1
|
Atasever A, Tekin S, Bolat İ, Bolat M, Dağ Y, ÇınaR B, Şengül E, Yıldırım S, Warda M, Çelebi F. The effects of melatonin on oxidative stress, inflammation, apoptosis and Nrf2/HO-1 in acrylamide-induced lung injury in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04292-8. [PMID: 40402233 DOI: 10.1007/s00210-025-04292-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025]
Abstract
This study is to investigate the effects of melatonin on lung inflammation, oxidative stress, apoptosis, tissue damage, and MT1 and MT2 receptors in acrylamide-induced lung toxicity. Fifty male rats were randomly divided into five groups. The control group received distilled water orally for 11 days, while the acrylamide group received acrylamide (50 mg/kg, i.g.) for 11 days. The MEL10 + ACR and MEL20 + ACR groups received intraperitoneal injections of melatonin at doses of 10 mg/kg and 20 mg/kg, respectively, followed by acrylamide (50 mg/kg, i.g.) administered 1 h after melatonin injection. The MEL20 group received melatonin injections (20 mg/kg) for 11 days. Lung tissues collected at the end of the study underwent biochemical, histopathological, immunohistochemical, immunofluorescence, and in silico analyses. Acrylamide caused oxidative stress, inflammation, apoptosis, and tissue damage in the lungs. Melatonin treatment alleviated acrylamide-induced lung damage by exhibiting antioxidant, anti-inflammatory, and anti-apoptotic effects. Melatonin significantly improved the histopathological changes caused by acrylamide in lung tissue. Melatonin may have protective effects on health by regulating cellular processes such as oxidative stress, antioxidant enzyme activity, inflammation, and apoptosis through MT1 and MT2 receptors. Melatonin mitigates oxidative stress, inflammation, apoptosis, and tissue damage in acrylamide-induced lung injury in rats.
Collapse
Affiliation(s)
- Aslıhan Atasever
- Department of Veterinary Medicine, Çayırlı Vocational High School, Erzincan University, Erzincan, Turkey
| | - Samet Tekin
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - İsmail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Merve Bolat
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey.
| | - Yusuf Dağ
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Burak ÇınaR
- Department of Pharmacology, Atatürk University Faculty of Medicine, Erzurum, Turkey
| | - Emin Şengül
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Serkan Yıldırım
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
- Department of Pathology, Faculty of Veterinary Medicine, Kyrgyzs-Turkish Manas University, Bishkek, Kyrgyzstan
| | - Mohamad Warda
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
- Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Fikret Çelebi
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
2
|
Salkin H, Satir-Basaran G, Korkmaz S, Burcin Gonen Z, Erdem Basaran K. Mesenchymal stem cell-derived conditioned medium and Methysergide give rise to crosstalk inhibition of 5-HT2A and 5-HT7 receptors in neuroblastoma cells. Brain Res 2023; 1808:148354. [PMID: 36997105 DOI: 10.1016/j.brainres.2023.148354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/04/2022] [Accepted: 03/26/2023] [Indexed: 03/30/2023]
Abstract
OBJECTIVE (s): We aimed to investigate the effects of mesenchymal stem cell secretome and methysergide combination on 5-hydroxytryptamine 2A, (5-HT2AR), 5-hydroxytryptamine 7 (5-HT7R), adenosine 2A (A2AR) receptors and CD73 on neuroblastoma cell line and how they affect biological characteristics. Methysergide was used as a serotonin antagonist on the neuroblastoma cells. MATERIALS AND METHODS Human dental pulp-derived stem cells (hDPSCs) used to obtain conditioned medium (CM). Methysergide drug was prepared in CM and applied to neuroblastoma cells. Analysis of 5-HT7R, 5-HT2AR, A2AR and CD73 expressions was performed by western blot and immunofluorescence staining. Total apoptosis, mitochondrial membrane depolarization, Ki-67 proliferation test, viability analysis, DNA damage and cell cycle analysis were performed in accordance with the product procedure by using biological activity test kits. RESULTS Our results showed that neuroblastoma cancer cells are normally on the Gs signaling axis via the serotonin 7 receptor and the adenosine 2A receptor. CM and Methysergide inhibited the 5-HT7 and A2A receptor levels in neuroblastoma cells. We found that CM and methysergide formed crosstalk inhibition between 5-HT2AR, 5-HT7R, A2AR and CD73. CM and Methysergide increased the total apoptosis in neuroblastoma cells and induced the mitochondrial membrane depolarization. CM and Methysergide induced the DNA damage and arrested in G0/G1 phase of cell cycle of the neuroblastoma cells. CONCLUSION These findings suggest that the combination of CM and methysergite may exert a therapeutic effect on neuroblastoma cancer cells, and future in vivo studies may be important in area of neuroblastoma research to support the findings.
Collapse
Affiliation(s)
- Hasan Salkin
- Beykent University, Vocational School, Department of Medical Services and Techniques, Program of Pathology Laboratory Techniques, Istanbul, Turkey; Erciyes University, Genome and Stem Cell Center, Kayseri, Turkey.
| | - Guzide Satir-Basaran
- Erciyes University, Faculty of Pharmacy, Department of Biochemistry, Kayseri, Turkey
| | - Seyda Korkmaz
- Erciyes University, Genome and Stem Cell Center, Kayseri, Turkey
| | - Zeynep Burcin Gonen
- Erciyes University, Genome and Stem Cell Center, Kayseri, Turkey; Erciyes University, Faculty of Dentistry, Department of Oral and Maxillofacial Surgery, Kayseri, Turkey
| | - Kemal Erdem Basaran
- Erciyes University, Faculty of Medicine, Department of Physiology, Kayseri, Turkey
| |
Collapse
|
3
|
Xie L, Luo M, Li J, Huang W, Tian G, Chen X, Ai Y, Zhang Y, He H, Jinyang He. Gastroprotective mechanism of modified lvdou gancao decoction on ethanol-induced gastric lesions in mice: Involvement of Nrf-2/HO-1/NF-κB signaling pathway. Front Pharmacol 2022; 13:953885. [PMID: 36120337 PMCID: PMC9475313 DOI: 10.3389/fphar.2022.953885] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/09/2022] [Indexed: 11/13/2022] Open
Abstract
Modified Lvdou Gancao decoction (MLG), a traditional Chinese medicine formula, has been put into clinical use to treat the diseases of the digestive system for a long run, showing great faculty in gastric protection and anti-inflammatory, whereas its protective mechanisms have not been determined. The current study puts the focus on the protective effect and its possible mechanisms of MLG on ethanol-induced gastric lesions in mice. In addition to various gastric lesion parameters and histopathology analysis, the activities of a list of relevant indicators in gastric mucosa were explored including ALDH, ADH, MDA, T-SOD, GSH-Px, and MPO, and the mechanisms were clarified using RT-qPCR, ELISA Western Blot and immunofluorescence staining. The results showed that MLG treatment induced significant increment of ADH, ALDH, T-SOD, GSH-Px, NO, PGE2 and SS activities in gastric tissues, while MPO, MDA, TNF-α and IL-1β levels were on the decline, both in a dose-dependent manner. In contrast to the model group, the mRNA expression of Nrf-2 and HO-1 in the MLG treated groups showed an upward trend while the NF-κB, TNFα, IL-1β and COX2 in the MLG treated groups had a downward trend simultaneously. Furthermore, the protein levels of p65, p-p65, IκBα, p-IκBα, iNOS, COX2 and p38 were inhibited, while Nrf2, HO-1, SOD1, SOD2 and eNOS were ramped up in MLG treatment groups. Immunofluorescence intensities of Nrf2 and HO-1 in the MLG treated groups were considerably enhanced, with p65 and IκBα diminished simultaneously, exhibiting similar trends to that of qPCR and western blot. To sum up, MLG could significantly ameliorate ethanol-induced gastric mucosal lesions in mice, which might be put down to the activation of alcohol metabolizing enzymes, attenuation of the oxidative damage and inflammatory response to maintain the gastric mucosa. The gastroprotective effect of MLG might be achieved through the diminution of damage factors and the enhancement of defensive factors involving NF-κB/Nrf2/HO-1 signaling pathway. We further confirmed that MLG has strong potential in preventing and treating ethanol-induced gastric lesions.
