601
|
Jesus AA, Passaglia P, Santos BM, Rodrigues-Santos I, Flores RA, Batalhão ME, Stabile AM, Cárnio EC. Chronic molecular hydrogen inhalation mitigates short and long-term memory loss in polymicrobial sepsis. Brain Res 2020; 1739:146857. [PMID: 32348775 DOI: 10.1016/j.brainres.2020.146857] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/04/2020] [Accepted: 04/24/2020] [Indexed: 01/13/2023]
Abstract
The central nervous system (CNS) is one of the first physiological systems to be affected in sepsis. During the exacerbated systemic inflammatory response at the early stage of sepsis, circulatory inflammatory mediators are able to reach the CNS leading to neuroinflammation and, consequently, long-term impairment in learning and memory formation is observed. The acute treatment with molecular hydrogen (H2) exerts important antioxidative, antiapoptotic, and anti-inflammatory effects in sepsis, but little is known about the mechanism itself and the efficacy of chronic H2 inhalation in sepsis treatment. Thus, we tested two hypotheses. We first hypothesized that chronic H2 inhalation is also an effective therapy to treat memory impairment induced by sepsis. The second hypothesis is that H2 treatment decreases sepsis-induced neuroinflammation in the hippocampus and prefrontal cortex, important areas related to short and long-term memory processing. Our results indicate that (1) chronic exposure of hydrogen gas is a simple, safe and promising therapeutic strategy to prevent memory loss in patients with sepsis and (2) acute H2 inhalation decreases neuroinflammation in memory-related areas and increases total nuclear factor E2-related factor 2 (Nrf2), a transcription factorthat regulates a vast group of antioxidant and inflammatory agents expression in these areas of septic animals.
Collapse
Affiliation(s)
- Aline A Jesus
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Patrícia Passaglia
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Bruna M Santos
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Isabelle Rodrigues-Santos
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Rafael A Flores
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil
| | - Marcelo E Batalhão
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900 Brazil
| | - Angelita M Stabile
- Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900 Brazil
| | - Evelin C Cárnio
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900, Brazil; Department of General and Specialized Nursing, School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14049-900 Brazil.
| |
Collapse
|
602
|
Deyts C, Clutter M, Pierce N, Chakrabarty P, Ladd TB, Goddi A, Rosario AM, Cruz P, Vetrivel K, Wagner SL, Thinakaran G, Golde TE, Parent AT. APP-Mediated Signaling Prevents Memory Decline in Alzheimer's Disease Mouse Model. Cell Rep 2020; 27:1345-1355.e6. [PMID: 31042463 DOI: 10.1016/j.celrep.2019.03.087] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 02/11/2019] [Accepted: 03/22/2019] [Indexed: 01/04/2023] Open
Abstract
Amyloid precursor protein (APP) and its metabolites play key roles in Alzheimer's disease (AD) pathophysiology. Whereas short amyloid-β (Aβ) peptides derived from APP are pathogenic, the APP holoprotein serves multiple purposes in the nervous system through its cell adhesion and receptor-like properties. Our studies focused on the signaling mediated by the APP cytoplasmic tail. We investigated whether sustained APP signaling during brain development might favor neuronal plasticity and memory process through a direct interaction with the heterotrimeric G-protein subunit GαS (stimulatory G-protein alpha subunit). Our results reveal that APP possesses autonomous regulatory capacity within its intracellular domain that promotes APP cell surface residence, precludes Aβ production, facilitates axodendritic development, and preserves cellular substrates of memory. Altogether, these events contribute to strengthening cognitive functions and are sufficient to modify the course of AD pathology.
Collapse
Affiliation(s)
- Carole Deyts
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Mary Clutter
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Nicholas Pierce
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Paramita Chakrabarty
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Thomas B Ladd
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Anna Goddi
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Awilda M Rosario
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Pedro Cruz
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Kulandaivelu Vetrivel
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Steven L Wagner
- Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA; Veterans Affairs San Diego Healthcare System, La Jolla, CA 92161, USA
| | - Gopal Thinakaran
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA
| | - Todd E Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, and McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA
| | - Angèle T Parent
- Department of Neurobiology, The University of Chicago, 924 East 57th Street, Chicago, IL 60637, USA.
| |
Collapse
|
603
|
Colasante G, Qiu Y, Massimino L, Di Berardino C, Cornford JH, Snowball A, Weston M, Jones SP, Giannelli S, Lieb A, Schorge S, Kullmann DM, Broccoli V, Lignani G. In vivo CRISPRa decreases seizures and rescues cognitive deficits in a rodent model of epilepsy. Brain 2020; 143:891-905. [PMID: 32129831 PMCID: PMC7089667 DOI: 10.1093/brain/awaa045] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 12/31/2019] [Accepted: 01/14/2020] [Indexed: 01/20/2023] Open
Abstract
Epilepsy is a major health burden, calling for new mechanistic insights and therapies. CRISPR-mediated gene editing shows promise to cure genetic pathologies, although hitherto it has mostly been applied ex vivo. Its translational potential for treating non-genetic pathologies is still unexplored. Furthermore, neurological diseases represent an important challenge for the application of CRISPR, because of the need in many cases to manipulate gene function of neurons in situ. A variant of CRISPR, CRISPRa, offers the possibility to modulate the expression of endogenous genes by directly targeting their promoters. We asked if this strategy can effectively treat acquired focal epilepsy, focusing on ion channels because their manipulation is known be effective in changing network hyperactivity and hypersynchronziation. We applied a doxycycline-inducible CRISPRa technology to increase the expression of the potassium channel gene Kcna1 (encoding Kv1.1) in mouse hippocampal excitatory neurons. CRISPRa-mediated Kv1.1 upregulation led to a substantial decrease in neuronal excitability. Continuous video-EEG telemetry showed that AAV9-mediated delivery of CRISPRa, upon doxycycline administration, decreased spontaneous generalized tonic-clonic seizures in a model of temporal lobe epilepsy, and rescued cognitive impairment and transcriptomic alterations associated with chronic epilepsy. The focal treatment minimizes concerns about off-target effects in other organs and brain areas. This study provides the proof-of-principle for a translational CRISPR-based approach to treat neurological diseases characterized by abnormal circuit excitability.
Collapse
Affiliation(s)
- Gaia Colasante
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Luca Massimino
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | | | - Jonathan H Cornford
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Mikail Weston
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Steffan P Jones
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Serena Giannelli
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy
| | - Andreas Lieb
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Stephanie Schorge
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK.,Department of Pharmacology, UCL School of Pharmacy, University College London, London, UK
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| | - Vania Broccoli
- San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milan, Italy.,CNR Institute of Neuroscience, Via Vanvitelli 32, 20129, Milan, Italy
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, University College London, London, UK
| |
Collapse
|
604
|
Ahnstedt H, Patrizz A, Chauhan A, Roy-O’Reilly M, Furr JW, Spychala MS, D’Aigle J, Blixt FW, Zhu L, Alegria JB, McCullough LD. Sex differences in T cell immune responses, gut permeability and outcome after ischemic stroke in aged mice. Brain Behav Immun 2020; 87:556-567. [PMID: 32058038 PMCID: PMC7590503 DOI: 10.1016/j.bbi.2020.02.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/15/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Stroke is a disease that presents with well-known sex differences. While women account for more stroke deaths, recent data show that after adjusting for age and pre-stroke functional status, mortality is higher in men. Immune responses are key determinants of stroke outcome and may differ by sex. This study examined sex differences in central and peripheral T cell immune responses, systemic effects on gut permeability and microbiota diversity and behavioral outcomes after stroke in aged mice. We hypothesized that there are sex differences in the immune response to stroke in aged animals. METHODS C57BL/6CR mice (20-22 months) were subjected to 60 min middle cerebral artery occlusion, or sham surgery. T cells were quantified in brain and blood at 3, 7 and 15 days (d) post-stroke by flow cytometry. Peripheral effects on gut permeability and microbiota diversity, as well as neurological function were assessed up to 14 d, and at 21 d (cognitive function) post-stroke. Brain glial fibrillary acidic protein (GFAP) expression was evaluated at 42 d post-stroke. RESULTS AND DISCUSSION Mortality (50% vs 14%, p < 0.05) and hemorrhagic transformation (44% vs 0%) were significantly higher in males than in females. No difference in infarct size at 3d were observed. Peripherally, stroke induced greater gut permeability of FITC-dextran in males at d3 (p < 0.05), and non-reversible alterations in microbiota diversity in males. Following the sub-acute phase, both sexes demonstrated a time-dependent increase of CD4+ and CD8+ T cells in the brain, with significantly higher levels of CD8+ T cells and Regulatory T cells in males at d15 (p < 0.01). Aged males demonstrated greater neurological deficits up to d5 and impaired sensorimotor function up to d15 when assessed by the corner asymmetry test (p < 0.001 and p < 0.01, respectively). A trend in greater cognitive decline was observed at d21 in males. Increased GFAP expression in the ischemic hemisphere, indicating astroglial activation and gliosis, was demonstrated in both males and females 42d post-stroke. Our findings indicate that despite a similar initial ischemic brain injury, aged male mice experience greater peripheral effects on the gut and ongoing central neuroinflammation past the sub-acute phase after stroke.
Collapse
Affiliation(s)
- Hilda Ahnstedt
- BRAINS Research Laboratory, Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
605
|
Developmental onset distinguishes three types of spontaneous recognition memory in mice. Sci Rep 2020; 10:10612. [PMID: 32606443 PMCID: PMC7326931 DOI: 10.1038/s41598-020-67619-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 06/09/2020] [Indexed: 11/20/2022] Open
Abstract
Spontaneous recognition memory tasks build on an animal’s natural preference for novelty to assess the what, where and when components of episodic memory. Their simplicity, ethological relevance and cross-species adaptability make them extremely useful to study the physiology and pathology of memory. Recognition memory deficits are common in rodent models of neurodevelopmental disorders, and yet very little is known about the expression of spontaneous recognition memory in young rodents. This is exacerbated by the paucity of data on the developmental onset of recognition memory in mice, a major animal model of disease. To address this, we characterized the ontogeny of three types of spontaneous recognition memory in mice: object location, novel object recognition and temporal order recognition. We found that object location is the first to emerge, at postnatal day (P)21. This was followed by novel object recognition (24 h delay), at P25. Temporal order recognition was the last to emerge, at P28. Elucidating the developmental expression of recognition memory in mice is critical to improving our understanding of the ontogeny of episodic memory, and establishes a necessary blueprint to apply these tasks to probe cognitive deficits at clinically relevant time points in animal models of developmental disorders.
Collapse
|
606
|
Chari T, Griswold S, Andrews NA, Fagiolini M. The Stage of the Estrus Cycle Is Critical for Interpretation of Female Mouse Social Interaction Behavior. Front Behav Neurosci 2020; 14:113. [PMID: 32714163 PMCID: PMC7340104 DOI: 10.3389/fnbeh.2020.00113] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/08/2020] [Indexed: 12/15/2022] Open
Abstract
Female animals in biomedical research have traditionally been excluded from research studies due to the perceived added complexity caused by the estrus cycle. However, given the importance of sex differences in a variety of neurological disorders, testing female mice is critical to identifying sex-linked effects in diseases. To determine the susceptibility of simple behaviors to hormonal fluctuations in the estrus cycle, we studied the effects of sex and the estrus cycle on a variety of behavioral tasks commonly used in mouse phenotyping laboratories. Male and female C57BL/6J mice were tested in a small battery of short duration tests and, immediately on completion of each test, females were classified using cytology of vaginal lavages as sexually-receptive (proestrus and estrus) or non-receptive (NR; metestrus and diestrus). We showed that there was a significant difference in 3-chamber social interaction (SI) between female mice at different stages of their estrus cycle, with sexually-receptive mice showing no preferential interest in a novel female mouse compared with an empty chamber. NR female mice showed the same level of preference for a novel female mouse as male mice did for a novel male mouse. No differences between or within sexes were found for tests of anxiety elevated plus maze (EPM; Hole board), working memory [Novel object recognition (NOR)], and motor learning (repeated tests on rotarod). We conclude that the stage of the estrus cycle may impact SI between same-sex conspecifics, and does not impact performance in the elevated plus-maze, hole board, NOR, and rotarod.
Collapse
Affiliation(s)
- Trishala Chari
- Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA, United States
| | - Sophie Griswold
- Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA, United States
| | - Nick A Andrews
- Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA, United States
| | - Michela Fagiolini
- Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
607
|
Kiffer F, Alexander T, Anderson J, Groves T, McElroy T, Wang J, Sridharan V, Bauer M, Boerma M, Allen A. Late Effects of 1H + 16O on Short-Term and Object Memory, Hippocampal Dendritic Morphology and Mutagenesis. Front Behav Neurosci 2020; 14:96. [PMID: 32670032 PMCID: PMC7332779 DOI: 10.3389/fnbeh.2020.00096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 05/22/2020] [Indexed: 11/17/2022] Open
Abstract
The space extending beyond Earth’s magnetosphere is subject to a complex field of high-energy charged nuclei, which are capable of traversing spacecraft shielding and human tissues, inducing dense ionization events. The central nervous system is a major area of concern for astronauts who will be exposed to the deep-space radiation environment on a mission to Mars, as charged-particle radiation has been shown to elicit changes to the dendritic arbor within the hippocampus of rodents, and related cognitive-behavioral deficits. We exposed 6-month-old male mice to whole-body 1H (0.5 Gy; 150 MeV/n; 18–19 cGy/minute) and an hour later to 16O (0.1Gy; 600 MeV/n; 18–33 Gy/min) at NASA’s Space Radiation Laboratory as a galactic cosmic ray-relevant model. Animals were housed with bedding which provides cognitive enrichment. Mice were tested for cognitive behavior 9 months after exposure to elucidate late radiation effects. Radiation induced significant deficits in novel object recognition and short-term spatial memory (Y-maze). Additionally, we observed opposing morphological differences between the mature granular and pyramidal neurons throughout the hippocampus, with increased dendritic length in the dorsal dentate gyrus and reduced length and complexity in the CA1 subregion of the hippocampus. Dendritic spine analyses revealed a severe reduction in mushroom spine density throughout the hippocampus of irradiated animals. Finally, we detected no general effect of radiation on single-nucleotide polymorphisms in immediate early genes, and genes involved in inflammation but found a higher variant allele frequency in the antioxidants thioredoxin reductase 2 and 3 loci.
