1
|
Tu J, Cheng W, Ban Z, Ning J, Tan X. Discovery of farnesoid X receptor antagonists from Salvia miltiorrhiza based on virtual screening and activity verification. Bioorg Med Chem Lett 2025; 123:130222. [PMID: 40199406 DOI: 10.1016/j.bmcl.2025.130222] [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: 01/05/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
The farnesoid X receptor (FXR) is a promising therapeutic target for the treatment of non-alcoholic fatty liver disease (NAFLD). Salvia miltiorrhiza, a traditional Chinese medicine, has demonstrated significant efficacy in the prevention and treatment of liver diseases. Consequently, investigating the potential effects of Salvia miltiorrhiza on FXR could provide new insights for NAFLD treatment. This study explores whether active ingredients from Salvia miltiorrhiza can target FXR and serve as therapeutic agents for treating NAFLD. The findings revealed that cynaroside and lithospermic acid displayed strong FXR antagonistic activity, with IC50 values of 5.41 ± 1.08 μM and 16.92 ± 2.68 μM, respectively. Salvianolic acid A also showed moderate activity (IC50 = 56.35 ± 4.54 μM). MTT assays demonstrated that these three compounds were non-toxic to HepG2 and LO2 cells at a concentration of 200 μM. Molecular dynamics simulations were conducted to elucidate the interaction mechanisms of cynaroside and lithospermic acid with FXR. These results suggest that cynaroside and lithospermic acid from Salvia miltiorrhiza may be potential candidates for targeting FXR in treating NAFLD.
Collapse
Affiliation(s)
- Jiaojiao Tu
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wa Cheng
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Zhenghu Ban
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Jiayi Ning
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Xiangduan Tan
- Guangxi Key Laboratory of Drug Discovery and Optimization, College of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
2
|
Mohan RD, Kulkarni NV. Recent developments in the design of functional derivatives of edaravone and exploration of their antioxidant activities. Mol Divers 2025; 29:1895-1910. [PMID: 39102113 DOI: 10.1007/s11030-024-10940-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/11/2024] [Indexed: 08/06/2024]
Abstract
Edaravone, a pyrazalone derivative, is an antioxidant and free radical scavenger used to treat oxidative stress-related diseases. It is a proven drug to mitigate conditions prevailing to oxidative stress by inhibiting lipid peroxidation, reducing inflammation, and thereby preventing endothelial cell death. In recent years, considerable interest has been given by researchers in the derivatization of edaravone by adding varieties of substituents of versatile steric and functional properties to improve its antioxidant and pharmacological activity. This review accounts all the important methods developed for the derivatization of edaravone and the impacts of the structural modifications on the antioxidant activity of the motif.
Collapse
Affiliation(s)
- R Divya Mohan
- Department of Chemistry, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, 690525, India
| | - Naveen V Kulkarni
- Department of Chemistry, Amrita Vishwa Vidyapeetham, Amritapuri, Kerala, 690525, India.
| |
Collapse
|
3
|
Tseng HF, Chao HN, Lin CH, Kuo CY. Danshensu Attenuates Palmitic Acid-Induced Activation of Hepatic Stellate Cells by Regulating Pyroptosis. Int J Med Sci 2025; 22:1865-1874. [PMID: 40225863 PMCID: PMC11983302 DOI: 10.7150/ijms.107564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/07/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction: We focused on examining the role of Danshensu in reducing reactive oxygen species (ROS) production and inhibiting NLRP3 inflammasome activation, which are key factors in liver fibrosis and inflammation. We sought to explore the potential of Danshensu as a therapeutic agent for liver fibrosis by targeting the pyroptosis-inflammasome signaling pathway, providing a basis for developing effective and safer NLRP3 inflammasome inhibitors. This study aimed to investigate whether Danshensu can mitigate palmitic acid (PA)-induced activation of hepatic stellate cells (HSCs) by regulating pyroptosis in HSC-T6 and LX-2 cells. Methods: HSC-T6 and LX-2 cell lines served as the cell models. A 2',7'-dichlorofluorescin diacetate reagent was used to measure ROS production within cells. Cell protein extraction was performed using radioimmunoprecipitation assay lysis buffer. The protein concentration in each sample was measured using a BCA assay kit. Western blot analysis was used with the SDS-polyacrylamide gel electrophoresis system. Results: PA-induced activation of HSC-T6 and LX-2 cells by upregulating alpha-smooth muscle actin, integrin-β1, and connective tissue growth factor. Danshensu mitigated PA-induced ROS accumulation in these cells. Moreover, Danshensu potentially reversed the upregulation of NLRP3, cleaved caspase 1, interleukin-1, GSDME, and ASC in PA-activated LX-2 cells via pyroptosis, suggesting its therapeutic potential. Pyroptosis inhibitor tetramethylthiuram disulfide reversed Danshensu attenuated PA activation of HSC-T6 and LX-2 cells, resulting in a 2-fold increase in alpha-smooth muscle actin, integrin-β1, and connective tissue growth factor. Conclusion: Danshensu effectively attenuates PA-induced HSC activation by reducing ROS production and inhibiting pyroptosis, offering a potential therapeutic strategy for liver fibrosis.