Collapse
Affiliation(s)
- Lei Xie
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Minyi Luo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Junlin Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wenguan Huang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Guangjun Tian
- Liver Diseases Center, Guangdong Provincial Hospital of Chinese Medicine, Zhuhai, Guangdong, China
| | - Xiuyun Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Ai
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yan Zhang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Haolan He
- Guangzhou Eighth People’s Hospital, Guangzhou, Guangdong, China
| | - Jinyang He
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
AL-Nasser MN, Mellor IR, Carter WG. Is L-Glutamate Toxic to Neurons and Thereby Contributes to Neuronal Loss and Neurodegeneration? A Systematic Review. Brain Sci 2022; 12:577. [PMID: 35624964 PMCID: PMC9139234 DOI: 10.3390/brainsci12050577] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023] Open
Abstract
L-glutamate (L-Glu) is a nonessential amino acid, but an extensively utilised excitatory neurotransmitter with critical roles in normal brain function. Aberrant accumulation of L-Glu has been linked to neurotoxicity and neurodegeneration. To investigate this further, we systematically reviewed the literature to evaluate the effects of L-Glu on neuronal viability linked to the pathogenesis and/or progression of neurodegenerative diseases (NDDs). A search in PubMed, Medline, Embase, and Web of Science Core Collection was conducted to retrieve studies that investigated an association between L-Glu and pathology for five NDDs: Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Together, 4060 studies were identified, of which 71 met eligibility criteria. Despite several inadequacies, including small sample size, employment of supraphysiological concentrations, and a range of administration routes, it was concluded that exposure to L-Glu in vitro or in vivo has multiple pathogenic mechanisms that influence neuronal viability. These mechanisms include oxidative stress, reduced antioxidant defence, neuroinflammation, altered neurotransmitter levels, protein accumulations, excitotoxicity, mitochondrial dysfunction, intracellular calcium level changes, and effects on neuronal histology, cognitive function, and animal behaviour. This implies that clinical and epidemiological studies are required to assess the potential neuronal harm arising from excessive intake of exogenous L-Glu.
Collapse
Affiliation(s)
- Maryam N. AL-Nasser
- Department of Biological Sciences, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia;
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| | - Ian R. Mellor
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Wayne G. Carter
- School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK
| |
Collapse
|
5
|
Sallam AAM, Darwish SF, El-Dakroury WA, Radwan E. Olmesartan niosomes ameliorates the Indomethacin-induced gastric ulcer in rats: Insights on MAPK and Nrf2/HO-1 signaling pathway. Pharm Res 2021; 38:1821-1838. [PMID: 34853982 DOI: 10.1007/s11095-021-03126-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/13/2021] [Indexed: 02/07/2023]
Abstract
AIMS Gastric ulcer is a continuous worldwide threat that inquires protective agents. Olmesartan (OLM) has potent anti-oxidant and anti-inflammatory characters, yet having limited bioavailability. We targeted the gastro-protective potential and probable mechanism of OLM and its niosomal form against indomethacin (IND) induced-gastric ulcer in rats. MAIN METHODS we prepared OLM niosomes (OLM-NIO) with different surfactant: cholesterol molar ratios. We evaluated particle size, zeta-potential, polydispersity, and entrapment efficiency. In-vitro release study, Fourier transform infrared spectroscopy, differential scanning calorimetry, and transmission electron microscopy were performed for selected niosomes. In-vivo, we used oral Omeprazole (30 mg/kg), OLM or OLM-NIO (10 mg/kg) for 3 days before IND (25 mg/kg) ingestion. We assessed gastric lesions, oxidative and inflammatory markers. KEY FINDINGS OLM-NIO prepared with span 60:cholesterol ratio (1:1) showed high entrapment efficiency 93 ± 2%, small particle size 159.3 ± 6.8 nm, low polydispersity 0.229 ± 0.009, and high zeta-potential -35.3 ± 1.2 mV, with sustained release mechanism by release data. In-vivo macroscopical and histological results showed gastro-protective effects of OLM pretreatment, which improved oxidative stress parameters and enhanced the gastric mucosal cyclooxygenase-1 (COX-1) and prostaglandin E2 (PGE2) contents. OLM pretreatment suppressed interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) contents and translocation of p38 mitogen-activated protein kinase (p38-MAPK). Besides, OLM substantially promoted the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) protective pathway. OLM-NIO furtherly improved all previous outcomes. SIGNIFICANCE We explored OLM anti-ulcerative effects, implicating oxidative stress and inflammation improvement, mediated by the Nrf2/HO-1 signaling pathway and p38-MAPK translocation. Meanwhile, the more bioavailable OLM-NIO achieved better gastro-protective effects compared to conventional OLM form.
Collapse
Affiliation(s)
- Al-Aliaa M Sallam
- Biochemistry Department, Faculty of Pharmacy, Ain-Shams University, Abassia, Cairo, 11566, Egypt
- Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Samar F Darwish
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt.
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo, 11829, Egypt
| | - Eman Radwan
- Department of Medical Biochemistry, Faculty of Medicine, Assiut University, 71515, Assiut, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Sphinx University, New Assiut City, Assiut 10, Egypt
| |
Collapse
|
6
|
Castañeda-Cabral JL, López-Ortega JG, Fajardo-Fregoso BF, Beas-Zárate C, Ureña-Guerrero ME. Glutamate induced neonatal excitotoxicity modifies the expression level of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins in various brain regions of the adult rat. Neurosci Lett 2020; 735:135237. [PMID: 32645399 DOI: 10.1016/j.neulet.2020.135237] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 07/03/2020] [Accepted: 07/04/2020] [Indexed: 02/08/2023]
Abstract
Glutamate-mediated excitatory synaptic signalling is primarily controlled by excitatory amino acid transporters (EAATs), such as EAAT1 and EAAT2, which are located mostly on astrocytes and, together, uptake more than 95 % of extracellular glutamate. Alterations in the functional expression levels of EAATs can lead to excessive extracellular glutamate accumulation, potentially triggering excitotoxicity and seizures, among other neurological disorders. Excitotoxicity induced in early developmental stages can lead to lasting changes in several neurotransmission systems, including the glutamatergic system, which could make the brain more susceptible to a second insult. In this study, the expression levels of EAAT1 (GLAST) and EAAT2 (GLT-1) proteins were assessed in the cerebral motor cortex (CMC), striatum, hippocampus and entorhinal cortex (EC) of male adult rats following the neonatal excitotoxic process triggered by monosodium glutamate (MSG)-treatment (4 g/kg of body weight at postnatal days 1,3,5 and 7, subcutaneously). Western blot analysis showed that neonatal MSG-treatment decreased EAAT1 expression levels in the CMC, striatum and hippocampus, while EAAT2 levels were increased in the striatum and EC and decreased in the CMC. Immunofluorescence staining confirmed the changes in EAAT1 and EAAT2 expression induced by neonatal MSG-treatment, which were accompanied by an increase in the glial fibrillary acidic protein (GFAP) immunofluorescence signalthat was particularly significant in the hippocampus. Our results show that a neonatal excitotoxic processes can induce lasting changes in the expression levels of EAAT1 and EAAT2 proteins and suggest that although astrogliosis occurs, glutamate uptake could be deficient, particularly in the CMC and hippocampus.