Collapse
Affiliation(s)
- Frederico Kiffer
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Tyler Alexander
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Julie Anderson
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Thomas Groves
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Taylor McElroy
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jing Wang
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Vijayalakshmi Sridharan
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Michael Bauer
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Antiño Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.,Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
608
|
Yan F, Tran DM, He H. Robotic Understanding of Object Semantics by Referringto a Dictionary. Int J Soc Robot 2020. [DOI: 10.1007/s12369-020-00657-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
609
|
Whittemore K, Derevyanko A, Martinez P, Serrano R, Pumarola M, Bosch F, Blasco MA. Telomerase gene therapy ameliorates the effects of neurodegeneration associated to short telomeres in mice. Aging (Albany NY) 2020; 11:2916-2948. [PMID: 31140977 PMCID: PMC6555470 DOI: 10.18632/aging.101982] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 05/17/2019] [Indexed: 12/26/2022]
Abstract
Neurodegenerative diseases associated with old age such as Alzheimer’s disease present major problems for society, and they currently have no cure. The telomere protective caps at the ends of chromosomes shorten with age, and when they become critically short, they can induce a persistent DNA damage response at chromosome ends, triggering secondary cellular responses such as cell death and cellular senescence. Mice and humans with very short telomeres owing to telomerase deficiencies have an earlier onset of pathologies associated with loss of the regenerative capacity of tissues. However, the effects of short telomeres in very low proliferative tissues such as the brain have not been thoroughly investigated. Here, we describe a mouse model of neurodegeneration owing to presence of short telomeres in the brain as the consequence of telomerase deficiency. Interestingly, we find similar signs of neurodegeneration in very old mice as the consequence of physiological mouse aging. Next, we demonstrate that delivery of telomerase gene therapy to the brain of these mice results in amelioration of some of these neurodegeneration phenotypes. These findings suggest that short telomeres contribute to neurodegeneration diseases with aging and that telomerase activation may have a therapeutic value in these diseases.
Collapse
Affiliation(s)
- Kurt Whittemore
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Aksinya Derevyanko
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Paula Martinez
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Rosa Serrano
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| | - Martí Pumarola
- Unit of Murine and Comparative Pathology (UPMiC), Department of Animal Medicine and Surgery, Veterinary Faculty, Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Barcelona, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Universitat Autònoma de Barcelona, 08193 Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | - Fàtima Bosch
- Center of Animal Biotechnology and Gene Therapy, Department of Animal Medicine and Surgery, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain.,Center of Animal Biotechnology and Gene Therapy, Department of Biochemistry and Molecular Biology, School of Veterinary Medicine, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid 28029, Spain
| |
Collapse
|
610
|
Droguerre M, Duchêne A, Picoli C, Portal B, Lejards C, Guiard BP, Meunier J, Villard V, Déglon N, Hamon M, Mouthon F, Charvériat M. Efficacy of THN201, a Combination of Donepezil and Mefloquine, to Reverse Neurocognitive Deficits in Alzheimer's Disease. Front Neurosci 2020; 14:563. [PMID: 32612499 PMCID: PMC7309601 DOI: 10.3389/fnins.2020.00563] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/07/2020] [Indexed: 01/15/2023] Open
Abstract
Donepezil (DPZ) is an acetylcholinesterase inhibitor used in Alzheimer’s disease to restore cognitive functions but is endowed with limited efficacy. Recent studies pointed out the implication of astroglial networks in cognitive processes, notably via astrocyte connexins (Cxs), proteins involved in gap junction intercellular communications. Hence, we investigated the impact on cognition of pharmacological or genetic modulations of those astrocyte Cxs during DPZ challenge in two rodent models of Alzheimer’s disease–like memory deficits. We demonstrated that the Cx modulator mefloquine (MEF) significantly enhanced the procognitive effect of DPZ in both models. In parallel, we determined that MEF potentiated DPZ-induced release of acetylcholine in hippocampus. Finally, local genetic silencing of astrocyte Cxs in the hippocampus was also found to enhance the procognitive effect of DPZ, pointing out the importance of Cx-dependent astrocyte networks in memory processes.
Collapse
Affiliation(s)
| | | | | | - Benjamin Portal
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Camille Lejards
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Bruno P Guiard
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | | | | | - Nicole Déglon
- Laboratory of Neurotherapies and NeuroModulation, Neuroscience Research Center (CRN), University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and NeuroModulation, Department of Clinical Neuroscience (DNC), Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | | | | | | |
Collapse
|
611
|
Zhong X, Xie L, Yang X, Liang F, Yang Y, Tong J, Zhong Y, Zhao K, Tang Y, Yuan C. Ethyl pyruvate protects against sepsis-associated encephalopathy through inhibiting the NLRP3 inflammasome. Mol Med 2020; 26:55. [PMID: 32517686 PMCID: PMC7285451 DOI: 10.1186/s10020-020-00181-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 05/18/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND With the advance of antibiotics and life support therapy, the mortality of sepsis has been decreasing in recent years. However, the incidence of sepsis-associated encephalopathy (SAE), a common complication of sepsis, is still high. There are few effective therapies to treat clinical SAE. We previously found that ethyl pyruvate (EP), a metabolite derivative, is able to effectively inhibit the NLRP3 inflammasome activation. Administration of ethyl pyruvate protects mice against polymicrobial sepsis in cecal ligation and puncture (CLP) model. The aim of present study is to investigate if ethyl pyruvate is able to attenuate SAE. METHODS After CLP, C57BL/6 mice were intraperitoneally or intrathecally injected with saline or ethyl pyruvate using the sham-operated mice as control. New Object Recognition (NOR) and Morris Water Maze (MWM) were conducted to determine the cognitive function. Brain pathology was assessed via immunohistochemistry. To investigate the mechanisms by which ethyl pyruvate prevent SAE, the activation of NLRP3 in the hippocampus and the microglia were determined using western blotting, and cognitive function, microglia activation, and neurogenesis were assessed using WT, Nlrp3-/- and Asc-/- mice in the sublethal CLP model. In addition, Nlrp3-/- and Asc-/- mice treated with saline or ethyl pyruvate were subjected to CLP. RESULTS Ethyl pyruvate treatment significantly attenuated CLP-induced cognitive decline, microglia activation, and impaired neurogenesis. In addition, EP significantly decreased the NLRP3 level in the hippocampus of the CLP mice, and inhibited the cleavage of IL-1β induced by NLRP3 inflammsome in microglia. NLRP3 and ASC deficiency demonstrated similar protective effects against SAE. Nlrp3-/- and Asc-/- mice significantly improved cognitive function and brain pathology when compared with WT mice in the CLP models. Moreover, ethyl pyruvate did not have additional effects against SAE in Nlrp3-/- and Asc-/- mice. CONCLUSION The results demonstrated that ethyl pyruvate confers protection against SAE through inhibiting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Xiaoli Zhong
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China
| | - Lingli Xie
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China
| | - Xiaolong Yang
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China
| | - Fang Liang
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
| | - Yanliang Yang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, 410000, P. R. China
| | - Jianbin Tong
- Department of Anesthesiology, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yanjun Zhong
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China
- ICU Center, The Second Xiangya Hospital, Central South University, No. 139 Renmin Middle Road, Furong, Changsha, 410011, Hunan, China
| | - Kai Zhao
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China
| | - Yiting Tang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China.
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, 410000, P. R. China.
| | - Chuang Yuan
- Department of Hematology and Critical Care Medicine, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan Province, 410000, P. R. China.
- Department of Pathophysiology, School of Basic Medical Science, Central South University, 138 Tong-zi-po Road, Changsha, Hunan Province, 410000, P. R. China.
| |
Collapse
|
612
|
Teixeira AE, Rocha-Gomes A, Pereira dos Santos T, Amaral BLS, da Silva AA, Malagutti AR, Leite FRF, Stuckert-Seixas SR, Riul TR. Cafeteria diet administered from lactation to adulthood promotes a change in risperidone sensitivity on anxiety, locomotion, memory, and social interaction of Wistar rats. Physiol Behav 2020; 220:112874. [DOI: 10.1016/j.physbeh.2020.112874] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 03/05/2020] [Accepted: 03/09/2020] [Indexed: 01/24/2023]
|
613
|
Hosseini-Sharifabad A, Rabbani M, Seyed-Yousefi Y, Safavi M. Magnesium Increases the Protective Effect of Citicoline on Aluminum Chloride-induced Cognitive Impairment. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2020; 18:241-248. [PMID: 32329305 PMCID: PMC7242111 DOI: 10.9758/cpn.2020.18.2.241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 09/04/2019] [Accepted: 09/07/2019] [Indexed: 11/18/2022]
Abstract
Objective Alzheimer's disease is a popular neurodegenerative disorder which is growing in the elderly people. Exposure to environmental pollutant like aluminum could trigger or accelerate its involved mechanisms like tau phosphorylation. The current study will evaluate the effect of alone or co-administration of Citicoline or/and magnesium on the aluminum chloride induced memory impairment. Methods Male albino mice were randomly divided into different groups (n = 7). Memory impairment was induced via orally administration of 300 mg/kg Aluminum Chloride for 28 days. Based on respective group, animals received 100, 250, 500 mg/kg of Citicoline or 50, 100, 150 mg/kg of Magnesium sulfate (MgSO4), intraperitoneally. In co-administration, 50 mg/kg of MgSO4 injected concomitantly with 100, 250, or 500 mg/kg of Citicoline. Rivastigmine (2 mg/kg intraperitoneally) was used as a positive control. Memory was evaluated using the Object Recognition Task (ORT) and Passive Avoidance Test (PAT). Results The studied doses of Citicoline or MgSO4 when administered individually showed significant increase in the discrimination index in ORT and latency time in the PAT compared to the Aluminium chloride (AlCl3) treated group. Concomitant injection of 50 mg/kg MgSO4 with the different doses of Citicoline strongly increased the above indices values in comparison to each alone. Conclusion The findings show, individual administration of Citicoline or MgSO4 inverted the AlCl3-induced memory impairment in a dose independent manner. The addition of MgSO4 to the Citicoline showed a synergistic effect in the PAT and likely additive effect in the ORT.
Collapse
Affiliation(s)
- Ali Hosseini-Sharifabad
- Department of Pharmacology and Toxicology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Rabbani
- Department of Pharmacology and Toxicology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Yasaman Seyed-Yousefi
- Department of Pharmacology and Toxicology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Maryam Safavi
- Department of Pharmacology and Toxicology and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
614
|
Heimfarth L, Dos Anjos KS, de Carvalho YMBG, Dos Santos BL, Serafini MR, de Carvalho Neto AG, Nunes PS, Beserra Filho JIA, da Silva SP, Ribeiro AM, Bezerra DP, Marreto RN, de Souza Siqueira Quintans J, de Souza Araújo AA, Melo Coutinho HD, Scotti MT, Scotti L, Quintans-Júnior LJ. Characterization of β-cyclodextrin/myrtenol complex and its protective effect against nociceptive behavior and cognitive impairment in a chronic musculoskeletal pain model. Carbohydr Polym 2020; 244:116448. [PMID: 32536383 DOI: 10.1016/j.carbpol.2020.116448] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/29/2022]
Abstract
Myrtenol has gained wide interest because of its pharmacological profiles, mainly for treatment of chronic diseases. To improve the solubility of myrtenol, the formation of inclusion complexes with β-cyclodextrin was performed by physical mixture, kneading process or slurry complexation (SC) methods and characterized using thermal analysis, XRD, SEM and NMR. From these results, myrtenol complexed by SC was successfully complexed into β-cyclodextrin cavity. The interaction between myrtenol and β-cyclodextrin was confirmed by molecular docking. Hence, the SC β-cyclodextrin-myrtenol complex was evaluate for its anti-hyperalgesic, anxiolytic and antioxidant activity in a fibromyalgia model. Results show that myrtenol and β-cyclodextrin form a stable complex and have anti-hyperalgesic effect, improve the cognitive impairment caused and have an anxiolytic-like effect. Furthermore, the β-cyclodextrin/myrtenol complex decrease lipoperoxidation, increased catalase activity and a reduce SOD/CAT ratio. Therefore, β-cyclodextrin/myrtenol complex reduce painful behavior, improves motor skills and emotional behavior and decreases oxidative stress in a fibromyalgia model.
