1
|
He XF, Yang XF, Li G, Zhao Y, Luo J, Xu JH, Zheng HQ, Zhang LY, Hu XQ. Physical Exercise Improves the Neuronal Function in Ischemic Stroke Via Microglial CB 2R/P2Y12 Signaling. Mol Neurobiol 2024:10.1007/s12035-024-04391-2. [PMID: 39066973 DOI: 10.1007/s12035-024-04391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
Physical exercise (PE) may be the single most important and accessible lifestyle habit throughout life, it inhibits the neuroinflammatory response and protects the brain against damage. As the innate cells in brain, microglia undergo morphological and functional changes to communicate with neurons protecting the neurons from injury. Herein, aiming at exploring the effects of PE on the communication between microglia-neuron during acute ischemic cerebral infarction, we carried out running wheel training before the conduction of transient middle cerebral artery occlusion (tMCAO) in C57BL/6 J and Cx3cr1-GFP mice. We found that microglial P2Y12 expression in the peri-infarct area was decreased, microglial dynamics and microglia-neuron communications were impaired, using in vivo two-photon imaging. PE up-regulated the microglial P2Y12 expression, increased the microglial dynamics, and promoted the contacts of microglia with neurons. As a result, PE inhibited neuronal Ca2+ overloads and protected against damage of the neuronal mitochondria in acute tMCAO. Mechanistically, PE increased the cannabinoid receptor 2 (CB2R) in microglia, promoted the phosphorylation of Nrf2 (NF-E2-related factor 2) at ser-344, increased the transcription factor level of Mafk, and up-regulated the level of P2Y12, whereby PE increased the levels of CB2R to promote microglia-neuron contacts to monitor and protect neuronal function.
Collapse
Affiliation(s)
- Xiao-Fei He
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiao-Feng Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, Guangdong, China
| | - Yun Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jing-Hui Xu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Hai-Qing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Li-Ying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Xi-Quan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
2
|
Yuan X, Ye W, Chen L, Luo D, Zhou L, Qiu Y, Zhuo R, Zhao Y, Peng L, Yang L, Jin X, Zhou Y. URB597 exerts neuroprotective effects against transient brain ischemia injury in mice by regulating autophagic flux and necroptosis. Eur J Pharmacol 2023; 957:175982. [PMID: 37572942 DOI: 10.1016/j.ejphar.2023.175982] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/14/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Ischemic stroke is a leading cause of death and disability, and medical treatments for ischemic stroke are very limited. URB597 is a potent and selective inhibitor of fatty acid amide hydrolase (FAAH). However, the effect of URB597 on ischemic stroke and the underlying molecular mechanisms remain little known. In this study, focal cerebral ischemia was induced by transient middle cerebral artery occlusion in mice. Our results showed that URB597 dose-dependently improved neurological function and reduced brain infarct volume and brain edema 24 h after brain ischemia. The most effective dose was 1 mg/kg and the therapeutic time window was within 3 h after ischemic stroke. To further investigate the underlying mechanism, necroptosis and autophagy flux were detected by Western blot and/or immunofluorescence staining with or without chloroquine, an autophagic flux inhibitor. Our results showed that URB597 promoted autophagic flux and reduced neuronal necroptosis after brain ischemia and these effects could be abolished by chloroquine. In addition, we found that peroxisome proliferator-activated receptor α (PPARα) antagonist GW6471 partly abolished the effect of URB597 against brain ischemia and URB597 upregulated the expressions of PPARα. In conclusion, URB597 exerts a neuroprotective effect in a dose- and time-dependent manner, and this effect may be related to its restoration of autophagic flux and inhibition of neuronal necroptosis. PPARα is involved in the neuroprotective effect of URB597. This study provides novel evidence that URB597 may be a promising agent for the clinical treatment of ischemic stroke.