Collapse
Affiliation(s)
- José Luis Castañeda-Cabral
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - José Guadalupe López-Ortega
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Blanca Fabiola Fajardo-Fregoso
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Carlos Beas-Zárate
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Mónica E Ureña-Guerrero
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico.
| |
Collapse
|
7
|
Yuksel TN, Yayla M, Halici Z, Cadirci E, Polat B, Kose D. Protective effect of 5-HT7 receptor activation against glutamate-induced neurotoxicity in human neuroblastoma SH-SY5Y cells via antioxidative and antiapoptotic pathways. Neurotoxicol Teratol 2019; 72:22-28. [PMID: 30685503 DOI: 10.1016/j.ntt.2019.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 12/23/2018] [Accepted: 01/17/2019] [Indexed: 12/29/2022]
Abstract
Serotonin exerts anti-inflammatory, antioxidant and antiapoptotic effects through 5-HT7 receptors. The present study determined the role of 5-HT7 receptors in glutamate-induced neurotoxicity by using human SH-SY5Y neuroblastoma cells. The cells were pretreated with different concentrations of 5-HT7 receptor agonist LP44 and antagonist SB269970 for 60 min, followed by treatment with glutamate. Cell proliferation was measured using xCELLigence system. Treatment with all the concentrations of LP44 significantly protected the cells from the toxic effects of glutamate after 24, 48 and 72 h. Although 5-HT7 receptor expression was significantly upregulated in glutamate-treated cells, it was downregulated in LP44-pretreated cells. Furthermore, LP44 treatment significantly decreased malondialdehyde levels and increased superoxide dismutase activities and glutathione levels. Moreover, LP44 treatment significantly decreased tumor necrosis factor alpha (TNF-α) levels and inhibited caspase 3 and caspase 9 mRNA expression. In contrast, SB269970 treatment exerted an insignificant effect on oxidative stress, inflammation and apoptosis. These findings suggest that exogenous stimulation of the 5-HT7 receptors may be protective in glutamate-induced neurotoxicity and that 5-HT7 receptor agonists can be used as therapeutic agents for preventing glutamate-induced neurological disorders.
Collapse
Affiliation(s)
- Tugba Nurcan Yuksel
- Namık Kemal University, Faculty of Medicine, Department of Pharmacology, Tekirdag, Turkey
| | - Muhammed Yayla
- Kafkas University, Faculty of Medicine, Department of Pharmacology, Kars, Turkey
| | - Zekai Halici
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey.
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Beyzagul Polat
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, Erzurum, Turkey
| | - Duygu Kose
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| |
Collapse
|
8
|
Rivera-Carvantes MC, Jarero-Basulto JJ, Feria-Velasco AI, Beas-Zárate C, Navarro-Meza M, González-López MB, Gudiño-Cabrera G, García-Rodríguez JC. Changes in the expression level of MAPK pathway components induced by monosodium glutamate-administration produce neuronal death in the hippocampus from neonatal rats. Neuroscience 2017; 365:57-69. [PMID: 28954212 DOI: 10.1016/j.neuroscience.2017.09.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 09/12/2017] [Accepted: 09/17/2017] [Indexed: 11/18/2022]
Abstract
Excessive Glutamate (Glu) release may trigger excitotoxic cellular death by the activation of intracellular signaling pathways that transduce extracellular signals to the cell nucleus, which determines the onset of a death program. One such signaling pathway is the mitogen-activated protein kinases (MAPK), which is involved in both survival and cell death. Experimental evidences from the use of specific inhibitors supports the participation of some MAPK pathway components in the excitotoxicity mechanism, but the complete process of this activation, which terminates in cell damage and death, is not clearly understood. The present work, we investigated the changes in the expression level of some MAPK-pathway components in hippocampal excitotoxic cell death in the neonatal rats using an experimental model of subcutaneous monosodium glutamate (MSG) administration on postnatal days (PD) 1, 3, 5 and 7. Data were collected at different ages through PD 14. Cell viability was evaluated using fluorescein diacetate mixed with propidium iodide (FDA-PI), and the Nissl-staining technique was used to evaluate histological damage. Transcriptional changes were also investigated in 98 components of the MAPK pathway that are associated with cell damage. These results are an evidence of that repetitive use of MSG, in neonatal rats, induces cell damage-associated transcriptional changes of MAPK components, that might reflect a differential stage of both biochemical and molecular brain maturation. This work also suggests that some of the proteins evaluated such as phosphorylated retinoblastoma (pRb) protein, which was up-regulated, could regulate the response to excitotoxic through modulation of the process of re-entry into the cell cycle in the hippocampus of rats treated with MSG.
Collapse
Affiliation(s)
- Martha Catalina Rivera-Carvantes
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico.
| | - José Jaime Jarero-Basulto
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Alfredo Ignacio Feria-Velasco
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Carlos Beas-Zárate
- Regeneration and Neural Development Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Mónica Navarro-Meza
- Department of Health and Wellness, CUSur, University of Guadalajara, Ciudad Guzman, Jal., Mexico
| | - Mariana Berenice González-López
- Cellular Neurobiology Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | - Graciela Gudiño-Cabrera
- Regeneration and Neural Development Laboratory, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Zapopan, Jal., Mexico
| | | |
Collapse
|
9
|
Castañeda-Cabral JL, Beas-Zarate C, Gudiño-Cabrera G, Ureña-Guerrero ME. Glutamate Neonatal Excitotoxicity Modifies VEGF-A, VEGF-B, VEGFR-1 and VEGFR-2 Protein Expression Profiles During Postnatal Development of the Cerebral Cortex and Hippocampus of Male Rats. J Mol Neurosci 2017; 63:17-27. [PMID: 28755050 DOI: 10.1007/s12031-017-0952-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
Vascular endothelial growth factor (VEGF) exerts both neuroprotective and proinflammatory effects in the brain, depending on the VEGF (A-E) and VEGF receptor (VEGFR1-3) types involved. Neonatal monosodium glutamate (MSG) treatment triggers an excitotoxic degenerative process associated with several neuropathological conditions, and VEGF messenger RNA (mRNA) expression is increased at postnatal day (PD) 14 in rat hippocampus (Hp) following the treatment. The aim of this work was to establish the changes in immunoreactivity to VEGF-A, VEGF-B, VEGFR-1 and VEGFR-2 proteins induced by neonatal MSG treatment (4 g/kg, subcutaneous, at PD1, 3, 5 and 7) in the cerebral motor cortex (CMC) and Hp. Samples collected from PD2 to PD60 from control and MSG-treated male Wistar rats were assessed by western blotting for each protein. Considering that immunoreactivity measured by western blotting is related to the protein expression level, we found that each protein in each cerebral region has a specific expression profile throughout the studied ages, and all profiles were differentially modified by MSG. Specifically, neonatal MSG treatment significantly increased the immunoreactivity to the following: (1) VEGF-A at PD8-PD10 in the CMC and at PD6-PD8 in the Hp; (2) VEGF-B at PD2, PD6 and PD10 in the CMC and at PD8-PD9 in the Hp; and (3) VEGFR-2 at PD6-PD8 in the CMC and at PD21-PD60 in the Hp. Also, MSG significantly reduced the immunoreactivity to the following: (1) VEGF-B at PD8-PD9 and PD45-PD60 in the CMC; and (2) VEGFR-1 at PD4-PD6 and PD14-PD21 in the CMC and at PD4, PD9-PD10 and PD60 in the Hp. Our results indicate that VEGF-mediated signalling is involved in the excitotoxic process triggered by neonatal MSG treatment and should be further characterized.
