1
|
Chen Y, Huang X, Hu R, Lu E, Luo K, Yan X, Zhang Z, Ma Y, Zhang M, Sha X. Inhalable biomimetic polyunsaturated fatty acid-based nanoreactors for peroxynitrite-augmented ferroptosis potentiate radiotherapy in lung cancer. J Nanobiotechnology 2025; 23:338. [PMID: 40340938 PMCID: PMC12060495 DOI: 10.1186/s12951-025-03409-8] [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: 01/26/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025] Open
Abstract
The limited efficacy and poor tumor accumulation remain crucial challenges for radiotherapy against lung cancer. To address these limitations, we rationally developed a polyunsaturated fatty acid (PUFA)-based nanoreactor (DHA-N@M) camouflaged with macrophage cell membrane to improve tumoral distribution and achieve peroxynitrite-augment ferroptosis for enhanced radiotherapy against lung cancer. After nebulization, the nanoreactors exhibited superior pulmonary accumulation in orthotopic lung cancer-bearing mice, with 70-fold higher than intravenously injected nanoreactors at 12 h post-administration, and distributed deeply in the tumors. DHA-N@M selectively released nitric oxide (NO) in glutathione (GSH)-enriched tumor cells, with consumption of GSH and subsequent inactivation of glutathione peroxidase 4 (GPX4). Under radiation, NO reacted with radiotherapy-induced reactive oxygen species (ROS) to generate peroxynitrite (ONOO-), resulting in redox homeostasis disruption. Combined with docosahexaenoic acid (DHA)-induced lipid metabolism disruption, overwhelming ferroptosis was induced both in vitro and in vivo. Notably, DHA-N@M mediated ferroptosis-radiotherapy significantly suppressed tumor growth with a 93.91% inhibition in orthotopic lung cancer models. Therefore, this design provides a nebulized ferroptosis-radiotherapy strategy for lung cancer.
Collapse
Affiliation(s)
- Yiting Chen
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xueli Huang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Ruining Hu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Enhao Lu
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Kuankuan Luo
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Xin Yan
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Zhiwen Zhang
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China
| | - Yan Ma
- Department of Pharmacy, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 201108, China.
| | - Minghe Zhang
- Naval Medical Center, Naval Medical University, Shanghai, Shanghai, 200052, China.
| | - Xianyi Sha
- Department of Pharmaceutics, School of Pharmacy, Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Lane 826, Zhangheng Road, Shanghai, 201203, China.
- Quzhou Fudan Institute, 108 Minjiang Avenue, Kecheng District, Quzhou, Zhejiang Province, 324002, China.
| |
Collapse
|
2
|
Yulak F, Joha Z, Öztürk A, İnan ZDŞ, Taşkıran AŞ. Enoxaparin Protects C6 Glioma Cells from Glutamate-Induced Cytotoxicity by Reducing Oxidative Stress and Apoptosis. Mol Neurobiol 2025; 62:4631-4640. [PMID: 39472385 DOI: 10.1007/s12035-024-04587-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 10/23/2024] [Indexed: 03/05/2025]
Abstract
Recent studies suggest enoxaparin may protect the central nervous system (CNS) from damage. However, its specific effects on glial cells and the underlying mechanisms involving cell death and oxidative stress require further investigation. Therefore, this research investigated enoxaparin's potential to safeguard C6 glioma cells against glutamate-induced cytotoxicity, specifically focusing on its influence on oxidative stress and apoptotic mechanisms. To investigate the neuroprotective effects of enoxaparin against glutamate-induced cytotoxicity in C6 cells, four groups were established: a control group, a group exposed to 10 mM glutamate, a group treated with enoxaparin at concentrations ranging from 25 to 200 µM, and a group receiving both 10 mM glutamate and enoxaparin at concentrations ranging from 25 to 200 µM. Cell viability was measured using an XTT assay. To evaluate the effects of enoxaparin on oxidative stress, superoxide dismutase (SOD) and malondialdehyde (MDA) levels were measured using ELISA, along with total antioxidant status (TAS) and total oxidant status (TOS). Apoptosis was evaluated using flow cytometry, and caspase-3 activity, a key marker of apoptosis, was assessed using caspase-3 immunofluorescence staining. Enoxaparin at 50, 100, and 200 µM markedly increased cell viability in the enoxaparin + glutamate group. Enoxaparin treatment in the enoxaparin + glutamate group also significantly elevated levels of SOD and TAS, while concurrently decreasing MDA and TOS levels. These changes indicate a reduction in oxidative stress. Enoxaparin treatment further resulted in a significant decline in cleaved caspase-3 levels, a marker of apoptosis. Enoxaparin pre-treatment reduced cell death according to flow cytometry analysis. This study suggests enoxaparin's potential to shield C6 glioma cells from glutamate-induced cell death by mitigating both oxidative stress and apoptotic pathways. More research is needed to confirm this effect.
