognostic value was assessed along with clinical features to develop predictive nomograms. This study further assessed the role of SRBS and SRBS genes in response to immunotherapy and identified personalized drug targets for distinct risk subgroups, with gene expression verified experimentally to ensure tailored medical interventions. Results Our analysis identified 79 Scissor+ B cell genes linked to LUAD prognosis, supporting the predictive aspect of PPPM. The SRBS model, which utilizes multiple machine learning algorithms, performed excellently in predicting prognosis and clinical transformation, embodying the preventive and personalized aspects of PPPM. Multifactorial analysis confirmed that SRBS was an independent prognostic factor. We observed varying biological functions and immune cell infiltration in the tumor immune microenvironment (TIME) between the high- and low-SRBS groups, underscoring personalized treatment approaches. Notably, patients with elevated SRBS may exhibit resistance to immunotherapy but show increased sensitivity to chemotherapy and targeted therapies. Additionally, we found that LDHA, as an SRBS gene with significant clinical implications, may regulate the sensitivity of LUAD cells to cisplatin. Conclusion This study presents a B cell-associated gene signature that serves as a prognostic marker to facilitate personalized treatment for patients with LUAD, adhering to the principles of PPPM. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00390-4.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Dingtao Hu
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Pu Fang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Min Qi
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| | - Gengyun Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022 Anhui Province China
| |
Collapse
|
23
|
Pitaloka DAE, Arfan A, Khairunnisa SF, Megantara S. In silico identification of a phosphate marine steroid from Indonesian marine compounds as a potential inhibitor of phosphatidylinositol mannosyltransferase (PimA) in Mycobacterium tuberculosis. Comput Biol Med 2025; 186:109677. [PMID: 39842238 DOI: 10.1016/j.compbiomed.2025.109677] [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: 10/11/2024] [Revised: 01/04/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025]
Abstract
A higher death rate is associated with multiple factors, including medication resistance and co-infection with the human immunodeficiency virus (HIV). This shows the need to obtain new and effective drug candidates in improving tuberculosis (TB) treatment. In addition, the phosphatidylinositol mannosyltransferase (PimA) enzyme starts the production of phosphatidyl-myo-inositol. PimA has been identified as a key enzyme and an important area for further research in the development of anti-TB drugs. Previous research investigated various applications including marine resources driven by a deeper understanding of the distinctive features of the ecosystem and the diverse array of organisms. Therefore, this research aims to investigate the potential of Indonesian marine compounds as inhibitors of PimA, with a focus on binding energy, interaction modes, and stability using docking and molecular dynamics (MD) investigation methodologies. The results show that a total of 84 Indonesian marine compounds are effectively docked to the PimA to obtain compounds 21, 27, and 33 for further investigation. Based on the MD analysis, compound 27 (desulfohaplosamate) is the most promising candidate among the new MTB-PimA inhibitors. Compounds bind to PimA, as shown by a strong affinity of -30.09 kJ/mol, and form hydrogen bonds with the key amino acid residue Gly16. Furthermore, a stable complex is formed to easily analyze the antibacterial agents targeting MTB in the future.
Collapse
Affiliation(s)
- Dian Ayu Eka Pitaloka
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia; Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, 45363, Indonesia.
| | - Arfan Arfan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Universitas Halu Oleo, Kendari, Indonesia
| | - Shafa Fitri Khairunnisa
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Sumedang, 45363, Indonesia; Pharmacist Professional Education, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Sandra Megantara
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia; Drug Development Study Centre, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| |
Collapse
|
24
|
Xing H, Li R, Huang Z, Gao Z, Mao Q, Shen Y, Huang G, Chu G, Wang Y. Engineered Cell Membrane-Coated Keratin Nanoparticles Attenuated Intervertebral Disc Degeneration by Remodeling the Disc Microenvironment. Adv Healthc Mater 2025; 14:e2404173. [PMID: 39876590 DOI: 10.1002/adhm.202404173] [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: 10/24/2024] [Revised: 12/24/2024] [Indexed: 01/30/2025]
Abstract
Characterized by a cascade of profound changes in nucleus pulposus (NP) cells, extracellular matrix (ECM), and biomechanics, intervertebral disc degeneration is a common multifactorial condition that may lead to various degenerative lumbar disorders. Therapeutic strategies targeting a single factor have shown limited efficacy in treating disc degeneration, and approaches that address multiple pathological ingredients are barely reported. In this study, engineered cell membrane-encapsulated keratin nanoparticles are developed to simultaneously alleviate NP cell senescence and promote ECM remodeling. To achieve this, salivary acid glycoengineered adipose mesenchymal stem cell membranes are used to coat keratin, a core protein for structural support and cellular protection. The synthesized cell membrane-coated keratin nanoparticles (MKNs) effectively protected mitochondrial integrity in NP cells from oxidative stress-induced damage. Moreover, MKNs modulate mitochondrial metabolism and attenuate NP cell senescence. In addition, MKNs activate integrins at the cell membrane and enhance the interactions between NP cells and ECM, resulting in increased ECM anabolism and decreased catabolism. The proposed multi-targeted strategy to block the degenerative cycle inside the disc is efficacious for treating disc degeneration and may have the potential for clinical application.
Collapse
Affiliation(s)
- Hongyuan Xing
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Run Li
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zizhan Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhongyang Gao
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qijiang Mao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Yifan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Guanrui Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Guangyu Chu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yue Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
25
|
Liu H, Huang C, Liu Z, Li Y, Zhu Y, Gao M, Chen J, Zhang H, Xiao Z, Zhao W. Systematic drug screening and target analysis identify digitoxin as a potential therapy for uveal melanoma. Br J Pharmacol 2025; 182:1275-1291. [PMID: 39617007 DOI: 10.1111/bph.17405] [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: 07/06/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE Cardiac glycosides (CGs), traditionally prescribed for heart failure and arrhythmias, show anticancer potential. However, their mechanisms for preferential inhibition of tumour tissue and constituent malignant cells are not fully elucidated. This study aims to elucidate the therapeutic benefits of CGs in targeting specific tumours and dissect their multi-targeting mechanisms that confer their cytotoxicity against malignant cells. EXPERIMENTAL APPROACH We designed an integrated workflow to identify therapeutic CGs with high toxicity to certain cancers, investigating their multi-target effects, assessing their toxicity to malignant cells and analysing the prognostic relevance of CGs' target genes. The computational findings were confirmed through gene knockdown, cell viability assays, reactive oxygen species (ROS) measurements and so forth. KEY RESULTS CGs modulate multiple genes crucial for ion homeostasis, oxidative stress and apoptosis, with a particularly strong inhibitory effects on uveal melanoma (UVM). Notably, digitoxin suppresses UVM cell proliferation and induces ROS levels by simultaneously targeting STAT3 and KLF5. Single-cell transcriptomic analysis revealed that malignant cells are likely more vulnerable to CGs due to their higher expression of CG target genes compared with surrounding cells in the UVM microenvironment. CONCLUSIONS AND IMPLICATIONS Given UVM's limited options, our study highlights the potential of digitoxin as a promising novel therapeutic agent for this aggressive and rare ocular cancer. Our comprehensive approach is effective in identifying the potent, cancer-specific therapeutic agents from herbal plants.
Collapse
Affiliation(s)
- Huilin Liu
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Chao Huang
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Zhenni Liu
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yuhan Li
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Yanan Zhu
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Min Gao
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Jing Chen
- Department of Obstetrics, Xi'an New Chang'an Maternity Hospital, Xi'an, Shaanxi, China
- Shaanxi Stem Cell Engineering Application Research Center, Shaanxi Jiuzhou Biomedical Science and Technology Group, Xi'an, Shaanxi, China
| | - Hui Zhang
- College of Life Sciences, Northwest Normal University, Lanzhou, Gansu, China
| | - Zhengtao Xiao
- Institute of Molecular and Translational Medicine (IMTM), Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education, Xi'an, Shaanxi, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
26
|
Abd Ali AAkool W, Kashanian S, Hadidi S. Molecular interaction of antiviral drug penciclovir with DNA and HSA insights from experimental and docking studies. J Biomol Struct Dyn 2025; 43:2585-2595. [PMID: 38263739 DOI: 10.1080/07391102.2024.2303382] [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: 07/31/2023] [Accepted: 10/11/2023] [Indexed: 01/25/2024]
Abstract
One approach to accelerate the availability of new cancer drugs is to test drugs approved for other conditions as anticancer agents. During recent decades, penciclovir (PNV) has been frequently utilized as a potent antiviral drug, in particular against infections caused by herpes viruses. Many antivirals interact with DNA and change their expression level, so determining the binding mode is of great importance. In our laboratory, we have focused our attention to design improved drugs that target cellular DNA, to understand the mechanism of action at the molecular level, and also to investigate the effect of antiviral drugs as anticancer agents. The results of ct-DNA-PNV interactions at physiological pH using fluorescence spectroscopy, UV-visible absorption spectroscopy, and molecular modeling reveal this drug binds well to ct-DNA through groove binding. The circular dichroism measurements displayed that PNV caused a slight change in the DNA structure which affirmed that the binding of PNV with the DNA occurs through the groove mode. Besides, multi-spectroscopic and molecular docking were used to evaluate how PNV interacts with human serum albumin under physiological conditions. The findings of fluorescence quenching suggested that static quenching was involved in the spontaneous development of HSA-PNV complex through hydrophobic force. The docking simulation results validated the findings of spectroscopic techniques.
