1
|
Dettori I, Bulli I, Venturini M, Magni G, Cherchi F, Rossi F, Lee H, Pedata F, Jacobson KA, Pugliese AM, Coppi E. MRS3997, a dual adenosine A 2A/A 2B receptor agonist, reduces brain ischemic damage and alleviates neuroinflammation in rats. Neuropharmacology 2025; 262:110214. [PMID: 39522676 PMCID: PMC11789432 DOI: 10.1016/j.neuropharm.2024.110214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The endogenous neuromodulator adenosine is massively released during hypoxic/ischemic insults and differentially modulates post-ischemic damage depending on the expression and recruitment of its four metabotropic receptor subtypes, namely A1, A2A, A2B and A3 receptors (A1Rs, A2ARs, A2BRs and A3Rs). We previously demonstrated, by using a model of transient middle cerebral artery occlusion (tMCAo) in rats, that selective activation of A2ARs, as well as A2BRs, ameliorates post-ischemic brain damage in contrast to neuroinflammation. In the present study, we investigated whether the multitarget nucleoside MRS3997, a full agonist at both A2ARs and A2BRs, would afford higher neuroprotection in post-ischemic damage. Chronic systemic treatment with MRS3997 reduced neurological deficit, body weight loss and infarct volume in the cortex and striatum measured 7 days after ischemia. The dual agonist counteracted neuronal loss, reduced myelin damage, and prevented morphological changes indicative of microglia and astrocyte activation. Finally, MRS3997 shifted plasma cytokine levels to an anti-inflammatory profile. These effects were preceded, at 2 days after the insult, by a reduced granulocyte infiltration in the ischemic cortex and, differently from what was observed with selective A2AR or A2BR agonism, also in striatum. In summary, we demonstrate here that MRS3997, systemically administered for 7 days after tMCAO, protects ischemic areas from neuronal and glial damage and inhibits neuroinflammation, therefore representing an attractive strategy to ameliorate post-stroke damage and neurological symptoms.
Collapse
Affiliation(s)
- Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Irene Bulli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Martina Venturini
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Giada Magni
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Sesto Fiorentino, Florence, Italy
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Francesca Rossi
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Sesto Fiorentino, Florence, Italy
| | - Hobin Lee
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabe-tes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabe-tes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anna Maria Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
2
|
Nairuz T, Heo JC, Lee JH. Differential Glial Response and Neurodegenerative Patterns in CA1, CA3, and DG Hippocampal Regions of 5XFAD Mice. Int J Mol Sci 2024; 25:12156. [PMID: 39596222 PMCID: PMC11594373 DOI: 10.3390/ijms252212156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/04/2024] [Accepted: 11/09/2024] [Indexed: 11/28/2024] Open
Abstract
In this study, the distinct patterns of glial response and neurodegeneration within the CA1, CA3, and dentate gyrus (DG) regions of the hippocampus were examined in 5XFAD mice at 6 and 12 months of age. The primary feature of this transgenic mouse model is the rapid onset of amyloid pathology. We employed quantitative assessments via immunohistochemistry, incorporating double staining techniques, followed by observation with light microscopy and subsequent digital analysis of microscopic images. We identified significantly increased Aβ deposition in these three hippocampal regions at 6 and 12 months of transgenic mice. Moreover, the CA1 and CA3 regions showed higher vulnerability, with signs of reactive astrogliosis such as increased astrocyte density and elevated GFAP expression. Additionally, we observed a significant rise in microglia density, along with elevated inflammatory markers (TNFα) in these hippocampal regions. These findings highlight a non-uniform glial and neuronal response to Aβ plaque deposition within the hippocampal regions of 5xFAD mice, potentially contributing to the neurodegenerative and memory deficit characteristics of Alzheimer's disease in this model.
Collapse
Affiliation(s)
| | | | - Jong-Ha Lee
- Department of Biomedical Engineering, Keimyung University, Daegu 42601, Republic of Korea; (T.N.); (J.-C.H.)
| |
Collapse
|
3
|
Lana D, Traini C, Bulli I, Sarti G, Magni G, Attorre S, Giovannini MG, Vannucchi MG. Chronic administration of prebiotics and probiotics ameliorates pathophysiological hallmarks of Alzheimer's disease in a APP/PS1 transgenic mouse model. Front Pharmacol 2024; 15:1451114. [PMID: 39166107 PMCID: PMC11333230 DOI: 10.3389/fphar.2024.1451114] [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: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
Introduction: The gut microbiota (MB), although one of the main producers of Aβ in the body, in physiological conditions contributes to the maintainance of a healthy brain. Dysbiosis, the dysbalance between Gram-negative and Gram-positive bacteria in the MB increases Aβ production, contributing to the accumulation of Aβ plaques in the brain, the main histopathological hallmark of Alzheimer's disease (AD). Administration of prebiotics and probiotics, maintaining or recovering gut-MB composition, could represent a nutraceutical strategy to prevent or reduce AD sympthomathology. Aim of this research was to evaluate whether treatment with pre- and probiotics could modify the histopathological signs of neurodegeneration in hippocampal CA1 and CA3 areas of a transgenic mouse model of AD (APP/PS1 mice). The hippocampus is one of the brain regions involved in AD. Methods: Tg mice and Wt littermates (Wt-T and Tg-T) were fed daily for 6 months from 2 months of age with a diet supplemented with prebiotics (a multi-extract of fibers and plant complexes, containing inulin/fruit-oligosaccharides) and probiotics (a 50%-50% mixture of Lactobacillus rhamnosus and Lactobacillus paracasei). Controls were Wt and Tg mice fed with a standard diet. Brain sections were immunostained for Aβ plaques, neurons, astrocytes, microglia, and inflammatory proteins that were evaluated qualitatively and quantitatively by immunofluorescence, confocal microscopy and digital imaging with ImageJ software. Results: Quantitative analyses demonstrated that: 1) The treatment with pre- and probiotics significantly decreased Aβ plaques in CA3, while in CA1 the reduction was not significant; 2) Neuronal damage in CA1 Stratum Pyramidalis was significantly prevented in Tg-T mice; no damage was found in CA3; 3) In both CA1 and CA3 the treatment significantly increased astrocytes density, and GFAP and IBA1 expression, especially around plaques; 4) Microglia reacted differently in CA1 and CA3: in CA3 of Tg-T mice there was a significant increase of CD68+ phagocytic microglia (ball-and-chain phenomic) and of CX3CR1 compared with CA1. Discussion: The higher microglia reactivity could be responsible for their more efficient scavenging activity towards Aβ plaques in CA3 in comparison to CA1. Treatment with pre- and probiotics, modifying many of the physiopathological hallmarks of AD, could be considered an effective nutraceutical strategy against AD symptomatology.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Chiara Traini
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Irene Bulli
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giorgia Sarti
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giada Magni
- Cnr — Istituto di Fisica Applicata “Nello Carrara”, Sesto Fiorentino, Italy
| | - Selene Attorre
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Maria Giuliana Vannucchi
- Research Unit of Histology and Embryology, Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
4
|
Gáll Z, Csüdör Á, Sável IG, Kelemen K, Kolcsár M. Cholecalciferol Supplementation Impacts Behavior and Hippocampal Neuroglial Reorganization in Vitamin D-Deficient Rats. Nutrients 2024; 16:2326. [PMID: 39064769 PMCID: PMC11279879 DOI: 10.3390/nu16142326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Vitamin D deficiency (VDD) is widespread around the world and has been extensively documented to affect various health conditions, including the cognitive functioning of the brain. Serum 25-hydroxylated forms of vitamin D are traditionally used to determine vitamin D status. However, there is now evidence that cholecalciferol activation can occur and be controlled by locally expressed enzymes in the brain. This study aimed to investigate the effects of cholecalciferol supplementation on cognitive function in rats who underwent transient VDD in adulthood. Thirty-six adult Wistar rats were administered paricalcitol (seven doses of 32 ng injected every other day) along with a "vitamin D-free" diet to induce VDD, which was confirmed using a LC-MS/MS serum analysis of the cholecalciferol and 25-hydroxyvitamin D3 levels. Treatment was performed by including 1000 IU/kg and 10,000 IU/kg cholecalciferol in the diet. Cognitive performance was evaluated using the novel object recognition (NOR), Morris water maze (MWM), and radial arm maze (RAM) tests. An immunohistochemical analysis of the brain regions involved in learning and memory was performed by quantifying the neurons, astrocytes, and microglia labelled with anti-neuronal nuclei (NeuN), glial fibrillary acidic protein (GFAP), and ionized calcium-binding adaptor molecule 1 (Iba-1) antibodies, respectively. The vitamin D deficient group showed the lowest performance in both the MWM and RAM tests. In contrast, the cholecalciferol-treated groups exhibited a faster learning curve. However, no difference was detected between the groups in the NOR test. On the other hand, differences in the cellular organization of the hippocampus and amygdala were observed between the groups. Cholecalciferol supplementation decreased the density of the Iba-1- and GFAP-labeled cells in the hilus and cornu Ammonis 3 (CA3) regions of the hippocampus and in the amygdala. These results support vitamin D's substantial role in learning and memory. They also highlight that subtle changes of cognitive function induced by transient VDD could be reversed by cholecalciferol supplementation. Further studies are needed to better understand VDD and cholecalciferol's effects on the brain structure and function.
Collapse
Affiliation(s)
- Zsolt Gáll
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Ágnes Csüdör
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - István-Gábor Sável
- Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Krisztina Kelemen
- Department of Physiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| | - Melinda Kolcsár
- Department of Pharmacology and Clinical Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Târgu Mureș, Romania;
| |
Collapse
|
5
|
Martini APR, Schlemmer LM, Lucio Padilha JA, Fabres RB, Couto Pereira NDS, Pereira LO, Dalmaz C, Netto CA. Acrobatic training prevents learning impairments and astrocyte remodeling in the hippocampus of rats undergoing chronic cerebral hypoperfusion: sex-specific benefits. FRONTIERS IN REHABILITATION SCIENCES 2024; 5:1375561. [PMID: 38939055 PMCID: PMC11208732 DOI: 10.3389/fresc.2024.1375561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
Background Chronic cerebral hypoperfusion (CCH) leads to memory and learning impairments associated with degeneration and gliosis in the hippocampus. Treatment with physical exercise carries different therapeutic benefits for each sex. We investigated the effects of acrobatic training on astrocyte remodeling in the CA1 and CA3 subfields of the hippocampus and spatial memory impairment in male and female rats at different stages of the two-vessel occlusion (2VO) model. Methods Wistar rats were randomly allocated into four groups of males and females: 2VO acrobatic, 2VO sedentary, sham acrobatic, and sham sedentary. The acrobatic training was performed for 4 weeks prior to the 2VO procedure. Brain samples were collected for morphological and biochemical analysis at 3 and 7 days after 2VO. The dorsal hippocampi were removed and prepared for Western blot quantification of Akt, p-Akt, COX IV, cleaved caspase-3, PARP, and GFAP. GFAP immunofluorescence was performed on slices of the hippocampus to count astrocytes and apply the Sholl's circle technique. The Morris water maze was run after 45 days of 2VO. Results Acutely, the trained female rats showed increased PARP expression, and the 2VO-trained rats of both sexes presented increased GFAP levels in Western blot. Training, mainly in males, induced an increase in the number of astrocytes in the CA1 subfield. The 2VO rats presented branched astrocytes, while acrobatic training prevented branching. However, the 2VO-induced spatial memory impairment was partially prevented by the acrobatic training. Conclusion Acrobatic training restricted the astrocytic remodeling caused by 2VO in the CA1 and CA3 subfields of the hippocampus. The improvement in spatial memory was associated with more organized glial scarring in the trained rats and better cell viability observed in females.