Collapse
Affiliation(s)
- Xiaoqian Yuan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China.
| | - Wenxuan Ye
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Ling Chen
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Doudou Luo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China; State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, 361102, China
| | - Li Zhou
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Yan Qiu
- Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Rengong Zhuo
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Yun Zhao
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Lu Peng
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Lichao Yang
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Xin Jin
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China
| | - Yu Zhou
- Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, China; Key Laboratory of Chiral Drugs, Xiamen, 361102, China; State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
3
|
Boboc IKS, Rotaru-Zavaleanu AD, Calina D, Albu CV, Catalin B, Turcu-Stiolica A. A Preclinical Systematic Review and Meta-Analysis of Behavior Testing in Mice Models of Ischemic Stroke. Life (Basel) 2023; 13:life13020567. [PMID: 36836924 PMCID: PMC9964520 DOI: 10.3390/life13020567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Stroke remains one of the most important causes of death and disability. Preclinical research is a powerful tool for understanding the molecular and cellular response to stroke. However, a lack of standardization in animal evaluation does not always ensure reproducible results. In the present study, we wanted to identify the best strategy for evaluating animal behavior post-experimental stroke. As such, a meta-analysis was made, evaluating behavioral tests done on male C57BL/6 mice subjected to stroke or sham surgery. Overall, fifty-six studies were included. Our results suggest that different types of tests should be used depending on the post-stroke period one needs to analyze. In the hyper-acute, post-stroke period, the best quantifier will be animal examination scoring, as it is a fast and inexpensive way to identify differences between groups. When evaluating stoke mice in the acute phase, a mix of animal examination and motor tests that focus on movement asymmetry (foot-fault and cylinder testing) seem to have the best chance of picking up differences between groups. Complex tasks (the rotarod test and Morris water maze) should be used within the chronic phase to evaluate differences between the late-subacute and chronic phases.
Collapse
Affiliation(s)
- Ianis Kevyn Stefan Boboc
- Department of Pharmacology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- U.M.F. Doctoral School Craiova, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Alexandra Daniela Rotaru-Zavaleanu
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Gastroenterology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Carmen Valeria Albu
- Department of Neurology, Clinical Hospital of Neuropsychiatry, 200473 Craiova, Romania
| | - Bogdan Catalin
- Experimental Research Centre for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Correspondence:
| | - Adina Turcu-Stiolica
- Department of Pharmaceutical Management and Marketing, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
4
|
The Effect of Oleoylethanolamide (OEA) Add-On Treatment on Inflammatory, Oxidative Stress, Lipid, and Biochemical Parameters in the Acute Ischemic Stroke Patients: Randomized Double-Blind Placebo-Controlled Study. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5721167. [PMID: 36120593 PMCID: PMC9477639 DOI: 10.1155/2022/5721167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/01/2022] [Accepted: 06/05/2022] [Indexed: 11/18/2022]
Abstract
Methods Sixty patients with a mean age of 68.60 ± 2.10 comprising 29 females (48.33%), who were admitted to an academic tertiary care facility within the first 12 hours poststroke symptoms onset or last known well (LKW), in case symptom onset time is not clear, were included in this study. AIS was confirmed based on a noncontrast head CT scan and also neurological symptoms. Patients were randomly and blindly assigned to OEA of 300 mg/day (n = 20) or 600 mg/day (n = 20) or placebo (n = 20) in addition to the standard AIS treatment for three days. A blood sample was drawn at 12 hours from symptoms onset or LKW as the baseline followed by the second blood sample at 72 hours post symptoms onset or LKW. Blood samples were assessed for inflammatory and biochemical parameters, oxidative stress (OS) biomarkers, and lipid profile. Results Compared to the baseline, there is a significant reduction in the urea, creatinine, triglyceride, high-density lipoprotein, cholesterol, alanine transaminase, total antioxidant capacity, malondialdehyde (MDA), total thiol groups (TTG), interleukin-6 (IL-6), and C-reactive protein levels on the follow-up blood testing in the OEA (300 mg/day) group. In patients receiving OEA (600 mg/day) treatment, there was only a significant reduction in the MDA level comparing baseline with follow-up blood testing. Also, the between-group analysis revealed a statistically significant difference between patients receiving OEA (300 mg/day) and placebo in terms of IL-6 and TTG level reduction when comparing them between baseline and follow-up blood testing. Conclusion OEA in moderate dosage, 300 mg/day, add-on to the standard stroke treatment improves short-term inflammatory, OS, lipid, and biochemical parameters in patients with AIS. This effect might lead to a better long-term neurological prognosis.
