1
|
Kim HJ, Kim H, Song J, Hong JY, Lee EH, Londhe AM, Choi JW, Park SJ, Oh E, Yoon H, Hwang H, Hahn D, Jung K, Kwon S, Kadayat TM, Ma MJ, Joo J, Kim J, Bae JH, Hwang H, Pae AN, Cho SJ, Park JH, Chin J, Kang H, Park KD. Highly potent and selective PPARδ agonist reverses memory deficits in mouse models of Alzheimer's disease. Theranostics 2024; 14:6088-6108. [PMID: 39431021 PMCID: PMC11488110 DOI: 10.7150/thno.96707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
Rationale: Alzheimer's disease (AD) is a progressive neurodegenerative disease accompanied by neurotoxicity, excessive inflammation, and cognitive impairment. The peroxisome proliferator-activated receptor (PPAR) δ is a potential target for AD. However, its regulatory mechanisms and therapeutic potential in AD remain unclear. We aimed to investigate if the activation of PPARδ using a highly selective and potent agonist could provide an effective therapeutic strategy against AD. Methods: We synthesized a novel PPARδ agonist, 5a, containing a selenazole group and determined the X-ray crystal structure of its complex with PPARδ. The drug-like properties of 5a were assessed by analyzing cytochrome P450 (CYP) inhibition, microsomal stability, pharmacokinetics, and mutagenicity. We investigated the anti-inflammatory effects of 5a using lipopolysaccharide (LPS)-stimulated BV-2 microglia and neuroinflammatory mouse model. The therapeutic efficacy of 5a was evaluated in AD mice with scopolamine-induced memory impairment and APP/PS1 by analyzing cognitive function, glial reactivity, and amyloid pathology. Results: Compound 5a, the most potent and selective PPARδ agonist, was confirmed to bind hPPARδ in a complex by X-ray crystallographic analysis. PPARδ activation using 5a showed potent anti-inflammatory effects in activated glial cells and mouse model of neuroinflammation. Administration of 5a inhibited amyloid plaque deposition by suppressing the expression of neuronal beta-site amyloid precursor protein cleaving enzyme 1 (BACE1), and reduced abnormal glial hyperactivation and inflammatory responses, resulting in improved learning and memory in the APP/PS1 mouse model of AD. Conclusion: We identified that specific activation of PPARδ provides therapeutic effects on multiple pathogenic phenotypes of AD, including neuroinflammation and amyloid deposition. Our findings suggest the potential of PPARδ as a promising drug target for treating AD.
Collapse
Affiliation(s)
- Hyeon Jeong Kim
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Haelee Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jaeyoung Song
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jun Young Hong
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- Department of Systems Biology, Yonsei University, Seoul 03722, Republic of Korea
| | - Elijah Hwejin Lee
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ashwini M. Londhe
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ji Won Choi
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Sun Jun Park
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Eunseok Oh
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
| | - Heeseok Yoon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Hoosang Hwang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
| | - Dongyup Hahn
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyungjin Jung
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Sugyeong Kwon
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Tara Man Kadayat
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Min Jung Ma
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jeongmin Joo
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jina Kim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Jae Hyun Bae
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Hayoung Hwang
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
| | - Ae Nim Pae
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Sung Jin Cho
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Jong-Hyun Park
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Jungwook Chin
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Republic of Korea
- Cureverse, lnc., H2 building, KIST, Seoul 02792, Republic of Korea
| | - Heonjoong Kang
- Laboratory of Marine Drugs, School of Earth and Environmental Sciences, Seoul National University, NS-80 Seoul 08826, Republic of Korea