Collapse
Affiliation(s)
- Ana Paula Rodrigues Martini
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Livia Machado Schlemmer
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Joelma Alves Lucio Padilha
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Bandeira Fabres
- Department of Pediatrics, NorthShore University HealthSystem, Evanston, IL, United States
- Pritzker School of Medicine, University of Chicago, Chicago, IL, United States
| | - Natividade de Sá Couto Pereira
- Psychological Neuroscience Laboratory, Psychology Research Center, School of Psychology, University of Minho, Braga, Portugal
| | - Lenir Orlandi Pereira
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carla Dalmaz
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Carlos Alexandre Netto
- Graduate Program in Neuroscience, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Graduate Program in Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
6
|
Zhao Z, Xie L, Shi J, Liu T, Wang S, Huang J, Wu D, Zhang X. Neuroprotective Effect of Zishen Huoxue Decoction treatment on Vascular Dementia by activating PINK1/Parkin mediated Mitophagy in the Hippocampal CA1 Region. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117172. [PMID: 37709106 DOI: 10.1016/j.jep.2023.117172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zishen Huoxue Decoction (ZSHXD) is a Traditional Chinese Medicine (TCM) prescription for the treatment of vascular dementia (VD). Although the clinical effects of ZSHXD have been demonstrated, the molecular mechanisms underlying the neuroprotective effects of ZSHXD remain unclear. AIM OF THE STUDY To explore whether the neuroprotective effect of Zishen Huoxue Decoction (ZSHXD) treatment is associated with the PINK1/Parkin pathway-mediated mitophagy in hippocampal CA1 region of 2-VO model rats. MATERIALS AND METHODS Seventy-two male SD rats were randomly divided into the sham group, model group, Donepezil (0.45 mg/kg) group, ZSHXD low dose group (8.9 g/kg), ZSHXD medium dose group (17.8 g/kg), and ZSHXD high dose group (35.6 g/kg). Two-vessel occlusion (2-VO) rat model is established to evaluate the therapeutic effect of ZSHXD pretreatment. Hematoxylin-eosin (HE) staining is conducted to detect the morphological changes of neurons and the number of normal neurons in the hippocampal CA1 region. Then, the mitochondrial function and structure were reflected by the mitochondrial membrane potential (MMP) levels and transmission electron microscopy (TEM). Meanwhile, the expression of mitophagy related proteins mediated by PINK1/Parkin was detected by western blot (WB). After that, malondialdehyde (MDA) and superoxide dismutase (SOD) levels were measured by Elisa. At last, the apoptosis-related proteins Caspase-3、Bax、Bcl-2 were measured by WB. RESULTS The results depict that ZSHXD has dose-dependently improved the cognitive function in 2-VO model rats. It has also been showed that ZSHXD can alleviate neuron damage, rescue the mitochondrial structural injury and dysfunction in hippocampal CA1 region. Besides, ZSHXD has increased the activity of SOD and decreased the activity of MDA. In addition, ZSHXD can inhibit apoptosis with Caspase-3, Bax decreasing and Bcl-2 increasing. Specially, the protection of ZSHXD showed in 2-VO model rats is along with the upregulation of PINK1, Parkin and LC3-Ⅱ/Ⅰ, and downregulation of p62 in the hippocampal CA1 region. CONCLUSIONS This study reveals that ZSHXD protects the 2-VO model rats from ischemic injury by activating the PINK1/Parkin-mediated mitophagy in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Ziting Zhao
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Le Xie
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China
| | - Jiayi Shi
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China
| | - Tonghe Liu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China
| | - Shiliang Wang
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China
| | - Jianhua Huang
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410006, Hunan Province, China
| | - Dahua Wu
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China.
| | - Xiuli Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China.
| |
Collapse
|
7
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
8
|
Yu M, Zhang M, Fu P, Wu M, Yin X, Chen Z. Research progress of mitophagy in chronic cerebral ischemia. Front Aging Neurosci 2023; 15:1224633. [PMID: 37600521 PMCID: PMC10434995 DOI: 10.3389/fnagi.2023.1224633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Chronic cerebral ischemia (CCI), a condition that can result in headaches, dizziness, cognitive decline, and stroke, is caused by a sustained decrease in cerebral blood flow. Statistics show that 70% of patients with CCI are aged > 80 years and approximately 30% are 45-50 years. The incidence of CCI tends to be lower, and treatment for CCI is urgent. Studies have confirmed that CCI can activate the corresponding mechanisms that lead to mitochondrial dysfunction, which, in turn, can induce mitophagy to maintain mitochondrial homeostasis. Simultaneously, mitochondrial dysfunction can aggravate the insufficient energy supply to cells and various diseases caused by CCI. Regulation of mitophagy has become a promising therapeutic target for the treatment of CCI. This article reviews the latest progress in the important role of mitophagy in CCI and discusses the induction pathways of mitophagy in CCI, including ATP synthesis disorder, oxidative stress injury, induction of reactive oxygen species, and Ca2+ homeostasis disorder, as well as the role of drugs in CCI by regulating mitophagy.
Collapse
Affiliation(s)
- Mayue Yu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Manqing Zhang
- School of Basic Medicine, Jiujiang University, Jiujiang, Jiangxi, China
| | - Peijie Fu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Moxin Wu
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Xiaoping Yin
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| | - Zhiying Chen
- Department of Neurology, Clinical Medical School of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, China
| |
Collapse
|
9
|
Wang H, Liu Y, Guo Z, Cui M, Pang P, Yang J, Wu C. Enhancement of oligodendrocyte autophagy alleviates white matter injury and cognitive impairment induced by chronic cerebral hypoperfusion in rats. Acta Pharm Sin B 2023; 13:2107-2123. [DOI: 10.1016/j.apsb.2023.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/23/2022] [Accepted: 02/28/2023] [Indexed: 03/18/2023] Open
|
10
|
Cannabidiol inhibits microglia activation and mitigates neuronal damage induced by kainate in an in-vitro seizure model. Neurobiol Dis 2022; 174:105895. [DOI: 10.1016/j.nbd.2022.105895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
|
11
|
Lana D, Landucci E, Mazzantini C, Magni G, Pellegrini-Giampietro DE, Giovannini MG. The Protective Effect of CBD in a Model of In Vitro Ischemia May Be Mediated by Agonism on TRPV2 Channel and Microglia Activation. Int J Mol Sci 2022; 23:12144. [PMID: 36292998 PMCID: PMC9603301 DOI: 10.3390/ijms232012144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 09/21/2023] Open
Abstract
Cannabinoids, used for centuries for recreational and medical purposes, have potential therapeutic value in stroke treatment. Cannabidiol (CBD), a non-psychoactive compound and partial agonist of TRPV2 channels, is efficacious in many neurological disorders. We investigated the effects of CBD or Δ9-tetrahydrocannabinol (THC) in rat organotypic hippocampal slices exposed to oxygen-glucose deprivation (OGD), an in vitro model of ischemia. Neuronal TRPV2 expression decreased after OGD, but it increased in activated, phagocytic microglia. CBD increased TRPV2 expression, decreased microglia phagocytosis, and increased rod microglia after OGD. THC had effects contrary to those of CBD. Our results show that cannabinoids have different effects in ischemia. CBD showed neuroprotective effects, mediated, at least in part, by TRPV2 channels, since the TRPV2 antagonist tranilast blocked them, while THC worsened the neurodegeneration caused by ischemia. In conclusion, our results suggest that different cannabinoid molecules play different roles in the mechanisms of post-ischemic neuronal death. These different effects of cannabinoid observed in our experiments caution against the indiscriminate use of cannabis or cannabinoid preparations for recreational or therapeutic use. It was observed that the positive effects of CBD may be counteracted by the negative effects caused by high levels of THC.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Costanza Mazzantini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Sesto Fiorentino, Italy
| | | | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy
| |
Collapse
|
12
|
Wang X, Xiao A, Yang Y, Zhao Y, Wang CC, Wang Y, Han J, Wang Z, Wen M. DHA and EPA Prevent Seizure and Depression-Like Behavior by Inhibiting Ferroptosis and Neuroinflammation via Different Mode-of-actions in a Pentylenetetrazole-Induced Kindling Model in Mice. Mol Nutr Food Res 2022; 66:e2200275. [PMID: 36099650 DOI: 10.1002/mnfr.202200275] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/07/2022] [Indexed: 11/06/2022]
Abstract
SCOPE It has been reported that eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) have anticonvulsant effects, yet the respective mechanism of EPA and DHA on epilepsy are still unclarified. This study aimed to investigate the effect of EPA and DHA on pentylenetetrazol (PTZ) induced seizures and depression. METHODS AND RESULTS The administration of EPA and DHA at a dose of 1% (w/w) significantly inhibited PTZ-induced seizures and depressive-like behavior, whereas EPA outcompetes DHA. Further mechanistic studies revealed that the higher effect of EPA can be partly attributed to the promotion of M2 polarization, inhibition of M1 polarization of microglia, and lower iron content in the brain, resulting from the stronger activation of nuclear factor E2-related factor 2 (Nrf2). We found that DHA and EPA comparably inhibited NLRP3 inflammasome activation but with different mode-of-actions: EPA preferred to inhibit the binding of NLRP3 and ASC, while DHA decreased the protein levels of ASC and Caspase-1. CONCLUSIONS These results indicated that DHA and EPA could efficaciously alleviate PTZ-induced seizure and depressive-like behavior but with different efficiency and molecular mechanisms. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xueyan Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| | - Aiai Xiao
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| | - Yueqi Yang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| | - Yingcai Zhao
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Cheng Cheng Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Yuming Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, 266003, China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, 252059, China
| |
Collapse
|
13
|
Anticonvulsant Action and Long-Term Effects of Chronic Cannabidiol Treatment in the Rat Pentylenetetrazole-Kindling Model of Epilepsy. Biomedicines 2022; 10:biomedicines10081811. [PMID: 36009358 PMCID: PMC9405483 DOI: 10.3390/biomedicines10081811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Cannabidiol (CBD) showed anticonvulsant action in several preclinical models and is currently approved by regulatory agencies to treat childhood epilepsy syndromes. However, CBD treatment has limited benefits, and its long-term effects on cognition are not fully understood yet. This study aimed to examine the impact of long-term CBD treatment in the pentylenetetrazole (PTZ)-kindling model of epilepsy. Adult male Wistar rats (N = 24) received PTZ (35 mg/kg intraperitoneally) every other day until two consecutive generalized seizures occurred. CBD (60 mg/kg body weight) was administered daily by the oral route until the kindled state was achieved (n = 12). To confirm that the formulation and administration techniques were not of concern, liquid chromatography–mass spectrometry was performed to test the brain penetration of the CBD formula. As a result of CBD treatment, a lower mortality rate and significantly prolonged generalized seizure latency (925.3 ± 120.0 vs. 550.1 ± 69.62 s) were observed, while the frequency and duration of generalized seizures were not influenced. The CBD-treated group showed a significant decrease in vertical exploration in the open field test and a significant decrease in the discrimination index in the novel object recognition (NOR) test (−0.01 ± 0.17 vs. 0.57 ± 0.15, p = 0.04). The observed behavioral characteristics may be connected to the decreased thickness of the stratum pyramidale or the decreased astrogliosis observed in the hippocampus. In conclusion, CBD treatment did not prevent kindling, nor did it affect seizure frequency or duration. However, it did increase the latency to the first seizure and decreased the prolonged status epilepticus-related mortality in PTZ-kindled rats. The cognitive impairment observed in the NOR test may be related to the high dose used in this study, which may warrant further investigation.