Collapse
Affiliation(s)
- Fatih Yulak
- Department of Physiology, School of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ziad Joha
- Department of Pharmacology, Faculty of Pharmacy, Sivas Cumhuriyet University, 58140, Sivas, Turkey.
| | - Ayşegül Öztürk
- Departments of Medical Services and Techniques, Vocational School of Health Services, Sivas Cumhuriyet University, Sivas, Turkey
| | - Zeynep Deniz Şahin İnan
- Department of Histology-Embryology, School of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ahmet Şevki Taşkıran
- Department of Physiology, School of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
3
|
Joha Z, Başgöz N, Özgür A, Taşkıran AŞ. Bromelain Protects Against PTZ-Induced Glial Damage and Inflammation: An In Vitro and In Silico Study. Cell Biochem Biophys 2025:10.1007/s12013-025-01703-8. [PMID: 40000586 DOI: 10.1007/s12013-025-01703-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
This study aimed to investigate how bromelain protects glial cells from pentylenetetrazole (PTZ)-induced damage, focusing on its anti-inflammatory effects. C6 glioma cells were treated with PTZ, bromelain, or a combination of PTZ and bromelain. The interactions of bromelain with iNOS (Inducible Nitric Oxide Synthase) and COX2 (Cyclooxygenase-2) were investigated using molecular docking calculations. Cell viability was measured using the XTT (Methoxynitrosulfophenyl-Tetrazolium Carboxanilide) assay. iNOS, NO (Nitric Oxide), and COX2 levels were assessed using ELISA and immunofluorescence staining. Bromelain at 50 and 100 µg/mL significantly increased cell viability (p < 0.001). On the other hand, bromelain at 50 µg/mL reduced inflammation, as indicated by lower levels of NO, iNOS, and COX2 (p < 0.001). In-silico predictions suggest that bromelain can effectively target iNOS and COX2, key inflammatory proteins. These findings indicate that bromelain protects glial cells by exerting anti-inflammatory effects. However, further research is needed to understand the underlying mechanisms fully.
Collapse
Affiliation(s)
- Ziad Joha
- Departments of Pharmacology, School of Medicine, Cumhuriyet University, Sivas, Turkey.
| | - Neslihan Başgöz
- Departments of Physiology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| | - Aykut Özgür
- Laboratory and Veterinary Health Program, Department of Veterinary Medicine, Artova Vocational School, Tokat Gaziosmanpaşa University, Tokat, Turkey
| | - Ahmet Şevki Taşkıran
- Departments of Physiology, School of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
4
|
Yulak F, Ergul M. Tannic acid protects neuroblastoma cells against hydrogen peroxide - triggered oxidative stress by suppressing oxidative stress and apoptosis. Brain Res 2024; 1844:149175. [PMID: 39168266 DOI: 10.1016/j.brainres.2024.149175] [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: 05/21/2024] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024]
Abstract
Recent investigations indicate that tannic acid is associated with a decrease in oxidative damage. Growing evidence supports the protective effects of tannic acid on the central nervous system (CNS). However, uncertainties persist regarding its influence on hydrogen peroxide (H2O2)-triggered oxidative impairment in nerve cells and its interaction with apoptosis. Hence, the objective of this work was to examine the neuroprotective impact of tannic acid on SH-SY5Y cell impairment following H2O2-induced oxidative stress, particularly concerning apoptotic pathways. The control group received no treatment, while the H2O2 group underwent treatment with 0.5 mM H2O2 for a duration of 24 h. The tannic acid group received treatment with different concentrations of tannic acid for a duration of 24 h. Meanwhile, the tannic acid + H2O2 group underwent pre-treatment with tannic acid for one hour and was subsequently subjected to 0.5 mM H2O2 for one day. Within the tannic acid + H2O2 group, the cell viability in SH-SY5Y cells was notably enhanced by tannic acid at concentrations of 2.5, 5, and 10 μM. It also resulted in a considerable rise in TAS (Total Antioxidant Status) levels and a concurrent decline in TOS (Total Oxidant Status) levels, serving as indicators of reduced oxidative stress. Additionally, tannic acid treatment resulted in decreased levels of apoptotic markers (Bax, cleaved PARP, and cleaved caspase 3) and oxidative DNA damage marker (8-oxo-dG), while increasing the anti-apoptotic marker Bcl-2. The findings from flow cytometry also revealed a significant reduction in the apoptosis rate following pretreatment with tannic acid. In summary, tannic acid demonstrates protective effects on SH-SY5Y cells in the face of H2O2-triggered oxidative damage by suppressing both oxidative stress and apoptosis. Nevertheless, additional research is warranted to assess the neuroprotective potential of tannic acid.
Collapse
Affiliation(s)
- Fatih Yulak
- Departments of Physiology, School of Medicine, Sivas Cumhuriyet University, 58140 Sivas, Turkey.
| | - Mustafa Ergul
- Department of Biochemistry, Faculty of Pharmacy, Sivas Cumhuriyet University, 58140 Sivas, Turkey.
| |
Collapse
|
5
|
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
|
6
|
Murugan R, Ramya Ranjan Nayak SP, Haridevamuthu B, Priya D, Chitra V, Almutairi BO, Arokiyaraj S, Saravanan M, Kathiravan MK, Arockiaraj J. Neuroprotective potential of pyrazole benzenesulfonamide derivative T1 in targeted intervention against PTZ-induced epilepsy-like condition in in vivo zebrafish model. Int Immunopharmacol 2024; 131:111859. [PMID: 38492342 DOI: 10.1016/j.intimp.2024.111859] [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: 12/22/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Epilepsy is a chronic neurological disease characterized by a persistent susceptibility to seizures. Pharmaco-resistant epilepsies, impacting around 30 % of patients, highlight the urgent need for improved treatments. Neuroinflammation, prevalent in epileptogenic brain regions, is a key player in epilepsy, prompting the search for new mechanistic therapies. Hence, in this study, we explored the anti-inflammatory potential of pyrazole benzenesulfonamide derivative (T1) against pentylenetetrazole (PTZ) induced epilepsy-like conditions in in-vivo zebrafish model. The results from the survival assay showed 79.97 ± 6.65 % at 150 µM of T1 compared to PTZ-group. The results from reactive oxygen species (ROS), apoptosis and histology analysis showed that T1 significantly reduces cellular damage due to oxidative stress in PTZ-exposed zebrafish. The gene expression analysis and neutral red assay results demonstrated a notable reduction in the inflammatory response in zebrafish pre-treated with T1. Subsequently, the open field test unveiled the anti-convulsant activity of T1, particularly at a concentration of 150 μM. Moreover, both RT-PCR and immunohistochemistry findings indicated a concentration-dependent potential of T1, which inhibited COX-2 in zebrafish exposed to PTZ. In summary, T1 protected zebrafish against PTZ-induced neuronal damage, and behavioural changes by mitigating the inflammatory response through the inhibition of COX-2.