- Research Institute of Oceanography, Seoul National University, NS-80, Seoul 08826, Republic of Korea
| | - Ki Duk Park
- Center for Brain Disorders, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
2
|
Golubska M, Paukszto Ł, Kurzyńska A, Mierzejewski K, Gerwel Z, Bogacka I. PPAR beta/delta regulates the immune response mechanisms in the porcine endometrium during LPS-induced inflammation - An in vitro study. Theriogenology 2024; 226:130-140. [PMID: 38878465 DOI: 10.1016/j.theriogenology.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024]
Abstract
Inflammation in the reproductive tract has become a serious threat to animal fertility. Recently, the role of peroxisome proliferator-activated receptor gamma (PPARγ) in the context of reproduction and the inflammatory response has been highlighted, but the role of PPARβ/δ has not been fully elucidated. The aim of the present study was to investigate the in vitro effect of PPARβ/δ ligands (agonist: L-165,041 and antagonist: GSK 3787) on the transcriptome profile of porcine endometrium during LPS-induced inflammation in the mid-luteal and follicular phases of the oestrous cycle (days 10-12 and 18-20, respectively) using the RNA-Seq method. During the mid-luteal phase of the oestrous cycle, the current study identified 145 and 143 differentially expressed genes (DEGs) after treatment with an agonist or antagonist, respectively. During the follicular phase of the oestrous cycle, 55 and 207 DEGs were detected after treatment with an agonist or antagonist, respectively. The detected DEGs are engaged in the regulation of various processes, such as the complement and coagulation cascade, NF-κB signalling pathway, or the pathway of 15-eicosatetraenoic acid derivatives synthesis. The results of the current study indicate that PPARβ/δ ligands are involved in the control of the endometrial inflammatory response.
Collapse
Affiliation(s)
- Monika Golubska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Łukasz Paukszto
- Department of Botany and Nature Protection, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Kurzyńska
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Karol Mierzejewski
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Zuzanna Gerwel
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Iwona Bogacka
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland.
| |
Collapse
|
3
|
Strosznajder AK, Wójtowicz S, Jeżyna MJ, Sun GY, Strosznajder JB. Recent Insights on the Role of PPAR-β/δ in Neuroinflammation and Neurodegeneration, and Its Potential Target for Therapy. Neuromolecular Med 2020; 23:86-98. [PMID: 33210212 PMCID: PMC7929960 DOI: 10.1007/s12017-020-08629-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) β/δ belongs to the family of hormone and lipid-activated nuclear receptors, which are involved in metabolism of long-chain fatty acids, cholesterol, and sphingolipids. Similar to PPAR-α and PPAR-γ, PPAR-β/δ also acts as a transcription factor activated by dietary lipids and endogenous ligands, such as long-chain saturated and polyunsaturated fatty acids, and selected lipid metabolic products, such as eicosanoids, leukotrienes, lipoxins, and hydroxyeicosatetraenoic acids. Together with other PPARs, PPAR-β/δ displays transcriptional activity through interaction with retinoid X receptor (RXR). In general, PPARs have been shown to regulate cell differentiation, proliferation, and development and significantly modulate glucose, lipid metabolism, mitochondrial function, and biogenesis. PPAR-β/δ appears to play a special role in inflammatory processes and due to its proangiogenic and anti-/pro-carcinogenic properties, this receptor has been considered as a therapeutic target for treating metabolic syndrome, dyslipidemia, carcinogenesis, and diabetes. Until now, most studies were carried out in the peripheral organs, and despite of its presence in brain cells and in different brain regions, its role in neurodegeneration and neuroinflammation remains poorly understood. This review is intended to describe recent insights on the impact of PPAR-β/δ and its novel agonists on neuroinflammation and neurodegenerative disorders, including Alzheimer’s and Parkinson’s, Huntington’s diseases, multiple sclerosis, stroke, and traumatic injury. An important goal is to obtain new insights to better understand the dietary and pharmacological regulations of PPAR-β/δ and to find promising therapeutic strategies that could mitigate these neurological disorders.