Collapse
Affiliation(s)
- Han-Fang Tseng
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Huan-Nung Chao
- Department of Nephrology, Hanming Christian Hospital, Changhua City 500, Taiwan
| | - Chih-Hung Lin
- Department of Internal Medicine, Cathay General Hospital, Taipei 106, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan
- Department of Nursing, Cardinal Tien College of Healthcare and Management, New Taipei City 231, Taiwan
- Institute of Oral Medicine and Materials, College of Medicine, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
4
|
Xiang Y, Kuang G, Gong X, Xie H, Lin Y, Zhang X, Chen Z, Wan J, Li Z. Dihydrotanshinone I Attenuates Diet-Induced Nonalcoholic Fatty Liver Disease via Up-Regulation of IRG1. Phytother Res 2025; 39:1531-1548. [PMID: 39853881 DOI: 10.1002/ptr.8443] [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: 09/12/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease, but effective therapeutic drugs are still lacking. Dihydrotanshinone I (DHTS), a natural product isolated from Salvia miltiorrhiza , has been shown to have ameliorative effects on NAFLD. The aim of this study was to investigate the hepatoprotective effect of DHTS on NAFLD and its mechanism. A model of NAFLD and DHTS treatment was established using a Western diet to observe the effect of DHTS on NAFLD, which were detected by immunohistochemical, immunofluorescence, and other experiments. The mechanism was further explored by constructing immune responsive gene 1 (IRG1) knockout mice, RNA sequence, and molecular docking. The results revealed that DHTS significantly improved diet-induced metabolic disorders in mice, notably alleviating liver inflammation, oxidative stress, and fibrosis. Further analysis revealed that the intervention of DHTS was associated with the activation of IRG1. Subsequent experiments confirmed that IRG1 gene deletion reversed the above protective effects of DHTS in NAFLD. Mechanistically, DHTS enhanced the antioxidant nuclear factor-erythroid 2-related factor 2 (Nrf2) pathway through IRG1/itaconate and blocked the oxidative stress response in the liver. In addition, DHTS also inhibited the activation of NACHT-, leucine-rich repeat (LRR)-, and pyrin domain (PYD)-containing protein 3 (NLRP3) inflammasome via IRG1/itaconate, blocking the inflammatory amplification effect in the liver. The study suggests that DHTS may be a potential drug for the treatment of NAFLD, which exerts protective regulatory effects mainly through the IRG1/itaconate molecular pathway.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Ge Kuang
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xia Gong
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Huang Xie
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Yan Lin
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Xijian Zhang
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Zhongpei Chen
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| | - Jingyuan Wan
- Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, China
| | - Zhenhan Li
- Department of Endocrinology, Chongqing Traditional Chinese Medicine Hospital, Chongqing, China
| |
Collapse
|
5
|
Liu X, Xie X, Wang K, Liu X, Gong J, Yang Z, Li J. Raddeanin A suppresses intracellular 5Methylcytosine DNA modification engaged the metastasis of hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119036. [PMID: 39515681 DOI: 10.1016/j.jep.2024.119036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/10/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Anemonoides Raddeana (Rege) Holubhe is commonly employed in clinical practice as a traditional Chinese medicine for the treatment of conditions such as rheumatism and limb numbness. Raddeanin A (RA), an active compound derived from this Traditional Chinese Medicine (TCM), demonstrates specific anticancer properties against many tumorigeneses. However, the molecular mechanism underlying its effects on hepatocellular carcinoma (HCC) remains unexplored. AIM OF THE STUDY The aim of this study is to investigate the inhibitory effects of RA in human HCC stimulated cells and its impact on DNA methylation in tumor cells, as well as to elucidate