Collapse
|
14
|
Martini APR, Hoeper E, Pedroso TA, Carvalho AVS, Odorcyk FK, Fabres RB, Pereira NDSC, Netto CA. Effects of acrobatic training on spatial memory and astrocytic scar in CA1 subfield of hippocampus after chronic cerebral hypoperfusion in male and female rats. Behav Brain Res 2022; 430:113935. [PMID: 35605797 DOI: 10.1016/j.bbr.2022.113935] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/07/2022] [Accepted: 05/17/2022] [Indexed: 12/22/2022]
Abstract
Chronic cerebral hypoperfusion leads to neuronal loss in the hippocampus and spatial memory impairments. Physical exercise is known to prevent cognitive deficits in animal models; and there is evidence of sex differences in behavioral neuroprotective approaches. The aim of present study was to investigate the effects of acrobatic training in male and female rats submitted to chronic cerebral hypoperfusion. Males and females rats underwent 2VO (two-vessel occlusion) surgery and were randomly allocated into 4 groups of males and 4 groups of females, as follows: 2VO acrobatic, 2VO sedentary, Sham acrobatic and Sham sedentary. The acrobatic training started 45 days after surgery and lasted 4 weeks; animals were then submitted to object recognition and water maze testing. Brain samples were collected for histological and morphological assessment and flow cytometry. 2VO causes cognitive impairments and acrobatic training prevented spatial memory deficits assessed in the water maze, mainly for females. Morphological analysis showed that 2VO animals had less NeuN labeling and acrobatic training prevented it. Increased number of GFAP positive cells was observerd in females; moreover, males had more branched astrocytes and acrobatic training prevented the branching after 2VO. Flow cytometry showed higher mitochondrial potential in trained animals and more reactive oxygen species production in males. Acrobatic training promoted neuronal survival and improved mitochondrial function in both sexes, and influenced the glial scar in a sex-dependent manner, associated to greater cognitive benefit to females after chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Ana Paula Rodrigues Martini
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| | - Eduarda Hoeper
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduation in Biological Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Thales Avila Pedroso
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduation in Physical Therapy, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Andrey Vinicios Soares Carvalho
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Felipe Kawa Odorcyk
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Rafael Bandeira Fabres
- Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Natividade de Sá Couto Pereira
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Graduate Program in Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil; Institute of Basic Health Sciences, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| |
Collapse
|
15
|
Choudhary RC, Ahmed U, Shoaib M, Alper E, Rehman A, Kim J, Shinozaki K, Volpe BT, Chavan S, Zanos S, Tracey KJ, Becker LB. Threshold adjusted vagus nerve stimulation after asphyxial cardiac arrest results in neuroprotection and improved survival. Bioelectron Med 2022; 8:10. [PMID: 35854394 PMCID: PMC9297561 DOI: 10.1186/s42234-022-00092-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/24/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Vagus nerve stimulation (VNS) has shown therapeutic potential in a variety of different diseases with many ongoing clinical trials. The role of VNS in reducing ischemic injury in the brain requires further evaluation. Cardiac arrest (CA) causes global ischemia and leads to the injury of vital organs, especially the brain. In this study, we investigated the protective effects of customized threshold-adjusted VNS (tVNS) in a rat model of CA and resuscitation. METHODS Sprague-Dawley rats underwent 12 min asphyxia-CA followed by resuscitation. Rats were assigned to either post-resuscitation tVNS for 2 h or no-tVNS (control). tVNS was applied by electrode placement in the left cervical vagus nerve. To optimize a threshold, we used animal's heart rate and determined a 15-20% drop from baseline levels as the effective and physiological threshold for each animal. The primary endpoint was 72 h survival; secondary endpoints included neurological functional recovery, reduction in brain cellular injury (histopathology), cardiac and renal injury parameters (troponin I and creatinine levels, respectively). RESULTS In comparison to the control group, tVNS significantly improved 72 h survival and brain functional recovery after 12 minutes of CA. The tVNS group demonstrated significantly reduced numbers of damaged neurons in the CA1 hippocampal region of the brain as compared to the control group. Similarly, the tVNS group showed decreased trend in plasma troponin I and creatinine levels as compared to the control group. CONCLUSIONS Our findings suggest that using tVNS for 2 h after 12 minutes of CA attenuates ischemia neuronal cell death, heart and kidney damage, and improves 72 h survival with improved neurological recovery.
Collapse
Affiliation(s)
- Rishabh C Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY, USA
| | - Umair Ahmed
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Muhammad Shoaib
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Eric Alper
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Abdul Rehman
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA
| | - Junhwan Kim
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Koichiro Shinozaki
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA
- Department of Emergency Medicine, Northwell Health, Manhasset, NY, USA
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Bruce T Volpe
- Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Center for Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
| | - Sangeeta Chavan
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Stavros Zanos
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Kevin J Tracey
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Lance B Becker
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, 350 Community Dr, Manhasset, NY, 11030, USA.
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
- Department of Emergency Medicine, Northwell Health, Manhasset, NY, USA.
| |
Collapse
|
16
|
Shoaib M, Choudhary RC, Chillale RK, Kim N, Miyara SJ, Haque S, Yin T, Frankfurt M, Molmenti EP, Zanos S, Kim J, Becker LB. Metformin-mediated mitochondrial protection post-cardiac arrest improves EEG activity and confers neuroprotection and survival benefit. FASEB J 2022; 36:e22307. [PMID: 35394702 DOI: 10.1096/fj.202200121r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 12/25/2022]
Abstract
Cardiac arrest (CA) produces global ischemia/reperfusion injury resulting in substantial multiorgan damage. There are limited efficacious therapies to save lives despite CA being such a lethal disease process. The small population of surviving patients suffer extensive brain damage that results in substantial morbidity. Mitochondrial dysfunction in most organs after CA has been implicated as a major source of injury. Metformin, a first-line treatment for diabetes, has shown promising results in the treatment for other diseases and is known to interact with the mitochondria. For the treatment of CA, prior studies have utilized metformin in a preconditioning manner such that animals are given metformin well before undergoing CA. As the timing of CA is quite difficult to predict, the present study, in a clinically relevant manner, sought to evaluate the therapeutic benefits of metformin administration immediately after resuscitation using a 10 min asphxyial-CA rat model. This is the first study to show that metformin treatment post-CA (a) improves 72 h survival and neurologic function, (b) protects mitochondrial function with a reduction in apoptotic brain injury without activating AMPK, and (c) potentiates earlier normalization of brain electrophysiologic activity. Overall, as an effective and safe drug, metformin has the potential to be an easily translatable intervention for improving survival and preventing brain damage after CA.
Collapse
Affiliation(s)
- Muhammad Shoaib
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Rishabh C Choudhary
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Emergency Medicine, Northwell Health, Manhasset, New York, USA
| | - Rupesh K Chillale
- Neural System Laboratory, University of Maryland, College Park, Maryland, USA
| | - Nancy Kim
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Santiago J Miyara
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Elmezzi Graduate School of Molecular Medicine, Manhasset, New York, USA
| | - Shabirul Haque
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Tai Yin
- Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Maya Frankfurt
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Molecular Medicine and Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | | | - Stavros Zanos
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Junhwan Kim
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Emergency Medicine, Northwell Health, Manhasset, New York, USA.,Molecular Medicine and Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Lance B Becker
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA.,Laboratory for Critical Care Physiology, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Department of Emergency Medicine, Northwell Health, Manhasset, New York, USA.,Molecular Medicine and Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
17
|
Blockade on Lin28a Prevents Cognitive Impairment and Disruption of the Blood-Brain Barrier Induced by Chronic Cerebral Hypoperfusion. Biomedicines 2022; 10:biomedicines10040852. [PMID: 35453602 PMCID: PMC9029709 DOI: 10.3390/biomedicines10040852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/22/2022] [Accepted: 04/01/2022] [Indexed: 12/10/2022] Open
Abstract
Lin28a is an RNA-binding protein involved in the translation and regulation of multiple mRNAs. Lin28a is overexpressed in animal models of brain injury. Similarly, our preliminary study found increased Lin28a expression levels in the animal models four to seven days after chronic cerebral hypoperfusion. Therefore, this current study aimed to evaluate the effects of modulating Lin28a on cognition and brain functions. Vascular dementia (VaD) was induced in 12-week-old male Wistar rats using permanent bilateral common carotid artery occlusion (BCCAO), and these rats were treated with Lin28a siRNA on the fourth and seventh day after BCCAO. From the 42nd day after BCCAO, cognitive behavioral experiments were performed for two weeks. VaD induced by BCCAO resulted in cognitive impairment and microglial activation. Lin28a expression was upregulated after BCCAO. Lin28a siRNA treatment alleviated cognitive impairment and overexpression of GFAP and Iba-1 in the brain. Furthermore, the treatment ameliorated the VaD-induced damage to the blood-brain barrier (BBB) components, including PECAM-1, PDGFRβ, occludin, claudin-9, and ZO-1. CCR6 activation after VaD, associated with BBB disruption, was diminished by treatment with Lin28a siRNA. The treatment inhibited VaD-induced microglial activity and alleviated BBB damage. Thus, blocking Lin28a may alleviate cognitive impairment caused by VaD.
Collapse
|
18
|
Sharifi MD, Karimi N, Karami M, Borhani Haghighi A, Shabani M, Bayat M. The Minocycline Ameliorated the Synaptic Plasticity Impairment in Vascular Dementia. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2022; 20:435-449. [PMID: 35194458 PMCID: PMC8842628 DOI: 10.22037/ijpr.2020.113942.14576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) leads to vascular dementia with progressive hippocampal damage and cognitive impairments. In the present study, we compared early and late Minocycline (MINO) treatment on cognitive function, long and short-term synaptic-plasticity following CCH. We used bilateral common carotid arteries occlusion model (2VO) for induction of hypoperfusion. Male Sprague-Dawley rats were divided into 5 following groups (each having 2 subgroups): 2VO + V (vehicle), 2VO+MINO-E (early treatment of MINO on days 0 to 3 after 2VO), 2VO+MINO-L (late-treatment on days 21 to 32 after 2VO), control, and sham. Passive-avoidance (PA) and radial arm maze (RAM) tests were used to investigate learning and memory. Long term and short term synaptic plasticity were assessed by field potential recording, the brains were removed after recording and preserved for histological study to count pyramidal cells in CA1 region.Cerebral hypoperfusion could impair memory performance, synaptic plasticity, and basal synaptic transmission (BST) along with hippocampal cell loss. Thus, we found a significant reduction in step-through latency (STL) of PA test with a higher number of working and reference errors in RAM in CCH rats. However, only late treatment with MINO improved memory performance, synaptic plasticity, hippocampal cell loss, and increased neurotransmitter pool (NP) in CCH rats, but early treatment could not produce long-lasting beneficial effects 32 days after 2VO. MINO may improve synaptic plasticity and memory performance in hypo-perfused rats directly and indirectly by increasing NP and/or suppressing inflammatory factors, respectively.
Collapse
Affiliation(s)
- Mohammad Davood Sharifi
- Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Narges Karimi
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Karami
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Shabani
- Neuroscience Research Center, Neuropharmacology Institute, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
19
|
Hypoxia/Ischemia-Induced Rod Microglia Phenotype in CA1 Hippocampal Slices. Int J Mol Sci 2022; 23:ijms23031422. [PMID: 35163344 PMCID: PMC8836225 DOI: 10.3390/ijms23031422] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
The complexity of microglia phenotypes and their related functions compels the continuous study of microglia in diseases animal models. We demonstrated that oxygen-glucose deprivation (OGD) induced rapid, time- and space-dependent phenotypic microglia modifications in CA1 stratum pyramidalis (SP) and stratum radiatum (SR) of rat organotypic hippocampal slices as well as the degeneration of pyramidal neurons, especially in the outer layer of SP. Twenty-four h following OGD, many rod microglia formed trains of elongated cells spanning from the SR throughout the CA1, reaching the SP outer layer where they acquired a round-shaped amoeboid phagocytic head and phagocytosed most of the pyknotic, damaged neurons. NIR-laser treatment, known to preserve neuronal viability after OGD, prevented rod microglia formation. In CA3 SP, pyramidal neurons were less damaged, no rod microglia were found. Thirty-six h after OGD, neuronal damage was more pronounced in SP outer and inner layers of CA1, rod microglia cells were no longer detectable, and most microglia were amoeboid/phagocytic. Damaged neurons, more numerous 36 h after OGD, were phagocytosed by amoeboid microglia in both inner and outer layers of CA1. In response to OGD, microglia can acquire different morphofunctional phenotypes which depend on the time after the insult and on the subregion where microglia are located.