Collapse
Affiliation(s)
- Anna K Strosznajder
- Faculty of Medicine, Medical University of Bialystok, 1 Kilinskiego st., 15-089, Białystok, Poland
| | - Sylwia Wójtowicz
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego st., 02-106, Warsaw, Poland
| | - Mieszko J Jeżyna
- Faculty of Medicine, Medical University of Bialystok, 1 Kilinskiego st., 15-089, Białystok, Poland
| | - Grace Y Sun
- Biochemistry Department, University of Missouri, Columbia, MO, 65211, USA
| | - Joanna B Strosznajder
- Department of Cellular Signaling, Mossakowski Medical Research Centre Polish Academy of Sciences, 5 Pawińskiego st., 02-106, Warsaw, Poland.
| |
Collapse
|
4
|
Gamdzyk M, Lenahan C, Tang J, Zhang JH. Role of peroxisome proliferator-activated receptors in stroke prevention and therapy-The best is yet to come? J Neurosci Res 2020; 98:2275-2289. [PMID: 32772463 DOI: 10.1002/jnr.24709] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/08/2020] [Accepted: 07/20/2020] [Indexed: 12/25/2022]
Abstract
Role of peroxisome proliferator-activated receptors (PPARs) in the pathophysiology of stroke and protective effects of PPAR ligands have been widely investigated in the last 20 years. Activation of all three PPAR isoforms, but especially PPAR-γ, was documented to limit postischemic injury in the numerous in vivo, as well as in in vitro studies. PPARs have been demonstrated to act on multiple mechanisms and were shown to activate multiple protective pathways related to inflammation, apoptosis, BBB protection, neurogenesis, and oxidative stress. The aim of this review was to summarize two decades of PPAR research in stroke with emphasis on in vivo animal studies. We focus on each PPAR receptor separately and detail their implication in stroke. This review also discusses recent clinical efforts in the field and the epidemiological data with regard to role of PPAR polymorphisms in susceptibility to stroke, and tries to draw conclusions and describe future perspectives.
Collapse
Affiliation(s)
- Marcin Gamdzyk
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Cameron Lenahan
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA, USA.,Department of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA, USA
| |
Collapse
|
5
|
Luo Y, Kuang S, Xue L, Yang J. The mechanism of 5-lipoxygenase in the impairment of learning and memory in rats subjected to chronic unpredictable mild stress. Physiol Behav 2016; 167:145-153. [PMID: 27640130 DOI: 10.1016/j.physbeh.2016.09.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Revised: 07/20/2016] [Accepted: 09/13/2016] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To examine the mechanism of 5-lipoxygenase (5-LO) in the learning and memory dysfunction in rats subjected to chronic unpredictable mild stress (CUMS). METHODS Eighty rats were divided into eight groups: the 0.5% sodium carboxymethyl cellulose solution (NaCMC)-treated group, empty vector (LV-Mock)-treated group, CUMS+NaCMC-treated group, CUMS+sertraline-treated group, CUMS+caffeic acid (10mg/kg)-treated group, CUMS+caffeic acid (30mg/kg)-treated group, CUMS+LV-Mock-treated group, and CUMS+5-LO-silencers lentiviral vectors (LV-si-5-LO)-treated group, n=10. Sucrose preference tests were performed to assess depression-like behavior. The Morris water maze and step-down tests were used to evaluate learning and memory performance. The levels of inflammatory cytokines, malondialdehyde, and the activity of superoxide dismutase (SOD) were detected to estimate inflammation and oxidative stress. Changes in 5-LO mRNA and protein were detected using reverse transcription polymerase chain reaction and Western blotting. The expression of synaptophysin, postsynaptic density-95 (PSD-95), and brain-derived neurotrophic factor (BDNF) in the hippocampus were measured using immunohistochemical staining. RESULTS Treatment with caffeic acid or LV-si-5-LO increased sucrose consumption, decreased escape latency and increased the number of platform crosses in the Morris water maze test, and decreased the number of errors and prolonged the latency in the step-down test. We observed a decreased expression of 5-LO, and levels of malondialdehyde, leukotriene-B4, tumor necrosis factor-α, and interleukin-6, while the protein levels of synaptophysin, PSD-95, BDNF, and the activity of SOD were increased in the hippocampus of the CUMS-treated rats. CONCLUSIONS CUMS-induced impairment in learning and memory could be triggered by an inflammatory response in the rat hippocampus, which results in oxidative stress injury and impacts the synaptic plasticity of hippocampal neurons. Inhibition of the activity or expression of 5-LO could suppress hippocampal inflammation, enhance synaptic plasticity, and improve learning and memory function in depressed rats.