Collapse
Affiliation(s)
| | | | - Saba Hadidi
- Inorganic Chemistry Department, Faculty of Chemistry, Razi University, Kermanshah, Iran
| |
Collapse
|
27
|
Faruque M, Siraj MA, Zilani MNH, Das AK, Anisuzzman M, Islam MM. Investigating small molecules in propolis as Nipah virus glycoprotein (NiV-G) inhibitors through molecular interaction studies. Heliyon 2025; 11:e42595. [PMID: 40051842 PMCID: PMC11883394 DOI: 10.1016/j.heliyon.2025.e42595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/08/2025] [Accepted: 02/09/2025] [Indexed: 03/09/2025] Open
Abstract
Despite the significant fatality rates associated with Nipah virus (NiV) outbreaks in South Asia, including Bangladesh, and India, till today, there is no approved medications to treat it. In this context, small molecules in propolis were computationally screened through pharmacokinetic and toxicity studies followed by molecular docking and dynamics simulation with Nipah virus glycoprotein (NiV-G protein) to assess their anti-Nipah potential. A thorough literature analysis was performed to identify antiviral compounds in propolis from a pool of 84 experimental articles. Following ADMET analysis, 27 molecules out of 34 were docked against NiV-G and compared with a control ligand, ribavirin, which is an investigational drug against Nipah. The molecular docking revealed that bauer-7-en-3β-yl acetate (BA) and moronic acid (MA) bound more strongly to the active site of NiV-G than ribavirin and other ligands. Investigation of root-mean-square deviation (RMSD), root mean square fluctuations (RMSF), radius of gyration (Rg), solvent accessible surface area (SASA), molecular surface area (MolSA), binding free energy (MM-PBSA), the complexity of hydrogen bonds (HBs), and secondary structure of ligand-target interactions for 100 ns by molecular dynamics (MD) simulation study further supported the docked complex's stability and compactness. Therefore, the in silico molecular interaction analysis reports that both molecules may be the possible candidates against Nipah infection.
Collapse
Affiliation(s)
- Muaz Faruque
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Md Afjalus Siraj
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
- Department of Pharmacy, Faculty of Health Sciences, Gono Bishwabidyalay, Dhaka, 1344, Bangladesh
| | - Md Nazmul Hasan Zilani
- Department of Pharmacy, Jashore University of Science & Technology, Jashore, 7408, Bangladesh
| | - Asish Kumar Das
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Md Anisuzzman
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| | - Md Monirul Islam
- Pharmacy Discipline, Life Science School, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
28
|
Zhu L, Zhu Q, Chen Z, Tao Y, Hu J, Wang D, Lin Y, Yang H, Gao C, Zhang W. Estrogen mitigates ischemia-reperfusion injury by inhibiting cardiomyocyte ferroptosis through the downregulation of PHLDA3 expression. Free Radic Biol Med 2025; 232:1-14. [PMID: 39961475 DOI: 10.1016/j.freeradbiomed.2025.01.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/08/2025]
Abstract
Ferroptosis represents a significant target for mitigating myocardial ischemia-reperfusion (I/R) injury. Existing literature indicates that estrogen (17β-estradiol, E2) can alleviate such injuries through various pathways. However, the specific mechanisms by which E2 may confer protection against myocardial I/R injury through the inhibition of ferroptosis remain to be fully elucidated. This study employed a mouse model of left anterior descending coronary artery ligation to investigate the protective effects of E2 on myocardial I/R injury, with a particular focus on its inhibitory effects on ferroptosis and PHLDA3 in both hypoxia-reoxygenation (H/R) and I/R models. A bioinformatics analysis was conducted to evaluate the impact of estrogen receptor GPER knockout on PHLDA3 expression and ferroptosis. Loss-of-function approaches were employed to elucidate the role of PHLDA3 in ferroptosis during myocardial I/R injury. Our findings demonstrate that E2 can ameliorate myocardial I/R injury, primarily by inhibiting ferroptosis. Notably, PHLDA3 expression levels were significantly elevated during ischemia-reperfusion events; however, E2 was observed to suppress this expression. Bioinformatics analysis indicated that PHLDA3 levels increased following GPER knockdown, and the inhibitory effect of E2 on PHLDA3 expression could be partially reversed by GPER inhibitors (G15) in animal models. Furthermore, the suppression of PHLDA3 reduced ferroptosis and mitigated the severity of myocardial I/R injury. Utilizing mass spectrometry and co-immunoprecipitation methodologies, we have elucidated a potential mechanism in which PHLDA3 directly binds to and interacts with proteins involved in the process of ferroptosis. Our findings demonstrate that E2 effectively suppresses ferroptosis and mitigates myocardial I/R injury by downregulating PHLDA3 expression through the activation of the GPER receptor.
Collapse
Affiliation(s)
- Lijie Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China; Department of Cardiology of Fuwai Central China Ccardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory for Coronary Heart Disease Prevention and Control, Zhengzhou, Henan, China
| | - Qiongjun Zhu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Zhezhe Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yecheng Tao
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jiayi Hu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Dan'an Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yutong Lin
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Honghui Yang
- Department of Cardiology of Fuwai Central China Ccardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory for Coronary Heart Disease Prevention and Control, Zhengzhou, Henan, China
| | - Chuanyu Gao
- Department of Cardiology of Fuwai Central China Ccardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory for Coronary Heart Disease Prevention and Control, Zhengzhou, Henan, China
| | - Wenbin Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, No 3 East of Qingchun Road, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
29
|
Ishikawa T, Matsumoto K, Hamada T, Koze H, Baba M, Okamoto M, Sudoh M. In Silico Discovery of SARS-CoV-2 Main Protease Inhibitors Using Docking, Molecular Dynamics, and Fragment Molecular Orbital Calculations. J Phys Chem B 2025; 129:1740-1749. [PMID: 39886917 DOI: 10.1021/acs.jpcb.4c07920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The 3C-like protease of severe acute respiratory syndrome coronavirus 2, known as the main protease (Mpro), is an attractive drug target for the treatment of coronavirus disease 2019. This study reports the discovery of novel Mpro inhibitors using several in silico techniques, including docking, molecular dynamics (MD), and fragment molecular orbital (FMO) calculations. We performed docking calculations on 5950 compounds with bioactivity, and 12 compounds were selected. An enzymatic assay was conducted, revealing that BP-1-102 exhibits significant Mpro inhibitory activity with an IC50 of 11.1 μM. The identification of seed compounds from the experiments on a few compounds demonstrates the effectiveness of our docking calculations. Furthermore, the detailed analyses using MD and FMO calculations suggested an interaction mechanism in which the hydroxyl group of BP-1-102 forms a hydrogen bond with E166 of Mpro. The Mpro inhibitory activity of SH-4-54, a derivative without the aforementioned hydroxyl group, was investigated and observed to be significantly reduced, with an IC50 of 81.5 μM. This result strongly supports the suggested interaction mechanism.