Collapse
Affiliation(s)
- Raghul Murugan
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - S P Ramya Ranjan Nayak
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - B Haridevamuthu
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - D Priya
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Vellapandian Chitra
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India
| | - Bader O Almutairi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Selvaraj Arokiyaraj
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Muthupandian Saravanan
- AMR and Nanotherapeutics Laboratory, Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, Tamil Nadu 600077, India
| | - M K Kathiravan
- Dr APJ Abdul Kalam Research Lab, Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur 603203, Chengalpattu District, Tamil Nadu, India.
| |
Collapse
|
7
|
Ran L, Xu M, Zhang Z, Zeng X. The association of nutrient intake with epilepsy: A cross-sectional study from NHANES, 2013-2014. Epilepsy Res 2024; 200:107297. [PMID: 38215605 DOI: 10.1016/j.eplepsyres.2024.107297] [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] [Revised: 12/18/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
BACKGROUND Dietary nutrient supplements are helpful in the treatment of many diseases, but their effect on epilepsy is still controversial. This study aimed to evaluate the association between dietary intake of multiple nutrients and epilepsy. METHODS A total of 3963 participants from the NHANES database were involved in this study. We compared the dietary intake of 14 nutrients between the normal population and those with epilepsy. Univariable and multivariable logistic regression were conducted to evaluate the association of these nutrients with epilepsy. RESULTS Compared with the normal population, the epilepsy patients showed lower intakes of protein, vitamin B1, vitamin B6, Fe, and Zn. Multivariable logistic regression showed the negative association of vitamin B1 (OR = 0.513, 95% CI: 0.293, 0.897) with epilepsy. When vitamin B1 was divided into 4 groups according to quartiles, the highest quartile showed a lower odds ratio (OR = 0.338, 95% CI: 0.115, 0.997) than that of the lowest quartile. In different population stratifications, the association of vitamin B1 with epilepsy was different. Vitamin B1 was negatively associated with the odds ratio of epilepsy among the elderly (OR = 0.243), low-income population (OR = 0.337), and current smokers (OR = 0.283). CONCLUSION Epilepsy patients had significantly lower intakes of vitamin B1, which was inversely associated with epilepsy risk. More detailed clinical trials are needed to accurately evaluate nutritional supplements for epilepsy.
Collapse
Affiliation(s)
- Liling Ran
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Mengchang Xu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, Hunan, China.
| | - Xin Zeng
- Department of Pharmacy, Hunan Children Hospital, Changsha, Hunan 410007, China.
| |
Collapse
|
8
|
Nazarinia D, Moslehi A, Hashemi P. (-)-α-bisabolol exerts neuroprotective effects against pentylenetetrazole-induced seizures in rats by targeting inflammation and oxidative stress. Physiol Behav 2023; 272:114351. [PMID: 37714321 DOI: 10.1016/j.physbeh.2023.114351] [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: 06/27/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Epilepsy is the most common neurological disorder which is accompanied with behavioral and psychiatric alternations. Current evidences have shown that (-)-α-bisabolol (BSB) possess anti-inflammatory and antioxidative effects in several animal studies. Here, we conducted present study to evaluate its neuroprotective effects against pentylenetetrazole (PTZ)-induced seizures in rats. We used fifty male rats and they were randomly assigned into 5 groups control, BSB100, PTZ, BSB50 + PTZ, BSB100 + PTZ. The animals intraperitoneally received PTZ (45 mg/kg) for ten consecutive days to induce epilepsy model. BSB in doses of 50 and 100 mg/kg was administrated orally one hour before PTZ administration for ten days. The elevated plus maze (EPM) test was carried out to assess anxiety-like behavior. The seizure intensity was evaluated according to modifies Racine's convulsion scale (RCS). Y-maze and passive avoidance were utilized to assess working memory and aversive memory. The expression of pro-inflammatory cytokines and oxidative stress factors were measured using the enzyme-linked immunosorbent assay (ELISA). The neuronal cell loss in the hilar region was assessed using Nissl staining. Results showed that PTZ-treated rats had more seizure intensity, anxiety-like behavior, memory deficits, higher levels of TNF-α, IL-1β, and oxidative markers. Pre-treatment with BSB 100 significantly inhibited seizure intensity, anxiety-like behavior, and memory deficits; reduced levels of TNF-α, IL-1β, and MDA oxidative markers. Collectively, outcome of this work shows that BSB at the dose of 100 mg/kg may exert neuroprotective effects by mitigating seizures, oxidative stress, and neuroinflammation, and ameliorates memory and anxiety disorders in the PTZ-induced seizure rats.