the molecular mechanisms underlying RA's anti-tumor activity. MATERIALS AND METHODS The inhibitory effects of RA on QGY-7703 and HepG2 cells were evaluated. The IC50 values were determined by employing non-linear sigmoidal curve fitting to analyze the normalized response. The impact of RA was investigated in cells overexpressing DNMT3A and DNMT3B. The effects of RA on cell cycle progression and apoptosis were assessed. Furthermore, the influence of RA on cellular methylation was determined, along with its effects on the expression levels of DNMT3A, DNMT3B, Bcl-2, Bax, and Caspase-3. RESULTS The findings demonstrate that RA induces cell cycle arrest at the G0/G1 phase and promotes apoptosis in hepatocellular carcinoma cells. Furthermore, RA effectively inhibits the invasion and migration of human HCC stimulated cells. The expression of DNMT3A and DNMT3B is downregulated by RA, effectively suppressing the intracellular 5mC DNA modification level. Moreover, the overexpression of these enzymes in RA-treated human HCC stimulated cells significantly impacts the overall 5mC level and hinders tumor metastasis by restricting migration and invasion. CONCLUSION The RA compound acts as an antagonist against HCC by reducing intracellular DNA 5mC levels through mechanisms mediated by methyltransferase. Moreover, RA demonstrates the capacity to induce apoptosis in tumor cells, thereby exerting its anti-tumor effects. The findings of this study provide valuable insights for enhancing the pharmacodynamic efficacy of RA in HCC treatment.
Collapse
Affiliation(s)
- Xin Liu
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China; Postgraduate School, Jilin Normal University, Siping, 136000, China
| | - Xiaoyan Xie
- Department of Pharmacy, The 3rd Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Kangyu Wang
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China
| | - Xiaokang Liu
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China
| | - Jiyu Gong
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Zizhao Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Department of General Surgery, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Jiannan Li
- Key Laboratory of Chinese Medicine Analysis, School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, 130117, China.
| |
Collapse
|
6
|
Marchini V, Paradisi F. Self-sufficient biocatalytic cascade for the continuous synthesis of danshensu in flow. Appl Microbiol Biotechnol 2025; 109:13. [PMID: 39836219 PMCID: PMC11750938 DOI: 10.1007/s00253-025-13407-3] [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: 08/29/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
A new strategy has been developed to successfully produce the active component danshensu ex vivo. For this purpose, phenylalanine dehydrogenase from Bacillus sphaericus was combined with the novel hydroxyphenylpyruvate reductase from Mentha x piperita, thereby providing an in situ cofactor regeneration throughout the conversion process. The purified enzymes were co-immobilized and subsequently employed in batch biotransformation, resulting in 60% conversion of 10 mM L-dopa within 24 h, with a catalytic amount of NAD+ as cofactor. Furthermore, the bienzymatic system was implemented as a packed-bed reactor in continuous flow, achieving a conversion rate up to 80% with 60 min retention time. The process was further intensified by implementing a 48-h flow bioreaction. The biocatalysts demonstrated remarkable stability, retaining 62% of their initial activity at the end of the process. The final productivity of the isolated compound (96% purity) was calculated to be 1.84 g L-1 h-1 yielding a sustainable synthesis of danshensu. KEY POINTS: • Characterization of the hydroxyphenylpyruvate reductase from Mentha x piperita • Bi-enzymatic system in a cascade reaction to produce danshensu • Purification and isolation of the active compound danshensu.
Collapse
Affiliation(s)
- Valentina Marchini
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland
- inSEIT AG, Freiestrasse 3, 3012, Bern, Switzerland
| | - Francesca Paradisi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Freiestrasse 3, 3012, Bern, Switzerland.