Collapse
Affiliation(s)
- Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Kenji Matsumoto
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Toshiyuki Hamada
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan
| | - Hinako Koze
- Department of Chemistry, Graduate School of Science and Engineering, Kagoshima University, 1-21-35 Korimoto, Kagoshima 890-0065, Japan
| | - Masanori Baba
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-8580, Japan
| | - Mika Okamoto
- Division of Infection Control Research, Center for Advanced Science Research and Promotion, Kagoshima University, 1-21-24, Korimoto, Kagoshima 890-8580, Japan
| | - Masayuki Sudoh
- Department of Translational Research, Joint Research Center for Human Retrovirus Infection, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| |
Collapse
|
30
|
Chukwuka AV, Adegboyegun AD, Adeogun AO. Algal bloom-mediated microplastic dispersion in coastal areas of West Africa: Integrated insights and risk projections from molecular models and remote-sensed evaluations. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137590. [PMID: 39954431 DOI: 10.1016/j.jhazmat.2025.137590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/01/2025] [Accepted: 02/10/2025] [Indexed: 02/17/2025]
Abstract
Algal blooms along the West African coast threaten ecosystems and human health due to nutrient enrichment and rising temperatures. This remote-sensing study examined the relationships between chlorophyll-a concentrations, environmental variables, and the potential for microplastic retention in blooms using molecular docking models for predictive insights. Correlation analyses revealed region-specific associations, with moderate positive correlations between chlorophyll and temperature along the southwest Nigeria-Togo coastline and near Liberia and Sierra Leone (r = 0.2-0.4) and strong correlations with particulate carbon across most regions (r = 0.6-0.8). Chlorophyll fluorescence correlations were generally low (r = 0.2), except for higher correlations in the Senegal-Gabon and Côte d'Ivoire-Ghana stretches, indicating that localized factors influence bloom dynamics. Molecular docking results predict that polycarbonate microplastics have the strongest binding affinities with algal proteins, particularly flagellin (-11.3 kcal/mol), suggesting significant retention potential within bloom matrices. In contrast, ethylene plastics displayed weaker interactions (up to -2.2 kcal/mol) and a high dissociation constant (Kd = 0.079 M), indicating minimal retention potential. The low Kd values for polycarbonateprotein interactions (e.g., 5.15e09 M for flagellin) predict a concerning scenario where microplastics become increasingly integrated into algal biomass, increasing exposure risks for marine life. Warm, nutrient-rich conditions along the West African coast, especially from southwest Nigeria to Togo and Côte d'Ivoire to Sierra Leone, are expected to increase the frequency and severity of algal blooms. This proliferation disrupts biodiversity and water quality while straining local fisheries by altering marine food webs. To mitigate microplastic entrapment from algal blooms and protect vulnerable marine ecosystems, targeted monitoring and intervention strategies are essential.
Collapse
Affiliation(s)
- Azubuike Victor Chukwuka
- Environmental Quality Control Department, National Environmental Standards and Regulations Enforcement Agency (NESREA), Nigeria.
| | - Ayotunde Daniel Adegboyegun
- Environmental Quality Control Department, National Environmental Standards and Regulations Enforcement Agency (NESREA), Nigeria
| | - Aina O Adeogun
- Hydrobiology and Fisheries Unit, Department of Zoology, University of Ibadan, Nigeria.
| |
Collapse
|
31
|
Yin WH, Liu Y, Huang HH, Li PY, Liu X, Bai FQ. Construction of Photosensitizer Candidates in Photodynamic Therapy: Computer Aided Design, Calculation, and Screening. J Org Chem 2025; 90:1825-1834. [PMID: 39877937 DOI: 10.1021/acs.joc.4c02428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Thiophene and pyrrole units are extensively utilized in light-responsive materials and have significantly advanced the field of organic photovoltaics (OPV). This progress has inspired our exploration of photosensitizers (PS) for photodynamic therapy (PDT). Currently, traditional PS face limitations in clinical application, including a restricted variety and narrow applicability. Drawing upon molecular design concepts from OPV, we aim to transcend these limitations in PDT. Given the abundance of candidate molecules, effective screening is crucial. Theoretical calculations and electronic structure analyses serve as precise and practical screening methods. In this study, we adopted strategies successfully employed in OPV molecular design, focusing on donor-acceptor (D-A) and acceptor-donor-acceptor (A-D-A) structures. Using density functional theory (DFT) and time-dependent density functional theory (TDDFT), we systematically designed combinations of promising organic fragments. These fragments include polythiophene and polypyrrole-dominated donor structures, paired with five electron acceptors: indene (Ind), diketopyrrole (DPP), naphthalimide (Ni), benzothiazole (Btd), and dithiazolyl diketopyrrole (Tbo). Through meticulous calculations, we obtained electronic structures and spectral properties for all candidate molecules, facilitating an efficient screening process. Our findings highlight that those combinations of polypyrrole-based frameworks with DPP, Ni, and Btd show significant promise for PS applications. Approximately 13% of candidates were selected through comprehensive comparison, markedly reducing molecular design time and experimental costs. This interdisciplinary approach holds potential to pave the way for more targeted and successful PS designs.
Collapse
Affiliation(s)
- Wei-Huang Yin
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
- Institute of Theoretical Chemistry and College of Chemistry, Jilin University, Changchun 130023, P.R. China
| | - Yang Liu
- Department of Orthopedics, Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Hou-Hou Huang
- Institute of Theoretical Chemistry and College of Chemistry, Jilin University, Changchun 130023, P.R. China
| | - Peng-Yuan Li
- Institute of Theoretical Chemistry and College of Chemistry, Jilin University, Changchun 130023, P.R. China
| | - Xin Liu
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Fu-Quan Bai
- Institute of Theoretical Chemistry and College of Chemistry, Jilin University, Changchun 130023, P.R. China
| |
Collapse
|
32
|
Li Y, Ding Z, Cheng T, Hu Y, Zhong F, Ren S, Wang S. Single-cell transcriptomics analysis reveals dynamic changes and prognostic signature in tumor microenvironment of PDAC. Sci Rep 2025; 15:4025. [PMID: 39894886 PMCID: PMC11788434 DOI: 10.1038/s41598-025-86950-8] [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: 09/03/2024] [Accepted: 01/15/2025] [Indexed: 02/04/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignant tumor characterized by a complex tumor microenvironment (TME) with significant heterogeneity, posing immense challenges for devising effective therapeutic strategies. This study aims to elucidate the dynamic changes in the TME during PDAC progression and develop a prognostic model using single-cell RNA sequencing (scRNA-seq) data. We utilized a previously published comprehensive dataset comprising 31 samples (including 8 PDAC I, 9 PDAC II, 6 PDAC III, and 8 PDAC IV) to characterize the changes in TME composition with PDAC progression through advanced scRNA-seq analysis. We found that as cancer progresses, immune cells gradually become a predominant component in late-stage PDAC. We defined a novel Treg and exhausted T cell signature gene, TNFRSF4. Additionally, we identified a prognostic gene set (RPS10, MIF, MT-ATP6, CSTB, IFI30, NPC2, BTG1, CTSD, FCGR2A, SEC61G, IER3, HSPB1, HMOX1, and ZFP36L1) and differentiated high-risk from low-risk PDAC patients based on median risk score threshold. Based on these findings, we developed a novel prognostic model that identifies poorer prognosis in high-risk groups. Furthermore, our analysis revealed significant interactions between cells at different stages of PDAC and identified three promising therapeutic agents (XR-11576, Ixabepilone, and AMONAFIDE) based on correlated genes. Finally, molecular docking studies validated their potential by confirming stable binding with key protein targets. This study not only provides insights into the evolving TME of PDAC but also offers a new prognostic model and potential therapeutic strategies, contributing to improved management and treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Yongsheng Li
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Zhilong Ding
- Department of Hepatobiliary Surgery, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China
| | - Tingxin Cheng
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Yihuai Hu
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China
| | - Fei Zhong
- Department of Laboratory Medicine, The Affiliated Huaian Hospital of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, Jiangsu, China.
| | - Shiying Ren
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| | - Shiyan Wang
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai'an, China.
| |
Collapse
|
33
|
Liu S, Guan H, Wang F. Genetic susceptibility and potential therapeutic targets of unruptured intracranial aneurysms: A genome-wide study based on Mendelian randomization. Clin Neurol Neurosurg 2025; 249:108749. [PMID: 39847889 DOI: 10.1016/j.clineuro.2025.108749] [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: 07/26/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND At present, although some studies have offered certain insights into the genetic factors related to unruptured intracranial aneurysms (uIAs), the potential genetic targets associated with uIAs remain largely unknown. Thus, this research adopted Mendelian randomization (MR) analysis to study two genome-wide association studies on uIAs, aiming to determine the reliable genetic susceptibility and potential therapeutic targets for uIAs. METHODS This study summarizes the data of expression quantitative trait loci (eQTL) as exposure data. The outcome data of uIAs were derived from the study by Bakker et al. and the FinnGen Biobank (version R10). The reliable genetic susceptibility and potential therapeutic targets of uIAs were identified by means of Mendelian randomization (MR) methods, with the inverse variance weighting (IVW) method as the primary analytical approach. Simultaneously, sensitivity and pleiotropy analyses were carried out, and the results were visualized. Subsequently, drug predictions and molecular docking were conducted for the potential gene targets to verify their reliability. RESULTS The MR analysis of the training cohort identified 100 targets related to uIAs. Then, these 100 gene targets and eQTL data were verified by MR Analysis again with the testing cohort. Finally, 7 gene targets were selected, namely MTMR3, SERINC1, CITED2, NKX3-1, ATOX1, MYADM and SLC20A1-DT.GO/KEGG enrichment analysis confirmed that the 7 gene targets mainly participate in the process Biological functions and pathways such as art development, cellular response to hypoxia, male Gonad development, RNA polymerase II specific DNA binding transcription factor binding, DNA binding transcription factor binding, Mineral absorption, Inositol phase metabolism, Photoshatidylinositol signaling system, etc.The protein-protein interaction(PPI) network describes the interactions between seven gene targets and related proteins.The molecular docking diagram shows good binding between candidate drugs and proteins related to gene targets. CONCLUSIONS The study identified 7 reliable gene susceptibility and potential therapeutic targets associated with uIAs, offering new insights for clinical diagnosis and treatment of uIAs, and suggesting novel research directions for understanding the etiology and molecular mechanisms of uIAs.