Collapse
Affiliation(s)
- Donya Nazarinia
- Department of Physiology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran.
| | - Ahmadreza Moslehi
- Student Research Committee, Dezful University of Medical Sciences, Dezful, Iran
| | - Paria Hashemi
- Cellular and Molecular Research Center, Research Institute for Health Development, KurdistanUniversity of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
9
|
Tran NB, Lee SJ. Effects of Gryllus bimaculatus and Oxya chinensis sinuosa extracts on brain damage via blood-brain barrier control and apoptosis in mice with pentylenetetrazol-induced epilepsy. PLoS One 2023; 18:e0291191. [PMID: 37695764 PMCID: PMC10495007 DOI: 10.1371/journal.pone.0291191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
The demand for environmentally friendly foods with high nutritional value and low carbon emissions is increasing with the aging of the global population and the crisis of food resources. Edible insects are becoming increasingly well-known as such foods. This study evaluated the effects and mechanisms of Gryllus bimaculatus (Cricket) (Gb) and Oxya chinensis sinuosa (Grasshopper) (Ocs) extracts on epilepsy. A pentylenetetrazol (PTZ)-induced seizure mouse model was used for the study, and Gb and Ocs extracts were administered for 29 days on alternate days at concentrations of 8 g/kg and 16 g/kg. The integrity of the blood-brain barrier (BBB) and brain edema was measured using the perfusion of Evans blue dye and brain water content. Gb and Ocs extracts prevented BBB permeabilization and cerebral edema through increasing the expression of tight junction-associated proteins in the endothelial cells and reducing water content in PTZ-treated mice. Additionally, Gb and Ocs extracts protected neurons from oxidative stress and apoptosis in different brain areas. These protective effects were demonstrated through the restoration of the expression of neuronal nuclear protein and postsynaptic density protein-95, thus increasing the levels of glutathione and superoxide dismutase, decreasing lipid peroxidation, and recovering apoptosis-associated proteins, such as Bax, cleaved PARP, and cleaved caspase-3, in epileptic mice. In addition, Gb and Ocs extracts rescued PTZ-induced hyperexcitable neurons to control mice level, as supported by the restored expression of gamma-aminobutyric acid (GABA) transporter 1, the metabotropic glutamate receptors-GRM2/3, and BDNF. This study suggested that Gb and Ocs extracts are novel medicinal candidates that can help ameliorate epilepsy by improving BBB health and preventing oxidative stress-mediated apoptosis.
Collapse
Affiliation(s)
- Ngoc Buu Tran
- Department of Bioactive Materials Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeollabuk-do, Korea
| | - Sook-Jeong Lee
- Department of Bioactive Materials Sciences and Research Center of Bioactive Materials, Jeonbuk National University, Jeonju, Jeollabuk-do, Korea
| |
Collapse
|
10
|
Ovey IS, Ozsimsek A, Velioglu HA, Altay O, Mardinoglu A, Yulug B. EGb 761 reduces Ca 2+ influx and apoptosis after pentylenetetrazole treatment in a neuroblastoma cell line. Front Cell Neurosci 2023; 17:1195303. [PMID: 37744878 PMCID: PMC10516604 DOI: 10.3389/fncel.2023.1195303] [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: 03/28/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
Background Transient receptor potential (TRP) channels have been found to have significant implications in neuronal outgrowth, survival, inflammatory neurogenic pain, and various epileptogenic processes. Moreover, there is a growing body of evidence indicating that transient receptor potential (TRP) channels have a significant impact on epilepsy and its drug-resistant subtypes. Objective We postulated that EGb 761 would modulate TRPA1 channels, thereby exhibiting anti-inflammatory and neuroprotective effects in a neuroblastoma cell line. Our rationale was to investigate the impact of EGb 761 in a controlled model of pentylenetetrazole-induced generalized epilepsy. Methodology We evaluated the neuroprotective, antioxidant and anti-apoptotic effects of EGb 761 both before and after the pentylenetetrazole application in a neuroblastoma cell line. Specifically, we focused on the effects of EGB 761 on the activity of Transient receptor potential (TRP) channels. Results EGb 761 applications both before and after the pentylenetetrazole incubation period reduced Ca release and restored apoptosis, ROS changes, mitochondrial depolarization and caspase levels, suggesting a prominent prophylactic and therapeutic effect of EGb 761 in the pentylenetetrazole-induced epileptogenesis process. Conclusion Our basic mechanistic framework for elucidating the pathophysiological significance of fundamental ion mechanisms in a pentylenetetrazole treated neuroblastoma cell line provided compelling evidence for the favorable efficacy and safety profile of Egb 761 in human-relevant in vitro model of epilepsy. To the best of our knowledge, this is the first study to investigate the combined effects of EGb 761 and pentylenetetrazole on TRP channels and measure their activation level in a relevant model of human epileptic diseases.