Collapse
|
20
|
Ameliorative effects of oyster (Crassostrea hongkongensis) protein hydrolysate on age-induced cognitive impairment via restoring glia cell dysfunction and neuronal injured in zebrafish. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
21
|
Santos VR, Melo IS, Pacheco ALD, Castro OWD. Life and death in the hippocampus: What's bad? Epilepsy Behav 2021; 121:106595. [PMID: 31759972 DOI: 10.1016/j.yebeh.2019.106595] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/13/2023]
Abstract
The hippocampal formation is crucial for the generation and regulation of several brain functions, including memory and learning processes; however, it is vulnerable to neurological disorders, such as epilepsy. Temporal lobe epilepsy (TLE), the most common type of epilepsy, changes the hippocampal circuitry and excitability, under the contribution of both neuronal degeneration and abnormal neurogenesis. Classically, neurodegeneration affects sensitive areas of the hippocampus, such as dentate gyrus (DG) hilus, as well as specific fields of the Ammon's horn, CA3, and CA1. In addition, the proliferation, migration, and abnormal integration of newly generated hippocampal granular cells (GCs) into the brain characterize TLE neurogenesis. Robust studies over the years have intensely discussed the effects of death and life in the hippocampus, though there are still questions to be answered about their possible benefits and risks. Here, we review the impacts of death and life in the hippocampus, discussing its influence on TLE, providing new perspectives or insights for the implementation of new possible therapeutic targets. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Victor Rodrigues Santos
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.
| | - Igor Santana Melo
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil
| | | | - Olagide Wagner de Castro
- Institute of Biological Sciences and Health, Federal University of Alagoas (UFAL), Maceio, Brazil.
| |
Collapse
|
22
|
Gerace E, Cialdai F, Sereni E, Lana D, Nosi D, Giovannini MG, Monici M, Mannaioni G. NIR Laser Photobiomodulation Induces Neuroprotection in an In Vitro Model of Cerebral Hypoxia/Ischemia. Mol Neurobiol 2021; 58:5383-5395. [PMID: 34319540 PMCID: PMC8497317 DOI: 10.1007/s12035-021-02496-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/12/2021] [Indexed: 12/25/2022]
Abstract
Brain photobiomodulation (PBM) is an innovative treatment for a variety of neurological conditions, including cerebral ischemia. However, the capability of PBM for ischemic stroke needs to be further explored and its mechanisms of action remain currently unclear. The aim of the present research was to identify a treatment protocol capable of inducing neuroprotection and to investigate the molecular mechanisms activated by a dual-wavelength near infrared (NIR) laser source in an organotypic hippocampal slice model of hypoxia/ischemia. Hippocampal slices were exposed to oxygen and glucose deprivation (OGD) for 30 min followed by NIR laser light (fluence 3.71, 7.42, or 14.84 J/cm2; wavelengths 808 nm and 905 nm) delivered immediately or 30 min or 60 min after OGD, in order to establish a therapeutic window. Neuronal injury was assessed by propidium iodide fluorescence 24 h later. Our results show that NIR laser irradiation attenuates OGD neurotoxicity once applied immediately or 30 min after OGD. Western blot analysis of proteins involved in neuroinflammation (iNOS, COX-2, NFkB subunit p65, and Bcl-2) and in glutamatergic-mediated synaptic activity (vGluT1, EAAT2, GluN1, and PSD95) showed that the protein modifications induced by OGD were reverted by NIR laser application. Moreover, CA1 confocal microscopy revealed that the profound morphological changes induced by OGD were reverted by NIR laser radiation. In conclusion, NIR laser radiation attenuates OGD neurotoxicity in organotypic hippocampal slices through attenuation of inflammatory mechanisms. These findings shed light on molecular definition of NIR neuroprotective mechanisms, thus underlining the potential benefit of this technique for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy.
| | - Francesca Cialdai
- ASAcampus Joint Laboratory, ASA Res. Div. - Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elettra Sereni
- ASAcampus Joint Laboratory, ASA Res. Div. - Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Monica Monici
- ASAcampus Joint Laboratory, ASA Res. Div. - Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
23
|
Tarudji AW, Gee CC, Romereim SM, Convertine AJ, Kievit FM. Antioxidant thioether core-crosslinked nanoparticles prevent the bilateral spread of secondary injury to protect spatial learning and memory in a controlled cortical impact mouse model of traumatic brain injury. Biomaterials 2021; 272:120766. [PMID: 33819812 DOI: 10.1016/j.biomaterials.2021.120766] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 03/04/2021] [Accepted: 03/14/2021] [Indexed: 01/19/2023]
Abstract
The secondary phase of traumatic brain injury (TBI) is partly caused by the release of excess reactive oxygen species (ROS) from the primary injury. However, there are currently no therapies that have been shown to reduce the secondary spread of injury beyond the primary insult. Nanoparticles offer the ability to rapidly accumulate and be retained in injured brain for improved target engagement. Here, we utilized systemically administered antioxidant thioether core-cross-linked nanoparticles (NP1) that scavenge and inactivate ROS to reduce this secondary spread of injury in a mild controlled cortical impact (CCI) mouse model of TBI. We found that NP1 treatment protected CCI mice from injury induced learning and memory deficits observed in the Morris water maze (MWM) test at 1-month post-CCI. This protection was likely a result of NP1-mediated reduction in oxidative stress in the ipsilateral hemisphere as determined by immunofluorescence imaging of markers of oxidative stress and the spread of neuroinflammation into the contralateral hippocampus as determined by immunofluorescence imaging of activated microglia and neuron-astrocyte-microglia triad formation. These data suggest NP1-mediated reduction in post-traumatic oxidative stress correlates with the reduction in the spread of injury to the contralateral hippocampus to protect spatial memory and learning in CCI mice. Therefore, these materials may offer an improved treatment strategy to reduce the secondary spread of TBI.
Collapse
Affiliation(s)
- Aria W Tarudji
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Connor C Gee
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Sarah M Romereim
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA
| | - Anthony J Convertine
- Department of Materials Science and Engineering, Missouri University of Science and Technology, 223 McNutt Hall, Rolla, MO, 65409, USA
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 200LW Chase Hall, Lincoln, NE, 68583, USA.
| |
Collapse
|
24
|
Dettori I, Gaviano L, Ugolini F, Lana D, Bulli I, Magni G, Rossi F, Giovannini MG, Pedata F. Protective Effect of Adenosine A 2B Receptor Agonist, BAY60-6583, Against Transient Focal Brain Ischemia in Rat. Front Pharmacol 2021; 11:588757. [PMID: 33643036 PMCID: PMC7905306 DOI: 10.3389/fphar.2020.588757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/21/2020] [Indexed: 01/03/2023] Open
Abstract
Cerebral ischemia is a multifactorial pathology characterized first by an acute injury, due to excitotoxicity, followed by a secondary brain injury that develops hours to days after ischemia. During ischemia, adenosine acts as an endogenous neuroprotectant. Few studies have investigated the role of A2B receptor in brain ischemia because of the low potency of adenosine for it and the few selective ligands developed so far. A2B receptors are scarcely but widely distributed in the brain on neurons, glial and endothelial cells and on hematopoietic cells, lymphocytes and neutrophils, where they exert mainly anti-inflammatory effects, inhibiting vascular adhesion and inflammatory cells migration. Aim of this work was to verify whether chronic administration of the A2B agonist, BAY60-6583 (0.1 mg/kg i.p., twice/day), starting 4 h after focal ischemia induced by transient (1 h) Middle Cerebral Artery occlusion (tMCAo) in the rat, was protective after the ischemic insult. BAY60-6583 improved the neurological deficit up to 7 days after tMCAo. Seven days after ischemia BAY60-6583 reduced significantly the ischemic brain damage in cortex and striatum, counteracted ischemia-induced neuronal death, reduced microglia activation and astrocytes alteration. Moreover, it decreased the expression of TNF-α and increased that of IL-10 in peripheral plasma. Two days after ischemia BAY60-6583 reduced blood cell infiltration in the ischemic cortex. The present study indicates that A2B receptors stimulation can attenuate the neuroinflammation that develops after ischemia, suggesting that A2B receptors may represent a new interesting pharmacological target to protect from degeneration after brain ischemia.
Collapse
Affiliation(s)
- Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Lisa Gaviano
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Irene Bulli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Giada Magni
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Florence, Italy
| | - Francesca Rossi
- Institute of Applied Physics "Nello Carrara", National Research Council (IFAC-CNR), Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Division of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
25
|
Lana D, Ugolini F, Giovannini MG. Space-Dependent Glia-Neuron Interplay in the Hippocampus of Transgenic Models of β-Amyloid Deposition. Int J Mol Sci 2020; 21:E9441. [PMID: 33322419 PMCID: PMC7763751 DOI: 10.3390/ijms21249441] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/14/2022] Open
Abstract
This review is focused on the description and discussion of the alterations of astrocytes and microglia interplay in models of Alzheimer's disease (AD). AD is an age-related neurodegenerative pathology with a slowly progressive and irreversible decline of cognitive functions. One of AD's histopathological hallmarks is the deposition of amyloid beta (Aβ) plaques in the brain. Long regarded as a non-specific, mere consequence of AD pathology, activation of microglia and astrocytes is now considered a key factor in both initiation and progression of the disease, and suppression of astrogliosis exacerbates neuropathology. Reactive astrocytes and microglia overexpress many cytokines, chemokines, and signaling molecules that activate or damage neighboring cells and their mutual interplay can result in virtuous/vicious cycles which differ in different brain regions. Heterogeneity of glia, either between or within a particular brain region, is likely to be relevant in healthy conditions and disease processes. Differential crosstalk between astrocytes and microglia in CA1 and CA3 areas of the hippocampus can be responsible for the differential sensitivity of the two areas to insults. Understanding the spatial differences and roles of glia will allow us to assess how these interactions can influence the state and progression of the disease, and will be critical for identifying therapeutic strategies.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy;
| |
Collapse
|
26
|
Gerace E, Ilari A, Caffino L, Buonvicino D, Lana D, Ugolini F, Resta F, Nosi D, Grazia Giovannini M, Ciccocioppo R, Fumagalli F, Pellegrini-Giampietro DE, Masi A, Mannaioni G. Ethanol neurotoxicity is mediated by changes in expression, surface localization and functional properties of glutamate AMPA receptors. J Neurochem 2020; 157:2106-2118. [PMID: 33107046 DOI: 10.1111/jnc.15223] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 10/13/2020] [Accepted: 10/19/2020] [Indexed: 11/28/2022]
Abstract
Modifications in the subunit composition of AMPA receptors (AMPARs) have been linked to the transition from physiological to pathological conditions in a number of contexts, including EtOH-induced neurotoxicity. Previous work from our laboratory showed that EtOH withdrawal causes CA1 pyramidal cell death in organotypic hippocampal slices and changes in the expression of AMPARs. Here, we investigated whether changes in expression and function of AMPARs may be causal for EtOH-induced neurotoxicity. To this aim, we examined the subunit composition, localization and function of AMPARs in hippocampal slices exposed to EtOH by using western blotting, surface expression assay, confocal microscopy and electrophysiology. We found that EtOH withdrawal specifically increases GluA1 protein signal in total homogenates, but not in the post-synaptic density-enriched fraction. This is suggestive of overall increase and redistribution of AMPARs to the extrasynaptic compartment. At functional level, AMPA-induced calcium influx was unexpectedly reduced, whereas AMPA-induced current was enhanced in CA1 pyramidal neurons following EtOH withdrawal, suggesting that increased AMPAR expression may lead to cell death because of elevated excitability, and not for a direct contribution on calcium influx. Finally, the neurotoxicity caused by EtOH withdrawal was attenuated by the non-selective AMPAR antagonist 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide disodium salt as well as by the selective antagonist of GluA2-lacking AMPARs 1-naphthyl acetyl spermine. We conclude that EtOH neurotoxicity involves changes in expression, surface localization and functional properties of AMPARs, and propose GluA2-lacking AMPARs as amenable specific targets for the development of neuroprotective drugs in EtOH-withdrawal syndrome.