| |
Collapse
|
7
|
Rijo P, Abuamara TMM, Ali Lashin LS, Kamar SA, Isca VMS, Mohammed TS, Abdrabo MSM, Amin MA, Abd El Maksoud AI, Hassan A. Glycyrrhizic Acid Nanoparticles Subside the Activity of Methicillin-Resistant Staphylococcus aureus by Suppressing PBP2a. Pharmaceuticals (Basel) 2024; 17:589. [PMID: 38794159 PMCID: PMC11123903 DOI: 10.3390/ph17050589] [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: 03/14/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Staphylococcus aureus and methicillin-resistant Staphylococcus aureus (MRSA) are classified as high-risk infections that can lead to death, particularly among older individuals. Nowadays, plant nanoparticles such as glycyrrhizic acid are recognized as efficient bactericides against a wide range of bacterial strains. Recently, scientists have shown interest in plant extract nanoparticles, derived from natural sources, which can be synthesized into nanomaterials. Interestingly, glycyrrhizic acid is rich in antioxidants as well as antibacterial agents, and it exhibits no adverse effects on normal cells. In this study, glycyrrhizic acid nanoparticles (GA-NPs) were synthesized using the hydrothermal method and characterized through physicochemical techniques such as UV-visible spectrometry, DLS, zeta potential, and TEM. The antimicrobial activity of GA-NPs was investigated through various methods, including MIC assays, anti-biofilm activity assays, ATPase activity assays, and kill-time assays. The expression levels of mecA, mecR1, blaR1, and blaZ genes were measured by quantitative RT-qPCR. Additionally, the presence of the penicillin-binding protein 2a (PBP2a) protein of S. aureus and MRSA was evaluated by a Western blot assay. The results emphasized the fabrication of GA nanoparticles in spherical shapes with a diameter in the range of 40-50 nm. The data show that GA nanoparticles exhibit great bactericidal effectiveness against S. aureus and MRSA. The treatment with GA-NPs remarkably reduces the expression levels of the mecA, mecR1, blaR1, and blaZ genes. PBP2a expression in MRSA was significantly reduced after treatment with GA-NPs. Overall, this study demonstrates that glycyrrhizic acid nanoparticles have potent antibacterial activity, particularly against MRSA. This research elucidates the inhibition mechanism of glycyrrhizic acid, which involves the suppressing of PBP2a expression. This work emphasizes the importance of utilizing plant nanoparticles as effective antimicrobial agents against a broad spectrum of bacteria.
Collapse
Affiliation(s)
- Patricia Rijo
- CBIOS—Lusófona University’s Research Center for Biosciences and Health Technologies, 1749-024 Lisbon, Portugal;
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Tamer M. M. Abuamara
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19111, Jordan; (T.M.M.A.); (L.S.A.L.); (S.A.K.)
- Department of Histology, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt
| | - Lashin Saad Ali Lashin
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19111, Jordan; (T.M.M.A.); (L.S.A.L.); (S.A.K.)
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Sherif A. Kamar
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman 19111, Jordan; (T.M.M.A.); (L.S.A.L.); (S.A.K.)
- Department of Anatomy and Embryology, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt
| | - Vera M. S. Isca
- CBIOS—Lusófona University’s Research Center for Biosciences and Health Technologies, 1749-024 Lisbon, Portugal;
| | - Tahseen S. Mohammed
- Department of Public Health and Community Medicine, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt; (T.S.M.); (M.S.M.A.)
| | - Mohamed S. M. Abdrabo
- Department of Public Health and Community Medicine, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt; (T.S.M.); (M.S.M.A.)
| | - Mohamed A. Amin
- Department of Basic Medical Science, Faculty of Dentistry, Zarqa University, Zarqa 13110, Jordan;
- Department of Microbiology and Immunology, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt
| | - Ahmed I. Abd El Maksoud
- College of Biotechnology, Misr University of Science and Technology, Giza 12573, Egypt;
- Department of Industrial Biotechnology, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat 32897, Egypt
| | - Amr Hassan
- Department of Bioinformatics, Genetic Engineering and Biotechnology Research Institute (GEBRI), University of Sadat City, Sadat 32897, Egypt
| |
Collapse
|
8
|
Hassan A, Mohsen R, Rezk A, Bangay G, Rijo P, Soliman MFM, G. A. Hablas M, Swidan KAK, Mohammed TS, Zoair MA, Mohamed AAK, Abdalrhman TI, Abdel-aleem Desoky AM, Mohamed DD, Mohamed DD, Abd El Maksoud AI, Mohamed AF. Enhancement of Vitamin C's Protective Effect against Thimerosal-Induced Neurotoxicity in the Cerebral Cortex of Wistar Albino Rats: An In Vivo and Computational Study. ACS OMEGA 2024; 9:8973-8984. [PMID: 38434836 PMCID: PMC10905602 DOI: 10.1021/acsomega.3c07239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/29/2023] [Accepted: 01/09/2024] [Indexed: 03/05/2024]