Collapse
Affiliation(s)
- Ishak Suat Ovey
- Department of Physiology, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Türkiye
| | - Ahmet Ozsimsek
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Türkiye
| | - Halil Aziz Velioglu
- Department of Neuroscience, Faculty of Medicine, Istanbul Medipol University, Istanbul, Türkiye
- Center for Psychiatric Neuroscience, Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Ozlem Altay
- KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Burak Yulug
- Department of Neurology and Neuroscience, Faculty of Medicine, Alanya Alaaddin Keykubat University, Antalya, Türkiye
- Department of Neuroscience, Faculty of Medicine, Istanbul Medipol University, Istanbul, Türkiye
| |
Collapse
|
11
|
Nazarinia D, Karimpour S, Hashemi P, Dolatshahi M. Neuroprotective effects of Royal Jelly (RJ) against pentylenetetrazole (PTZ)-induced seizures in rats by targeting inflammation and oxidative stress. J Chem Neuroanat 2023; 129:102255. [PMID: 36878412 DOI: 10.1016/j.jchemneu.2023.102255] [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: 01/06/2023] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023]
Abstract
Epilepsy is a chronic neurological condition in which inflammation and oxidative stress play a key role in the pathogenesis. Recently, several studies have suggested that Royal Jelly (RJ) has antioxidant effects. Nevertheless, there is no evidence of its effectiveness against epilepsy. Here, we evaluated its neuroprotective effects at different doses (100 and 200 mg/kg) against pentylenetetrazole (PTZ)-induced seizures. Fifty male Wistar rats were randomly divided into five groups: control, PTZ, RJ100 + PTZ, RJ200 + PTZ and RJ100. In order to establish epilepsy model, 45 mg/kg of PTZ was injected intraperitoneally for 10 consecutive days. Seizure parameters were graded based on Racine's 7-point classification. Elevated-plus maze, Y maze and shuttle box tests were carried out to assess anxiety-like behavior, short-term memory, and passive avoidance memory, respectively. We used ELISA technique to measure the expression of the pro-inflammatory cytokines and oxidative stress factors. Also, neuronal loss in the hippocampal CA3 region was determined using Nissl staining. Our findings showed that PTZ-treated rats had more seizure intensity, anxiety-like behavior, memory dysfunction, higher levels of TNF-α, IL-1β, and oxidative markers. RJ could allay seizure severity and duration. It also improved memory function as well as anxiety level. In terms of biochemical assessment, RJ gave rise to a significant decrease in the level of IL-1β, TNF-α and MDA and it restored the activities of GPX and SOD enzymes. Hence, our study shows that RJ contains anti-inflammatory and antioxidative effects which contribute to less neuronal damage in the PTZ-induced epilepsy model.
Collapse
Affiliation(s)
- Donya Nazarinia
- Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran.
| | - Sepideh Karimpour
- Department of Physiology, School of Paramedical Sciences, Dezful University of Medical Sciences, Dezful, Iran
| | - Paria Hashemi
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mojtaba Dolatshahi
- Department of Physiology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| |
Collapse
|
12
|
Yang MT, Chou IC, Wang HS. Role of vitamins in epilepsy. Epilepsy Behav 2023; 139:109062. [PMID: 36577336 DOI: 10.1016/j.yebeh.2022.109062] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 12/27/2022]
Abstract
Epilepsy is a chronic neurological disorder that presents as recurrent, unprovoked seizures. Pharmacotherapy is the main treatment for epilepsy, but at least 30% of patients with epilepsy have pharmacoresistant epilepsy. Therefore, non-pharmacological treatments are still required. In addition to electrophysiological aberrations contributing to epileptogenesis and pathophysiology in epilepsy, neuroinflammation, oxidative stress, and metabolic derangement have been investigated as drug targets in the treatment of epilepsy. Vitamins have antioxidant, anti-inflammatory, and immunomodulatory effects, which can be beneficial for the treatment of epilepsy. Herein, we comprehensively review the role of vitamins in epilepsy. Certain epilepsies are vitamin-dependent or vitamin-responsive. Most studies on vitamins in epilepsy are of low evidence level or limited to animal studies. Nevertheless, vitamin supplementation should be considered in epilepsy therapy. Additionally, certain anti-seizure medications may alter the serum levels of certain vitamins. Monitoring the serum levels of vitamins and supplementing vitamins when needed are suggested during the follow-up of patients with epilepsy.
Collapse
Affiliation(s)
- Ming-Tao Yang
- Department of Pediatrics, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan, Taiwan.
| | - I-Ching Chou
- Division of Pediatrics Neurology, China Medical University Children's Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Huei-Shyong Wang
- Division of Pediatric Neurology, Chang Gung Children's Hospital at Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
13
|
Albrakati A. Monosodium glutamate induces cortical oxidative, apoptotic, and inflammatory challenges in rats: the potential neuroprotective role of apigenin. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:24143-24153. [PMID: 36334201 DOI: 10.1007/s11356-022-23954-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
Monosodium glutamate (MSG) is used as a flavor, and a taste enhancer was reported to evoke marked neuronal impairments. This study investigated the neuroprotective ability of flavonoid apigenin against neural damage in MSG-administered rats. Adult male rats were allocated into four groups: control, apigenin (20 mg/kg b.wt, orally), MSG (4 g/kg b.wt, orally), and apigenin + MSG at the aforementioned doses for 30 days. Regarding the levels of neurotransmitters, our results revealed that apigenin augmented the activity of acetylcholinesterase (AChE) markedly, and levels of brain monoamines (dopamine, norepinephrine, and serotonin) accompanied by lessening the activity of monoamine oxidase (MAO) as compared to MSG treatment. Moreover, apigenin counteracted the MSG-mediated oxidative stress by decreasing the malondialdehyde (MDA) levels together with elevating the glutathione (GSH) levels. In addition, pretreatment with apigenin induced notable increases in the activities of cortical superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and glutathione reductase (GR). Furthermore, apigenin attenuated the cortical inflammatory stress as indicated by lower levels of pro-inflammatory mediators such as interleukin-1 b (IL-1b), tumor necrosis factor-α (TNF-α), and nitric oxide (NO) as well as downregulated inducible nitric oxide synthase (iNOS) expression levels. Histopathological screening validated the abovementioned results and revealed that apigenin restored the distorted cytoarchitecture of the brain cortex. Thus, the present findings collectively suggest that apigenin exerted significant protection against MSG-induced neurotoxicity by enhancing the cellular antioxidant response and attenuating inflammatory machineries in the rat brain cortex.