Collapse
Affiliation(s)
- Elisabetta Gerace
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Alice Ilari
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy.,School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | - Daniela Buonvicino
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Francesco Resta
- European Laboratory for Non-Linear Spectroscopy, Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Roberto Ciccocioppo
- School of Pharmacy, Pharmacology Unit, University of Camerino, Camerino, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Italy
| | | | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Guido Mannaioni
- Department of Neuroscience, Psychology, Drug Research and Child Health (NeuroFarBa), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
27
|
Lana D, Ugolini F, Giovannini MG. An Overview on the Differential Interplay Among Neurons-Astrocytes-Microglia in CA1 and CA3 Hippocampus in Hypoxia/Ischemia. Front Cell Neurosci 2020; 14:585833. [PMID: 33262692 PMCID: PMC7686560 DOI: 10.3389/fncel.2020.585833] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Neurons have been long regarded as the basic functional cells of the brain, whereas astrocytes and microglia have been regarded only as elements of support. However, proper intercommunication among neurons-astrocytes-microglia is of fundamental importance for the functional organization of the brain. Perturbation in the regulation of brain energy metabolism not only in neurons but also in astrocytes and microglia may be one of the pathophysiological mechanisms of neurodegeneration, especially in hypoxia/ischemia. Glial activation has long been considered detrimental for survival of neurons, but recently it appears that glial responses to an insult are not equal but vary in different brain areas. In this review, we first take into consideration the modifications of the vascular unit of the glymphatic system and glial metabolism in hypoxic conditions. Using the method of triple-labeling fluorescent immunohistochemistry coupled with confocal microscopy (TIC), we recently studied the interplay among neurons, astrocytes, and microglia in chronic brain hypoperfusion. We evaluated the quantitative and morpho-functional alterations of the neuron-astrocyte-microglia triads comparing the hippocampal CA1 area, more vulnerable to ischemia, to the CA3 area, less vulnerable. In these contiguous and interconnected areas, in the same experimental hypoxic conditions, astrocytes and microglia show differential, finely regulated, region-specific reactivities. In both areas, astrocytes and microglia form triad clusters with apoptotic, degenerating neurons. In the neuron-astrocyte-microglia triads, the cell body of a damaged neuron is infiltrated and bisected by branches of astrocyte that create a microscar around it while a microglial cell phagocytoses the damaged neuron. These coordinated actions are consistent with the scavenging and protective activities of microglia. In hypoxia, the neuron-astrocyte-microglia triads are more numerous in CA3 than in CA1, further indicating their protective effects. These data, taken from contiguous and interconnected hippocampal areas, demonstrate that glial response to the same hypoxic insult is not equal but varies significantly. Understanding the differences of glial reactivity is of great interest to explain the differential susceptibility of hippocampal areas to hypoxia/ischemia. Further studies may evidence the differential reactivity of glia in different brain areas, explaining the higher or lower sensitivity of these areas to different insults and whether glia may represent a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Anatomopathology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
28
|
Radenovic L, Nenadic M, Ułamek-Kozioł M, Januszewski S, Czuczwar SJ, Andjus PR, Pluta R. Heterogeneity in brain distribution of activated microglia and astrocytes in a rat ischemic model of Alzheimer's disease after 2 years of survival. Aging (Albany NY) 2020; 12:12251-12267. [PMID: 32501292 PMCID: PMC7343500 DOI: 10.18632/aging.103411] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/01/2020] [Indexed: 01/07/2023]
Abstract
The present study was designed to follow neuroinflammation after ischemic brain injury in the long-term survival rat model. Immunohistochemistry was performed 2 years after 10 min global brain ischemia due to cardiac arrest. For the visualization of the cellular inflammatory reaction microglial marker Iba1 and astrocyte marker GFAP were used. In post-ischemic animals our study revealed significant activation of astrocytes in all tested brain regions (hippocampal CA1 and CA3 areas and dentate gyrus, motor and somatosensory cortex, striatum and thalamus), while microglial activation was only found in CA1 and CA3 areas, and the motor cortex. In the specifically sensitive brain areas microglia and astrocytes showed simultaneously significant activation, while in the resistant brain areas only astrocytes were activated. Thus, there was clear evidence of less intensive neuroinflammation in brain areas resistant to ischemia. Such neuroinflammatory processes are backed by microglia and astrocytes activity even up to 2 years after ischemia-reperfusion brain injury. Our study thus revealed a chronic effect of global cerebral ischemia on the neuroinflammatory reaction in the rat brain even 2 years after the insult.
Collapse
Affiliation(s)
- Lidija Radenovic
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marija Nenadic
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Marzena Ułamek-Kozioł
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| | | | - Pavle R. Andjus
- Center for Laser Microscopy, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Centre, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
29
|
Ma C, Wang X, Xu T, Zhang S, Liu S, Zhai C, Wang Z, Mu J, Li C, Cheng F, Wang Q. An Integrative Pharmacology-Based Analysis of Refined Qingkailing Injection Against Cerebral Ischemic Stroke: A Novel Combination of Baicalin, Geniposide, Cholic Acid, and Hyodeoxycholic Acid. Front Pharmacol 2020; 11:519. [PMID: 32457601 PMCID: PMC7227481 DOI: 10.3389/fphar.2020.00519] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/02/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is the second leading cause of death after heart disease globally and cerebral ischemic stroke accounts for approximately 70% of all incident stroke cases. We selected four main compounds from a patent Chinese medicine, Qingkailing (QKL) injection, including baicalin from Scutellaria baicalensis Georgi (Huang Qin), geniposide from Gardenia jasminoides J. Ellis (Zhizi), and cholic acid and hyodeoxycholic acid from Bovis Calculus (Niuhuang) with a ratio of 4.4:0.4:3:2.6 m/m, to develop a more efficacious and safer modern Chinese medicine injection against ischemic stroke, refined QKL (RQKL). In this study, we investigated multiple targets, levels, and pathways of RQKL by using an integrative pharm\acology combining experimental validation approach. In silica study showed that RQKL may regulate PI3K-Akt, estrogen, neurotrophin, HIF-1, MAPK, Hippo, FoxO, TGF-beta, NOD-like receptor, apoptosis, NF-kappa B, Wnt, chemokine, TNF, Toll-like receptor signaling pathways against ischemic stroke. The experimental results showed that RQKL improved neurological function and prevented infract volume and blood-brain-barrier damage. RQKL inhibited microgliosis and astrogliosis, and protected neurons from ischemic/reperfusion injury. RQKL also inhibited cell apoptosis and affecting the ratio of the anti-apoptosis protein B-cell lymphoma-2 (Bcl2) and pro-apoptosis protein Bcl2-associated X protein (Bax). Western blot analysis showed that RQKL activated AKT/PI3K signaling pathway and antibody array showed RQKL inhibited inflammatory response and decreased proinflammatory factor Tnf, Il6, and Il1b, and chemokines Ccl2, Cxcl2, and Cxcl3, and increased anti-inflammatory cytokine Il10. In conclusion, RQKL protected tissue against ischemic stroke through multiple-target, multiple signals, and modulating multiple cell-types in brain. This study not only promoted our understanding of the role of RQKL against ischemic stroke, but also provided a pattern for the study of Chinese medicine combining pharmaceutical Informatics and system biology methods.
Collapse
Affiliation(s)
- Chongyang Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xueqian Wang
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Tian Xu
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Shuang Zhang
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Shuling Liu
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Changming Zhai
- Department of Liver Disease, Guangdong Province Hospital of Traditional Chinese Medicine Zhuhai Branch, Zhuhai, China
| | - Zisong Wang
- Department of Traditional Chinese Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jie Mu
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Changxiang Li
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| | - Qingguo Wang
- School of Traditional Chinese Medicine Department, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Heterogeneous Disease Progression in a Mouse Model of Vascular Cognitive Impairment. Int J Mol Sci 2020; 21:ijms21082820. [PMID: 32316637 PMCID: PMC7215687 DOI: 10.3390/ijms21082820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 11/17/2022] Open
Abstract
Recently, an asymmetric vascular compromise approach that replicates many aspects of human vascular cognitive impairment (VCI) has been reported. The present study aimed to first investigate on the reproducibility in the disease progression of this newly reported VCI model using wild-type C57BL6/J mice. The second aim was to assess how this approach will affect the disease progression of transgenic Alzheimer’s disease (AD) 5XFAD mice subjected to VCI. C57BL6/J and 5XFAD mice were subjected to VCI by placing an ameroid constrictor on the right CCA and a microcoil on the left CCA. Infarcts and hippocampal neuronal loss did not appear predominantly in the right (ameroid side) as expected but randomly in both hemispheres. The mortality rate of C57BL6/J mice was unexpectedly high. Inducing VCI reduced amyloid burden in the hippocampi of 5XFAD mice. Since VCI is known to be complex and complicated, the heterogeneous disease progression observed from this current study shares close resemblance to the clinical manifestation of VCI. This heterogeneity, however, makes it challenging to test novel treatment options using this model. Further study is warranted to tackle the heterogeneous nature of VCI.
Collapse
|
31
|
Cavallo D, Landucci E, Gerace E, Lana D, Ugolini F, Henley JM, Giovannini MG, Pellegrini-Giampietro DE. Neuroprotective effects of mGluR5 activation through the PI3K/Akt pathway and the molecular switch of AMPA receptors. Neuropharmacology 2020; 162:107810. [PMID: 31600563 DOI: 10.1016/j.neuropharm.2019.107810] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/23/2019] [Accepted: 10/03/2019] [Indexed: 12/19/2022]
Abstract
Previous studies have demonstrated that antagonists of mGluR1, but not mGluR5, are neuroprotective in models of cerebral ischemia. To investigate the individual roles of mGlu1 and mGlu5 receptors in in vitro model of cerebral ischemia we used low doses of the non-selective group I agonist DHPG and mGlu1 and mGlu5 selective positive allosteric modulators (PAMs). In hippocampal slices subjected to 30 min oxygen-glucose deprivation (OGD), DHPG (1 μM) and the mGluR5 PAM (VU0092273) significantly reduced OGD-induced CA1 injury monitored by propidium iodide staining of the slices and quantitative analysis of CA1 neurons. In contrast, the mGluR1 PAM (VU0483605) showed no neuroprotection. These protective effects of DHPG and VU0092273 were prevented by inhibition of PI3K/Akt pathway by LY294002. The mGluR5 PAM (VU0092273) also prevented GluA2 down-regulation triggered by ischemic injury, via PI3K/Akt pathway, revealing a further contribution to its neuroprotective effects by reducing the excitotoxic effects of increased Ca2+ influx through GluA2-lacking AMPA receptors. Furthermore, immunohistochemical assays confirmed the neuroprotective effect of VU0092273 and revealed activation of glia, indicating the involvement reactive astrogliosis in the mechanisms of neuroprotection. Our data suggest that selective activation/potentiation of mGluR5 signalling represents a promising strategy for the development of new interventions to reduce or prevent ischemia-induced neuronal death.
Collapse
Affiliation(s)
- Damiana Cavallo
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy; School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| | - Elisa Landucci
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Elisabetta Gerace
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Maria Grazia Giovannini
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| | - Domenico E Pellegrini-Giampietro
- Department of Health Sciences, Unit of Clinical Pharmacology and Oncology, University of Florence, Viale G. Pieraccini 6, 50139, Florence, Italy
| |
Collapse
|
32
|
Wen M, Ding L, Zhang L, Zhang T, Teruyoshi Y, Wang Y, Xue C. Eicosapentaenoic Acid-Enriched Phosphatidylcholine Mitigated Aβ1-42-Induced Neurotoxicity via Autophagy-Inflammasome Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:13767-13774. [PMID: 31722531 DOI: 10.1021/acs.jafc.9b05947] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Recent studies indicated that neuroinflammation contributes to the exacerbation of Alzheimer's disease (AD) and plays an important role in AD. The NOD-like receptor protein 3 (NLRP3) inflammasome, which is an important component of innate immune system, is associated with a wide range of human central nervous system disorders, including AD. Most of the studies focus on the protective effects of docosahexaenoic acid (DHA) in AD, but eicosapentaenoic acid (EPA) has rarely been involved. Here, we investigate the effects of EPA in the forms of phosphatidylcholine (EPA-PC) and ethyl esters (EPA-EE) in improving Aβ1-42-induced neurotoxicity. The spatial memory ability and the biochemical changes in the hippocampus were measured, including glial cell activation, tumor necrosis factor α production, NLRP3 inflammasome activation, and autophagic flux. The present results showed that the AD rats were significantly protected from spatial memory loss by the supplementation (EPA + DHA = 60 mg/kg, i.g., 20 days) of EPA-PC, while EPA-EE showed no significant benefit. Further mechanism studies suggested that EPA-PC could inhibit Aβ-induced neurotoxicity by alleviating NLRP3 inflammasome activation and enhancing autophagy. These findings indicate that EPA could improve cognitive deficiency in Aβ1-42-induced AD rats via autophagic inflammasomal pathway and the bioactivity differs in its molecular form.