Collapse
Affiliation(s)
- Jose Luis Castañeda-Cabral
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Carlos Beas-Zarate
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico. .,Laboratorio de Regeneración y Desarrollo Neural, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Km 15.5 Carretera a Nogales, Camino Ing. Ramón Padilla Sánchez Km 2, 45221, Zapopan, Jalisco, Mexico.
| | - Graciela Gudiño-Cabrera
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico
| | - Monica E Ureña-Guerrero
- Departamento de Biología Celular y Molecular, Centro Universitario de Ciencias Biológicas y Agropecuarias (CUCBA), Universidad de Guadalajara, Zapopan, Jalisco, Mexico. .,Laboratorio de Biología de la Neurotransmisión, Edificio de Posgrado, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Km 15.5 Carretera a Nogales, Camino Ing. Ramón Padilla Sánchez Km 2, 45221, Zapopan, Jalisco, Mexico.
| |
Collapse
|
10
|
Protective effects of tropisetron on cerulein-induced acute pancreatitis in mice. Biomed Pharmacother 2017; 93:589-595. [PMID: 28686973 DOI: 10.1016/j.biopha.2017.06.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) causes morbidity and mortality. The aim of the present study was to investigate the protective effect of tropisetron against AP induced by cerulein. Cerulein (50μg/kg, 5 doses) was used to induce AP in mice. Six hours after final cerulein injection, animals were decapitated. Hepatic/pancreatic enzymes in the serum, pancreatic content of malondialdehyde (MDA), pro-inflammatory cytokines and myeloperoxidase (MPO) activity were measured. Tropisetron significantly attenuated pancreatic injury markers and decreased the amount of elevated serum amylase, lipase, alanine aminotransferase (ALT), aspartate aminotransferase (AST), MPO activities and pro-inflammatory cytokines levels caused by AP in mice. Tropisetron didn't affect the pancreatic levels of MDA. Our results suggest that tropisetron could attenuate cerulein-induced AP by combating inflammatory signaling. Further clinical studies are needed to confirm its efficacy in patients with AP.
Collapse
|
11
|
Tanshinone IIA Inhibits Glutamate-Induced Oxidative Toxicity through Prevention of Mitochondrial Dysfunction and Suppression of MAPK Activation in SH-SY5Y Human Neuroblastoma Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4517486. [PMID: 28690763 PMCID: PMC5485345 DOI: 10.1155/2017/4517486] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/17/2017] [Accepted: 05/02/2017] [Indexed: 01/27/2023]
Abstract
Glutamate excitotoxicity is associated with many neurological diseases, including cerebral ischemia and neurodegenerative diseases. Tanshinone IIA, a diterpenoid naphthoquinone from Salvia miltiorrhiza, has been shown to suppress presynaptic glutamate release, but its protective mechanism against glutamate-induced neurotoxicity is lacking. Using SH-SY5Y human neuroblastoma cells, we show here that excessive glutamate exposure decreases cell viability and proliferation and increases LDH release. Pretreatment with tanshinone IIA, however, prevents the decrease in cell viability and proliferation and the increase in LDH release induced by glutamate. Tanshinone IIA also attenuates glutamate-induced oxidative stress by reducing reactive oxygen species level and malondialdehyde and protein carbonyl contents and by enhancing activities and protein levels of superoxide dismutase and catalase. We then show that tanshinone IIA prevents glutamate-induced mitochondrial dysfunction by increasing mitochondrial membrane potential and ATP content and by reducing mitochondrial protein carbonyl content. Moreover, tanshinone IIA can inhibit glutamate-induced apoptosis through regulation of apoptosis-related protein expression and MAPK activation, including elevation of Bcl-2 protein level, decrease in Bax and cleaved caspase-3 levels, and suppression of JNK and p38 MAPK activation. Collectively, our findings demonstrate that tanshinone IIA protects SH-SY5Y cells against glutamate toxicity by reducing oxidative stress and regulating apoptosis and MAPK pathways.
Collapse
|
12
|
Shah SZA, Zhao D, Hussain T, Yang L. The Role of Unfolded Protein Response and Mitogen-Activated Protein Kinase Signaling in Neurodegenerative Diseases with Special Focus on Prion Diseases. Front Aging Neurosci 2017; 9:120. [PMID: 28507517 PMCID: PMC5410568 DOI: 10.3389/fnagi.2017.00120] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 04/12/2017] [Indexed: 12/25/2022] Open
Abstract
Prion diseases are neurodegenerative pathologies characterized by the accumulation of a protease-resistant form of the cellular prion protein named prion protein scrapie (PrPSc) in the brain. PrPSc accumulation in the endoplasmic reticulum (ER) result in a dysregulated calcium (Ca2+) homeostasis and subsequent initiation of unfolded protein response (UPR) leading to neuronal dysfunction and apoptosis. The molecular mechanisms for the transition between adaptation to ER stress and ER stress-induced apoptosis are still unclear. Mitogen-activated protein kinases (MAPKs) are serine/threonine protein kinases that rule the signaling of many extracellular stimuli from plasma membrane to the nucleus. However the identification of numerous points of cross talk between the UPR and MAPK signaling pathways may contribute to our understanding of the consequences of ER stress in prion diseases. Indeed the MAPK signaling network is known to regulate cell cycle progression and cell survival or death responses following a variety of stresses including misfolded protein response stress. In this article, we review the UPR signaling in prion diseases and discuss the triad of MAPK signaling pathways. We also describe the role played by MAPK signaling cascades in Alzheimer’s (AD) and Parkinson’s disease (PD). We will also overview the mechanisms of cell death and the role of MAPK signaling in prion disease progression and highlight potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural UniversityBeijing, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural UniversityBeijing, China
| | - Tariq Hussain
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural UniversityBeijing, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural UniversityBeijing, China
| |
Collapse
|
13
|
N-Adamantyl-4-Methylthiazol-2-Amine Attenuates Glutamate-Induced Oxidative Stress and Inflammation in the Brain. Neurotox Res 2017; 32:107-120. [PMID: 28285348 DOI: 10.1007/s12640-017-9717-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/25/2017] [Accepted: 02/28/2017] [Indexed: 12/15/2022]
Abstract
In this study, we explored the possible mechanisms underlying the neuroprotective and anti-oxidative effects of N-adamantyl-4-methylthiazol-2-amine (KHG26693) against in vivo glutamate-induced toxicity in the rat cerebral cortex. Our results showed that pretreatment with KHG26693 significantly attenuated glutamate-induced elevation of lipid peroxidation, tumor necrosis factor-α, interferon gamma, IFN-γ, interleukin-1β, nitric oxide, reactive oxygen species, NADPH oxidase, caspase-3, calpain activity, and Bax. Furthermore, KHG26693 pretreatment attenuated key antioxidant parameters such as levels of superoxide dismutase, catalase, glutathione, and glutathione reductase. KHG26693 also attenuated the protein levels of inducible nitric oxide synthase, neuronal nitric oxide synthase, nuclear factor erythroid 2-related factor 2, heme oxygenase-1, and glutamate cysteine ligase catalytic subunit caused by glutamate toxicity. Finally, KHG26693 mitigated glutamate-induced changes in mitochondrial ATP level and cytochrome oxidase c. Thus, KHG26693 functions as neuroprotective and anti-oxidative agent against glutamate-induced toxicity through its antioxidant and anti-inflammatory activities in rat brain at least in part.