Collapse
Affiliation(s)
- Ying Luo
- Department of Pharmacology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China.
| | - Shengnan Kuang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Lai Xue
- Department of Pharmacology, Chongqing Medical University, Chongqing, China.
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, Chongqing, China; Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, China.
| |
Collapse
|
6
|
Cheng O, Li R, Zhao L, Yu L, Yang B, Wang J, Chen B, Yang J. Short-term sleep deprivation stimulates hippocampal neurogenesis in rats following global cerebral ischemia/reperfusion. PLoS One 2015; 10:e0125877. [PMID: 26039740 PMCID: PMC4454510 DOI: 10.1371/journal.pone.0125877] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/26/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Sleep deprivation (SD) plays a complex role in central nervous system (CNS) diseases. Recent studies indicate that short-term SD can affect the extent of ischemic damage. The aim of this study was to investigate whether short-term SD could stimulate hippocampal neurogenesis in a rat model of global cerebral ischemia/reperfusion (GCIR). METHODS One hundred Sprague-Dawley rats were randomly divided into Sham, GCIR and short-term SD groups based on different durations of SD; the short-term SD group was randomly divided into three subgroups: the GCIR+6hSD*3d-treated, GCIR+12hSD-treated and GCIR+12hSD*3d-treated groups. The GCIR rat model was induced via the bilateral occlusion of the common carotid arteries and hemorrhagic hypotension. The rats were sleep-deprived starting at 48 h following GCIR. A Morris water maze test was used to assess learning and memory ability; cell proliferation and differentiation were analyzed via 5-bromodeoxyuridine (BrdU) and neuron-specific enolase (NSE), respectively, at 14 and 28 d; the expression of hippocampal BDNF was measured after 7 d. RESULTS The different durations of short-term SD designed in our experiment exhibited improvement in cognitive function as well as increased hippocampal BDNF expression. Additionally, the short-term SD groups also showed an increased number of BrdU- and BrdU/NSE-positive cells compared with the GCIR group. Of the three short-term SD groups, the GCIR+12hSD*3d-treated group experienced the most substantial beneficial effects. CONCLUSIONS Short-term SD, especially the GCIR+12hSD*3d-treated method, stimulates neurogenesis in the hippocampal dentate gyrus (DG) of rats that undergo GCIR, and BDNF may be an underlying mechanism in this process.