Collapse
Affiliation(s)
- Min Wen
- Institute of Biopharmaceutical Research , Liaocheng University , Liaocheng 252059 , P. R. China
| | - Lin Ding
- College of Food Science and Engineering , Ocean University of China , Qingdao 266003 , P. R. China
| | - Lingyu Zhang
- College of Food Science and Engineering , Ocean University of China , Qingdao 266003 , P. R. China
| | - Tiantian Zhang
- College of Food Science and Engineering , Ocean University of China , Qingdao 266003 , P. R. China
| | - Yanagita Teruyoshi
- Laboratory of Nutrition Biochemistry, Department of Applied Biochemistry and Food Science , Saga University , Saga 840-8502 , Japan
| | - Yuming Wang
- College of Food Science and Engineering , Ocean University of China , Qingdao 266003 , P. R. China
- Laboratory for Marine Drugs and Bioproducts , Pilot National Laboratory for Marine Science and Technology (Qingdao) , Qingdao 266237 , P. R. China
| | - Changhu Xue
- College of Food Science and Engineering , Ocean University of China , Qingdao 266003 , P. R. China
- Laboratory for Marine Drugs and Bioproducts , Pilot National Laboratory for Marine Science and Technology (Qingdao) , Qingdao 266237 , P. R. China
| |
Collapse
|
33
|
Mustapha M, Nassir CMNCM, Aminuddin N, Safri AA, Ghazali MM. Cerebral Small Vessel Disease (CSVD) - Lessons From the Animal Models. Front Physiol 2019; 10:1317. [PMID: 31708793 PMCID: PMC6822570 DOI: 10.3389/fphys.2019.01317] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cerebral small vessel disease (CSVD) refers to a spectrum of clinical and imaging findings resulting from pathological processes of various etiologies affecting cerebral arterioles, perforating arteries, capillaries, and venules. Unlike large vessels, it is a challenge to visualize small vessels in vivo, hence the difficulty to directly monitor the natural progression of the disease. CSVD might progress for many years during the early stage of the disease as it remains asymptomatic. Prevalent among elderly individuals, CSVD has been alarmingly reported as an important precursor of full-blown stroke and vascular dementia. Growing evidence has also shown a significant association between CSVD's radiological manifestation with dementia and Alzheimer's disease (AD) pathology. Although it remains contentious as to whether CSVD is a cause or sequelae of AD, it is not far-fetched to posit that effective therapeutic measures of CSVD would mitigate the overall burden of dementia. Nevertheless, the unifying theory on the pathomechanism of the disease remains elusive, hence the lack of effective therapeutic approaches. Thus, this chapter consolidates the contemporary insights from numerous experimental animal models of CSVD, to date: from the available experimental animal models of CSVD and its translational research value; the pathomechanical aspects of the disease; relevant aspects on systems biology; opportunities for early disease biomarkers; and finally, converging approaches for future therapeutic directions of CSVD.
Collapse
Affiliation(s)
- Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Niferiti Aminuddin
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Department of Basic Medical Sciences, Kulliyyah of Pharmacy, International Islamic University Malaysia, Kuantan, Malaysia
| | - Amanina Ahmad Safri
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Mazira Mohamad Ghazali
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
34
|
Central Noradrenergic Agonists in the Treatment of Ischemic Stroke-an Overview. Transl Stroke Res 2019; 11:165-184. [PMID: 31327133 DOI: 10.1007/s12975-019-00718-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 02/03/2023]
Abstract
Ischemic stroke is the leading cause of morbidity and mortality with a significant health burden worldwide and few treatment options. Among the short- and long-term effects of ischemic stroke is the cardiovascular sympathetic autonomic dysfunction, presented in part as the by-product of the ischemic damage to the noradrenergic centers of the brain. Unlike high levels in the plasma, the brain may face suboptimal levels of norepinephrine (NE), with adverse effects on the clinical and functional outcomes of ischemic stroke. The intravenous administration of NE and other sympathomimetic agents, in an attempt to increase cerebral perfusion pressure, often aggravates the ischemia-induced rise in blood pressure (BP) with life-threatening consequences for stroke patients, the majority of whom present with hypertension at the time of admission. Unlike the systemic administration, the central administration of NE reduces BP while exerting anti-inflammatory and neuroprotective effects. These characteristics of centrally administered NE, combined with the short latency of response, make it an ideal candidate for use in the acute phase of stroke, followed by the use of centrally acting noradrenergic agonists, such as NE reuptake inhibitors and B2-adrenergic receptor agonists for stroke rehabilitation. In addition, a number of nonpharmacological strategies, such as transcutaneous vagus nerve stimulation (tVNS) and trigeminal nerve stimulation (TNS), have the potential to enhance the central noradrenergic functional activities and improve stroke clinical outcomes. Many factors could influence the efficacy of the noradrenergic treatment in stroke patients. These factors include the type of the noradrenergic agent; the dose, frequency, and duration of administration; the timing of administration in relation to the acute event; and the site and characteristics of the ischemic lesions. Having this knowledge, combined with the better understanding of the regulation of noradrenergic receptors in different parts of the brain, would pave the path for the successful use of the centrally acting noradrenergic agents in the management of ischemic stroke.
Collapse
|
35
|
Lana D, Ugolini F, Wenk GL, Giovannini MG, Zecchi-Orlandini S, Nosi D. Microglial distribution, branching, and clearance activity in aged rat hippocampus are affected by astrocyte meshwork integrity: evidence of a novel cell-cell interglial interaction. FASEB J 2018; 33:4007-4020. [PMID: 30496700 DOI: 10.1096/fj.201801539r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aging and neurodegenerative diseases share a condition of neuroinflammation entailing the production of endogenous cell debris in the CNS that must be removed by microglia ( i.e., resident macrophages), to restore tissue homeostasis. In this context, extension of microglial cell branches toward cell debris underlies the mechanisms of microglial migration and phagocytosis. Amoeboid morphology and the consequent loss of microglial branch functionality characterizes dysregulated microglia. Microglial migration is assisted by another glial population, the astroglia, which forms a dense meshwork of cytoplasmic projections. Amoeboid microglia and disrupted astrocyte meshwork are consistent traits in aged CNS. In this study, we assessed a possible correlation between microglia and astroglia morphology in rat models of chronic neuroinflammation and aging, by 3-dimensional confocal analysis implemented with particle analysis. Our findings suggest that a microglia-astroglia interaction occurs in rat hippocampus via cell-cell contacts, mediating microglial cell branching in the presence of inflammation. In aged rats, the impairment of such an interaction correlates with altered distribution, morphology, and inefficient clearance by microglia. These data support the idea that generally accepted functional boundaries between microglia and astrocytes should be re-evaluated to better understand how their functions overlap and interact.-Lana, D., Ugolini, F., Wenk, G. L., Giovannini, M. G., Zecchi-Orlandini, S., Nosi, D. Microglial distribution, branching, and clearance activity in aged rat hippocampus are affected by astrocyte meshwork integrity: evidence of a novel cell-cell interglial interaction.
Collapse
Affiliation(s)
- Daniele Lana
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, Columbus, Ohio, USA
| | - Maria Grazia Giovannini
- Section of Clinical Pharmacology and Oncology, Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
36
|
Ugolini F, Lana D, Nardiello P, Nosi D, Pantano D, Casamenti F, Giovannini MG. Different Patterns of Neurodegeneration and Glia Activation in CA1 and CA3 Hippocampal Regions of TgCRND8 Mice. Front Aging Neurosci 2018; 10:372. [PMID: 30483118 PMCID: PMC6243135 DOI: 10.3389/fnagi.2018.00372] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 10/26/2018] [Indexed: 01/24/2023] Open
Abstract
We investigated the different patterns of neurodegeneration and glia activation in CA1 and CA3 hippocampal areas of TgCRND8 mice. The main feature of this transgenic model is the rapid development of the amyloid pathology, which starts already at 3 months of age. We performed immunohistochemical analyses to compare the different sensibility of the two hippocampal regions to neurodegeneration. We performed qualitative and quantitative evaluations by fluorescence immunohistochemistry with double or triple staining, followed by confocal microscopy and digital image analysis in stratum pyramidale (SP) and stratum radiatum (SR) of CA1 and CA3, separately. We evaluated time-dependent Aβ plaques deposition, expression of inflammatory markers, as well as quantitative and morphological alterations of neurons and glia in transgenic mice at 3 (Tg 3M) and 6 (Tg 6M) months of age, compared to WT mice. In CA1 SR of Tg 6M mice, we found significantly more Medium and Large plaques than in CA3. The pattern of neurodegeneration and astrocytes activation was different in the two areas, indicating higher sensitivity of CA1. In the CA1 SP of Tg 6M mice, we found signs of reactive astrogliosis, such as increase of astrocytes density in SP, increase of GFAP expression in SR, and elongation of astrocytes branches. We found also common patterns of glia activation and neurodegenerative processes in CA1 and CA3 of Tg 6M mice: significant increase of total and reactive microglia density in SP and SR, increased expression of TNFα, of iNOS, and IL1β in astrocytes and increased density of neurons-astrocytes-microglia triads. In CA1 SP, we found decrease of volume and number of pyramidal neurons, paralleled by increase of apoptosis, and, consequently, shrinkage of CA1 SP. These data demonstrate that in TgCRND8 mice, the responses of neurons and glia to neurodegenerative patterns induced by Aβ plaques deposition is not uniform in the two hippocampal areas, and in CA1 pyramidal neurons, the higher sensitivity may be related to the different plaque distribution in this area. All these modifications may be at the basis of memory loss, the peculiar symptom of AD, which was demonstrated in this transgenic mouse model of Aβ deposition, even at early stages.
Collapse
Affiliation(s)
- Filippo Ugolini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Pamela Nardiello
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Daniela Pantano
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Fiorella Casamenti
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| |
Collapse
|
37
|
Fusco I, Ugolini F, Lana D, Coppi E, Dettori I, Gaviano L, Nosi D, Cherchi F, Pedata F, Giovannini MG, Pugliese AM. The Selective Antagonism of Adenosine A 2B Receptors Reduces the Synaptic Failure and Neuronal Death Induced by Oxygen and Glucose Deprivation in Rat CA1 Hippocampus in Vitro. Front Pharmacol 2018; 9:399. [PMID: 29740323 PMCID: PMC5928446 DOI: 10.3389/fphar.2018.00399] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 04/06/2018] [Indexed: 01/02/2023] Open
Abstract
Ischemia is a multifactorial pathology characterized by different events evolving in time. Immediately after the ischemic insult, primary brain damage is due to the massive increase of extracellular glutamate. Adenosine in the brain increases dramatically during ischemia in concentrations able to stimulate all its receptors, A1, A2A, A2B, and A3. Although adenosine exerts clear neuroprotective effects through A1 receptors during ischemia, the use of selective A1 receptor agonists is hampered by their undesirable peripheral side effects. So far, no evidence is available on the involvement of adenosine A2B receptors in cerebral ischemia. This study explored the role of adenosine A2B receptors on synaptic and cellular responses during oxygen and glucose deprivation (OGD) in the CA1 region of rat hippocampus in vitro. We conducted extracellular recordings of CA1 field excitatory post-synaptic potentials (fEPSPs); the extent of damage on neurons and glia was assessed by immunohistochemistry. Seven min OGD induced anoxic depolarization (AD) in all hippocampal slices tested and completely abolished fEPSPs that did not recover after return to normoxic condition. Seven minutes OGD was applied in the presence of the selective adenosine A2B receptor antagonists MRS1754 (500 nM) or PSB603 (50 nM), separately administered 15 min before, during and 5 min after OGD. Both antagonists were able to prevent or delay the appearance of AD and to modify synaptic responses after OGD, allowing significant recovery of neurotransmission. Adenosine A2B receptor antagonism also counteracted the reduction of neuronal density in CA1 stratum pyramidale, decreased apoptosis at least up to 3 h after the end of OGD, and maintained activated mTOR levels similar to those of controls, thus sparing neurons from the degenerative effects caused by the simil-ischemic conditions. Astrocytes significantly proliferated in CA1 stratum radiatum already 3 h after the end of OGD, possibly due to increased glutamate release. A2Breceptor antagonism significantly prevented astrocyte modifications. Both A2B receptor antagonists did not protect CA1 neurons from the neurodegeneration induced by glutamate application, indicating that the antagonistic effect is upstream of glutamate release. The selective antagonists of the adenosine A2B receptor subtype may thus represent a new class of neuroprotective drugs in ischemia.