Collapse
Affiliation(s)
- Shuming Liu
- Department of Interventional Therapy, the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Huiyuan Guan
- Department of Breast Surgery,the Huzhou Maternal and Child Health Hospital, Huzhou, China
| | - Feng Wang
- Department of Interventional Therapy, the First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
34
|
Haider M, Sharma S, Agrahari AK, Dikshit M, Pathak DP, Asthana S. Crystallographic mining driven computer-guided approach to identify the ASK1 inhibitor likely to perturb the catalytic region. J Biomol Struct Dyn 2025; 43:1290-1304. [PMID: 38069610 DOI: 10.1080/07391102.2023.2291545] [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: 10/16/2023] [Accepted: 11/18/2023] [Indexed: 01/16/2025]
Abstract
The pathological levels of reactive oxygen species (ROS) and oxidative stress has been recognized as a critical driver for inflammatory disorders. Apoptosis signal-regulating kinase 1 (ASK1) has been reported to be activated by intracellular ROS and its inhibition leads to a down regulation of p38-and JNK-dependent signaling. ASK1 inhibitors are reported to have the potential to treat clinically important inflammatory pathologies including liver, pulmonary and renal disorders. In view of its biological and pathological significance, inhibition of ASK1 with small molecules has been pursued as an attractive strategy to combat human diseases such as non-alcoholic steatohepatitis (NASH). Despite several ASK1 inhibitors being developed, the failure in Phase 3 clinical trials of most advanced candidate selonsertib's, underscores to discover therapeutic agents with diverse chemical moiety. Here, by using structural pharmacophore and enumeration strategy on mining co-crystals of ASK1, different scaffolds were generated to enhance the chemical diversity keeping the critical molecular interaction in the catalytic site intact. A total of 15,772 compounds were generated from diverse chemical scaffolds and were evaluated using a virtual screening pipeline. Based on docking and MM-GBSA scores, a lead candidate, S3C-1-D424 was identified from top hits. A comparative molecular dynamics simulations (MD) of APO, Selonsertib and shortlisted potential candidates combined with pharmacokinetics profiling and thermodynamic analysis, demonstrating their suitability as potential ASK1 inhibitors to explore further for establishment towards hit-to-lead campaign.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mohamad Haider
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSR University, New Delhi, India
| | - Shilpa Sharma
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Ashish Kumar Agrahari
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Madhu Dikshit
- Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
- Pharmacology Division, Central Drug Research Institute, Lucknow, India
| | - Dharam Pal Pathak
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research (DIPSAR), DPSR University, New Delhi, India
| | - Shailendra Asthana
- Computational Biophysics and CADD Group, Computational and Mathematical Biology Center (CMBC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| |
Collapse
|
35
|
Sahota JS, Guleria K, Sambyal V. XRCC1 Polymorphisms p.Arg194Trp, p.Arg280His, and p.Arg399Gln, Polycyclic Aromatic Hydrocarbons, and Infertility: A Case-Control and In Silico Study. Biochem Genet 2025; 63:730-760. [PMID: 38514504 DOI: 10.1007/s10528-024-10743-3] [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: 09/27/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024]
Abstract
XRCC1 is involved in repair of single-strand breaks generated by mutagenic exposure. Polymorphisms within XRCC1 affect its ability to efficiently repair DNA damage. Polycyclic aromatic hydrocarbons or PAHs are genotoxic compounds which form bulky DNA adducts that are linked with infertility. Few reports suggest combined role of XRCC1 polymorphisms and PAHs in infertility. Present study investigates association of three XRCC1 polymorphisms (p.Arg194Trp, p.Arg280His, p.Arg399Gln) with male and female infertility in a North-West Indian population using case-control approach. Additionally, in silico approach has been used to predict whether XRCC1 polymorphisms effect interaction of XRCC1 with different PAHs. For case-control study, XRCC1 polymorphisms were screened in peripheral blood samples of age- and gender-matched 201 infertile cases (♂-100, ♀-101) and 201 fertile controls (♂-100, ♀-101) using PCR-RFLP method. For in silico study, AutoDock v4.2.6 was used for molecular docking of B[a]P, BPDE-I, ( ±)-anti-BPDE, DB[a,l]P, 1-N, 2-N, 1-OHP, 2-OHF with XRCC1 and assess effect of XRCC1 polymorphisms on their interaction. In case-control study, statistical analysis showed association of XRCC1 p.Arg280His GA genotype (p = 0.027), A allele (p = 0.019) with reduced risk of male infertility. XRCC1 p.Arg399Gln AA genotype (p = 0.021), A allele (p = 0.014) were associated with reduced risk for female primary infertility. XRCC1 p.Arg194Trp T allele was associated with increased risk for female infertility (p = 0.035). In silico analysis showed XRCC1-PAH interaction with non-significant effect of XRCC1 polymorphisms on predicted binding. Therefore, present study concludes that XRCC1 polymorphism-modified risk for male and female infertility in North-West Indians without significant effect on predicted XRCC1-PAH interactions. This is the first report on XRCC1 in female infertility.
Collapse
Affiliation(s)
- Jatinder Singh Sahota
- Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Kamlesh Guleria
- Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India
| | - Vasudha Sambyal
- Cytogenetics Laboratory, Department of Human Genetics, Guru Nanak Dev University (GNDU), Amritsar, Punjab, 143005, India.
| |
Collapse
|
36
|
Zhou H, Zhang M, Lian J, Wang R, Yang Z, Wang J, Bi X. DSN1 Interaction With Centromere-Associated Proteins Promotes Chromosomal Instability in Hepatocellular Carcinoma. Mol Carcinog 2025; 64:329-343. [PMID: 39560395 DOI: 10.1002/mc.23845] [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: 10/03/2024] [Accepted: 10/26/2024] [Indexed: 11/20/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most prevalent type of liver cancer. Dosage suppressor of NNF1 (DSN1), a component of the MIS12 kinetochore complex, encodes a kinetochore protein crucial for proper mitotic assembly. The role of DSN1 in HCC remains to be elucidated. In this study, we utilized The Cancer Genome Atlas, the Hepatocellular carcinoma Cell Database, and other databases to analyze DSN1 expression and prognosis in samples from patients with HCC. We investigated the signaling pathways regulated by DSN1 and their implications in HCC. Additionally, we engineered siRNA/shRNA and overexpression vectors for DSN1 and assessed the specific mechanisms of regulatory pathways of DSN1 in hepatoma cell lines and subcutaneous tumor xenograft model. Our findings revealed that DSN1 expression was significantly upregulated in patients with HCC, correlating with decreased survival rates. Elevated DSN1 expression led to the overproduction of cell cycle-related proteins through direct interaction with Centromere Protein T. This interaction contributes to chromosomal instability in patients with HCC, resulting in an aberrant cell cycle and fostering the development and progression of HCC. Increased DSN1 expression is pivotal in HCC initiation and progression. Investigating DSN1 offers valuable insights into the pathogenesis, treatment, and prevention of HCC.