Collapse
|
5
|
Nuclear Receptors in Myocardial and Cerebral Ischemia-Mechanisms of Action and Therapeutic Strategies. Int J Mol Sci 2021; 22:ijms222212326. [PMID: 34830207 PMCID: PMC8617737 DOI: 10.3390/ijms222212326] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Nearly 18 million people died from cardiovascular diseases in 2019, of these 85% were due to heart attack and stroke. The available therapies although efficacious, have narrow therapeutic window and long list of contraindications. Therefore, there is still an urgent need to find novel molecular targets that could protect the brain and heart against ischemia without evoking major side effects. Nuclear receptors are one of the promising targets for anti-ischemic drugs. Modulation of estrogen receptors (ERs) and peroxisome proliferator-activated receptors (PPARs) by their ligands is known to exert neuro-, and cardioprotective effects through anti-apoptotic, anti-inflammatory or anti-oxidant action. Recently, it has been shown that the expression of aryl hydrocarbon receptor (AhR) is strongly increased after brain or heart ischemia and evokes an activation of apoptosis or inflammation in injury site. We hypothesize that activation of ERs and PPARs and inhibition of AhR signaling pathways could be a promising strategy to protect the heart and the brain against ischemia. In this Review, we will discuss currently available knowledge on the mechanisms of action of ERs, PPARs and AhR in experimental models of stroke and myocardial infarction and future perspectives to use them as novel targets in cardiovascular diseases.
Collapse
|
6
|
Jiang Q, Ji A, Li D, Shi L, Gao M, Lv N, Zhang Y, Zhang R, Chen R, Chen W, Zheng Y, Cui L. Mitochondria damage in ambient particulate matter induced cardiotoxicity: Roles of PPAR alpha/PGC-1 alpha signaling. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 288:117792. [PMID: 34280742 DOI: 10.1016/j.envpol.2021.117792] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/06/2021] [Accepted: 07/12/2021] [Indexed: 06/13/2023]
Abstract
Particulate matter (PM) had been associated with cardiotoxicity, while the mechanism of toxicity has yet to be elucidated, with mitochondria dysfunction as a potential candidate. To investigate the potential cardiotoxic effects of ambient PM exposure and assess the damage to cardiac mitochondria, C57/B6 mice were exposed to filtered air or real ambient PM for three or six weeks. Furthermore, to reveal the role of peroxisome proliferators-activated receptor alpha (PPAR alpha) in PM exposure induced cardiotoxicity/mitochondria damage, animals were also co-treated with PPAR alpha agonist WY 14,643 or PPAR alpha antagonist GW 6471. Cardiotoxicity was assessed with echocardiography and histopathology, while mitochondria damage was evaluated with mitochondria membrane potential measurement and transmission electron microscopy. Potential impacts of PM exposure to PPAR alpha signaling were detected with co-immunoprecipitation and western blotting. The results indicated that exposure to ambient PM exposure induced cardiotoxicity in C57/B6 mice, including altered cardiac functional parameters and morphology. Cardiac mitochondria damage is detected, in the form of compromised mitochondria membrane potential and morphology. Molecular investigations revealed disruption of PPAR alpha interaction with peroxisome proliferator-activated receptor gamma coactivator-1A (PGC-1a) as well as altered expression levels of PPAR alpha downstream genes. Co-treatment with WY 14,643 alleviated the observed toxicities, while co-treatment with GW 6471 had mixed results, exaggerating most cardiotoxicity and mitochondrial damage endpoints but alleviating some cardiac functional parameters. Interestingly, WY 14,643 and GW 6471 co-treatment seemed to exhibit similar regulative effects towards PPAR alpha signaling in animals exposed to PM. In conclusion, ambient PM exposure indeed induced cardiotoxicity in C57/B6 mice, in which cardiac mitochondria damage and disrupted PPAR alpha signaling are contributors.