Collapse
|
14
|
Sadek K, Abouzed T, Nasr S. Lycopene modulates cholinergic dysfunction, Bcl-2/Bax balance, and antioxidant enzymes gene transcripts in monosodium glutamate (E621) induced neurotoxicity in a rat model. Can J Physiol Pharmacol 2016; 94:394-401. [DOI: 10.1139/cjpp-2015-0388] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The effect of monosodium glutamate (MSG) on brain tissue and the relative ability of lycopene to avert these neurotoxic effects were investigated. Thirty-two male Wistar rats were distributed into 4 groups: group I, untreated (placebo); group II, injected with MSG (5 mg·kg−1) s.c.; group III, gastrogavaged with lycopene (10 mg·kg−1) p.o.; and group IV received MSG with lycopene with the same mentioned doses for 30 days. The results showed that MSG induced elevation in lipid peroxidation marker and perturbation in the antioxidant homeostasis and increased the levels of brain and serum cholinesterase (ChE), total creatine phosphokinase (CPK), creatine phosphokinase isoenzymes BB (CPK-BB), and lactate dehydrogenase (LDH). Glutathione S-transferase (GST), superoxide dismutase (SOD), and catalase (CAT) activities and gene expression were increased and glutathione content was reduced in the MSG-challenged rats, and these effects were ameliorated by lycopene. Furthermore, MSG induced apoptosis in brain tissues reflected in upregulation of pro-apoptotic Bax while lycopene upregulated the anti-apoptotic Bcl-2. Our results indicate that lycopene appears to be highly effective in relieving the toxic effects of MSG by inhibiting lipid peroxidation and inducing modifications in the activity of cholinesterase and antioxidant pathways. Interestingly, lycopene protects brain tissue by inhibiting apoptosis signaling induced by MSG.
Collapse
Affiliation(s)
- Kadry Sadek
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Al-Buhiyra Governorate, Egypt
| | - Tarek Abouzed
- Department of Biochemistry, Faculty of Veterinary Medicine, Kafr El-sheikh University, Egypt
| | - Sherif Nasr
- Department of Veterinary Genetics and Molecular Biology, Faculty of Veterinary Medicine, Damanhour University, Egypt
| |
Collapse
|
15
|
Fan LY, Wang ZC, Wang P, Lan YY, Tu L. Exogenous nerve growth factor protects the hypoglossal nerve against crush injury. Neural Regen Res 2016; 10:1982-8. [PMID: 26889186 PMCID: PMC4730822 DOI: 10.4103/1673-5374.172316] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Studies have shown that sensory nerve damage can activate the p38 mitogen-activated protein kinase (MAPK) pathway, but whether the same type of nerve injury after exercise activates the p38MAPK pathway remains unclear. Several studies have demonstrated that nerve growth factor may play a role in the repair process after peripheral nerve injury, but there has been little research focusing on the hypoglossal nerve injury and repair. In this study, we designed and established rat models of hypoglossal nerve crush injury and gave intraperitoneal injections of exogenous nerve growth factor to rats for 14 days. p38MAPK activity in the damaged neurons was increased following hypoglossal nerve crush injury; exogenous nerve growth factor inhibited this increase in acitivity and increased the survival rate of motor neurons within the hypoglossal nucleus. Under transmission electron microscopy, we found that the injection of nerve growth factor contributed to the restoration of the morphology of hypoglossal nerve after crush injury. Our experimental findings indicate that exogenous nerve growth factor can protect damaged neurons and promote hypoglossal nerve regeneration following hypoglossal nerve crush injury.
Collapse
Affiliation(s)
- Li-Yuan Fan
- Department of Prosthodontics, Stomatological Hospital of Sichuan Medical University, Luzhou, Sichuan Province, China; Orofacial Reconstruction and Regeneration Laboratory, Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Zhong-Chao Wang
- Orofacial Reconstruction and Regeneration Laboratory, Sichuan Medical University, Luzhou, Sichuan Province, China; Department of Endodontics, Stomatological Hospital of Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Pin Wang
- Department of Prosthodontics, Stomatological Hospital of Sichuan Medical University, Luzhou, Sichuan Province, China; Orofacial Reconstruction and Regeneration Laboratory, Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Yu-Yan Lan
- Department of Prosthodontics, Stomatological Hospital of Sichuan Medical University, Luzhou, Sichuan Province, China; Orofacial Reconstruction and Regeneration Laboratory, Sichuan Medical University, Luzhou, Sichuan Province, China
| | - Ling Tu
- Department of Anatomy and Physiology, College of Stomatology, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
16
|
Roux C, Aligny C, Lesueur C, Girault V, Brunel V, Ramdani Y, Genty D, Driouich A, Laquerrière A, Marret S, Brasse-Lagnel C, Gonzalez BJ, Bekri S. NMDA receptor blockade in the developing cortex induces autophagy-mediated death of immature cortical GABAergic interneurons: An ex vivo and in vivo study in Gad67-GFP mice. Exp Neurol 2015; 267:177-93. [PMID: 25795167 DOI: 10.1016/j.expneurol.2015.02.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 01/14/2015] [Accepted: 02/05/2015] [Indexed: 01/16/2023]
Abstract
In neonates, excitotoxicity is a major process involved in hypoxic-ischemic brain lesions, and several research groups have suggested the use of NMDA antagonists for neuroprotection. However, despite their clinical interest, there is more and more evidence suggesting that, in the immature brain, these molecules exert deleterious actions on migrating GABAergic interneurons by suppressing glutamatergic trophic inputs. Consequently, preventing the side effects of NMDA antagonists would be therapeutically useful. Because macroautophagy is involved in the adaptive response to trophic deprivation, the aim of the present study was to investigate the impact of autophagy modulators on the MK801-induced death of immature GABAergic interneurons and to characterize the crosstalk between autophagic and apoptotic mechanisms in this cell type. Ex vivo, using cortical slices from NMRI and Gad67-GFP mice, we show that blockade of the NMDA receptor results in an accumulation of autophagosomes due to the disruption of the autophagic flux. This effect precedes the activation of the mitochondrial apoptotic pathway, and the degeneration of immature GABAergic neurons present in developing cortical layers II-IV and is prevented by 3-MA, an autophagy inhibitor. In contrast, modulators of autophagy (3-MA, rapamycin) do not interfere with the anti-excitotoxic and neuroprotective effect of MK801 observed in deep layers V and VI. In vivo, 3-MA blocks the rapid increase in caspase-3 cleavage induced by the blockade of NMDA receptors and prevents the resulting long-term decrease in Gad67-GFP neurons in layers II-IV. Together, these data suggest that, in the developing cortex, the suppression of glutamatergic inputs through NMDA receptor inhibition results in the impairment of the autophagic flux and the subsequent switch to apoptotic death of immature GABAergic interneurons. The concomitant inhibition of autophagy prevents this pro-apoptotic action of the NMDA blocker and favors the long-term rescue of GABAergic interneurons without interfering with its neuroprotective actions. The use of autophagy modulators in the developing brain would create new opportunities to prevent the side effects of NMDA antagonists used for neuroprotection or anesthesia.