Collapse
Affiliation(s)
- Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia.
| |
Collapse
|
14
|
Ahlatcı A, Yıldızhan K, Tülüce Y, Bektaş M. Valproic Acid Attenuated PTZ-induced Oxidative Stress, Inflammation, and Apoptosis in the SH-SY5Y Cells via Modulating the TRPM2 Channel. Neurotox Res 2022; 40:1979-1988. [PMID: 36536269 DOI: 10.1007/s12640-022-00622-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
Valproic acid (VPA) is one of the most widely used antiepileptic drugs. The protective role of VPA and the role of the TRPM2 channel in this mechanism in developing neuronal damage due to increased pentylenetetrazol (PTZ)-induced neurotoxicity in SH-SY5Y cells were not clarified. Here, we investigated the role of VPA via modulation of TRPM2 channel on cell death and oxidative neurotoxicity in SH-SY5Y cells. The SH-SY5Y cell toxicity model was constructed by treating SH-SY5Y cells with PTZ. The VPA and TRPM2 channel antagonist N-(p-amylcinnamoyl) anthranilic acid (ACA) were added to prevent neurotoxicity in PTZ-induced SH-SY5Y cells. The role of the VPA and TRPM2 channel was evaluated using an ELISA kit and patch-clamp. Primarily, antioxidant (GSH and GSH-Px) and oxidative stress (MDA and ROS) levels and inflammatory factors (IL-1β, IL-6, and TNF-α) in cells were determined by ELISA kits. Then, TRPM2 channel activation in cells was detected using both the ELISA kit and patch-clamp methods. In addition, apoptosis and cell viability levels in cells were determined by performing PARP1, caspase-3, caspase-9, and CCK-8 assays by ELISA kits. Our results showed that the TRPM2 channel is vital in damage formation in PTZ-induced cells. Furthermore, we observed that VPA attenuated PTZ-induced neurotoxicity by suppressing cells' oxidative stress and inflammation, and reducing TRPM2 channel activation. In our study, in which the protective effect of VPA and the role of the TRPM2 channel in PTZ-induced SH-SY5Y cells were investigated for the first time, we can conclude that VPA treatment and TRPM2 channel blockade can suppress PTZ-induced neurotoxicity.
Collapse
Affiliation(s)
- Adem Ahlatcı
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Kenan Yıldızhan
- Department of Biophysics, Faculty of Medicine, Van Yuzuncu Yil University, TR- 65080, Van, Turkey.
| | - Yasin Tülüce
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Muhammet Bektaş
- Department of Biophysics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
15
|
Moradi Jafari A, Hassanpourezatti M. Influence of methadone on the anticonvulsant efficacy of valproate sodium gabapentin against maximal electroshock seizure in mice by regulation of brain MDA TNF-α. Front Neurol 2022; 13:920107. [PMID: 36081867 PMCID: PMC9445582 DOI: 10.3389/fneur.2022.920107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
Abstract
Methadone is the most frequently used opioid therapy worldwide, with controversial effects on oxidative stress homeostasis. This study investigated the effects of intraperitoneal (i.p.) co-administration of methadone (0.1, 0.3, 1, and 3 mg/kg) and valproate sodium (300 mg/kg) or gabapentin (50 mg/kg) in the mice maximal electroshock (MES)-induced seizure model. The adverse effect of drugs was assessed using the chimney test. The levels of tumor necrosis factor-alpha (TNF-α) and malondialdehyde (MDA) contents were measured in mice brains after a single seizure. Administration of methadone alone resulted in a significant reduction in the duration of hind limb extension (HLE) than that in the control group. Methadone pretreatment at doses of 0.1 and 0.3 mg/kg i.p. decreased, and at doses of 1 and 3 mg/kg i.p. had an increasing effect on anticonvulsant efficacy of gabapentin. Pretreatment with all doses of methadone significantly decreased the valproate anticonvulsive efficacy. At doses of 1 and 3 mg/kg i.p. methadone per se increased brain MDA levels after MES-induced seizure. Administration of methadone (0.3 mg/kg i.p.) enhanced and at 3 mg/kg decreased gabapentin effect on brain MDA level, but their co-treatment did not lead to further increase in MDA. Methadone at 0.3–3 mg/kg enhanced the effect of sodium valproate on MDA levels in the brain, but at all doses significantly potentiated its effect on brain TNF-α levels. The drugs did not produce any side effects on motor coordination in experimental animals. In conclusion, methadone showed different effects on anticonvulsant actions of gabapentin and valproate through regulation of brain levels of MDA and TNF-α.