Abstract
Vitamin C was examined to ameliorate the neurotoxicity of thimerosal (THIM) in an animal model (Wistar albino rats). In our work, oxidative and antioxidative biomarkers such as SOD, LPO, and GSH were investigated at various doses of THIM with or without concurrent vitamin C administration. Furthermore, the adverse effects of THIM on hepatic tissue and cerebral cortex morphology were examined in the absence or presence of associated vitamin C administration. Also, we studied the effect of vitamin C on the metallothionein isoforms (MT-1, MT-2, and MT-3) in silico and in vivo using the RT-PCR assay. The results showed that the antioxidant biomarker was reduced as the THIM dose was raised and vice versa. THIM-associated vitamin C reduced the adverse effects of the THIM dose. The computation studies demonstrated that vitamin C has a lower ΔG of -4.9 kcal/mol compared to -4.1 kcal/mol for THIM to bind to the MT-2 protein, which demonstrated that vitamin C has a greater ability to bind with MT-2 than THIM. This is due to multiple hydrogen bonds that exist between vitamin C and MT-2 residues Lys31, Gln23, Cys24, and Cys29, and the sodium ion represents key stabilizing interactions. Hydrogen bonds involve electrostatic interactions between hydrogen atom donors (e.g., hydroxyl groups) and acceptors (e.g., carbonyl oxygens). The distances between heavy atoms are typically 2.5-3.5 Å. H-bonds provide directed, high-affinity interactions to anchor the ligand to the binding site. The five H-bonds formed by vitamin C allow it to form a stable complex with MT, while THIM can form two H-bonds with Gln23 and Cys24. This provides less stabilization in the binding pocket, contributing to the lower affinity compared to vitamin C. The histopathological morphologies in hepatic tissue displayed an expansion in the portal tract and the hepatocytes surrounding the portal tract, including apoptosis, binucleation, and karyomegaly. The histopathological morphologies in the brain tissue revealed a significant decrease in the number of Purkinje cells due to THIM toxicity. Interestingly, THIM toxicity was associated with hemorrhage and astrogliosis. Both intracellular and vasogenic edema appeared as the concentrations of THIM rose. Finally, vitamin C ameliorated the adverse effect on the cerebral cortex in Wistar albino rats.
Collapse
Affiliation(s)
- Amr Hassan
- Department
of Bioinformatics, Genetic Engineering and Biotechnology Research
Institute (GEBRI), University of Sadat City, Sadat 32897, Egypt
| | - Reham Mohsen
- College
of Biotechnology, October University for Modern Science and Arts (MSA), University Giza, Giza 11456, Egypt
| | - Ahmed Rezk
- College
of Biotechnology, October University for Modern Science and Arts (MSA), University Giza, Giza 11456, Egypt
| | - Gabrielle Bangay
- CBIOS—Research
Center for Biosciences & Health Technologies, Universidade Lusófona de Humanidades e Tecnologias, Lisboa 1749-024, Portugal
- Facultad
de Farmacia, Departamento de Ciencias Biomédicas (Área
de Farmacología; Nuevos agentes antitumorales, Acción
tóxica sobre células leucémicas), Universidad de Alcalá de Henares, Ctra. Madrid-Barcelona km. 33, Alcalá de Henares 600 28805, Madrid, Espana
| | - Patrícia Rijo
- CBIOS—Research
Center for Biosciences & Health Technologies, Universidade Lusófona de Humanidades e Tecnologias, Lisboa 1749-024, Portugal
- Instituto
de Investigação do Medicamento (iMed.ULisboa), Faculdade
de Farmácia, Universidade de Lisboa, Lisbon 1749-024, Portugal
| | - Mona F. M. Soliman
- Department
of Histology and Cell Biology, Faculty Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed G. A. Hablas
- Department
of Histology and Cell Biology, Faculty of Medicine, Suez University, Suez 43221, Egypt
| | | | - Tahseen S. Mohammed
- Department
of Public Health and Community Medicine, Faculty of Medicine, Al-Azhar University, Cairo 11884, Egypt
| | - Mohammad A. Zoair
- Department
of Physiology, Faculty of Medicine, Al-Azhar
University, Cairo 11884, Egypt
| | - Abir A. Khalil Mohamed
- Department
of Zoology, Faculty of Science, Girls Branch, Al-Azhar University, Cairo 11884, Egypt
| | - Tamer I. Abdalrhman
- Department of Histology, Faculty of Medicine, Al-Azhar University, Assiut 71524, Egypt
| | | | - Dalia D. Mohamed
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Doaa D. Mohamed
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Ahmed I. Abd El Maksoud
- Department of Industrial Biotechnology,
Genetic Engineering and Biotechnology
Research Institute (GEBRI), University of
Sadat City, Sadat 32897, Egypt
| | - Aly F. Mohamed
- Holding
Company for Vaccine and Sera Production (VACSERA), Giza 22311, Egypt
| |
Collapse
|