1
|
Abd-Allah WH, Abdel-Maksoud MS, Elbaset MA, Korany RMS, Ibrahim IM, Hassan RM. Design and synthesis of novel cyclohexanecarboxamides with anticonvulsant effect by activating Nrf2-ARE pathway. Bioorg Chem 2025; 159:108357. [PMID: 40088686 DOI: 10.1016/j.bioorg.2025.108357] [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: 01/09/2025] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
This study investigated a series of novel cyclohexanecarboxamide derivatives 4a-e, 5a-e and 6a-e as anticonvulsant and neuroprotective agents. Compounds 4a-e, 5a-e, and 6a-e were synthesized starting from cyclohexanone and aniline derivatives and evaluated for their anticonvulsant effects using maximal electroshock (MES) and subcutaneous pentylenetetrazole (scPTZ) seizure models. The most potent compounds 4b, 5c, and 6d demonstrated 83.33 % protection in the scPTZ test, while compounds 5a and 6b showed 100 % protection in the MES test. Further quantitative evaluation showed that compound 6d was the most active derivative in scPTZ test (ED50 = 0.04 mmol/kg) and it was more potent than the two reference drugs phenobarbital and ethosuximide by 1.7 and 25.7-fold, respectively. Notably, all the compounds were free from neurotoxic side effects. Mechanistic studies indicated that the compounds 4b, 5c, and 6d exerted neuroprotective effects by modulating oxidative stress markers and activating the Nrf2/ARE pathway. Histopathological examination of brain of animals treated with compounds 4b, 5c, or 6d corroborated the neuroprotective properties. Additionally, molecular docking study and dynamic simulations were also carried out to study the mechanism of Nrf2 activation by the most active compound 6d. These results spotlight the potential of these derivatives as promising candidates for further development as anti-epileptic and neuroprotective agents.
Collapse
Affiliation(s)
- Walaa H Abd-Allah
- Pharmaceutical Chemistry Department, Collage of Pharmaceutical Science and Drug Manufacturing, Misr University for Science and Technology, P.O. 77, 6th of October City, Giza, Egypt.
| | - Mohammed S Abdel-Maksoud
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Marawan A Elbaset
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Centre, Giza, Egypt; Neurology Department, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Reda M S Korany
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Egypt
| | - Ibrahim M Ibrahim
- Biophysics Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Rasha M Hassan
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Institute, National Research Centre (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| |
Collapse
|
2
|
Alzarea SI, Afzal M, Moglad E, Alhassan HH, Alzarea AI, Alsaidan OA, Sayyed N, Kazmi I. In silico and in vivo evaluation of erucic acid against pentylenetetrazole-induced seizures in mice by modulating oxidative stress, neurotransmitters and neuroinflammation markers. Nutr Neurosci 2025:1-16. [PMID: 40022513 DOI: 10.1080/1028415x.2025.2463677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
BACKGROUND Pentylenetetrazole (PTZ) is a commonly used chemical to induce epileptic seizures in experimental animals. AIM To investigate the neuroprotective effects of erucic acid against PTZ-induced seizures in mice and explore its underlying mechanisms. METHODOLOGY The mice were randomly allocated into four groups: normal control, PTZ-treated (35 mg/kg via intraperitoneal injection), and PTZ + erucic acid (at doses of 10 and 20 mg/kg). Various parameters were assessed, including the percentage of animals experiencing convulsions, latency to death, percentage of deaths, levels of neurotransmitters, pro-inflammatory cytokines such as interleukin-6 (IL-6), interleukin-1 beta (IL-1β), tumor necrosis factor-alpha (TNF-α), nuclear factor kappa B (NF-κB), oxidative stress marker malondialdehyde (MDA), antioxidant enzymes catalase (CAT), superoxide dismutase (SOD), reduced glutathione (GSH), and caspase-3. The docking analysis was performed using AutoDock Vina software. RESULTS Erucic acid markedly reduced the severity and frequency of PTZ-induced seizures, significantly decreased mortality rates, and restored altered neurotransmitter levels in mice. It alleviated oxidative stress by increasing the activity of antioxidant enzymes and reducing malondialdehyde (MDA) levels. Additionally, erucic acid mitigated neuroinflammation by downregulating pro-inflammatory cytokine production and inhibiting NF-κB activation. Molecular docking studies demonstrated that erucic acid exhibited strong binding affinities toward key molecular targets, including GABA (-4.546), NF-κB (-5.982), and caspase-3 (-5.22), suggesting its potential as a neuroprotective agent. CONCLUSION Erucic acid may be an effective natural compound in PTZ-induced seizures in mice by restoring neurotransmitters, oxidative stress and neuroinflammatory mediators. It could prove to be a better alternative in the treatment of epilepsy.