Collapse
Affiliation(s)
- Hongrui Zhou
- College of Life Science, Liaoning University, Shenyang, China
| | - Mengxue Zhang
- College of Life Science, Liaoning University, Shenyang, China
| | - Jiabing Lian
- College of Life Science, Liaoning University, Shenyang, China
| | - Ruichang Wang
- College of Life Science, Liaoning University, Shenyang, China
| | - Zhe Yang
- College of Life Science, Liaoning University, Shenyang, China
| | - Jin Wang
- College of Life Science, Liaoning University, Shenyang, China
| | - Xiuli Bi
- College of Life Science, Liaoning University, Shenyang, China
- Key Laboratory of Chronic Disease Occurrence and Nutrition Intervention, Liaoning University, Shenyang, China
| |
Collapse
|
37
|
Ding X, Tan D, Wang Z, Yin H. Ginkgolide B regulates apoptosis, oxidative stress, and mitochondrial dysfunction in MPP +-induced SK-N-SH cells by targeting HDAC4/JNK pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03815-7. [PMID: 39878815 DOI: 10.1007/s00210-025-03815-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/14/2025] [Indexed: 01/31/2025]
Abstract
Ginkgolide B (GB) is a bioactive constituent found in Ginkgo biloba leaves that has been long recognized as a protective agent against many neurological disorders. Our study aimed to examine the effect of GB in an in vitro Parkinson's disease (PD) model and to investigate its neuroprotective mechanism as a primary objective. SK-N-SH cells were challenged with 1-methyl-4-phenylpyridinium (MPP+) to act as a PD-like model of neuronal damage. CCK-8 method, flow cytometry assay, and fluorescent probe JC-1 respectively measured cell viability, apoptosis, and mitochondrial membrane potential (MMP). Oxidative stress parameters were examined with assay kits. Nicotinamide adenine dinucleotide phosphate level and adenosine triphosphate (ATP) synthesis were also appraised. RT-qPCR examined mitochondrial DNA (mtDNA) release. Western blotting analyzed the proteins implicated in apoptosis and the histone deacetylase 4 (HDAC4)/Jun N-terminal kinase (JNK) pathway. GB concentration-dependently alleviated MPP+-stimulated viability loss, apoptosis, oxidative stress, and mitochondrial dysfunction in SK-N-SH cells. GB docked with HDAC4 and downregulated the HDAC4/JNK pathway. HDAC4 overexpression further reduced the viability and aggravated apoptosis, oxidative stress, and mitochondrial dysfunction in GB-treated SK-N-SH cells challenged with MPP+. Altogether, GB might inactivate the HDAC4/JNK pathway to protect against MPP+-triggered neuronal damage in PD.
Collapse
Affiliation(s)
- Xu Ding
- School of Traditional Chinese Medicine, Jiangsu College of Nursing, No. 9 Keji Avenue, Huai'an City, Jiangsu Province, China
| | - Dongming Tan
- School of Traditional Chinese Medicine, Jiangsu College of Nursing, No. 9 Keji Avenue, Huai'an City, Jiangsu Province, China
| | - Zhao Wang
- School of Traditional Chinese Medicine, Jiangsu College of Nursing, No. 9 Keji Avenue, Huai'an City, Jiangsu Province, China
| | - Hongying Yin
- Huai'an Hospital Affiliated to Yangzhou University, The Fifth People's Hospital of Huai'an), 1 Huaihe East Road, Huaiyin District, Huai'an City, Jiangsu Province, China.
| |
Collapse
|
38
|
Zhang H, Xu J, Chen M, Yin J, Hou Y, Tang B. The OnSPN2 from the nipa palm hispid beetle Octodonta nipae is a multipurpose defense tool against proteases from different peptidase families. INSECT SCIENCE 2025. [PMID: 39828949 DOI: 10.1111/1744-7917.13483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 01/22/2025]
Abstract
Serpins (serine protease inhibitors) constitute a superfamily of proteins with functional diversity and unusual conformational flexibility. In insects, serpins act as multiple inhibitors, by forming inactive acyl-enzyme complexes, in regulating Spätzles activation, phenoloxidases (POs) activity, and other cytokines. In this study, we present the cloning and characterization of Octodonta nipae serpin2 (OnSPN2), a 415 residues protein homologous to Tenebrio molitor 42Dd-like. Notably, OnSPN2 features an arginine residue (R364) at the P1 position, and additional arginine residues (R362, R367) at the P3 and P3' positions, respectively which is crucial for protease inhibition. Immunohistochemistry (IHC) and Western blot analyses revealed that OnSPN2 is primarily synthesized in plasmatocytes and then released into the plasma to exert its function. RNA interference results indicated that OnSPN2 knockdown may depress serine protease in melanization and remarkably increase the transcript level of Attacin in hemolymph, but its messenger RNA levels were not changed upon immune induction. Reciprocal co-immunoprecipitation assay results confirmed that OnSPN2 binds to OnPPAF1 and OnSP8, indicating its role as a negative regulator in the PO and AMP pathway. Intriguingly, several cathepsin-L isoforms were identified in the OnSPN2 immunoprecipitated samples. The cathepsin-L inhibition assays and protein-protein docking results, identified cathepsin-L as a potential target of OnSPN2. These results indicate that OnSPN2 is produced as an intracellular resident and additionally is associated with the PO and AMP pathway. OnSPN2 represents a multiple defense tool that may provide multiple antiproteolytic functions.
Collapse
Affiliation(s)
- Huajian Zhang
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural, School of Rural Revitalization), Hainan University, Haikou, China
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University
| | - Jiawei Xu
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural, School of Rural Revitalization), Hainan University, Haikou, China
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University
| | - Mintao Chen
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural, School of Rural Revitalization), Hainan University, Haikou, China
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University
| | - Jiawei Yin
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural, School of Rural Revitalization), Hainan University, Haikou, China
- School of Breeding and Multiplication (Sanya Institute of Breeding and Multiplication), Hainan University
| | - Youming Hou
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Insect Ecology, Department of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Baozhen Tang
- State Key Laboratory of Agricultural and Forestry Biosecurity, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Insect Ecology, Department of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
39
|
Dong ZY, He MJ, Yu YK, Wang F, Zhao PY, Ran DL, Fu DS, He Q, Yang RP, Zhang JA. Integrative genetics and multiomics analysis reveal mechanisms and therapeutic targets in vitiligo highlighting JAK STAT pathway regulation of CTSS. Sci Rep 2025; 15:2245. [PMID: 39824912 PMCID: PMC11742684 DOI: 10.1038/s41598-025-86134-4] [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: 09/15/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
Vitiligo is a complex autoimmune disease characterized by the loss of melanocytes, leading to skin depigmentation. Despite advances in understanding its genetic and molecular basis, the precise mechanisms driving vitiligo remain elusive. Integrating multiple layers of omics data can provide a comprehensive view of disease pathogenesis and identify potential therapeutic targets. The study aims to delineate the genetic and molecular mechanisms of vitiligo pathogenesis using an integrative multiomics strategy. We focus on exploring the regulatory influence of the JAK/STAT pathway on Cathepsin S, a potential therapeutic target in vitiligo. Our GWAS-meta analysis pinpointed five druggable genes: ERBB3, RHOH, CDK10, MC1R, and NDUFAF3, and underwent drug target exploration and molecular docking. SMR analysis linked CTSS, CTSH, STX8, KIR2DL3, and GRHPR to vitiligo through pQTL and eQTL associations. Microarray and single-cell RNA-seq data showed differential expression of CTSS and STAT1/3 in vitiligo patients' blood and skin lesions. Our study offers novel perspectives on vitiligo's genetic and molecular basis, highlighting the JAK/STAT pathway's role in regulating CTSS for antigen processing in melanocytes. Further research is needed to confirm these results and assess the therapeutic potential of CTSS and related genes.
Collapse
Affiliation(s)
- Zi-Yue Dong
- Department of Dermatology, Zhengzhou People's Hospital, Zhengzhou, Henan, China
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - Ming-Jie He
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - Yong-Kai Yu
- Department of Dermatology, Jiangsu Province Hospital and Nanjing Medical University First Affiliated Hospital, Nanjing, Jiangsu, China
| | - Fang Wang
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - Peng-Yuan Zhao
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - De-Long Ran
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - De-Shuang Fu
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - Qing He
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China
| | - Run-Ping Yang
- Department of Dermatology, The Sixth Medical Center of Chinese, PLA General Hospital, 6 Fucheng Road, Haidian District, Beijing, 100048, China.
| | - Jiang-An Zhang
- Department of Dermatology, First Affiliated Hospital of Zhengzhou University, No.1 Longhu Outer Ring Road, Jinshui District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
40
|
Han D, Wu Z, Zhang C, Wei Z, Chao F, Xie X, Liu J, Song Y, Song X, Shao D, Wang S, Xu G, Chen G. GILT stabilizes cofilin to promote the metastasis of prostate cancer. Cell Death Discov 2025; 11:10. [PMID: 39820478 PMCID: PMC11739388 DOI: 10.1038/s41420-025-02288-0] [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: 06/03/2024] [Revised: 12/11/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Gamma-interferon-induced lysosomal thiol reductase (GILT), known for catalyzing disulfide bond reduction, is involved in various physiological processes. While the involvement of GILT in the development of various tumors has been demonstrated, the mechanisms underlying its regulation in prostate cancer (PCa) are not fully understood. In the present study, we confirmed that GILT was significantly upregulated in PCa and facilitated tumor metastasis. Mechanistically, GILT stabilized the cofilin protein by competitively binding to cofilin with Src family tyrosine kinase (SRC), inhibiting SRC-mediated tyrosine phosphorylation of cofilin, thereby suppressing the ubiquitination pathway degradation of cofilin. GILT overexpression stabilized and increased the protein level of cofilin in PCa cells and promoted the metastasis of PCa cells by accelerating actin dynamics through cofilin-mediated actin severing. Our findings reveal a novel mechanism of GILT in PCa and provide a new potential target for the diagnosis and treatment of PCa patients.