Collapse
Affiliation(s)
- Oumei Cheng
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Rong Li
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Lei Zhao
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Lijuan Yu
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Bin Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Jia Wang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
| | - Beibei Chen
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing 400016, China
- * E-mail:
| |
Collapse
|
7
|
Pan Y, Yu L, Lei W, Guo Y, Wang J, Yu H, Tang Y, Yang J. Beraprost sodium protects against chronic brain injury in aluminum-overload rats. Behav Brain Funct 2015; 11:6. [PMID: 25888780 PMCID: PMC4326490 DOI: 10.1186/s12993-014-0051-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 12/29/2014] [Indexed: 11/25/2022] Open
Abstract
Background Aluminum overload can cause severe brain injury and neurodegeneration. Previous studies suggest that prostacyclin synthase (PGIS) expression and prostacyclin receptor (IP) activation are beneficial for treatment of acute traumatic and ischemic brain injury. However, the potential value of PGIS/IP signaling pathway to chronic brain injury is still unclear. In this study, we investigated the change of PGIS/IP signaling pathway and the effect of beraprost sodium (BPS) on chronic brain injury in chronic aluminum-overload rats. Methods Rat model of chronic cerebral injury was established by chronic intragastric administration of aluminum gluconate(Al3+ 200 mg/kg per day,5d a week for 20 weeks). The methods of ELISA, qRT-PCR and Western blotting were used to detect the PGI2 level and the PGIS and IP mRNA and protein levels in hippocampi of chronic aluminum-overload rats, respectively. Rat hippocampal superoxide dismutase (SOD) activity and malondialdehyde (MDA) content also were measured. The effects of BPS (6, 12 and 24 μg⋅kg-1) on brain injury in chronic aluminum-overload rats were evaluated. Results Compared with the control group, PGIS mRNA expression, PGI2 level, and the IP mRNA and protein expressions significantly increased in hippocampi of chronic aluminum-overload rats. Administration of BPS significantly improved spatial learning and memory function impairment and hippocampal neuron injury induced by chronic aluminum overload in rats. Meanwhile, administration of BPS resulted in a decrease of PGI2 level and downregulation of PGIS and IP expressions in a dose-dependent manner. Aluminum overload also caused a decrease of SOD activity and an increase of MDA content. Administration of BPS significantly blunted the decrease of SOD activity and the increase of MDA content induced by aluminum overload in rats. Conclusions BPS has a significant neuroprotective effect on chronic brain injury induced by aluminum overload in rats. Remodeling the balance of PGIS/IP signaling pathway and inhibition of oxidative stress involve in the neuroprotective mechanism of BPS in aluminum-overload rats. The PGIS/IP signaling pathway is a potential therapeutic strategy for chronic brain injury patients.
Collapse
Affiliation(s)
- Yongquan Pan
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China. .,Laboratory Animal Center, Chongqing Medical University, Chongqing, 400016, China.
| | - Lijuan Yu
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Wenjuan Lei
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Yuanxin Guo
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Jianfeng Wang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China.
| | - Huarong Yu
- Department of Basic College, Chongqing Medical University, Chongqing, 400016, China.
| | - Yong Tang
- Department of Basic College, Chongqing Medical University, Chongqing, 400016, China.
| | - Junqing Yang
- Department of Pharmacology, Chongqing Medical University, the Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing, 400016, China. .,Department of Pharmacology, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
8
|
Dabaghian FH, Hashemi M, Entezari M, Movassaghi S, Goushegir SA, Kalantari S, Movafagh A, Sharifi ZN. Effect of Cyperus rotundus on ischemia-induced brain damage and memory dysfunction in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:199-204. [PMID: 25825638 PMCID: PMC4366733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 12/22/2014] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Global cerebral ischemia-reperfusion injury causes loss of pyramidal cells in CA1 region of hippocampus. In this study, we investigated the possible neuroprotective effects of the ethanol extract of Cyperus rotundus (EECR) on a model of global transient ischemia in rat, by evaluating the pathophysiology of the hippocampal tissue and spatial memory. MATERIALS AND METHODS Treatment group (EECR, 100 mg/kg/day) was gavaged from 4 days before, to 3 days after ischemia. Morris water maze test was performed 1 week after ischemia for 4 days. Brain tissue was prepared for Nissl staining. RESULTS Our data showed no statistical difference between the treatment and ischemia groups in water maze task. So, treatment of ischemia with EECR cannot improve spatial learning and memory. On the contrary EECR ameliorated the CA1 pyramidal cell loss due to transient global ischemia/reperfusion injury. CONCLUSION These results suggest that EECR cannot reduce the ischemia-induced, cognitive impairments seen after transient, global cerebral ischemia but can prevent pyramidal cell loss in CA1 region of hippocampus.