Collapse
Affiliation(s)
- Luana Heimfarth
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | | | | | | | | | - Paula Santos Nunes
- Department of Physiology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | - Sara Pereira da Silva
- Department of Biosciences, Federal University of São Paulo/UNIFESP, Santos, SP, Brazil
| | | | - Daniel Pereira Bezerra
- Oswaldo Cruz Foundation, Laboratory of Tissue Engineering and Immunopharmacology, Salvador, BA, Brazil
| | | | | | | | - Henrique Douglas Melo Coutinho
- Department of Biological Chemistry, Regional University of Cariri, URCA, Av Cel. Antonio Luiz, 1161, Pimenta, Crato, CE, 63105-000, Brazil.
| | - Marcus T Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | - Luciana Scotti
- Cheminformatics Laboratory- Postgraduate Program in Natural Products and Synthetic Bioactive, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | | |
Collapse
|
615
|
Di Domenico M, Benevenuto SGDM, Tomasini PP, Yariwake VY, de Oliveira Alves N, Rahmeier FL, da Cruz Fernandes M, Moura DJ, Nascimento Saldiva PH, Veras MM. Concentrated ambient fine particulate matter (PM 2.5) exposure induce brain damage in pre and postnatal exposed mice. Neurotoxicology 2020; 79:127-141. [PMID: 32450181 DOI: 10.1016/j.neuro.2020.05.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/20/2022]
Abstract
Air pollution is a public health concern that has been associated with adverse effects on the development and functions of the central nervous system (CNS). However, studies on the effects of exposure to pollutants on the CNS across the entire developmental period still remain scarce. In this study, we investigated the impacts of prenatal and/or postnatal exposure to fine particulate matter (PM2.5) from São Paulo city, on the brain structure and behavior of juvenile male mice. BALB/c mice were exposed to PM2.5 concentrated ambient particles (CAP) at a daily concentration of 600 μg/m³ during the gestational [gestational day (GD) 1.5-18.5] and the postnatal periods [postnatal day (PND) 22-90] to filtered air (FA) in both periods (FA/FA), to CAP only in the postnatal period (FA/CAP), to CAP only in the gestational period (CAP/FA), and to CAP in both periods (CAP/CAP). Behavioral tests were performed when animals were at PND 30 and PND 90. Glial activation, brain volume, cortical neuron number, serotonergic and GABAergic receptors, as well as oxidative stress, were measured. Mice at PND 90 presented greater behavioral changes in the form of greater locomotor activity in the FA-CAP and CAP-CAP groups. In general, these same groups explored objects longer and the CAP-FA group presented anxiolytic behavior. There was no difference in total brain volume among groups, but a lower corpus callosum (CC) volume was observed in the CAP-FA group. Also, the CAP-CAP group presented an increase in microglia in the cortex and an increased in astrocytes in the cortex, CC, and C1A and dentate gyrus of hippocampus regions. Gene expression analysis showed a decrease in BDNF in the hippocampus of CAP-CAP group. Treatment of immortalized glial cells with non-cytotoxic doses of ambient PM2.5 increased micronuclei frequencies, indicating genomic instability. These findings highlight the potential for negative neurodevelopmental outcomes induced by exposure to moderate levels of PM2.5 in Sao Paulo city.
Collapse
Affiliation(s)
- Marlise Di Domenico
- Department of Pathology, LIM05-HCFMUSP, Laboratory of Experimental Air Pollution, School of Medicine, University of São Paulo, São Paulo, Brazil.
| | | | - Paula Pellenz Tomasini
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Yuji Yariwake
- Department of Pathology, LIM05-HCFMUSP, Laboratory of Experimental Air Pollution, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Nilmara de Oliveira Alves
- Department of Pathology, LIM05-HCFMUSP, Laboratory of Experimental Air Pollution, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Francine Luciano Rahmeier
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Marilda da Cruz Fernandes
- Pathology Research Laboratory, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Dinara Jaqueline Moura
- Laboratory of Genetic Toxicology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Department of Pathology, LIM05-HCFMUSP, Laboratory of Experimental Air Pollution, School of Medicine, University of São Paulo, São Paulo, Brazil
| | - Mariana Matera Veras
- Department of Pathology, LIM05-HCFMUSP, Laboratory of Experimental Air Pollution, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
616
|
Chen J, Tan L, Liao Y, Long J, Zhou Y, Wei J, Zhou Y. Chemokine CCL2 impairs spatial memory and cognition in rats via influencing inflammation, glutamate metabolism and apoptosis-associated genes expression- a potential mechanism for HIV-associated neurocognitive disorder. Life Sci 2020; 255:117828. [PMID: 32454160 DOI: 10.1016/j.lfs.2020.117828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 01/16/2023]
Abstract
AIMS To explore the role of chemokine CC motif ligand 2 (CCL2) in spatial memory and cognition impairment, and the underlying mechanisms focused on inflammatory, glutamate metabolistic and apoptotic- associated mRNA expression. MATERIALS AND METHODS Stereotaxic surgery was performed here to establish a rat model by bilateral intra-hippocampal injection of CCL2. Morris water maze (MWM) and Novel object recognition test (NORT) were used to assess the learning, memory and cognitive ability respectively. RT-PCR was used to detect the relative mRNA expression of inflammatory, glutamate metabolistic and apoptotic- associated indexes. Nissl and TUNEL staining were performed to observe the morphological changes of hippocampal CA1 zone and quantified the apoptosis of hippocampal neurons of CA1 zones respectively. KEY FINDINGS We found CCL2 injured cognitive function in rats. Six days after CCL2 injection, we revealed the following obvious mRNA expression changes: (1) increasing of the neuroinflammatory cytokines IL-1β, CXCL-10, IL-6; (2) decreasing of the glutamate transporters GLT-1 and GLAST and increasing of PAG; (3) increasing of the apoptotic genes caspase-8, caspase-3 and Bax, while decreasing the anti-apoptotic gene Bcl-2. Further, Nissl staining and TUNEL confirmed the injury of the structure of hippocampal CA1 zones and the apoptosis of hippocampal neurons. SIGNIFICANCE Our results indicated that CCL2 impaired spatial memory and cognition, the involving mechanisms may link to the up-regulation of mRNA expression of the three major pathological events: inflammation, excitotoxicity and neuronal apoptosis, which were involved in HIV-associated neurocognitive disorder (HAND). Taken together, these findings suggest a potential therapeutic strategy against CCL2.
Collapse
Affiliation(s)
- Jianmin Chen
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Liqiu Tan
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Yuanjun Liao
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Jiangyi Long
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Yinjun Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China
| | - Jinbin Wei
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China.
| | - Yan Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, Guangxi 530021, People's Republic of China.
| |
Collapse
|
617
|
de Almeida RF, Pocharski CB, Rodrigues ALS, Elisabetsky E, Souza DO. Guanosine fast onset antidepressant-like effects in the olfactory bulbectomy mice model. Sci Rep 2020; 10:8429. [PMID: 32439951 PMCID: PMC7242421 DOI: 10.1038/s41598-020-65300-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 04/28/2020] [Indexed: 12/14/2022] Open
Abstract
The treatment of major depressive disorder (MDD) is still a challenge. In the search for novel antidepressants, glutamatergic neuromodulators have been investigated as possible fast-acting antidepressants. Innovative studies suggest that the purine cycle and/or the purinergic signaling can be dysregulated in MDD, and the endogenous nucleoside guanosine has gained attention due to its extracellular effects. This study aimed to verify if guanosine produces fast-onset effects in the well-validated, reliable and sensitive olfactory bulbectomy (OBX) model of depression. The involvement of the mTOR pathway, a key target for the fast-onset effect of ketamine, was also investigated. Results show that a single i.p. injection of guanosine, or ketamine, completely reversed the OBX-induced anhedonic-like behavior 24 or 48 h post treatment, as well as the short-term recognition memory impairment 48 h post treatment. The antidepressant-like effects of guanosine and ketamine were completely abolished by rapamycin. This study shows, for the first time, that guanosine, in a way similar to ketamine, is able to elicit a fast antidepressant response in the OBX model in mice. The results support the notion that guanosine represents a new road for therapeutic improvement in MDD.
Collapse
Affiliation(s)
- Roberto Farina de Almeida
- Departamento de Ciências Biológicas, Programa de Pós-Graduação em Ciências Biológicas, Universidade Federal de Ouro Preto (UFOP), Ouro Preto, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - Camila Barbosa Pocharski
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Ana Lúcia S Rodrigues
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elaine Elisabetsky
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Diogo O Souza
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
618
|
Sex Differences in Cognitive Impairment Induced by Cerebral Microhemorrhage. Transl Stroke Res 2020; 12:316-330. [PMID: 32440818 DOI: 10.1007/s12975-020-00820-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 10/24/2022]
Abstract
It has been suggested that cerebral microhemorrhages (CMHs) could be involved in cognitive decline. However, little is known about the sex-dependency of this effect. Using a multimodal approach combining behavioral tests, in vivo imaging, biochemistry, and molecular biology, we studied the cortical and hippocampal impact of a CMH in male and female mice (C57BL/6J) 6 weeks post-induction using a collagenase-induced model. Our work shows for the first time that a single cortical CMH exerts sex-specific effects on cognition. It notably induced visuospatial memory impairment in males only. This sex difference might be explained by cortical changes secondary to the lesion. In fact, the CMH induced an upregulation of ERα mRNA only in the female cortex. Besides, in male mice, we observed an impairment of pathways associated to neuronal, glial, or vascular functions: decrease in the P-GSK3β/GSK3β ratio, in BDNF and VEGF levels, and in microvascular water mobility. The CMH also exerted spatial remote effects in the hippocampus by increasing the number of astrocytes in both sexes, increasing the mean area occupied by each astrocyte in males, and decreasing hippocampal BDNF in females suggesting a cortical-hippocampal network impairment. This work demonstrates that a CMH could directly affect cognition in a sex-specific manner and highlights the need to study both sexes in preclinical models.
Collapse
|
619
|
Turlova E, Wong R, Xu B, Li F, Du L, Habbous S, Horgen FD, Fleig A, Feng ZP, Sun HS. TRPM7 Mediates Neuronal Cell Death Upstream of Calcium/Calmodulin-Dependent Protein Kinase II and Calcineurin Mechanism in Neonatal Hypoxic-Ischemic Brain Injury. Transl Stroke Res 2020; 12:164-184. [PMID: 32430797 DOI: 10.1007/s12975-020-00810-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 03/12/2020] [Accepted: 03/18/2020] [Indexed: 11/25/2022]
Abstract
Transient receptor potential melastatin 7 (TRPM7), a calcium-permeable, ubiquitously expressed ion channel, is critical for axonal development, and mediates hypoxic and ischemic neuronal cell death in vitro and in vivo. However, the downstream mechanisms underlying the TRPM7-mediated processes in physiology and pathophysiology remain unclear. In this study, we employed a mouse model of hypoxic-ischemic brain cell death which mimics the pathophysiology of hypoxic-ischemic encephalopathy (HIE). HIE is a major public health issue and an important cause of neonatal deaths worldwide; however, the available treatments for HIE remain limited. Its survivors face life-long neurological challenges including mental retardation, cerebral palsy, epilepsy and seizure disorders, motor impairments, and visual and auditory impairments. Through a proteomic analysis, we identified calcium/calmodulin-dependent protein kinase II (CaMKII) and phosphatase calcineurin as potential mediators of cell death downstream from TRPM7 activation. Further analysis revealed that TRPM7 mediates cell death through CaMKII, calmodulin, calcineurin, p38, and cofilin cascade. In vivo, we found a significant reduction of brain injury and improvement of short- and long-term functional outcomes after HI after administration of specific TRPM7 blocker waixenicin A. Our data demonstrate a molecular mechanism of TRPM7-mediated cell death and identifies TRPM7 as a promising therapeutic and drug development target for HIE.
Collapse
Affiliation(s)
- Ekaterina Turlova
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Raymond Wong
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Baofeng Xu
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Feiya Li
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Lida Du
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - Steven Habbous
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, HI, 96744, USA
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, 96720, USA
| | - Zhong-Ping Feng
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
| | - Hong-Shuo Sun
- Department of Surgery, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, M5S 1A8, Canada.
- Department of Pharmacology, University of Toronto, 1 King's College Circle, Toronto, M5S 1A8, Canada.
- Leslie Dan Faculty of Pharmacy, University of Toronto, University of Toronto, Toronto, Canada.
| |
Collapse
|
620
|
Prefrontal-hippocampal functional connectivity encodes recognition memory and is impaired in intellectual disability. Proc Natl Acad Sci U S A 2020; 117:11788-11798. [PMID: 32393630 DOI: 10.1073/pnas.1921314117] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Down syndrome (DS) is the most common form of intellectual disability. The cognitive alterations in DS are thought to depend on brain regions critical for learning and memory such as the prefrontal cortex (PFC) and the hippocampus (HPC). Neuroimaging studies suggest that increased brain connectivity correlates with lower intelligence quotients (IQ) in individuals with DS; however, its contribution to cognitive impairment is unresolved. We recorded neural activity in the PFC and HPC of the trisomic Ts65Dn mouse model of DS during quiet wakefulness, natural sleep, and the performance of a memory test. During rest, trisomic mice showed increased theta oscillations and cross-frequency coupling in the PFC and HPC while prefrontal-hippocampal synchronization was strengthened, suggesting hypersynchronous local and cross-regional processing. During sleep, slow waves were reduced, and gamma oscillations amplified in Ts65Dn mice, likely reflecting prolonged light sleep. Moreover, hippocampal sharp-wave ripples were disrupted, which may have further contributed to deficient memory consolidation. Memory performance in euploid mice correlated strongly with functional connectivity measures that indicated a hippocampal control over memory acquisition and retrieval at theta and gamma frequencies, respectively. By contrast, trisomic mice exhibited poor memory abilities and disordered prefrontal-hippocampal functional connectivity. Memory performance and key neurophysiological alterations were rescued after 1 month of chronic administration of a green tea extract containing epigallocatequin-3-gallate (EGCG), which improves executive function in young adults with DS and Ts65Dn mice. Our findings suggest that abnormal prefrontal-hippocampal circuit dynamics are candidate neural mechanisms for memory impairment in DS.