Collapse
Affiliation(s)
- Christian Roux
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Caroline Aligny
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Céline Lesueur
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Virginie Girault
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Valery Brunel
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Yasmina Ramdani
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France
| | - Damien Genty
- Department of Pathology, Rouen University Hospital, Rouen, France
| | - Azeddine Driouich
- Research Platform of Cell Imagery (PRIMACEN), France; Laboratory of Glycobiology and Plant Extracellular Matrix (GLYCOMEV) EA 4358, France
| | - Annie Laquerrière
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Pathology, Rouen University Hospital, Rouen, France
| | - Stéphane Marret
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Neonatal Paediatrics and Intensive Care, Rouen University Hospital, Rouen, France
| | - Carole Brasse-Lagnel
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| | - Bruno J Gonzalez
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France.
| | - Soumeya Bekri
- Region-Inserm Team NeoVasc ERI28, Laboratory of Microvascular Endothelium and Neonate Brain Lesions, Institute of Research for Innovation in Biomedicine, Normandy University, Rouen, France; Department of Medical Biochemistry, Rouen University Hospital, Rouen, France
| |
Collapse
|
17
|
Rivera-Cervantes MC, Castañeda-Arellano R, Castro-Torres RD, Gudiño-Cabrera G, Feria y Velasco AI, Camins A, Beas-Zárate C. P38 MAPK inhibition protects against glutamate neurotoxicity and modifies NMDA and AMPA receptor subunit expression. J Mol Neurosci 2014; 55:596-608. [PMID: 25172309 DOI: 10.1007/s12031-014-0398-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/05/2014] [Indexed: 11/28/2022]
Abstract
NMDA and AMPA receptors are thought to be responsible for Ca(++) influx during glutamate-induced excitotoxicity and, therefore, hippocampal neuronal death. We assessed whether excitotoxicity induced by neonatal treatment with monosodium glutamate in rats at postnatal age of 1, 3, 5, and 7 modifies the hippocampal expression of the NMDAR subunit NR1 and the AMPAR subunits GluR1/GluR2 at postnatal days 8, 10, 12, and 14. We also assessed the involvement of MAPK signaling by using the p38 inhibitor SB203580. Our results showed that monosodium glutamate induces neuronal death and alters the expression of the subunits evaluated in the hippocampus at all ages studied, which could be prevented by SB203580 treatment.Furthermore, expression of the NRSF gene silencing factor also increased in response to excitotoxicity, suggesting a relationship in suppressing GluR2-expression, which was regulated by the p38-MAPK pathway inhibitor SB203580. This result suggests that selectively blocking the pro-death signaling pathway may reduce neuronal death in some neurodegenerative diseases in which these neurotoxic processes are present and produce major clinical benefits in the treatment of these pathologies.
Collapse
|
18
|
Xing B, Bachstetter AD, Van Eldik LJ. Inhibition of neuronal p38α, but not p38β MAPK, provides neuroprotection against three different neurotoxic insults. J Mol Neurosci 2014; 55:509-18. [PMID: 25012593 PMCID: PMC4303701 DOI: 10.1007/s12031-014-0372-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/01/2014] [Indexed: 12/13/2022]
Abstract
The p38 mitogen-activated protein kinase (MAPK) pathway plays a key role in pathological glial activation and neuroinflammatory responses. Our previous studies demonstrated that microglial p38α and not the p38β isoform is an important contributor to stressor-induced proinflammatory cytokine upregulation and glia-dependent neurotoxicity. However, the contribution of neuronal p38α and p38β isoforms in responses to neurotoxic agents is less well understood. In the current study, we used cortical neurons from wild-type or p38β knockout mice, and wild-type neurons treated with two highly selective inhibitors of p38α MAPK. Neurons were treated with one of three neurotoxic insults (L-glutamate, sodium nitroprusside, and oxygen-glucose deprivation), and neurotoxicity was assessed. All three stimuli led to neuronal death and neurite degeneration, and the degree of neurotoxicity induced in wild-type and p38β knockout neurons was not significantly different. In contrast, selective inhibition of neuronal p38α was neuroprotective. Our results show that neuronal p38β is not required for neurotoxicity induced by multiple toxic insults, but that p38α in the neuron contributes quantitatively to the neuronal dysfunction responses. These data are consistent with our previous findings of the critical importance of microglia p38α compared to p38β, and continue to support selective targeting of the p38α isoform as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Bin Xing
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | | | | |
Collapse
|
19
|
Ramesh G. Novel Therapeutic Targets in Neuroinflammation and Neuropathic Pain. INFLAMMATION AND CELL SIGNALING 2014; 1. [PMID: 26052540 DOI: 10.14800/ics.111] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
There is abounding evidence that neuroinflammation plays a major role in the pathogenesis of neurodegeneration and neuropathic pain. Chemokine-induced recruitment of peripheral immune cells is a central feature in inflammatory neurodegenerative disorders. Immune cells, glial cells and neurons constitute an integral network that coordinates the immune response by releasing inflammatory mediators that in turn modulate inflammation, neurodegeneration and the signal transduction of pain, via interaction with neurotransmitters and their receptors. The chemokine monocyte chemoattractant protein-1/ chemokine (C-C motif) ligand (MCP-1/CCL2) and its receptor C-C chemokine receptor (CCR2) play a major role in mediating neuroinflammation and targeting CCL2/CCR2 represents a promising strategy to limit neuroinflammation-induced neuropathy. In addition, the CCL2/CCR2 axis is also involved in mediating the pain response. Key cellular signaling events such as phosphorylation and subsequent activation of mitogen activated protein kinase (MAPK) p38 and its substrate MAPK-activated protein MAPKAP Kinase (MK) MK-2, regulate neuroinflammation, neuronal survival and synaptic activity. Further, MAPKs such as extracellular signal-regulated kinases (ERK), c-jun N-terminal kinase (JNK) and p38 play vital roles in mediating the pain signaling cascade and contribute to the maintenance of peripheral and central neuronal sensitization associated with chronic pain. This review outlines the rationale for developing therapeutic strategies against CCL2/CCR2 and MAPK signaling networks, identifying them as novel therapeutic targets for limiting neuroinflammation and neuropathic pain.