Collapse
Affiliation(s)
- Dunsheng Han
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhiming Wu
- Department of Urology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Cong Zhang
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ziwei Wei
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fan Chao
- Department of Urology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Xuefeng Xie
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jinke Liu
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yufeng Song
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xiaoming Song
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Dingchang Shao
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Shiyu Wang
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Guoxiong Xu
- Research Center for Clinical Medicine, Jinshan Hospital, Fudan University, Shanghai, China.
| | - Gang Chen
- Department of Urology, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
41
|
Chen J, He J, Wang X, Bai L, Yang X, Chen J, He Y, Chen K. Glis1 inhibits RTEC cellular senescence and renal fibrosis by downregulating histone lactylation in DKD. Life Sci 2025; 361:123293. [PMID: 39643036 DOI: 10.1016/j.lfs.2024.123293] [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: 09/10/2024] [Revised: 11/17/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Accelerated senescence of renal tubular epithelial cells (RTEC) is critical in the progression of diabetic kidney disease (DKD). GLIS family zinc finger 1 (Glis1) alleviates age-related renal fibrosis in naturally aged mice. However, the role and associated mechanism of Glis1 in accelerated senescence of RTEC and the development of DKD remain unclear. METHODS Glis1 expression was examined in the renal tubules of patients with DKD and STZ-induced DKD mice. Glis1-CKO and Glis1-overexpression mice were generated to assess the effect of Glis1 on renal dysfunction and senescence in RTEC. The interplay between Glis1 and histone lactylation during cellular senescence was elucidated in vivo and in vitro. RESULTS Glis1 expression was significantly decreased in RTEC of DKD and DKD mice. Glis1 overexpression alleviated renal dysfunction and accelerated RTEC senescence in DKD mice. Histone lactylation levels significantly increased in the kidneys of mice with DKD. Lactylation enhancers, including rotenone and nala, diminished the protective effects of Glis1 against cellular senescence, whereas treatment with sodium dichloroacetate, a lactylation inhibitor, enhanced Glis1's anti-senescence capabilities. Protein-protein interaction tools, AlphaFold2 and AutoDock, indicated that Glis1 might directly bind to the lactyltransferase KAT5 with multiple interaction sites. In vitro, the interaction between KAT5 and histone H3 was enhanced under high glucose conditions. However, Glis1 overexpression led to a significant reduction in the binding affinity between histones and KAT5, which could decrease lactylation levels. CONCLUSIONS Glis1 inhibits tubular accelerated senescence by downregulating histone lactylation and alleviating kidney fibrosis during DKD progression.
Collapse
Affiliation(s)
- Juan Chen
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Junling He
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Xiaoyue Wang
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Lihua Bai
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Xin Yang
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Jia Chen
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases
| | - Yani He
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases; State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China
| | - Kehong Chen
- Department of Nephrology, Daping Hospital, Army Medical University, Chongqing 400042, China; Chongqing Key Laboratory of Precision Diagnosis and Treatment for Kidney Diseases; State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China.
| |
Collapse
|
42
|
Zhang L, Wang K, Huang L, Deng B, Chen C, Zhao K, Wang W. Ganoderic Acid A Alleviates Severe Acute Pancreatitis by Modulating Gut Homeostasis and Inhibiting TLR4-NLRP3 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1563-1579. [PMID: 39811933 PMCID: PMC11740897 DOI: 10.1021/acs.jafc.4c07635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025]
Abstract
Background Severe acute pancreatitis (SAP) manifests as a critical state marked by acute abdominal symptoms, often associated with intestinal barrier dysfunction, exacerbating SAP retroactively. Ganoderic acid A (GAA) demonstrates anti-inflammatory properties in various inflammatory disorders. Nonetheless, its potential therapeutic impact on SAP and the underlying mechanisms remain unexplored. Methods In both wild-type and TLR4-/- mice, experimental SAP was induced using caerulein plus lipopolysaccharide. Caerulein injections were administered intraperitoneally following 7 days of intragastric GAA administration. Additionally, the potential mechanisms by which GAA ameliorates SAP were further investigated using fecal microbiota transplantation and TLR4-overexpressing IEC-6 cells. Results We observed that GAA treatment significantly ameliorated serum levels of amylase, lipase, and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α) in SAP mice. Pretreatment with GAA mitigated pathological injuries and reduced M1 macrophage and neutrophil infiltration in pancreatic or ileal tissues. Additionally, GAA treatment down-regulated TLR4-MAPK/NF-κB signaling and NLRP3 inflammasome activation in the pancreatic and ileal tissues of SAP mice. The results further revealed that the gavage of GAA decreased bacterial translocation (Escherichia coli and EUB338), repaired intestinal barrier dysfunction (ZO-1, occludin, DAO, and FITC), increased lysozyme and MUC2 expression, and raised the levels of short-chain fatty acids. Analysis of the gut microbiome showed that the beneficial effects of GAA treatment were associated with improvements in pancreatitis-associated gut microbiota dysbiosis, characterized by notable increases in α-diversity and the abundance of probiotics such as Akkermansia, GCA-900066575, and Parvibacter. Fecal transplantation experiments further confirmed that GAA exerts protective effects by modulating intestinal flora. The protective role of GAA in intestinal and pancreatic injuries is mediated by the inhibition of TLR4 signaling, as further evidenced in TLR4-deficient mice and TLR4-overexpressed IEC-6 cells. The results of docking indicated that GAA interacts with TLR4 via a hydrophobic interaction. Conclusions The study demonstrates that GAA significantly alleviates SAP through its anti-inflammatory and antioxidant capacities, as well as by restoring intestinal homeostasis, thereby providing insights into novel treatments for SAP.
Collapse
Affiliation(s)
- Lilong Zhang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kunpeng Wang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Li Huang
- Department
of Gastroenterology, Renmin Hospital of
Wuhan University, Wuhan, Hubei 430060, China
| | - Beiying Deng
- Department
of Gastroenterology, Renmin Hospital of
Wuhan University, Wuhan, Hubei 430060, China
| | - Chen Chen
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Kailiang Zhao
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Weixing Wang
- Department
of General Surgery, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Hubei
Key Laboratory of Digestive System Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- General
Surgery Laboratory, Renmin Hospital of Wuhan
University, Wuhan, Hubei 430060, China
- Central
Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
43
|
Lee S, Ryu G, Shin S, Kim W, Yoon M, Kim Y, Park S, Kim Y, Cho SY. Clinically-Driven Rapidly Developed Nanoparticle Corona for Label-Free Cerebrospinal Fluid Leakage Detection. ACS NANO 2025; 19:950-962. [PMID: 39714438 DOI: 10.1021/acsnano.4c12364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Rapid diagnosis of cerebrospinal fluid (CSF) leaks is critical as endoscopic endonasal skull base surgery gains global prominence. Current clinical methods such as endoscopic examination with and without intrathecal injection of fluorescent dye are invasive and rely on subjective judgment by physicians, highlighting the clinical need for label-free point-of-care (POC). However, a viable solution remains undeveloped due to the molecular complexity of CSF rhinorrhea mixed with nasal discharge and the scarcity of specific biomarkers, delaying sensor development. In this study, we accelerated the development of a label-free CSF detection method for clinical use using a nanoparticle corona. We engineered corona nanointerfaces on near-infrared (nIR) fluorescent single-walled carbon nanotubes (SWCNTs) through noncovalent functionalization with 12 custom-designed poly(ethylene glycol) (PEG) lipids. By high-throughput screening of the corona library for the CSF biomarker β-trace protein (βTP), we selected the optimal corona, achieving a limit of detection (LOD) down to 1.46 mg/L, maintaining its selectivity even in human nasal discharge. Using molecular dynamics and docking simulations, we characterized the 3D morphology and βTP binding energy of the optimal corona in a quantified way. The corona nanosensor accurately diagnosed CSF leakages from eight patients having lumbar drainage and one patient with CSF leakage due to diverse diseases without any sample preparations. By integrating the nanosensor with custom-designed in vivo and in vitro form factors such as a camera and endoscope, we showed its potential for versatile and practical use in clinical settings. This accelerated sensor development platform can meet future urgent clinical demands for various diseases and conditions.