Collapse
|
621
|
Endonuclease VIII-like 1 deficiency impairs survival of newly generated hippocampal neurons and memory performance in young-adult male mice. Life Sci 2020; 254:117755. [PMID: 32437792 DOI: 10.1016/j.lfs.2020.117755] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/29/2020] [Accepted: 05/04/2020] [Indexed: 11/20/2022]
Abstract
AIMS Efficient memory formation in rodents depends on adult neurogenesis in the subgranular zone of the hippocampus, and mounting evidence suggests that deficiencies in initiating repair of oxidatively induced DNA damage may impair neurogenesis. Hence, we aimed to determine whether loss of the DNA glycosylase, endonuclease VIII-like 1 (Neil1), affects hippocampal neurogenesis and memory performance in young-adult mice. MAIN METHODS Eight-week-old male wild-type and Neil1-deficient (Neil1-/-) mice were treated with bromodeoxyuridine to track neuronal proliferation and differentiation. A neurosphere formation assay was further used to measure neuroprogenitor proliferative capacity. Hippocampus-related memory functions were assessed with Y-maze spontaneous alternation and novel object recognition tests. KEY FINDINGS Young-adult male Neil1-/- mice exhibited diminished adult hippocampal neurogenesis in the dentate gyrus, probably as a result of poor survival of newly proliferated neurons. Furthermore, the Y-maze spontaneous alternation and novel object recognition tests respectively revealed that Neil1 deficiency impairs spatial and non-spatial hippocampus-related memory functions. We also found that expression of p53, a central regulator of apoptosis, was upregulated in the dentate gyrus of Neil1-/- mice, while the level of β-catenin, a key cell survival molecule, was downregulated. SIGNIFICANCE The DNA glycosylase, Neil1, promotes successful hippocampal neurogenesis and learning and memory in young-adult mice.
Collapse
|
622
|
Khan M, Kumar P, Qiao F, Islam SMT, Singh AK, Won JS, Feng W, Singh I. Targeting GSNOR for functional recovery in a middle-aged mouse model of stroke. Brain Res 2020; 1741:146879. [PMID: 32418890 DOI: 10.1016/j.brainres.2020.146879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/01/2023]
Abstract
The nitric oxide (NO) metabolome and the NO metabolite-based neurovascular protective pathways are dysregulated after stroke. The major NO metabolite S-nitrosoglutahione (GSNO) is essential for S-nitrosylation-based signaling events and the inhibition of S-nitrosoglutahione (GSNO)-metabolizing enzyme GSNO reductase (GSNOR) provides protective effects following cardiac ischemia. However, the role of GSNOR and GSNOR inhibition-mediated increased GSNO/S-nitrosylation is not understood in neurovascular diseases such as stroke. Because age is the major risk factor of stroke and recovery in aged stroke patients is low and slow, we investigated the efficacy of GSNOR inhibition using a GSNOR selective inhibitor N6022 in a clinically relevant middle-aged cerebral ischemia and reperfusion (IR) mouse model of stroke. N6022 (5 mg/kg; iv) treatment of IR mice at 2 h after reperfusion followed by the treatment of the same dose daily for 3 days reduced the infarct volume and decreased the neurological score. Daily treatment of IR animals with N6022 for 2 weeks significantly improved neurological score, brain infarctions/atrophy, survival rate, motor (measured by cylinder test) and cognitive (evaluated by novel object recognition test) functions which paralleled the decreased activity of GSNOR, reduced levels of peroxynitrite and decreased neurological score. These results are the first evidence of a new pathway for the treatment of stroke via the inhibition of GSNOR. Based on the efficacy of N6022 in the stroke animal model and its use in human therapeutic studies without toxicity, we submit that GSNOR is a druggable target, and N6022 is a promising drug candidate for human stroke therapy.
Collapse
Affiliation(s)
- Mushfiquddin Khan
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Pavan Kumar
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States.
| | - Fei Qiao
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - S M Touhidul Islam
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| | - Je-Seong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, United States.
| | - Wayne Feng
- Department of Neurology, Duke University School of Medicine, Durham, NC, United States.
| | - Inderjit Singh
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC, United States; Ralph H. Johnson VA Medical Center, Charleston, SC, United States.
| |
Collapse
|
623
|
Wu M, Liao M, Huang R, Chen C, Tian T, Wang H, Li J, Li J, Sun Y, Wu C, Li Q, Xiao X. Hippocampal overexpression of TREM2 ameliorates high fat diet induced cognitive impairment and modulates phenotypic polarization of the microglia. Genes Dis 2020; 9:401-414. [PMID: 35224156 PMCID: PMC8843863 DOI: 10.1016/j.gendis.2020.05.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 01/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) share several common pathophysiological features. Rare variants of triggering receptor expressed on myeloid cells 2 (TREM2) increase the risk of developing AD, suggesting the involvement of TREM2 and innate immunity in AD development. It is still unknown whether TREM2 is related to cognitive impairment in T2DM. Here, we investigated the effects of the hippocampal overexpression of TREM2 on cognitive in long-term high-fat diet (HFD)-fed mice. Male C57BL/6J mice were maintained on HFD for 50 weeks. TREM2 was overexpressed in the hippocampus 36 weeks after HFD feeding using adeno-associated virus vector (AAV)-mediated gene delivery. The results showed that the HFD feeding induced rapid and persistent weight gain, glucose intolerance and significant impairments in learning and memory. Compared with AAV-con, AAV-TREM2 significantly ameliorated cognitive impairment without altering body weight and glucose homeostasis in HFD mice. The overexpression of TREM2 upregulated the synaptic proteins spinophilin, PSD95 and synaptophysin, suggesting the improvement in synaptic transmission. Dendritic complexity and spine density in the CA1 region were rescued after TREM2 overexpression. Furthermore, TREM2 markedly increased the number of iba-1/Arg-1-positive microglia in the hippocampus, suppressed neuroinflammation and microglial activation. In sum, hippocampal TREM2 plays an important role in improving HFD-induced cognitive dysfunction and promoting microglial polarization towards the M2 anti-inflammatory phenotype. Our study also suggests that TREM2 might be a novel target for the intervention of obesity/diabetes-associated cognitive decline.
Collapse
Affiliation(s)
- Min Wu
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Maolin Liao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Rongfeng Huang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Chunxiu Chen
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Tian Tian
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Hongying Wang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jiayu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Jibin Li
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuxiang Sun
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Qifu Li
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaoqiu Xiao
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Corresponding author. Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
624
|
In silico, in vitro and in vivo studies indicate resveratrol analogue as a potential alternative for neuroinflammatory disorders. Life Sci 2020; 249:117538. [DOI: 10.1016/j.lfs.2020.117538] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 03/03/2020] [Accepted: 03/08/2020] [Indexed: 12/18/2022]
|
625
|
Wang C, Liu Z, Hu T, Li Y, Liu R, Zhang J, He H. Potential neurotoxicity of 5-hydroxymethylfurfural and its oligomers: widespread substances in carbohydrate-containing foods. Food Funct 2020; 11:4216-4223. [PMID: 32352452 DOI: 10.1039/c9fo02526j] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
5-Hydroxymethylfurfural (5-HMF) is present in numerous carbohydrate-containing consumer products and is readily converted into two oligomers (II and III) by acid-catalyzed transformations. Previous studies have demonstrated various undesirable effects of 5-HMF at relatively high concentrations. In this study, we demonstrate that 5-HMF and its two oligomers exert neurotoxic effects in vivo and in vitro. All three substances blocked the proliferation of PC12 and HT22 cells at the S or G2-M phase in dose- and time-dependent manners. In addition, [Ca2+]i and reactive oxygen species levels were both significantly increased by treatment with these substances at 100 μM, individually, compared with the control group. Although no motor and cognitive deficits are observed, 5-HMF and III can induce anxiety- and depression-like behavior in adolescent mice at administered doses of 0.15 mg kg-1 and 1.5 mg kg-1in vivo, which are close to or less than the reported 24 h dietary intake of 5-HMF in humans. Together, our findings suggest the need for close monitoring of the content of these substances in food, as well as the need for studies on the effects of long-term exposure to them.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710061, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
626
|
Corsetti V, Borreca A, Latina V, Giacovazzo G, Pignataro A, Krashia P, Natale F, Cocco S, Rinaudo M, Malerba F, Florio R, Ciarapica R, Coccurello R, D’Amelio M, Ammassari-Teule M, Grassi C, Calissano P, Amadoro G. Passive immunotherapy for N-truncated tau ameliorates the cognitive deficits in two mouse Alzheimer's disease models. Brain Commun 2020; 2:fcaa039. [PMID: 32954296 PMCID: PMC7425324 DOI: 10.1093/braincomms/fcaa039] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/05/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and neuropathological studies have shown that tau pathology better correlates with the severity of dementia than amyloid plaque burden, making tau an attractive target for the cure of Alzheimer's disease. We have explored whether passive immunization with the 12A12 monoclonal antibody (26-36aa of tau protein) could improve the Alzheimer's disease phenotype of two well-established mouse models, Tg2576 and 3xTg mice. 12A12 is a cleavage-specific monoclonal antibody which selectively binds the pathologically relevant neurotoxic NH226-230 fragment (i.e. NH2htau) of tau protein without cross-reacting with its full-length physiological form(s). We found out that intravenous administration of 12A12 monoclonal antibody into symptomatic (6 months old) animals: (i) reaches the hippocampus in its biologically active (antigen-binding competent) form and successfully neutralizes its target; (ii) reduces both pathological tau and amyloid precursor protein/amyloidβ metabolisms involved in early disease-associated synaptic deterioration; (iii) improves episodic-like type of learning/memory skills in hippocampal-based novel object recognition and object place recognition behavioural tasks; (iv) restores the specific up-regulation of the activity-regulated cytoskeleton-associated protein involved in consolidation of experience-dependent synaptic plasticity; (v) relieves the loss of dendritic spine connectivity in pyramidal hippocampal CA1 neurons; (vi) rescues the Alzheimer's disease-related electrophysiological deficits in hippocampal long-term potentiation at the CA3-CA1 synapses; and (vii) mitigates the neuroinflammatory response (reactive gliosis). These findings indicate that the 20-22 kDa NH2-terminal tau fragment is crucial target for Alzheimer's disease therapy and prospect immunotherapy with 12A12 monoclonal antibody as safe (normal tau-preserving), beneficial approach in contrasting the early Amyloidβ-dependent and independent neuropathological and cognitive alterations in affected subjects.
Collapse
Affiliation(s)
| | - Antonella Borreca
- Humanitas University Laboratory of Pharmacology and Brain Pathology, Neuro Center, 20089 Milan, Italy
- Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | - Paraskevi Krashia
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | - Francesca Natale
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Rita Florio
- European Brain Research Institute (EBRI), 00161 Rome, Italy
| | | | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Institute for Complex Systems (ISC), CNR, 00185 Rome, Italy
| | - Marcello D’Amelio
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | | | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT)–National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
627
|
Jiang LX, Huang GD, Su F, Wang H, Zhang C, Yu X. Vortioxetine administration attenuates cognitive and synaptic deficits in 5×FAD mice. Psychopharmacology (Berl) 2020; 237:1233-1243. [PMID: 31953648 DOI: 10.1007/s00213-020-05452-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 01/02/2020] [Indexed: 01/10/2023]
Abstract
RATIONALE AND OBJECTIVE Vortioxetine has been reported to exhibit a variety of neurobiological functions and neuroprotective effects. In the present study, we aimed to investigate the effects of vortioxetine on cognitive performance in a transgenic mouse model of Alzheimer's disease (AD). METHODS We administered vortioxetine (10 mg/kg, i.p., every day, for approximately 6 weeks), which acts on multiple 5-serotonin (5-HT) receptors, to 3.5-month-old 5×FAD mice. Subsequently, we used the open field (OF) test to detect anxiety-like behavior in the mice. The novel object recognition (NOR) test and Morris water maze (MWM) were used to assess the cognitive states of the 5×FAD mice. We also measured the levels of insoluble amyloid plaques and soluble β-amyloid (Aβ) plaques. Finally, we explored the expression levels of postsynaptic density protein 95 (PSD95), synaptophysin (SYP), and synaptotagmin-1 (SYT1) in the hippocampus of the mice. RESULTS The administration of vortioxetine effectively reversed the reduction in anxiety-type behaviors in 5×FAD mice and improved the impairment in recognition memory and spatial reference memory. However, we did not find that vortioxetine decreased or delayed the formation of amyloid plaques or Aβ. Interestingly, we found a significant increase in the expression levels of PSD95, SYP, and SYT1 in the 5×FAD mice after vortioxetine treatment compared with the control group. CONCLUSION These results demonstrate that vortioxetine may improve cognitive impairment in 5×FAD mice. The role in cognitive improvement may be related to the beneficial effects of vortioxetine on synaptic function.