Collapse
Affiliation(s)
- Qixiao Jiang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Andong Ji
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Limei Shi
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Mengyu Gao
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Na Lv
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Ying Zhang
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Rong Zhang
- Department of Toxicology, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Rui Chen
- Department of Toxicology, School of Public Health, Capital Medical University, Beijing, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Yuxin Zheng
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China
| | - Lianhua Cui
- Department of Toxicology, School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
7
|
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
|
8
|
Gaspar JC, Okine BN, Llorente-Berzal A, Roche M, Finn DP. Pharmacological Blockade of PPAR Isoforms Increases Conditioned Fear Responding in the Presence of Nociceptive Tone. Molecules 2020; 25:molecules25041007. [PMID: 32102354 PMCID: PMC7070536 DOI: 10.3390/molecules25041007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 12/29/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors with three isoforms (PPARα, PPARβ/δ, PPARγ) and can regulate pain, anxiety, and cognition. However, their role in conditioned fear and pain-fear interactions has not yet been investigated. Here, we investigated the effects of systemically administered PPAR antagonists on formalin-evoked nociceptive behaviour, fear-conditioned analgesia (FCA), and conditioned fear in the presence of nociceptive tone in rats. Twenty-three and a half hours following fear conditioning to context, male Sprague-Dawley rats received an intraplantar injection of formalin and intraperitoneal administration of vehicle, PPARα (GW6471), PPARβ/δ (GSK0660) or PPARγ (GW9662) antagonists, and 30 min later were re-exposed to the conditioning arena for 15 min. The PPAR antagonists did not alter nociceptive behaviour or fear-conditioned analgesia. The PPARα and PPARβ/δ antagonists prolonged context-induced freezing in the presence of nociceptive tone without affecting its initial expression. The PPARγ antagonist potentiated freezing over the entire trial. In conclusion, pharmacological blockade of PPARα and PPARβ/δ in the presence of formalin-evoked nociceptive tone, impaired short-term, within-trial fear-extinction in rats without affecting pain response, while blockade of PPARγ potentiated conditioned fear responding. These results suggest that endogenous signalling through these three PPAR isoforms may reduce the expression of conditioned fear in the presence of nociceptive tone.
Collapse
Affiliation(s)
- Jessica C. Gaspar
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Bright N. Okine
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Alvaro Llorente-Berzal
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - Michelle Roche
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Centre for Pain Research, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
- Physiology Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
| | - David P. Finn
- Pharmacology and Therapeutics Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland; (J.C.G.); (B.N.O.); (A.L.-B.)
- Galway Neuroscience Centre, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland;
- Physiology Department, National University of Ireland Galway, University Road, H91 W5P7 Galway, Ireland
- Correspondence: ; Tel.: +353-(0)91-495-280; Fax: +353-(0)91-495-586
| |
Collapse
|
9
|
Luo D, Zhang Y, Yuan X, Pan Y, Yang L, Zhao Y, Zhuo R, Chen C, Peng L, Li W, Jin X, Zhou Y. Oleoylethanolamide inhibits glial activation via moudulating PPARα and promotes motor function recovery after brain ischemia. Pharmacol Res 2019; 141:530-540. [PMID: 30660821 DOI: 10.1016/j.phrs.2019.01.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/29/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
Glial activation and scar formation impede the neurological function recovery after cerebral ischemia. Oleoylethanolamide (OEA), a bioactive lipid mediator, shows neuroprotection against acute brain ischemia, however, its long-term effect, especially on glial scar formation, has not been characterized. In this research, we investigate the effect of OEA on glial activation and scar formation after cerebral ischemia in vitro and in vivo experiments. Glial scar formation in vitro model was induced by transforming growth factor β1 (TGF-β1) in C6 glial cell culture, and experiment model in vivo was induced by middle cerebral artery occlusion (MCAO) in mice. The protein expressions of the markers of glial activation (S100β, GFAP, or pSmads) and glial scar (neurocan) were detected by Western blot and/or immunofluorescence staining; To evaluate the role of PPARɑ in the effect of OEA on glial activation, the PPARɑ antagonist GW6471 was used. Behavior tests were used to assay the effect of OEA on motor function recovery 14 days after brain ischemia in mice. Our results show that OEA (10-50 μM) concentration-dependently inhibited the upregulation of S100β, GFAP, pSmads and neurocan induced by TGF-β1 in C6 glial cells. At the same time, OEA promoted the protein expression and nuclear transportation of PPARɑ in glial cells. PPARα antagonist GW6471 abolished the effect of OEA on glial activation. In addition, we found that delay administration of OEA inhibited the astrocyte activation and promoted the recovery of motor function after brain ischemia in mice. These results indicate that OEA may be developed into a new candidate for attenuating astrocytic scar formation and improving motor function after ischemic stroke.