Collapse
Affiliation(s)
- Irene Fusco
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Elisabetta Coppi
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Ilaria Dettori
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Lisa Gaviano
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Federica Cherchi
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Felicita Pedata
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Maria G Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | - Anna M Pugliese
- Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA, Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| |
Collapse
|
38
|
Li X, Liu RZ, Zeng Q, Huang ZH, Zhang JD, Liu ZL, Zeng J, Xiao H. 3'-Daidzein sulfonate sodium protects against memory impairment and hippocampal damage caused by chronic cerebral hypoperfusion. Neural Regen Res 2018; 13:1561-1567. [PMID: 30127116 PMCID: PMC6126135 DOI: 10.4103/1673-5374.237119] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
3′-Daidzein sulfonate sodium (DSS) is a new synthetic water-soluble compound derived from daidzein, a soya isoflavone that plays regulatory roles in neurobiology. In this study, we hypothesized that the regulatory role of DSS in neurobiology exhibits therapeutic effects on hippocampal damage and memory impairment. To validate this hypothesis, we established rat models of chronic cerebral hypoperfusion (CCH) by the permanent occlusion of the common carotid arteries using the two-vessel occlusion method. Three weeks after modeling, rat models were intragastrically administered 0.1, 0.2, and 0.4 mg/kg DSS, once a day, for 5 successive weeks. The Morris water maze test was performed to investigate CCH-induced learning and memory deficits. TUNEL assay was used to analyze apoptosis in the hippocampal CA1, CA3 regions and dentate gyrus. Hematoxylin-eosin staining was performed to observe the morphology of neurons in the hippocampal CA1, CA3 regions and dentate gyrus. Western blot analysis was performed to investigate the phosphorylation of PKA, ERK1/2 and CREB in the hippocampal PKA/ERK1/2/CREB signaling pathway. Results showed that DSS treatment greatly improved the learning and memory deficits of rats with CCH, reduced apoptosis of neurons in the hippocampal CA1, CA3 regions and dentate gyrus, and increased the phosphorylation of PKA, ERK1/2, and CREB in the hippocampus. These findings suggest that DSS protects against CCH-induced memory impairment and hippocampal damage possibly through activating the PKA/ERK1/2/CREB signaling pathway.
Collapse
Affiliation(s)
- Xiao Li
- Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Rui-Zhen Liu
- Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Qi Zeng
- Department of Ultrasound, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Zhi-Hua Huang
- Gannan Medical University, Ganzhou, Jiangxi Province, China
| | | | - Zong-Liang Liu
- Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Jing Zeng
- Gannan Medical University, Ganzhou, Jiangxi Province, China
| | - Hai Xiao
- Department of Pathology, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi Province, China
| |
Collapse
|
39
|
Bordeleau M, ElAli A, Rivest S. Severe chronic cerebral hypoperfusion induces microglial dysfunction leading to memory loss in APPswe/PS1 mice. Oncotarget 2017; 7:11864-80. [PMID: 26918610 PMCID: PMC4914254 DOI: 10.18632/oncotarget.7689] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022] Open
Abstract
Cerebral vasculature plays a key role in controlling brain homeostasis. Cerebral vasculature dysfunction, associated to irregularities in cerebral blood perfusion, has been proposed to directly contribute to Alzheimer's disease (AD) pathogenesis. More precisely, chronic cerebral hypoperfusion, which impairs brain homeostasis, was demonstrated to take place even before cognitive decline. However, the mechanisms underlying the implication of chronic cerebral hypoperfusion in AD pathogenesis remain elusive. Therefore, this study aims at investigating the role of severe chronic cerebral hypoperfusion (SCCH) in AD pathogenesis. For this purpose, SCCH was induced in young APPswe/PS1 in order to evaluate the progression of AD-like pathology in these mice. We observed that SCCH accelerated the cognitive decline of young APPswe/PS1 mice, which was associated with an increased amyloid plaque number in brain parenchyma. In addition, SCCH reduced the activity of extracellular signal-regulated kinases 1/2 (ERK1/2), which has been shown to play an important role in the adaptive responses of neurons. Importantly, SCCH impaired the function of microglial cells, which are implicated in amyloid-β (Aβ) elimination. In vitro approaches underlined the ability of a low-glucose microenvironment to decrease the general activity and phagocytic capacity of microglia. By using a new model of SCCH, our study unravels new insights into the implication of severe chronic cerebral hypoperfusion in AD pathogenesis, mainly by altering microglial cell activity and consequently Aβ clearance.
Collapse
Affiliation(s)
- Maude Bordeleau
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada
| | - Ayman ElAli
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Psychiatry and Neuroscience, Faculty of Medicine, Laval University, Québec, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Québec Research Center (CHUL), Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, Canada
| |
Collapse
|
40
|
Lana D, Ugolini F, Nosi D, Wenk GL, Giovannini MG. Alterations in the Interplay between Neurons, Astrocytes and Microglia in the Rat Dentate Gyrus in Experimental Models of Neurodegeneration. Front Aging Neurosci 2017; 9:296. [PMID: 28955220 PMCID: PMC5601988 DOI: 10.3389/fnagi.2017.00296] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
The hippocampus is negatively affected by aging and neurodegenerative diseases leading to impaired learning and memory abilities. A diverse series of progressive modifications in the intercellular communication among neurons, astrocytes and microglia occur in the hippocampus during aging or inflammation. A detailed understanding of the neurobiological modifications that contribute to hippocampal dysfunction may reveal new targets for therapeutic intervention. The current study focussed on the interplay between neurons and astroglia in the Granule Layer (GL) and the Polymorphic Layer (PL) of the Dentate Gyrus (DG) of adult, aged and LPS-treated rats. In GL and PL of aged and LPS-treated rats, astrocytes were less numerous than in adult rats. In GL of LPS-treated rats, astrocytes acquired morphological features of reactive astrocytes, such as longer branches than was observed in adult rats. Total and activated microglia increased in the aged and LPS-treated rats, as compared to adult rats. In the GL of aged and LPS-treated rats many neurons were apoptotic. Neurons decreased significantly in GL and PL of aged but not in rats treated with LPS. In PL of aged and LPS-treated rats many damaged neurons were embraced by microglia cells and were infiltrated by branches of astrocyte, which appeared to be bisecting the cell body, forming triads. Reactive microglia had a scavenging activity of dying neurons, as shown by the presence of neuronal debris within their cytoplasm. The levels of the chemokine fractalkine (CX3CL1) increased in hippocampal homogenates of aged rats and rats treated with LPS, and CX3CL1 immunoreactivity colocalized with activated microglia cells. Here we demonstrated that in the DG of aged and LPS-treated rats, astrocytes and microglia cooperate and participate in phagocytosis/phagoptosis of apoptotic granular neurons. The differential expression/activation of astroglia and the alteration of their intercommunication may be responsible for the different susceptibility of the DG in comparison to the CA1 and CA3 hippocampal areas to neurodegeneration during aging and inflammation.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| | - Filippo Ugolini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of FlorenceFlorence, Italy
| | - Gary L Wenk
- Department of Psychology, The Ohio State UniversityColumbus, OH, United States
| | - Maria G Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of FlorenceFlorence, Italy
| |
Collapse
|
41
|
Moghaddasi M, Taati M, Asadian P, Khalatbary AR, Asaei R, Pajouhi N. The effects of two-stage carotid occlusion on spatial memory and pro-inflammatory markers in the hippocampus of rats. J Physiol Sci 2017; 67:415-423. [PMID: 27470129 PMCID: PMC10717598 DOI: 10.1007/s12576-016-0474-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Accepted: 07/13/2016] [Indexed: 02/07/2023]
Abstract
The purpose of this study was to evaluate the effects of cerebral hypoperfusion on cognitive ability, TNFα, IL1β and PGE2 levels in both hippocampi in a modified two-vessel occlusion model. Both common carotid arteries of adult male Wistar rats were permanently occluded with an interval of 1 week between occlusions. Learning and memory were significantly decreased after 1 month. This reduction was not significant after 2 months, which may be attributed to blood flow compensation. The TNFα level was significantly increased after 3 h and 1 day. IL1β was significantly increased after 1 day. After a week there was no significant difference in pro-inflammatory levels. Furthermore, there was no difference between right and left hippocampi. It is possible that TNFα and IL1β elevation initiates pathologic processes that contribute to memory impairment.
Collapse
Affiliation(s)
- Mehrnoush Moghaddasi
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| | - Majid Taati
- Department of Pathobiology, Faculty of Veterinary Medicine, Lorestan University, Khorramabad, Iran
| | - Payman Asadian
- Ontario Veterinary College, University of Guelph, Guelph, Canada
| | - Ali Reza Khalatbary
- Department of Anatomy, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Raheleh Asaei
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Naser Pajouhi
- Department of Physiology, Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
42
|
Matyash M, Zabiegalov O, Wendt S, Matyash V, Kettenmann H. The adenosine generating enzymes CD39/CD73 control microglial processes ramification in the mouse brain. PLoS One 2017; 12:e0175012. [PMID: 28376099 PMCID: PMC5380357 DOI: 10.1371/journal.pone.0175012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 03/20/2017] [Indexed: 02/03/2023] Open
Abstract
Microglial cells invade the brain as amoeboid precursors and acquire a highly ramified morphology in the postnatal brain. Microglia express all essential purinergic elements such as receptors, nucleoside transporters and ecto-enzymes, including CD39 (NTPDase1) and CD73 (5'-nucleotidase), which sequentially degrade extracellular ATP to adenosine. Here, we show that constitutive deletion of CD39 and CD73 or both caused an inhibition of the microglia ramified phenotype in the brain with a reduction in the length of processes, branching frequency and number of intersections with Sholl spheres. In vitro, unlike wild-type microglia, cd39-/- and cd73-/- microglial cells were less complex and did not respond to ATP with the transformation into a more ramified phenotype. In acute brain slices, wild-type microglia retracted approximately 50% of their processes within 15 min after slicing of the brain, and this phenomenon was augmented in cd39-/- mice; moreover, the elongation of microglial processes towards the source of ATP or towards a laser lesion was observed only in wild-type but not in cd39-/- microglia. An elevation of extracellular adenosine 1) by the inhibition of adenosine transport with dipyridamole, 2) by application of exogenous adenosine or 3) by degradation of endogenous ATP/ADP with apyrase enhanced spontaneous and ATP-induced ramification of cd39-/- microglia in acute brain slices and facilitated the transformation of cd39-/- and cd73-/- microglia into a ramified process-bearing phenotype in vitro. These data indicate that under normal physiological conditions, CD39 and CD73 nucleotidases together with equilibrative nucleoside transporter 1 (ENT1) control the fate of extracellular adenosine and thereby the ramification of microglial processes.
Collapse
Affiliation(s)
- Marina Matyash
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oleksandr Zabiegalov
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Stefan Wendt
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Vitali Matyash
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Helmut Kettenmann
- Cellular Neurosciences, Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- * E-mail:
| |
Collapse
|
43
|
Zhao SC, Ma LS, Chu ZH, Xu H, Wu WQ, Liu F. Regulation of microglial activation in stroke. Acta Pharmacol Sin 2017; 38:445-458. [PMID: 28260801 DOI: 10.1038/aps.2016.162] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 11/06/2016] [Indexed: 12/16/2022]
Abstract
When ischemic stroke occurs, oxygen and energy depletion triggers a cascade of events, including inflammatory responses, glutamate excitotoxicity, oxidative stress, and apoptosis that result in a profound brain injury. The inflammatory response contributes to secondary neuronal damage, which exerts a substantial impact on both acute ischemic injury and the chronic recovery of the brain function. Microglia are the resident immune cells in the brain that constantly monitor brain microenvironment under normal conditions. Once ischemia occurs, microglia are activated to produce both detrimental and neuroprotective mediators, and the balance of the two counteracting mediators determines the fate of injured neurons. The activation of microglia is defined as either classic (M1) or alternative (M2): M1 microglia secrete pro-inflammatory cytokines (TNFα, IL-23, IL-1β, IL-12, etc) and exacerbate neuronal injury, whereas the M2 phenotype promotes anti-inflammatory responses that are reparative. It has important translational value to regulate M1/M2 microglial activation to minimize the detrimental effects and/or maximize the protective role. Here, we discuss various regulators of microglia/macrophage activation and the interaction between microglia and neurons in the context of ischemic stroke.