Collapse
Affiliation(s)
- Li-Xin Jiang
- School of Mental Health, Wenzhou Medical University, Chashan Higher Education Park, Ouhai District, Wenzhou, 325035, China
- Peking University Institute of Mental Health (Sixth Hospital), No. 51 Huayuanbei Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health, Peking University, Beijing, 100191, China
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, 100191, China
| | - Geng-Di Huang
- Peking University Institute of Mental Health (Sixth Hospital), No. 51 Huayuanbei Road, Haidian District, Beijing, 100191, China
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health, Peking University, Beijing, 100191, China
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, 100191, China
| | - Feng Su
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, No.10 Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100069, China
| | - Huali Wang
- Peking University Institute of Mental Health (Sixth Hospital), No. 51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health, Peking University, Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| | - Chen Zhang
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, 100191, China.
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, No.10 Xitoutiao, You'anmenwai, Fengtai District, Beijing, 100069, China.
| | - Xin Yu
- School of Mental Health, Wenzhou Medical University, Chashan Higher Education Park, Ouhai District, Wenzhou, 325035, China.
- Peking University Institute of Mental Health (Sixth Hospital), No. 51 Huayuanbei Road, Haidian District, Beijing, 100191, China.
- National Clinical Research Center for Mental Disorders & NHC Key Laboratory of Mental Health, Peking University, Beijing, 100191, China.
- Beijing Municipal Key Laboratory for Translational Research on Diagnosis and Treatment of Dementia, Beijing, 100191, China.
| |
Collapse
|
628
|
Church JS, Chace-Donahue F, Blum JL, Ratner JR, Zelikoff JT, Schwartzer JJ. Neuroinflammatory and Behavioral Outcomes Measured in Adult Offspring of Mice Exposed Prenatally to E-Cigarette Aerosols. ENVIRONMENTAL HEALTH PERSPECTIVES 2020; 128:47006. [PMID: 32293200 PMCID: PMC7228099 DOI: 10.1289/ehp6067] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
BACKGROUND In an effort to decrease the rates of smoking conventional tobacco cigarettes, electronic cigarettes (e-cigarettes) have been proposed as an effective smoking cessation tool. However, little is known about their toxicological impacts. This is concerning given that e-cigarette use is perceived as less harmful than conventional tobacco cigarettes during pregnancy for both the mother and fetus. OBJECTIVE The goal of this study was to test the neurodevelopmental consequences of maternal e-cigarette use on adult offspring behavior and neuroimmune outcomes. METHODS Pregnant female CD-1 mice were randomly assigned to one of three treatment groups (n=8-10 per group) and exposed daily to either filtered air, propylene glycol and vegetable glycerol (50:50 PG/VG vehicle), or to PG/VG with 16mg/mL nicotine (+Nic). Whole-body exposures were carried out for 3 h/d, 7 d/week, from gestational day (GD)0.5 until GD17.5. Adult male and female offspring (8 weeks old) were assessed across a battery of behavioral assessments followed by region-specific quantification of brain cytokines using multiplex immunoassays. RESULTS Adult offspring of both sexes exposed to +Nic exhibited elevated locomotor activity in the elevated plus maze and altered stress-coping strategies in the forced swim task. Moreover, male and female offspring exposed to PG/VG with and without nicotine had a 5.2% lower object discrimination score in the novel object recognition task. In addition to differences in offspring behavior, maternal e-cigarette exposure with nicotine led to a reduction in interleukin (IL)-4 and interferon-gamma (IFNγ) in the diencephalon, as well as lower levels of hippocampal IFNγ (females only). E-cigarette exposure without nicotine resulted in a 2-fold increase of IL-6 in the cerebellum. DISCUSSION These findings support previous adverse findings of e-cigarette exposure on neurodevelopment in a mouse model and provide substantial evidence of persistent adverse behavioral and neuroimmunological consequences to adult offspring following maternal e-cigarette exposure during pregnancy. https://doi.org/10.1289/EHP6067.
Collapse
Affiliation(s)
- Jamie S. Church
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Fiona Chace-Donahue
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Jason L. Blum
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, USA
| | - Jill R. Ratner
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, USA
| | - Judith T. Zelikoff
- Department of Environmental Medicine, New York University School of Medicine, New York, New York, USA
| | - Jared J. Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, South Hadley, Massachusetts, USA
| |
Collapse
|
629
|
Salience processing by glutamatergic neurons in the ventral pallidum. Sci Bull (Beijing) 2020; 65:389-401. [PMID: 36659230 DOI: 10.1016/j.scib.2019.11.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/07/2019] [Accepted: 11/25/2019] [Indexed: 01/21/2023]
Abstract
Organisms must make sense of a constant stream of sensory inputs from both internal and external sources which compete for attention by determining which ones are salient. The ability to detect and respond appropriately to potentially salient stimuli in the environment is critical to all organisms. However, the neural circuits that process salience are not fully understood. Here, we identify a population of glutamatergic neurons in the ventral pallidum (VP) that play a unique role in salience processing. Using cell-type-specific fiber photometry, we find that VP glutamatergic neurons are robustly activated by a variety of aversion- and reward-related stimuli, as well as novel social and non-social stimuli. Inhibition of the VP glutamatergic neurons reduces the ability to detect salient stimuli in the environment, such as aversive cue, novel conspecific and novel object. Besides, VP glutamatergic neurons project to both the lateral habenula (LHb) and the ventral tegmental area (VTA). Together, our findings demonstrate that the VP glutamatergic neurons participate in salience processing and therefore provide a new perspective on treating several neuropsychiatric disorders, including dementia and psychosis.
Collapse
|
630
|
Knopp RC, Lee SH, Hollas M, Nepomuceno E, Gonzalez D, Tam K, Aamir D, Wang Y, Pierce E, BenAissa M, Thatcher GRJ. Interaction of oxidative stress and neurotrauma in ALDH2 -/- mice causes significant and persistent behavioral and pro-inflammatory effects in a tractable model of mild traumatic brain injury. Redox Biol 2020; 32:101486. [PMID: 32155582 PMCID: PMC7063127 DOI: 10.1016/j.redox.2020.101486] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/17/2020] [Accepted: 02/29/2020] [Indexed: 11/18/2022] Open
Abstract
Oxidative stress induced by lipid peroxidation products (LPP) accompanies aging and has been hypothesized to exacerbate the secondary cascade in traumatic brain injury (TBI). Increased oxidative stress is a contributor to loss of neural reserve that defines the ability to maintain healthy cognitive function despite the accumulation of neuropathology. ALDH2−/− mice are unable to clear aldehyde LPP by mitochondrial aldehyde dehydrogenase-2 (Aldh2) detoxification and provide a model to study mild TBI (mTBI), therapeutic interventions, and underlying mechanisms. The ALDH2−/− mouse model presents with elevated LPP-mediated protein modification, lowered levels of PSD-95, PGC1-α, and SOD-1, and mild cognitive deficits from 4 months of age. LPP scavengers are neuroprotective in vitro and in ALDH2−/− mice restore cognitive performance. A single-hit, closed skull mTBI failed to elicit significant effects in WT mice; however, ALDH2−/− mice showed a significant inflammatory cytokine surge in the ipsilateral hemisphere 24 h post-mTBI, and increased GFAP cleavage, a biomarker for TBI. Known neuroprotective agents, were able to reverse the effects of mTBI. This new preclinical model of mTBI, incorporating significant perturbations in behavior, inflammation, and clinically relevant biomarkers, allows mechanistic study of the interaction of LPP and neurotrauma in loss of neural reserve. ALDH2−/− mice have elevated brain LPP adducts and mild cognitive impairment. The effects of a “2nd hit” via LPS are exacerbated by LPP in vitro and in vivo. ALDH2−/− mice + mTBI show amplified/prolonged cognitive deficits and neuroinflammation. This new preclinical model for mTBI supports a role for LPP in reduced neural reserve.
Collapse
Affiliation(s)
- Rachel C Knopp
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sue H Lee
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael Hollas
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA; UICentre (Drug Discovery @ UIC), University of Illinois at Chicago, 833 S. Wood St, Chicago, IL, 60612, USA
| | - Emily Nepomuceno
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - David Gonzalez
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kevin Tam
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Daniyal Aamir
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Yueting Wang
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Emily Pierce
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Manel BenAissa
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA; UICentre (Drug Discovery @ UIC), University of Illinois at Chicago, 833 S. Wood St, Chicago, IL, 60612, USA
| | - Gregory R J Thatcher
- Department of Pharmaceutical Science, College of Pharmacy, University of Illinois at Chicago, Chicago, IL, 60612, USA; UICentre (Drug Discovery @ UIC), University of Illinois at Chicago, 833 S. Wood St, Chicago, IL, 60612, USA.
| |
Collapse
|
631
|
Wang W, Kiyoshi CM, Du Y, Taylor AT, Sheehan ER, Wu X, Zhou M. TREK-1 Null Impairs Neuronal Excitability, Synaptic Plasticity, and Cognitive Function. Mol Neurobiol 2020; 57:1332-1346. [PMID: 31728930 PMCID: PMC8808335 DOI: 10.1007/s12035-019-01828-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023]
Abstract
TREK-1, a two-pore-domain K+ channel, is highly expressed in the central nervous system. Although aberrant expression of TREK-1 is implicated in cognitive impairment, the cellular and functional mechanism underlying this channelopathy is poorly understood. Here we examined TREK-1 contribution to neuronal morphology, excitability, synaptic plasticity, and cognitive function in mice deficient in TREK-1 expression. TREK-1 immunostaining signal mainly appeared in hippocampal pyramidal neurons, but not in astrocytes. TREK-1 gene knockout (TREK-1 KO) increases dendritic sprouting and the number of immature spines in hippocampal CA1 pyramidal neurons. Functionally, TREK-1 KO increases neuronal excitability and enhances excitatory and inhibitory postsynaptic currents (EPSCs and IPSCs). The increased EPSCs appear to be attributed to an increased release probability of presynaptic glutamate and functional expression of postsynaptic AMPA receptors. TREK-1 KO decreased the paired-pulse ratio and severely occluded the long-term potentiation (LTP) in the CA1 region. These altered synaptic transmission and plasticity are associated with recognition memory deficit in TREK-1 KO mice. Although astrocytic expression of TREK-1 has been reported in previous studies, TREK-1 KO does not alter astrocyte membrane K+ conductance or the syncytial network function in terms of syncytial isopotentiality. Altogether, TREK-1 KO profoundly affects the cellular structure and function of hippocampal pyramidal neurons. Thus, the impaired cognitive function in diseases associated with aberrant expression of TREK-1 should be attributed to the failure of this K+ channel in regulating neuronal morphology, excitability, synaptic transmission, and plasticity.
Collapse
Affiliation(s)
- Wei Wang
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Conrad M Kiyoshi
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yixing Du
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anne T Taylor
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Erica R Sheehan
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Xiao Wu
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Min Zhou
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
632
|
Long JY, Chen JM, Liao YJ, Zhou YJ, Liang BY, Zhou Y. Naringin provides neuroprotection in CCL2-induced cognition impairment by attenuating neuronal apoptosis in the hippocampus. Behav Brain Funct 2020; 16:4. [PMID: 32103758 PMCID: PMC7045422 DOI: 10.1186/s12993-020-00166-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/18/2020] [Indexed: 01/21/2023] Open
Abstract
Background Chemokine C–C motif ligand 2 (CCL2) is one of the most widely recognised proinflammatory chemokines in cognitive disorders. Currently, CCL2-targeting drugs are extremely limited. Thus, this study aimed to explore the neuroprotection afforded by naringin in CCL2-induced cognitive impairment in rats. Methods Before the CCL2 intra-hippocampal injection, rats were treated with naringin for 3 consecutive days via intraperitoneal injection. Two days post-surgery, the Morris water maze (MWM) and novel object recognition (NORT) tests were performed to detect spatial learning and memory and object cognition, respectively. Nissl staining and dUTP nick-end labelling (TUNEL) staining were performed to assess histopathological changes in the hippocampus. Commercial kits were used to measure the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) and the content of malondialdehyde (MDA). Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to examine the relative mRNA expression of interleukin 1β, (IL-1β), interleukin 6 (IL-6), glutamate/aspartate transporter (GLAST), glutamate transporter-1 (GLT-1), phosphate-activated glutaminase (PAG), cysteine aspartic acid-specific protease 8 (caspase-8), cysteine aspartic acid-specific protease 3 (caspase-3), cell lymphoma/leukaemia-2 (Bcl-2), and Bcl-2 associated X protein (Bax). Results In the MWM, the average escape latency and average swimming distance were significantly reduced and the crossing times were increased in the naringin-treated groups, compared with the CCL2 group. The NORT results revealed that, compared with the CCL2 rats, the discrimination index in the naringin-treated rats increased significantly. Nissl and TUNEL staining revealed that naringin protected the structure and survival of the neurons in the CA1 zone of the hippocampus. In the naringin-treated groups, the SOD and GSH-Px activities were increased, whereas the MDA levels were decreased. Furthermore, in the naringin-treated groups, the relative mRNA expression of IL-1β and IL-6 was significantly decreased; GLAST and GLT-1 mRNA expression levels were increased, whereas PAG was decreased. In the naringin-treated groups, the relative mRNA expression levels of caspase-8, caspase-3, and Bax were decreased, whereas that of Bcl-2 was increased. Conclusion Collectively, these data indicated that naringin alleviated the CCL2-induced cognitive impairment. The underlying mechanisms could be associated with the inhibition of neuroinflammation, oxidative stress, apoptosis, and the regulation of glutamate metabolism.