Collapse
Affiliation(s)
- Fataneh Hashem Dabaghian
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Science, Tehran, Iran
| | - Mehrdad Hashemi
- Genetic Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Maliheh Entezari
- Biology Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Shabnam Movassaghi
- Anatomy Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Seyed Ashrafadin Goushegir
- Research Institute for Islamic and Complementary Medicine, Iran University of Medical Science, Tehran, Iran
| | - Samaneh Kalantari
- Biology Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Abolfazl Movafagh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Nadia Sharifi
- Anatomy Department, Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran,*Corresponding author: Zahra Nadia Sharifi. Anatomy Department, School of Medicine, Islamic Azad University Tehran Medical Branch, Zargandeh St, Shariati St, Tehran, Iran. Tel: +98-21-22006660-7; Fax: +98-21-22600714; ,
| |
Collapse
|
9
|
Yu L, Yang B, Wang J, Zhao L, Luo W, Jiang Q, Yang J. Time course change of COX2-PGI2/TXA2 following global cerebral ischemia reperfusion injury in rat hippocampus. Behav Brain Funct 2014; 10:42. [PMID: 25388440 PMCID: PMC4240876 DOI: 10.1186/1744-9081-10-42] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Accepted: 10/22/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neuroinflammation plays pivotal roles in the progression of cerebral ischemia injury. Prostaglandins (PGs) as the major inflammatory mediators in the brain participate in the pathophysiological processes of cerebral ischemia injury. Cyclooxygenase-2 (COX2) is the rate-limiting enzyme of PGs, and thus it is necessary to characterize of the expression patterns of COX2 and its downstream products at the same time in a cerebral ischemia/reperfusion (I/R) model. METHODS The levels of prostacyclin (PGI2) and thromboxane (TXA2) and the expression of COX2 were detected in the rat hippocampus at different time points after reperfusion (30 min, 2 h, 6 h, 24 h, 48 h, 7 d, and 15 d). RESULTS The COX2 mRNA and protein expressions in hippocampus both remarkably increased at 30 min, and peaked at 7 d after global cerebral I/R compared with the sham-operated group. The level of PGI2 significantly increased at 2 h after reperfusion, with a peak at 48 h, but was still significantly higher than the sham-operated animals at 15 d. TXA2 level decreased at 30 min and 2 h after reperfusion, but significantly increased at 6 h and peaked at 48 h. PGI2/TXA2 ratio increased at 30 min after reperfusion, and peaked at 48 h compared with the sham-operated animals. CONCLUSIONS I/R injury significantly increased the COX2 expression, PGI2 and TXA2 levels, and the PGI2/TXA2 ratio in rat hippocampus in a time-dependent manner. As a consequence, the increased PGI2 level and PGI2/TXA2 ratio may represent a physiological mechanism to protect the brain against the neuronal damage produced by I/R injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Junqing Yang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Medical College Rd, No 1, Chongqing 400016, China.
| |
Collapse
|
10
|
Feng T, Liu Y, Li C, Li Z. Protective Effects of Nigranoic Acid on Cerebral Ischemia–Reperfusion Injury and its Mechanism Involving Apoptotic Signaling Pathway. Cell Biochem Biophys 2014; 71:345-51. [DOI: 10.1007/s12013-014-0204-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Luo Y, He Q, Kuang G, Jiang Q, Yang J. PPAR-alpha and PPAR-beta expression changes in the hippocampus of rats undergoing global cerebral ischemia/reperfusion due to PPAR-gamma status. Behav Brain Funct 2014; 10:21. [PMID: 24934302 PMCID: PMC4167308 DOI: 10.1186/1744-9081-10-21] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/08/2014] [Indexed: 12/19/2022] Open
Abstract