Collapse
Affiliation(s)
- Doudou Luo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yali Zhang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China; Medical College, Xuchang University, Xuchang, 461000, PR China
| | - Xiaoqian Yuan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yilin Pan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China
| | - Lichao Yang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yun Zhao
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Rengong Zhuo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Caixia Chen
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Lu Peng
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Wenjun Li
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Xin Jin
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| | - Yu Zhou
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| |
Collapse
|
10
|
Holubiec MI, Romero JI, Suárez J, Portavella M, Fernández-Espejo E, Blanco E, Galeano P, de Fonseca FR. Palmitoylethanolamide prevents neuroinflammation, reduces astrogliosis and preserves recognition and spatial memory following induction of neonatal anoxia-ischemia. Psychopharmacology (Berl) 2018; 235:2929-2945. [PMID: 30058012 DOI: 10.1007/s00213-018-4982-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 07/23/2018] [Indexed: 11/27/2022]
Abstract
RATIONAL Neonatal anoxia-ischemia (AI) particularly affects the central nervous system. Despite the many treatments that have been tested, none of them has proven to be completely successful. Palmitoylethanolamide (PEA) and oleoylethanolamide (OEA) are acylethanolamides that do not bind to CB1 or CB2 receptors and thus they do not present cannabinoid activity. These molecules are agonist compounds of peroxisome proliferator-activator receptor alpha (PPARα), which modulates the expression of different genes that are related to glucose and lipid metabolism, inflammation, differentiation and proliferation. OBJECTIVE In the present study, we analyzed the effects that the administration of PEA or OEA, after a neonatal AI event, has over different areas of the hippocampus. METHODS To this end, 7-day-old rats were subjected to AI and then treated with vehicle, OEA (2 or 10 mg/kg) or PEA (2 or 10 mg/kg). At 30 days of age, animals were subjected to behavioral tests followed by immunohistochemical studies. RESULTS Results showed that neonatal AI was associated with decreased locomotion, as well as recognition and spatial memory impairments. Furthermore, these deficits were accompanied with enhanced neuroinflammation and astrogliosis, as well as a decreased PPARα expression. PEA treatment was able to prevent neuroinflammation, reduce astrogliosis and preserve cognitive functions. CONCLUSIONS These results indicate that the acylethanolamide PEA may play an important role in the mechanisms underlying neonatal AI, and it could be a good candidate for further studies regarding neonatal AI treatments.
Collapse
Affiliation(s)
- Mariana I Holubiec
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan I Romero
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain
| | - Manuel Portavella
- Laboratorio de Conducta Animal y Neurociencia, Departamento de Psicología Experimental, Facultad de Psicología, Universidad de Sevilla, C/Camilo José Cela s/n, 41018, Sevilla, Spain
| | - Emilio Fernández-Espejo
- Laboratorio de Neurofisiología y Neurología Molecular, Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Av. Sánchez Pizjuán 4, 41009, Sevilla, Spain
| | - Eduardo Blanco
- Lleida Institute for Biomedical Research, Dr. Pifarré Foundation (IRBLleida), University of Lleida, Av. Alcalde Rovira Roure 80, 25198, Lleida, Spain
| | - Pablo Galeano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain.
- Instituto de Investigaciones Bioquímicas de Buenos Aires (IIBBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Av. Patricias Argentinas 435, C1405BWE, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Avenida Carlos Haya 82, 29010, Málaga, Spain.
| |
Collapse
|