Collapse
Affiliation(s)
- Geeta Ramesh
- Division of Bacteriology and Parasitology, Tulane National Primate Research Center, Tulane University, 18703 Three Rivers Road, Covington, LA, USA
| |
Collapse
|
20
|
Kim EA, Choi J, Han AR, Choi SY, Hahn HG, Cho SW. Anti-oxidative and anti-inflammatory effects of 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride on glutamate-induced neurotoxicity in rat brain. Neurotoxicology 2013; 38:106-14. [DOI: 10.1016/j.neuro.2013.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/04/2013] [Accepted: 07/05/2013] [Indexed: 01/13/2023]
|
21
|
Combined treatment with capsaicin and resveratrol enhances neuroprotection against glutamate-induced toxicity in mouse cerebral cortical neurons. Food Chem Toxicol 2012; 50:3877-85. [DOI: 10.1016/j.fct.2012.08.040] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 08/16/2012] [Accepted: 08/16/2012] [Indexed: 01/08/2023]
|
22
|
Ju X, Mallet RT, Downey HF, Metzger DB, Jung ME. Intermittent hypoxia conditioning protects mitochondrial cytochrome c oxidase of rat cerebellum from ethanol withdrawal stress. J Appl Physiol (1985) 2012; 112:1706-14. [PMID: 22403345 PMCID: PMC3365408 DOI: 10.1152/japplphysiol.01428.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 03/05/2012] [Indexed: 12/17/2022] Open
Abstract
Intermittent hypoxia (IH) conditioning minimizes neurocognitive impairment and stabilizes brain mitochondrial integrity during ethanol withdrawal (EW) in rats, but the mitoprotective mechanism is unclear. We investigated whether IH conditioning protects a key mitochondrial enzyme, cytochrome c oxidase (COX), from EW stress by inhibiting mitochondrially directed apoptotic pathways involving cytochrome c, Bax, or phosphor-P38 (pP38). Male rats completed two cycles of a 4-wk ethanol diet (6.5%) and 3 wk of EW. An IH program consisting of 5-10 bouts of 5-8 min of mild hypoxia (9.5-10% inspired O(2)) and 4 min of reoxygenation for 20 consecutive days began 3 days before the first EW period. For some animals, vitamin E replaced IH conditioning to test the contributions of antioxidant mechanisms to IH's mitoprotection. During the second EW, cerebellar-related motor function was evaluated by measuring latency of fall from a rotating rod (Rotarod test). After the second EW, COX activity in cerebellar mitochondria was measured by spectrophotometry, and COX, cytochrome c, Bax, and pP38 content were analyzed by immunoblot. Mitochondrial protein oxidation was detected by measuring carbonyl contents and by immunochemistry. Earlier IH conditioning prevented motor impairment, COX inactivation, depletion of COX subunit 4, protein carbonylation, and P38 phosphorylation during EW. IH did not prevent cytochrome c depletion during EW, and Bax content was unaffected by EW ± IH. Vitamin E treatment recapitulated IH protection of COX, and P38 inhibition attenuated protein oxidation during EW. Thus IH protects COX and improves cerebellar function during EW by limiting P38-dependent oxidative damage.
Collapse
Affiliation(s)
- Xiaohua Ju
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas 76107-2699, USA
| | | | | | | | | |
Collapse
|
23
|
HIF-1α expression in the hippocampus and peripheral macrophages after glutamate-induced excitotoxicity. J Neuroimmunol 2011; 238:12-8. [DOI: 10.1016/j.jneuroim.2011.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 05/30/2011] [Accepted: 06/01/2011] [Indexed: 01/13/2023]
|
24
|
Bachstetter AD, Xing B, de Almeida L, Dimayuga ER, Watterson DM, Van Eldik LJ. Microglial p38α MAPK is a key regulator of proinflammatory cytokine up-regulation induced by toll-like receptor (TLR) ligands or beta-amyloid (Aβ). J Neuroinflammation 2011; 8:79. [PMID: 21733175 PMCID: PMC3142505 DOI: 10.1186/1742-2094-8-79] [Citation(s) in RCA: 201] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 07/06/2011] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Overproduction of proinflammatory cytokines from activated microglia has been implicated as an important contributor to pathophysiology progression in both acute and chronic neurodegenerative diseases. Therefore, it is critical to elucidate intracellular signaling pathways that are significant contributors to cytokine overproduction in microglia exposed to specific stressors, especially pathways amenable to drug interventions. The serine/threonine protein kinase p38α MAPK is a key enzyme in the parallel and convergent intracellular signaling pathways involved in stressor-induced production of IL-1β and TNFα in peripheral tissues, and is a drug development target for peripheral inflammatory diseases. However, much less is known about the quantitative importance of microglial p38α MAPK in stressor-induced cytokine overproduction, or the potential of microglial p38α MAPK to be a druggable target for CNS disorders. Therefore, we examined the contribution of microglial p38αMAPK to cytokine up-regulation, with a focus on the potential to suppress the cytokine increase by inhibition of the kinase with pharmacological or genetic approaches. METHODS The microglial cytokine response to TLR ligands 2/3/4/7/8/9 or to Aβ1-42 was tested in the presence of a CNS-penetrant p38α MAPK inhibitor, MW01-2-069A-SRM. Primary microglia from mice genetically deficient in p38α MAPK were used to further establish a linkage between microglia p38α MAPK and cytokine overproduction. The in vivo significance was determined by p38α MAPK inhibitor treatment in a LPS-induced model of acute neuroinflammation. RESULTS Increased IL-1β and TNFα production by the BV-2 microglial cell line and by primary microglia cultures was inhibited in a concentration-dependent manner by the p38α MAPK-targeted inhibitor. Cellular target engagement was demonstrated by the accompanying decrease in the phosphorylation state of two p38α MAPK protein substrates, MK2 and MSK1. Consistent with the pharmacological findings, microglia from p38α-deficient mice showed a diminished cytokine response to LPS. Further, oral administration of the inhibitor blocked the increase of IL-1β in the cerebral cortex of mice stressed by intraperitoneal injection of LPS. CONCLUSION The p38α MAPK pathway is an important contributor to the increased microglial production of proinflammatory cytokines induced by diverse stressors. The results also indicate the feasibility of targeting p38α MAPK to modulate CNS proinflammatory cytokine overproduction.
Collapse
Affiliation(s)
- Adam D Bachstetter
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | | | | | | | | | | |
Collapse
|
25
|
The cycad genotoxin MAM modulates brain cellular pathways involved in neurodegenerative disease and cancer in a DNA damage-linked manner. PLoS One 2011; 6:e20911. [PMID: 21731631 PMCID: PMC3121718 DOI: 10.1371/journal.pone.0020911] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 05/16/2011] [Indexed: 02/02/2023] Open
Abstract
Methylazoxymethanol (MAM), the genotoxic metabolite of the cycad azoxyglucoside cycasin, induces genetic alterations in bacteria, yeast, plants, insects and mammalian cells, but adult nerve cells are thought to be unaffected. We show that the brains of adult C57BL6 wild-type mice treated with a single systemic dose of MAM acetate display DNA damage (O6-methyldeoxyguanosine lesions, O6-mG) that remains constant up to 7 days post-treatment. By contrast, MAM-treated mice lacking a functional gene encoding the DNA repair enzyme O6-mG DNA methyltransferase (MGMT) showed elevated O6-mG DNA damage starting at 48 hours post-treatment. The DNA damage was linked to changes in the expression of genes in cell-signaling pathways associated with cancer, human neurodegenerative disease, and neurodevelopmental disorders. These data are consistent with the established developmental neurotoxic and carcinogenic properties of MAM in rodents. They also support the hypothesis that early-life exposure to MAM-glucoside (cycasin) has an etiological association with a declining, prototypical neurodegenerative disease seen in Guam, Japan, and New Guinea populations that formerly used the neurotoxic cycad plant for food or medicine, or both. These findings suggest environmental genotoxins, specifically MAM, target common pathways involved in neurodegeneration and cancer, the outcome depending on whether the cell can divide (cancer) or not (neurodegeneration). Exposure to MAM-related environmental genotoxins may have relevance to the etiology of related tauopathies, notably, Alzheimer's disease.