Background Peroxisome proliferator-activated receptors (PPARs, including alpha, beta and gamma subtypes) and their agonists have a protective role in treatment of central nervous system (CNS) diseases. The present study was designed to investigate the expression changes of PPAR-alpha, -beta, -gamma and NF-kappa B in the hippocampus of rats with global cerebral ischemia/reperfusion injury (GCIRI) after treatment with agonists or antagonists of PPAR-gamma. Methods A rat GCIRI model was established by occlusion of bilateral common carotid arteries and cervical vena retransfusion. GW9662 (5 μg), a selective PPAR- gamma antagonist, was intraventricularly injected at 0.5 h before GCIR; Rosiglitazone (0.8, 2.4 and 7.2 mg/kg), a selective PPAR- gamma agonist, was injected intraperitoneally at 1 h before GCIRI. The expression changes of PPAR-alpha, -beta and -gamma at mRNA and protein levels were detected by RT-PCR and western blotting. The changes of spatial learning and memory (SLM) functions were assessed by using a Morris water maze; the pathohistological changes of hippocampal neurons were evaluated by hematoxylin-eosin (HE) staining; the contents of IL-1, IL-6, IL-10 and TNF-alpha, and the NF- kappa B expression were measured by enzyme-linked immunosorbent assay (ELISA) and immunohistochemical staining. The superoxide dismutase (SOD) activity and malondialdehyde (MDA) content were also detected. Results The SLM function and hippocampal neurons were significantly impaired after the occurrence of GCIRI. The MDA, IL-1, IL-6, IL-10, TNF-alpha content and expression of PPARs increased significantly, but the SOD activity and NF-kappa B expression were weakened in the hippocampus. Rosiglitazone treatment significantly protected rats from SLM function impairment and neuron death, and resulted in higher expressions of SOD activity and NF-kappa B, but lower contents of MDA and inflammatory factors. After treatment with rosiglitazone or GW9662, no significant change in PPAR-alpha or -beta expression was detected. Conclusions Rosiglitazone, a PPAR-gamma agonist, plays a protective role in hippocampal neuron damage of GCIRI rats by inhibiting the oxidative stress response and inflammation. The activation or antagonism of PPAR-gamma did not affect the expression of PPAR-alpha or -beta, indicating that the three subtypes of PPARs act in independent pathways in the CNS.
Collapse
Affiliation(s)
| | | | | | | | - Junqing Yang
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Medical College Rd, No 1, Chongqing 400016, P, R, China.
| |
Collapse
|
12
|
Yu L, Jiang R, Su Q, Yu H, Yang J. Hippocampal neuronal metal ion imbalance related oxidative stress in a rat model of chronic aluminum exposure and neuroprotection of meloxicam. Behav Brain Funct 2014; 10:6. [PMID: 24618126 PMCID: PMC3995718 DOI: 10.1186/1744-9081-10-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 02/28/2014] [Indexed: 01/22/2023] Open
Abstract
Neurodegenerative diseases remain a significant unresolved societal burden afflicting millions of people worldwide. Neurons in the brain are highly sensitive to oxidative stress, which can be induced by metal toxicity. In this paper, a chronic aluminum overload-induced model of neurodegeneration was used to investigate whether metal ions (Al, Fe, Mn, Cu and Zn)-related oxidative stress was involved in neurodegenerative mechanism and to identify the protective action of meloxicam against rat hippocampal neuronal injury. The metal ion contents, activity of superoxide dismutase (SOD), and content of malondialdehyde (MDA) were detected. The results showed that the spatial learning and memory (SLM) function was significantly impaired in chronic aluminum overload rats. Considerable karyopycnosis was observed in hippocampal neurons. The SOD activity was weakened and the MDA content increased both significantly. In the hippocampus, Al, Fe, Mn, Cu, and Zn contents increased by 184.1%, 186.1%, 884.2%, 199.4% and 149.2%, respectively. Meloxicam administration (without Al) had no effect compared with the control group, while meloxicam treatment with aluminum exposure significantly protected rats from SLM function impairment, neuron death, lower SOD activity, higher MDA content and brain metal ion imbalance. Our findings suggest that the cerebral metal ion imbalance-related oxidative stress is involved in mechanism of cerebral injury and neurodegeneration induced by chronic Al overload in rats, and that meloxicam protects neurons by reducing metal ion imbalance-related oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Huarong Yu
- Department of Pharmacology, Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Medical College Rd, No 1, Chongqing Medical University, Chongqing 400016, P, R, China.
| | | |
Collapse
|