Collapse
Affiliation(s)
- Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Hassan H Alhassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | | | - Omar Awad Alsaidan
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka, Saudi Arabia
| | - Nadeem Sayyed
- School of Pharmacy, Glocal University, Saharanpur, India
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
3
|
Althagafi HA. Neuroprotective role of chlorogenic acid against hippocampal neuroinflammation, oxidative stress, and apoptosis following acute seizures induced by pentylenetetrazole. Metab Brain Dis 2024; 39:1307-1321. [PMID: 39133453 DOI: 10.1007/s11011-024-01400-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/19/2024] [Indexed: 08/13/2024]
Abstract
This study investigated the neuroprotective effect of chlorogenic acid (CGA) on pentylenetetrazole (PTZ)-induced acute epileptic seizures in mice. Epileptic animals received CGA (200 mg/kg) or sodium valproate (standard antiepileptic agent, 200 mg/kg) for four weeks. Results revealed that pre-administration of CGA significantly reversed the behavioral changes following pentylenetetrazole (PTZ) injection. Further, CGA pre-treatment caused significant increases in acetylcholinesterase (AChE) activity and brain-derived neurotrophic factor (BDNF) levels, along with marked increases in dopamine, norepinephrine, and serotonin levels. Additionally, the increased antioxidant enzymes activities, along with higher glutathione (GSH) contents and upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) gene expression, were indicative of a notable improvement in the cellular antioxidant defense in mice treated with CGA. These results were associated with lowered malondialdehyde (MDA) and nitric oxide (NO) levels. Moreover, epileptic mice that received CGA showed significant declines in the content of interleukin-1 beta (IL-1β), interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and nuclear factor kappa-B (NF-κB), besides downregulating inducible nitric oxide synthase (iNOS) expression. Remarkably, CGA counteracted hippocampal apoptosis by lessening the levels of pro-apoptotic biomarkers [Bcl-2-associated X protein (Bax) and caspase-3] and increasing the anti-apoptogenic marker level of B-cell lymphoma 2 (Bcl-2). The hippocampal histopathological findings corroborated the abovementioned changes. In sum, these findings suggest that CGA could mediate the neuroprotective effect against PTZ-induced epilepsy via modulation of neurotransmitters, oxidative damage, neuroinflammation, and apoptosis. CGA, therefore, could be considered a valuable antiepileptic therapeutic supplement.
Collapse
Affiliation(s)
- Hussam A Althagafi
- Department of Biology, Faculty of Science, Al-Baha University, Al Baha, Saudi Arabia.