Collapse
Affiliation(s)
- Seungju Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gwanghui Ryu
- Department of Otorhinolaryngology-Head and Neck Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Seyoung Shin
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Woojin Kim
- Department of Materials Science and Engineering, Kookmin University, Seoul 02707, Republic of Korea
| | - Minyeong Yoon
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yeji Kim
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seongjun Park
- College of Transdisciplinary Innovations, Seoul National University, Seoul 08826, Republic of Korea
- Department of Biomedical Sciences, Seoul National University, Seoul 03080, Republic of Korea
| | - YongJoo Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Soo-Yeon Cho
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
44
|
Wang F, Liu L, Wang J, Zhou Y, Feng X, Liu K. Therapeutic Potential of Curcumin in Diabetic Cardiomyopathy: Modulation of Pyroptosis Pathways. Cardiovasc Drugs Ther 2025:10.1007/s10557-024-07644-3. [PMID: 39786506 DOI: 10.1007/s10557-024-07644-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Cardiac inflammation is a basic pathological process of diabetic cardiomyopathy (DCM). Inflammatory response is closely related to pyroptosis, which is a recently identified programmed cell death type. Curcumin (CUR) is a polyphenol extracted from turmeric and has been reported to be crucial in alleviating pyroptosis in DCM. However, the exact mechanism by which CUR improves pyroptosis remains unclear. Therefore, we aimed to investigate the effect of CUR on pyroptosis in DCM and explore the potential mechanisms. METHODS The molecular docking (MOD) analysis was performed using AutoDock Tools to evaluate the binding patterns and affinities between CUR and tripartite motif containing 21 (TRIM21), as well as between TRIM21 and gasdermin D (GSDMD). Subsequently, DCM models were established in Sprague-Dawley (SD) rats (in vivo) by administering streptozotocin (STZ) and feeding them a high-fat diet. In addition, H9C2 cells were cultured in a high glucose and palmitate environment to construct in vitro models of DCM. Rats or cells were treated by CUR directly. Subsequently, body weight (BW), heart weight (HW)/BW ratio, fasting blood glucose level, and lipid metabolism were measured. Pathological changes were analyzed using hematoxylin and eosin (H&E) and Masson staining. Small interfering RNA (si-RNA) was used to knockdown TRIM21 expression, and the pyroptosis protein expression and cellular activity were evaluated in different groups. RESULTS MOD analysis revealed that CUR had a strong binding affinity with TRIM21, and TRIM21 showed a robust interaction with GSDMD. STZ-induced diabetic SD rats showed metabolic abnormalities, structural changes in the ventricle, and the expression of TRIM21 and pyroptosis markers, including nod-like receptor protein-3 (NLRP3), Caspase-1, and GSDMD, were upregulated. CUR reduced cardiac remodeling and improved cardiac function in vivo. CUR inhibited pyroptosis by regulating TRIM21 through in vivo and in vitro studies. CONCLUSION CUR improves DCM by regulating TRIM21 expression to inhibit pyroptosis. Furthermore, this study provides novel approaches and experimental evidence for the research and treatment of DCM and presents new insights into its potential mechanisms.
Collapse
Affiliation(s)
- Fei Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, Jiangsu, China
| | - Lehan Liu
- Medical School of Nantong University, Nantong, 226000, Jiangsu, China
| | - Jiaxin Wang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, Jiangsu, China
| | - Yizhu Zhou
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, Jiangsu, China
| | - Xiaochun Feng
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, Jiangsu, China.
| | - Kun Liu
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226000, Jiangsu, China.
| |
Collapse
|
45
|
Xiong K, Pan B, Fang H, Tao Z. Single-cell sequencing analysis reveals cancer-associated pericyte subgroup in esophageal squamous cell carcinoma to predict prognosis. Front Immunol 2025; 15:1474673. [PMID: 39835116 PMCID: PMC11743493 DOI: 10.3389/fimmu.2024.1474673] [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: 08/02/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025] Open
Abstract
Background The role of cancer-associated pericytes (CAPs) in tumor microenvironment (TME) suggests that they are potential targets for cancer treatment. The mechanism of CAP heterogeneity in esophageal squamous cell carcinoma (ESCC) remains unclear, which has limited the development of treatments for tumors through CAPs. Therefore, a comprehensive understanding of the classification, function, cellular communication and spatial distribution of CAP subpopulations in ESCC is urgently needed. Methods This study used large-sample single-cell transcriptome sequencing (scRNA-seq) data to investigate pericytes' subpopulation characteristics, functions, upstream and downstream regulation and interactions with other components of the TME in the ESCC, and analyzed prognostically in conjunction with Bulk RNA-seq data. In addition, pericyte subpopulations were validated and their spatial distribution in the ESCC TME was observed by multiplex immunofluorescence. Drug prediction and molecular docking was further used to validate the medicinal value of drug targets. Results CAPs in the ESCC TME were found to be highly heterogeneous, and we identified six pericyte subtypes: c1_ARHGDIB, c2_BCAM, c3_LUM, c4_SOD2, c5_TYMS, and c6_KRT17, which have commonality in a part of their functions, and each of them has a major function to play, by having different strengths of interaction with different components in the TME. In addition, we found that c4_SOD2 was negatively correlated with prognosis, conversely, c5_TYMS was positively correlated with prognosis. The drug with a better effect on c5_TYMS was docetaxel (binding energy = -8.1, -8.7 kcal/mol); raloxifene may be more effective against c4_SOD2, although raloxifene has a slightly lower binding energy to SOD2 (-6.4 kcal/mol), it has a higher binding energy to PDGFRβ (-8.1 kcal/mol). Conclusion The present study identified and discovered pericyte subpopulations that were significantly associated with prognosis, which provides new biomarkers for predicting patient prognosis and adds usable targets for immunotherapy, and it is also important for gaining insights into the composition of the TME in ESCC.
Collapse
Affiliation(s)
- Kai Xiong
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Bing Pan
- Tianjia Genomes Tech Cor. Ltd., Hefei, China
| | - Hao Fang
- Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ziyou Tao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
46
|
Visitsatthawong S, Anuwan P, Lawan N, Chaiyen P, Wongnate T. Mechanistic insights into allosteric regulation of the reductase component of p-hydroxyphenylacetate 3-hydroxylase by p-hydroxyphenylacetate: a model for effector-controlled activity of redox enzymes. RSC Chem Biol 2025; 6:81-93. [PMID: 39649338 PMCID: PMC11618861 DOI: 10.1039/d4cb00213j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 11/30/2024] [Indexed: 12/10/2024] Open
Abstract
Understanding how an enzyme regulates its function through substrate or allosteric regulation is crucial for controlling metabolic pathways. Some flavin-dependent monooxygenases (FDMOs) have evolved an allosteric mechanism to produce reduced flavin while minimizing the use of NADH and the production of harmful hydrogen peroxide (H2O2). In this work, we investigated in-depth mechanisms of how the reductase component (C1) of p-hydroxyphenylacetate (HPA) 3-hydroxylase (HPAH) from Acinetobacter baumanii is allosterically controlled by the binding of HPA, which is a substrate of its monooxygenase counterpart (C2). The C1 structure can be divided into three regions: the N-terminal domain (flavin reductase); a flexible loop; and the C-terminal domain, which is homologous to NadR, a repressor protein having HPA as an effector. The binding of HPA to NadR induces a conformational change in the recognition helix, causing it to disengage from the NadA gene. The HPA binding site of C1 is located at the C-terminal domain, which can be divided into five helices. Molecular dynamics simulations performed on HPA-docked C1 elucidated the allosteric mechanism. The carboxylate group of HPA maintains the salt bridge between helix 2 and the flexible loop. This maintenance shortens the loop between helices 2 and 3, causing helix 3 to disengage from the N-terminal domain. The aromatic ring of HPA induces a conformational change in helices 1 and 5, pulling helix 4, analogous to the recognition helix in NadR, away from the N-terminal domain. A Y189A mutation, obtained from site-saturation mutagenesis, confirms that HPA with an unsuitable conformation cannot induce the conformational change of C1. Additionally, an HPA-independent effect is observed, in which Arg20, an NADH binding residue on the N-terminal domain, occasionally disengages from helix 4. This model provides valuable insights into the allosteric regulation of two-component FDMOs and aromatic effector systems.