Collapse
|
16
|
Research progress on oxidative stress regulating different types of neuronal death caused by epileptic seizures. Neurol Sci 2022; 43:6279-6298. [DOI: 10.1007/s10072-022-06302-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/24/2022] [Indexed: 12/09/2022]
|
17
|
Zeng C, Hu J, Chen F, Huang T, Zhang L. The Coordination of mTOR Signaling and Non-Coding RNA in Regulating Epileptic Neuroinflammation. Front Immunol 2022; 13:924642. [PMID: 35898503 PMCID: PMC9310657 DOI: 10.3389/fimmu.2022.924642] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy accounts for a significant proportion of the burden of neurological disorders. Neuroinflammation acting as the inflammatory response to epileptic seizures is characterized by aberrant regulation of inflammatory cells and molecules, and has been regarded as a key process in epilepsy where mTOR signaling serves as a pivotal modulator. Meanwhile, accumulating evidence has revealed that non-coding RNAs (ncRNAs) interfering with mTOR signaling are involved in neuroinflammation and therefore articipate in the development and progression of epilepsy. In this review, we highlight recent advances in the regulation of mTOR on neuroinflammatory cells and mediators, and feature the progresses of the interaction between ncRNAs and mTOR in epileptic neuroinflammation.
Collapse
Affiliation(s)
- Chudai Zeng
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jason Hu
- Department of Neonatology, Yale School of Medicine, New Haven, CT, United States
| | - Fenghua Chen
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| | - Tianxiang Huang
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| | - Longbo Zhang
- Departments of Neurosurgery, and National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Fenghua Chen, ; Tianxiang Huang, ; Longbo Zhang,
| |
Collapse
|
18
|
Karabulut S, Filiz AK, Akkaya R. Thiamine alleviates cognitive impairment and epileptogenesis by relieving brain inflammation in PTZ-induced kindling rat model. Neurol Res 2022; 44:902-909. [PMID: 35446240 DOI: 10.1080/01616412.2022.2066785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Epileptogenesis, the process by which the brain becomes epileptic, is related to neuroinflammation, hyperexcitability cognitive deficits. Evidence suggests that improving brain inflammation can inhibit the epileptogenesis process and help the emergence of new drugs for the treatment of epilepsy. Therefore, the PTZ kindling model of epilepsy was utilized to assess the neuroprotective role of thiamine in epileptogenesis. METHODS Male rats were exposed to PTZ-induced kindling and pretreated with low thiamine (25 mg/kg) or high thiamine (50 mg/kg). Cyclooxygenase (COX-1 and COX-2), interleukin 1-beta (IL-1β), tumor necrosis factor-alpha (TNF-α), and nuclear factor-κB (NF-κB) concentrations in the brain were analyzed using biochemical assays. Cognitive function was evaluated using the passive avoidance test. RESULTS Thiamine ameliorated epileptogenesis and enhanced the rats' performance in the passive avoidance test. Also, thiamine significantly decreased the level of neuroinflammatory mediators in the brain induced by PTZ. CONCLUSION These results provide evidence that thiamine alleviates PTZ-induced neuroinflammation and cognitive impairments.
Collapse
Affiliation(s)
- Sebahattin Karabulut
- Department of Medical Services and Techniques, Vocational School of Health Services, Sivas Cumhuriyet University, Sivas, Turkey
| | - Ahmet Kemal Filiz
- Department of Medical Physiology, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| | - Recep Akkaya
- Department of Biophysics, Faculty of Medicine, Sivas Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
19
|
Bai G, Qiao Y, Lo PC, Song L, Yang Y, Duan L, Wei S, Li M, Huang S, Zhang B, Wang Q, Yang C. Anti-depressive effects of Jiao-Tai-Wan on CORT-induced depression in mice by inhibiting inflammation and microglia activation. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114717. [PMID: 34627986 DOI: 10.1016/j.jep.2021.114717] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 09/29/2021] [Accepted: 10/05/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jiao-Tai-Wan (JTW) is a very famous traditional Chinese medicine formula for the treatment of psychiatric disorders, especially in anxiety, insomnia and depression. However, its molecular mechanism of treatment remains indistinct. AIM OF THE STUDY We aimed to reveal the action mechanism of JTW on anti-depression via inhibiting microglia activation and pro-inflammatory response both in vivo and in vitro. MATERIAL AND METHODS The corticosterone (CORT)-induced depression mouse model was used to evaluate the therapeutic efficacy of JTW. Behavioral tests (open field, elevated plus maze, tail suspension and forced swim test) were conducted to evaluate the effect of JTW on depressive-like behaviors. The levels of inflammatory factors and the concentration of neurotransmitters were detected by RT-qPCR or ELISA assays. Then three hippocampal tissue samples per group (Control, CORT, and JTW group) were sent for RNA sequencing (RNA-seq). Transcriptomics data analysis was used to screen the key potential therapeutic targets and signaling pathways of JTW. Based on 8 bioactive species of JTW by our previous study using High-performance liquid chromatography (HPLC) analysis, molecular docking analyses were used to predict the interaction of JTW-derived compounds and depression targets. Finally, the results of transcriptome and molecular docking analyses were combined to verify the targets, key pathways, and efficacy of JTW treatment in vivo and vitro. RESULTS JTW ameliorated CORT-induced depressive-like behaviors, neuronal damage and enhanced the levels of monoamine neurotransmitters in the serum of mice. JTW also inhibited CORT-induced inflammatory activation of microglia and decreased the serum levels of interleukin- 6(IL-6) and interleukin- 1β (IL-1β) in vivo. Transcriptomic data analysis showed there were 10 key driver analysis (KDA) genes with the strongest correlation which JTW regulated in depression mice. Molecular docking analysis displayed bioactive compound Magnoflorine had the strongest binding force to the key gene colony-stimulating factor 1 receptor (CSF1R), which is the signaling microglia dependent upon for their survival. Meanwhile, CSF1R staining showed it was consistent with inflammatory activation of microglia. Our vitro experiment also showed JTW and CSF1R inhibitor significantly reduced lipopolysaccharide (LPS)/interferon-gamma (IFNɣ)-induced inflammatory activation response in macrophage cells. CONCLUSIONS Our study suggests that JTW might ameliorate CORT-induced neuronal damage in depression mice by inhibiting CSF1R mediated microglia activation and pro-inflammatory response.