Collapse
Affiliation(s)
- Jiang-Yi Long
- Department of Pharmacology, Guangxi Medical University, Nanning, 53002, Guangxi, China
| | - Jian-Min Chen
- Department of Pharmacology, Guangxi Medical University, Nanning, 53002, Guangxi, China
| | - Yuan-Jun Liao
- Department of Pharmacology, Guangxi Medical University, Nanning, 53002, Guangxi, China
| | - Yi-Jun Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, 53002, Guangxi, China
| | - Bing-Yu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, 530021, Guang, China
| | - Yan Zhou
- Department of Pharmacology, Guangxi Medical University, Nanning, 53002, Guangxi, China.
| |
Collapse
|
633
|
Li Y, Bao H, Luo Y, Yoan C, Sullivan HA, Quintanilla L, Wickersham I, Lazarus M, Shih YYI, Song J. Supramammillary nucleus synchronizes with dentate gyrus to regulate spatial memory retrieval through glutamate release. eLife 2020; 9:e53129. [PMID: 32167473 PMCID: PMC7069722 DOI: 10.7554/elife.53129] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 02/24/2020] [Indexed: 12/29/2022] Open
Abstract
The supramammillary nucleus (SuM) provides substantial innervation to the dentate gyrus (DG). It remains unknown how the SuM and DG coordinate their activities at the circuit level to regulate spatial memory. Additionally, SuM co-releases GABA and glutamate to the DG, but the relative role of GABA versus glutamate in regulating spatial memory remains unknown. Here we report that SuM-DG Ca2+ activities are highly correlated during spatial memory retrieval as compared to the moderate correlation during memory encoding when mice are performing a location discrimination task. Supporting this evidence, we demonstrate that the activity of SuM neurons or SuM-DG projections is required for spatial memory retrieval. Furthermore, we show that SuM glutamate transmission is necessary for both spatial memory retrieval and highly-correlated SuM-DG activities during spatial memory retrieval. Our studies identify a long-range SuM-DG circuit linking two highly correlated subcortical regions to regulate spatial memory retrieval through SuM glutamate release.
Collapse
Affiliation(s)
- Yadong Li
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Neuroscience Center, University of North CarolinaChapel HillUnited States
| | - Hechen Bao
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Neuroscience Center, University of North CarolinaChapel HillUnited States
| | - Yanjia Luo
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Neuroscience Center, University of North CarolinaChapel HillUnited States
| | - Cherasse Yoan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of TsukubaTsukubaJapan
| | - Heather Anne Sullivan
- The McGovern Institute for Brain Research, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Luis Quintanilla
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Neuroscience Center, University of North CarolinaChapel HillUnited States
- Neurobiology Curriculum, University of North CarolinaChapel HillUnited States
| | - Ian Wickersham
- The McGovern Institute for Brain Research, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Michael Lazarus
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of TsukubaTsukubaJapan
| | - Yen-Yu Ian Shih
- Department of Neurology and Biomedical Research Imaging Center, University of North CarolinaChapel HillUnited States
| | - Juan Song
- Department of Pharmacology, University of North CarolinaChapel HillUnited States
- Neuroscience Center, University of North CarolinaChapel HillUnited States
| |
Collapse
|
634
|
Shin S, Kim D, Song JY, Jeong H, Hyeon SJ, Kowall NW, Ryu H, Pae AN, Lim S, Kim YK. Visualization of soluble tau oligomers in TauP301L-BiFC transgenic mice demonstrates the progression of tauopathy. Prog Neurobiol 2020; 187:101782. [PMID: 32105751 DOI: 10.1016/j.pneurobio.2020.101782] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/16/2020] [Accepted: 02/21/2020] [Indexed: 11/30/2022]
Abstract
Accumulation of abnormal tau aggregates in the brain is a pathological hallmark of multiple neurodegenerative disorders including Alzheimer's disease. Increasing evidence suggests that soluble tau aggregates play a key role in tau pathology as neurotoxic species causing neuronal cell death and act as prion-like seeds mediating tau propagation. Despite the pathological relevance, there is a paucity of methods to monitor tau oligomerization in the brain. As a tool to monitor tau self-assembly in the brain, we generated a novel tau transgenic mouse, named TauP301L-BiFC. By introducing bimolecular fluorescence complementation technique to human tau containing a P301L mutation, we were able to monitor and quantify tau self-assembly, represented by BiFC fluorescence in the brains of transgenic TauP301L-BiFC mice. TauP301L-BiFC mice showed soluble tau oligomerization from 3 months, showing significantly enriched BiFC fluorescence in the brain. Then, massive tau fragmentation occured at 6 months showing dramatically decreased TauP301L-BiFC fluorescence. The fragmented tau species served as a seed for insoluble tau aggregation. In a result, insoluble TauP301L-BiFC aggregates coaggregated with endogenous mouse tau accumulated in the brain, showing subsequently increased BiFC fluorescence from 9 months. Neuronal degeneration and cognitive deficits were observed from 12 months of age. TauP301L-BiFC mouse model demonstrated that methylene blue reduced the amount of soluble tau oligomers in the brain, resulting in the prevention of cognitive impairments. We assure that TauP301L-BiFC mice are a bona-fide animal tool to monitor pathological tau oligomerization in AD and other tauopathies.
Collapse
Affiliation(s)
- Seulgi Shin
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Dohee Kim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Ji Yeon Song
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hyeanjeong Jeong
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Department of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Seung Jae Hyeon
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Neil W Kowall
- Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Hoon Ryu
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Ae Nim Pae
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Sungsu Lim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Yun Kyung Kim
- Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
635
|
Abstract
Postoperative cognitive dysfunction (POCD) is a major complication affecting patients of any age undergoing surgery. This syndrome impacts everyday life up to months after hospital discharge, and its pathophysiology still remains unclear. Translational research focusing on POCD is based on a wide variety of rodent models, such as the murine tibial fracture, whose severity can limit mouse locomotion and proper behavioral assessment. Besides, influence of skeletal muscle injury, a lesion encountered in a wide range of surgeries, has not been explored in POCD occurrence. We propose a physical model of muscle injury in CX3CR1GFP/+ mice (displaying green fluorescent microglial cells) to study POCD, with morphological, behavioral and molecular approaches. We highlighted: alteration of short- and long-term memory after muscle regeneration, wide microglial reactivity in the brain, including hippocampus area, 24 hours after muscle injury, and an alteration of central brain derived neurotrophic factor (BDNF) and nerve growth factor (NGF) balance, 28 days after muscle injury. Our results suggest for the first time that muscle injury can have early as well as late impacts on the brain. Our CX3CR1GFP/+ model can also facilitate microglial investigation, more specifically their pivotal role in neuroinflammation and synaptic plasticity, in the pathophysiology of POCD.
Collapse
|
636
|
Meng B, Li X, Lu B, Liu R, Yuan H, Zhai X, Qin J, Chen Z, Zheng J, Chen J. The Investigation of Hippocampus-Dependent Cognitive Decline Induced by Anesthesia/Surgery in Mice Through Integrated Behavioral Z-Scoring. Front Behav Neurosci 2020; 13:282. [PMID: 32038186 PMCID: PMC6987045 DOI: 10.3389/fnbeh.2019.00282] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 12/11/2019] [Indexed: 01/11/2023] Open
Abstract
Objective Patients undergoing major surgeries may experience certain cognitive decline, which is known as postoperative delirium (POD) or postoperative cognitive dysfunction (POCD). We employed integrated behavioral Z-scoring introduced by Guilloux et al. (2011) to investigate the effects of fracture fixation under anesthesia on hippocampus-dependent memory in mice. Methods ICR mice (12-14 months) underwent stabilized tibial fracture operation under sevoflurane anesthesia. They were subjected to a battery of successive hippocampus-dependent tests following surgery, including open field test (OF), novel object recognition (NOR), fear conditioning test (FC), and Morris water maze (MWM). The integrated behavioral Z-scoring was applied to assess the hippocampus-dependent memory after anesthesia/surgery, and the association between the integrated behavioral Z-scores and hippocampal pro-inflammatory cytokines was explored. Results Mice after anesthesia/surgery were found to have impaired hippocampus-dependent memory in NOR, FC, and MWM but with different degrees in these aspects as represented by P-value and effect size. The integrated memory Z-scores based on principal parameters of the above three tests can reduced the variability and increase the comprehensiveness of behavioral results. However, we found no statistic associations between hippocampal pro-inflammatory cytokines and the integrated Z-scores, as the elevated cytokines quickly return to normal on postoperative day 3 and/or day 7. Conclusion The integrated Z-score methodology could facilitate the interpretation of the anesthesia/surgery induced cognitive decline in mice and robustly quantify the behavioral phenotyping of hippocampus-dependent memory.
Collapse
Affiliation(s)
- Bo Meng
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiaoyu Li
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Bo Lu
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Rongjun Liu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Hui Yuan
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiaojie Zhai
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jinling Qin
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Zhang Chen
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Jinwei Zheng
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Junping Chen
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
637
|
Rochais C, Lecoutey C, Hamidouche K, Giannoni P, Gaven F, Cem E, Mignani S, Baranger K, Freret T, Bockaert J, Rivera S, Boulouard M, Dallemagne P, Claeysen S. Donecopride, a Swiss army knife with potential against Alzheimer's disease. Br J Pharmacol 2020; 177:1988-2005. [PMID: 31881553 DOI: 10.1111/bph.14964] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/21/2019] [Accepted: 11/28/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE We recently identified donecopride as a pleiotropic compound able to inhibit AChE and to activate 5-HT4 receptors. Here, we have assessed the potential therapeutic effects of donecopride in treating Alzheimer's disease (AD). EXPERIMENTAL APPROACH We used two in vivo animal models of AD, transgenic 5XFAD mice and mice exposed to soluble amyloid-β peptides and, in vitro, primary cultures of rat hippocampal neurons. Pro-cognitive and anti-amnesic effects were evaluated with novel object recognition, Y-maze, and Morris water maze tests. Amyloid load in mouse brain was measured ex vivo and effects of soluble amyloid-β peptides on neuronal survival and neurite formation determined in vitro. KEY RESULTS In vivo, chronic (3 months) administration of donecopride displayed potent anti-amnesic properties in the two mouse models of AD, preserving learning capacities, including working and long-term spatial memories. These behavioural effects were accompanied by decreased amyloid aggregation in the brain of 5XFAD mice and, in cultures of rat hippocampal neurons, reduced tau hyperphosphorylation. In vitro, donecopride increased survival in neuronal cultures exposed to soluble amyloid-β peptides, improved the neurite network and provided neurotrophic benefits, expressed as the formation of new synapses. CONCLUSIONS AND IMPLICATIONS Donecopride acts like a Swiss army knife, exhibiting a range of sustainable symptomatic therapeutic effects and potential disease-modifying effects in models of AD. Clinical trials with this promising drug candidate will soon be undertaken to confirm its therapeutic potential in humans.
Collapse
Affiliation(s)
- Christophe Rochais
- Normandie Univ, Caen, France.,UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), Caen, France
| | - Cédric Lecoutey
- Normandie Univ, Caen, France.,UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), Caen, France
| | - Katia Hamidouche
- Normandie Univ, Caen, France.,UNICAEN, INSERM U1075 COMETE, Caen, France
| | - Patrizia Giannoni
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France.,Equipe Chrome, EA7352, Université de Nîmes, Nîmes, France
| | - Florence Gaven
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France.,CRBM, CNRS UMR5237, Montpellier, France
| | - Eleazere Cem
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Serge Mignani
- Normandie Univ, Caen, France.,UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), Caen, France
| | - Kevin Baranger
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Thomas Freret
- Normandie Univ, Caen, France.,UNICAEN, INSERM U1075 COMETE, Caen, France
| | - Joël Bockaert
- IGF, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | - Santiago Rivera
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Michel Boulouard
- Normandie Univ, Caen, France.,UNICAEN, INSERM U1075 COMETE, Caen, France
| | - Patrick Dallemagne
- Normandie Univ, Caen, France.,UNICAEN, CERMN (Centre d'Etudes et de Recherche sur le Médicament de Normandie), Caen, France
| | | |
Collapse
|
638
|
Wang J, Song HR, Guo MN, Ma SF, Yun Q, Zhang WN. Adult conditional knockout of PGC-1α in GABAergic neurons causes exaggerated startle reactivity, impaired short-term habituation and hyperactivity. Brain Res Bull 2020; 157:128-139. [PMID: 32057952 DOI: 10.1016/j.brainresbull.2020.02.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/05/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023]
Abstract
Interneurons not only contribute to the global balance of activity in cortical networks but also mediate the precise gating of information through specific proteins. Accumulating evidence demonstrates that peroxisome-proliferator-activated receptor-gamma co-activator 1 alpha (PGC-1α) is concentrated in inhibitory interneurons and that it plays an important role in neuropsychiatric diseases. However, the functions of the transcriptional coactivator PGC-1α in sensorimotor gating, short-term habituation and spatial reference memory are still not entirely clear. To investigate the precise involvement of PGC-1α in the progression of psychiatric disorders, we first generated PGC-1α conditional knockout mice through transgenic expression of Cre recombinase under the control of dlx5/6 promoter, Cre-mediated excision events occurred specifically in γ-amino-butyric-acid-(GABA)ergic neurons. Short-term habituation and spatial reference memory in Dlx5/6-Cre::PGC-1αfl/fl mice were evaluated using the novel object recognition test and the Morris water maze test, and sensorimotor gating was measured by prepulse inhibition of the acoustic startle reflex. Protein expression of parvalbumin (PV) in specific brain regions was studied by western blotting, immunofluorescence and immunohistochemistry. Here, we show that mice lacking the PGC-1α gene in GABAergic neurons exhibit deficits in short-term habituation, hyperactivity, reduced prepulse inhibition and exaggerated startle reactivity but normal associative spatial reference memory. In particular, these mice display aberrant salience, whereby more attention is paid to a further copy of the original object (now familiar) (relative to the first presentation of the original object, and relative to the presentation of the novel object). These behavioral dysfunctions were associated with decreased PV expression in the cortex (including somatosensory and motor cortex) as well as in the hippocampus, especially in its CA1 and CA3 regions. Together, these findings draw attention to a hyper-response phenotype of PGC-1α conditional knockout mice and indicate that PGC-1α is a novel regulator of gene expression and function in PV-positive interneurons and a potential therapeutic target for psychiatric disorders associated with PGC-1α dysregulation.