Collapse
Affiliation(s)
- Surawit Visitsatthawong
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC) Wangchan Valley Rayong Thailand
| | - Piyanuch Anuwan
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC) Wangchan Valley Rayong Thailand
| | - Narin Lawan
- Department of Chemistry, Faculty of Science, Chiang Mai University Chiang Mai Thailand
| | - Pimchai Chaiyen
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC) Wangchan Valley Rayong Thailand
| | - Thanyaporn Wongnate
- School of Biomolecular Science and Engineering, Vidyasirimedhi Institute of Science and Technology (VISTEC) Wangchan Valley Rayong Thailand
| |
Collapse
|
47
|
Aroua LM, Alkhaibari IS, Alminderej FM, Messaoudi S, Chigurupati S, Al-mahmoud SA, Albadri AE, Emwas AH, Mohammed HA. Synthesis, bioactivity, and molecular docking of pyrazole bearing Schiff-bases as prospective dual alpha-amylase and alpha-glucosidase inhibitors with antioxidant activity. J Mol Struct 2025; 1320:139291. [DOI: 10.1016/j.molstruc.2024.139291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
48
|
Kahvecioglu D, Ozguven SY, Sicak Y, Tok F, Öztürk M, Kocyigit-Kaymakcioglu B. Synthesis and molecular docking analysis of novel hydrazone and thiosemicarbazide derivatives incorporating a pyrimidine ring: exploring neuroprotective activity. J Biomol Struct Dyn 2024:1-15. [PMID: 39731533 DOI: 10.1080/07391102.2024.2442758] [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: 12/29/2023] [Accepted: 04/29/2024] [Indexed: 12/30/2024]
Abstract
The increasing global prevalence of Alzheimer's disease necessitates the development of novel therapeutic approaches. Neurodegenerative diseases are associated with increased oxidative stress and levels of cholinesterase enzymes. Hence, the development of cholinesterase inhibitors and antioxidants may provide neuroprotective effects. Our study focused on the synthesis of a new series of hydrazone and thiosemicarbazide derivatives bearing a pyrimidine ring. The compounds of structures were characterized by FT-IR, 1H NMR, 13C NMR, and HR-MS spectroscopic methods. Compounds 3a and 4f were determined using COSY and HSQC spectra. Compared to the standard drug galantamine (IC50 = 4.82 ± 0.75 µM), compound 3d exhibited remarkable inhibitory activity against AChE (IC50 values of 20.15 ± 0.44 µM). This compound was more effective against BChE (IC50 = 36.42 ± 0.73 µM) than galantamine (IC50 = 45.54 ± 0.18 µM). Antioxidant assays revealed the robust antioxidant activity of compound 3d. Furthermore, docking studies have shown that the active site of enzymes interacts strongly with electron donors through hydrogen bonds, while the aromatic ring structure plays an active role in π interactions.
Collapse
Affiliation(s)
- Dilay Kahvecioglu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Türkiye
- Institute of Health Sciences, Marmara University, Istanbul, Türkiye
- Edirne Sultan 1. Murat State Hospital, Republic of Türkiye Ministry of Health, Edirne, Türkiye
| | - Serap Yilmaz Ozguven
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Trakya University, Edirne, Türkiye
| | - Yusuf Sicak
- Department of Herbal and Animal Production, Koycegiz Vocational School, Mugla Sitki Kocman University, Mugla, Türkiye
| | - Fatih Tok
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Istanbul, Türkiye
| | - Mehmet Öztürk
- Department of Chemistry, Faculty of Science, Mugla Sitki Kocman University, Mugla, Türkiye
| | | |
Collapse
|
49
|
Xu X, Ying H, Wang X, Hong W, Zhang M. Identification of Angiogenesis-Related Gene Signatures and Prediction of Potential Therapeutic Targets in Ulcerative Colitis Using Integrated Bioinformatics. J Inflamm Res 2024; 17:11699-11717. [PMID: 39741751 PMCID: PMC11687120 DOI: 10.2147/jir.s478880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/10/2024] [Indexed: 01/03/2025] Open
Abstract
Objective This study aims to clarify angiogenesis mechanisms in ulcerative colitis and identify potential therapeutic targets. Methods The Gene Expression Omnibus (GEO) database was used to obtain expression profiles and clinical data for UC and healthy colon tissues. Angiogenesis-related gene sets were acquired from GeneCards. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) identified UC-associated hub genes. The CIBERSORT algorithm assessed immune cell infiltration. Analyses of Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed to determine biological mechanisms. External datasets were utilized to validate and characterize the angiogenesis-related genes in relation to biological agents. Additionally, an ulcerative colitis mouse model was constructed to verify the key genes' expression using real-time quantitative PCR. To predict potential therapeutic agents, we used the DGIdb database. Molecular docking modeled small molecule binding conformations to key gene targets. Results This study identified 1,247 DEGs enriched in inflammatory/immune pathways from UC and healthy colon samples. WGCNA indicated the black and light cyan modules were most relevant. Intersecting these with 89 angiogenesis genes revealed 5 UC-associated hub genes (pdgfrb, vegfc, angpt2, tnc, hgf). Validation via ROC analysis, differential expression, and a mouse model confirmed upregulation, supporting their potential as UC diagnostic biomarkers. Bioinformatics approaches like protein-protein interaction, enrichment analysis, and GSEA revealed involvement in PDGFR and PI3K-Akt signaling pathways. CIBERSORT analysis of immune cell infiltration showed positive correlations between the key genes and various immune cells, especially neutrophils, highlighting angiogenesis-inflammation interplay in UC. A ceRNA network was constructed. Drug prediction and molecular docking revealed potential UC therapies like sunitinib and imatinib targeting angiogenesis. Conclusion This study identified and validated five angiogenesis-related genes (pdgfrb, vegfc, angpt2, tnc, hgf) that may serve as diagnostic biomarkers and drug targets for UC.
Collapse
Affiliation(s)
- Xijuan Xu
- Department of Anus & Intestine Surgery, Taizhou First People’s Hospital, Taizhou, Zhejiang, People’s Republic of China
| | - Hongan Ying
- Department of Geriatrics, Taizhou First People’s Hospital, Taizhou, People’s Republic of China
| | - Xiaozhi Wang
- Department of Anus & Intestine Surgery, Taizhou First People’s Hospital, Taizhou, Zhejiang, People’s Republic of China
| | - Weiwen Hong
- Department of Anus & Intestine Surgery, Taizhou First People’s Hospital, Taizhou, Zhejiang, People’s Republic of China
| | - Meng Zhang
- Department of General Surgery, Taizhou First People’s Hospital, Taizhou, Zhejiang, People’s Republic of China
| |
Collapse
|
50
|
Zhuang M, Zhu S, Su L, Liu L, Ji M, Dai C, Liu J, Zhang W, Pu H. PKCδ modulates SP1 mediated mitochondrial autophagy to exacerbate diacetylmorphine-induced ferroptosis in neurons. Int Immunopharmacol 2024; 143:113468. [PMID: 39490143 DOI: 10.1016/j.intimp.2024.113468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/14/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
Diacetylmorphine (DA) is widely implicated in neuronal injury; however, the underlying mechanisms remain unclear. We investigated the role of iron metamorphosis in DA-induced neurotoxicity using Sprague-Dawley rats and PC12 and SH-SY5Y cells. Tandem mass tag proteomics analysis showed that the upregulation of protein kinase C delta (PKCδ) and iron metabolism-related protein transferrin receptor (TFRC) significantly the enriched iron metabolism pathway. Subsequent experiments showed that DA exposure significantly upregulated PKCδ in PC12 cells, which increased the nuclear translocation of specificity protein 1 (SP1), and the intracellular free iron and lipid peroxide levels. In addition, silencing of PKCδ in rats improved behaviour and restored the expression level of glutathione peroxidase 4 (GPX4). In addition, DA exposure activated mitochondrial autophagy in PC12 cells, leading to a decrease in the mitochondrial membrane potential, accumulation of reactive oxygen species (ROS), elevation of LC3 (which plays a key role in autophagy), and a decrease in p62 expression. Following the inhibition of autophagy, the mitochondrial membrane potential and ROS were restored, as was the expression of voltage-dependent anion channel 1 (VDAC1) and GPX4. In conclusion, the present study suggests that PKCδ regulates SP1, further exacerbating DA-induced neuronal ferroptosis. Therefore, inhibition of PKCδ and mitochondrial autophagy or ferroptosis may be a key therapeutic target to ameliorate neurotoxicity following DA exposure.
Collapse
Affiliation(s)
- Mengjie Zhuang
- Xinjiang Medical University, School of Basic Medical Science, Urumqi 830017, China
| | - Sensen Zhu
- Xinjiang Medical University, School of Basic Medical Science, Urumqi 830017, China
| | - Liping Su
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Li Liu
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Min Ji
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China
| | - Chenlu Dai
- Xinjiang Medical University, School of Basic Medical Science, Urumqi 830017, China
| | - Jingyu Liu
- Xinjiang Medical University, School of Basic Medical Science, Urumqi 830017, China
| | - Wei Zhang
- Department of Pathology, The First Affiliated Hospital of Xinjiang Medical University, Xinjiang, China.
| | - Hongwei Pu
- Xinjiang Medical University, School of Basic Medical Science, Urumqi 830017, China; Key Laboratory of Forensic Medicine, School of Basic Medical Sciences, Xinjiang Medical University, China; Department of Discipline Construction, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China.
| |
Collapse
|