Collapse
Affiliation(s)
- Guiqin Bai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Yiqi Qiao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Po-Chieh Lo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Lei Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Yuna Yang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Lining Duan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Sufen Wei
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Min Li
- Clinical Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Shuiqing Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Beiping Zhang
- Guangdong Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Qi Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| | - Cong Yang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
20
|
Wang P, Ma K, Yang L, Zhang G, Ye M, Wang S, Wei S, Chen Z, Gu J, Zhang L, Niu J, Tao S. Predicting signaling pathways regulating demyelination in a rat model of lithium-pilocarpine-induced acute epilepsy: A proteomics study. Int J Biol Macromol 2021; 193:1457-1470. [PMID: 34742844 DOI: 10.1016/j.ijbiomac.2021.10.209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
Demyelination is observed in animal models of intractable epilepsy (IE). Epileptogenesis damages the myelin sheath and dysregulates oligodendrocyte precursor cell (OPC) development. However, the molecular pathways regulating demyelination in epilepsy are unclear. Here, we predicted the molecular mechanisms regulating demyelination in a rat model of lithium-pilocarpine hydrochloride-induced epilepsy. We identified DGKA/Mboat2/Inpp5j and NOS/Keratin 28 as the main target molecules that regulate demyelination via glycerolipid and glycerophospholipid metabolism, phosphatidylinositol signaling, and estrogen signaling in demyelinated forebrain slice cultures (FSCs). In seizure-like FCSs, the actin cytoskeleton was regulated by Cnp and MBP via Pak4/Tmsb4x (also known as Tβ4) and Kif5c/Kntc1. Tβ4 possibly prevented OPC differentiation and maturation and inhibited MBP phosphorylation via the p38MAPK/ERK1/JNK1 pathway. The MAPK signaling pathway was more likely activated in seizure-like FCSs than in demyelinated FCSs. pMBP expression was decreased in the hippocampus of lithium-pilocarpine hydrochloride-induced acute epilepsy rats. The expression of remyelination-related factors was suppressed in the hippocampus and corpus callosum in lithium-pilocarpine hydrochloride-induced epilepsy rats. These findings suggest that the actin cytoskeleton, Tβ4, and MAPK signaling pathways regulate the decrease in pMBP in the hippocampus in a rat model of epilepsy. Our results indicate that regulating the actin cytoskeleton, Tβ4, and MAPK signaling pathways may facilitate the prevention of demyelination in IE.
Collapse
Affiliation(s)
- Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China.
| | - Kang Ma
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Lu Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Guodong Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Mengyi Ye
- Ningxia Medical University College of Traditional Chinese Medicine, Yinchuan 750004, Ningxia, China
| | - Siqi Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Shuangshuang Wei
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Zhangping Chen
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Jinghai Gu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Lianxiang Zhang
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China.
| | - Sun Tao
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China; Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
21
|
Ergül M, Taşkıran AŞ. Thiamine Protects Glioblastoma Cells against Glutamate Toxicity by Suppressing Oxidative/Endoplasmic Reticulum Stress. Chem Pharm Bull (Tokyo) 2021; 69:832-839. [PMID: 34470947 DOI: 10.1248/cpb.c21-00169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Thiamine (vitamin B1), which is synthesized only in bacteria, fungi and plants and which humans should take with diet, participates in basic biochemical and physiological processes in a versatile way and its deficiency is associated with neurological problems accompanied by cognitive dysfunctions. The rat glioblastoma (C6) model was used, which was exposed to a limited environment and toxicity with glutamate. The cells were stressed by exposure to glutamate in the presence and absence of thiamine. The difference in cell proliferation was evaluated in the XTT assay. Oxidative stress (OS) markers malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) levels, as well as endoplasmic reticulum (ER) stress markers 78-kDa glucose-regulated protein (GRP78), activating transcription factor-4 (ATF-4), and C/EBP homologous protein (CHOP) levels, were measured with commercial kits. Apoptosis determined by flow cytometry was confirmed by 4',6-diamidino-2-phenylindole (DAPI) staining. At all concentrations, thiamine protects the cells and increased the viability against glutamate-induced toxicity. Thiamine also significantly decreased the levels of MDA, while increasing SOD and CAT levels. Moreover, thiamine reduced ER stress proteins' levels. Moreover, it lessened the apoptotic cell amount and enhanced the live-cell percentage in the flow cytometry and DAPI staining. As a result, thiamine may be beneficial nutritional support for individuals with a predisposition to neurodegenerative disorders due to its protective effect on glutamate cytotoxicity in glioblastoma cells by suppressing OS and ER stress.
Collapse
Affiliation(s)
- Merve Ergül
- Department of Pharmacology, Faculty of Pharmacy, Sivas Cumhuriyet University
| | | |
Collapse
|