Collapse
|
44
|
Protective Effect of 17β-Estradiol Upon Hippocampal Spine Density and Cognitive Function in an Animal Model of Vascular Dementia. Sci Rep 2017; 7:42660. [PMID: 28205591 PMCID: PMC5311994 DOI: 10.1038/srep42660] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 01/12/2017] [Indexed: 12/14/2022] Open
Abstract
The current study examined whether the steroid hormone, 17β-estradiol (E2) can exert long-lasting beneficial effects upon axonal health, synaptic plasticity, dementia-related amyloid-beta (Aβ) protein expression, and hippocampal-dependent cognitive function in an animal model of chronic cerebral hypoperfusion and vascular dementia (VaD). Chronic cerebral hypoperfusion and VaD was induced by bilateral common carotid artery occlusion (BCCAO) in adult male Sprague Dawley rats. Low dose E2 administered for the first 3-months after BCCAO exerted long-lasting beneficial effects, including significant neuroprotection of hippocampal CA1 neurons and preservation of hippocampal-dependent cognitive function when examined at 6-months after BCCAO. E2 treatment also prevented BCCAO-induced damage to hippocampal myelin sheaths and oligodendrocytes, enhanced expression of the synaptic proteins synaptophysin and PSD95 in the hippocampus, and prevented BCCAO-induced loss of total and mushroom dendritic spines in the hippocampal CA1 region. Furthermore, E2-treatment also reduced BCCAO induction of dementia-related proteins expression such as p-tau (PHF1), total ubiquitin, and Aβ1-42, when examined at 6 m after BCCAO. Taken as a whole, the results suggest that low-dose E2 replacement might be a potentially promising therapeutic modality to attenuate or block negative neurological consequences of chronic cerebral hypoperfusion and VaD.
Collapse
|
45
|
Acetyl-L-Carnitine via Upegulating Dopamine D1 Receptor and Attenuating Microglial Activation Prevents Neuronal Loss and Improves Memory Functions in Parkinsonian Rats. Mol Neurobiol 2016; 55:583-602. [PMID: 27975173 DOI: 10.1007/s12035-016-0293-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023]
Abstract
Parkinson's disease is accompanied by nonmotor symptoms including cognitive impairment, which precede the onset of motor symptoms in patients and are regulated by dopamine (DA) receptors and the mesocorticolimbic pathway. The relative contribution of DA receptors and astrocytic glutamate transporter (GLT-1) in cognitive functions is largely unexplored. Similarly, whether microglia-derived increased immune response affects cognitive functions and neuronal survival is not yet understood. We have investigated the effect of acetyl-L-carnitine (ALCAR) on cognitive functions and its possible underlying mechanism of action in 6-hydroxydopamine (6-OHDA)-induced hemiparkinsonian rats. ALCAR treatment in 6-OHDA-lesioned rats improved memory functions as confirmed by decreased latency time and path length in the Morris water maze test. ALCAR further enhanced D1 receptor levels without altering D2 receptor levels in the hippocampus and prefrontal cortex (PFC) regions, suggesting that the D1 receptor is preferentially involved in the regulation of cognitive functions. ALCAR attenuated microglial activation and release of inflammatory mediators through balancing proinflammatory and anti-inflammatory cytokines, which subsequently enhanced the survival of mature neurons in the CA1, CA3, and PFC regions and improved cognitive functions in hemiparkinsonian rats. ALCAR treatment also improved glutathione (GSH) content, while decreasing oxidative stress indices, inducible nitrogen oxide synthase (iNOS) levels, and astrogliosis resulting in the upregulation of GLT-1 levels. Additionally, ALCAR prevented the loss of dopaminergic (DAergic) neurons in ventral tagmental area (VTA)/substantia nigra pars compacta (SNpc) regions of 6-OHDA-lesioned rats, thus maintaining the integrity of the nigrostriatal pathway. Together, these results demonstrate that ALCAR treatment in hemiparkinsonian rats ameliorates neurodegeneration and cognitive deficits, hence suggesting its therapeutic potential in neurodegenerative diseases.
Collapse
|
46
|
Kim MS, Bang JH, Lee J, Han JS, Baik TG, Jeon WK. Ginkgo biloba L. extract protects against chronic cerebral hypoperfusion by modulating neuroinflammation and the cholinergic system. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1356-1364. [PMID: 27765355 DOI: 10.1016/j.phymed.2016.07.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/10/2016] [Accepted: 07/31/2016] [Indexed: 05/27/2023]
Abstract
BACKGROUND Ginkgo biloba extract (GBE)-a widely used nutraceutical-is reported to have diverse functions, including positive effects on memory and vasodilatory properties. Although numerous studies have assessed the neuroprotective properties of GBE in ischemia, only a few studies have investigated the neuro-pharmacological mechanisms of action of GBE in chronic cerebral hypoperfusion (CCH). PURPOSE In the present study, we sought to determine the effects of GBE on CCH-induced neuroinflammation and cholinergic dysfunction in a rat model of bilateral common carotid artery occlusion (BCCAo). METHODS Chronic BCCAo was induced in adult male Wistar rats to reflect the CCH conditions. On day 21 after BCCAo, the animals were treated orally with saline or GBE (5, 10, 20, and 40mg/kg) daily for 42 days. After the final treatment, brain tissues were isolated for the immunohistochemical analysis of glial markers and choline acetyltransferase (ChAT), as well as for the western blot analysis of proinflammatory cytokines, toll-like receptor (TLR)-related pathway, receptor for advanced glycation end products (RAGE), angiotensin-II (Ang-II), and phosphorylated mitogen-activated protein kinases (MAPKs). RESULTS BCCAo increased glial proliferation in the hippocampus and white matter, whereas proliferation was significantly attenuated by GBE treatment. GBE also attenuated the BCCAo-related increases in the hippocampal expression of proinflammatory cytokines (TNF-α, IL-1β, and IL-6), TLR4, myeloid differentiation primary response gene 88, RAGE, Ang-II, and phosphorylated MAPKs (ERK, p38, and JNK). Furthermore, GBE treatment restored the ChAT expression in the basal forebrain following BCCAo. CONCLUSIONS These findings suggest that GBE has specific neuroprotective effects that may be useful for the treatment of CCH. The pharmacological mechanism of GBE partly involves the modulation of inflammatory mediators and the cholinergic system.
Collapse
Affiliation(s)
- Min-Soo Kim
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea
| | - Ji Hye Bang
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea
| | - Jun Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea
| | - Jung-Soo Han
- Department of Biological Science, Konkuk University, Seoul 05029, The Republic of Korea
| | - Tae Gon Baik
- Central Research Center, Yuyu Pharma. Inc., Seoul 04598, The Republic of Korea
| | - Won Kyung Jeon
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, The Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul 02792, The Republic of Korea.
| |
Collapse
|
47
|
Lana D, Iovino L, Nosi D, Wenk GL, Giovannini MG. The neuron-astrocyte-microglia triad involvement in neuroinflammaging mechanisms in the CA3 hippocampus of memory-impaired aged rats. Exp Gerontol 2016; 83:71-88. [PMID: 27466072 DOI: 10.1016/j.exger.2016.07.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/23/2016] [Accepted: 07/20/2016] [Indexed: 01/08/2023]
Abstract
We examined the effects of inflammaging on memory encoding, and qualitative and quantitative modifications on proinflammatory proteins, apoptosis, neurodegeneration and morphological changes of neuron-astrocyte-microglia triads in CA3 Stratum Pyramidale (SP), Stratum Lucidum (SL) and Stratum Radiatum (SR) of young (3months) and aged rats (20months). Aged rats showed short-term memory impairments in the inhibitory avoidance task, increased expression of iNOS and activation of p38MAPK in SP, increase of apoptotic neurons in SP and of ectopic neurons in SL, and decrease of CA3 pyramidal neurons. The number of astrocytes and their branches length decreased in the three CA3 subregions of aged rats, with morphological signs of clasmatodendrosis. Total and activated microglia increased in the three CA3 subregions of aged rats. In aged rats CA3, astrocytes surrounded ectopic degenerating neurons forming "micro scars" around them. Astrocyte branches infiltrated the neuronal cell body, and, together with activated microglia formed "triads". In the triads, significantly more numerous in CA3 SL and SR of aged rats, astrocytes and microglia cooperated in fragmentation and phagocytosis of ectopic neurons. Inflammaging-induced modifications of astrocytes and microglia in CA3 of aged rats may help clearing neuronal debris derived from low-grade inflammation and apoptosis. These events might be common mechanisms underlying many neurodegenerative processes. The frequency to which they appear might depend upon, or might be the cause of, the burden and severity of neurodegeneration. Targeting the triads may represent a therapeutic strategy which may control inflammatory processes and spread of further cellular damage to neighboring cells.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| | - Ludovica Iovino
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, Section of Physiological Sciences, Viale Morgagni 63 and Section of Anatomy and Histology, Largo Brambilla 3, University of Florence, 50134 Firenze, Italy.
| | - Gary L Wenk
- Department of Psychology, The Ohio State University, OH, USA..
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Pharmacology and Clinical Oncology, University of Florence, Viale Pieraccini 6, 50139 Firenze, Italy.
| |
Collapse
|
48
|
Mu S, Liu B, Ouyang L, Zhan M, Chen S, Wu J, Chen J, Wei X, Wang W, Zhang J, Lei W. Characteristic Changes of Astrocyte and Microglia in Rat Striatum Induced by 3-NP and MCAO. Neurochem Res 2015; 41:707-14. [PMID: 26586406 DOI: 10.1007/s11064-015-1739-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/28/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Our previous studies had confirmed that both 3-NP and MCAO induced the behavioral defect as well as striatal neuronal injury and loss in experimental rats. This study aimed to examine different response forms of striatal astrocyte and microglia in 3-NP and MCAO rat models. The present results showed that the immunoreaction for GFAP was extremely weak in the lesioned core of striatum, but in the transition zone of 3-NP model and the penumbra zone of MCAO model, GFAP+ cells showed strong hypertrophic and proliferative changes. Statistical analysis for the number, size and integral optical density (IOD) of GFAP+ cells showed significant differences when compared with their controls and compared between the core and the transition zone or the penumbra zone, respectively, but no differences between the 3-NP and MCAO groups. However, Iba-1+ cells showed obvious hypertrophy and proliferation in the injured striatum in the 3-NP and the MCAO models, especially in the transition zone of 3-NP model and the penumbra zone of MCAO model. These Iba-1+ cells displayed two characteristic forms as branching cells with thick processes and amoeboid cells with thin processes. Statistical analysis showed that the number, size and IOD of Iba-1+ cells were significantly increased in the cores and the transition zone of 3-NP group and the penumbra zone of MCAO group than that of the controls, and the immune response of Iba-1 was stronger in the MCAO group than in the 3-NP group. The present results suggested that characteristic responses of astrocyte and microglia in the 3-NP and the MCAO models display their different effects on the pathological process of brain injury.
Collapse
Affiliation(s)
- Shuhua Mu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China
| | - Bingbing Liu
- Department of Anesthesiology, Guangdong No. 2 Provincial People's Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China
| | - Lisi Ouyang
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Mali Zhan
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Si Chen
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Jiajia Wu
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Jiachang Chen
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Xianyou Wei
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Weiping Wang
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China
| | - Jian Zhang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China. .,School of Medicine, Shenzhen University, Nanhai Ave 3688, Shenzhen, 518060, China.
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, SUN Yat-sen University, 74 Zhongshan Rd 2, Guangzhou, 510080, China.
| |
Collapse
|
49
|
Su SH, Wu YF, Lin Q, Yu F, Hai J. Cannabinoid receptor agonist WIN55,212-2 and fatty acid amide hydrolase inhibitor URB597 suppress chronic cerebral hypoperfusion-induced neuronal apoptosis by inhibiting c-Jun N-terminal kinase signaling. Neuroscience 2015; 301:563-75. [DOI: 10.1016/j.neuroscience.2015.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/13/2015] [Accepted: 03/11/2015] [Indexed: 11/15/2022]
|