Collapse
Affiliation(s)
- Jia Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China.
| | - Huang-Rong Song
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China
| | - Mei-Na Guo
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China
| | - Si-Fei Ma
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China
| | - Qi Yun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China
| | - Wei-Ning Zhang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu Province 212013, PR China.
| |
Collapse
|
639
|
Wlodarek L, Cao F, Alibhai FJ, Fekete A, Noyan N, Tobin SW, Marvasti TB, Wu J, Li SH, Weisel RD, Wang LY, Jia Z, Li RK. Rectification of radiotherapy-induced cognitive impairments in aged mice by reconstituted Sca-1 + stem cells from young donors. J Neuroinflammation 2020; 17:51. [PMID: 32028989 PMCID: PMC7006105 DOI: 10.1186/s12974-019-1681-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/17/2019] [Indexed: 01/03/2023] Open
Abstract
Background Radiotherapy is widely used and effective for treating brain tumours, but inevitably impairs cognition as it arrests cellular processes important for learning and memory. This is particularly evident in the aged brain with limited regenerative capacity, where radiation produces irreparable neuronal damage and activation of neighbouring microglia. The latter is responsible for increased neuronal death and contributes to cognitive decline after treatment. To date, there are few effective means to prevent cognitive deficits after radiotherapy. Methods Here we implanted hematopoietic stem cells (HSCs) from young or old (2- or 18-month-old, respectively) donor mice expressing green fluorescent protein (GFP) into old recipients and assessed cognitive abilities 3 months post-reconstitution. Results Regardless of donor age, GFP+ cells homed to the brain of old recipients and expressed the macrophage/microglial marker, Iba1. However, only young cells attenuated deficits in novel object recognition and spatial memory and learning in old mice post-irradiation. Mechanistically, old recipients that received young HSCs, but not old, displayed significantly greater dendritic spine density and long-term potentiation (LTP) in CA1 neurons of the hippocampus. Lastly, we found that GFP+/Iba1+ cells from young and old donors were differentially polarized to an anti- and pro-inflammatory phenotype and produced neuroprotective factors and reactive nitrogen species in vivo, respectively. Conclusion Our results suggest aged peripherally derived microglia-like cells may exacerbate cognitive impairments after radiotherapy, whereas young microglia-like cells are polarized to a reparative phenotype in the irradiated brain, particularly in neural circuits associated with rewards, learning, and memory. These findings present a proof-of-principle for effectively reinstating central cognitive function of irradiated brains with peripheral stem cells from young donor bone marrow. Electronic supplementary material The online version of this article (10.1186/s12974-019-1681-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lukasz Wlodarek
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Feng Cao
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Faisal J Alibhai
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Adam Fekete
- Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada
| | - Nima Noyan
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Stephanie W Tobin
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Tina B Marvasti
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Jun Wu
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Shu-Hong Li
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada
| | - Richard D Weisel
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada.,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada
| | - Lu-Yang Wang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | - Zhengping Jia
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Program in Neurosciences & Mental Health, SickKids Research Institute, Floor 5, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | - Ren-Ke Li
- Toronto General Hospital Research Institute, University Health Network, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, M5G 1L7, Canada. .,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada. .,Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Department of Surgery, Division of Cardiac Surgery, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
640
|
Wang T, Niu K, Fan A, Bi N, Tao H, Chen XT, Wang HL. Dietary intake of polyunsaturated fatty acids alleviates cognition deficits and depression-like behaviour via cannabinoid system in sleep deprivation rats. Behav Brain Res 2020; 384:112545. [PMID: 32035867 DOI: 10.1016/j.bbr.2020.112545] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/02/2020] [Accepted: 02/05/2020] [Indexed: 01/01/2023]
Abstract
Sleep deprivation (SD) is a common feature in modern society. Prolonged sleep deprivation causes cognition deficits and depression-like behavior in the model of animal experiments. Endocannabinoid system are key modulators of synaptic function, which were related to memory and mood. Although the underlying mechanism remains unknown, several studies indicated the benefits of polyunsaturated fatty acids (PUFAs, linolenic acid, 39.7 %; linoleic acid, 28 %; and oleic acid, 22 %) on brain function through the endocannabinoid system. The present study aimed to evaluate the influence of dietary PUFAs on cognition deficits induced by sleep deprivation in Sprague Dawley rats. The rats were sleep deprivation continuously for 7 days and fed with PUFAs at three different dosages (2, 4 and 8 μl/g body weight) at the meantime. The effect of PUFAs on cognition was investigated by object recognition test while depressive-like behavior were detected using sucrose preference test and forced swim test. The mechanism of PUFAs was elucidated by hippocampal synaptic transmission analyses. The resluts revealed that SD led to the disorder of cognition and mood which was improved by the supplement of PUFAs. SD significantly increased the mEPSC frequency, and decreased the protein level of cannabinoid type-1 receptors (CB1R). These changes were restored by supplement of PUFAs, which showed a similar level to the control group. Behaviour tests showed that the positive effects on repairing cognition and anxiety disorders were almost completely abolished when the CB1R receptor antagonist rimonabant was applied to the SD rats. These findings indicated that PUFAs are a factor regulating cognition deficits and depression induced by SD via cannabinoid type-1 receptors.
Collapse
Affiliation(s)
- Tiandong Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Kang Niu
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Anni Fan
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Nanxi Bi
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Han Tao
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China
| | - Xiang-Tao Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230031, PR China.
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, 193 Tunxi Road, Hefei, Anhui 230009, PR China; School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, PR China.
| |
Collapse
|
641
|
Bellantuono I, de Cabo R, Ehninger D, Di Germanio C, Lawrie A, Miller J, Mitchell SJ, Navas-Enamorado I, Potter PK, Tchkonia T, Trejo JL, Lamming DW. A toolbox for the longitudinal assessment of healthspan in aging mice. Nat Protoc 2020; 15:540-574. [PMID: 31915391 PMCID: PMC7002283 DOI: 10.1038/s41596-019-0256-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022]
Abstract
The number of people aged over 65 is expected to double in the next 30 years. For many, living longer will mean spending more years with the burdens of chronic diseases such as Alzheimer's disease, cardiovascular disease, and diabetes. Although researchers have made rapid progress in developing geroprotective interventions that target mechanisms of aging and delay or prevent the onset of multiple concurrent age-related diseases, a lack of standardized techniques to assess healthspan in preclinical murine studies has resulted in reduced reproducibility and slow progress. To overcome this, major centers in Europe and the United States skilled in healthspan analysis came together to agree on a toolbox of techniques that can be used to consistently assess the healthspan of mice. Here, we describe the agreed toolbox, which contains protocols for echocardiography, novel object recognition, grip strength, rotarod, glucose tolerance test (GTT) and insulin tolerance test (ITT), body composition, and energy expenditure. The protocols can be performed longitudinally in the same mouse over a period of 4-6 weeks to test how candidate geroprotectors affect cardiac, cognitive, neuromuscular, and metabolic health.
Collapse
Affiliation(s)
- I Bellantuono
- Department of Oncology and Metabolism, Healthy Lifespan Institute and MRC-Arthritis Research UK Centre for Integrated research into Musculoskeletal Ageing, University of Sheffield, Sheffield, UK.
| | - R de Cabo
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - D Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1, Bonn, Germany
| | - C Di Germanio
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - A Lawrie
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - J Miller
- Robert and Arlene KogodCenter on Aging, Mayo Clinic, Rochester, MN, USA
| | - S J Mitchell
- Department of Molecular Medicine, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - I Navas-Enamorado
- Translational Gerontology Branch, National Institutes of Health, Baltimore, MD, USA
| | - P K Potter
- Department of Biological and Life Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Oxfordshire, UK
| | - T Tchkonia
- Robert and Arlene KogodCenter on Aging, Mayo Clinic, Rochester, MN, USA
| | - J L Trejo
- Department of Translational Neuroscience, Cajal Institute (CSIC), Madrid, Spain
| | - D W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
642
|
Moura CA, Oliveira MC, Costa LF, Tiago PRF, Holanda VAD, Lima RH, Cagni FC, Lobão-Soares B, Bolaños-Jiménez F, Gavioli EC. Prenatal restraint stress impairs recognition memory in adult male and female offspring. Acta Neuropsychiatr 2020; 32:1-6. [PMID: 31992385 DOI: 10.1017/neu.2020.3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Accumulating evidence from preclinical and clinical studies indicates that prenatal exposure to stress impairs the development of the offspring brain and facilitates the emergence of mental illness. This study aims to describe the impact of prenatal restraint stress on cognition and exploration to an unfamiliar environment at adulthood in an outbred strain of mice. METHODS Late pregnant mice were exposed to restraint stress and adult offspring (60 days of age) behaviours were assessed in the object recognition task and open field test. FINDINGS Prenatal stress (PNS) impaired new object recognition in male and female mice. Importantly, the learning deficits in female PNS mice were linked to their estrous cycle. Actually, PNS females in metestrus/diestrus but not in proestrus/estrus phases displayed recognition deficits compared to controls. Concerning locomotion in an unfamiliar environment, male but not female PNS mice displayed significant increase, but showed no differences in the distance travelled within the centre zone of the arena. CONCLUSION Present findings support the view that maternal restraint-stress during late pregnancy impairs recognition memory in both male and female offspring, and in females, this cognitive deficit is dependent on the estrous cycle phase. Ultimately, these data reinforce that PNS is an aetiological component of psychiatric disorders associated with memory deficits.
Collapse
Affiliation(s)
- Clarissa A Moura
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Matheus C Oliveira
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Layse F Costa
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Pamella R F Tiago
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Victor A D Holanda
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Ramon H Lima
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Fernanda C Cagni
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Bruno Lobão-Soares
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Elaine C Gavioli
- Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
643
|
Zhao J, Fok AHK, Fan R, Kwan PY, Chan HL, Lo LHY, Chan YS, Yung WH, Huang J, Lai CSW, Lai KO. Specific depletion of the motor protein KIF5B leads to deficits in dendritic transport, synaptic plasticity and memory. eLife 2020; 9:53456. [PMID: 31961321 PMCID: PMC7028368 DOI: 10.7554/elife.53456] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
The kinesin I family of motor proteins are crucial for axonal transport, but their roles in dendritic transport and postsynaptic function are not well-defined. Gene duplication and subsequent diversification give rise to three homologous kinesin I proteins (KIF5A, KIF5B and KIF5C) in vertebrates, but it is not clear whether and how they exhibit functional specificity. Here we show that knockdown of KIF5A or KIF5B differentially affects excitatory synapses and dendritic transport in hippocampal neurons. The functional specificities of the two kinesins are determined by their diverse carboxyl-termini, where arginine methylation occurs in KIF5B and regulates its function. KIF5B conditional knockout mice exhibit deficits in dendritic spine morphogenesis, synaptic plasticity and memory formation. Our findings provide insights into how expansion of the kinesin I family during evolution leads to diversification and specialization of motor proteins in regulating postsynaptic function. Transporting molecules within a cell becomes a daunting task when the cell is a neuron, with fibers called axons and dendrites that can stretch as long as a meter. Neurons use many different molecules to send messages across the body and store memories in the brain. If the right molecules cannot be delivered along the length of nerve cells, connections to neighboring neurons may decay, which may impair learning and memory. Motor proteins are responsible for transporting molecules within cells. Kinesins are a type of motor protein that typically transports materials from the body of a neuron to the cell’s periphery, including the dendrites, which is where a neuron receives messages from other nerve cells. Each cell has up to 45 different kinesin motors, but it is not known whether each one performs a distinct task or if they have overlapping roles. Now, Zhao, Fok et al. have studied two similar kinesins, called KIF5A and KIF5B, in rodent neurons to determine their roles. First, it was shown that both proteins were found at dendritic spines, which are small outgrowths on dendrites where contact with other cells occurs. Next, KIF5A and KIF5B were depleted, one at a time, from neurons extracted from a brain region called the hippocampus. Removing KIF5B interfered with the formation of dendritic spines, but removing KIF5A did not have an effect. Dendritic spines are essential for learning and memory, so several behavioral tests were conducted on mice that had been genetically modified to express less KIF5B in the forebrain. These tests revealed that the mice performed poorly in tasks that tested their memory recall. This work opens a new area of research studying the specific roles of different kinesin motor proteins in nerve cells. This could have important implications because certain kinesin motor proteins such as KIF5A are known to be defective in some inherited neurodegenerative diseases.