| |
Collapse
|
4
|
Qu J, Pei H, Li XZ, Li Y, Chen JM, Zhang M, Lu ZQ. Erythrocyte membrane biomimetic EGCG nanoparticles attenuate renal injury induced by diquat through the NF-κB/NLRP3 inflammasome pathway. Front Pharmacol 2024; 15:1414918. [PMID: 39045044 PMCID: PMC11263105 DOI: 10.3389/fphar.2024.1414918] [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: 04/09/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Diquat (DQ) poisoning can cause multiple organ damage, and the kidney is considered to be the main target organ. Increasing evidence shows that alleviating oxidative stress and inflammatory response has promising application prospects. Epigallocatechin gallate (EGCG) has potent antioxidant and anti-inflammatory effects. In this study, red blood cell membrane (RBCm)-camouflaged polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) were synthesized to deliver EGCG (EGCG-RBCm/NPs) for renal injury induced by DQ. Human renal tubular epithelial cells (HK-2 cells) were stimulated with 600 μM DQ for 12 h and mice were intraperitoneally injected with 50 mg/kg b.w. DQ, followed by 20 mg/kg b.w./day EGCG or EGCG-RBCM/NPs for 3 days. The assessment of cellular vitality was carried out using the CCK-8 assay, while the quantification of reactive oxygen species (ROS) was performed through ROS specific probes. Apoptosis analysis was conducted by both flow cytometry and TUNEL staining methods. Pathological changes in renal tissue were observed. The expressions of NLRP3, IL-1β, IL-18, NFκB and Caspase1 were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunohistochemistry, immunofluorescence, and Western blot. The results showed that the DQ group had increased ROS expression, increased the level of oxidative stress, and increased apoptosis rate compared with the control group. Histopathological analysis of mice in the DQ group showed renal tubular injury and elevated levels of blood urea nitrogen (BUN), serum creatinine (SCr), kidney injury molecule-1 (KIM-1), and cystatin C (Cys C). Furthermore, the DQ group exhibited heightened expression of NLRP3, p-NFκB p65, Caspase1 p20, IL-1β, and IL-18. However, EGCG-RBCm/NPs treatment mitigated DQ-induced increases in ROS, apoptosis, and oxidative stress, as well as renal toxicity and decreases in renal biomarker levels. Meanwhile, the expression of the above proteins were significantly decreased, and the survival rate of mice was ultimately improved, with an effect better than that of the EGCG treatment group. In conclusion, EGCG-RBCm/NPs can improve oxidative stress, inflammation, and apoptosis induced by DQ. This effect is related to the NF-κB/NLRP3 inflammasome pathway. Overall, this study provides a new approach for treating renal injury induced by DQ.
Collapse
Affiliation(s)
- Jie Qu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Hui Pei
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Xin-Ze Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Yan Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Jian-Ming Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Min Zhang
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Zhong-Qiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| |
Collapse
|
5
|
Peng X, McClements DJ, Liu X, Liu F. EGCG-based nanoparticles: synthesis, properties, and applications. Crit Rev Food Sci Nutr 2024; 65:2177-2198. [PMID: 38520117 DOI: 10.1080/10408398.2024.2328184] [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] [Indexed: 03/25/2024]
Abstract
(-)-Epigallocatechin-3-gallate (EGCG) is a natural phenolic substance found in foods and beverages (especially tea) that exhibits a broad spectrum of biological activities, including antioxidant, antimicrobial, anti-obesity, anti-inflammatory, and anti-cancer properties. Its potential in cardiovascular and brain health has garnered significant attention. However, its clinical application remains limited due to its poor physicochemical stability and low oral bioavailability. Nanotechnology can be used to improve the stability, efficacy, and pharmacokinetic profile of EGCG by encapsulating it within nanoparticles. This article reviews the interactions of EGCG with various compounds, the synthesis of EGCG-based nanoparticles, the functional attributes of these nanoparticles, and their prospective applications in drug delivery, diagnosis, and therapy. The potential application of nanoencapsulated EGCG in functional foods and beverages is also emphasized. Top-down and bottom-up approaches can be used to construct EGCG-based nanoparticles. EGCG-based nanoparticles exhibit enhanced stability and bioavailability compared to free EGCG, making them promising candidates for biomedical and food applications. Notably, the non-covalent and covalent interactions of EGCG with other substances significantly contribute to the improved properties of these nanoparticles. EGCG-based nanoparticles appear to have a wide range of applications in different industries, but further research is required to enhance their efficacy and ensure their safety.
Collapse
Affiliation(s)
- Xiaoke Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | | | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|