Collapse
|
26
|
Mitogen-activated protein kinase p38 in HIV infection and associated brain injury. J Neuroimmune Pharmacol 2011; 6:202-15. [PMID: 21286833 DOI: 10.1007/s11481-011-9260-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 01/23/2011] [Indexed: 02/05/2023]
Abstract
Infection with human immunodeficiency virus-1 (HIV-1) often leads to HIV-associated neurocognitive disorders (HAND) prior to the progression to acquired immunodeficiency syndrome (AIDS). At the cellular level, mitogen-activated protein kinases (MAPK) provide a family of signal transducers that regulate many processes in response to extracellular stimuli and environmental stress, such as viral infection. Recently, evidence has accumulated suggesting that p38 MAPK plays crucial roles in various pathological processes associated with HIV infection, ranging from macrophage activation to neurotoxicity and impairment of neurogenesis to lymphocyte apoptosis. Thus, p38 MAPK, which has generally been linked to stress-related signal transduction, may be an important mediator in the development of AIDS and HAND.
Collapse
|
27
|
Zhou H, Chen Q, Kong DL, Guo J, Wang Q, Yu SY. Effect of resveratrol on gliotransmitter levels and p38 activities in cultured astrocytes. Neurochem Res 2010; 36:17-26. [PMID: 20842424 DOI: 10.1007/s11064-010-0254-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2010] [Indexed: 10/19/2022]
Abstract
Accumulating evidence suggests that resveratrol may have beneficial effects against traumatic brain injury. However, its effect on the regulation of extracellular levels of gliotransmitter and on the activation of p38 MAPK in astrocytes is still unknown. We have examined whether resveratrol regulates extracellular levels of gliotransmitter as well as the activation of p38 MAPK in cultured astrocytes before and after stretch injury. The extracellular levels of glutamate, D-/L-serine and D-serine were apparently reduced by 100 μM resveratrol in control astrocyte cultures. The dramatic increase of glutamate and D-serine release induced by stretch injury was also clearly inhibited by resveratrol. Resveratrol mediates this response by reduction of release through inhibition of extracellular calcium influx and increment of gliotransmitter uptake through enhancement of amino acid transporter expressed in the membrane of astrocyte. In addition, resveratrol definitely reduced the activation of p38 MAPK in cultured astrocytes following stretch injury. AMPA receptor is involved in the activation of p38 following injury. Conversely, the levels of glutamine and glycine were not obviously affected by resveratrol before and after injury. Intracellular levels of glutamate and D-serine are not apparently changed by stretch injury. Collectively, our data suggest that resveratrol might play an important role in protection of the nervous system after injury by decreasing the extracellular levels of gliotransmitter and inhibiting activation of p38 MAPK following injury.
Collapse
Affiliation(s)
- Hao Zhou
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | | | |
Collapse
|
28
|
Zhu W, Zheng H, Shao X, Wang W, Yao Q, Li Z. Excitotoxicity of TNFalpha derived from KA activated microglia on hippocampal neurons in vitro and in vivo. J Neurochem 2010; 114:386-96. [PMID: 20438614 DOI: 10.1111/j.1471-4159.2010.06763.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Highly activated microglia and followed excessive expression of inflammatory cytokines are associated with neuroexcitotoxic injuries. We use electrophysiological techniques, ELISA, western-blot, RT-PCR assay and TUNEL method to explore whether over-produced tumor necrosis factor alpha (TNFalpha) released from activated microglia results in neuronal injuries, and further causes apoptosis through increasing excitotoxicity of hippocampal neurons. Our data showed that kainic acid (KA) activated microglia highly expressed TNFalpha, mRNA and protein. KA activated microglia conditioned media ((KA-MCM) significantly enhanced the amplitude of the population spike at rat's hippocampal CA3 region. It also increased the Ca(2+) current amplitude and density in cultured hippocampal neurons, as well as the high expression of NMDAR1, iNOS, and caspase 3 mRNA and protein at both hippocampal neurons and tissues. KA-MCM also increased TUNEL-positive cells in hippocampal neurons, whereas addition of anti-TNFalpha to the KA-MCM before its application significantly reduced those effects. These studies suggest that TNFalpha derived from KA activated microglia increases excitotoxicity of hippocampal neurons, and might induce neuronal apoptosis in vitro and in vivo.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Brain Research, Division of Anatomy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
29
|
Chronic pretreatment with acetyl-L-carnitine and ±DL-α-lipoic acid protects against acute glutamate-induced neurotoxicity in rat brain by altering mitochondrial function. Neurotox Res 2010; 19:319-29. [PMID: 20217290 DOI: 10.1007/s12640-010-9165-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 12/23/2009] [Accepted: 02/20/2010] [Indexed: 01/05/2023]
Abstract
Cellular oxidative stress and energy failure were shown to be involved in Glutamate (L-Glu) neurotoxicity, whereas, acetyl-L-carnitine (ALCAR) and ±DL-α-lipoic acid (LA) are known to be key players in the mitochondrial energy production. To evaluate the effects of the above antioxidants, adult rats were pretreated with ALCAR (100 mg/kg i.p for 21 days) and both ALCAR and LA (100 mg/kg i.p + 50 mg/kg i.p for 21 days), before stereotactically administering L-Glu bolus (1 μmole/1 μl) in the cerebral cortex. Results showed that acute L-Glu increased ROS (P < 0.001), LPO (P < 0.001), Ca(2+) (P < 0.001), TNF-α (P < 0.001), IFN-γ (P < 0.001), NO (P < 0.001) levels and mRNA expression of Caspase-3, Casapase-9, iNOS, and nNOS genes with respect to saline-injected control group. Key antioxidant parameters such as SOD, CAT, GSH, GR along with mitochondrial transmembrane potential (Ψ∆m) were decreased (P < 0.05), while ALCAR pretreatment prevented these effects by significantly inhibiting ROS (P < 0.001), LPO (P < 0.001), Ca(2+) (P < 0.05), TNF-α (P < 0.05), IFN-γ (P < 0.001), NO (P < 0.01) levels and expression of the above genes. This chronic pretreatment of ALCAR also increased SOD, CAT, GSH, GR, and Ψ∆m (P < 0.0.01, P < 0.0.01, P < 0.05, P < 0.05, and P < 0.001, respectively) with respect to L: -Glu group. The addition of LA to ALCAR resulted in further increases in CAT (P < 0.05), GSH (P < 0.01), GR (P < 0.05), Ψ∆m (P < 0.05) and additional decreases in ROS (P < 0.001), LPO (P < 0.05), Ca(2+) (P < 0.05), TNF-α (P < 0.05) and mRNA expression of iNOS and nNOS genes with respect to ALCAR group. Hence, this "one-two punch" of ALCAR + LA may help in ameliorating the deleterious cellular events that occur after L-Glu.
Collapse
|
30
|
Lin L, Wu XD, Davey AK, Wang J. The anti-inflammatory effect of baicalin on hypoxia/reoxygenation and TNF-α induced injury in cultural rat cardiomyocytes. Phytother Res 2009; 24:429-37. [DOI: 10.1002/ptr.3003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|