Collapse
Affiliation(s)
- Junjun Zhao
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Albert Hiu Ka Fok
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Ruolin Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Pui-Yi Kwan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Hei-Lok Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Louisa Hoi-Ying Lo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Ying-Shing Chan
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Wing-Ho Yung
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jiandong Huang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.,Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Cora Sau Wan Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
644
|
Boillat M, Hammoudi PM, Dogga SK, Pagès S, Goubran M, Rodriguez I, Soldati-Favre D. Neuroinflammation-Associated Aspecific Manipulation of Mouse Predator Fear by Toxoplasma gondii. Cell Rep 2020; 30:320-334.e6. [PMID: 31940479 PMCID: PMC6963786 DOI: 10.1016/j.celrep.2019.12.019] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 10/27/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022] Open
Abstract
In rodents, the decrease of felid aversion induced by Toxoplasma gondii, a phenomenon termed fatal attraction, is interpreted as an adaptive manipulation by the neurotropic protozoan parasite. With the aim of understanding how the parasite induces such specific behavioral modifications, we performed a multiparametric analysis of T. gondii-induced changes on host behavior, physiology, and brain transcriptome as well as parasite cyst load and distribution. Using a set of complementary behavioral tests, we provide strong evidence that T. gondii lowers general anxiety in infected mice, increases explorative behaviors, and surprisingly alters predator aversion without selectivity toward felids. Furthermore, we show a positive correlation between the severity of the behavioral alterations and the cyst load, which indirectly reflects the level of inflammation during brain colonization. Taken together, these findings refute the myth of a selective loss of cat fear in T. gondii-infected mice and point toward widespread immune-related alterations of behaviors.
Collapse
Affiliation(s)
- Madlaina Boillat
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland
| | - Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland
| | - Stéphane Pagès
- Wyss Center for Bio- and Neuroengineering, Geneva, Switzerland; Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Maged Goubran
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada
| | - Ivan Rodriguez
- Department of Genetics and Evolution, Faculty of Sciences, University of Geneva, 1211 Geneva, Switzerland.
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva CMU, 1 rue Michel-Servet 1211 Geneva 4, Switzerland.
| |
Collapse
|
645
|
Won S, Park K, Lim H, Lee SJ. Sexual dimorphism in cognitive disorders in a murine model of neuropathic pain. Behav Brain Funct 2020; 16:1. [PMID: 31901234 PMCID: PMC6942364 DOI: 10.1186/s12993-019-0164-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/18/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A sex-difference in susceptibility to chronic pain is well-known. Although recent studies have begun to reveal the sex-dependent mechanisms of nerve injury-induced pain sensitization, sex differences in the affective and cognitive brain dysfunctions associated with chronic pain have not been investigated. Therefore, we tested whether chronic pain leads to affective and cognitive disorders in a mouse neuropathic pain model and whether those disorders are sexually dimorphic. METHODS Chronic neuropathic pain was induced in male and female mice by L5 spinal nerve transection (SNT) injury. Pain sensitivity was measured with the von Frey test. Affective behaviors such as depression and anxiety were assessed by the forced swim, tail suspension, and open field tests. Cognitive brain function was assessed with the Morris water maze and the novel object location and novel object recognition tests. RESULTS Mechanical allodynia was induced and maintained for up to 8 weeks after SNT in both male and female mice. Depressive- and anxiety-like behaviors were observed 8 weeks post-SNT injury regardless of sex. Chronic pain-induced cognitive deficits measured with the Morris water maze and novel object location test were seen only in male mice, not in female mice. CONCLUSIONS Chronic neuropathic pain is accompanied by anxiety- and depressive-like behaviors in a mouse model regardless of sex, and male mice are more vulnerable than female mice to chronic pain-associated cognitive deficits.
Collapse
Affiliation(s)
- Soonmi Won
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Keebum Park
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea.,Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hyoungsub Lim
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea
| | - Sung Joong Lee
- Department of Neuroscience and Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, Republic of Korea. .,Interdisciplinary Program in Neuroscience, College of Natural Science, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
646
|
Ali M, Zahid S. The neurogenic effects of rosmarinic acid in a mouse model of type 2 diabetes mellitus. BRAZ J PHARM SCI 2020. [DOI: 10.1590/s2175-979020200003180772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Mahnoor Ali
- National University of Sciences and Technology, Pakistan
| | - Saadia Zahid
- National University of Sciences and Technology, Pakistan
| |
Collapse
|
647
|
Chen MX, Liu Q, Cheng S, Lei L, Lin AJ, Wei R, K Hui TC, Li Q, Ao LJ, Sham PC. Interleukin-18 levels in the hippocampus and behavior of adult rat offspring exposed to prenatal restraint stress during early and late pregnancy. Neural Regen Res 2020; 15:1748-1756. [PMID: 32209782 PMCID: PMC7437598 DOI: 10.4103/1673-5374.276358] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Exposure to maternal stress during prenatal life is associated with an increased risk of neuropsychiatric disorders, such as depression and anxiety, in offspring. It has also been increasingly observed that prenatal stress alters the phenotype of offspring via immunological mechanisms and that immunological dysfunction, such as elevated interleukin-18 levels, has been reported in cultures of microglia. Prenatal restraint stress (PRS) in rats permits direct experimental investigation of the link between prenatal stress and adverse outcomes. However, the majority of studies have focused on the consequences of PRS delivered in the second half of pregnancy, while the effects of early prenatal stress have rarely been examined. Therefore, pregnant rats were subjected to PRS during early/middle and late gestation (days 8-14 and 15-21, respectively). PRS comprised restraint in a round plastic transparent cylinder under bright light (6500 lx) three times per day for 45 minutes. Differences in interleukin-18 expression in the hippocampus and in behavior were compared between offspring rats and control rats on postnatal day 75. We found that adult male offspring exposed to PRS during their late prenatal periods had higher levels of anxiety-related behavior and depression than control rats, and both male and female offspring exhibited higher levels of depression-related behavior, impaired recognition memory and diminished exploration of novel objects. Moreover, an elevated level of interleukin-18 was observed in the dorsal and ventral hippocampus of male and female early- and late-PRS offspring rats. The results indicate that PRS can cause anxiety and depression-related behaviors in adult offspring and affect the expression of interleukin-18 in the hippocampus. Thus, behavior and the molecular biology of the brain are affected by the timing of PRS exposure and the sex of the offspring. All experiments were approved by the Animal Experimentation Ethics Committee at Kunming Medical University, China (approval No. KMMU2019074) in January 2019.
Collapse
Affiliation(s)
- Mo-Xian Chen
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Qiang Liu
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shu Cheng
- Department of Rehabilitation, China Resources & WISCO General Hospital, Wuhan, Hubei Province, China
| | - Lei Lei
- Department of Rehabilitation Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ai-Jin Lin
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Ran Wei
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Tomy C K Hui
- Department of Psychiatry, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qi Li
- Department of Psychiatry; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Li-Juan Ao
- School of Rehabilitation, Kunming Medical University, Kunming, Yunnan Province, China
| | - Pak C Sham
- Department of Psychiatry; State Key Laboratory of Brain and Cognitive Sciences; Centre for Genomic Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
648
|
Jin Y, Cai S, Jiang Y, Zhong K, Wen C, Ruan Y, Chew LA, Khanna R, Xu Z, Yu J. Tetramethylpyrazine Reduces Epileptogenesis Progression in Electrical Kindling Models by Modulating Hippocampal Excitatory Neurotransmission. ACS Chem Neurosci 2019; 10:4854-4863. [PMID: 31756074 DOI: 10.1021/acschemneuro.9b00575] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Antiepileptic drugs (AEDs) are the primary agents prescribed for clinical management of limbic epilepsy. However, high incidence of pharmacoresistance and a limited armory of drugs for inhibiting the pathological progression of epilepsy pose major obstacles to managing epilepsy. Here, we investigated the effect of tetramethylpyrazine (TMP), the main bioactive alkaloid isolated from the oriental medicine Ligusticum chuanxiong Hort., against the epileptogenesis progression of acute hippocampal and corneal (6 Hz) electrical kindling models of TLE. TMP dose-dependently limited the progression of seizures and reduced the after-discharge duration (ADDs) in a hippocampal mouse kindling model. Mice treated with TMP (20, 50 mg/kg, i.p.) remained in stage 1 of epileptic progression for a protracted period, requiring additional stimulation to induce stages 2-5 epileptic phenotypes. TMP (50 mg/kg) also inhibited 6 Hz corneal kindling progression. In contrast, TMP did not reverse the phenotypes induced in a generalized seizures (GS) model, or the maximal electroshock (MES) or pentylenetetrazole (PTZ)-induced models of epilepsy. Furthermore, patch clamp recordings revealed no effect of TMP (10 μM) on CA1 hippocampal neurons' intrinsic properties but suppressed the (i) frequency of spontaneous excitatory post synaptic currents (sEPSCs), (ii) paired pulse ratio (PPR), and (iii) long-term potentiation (LTP) induction in the Schaffer collateral-CA1 pathway. TMP suppressed the activity of calcium, but not sodium, channels. Taken together, these results suggest that TMP has an antiepileptogenic effect, likely through suppression of excitatory synaptic transmission by its effects on inhibition of calcium channels; these traits distinguish TMP from currently available AEDs. As mice administered TMP did not show any neurologic impairment in the object recognition and open field tests, the data support further development of TMP as a promising treatment for epilepsy.
Collapse
Affiliation(s)
| | - Song Cai
- Department of Anatomy, Histology & Developmental Biology, School of Basic Medical Sciences, Shenzhen University Health Science Centre, Shenzhen 518055, China
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, Arizona 85724, United States
| | | | - Kai Zhong
- Hangzhou Medical College, Hangzhou 310053, China
| | | | | | - Lindsey A. Chew
- School of Medicine, Duke University, Durham, North Carolina 27710, United States
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona College of Medicine and College of Pharmacy, Tucson, Arizona 85724, United States
- The Center for Innovation in Brain Sciences, The University of Arizona Health Sciences, Tucson, Arizona 85724, United States
| | | | | |
Collapse
|
649
|
Lu Z, Zhao T, Tao L, Yu Q, Yang Y, Cheng J, Lu S, Ding Q. Cystathionine β-Synthase-Derived Hydrogen Sulfide Correlates with Successful Aging in Mice. Rejuvenation Res 2019; 22:513-520. [PMID: 30799778 DOI: 10.1089/rej.2018.2166] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Zhihong Lu
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tianzhi Zhao
- Department of Neurosurgery, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Lei Tao
- Department of Anesthesiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Qian Yu
- Department of Anesthesiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Yonghui Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Jin Cheng
- Department of Cardiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Shaoping Lu
- Department of Cardiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| | - Qian Ding
- Department of Anesthesiology, the Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
650
|
Wang KW, Ye XL, Huang T, Yang XF, Zou LY. Optogenetics-induced activation of glutamate receptors improves memory function in mice with Alzheimer's disease. Neural Regen Res 2019; 14:2147-2155. [PMID: 31397354 PMCID: PMC6788230 DOI: 10.4103/1673-5374.262593] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 02/18/2019] [Indexed: 12/21/2022] Open
Abstract
Optogenetics is a combination of optics and genetics technology that can be used to activate or inhibit specific cells in tissues. It has been used to treat Parkinson's disease, epilepsy and neurological diseases, but rarely Alzheimer's disease. Adeno-associated virus carrying the CaMK promoter driving the optogenetic channelrhodopsin-2 (CHR2) gene (or without the CHR2 gene, as control) was injected into the bilateral dentate gyri, followed by repeated intrahippocampal injections of soluble low-molecular-weight amyloid-β1-42 peptide (Aβ1-42). Subsequently, the region was stimulated with a 473 nm laser (1-3 ms, 10 Hz, 5 minutes). The novel object recognition test was conducted to test memory function in mice. Immunohistochemical staining was performed to analyze the numbers of NeuN and synapsin Ia/b-positive cells in the hippocampus. Western blot assay was carried out to analyze the expression levels of glial fibrillary acidic protein, NeuN, synapsin Ia/b, metabotropic glutamate receptor-1a (mGluR-1a), mGluR-5, N-methyl-D-aspartate receptor subunit NR1, glutamate receptor 2, interleukin-1β, interleukin-6 and interleukin-10. Optogenetic stimulation improved working and short-term memory in mice with Alzheimer's disease. This neuroprotective effect was associated with increased expression of NR1, glutamate receptor 2 and mGluR-5 in the hippocampus, and decreased expression of glial fibrillary acidic protein and interleukin-6. Our results show that optogenetics can be used to regulate the neuronal-glial network to ameliorate memory functions in mice with Alzheimer's disease. The study was approved by the Animal Resources Committee of Jinan University, China (approval No. LL-KT-2011134) on February 28, 2011.
Collapse
Affiliation(s)
- Ke-Wei Wang
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Xiao-Lin Ye
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Ting Huang
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| | - Xi-Fei Yang
- Key Laboratory of Modern Toxicology of Shenzhen, Shenzhen Center for Disease Control and Prevention, Shenzhen, Guangdong Province, China
| | - Liang-Yu Zou
- Department of Neurology, Shenzhen People's Hospital (First Affiliated Hospital of Southern University of Science and Technology), Second Clinical College, Jinan University, Shenzhen, Guangdong Province, China
| |
Collapse
|