651
|
Ge X, Guo M, Hu T, Li W, Huang S, Yin Z, Li Y, Chen F, Zhu L, Kang C, Jiang R, Lei P, Zhang J. Increased Microglial Exosomal miR-124-3p Alleviates Neurodegeneration and Improves Cognitive Outcome after rmTBI. Mol Ther 2019; 28:503-522. [PMID: 31843449 PMCID: PMC7001001 DOI: 10.1016/j.ymthe.2019.11.017] [Citation(s) in RCA: 152] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 11/10/2019] [Accepted: 11/21/2019] [Indexed: 01/20/2023] Open
Abstract
Repetitive mild traumatic brain injury (rmTBI) is considered to be an important risk factor for long-term neurodegenerative disorders such as Alzheimer's disease, which is characterized by β-amyloid abnormalities and impaired cognitive function. Microglial exosomes have been reported to be involved in the transportation, distribution, and clearance of β-amyloid in Alzheimer's disease. However, their impacts on the development of neurodegeneration after rmTBI are not yet known. The role of miRNAs in microglial exosomes on regulating post-traumatic neurodegeneration was investigated in the present study. We demonstrated that miR-124-3p level in microglial exosomes from injured brain was significantly altered in the acute, sub-acute, and chronic phases after rmTBI. In in vitro experiments, microglial exosomes with upregulated miR-124-3p (EXO-124) alleviated neurodegeneration in repetitive scratch-injured neurons. The effects were exerted by miR-124-3p targeting Rela, an inhibitory transcription factor of ApoE that promotes the β-amyloid proteolytic breakdown, thereby inhibiting β-amyloid abnormalities. In mice with rmTBI, the intravenously injected microglial exosomes were taken up by neurons in injured brain. Besides, miR-124-3p in the exosomes was transferred into hippocampal neurons and alleviated neurodegeneration by targeting the Rela/ApoE signaling pathway. Consequently, EXO-124 treatments improved the cognitive outcome after rmTBI, suggesting a promising therapeutic strategy for future clinical translation.
Collapse
Affiliation(s)
- Xintong Ge
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China
| | - Mengtian Guo
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Tianpeng Hu
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Wenzhu Li
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Shan Huang
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Zhenyu Yin
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Li
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China
| | - Fanglian Chen
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China
| | - Luoyun Zhu
- Department of Medical Examination, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chunsheng Kang
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China.
| | - Ping Lei
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin 300052, China; Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China; Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin 300052, China; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Tianjin 300052, China.
| |
Collapse
|
652
|
Moon GJ, Kim S, Jeon MT, Lee KJ, Jang IS, Nakamura M, Kim SR. Therapeutic Potential of AAV1-Rheb(S16H) Transduction Against Alzheimer's Disease. J Clin Med 2019; 8:jcm8122053. [PMID: 31766645 PMCID: PMC6947419 DOI: 10.3390/jcm8122053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/14/2019] [Accepted: 11/19/2019] [Indexed: 12/22/2022] Open
Abstract
We recently reported that adeno-associated virus serotype 1-constitutively active Ras homolog enriched in brain [AAV1-Rheb(S16H)] transduction of hippocampal neurons could induce neuron-astroglia interactions in the rat hippocampus in vivo, resulting in neuroprotection. However, it remains uncertain whether AAV1-Rheb(S16H) transduction induces neurotrophic effects and preserves the cognitive memory in an animal model of Alzheimer’s disease (AD) with characteristic phenotypic features, such as β-amyloid (Aβ) accumulation and cognitive impairments. To assess the therapeutic potential of Rheb(S16H) in AD, we have examined the beneficial effects of AAV1-Rheb(S16H) administration in the 5XFAD mouse model. Rheb(S16H) transduction of hippocampal neurons in the 5XFAD mice increased the levels of neurotrophic signaling molecules, including brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF), and their corresponding receptors, tropomyosin receptor kinase B (TrkB) and CNTF receptor α subunit (CNTFRα), respectively. In addition, Rheb(S16H) transduction inhibited Aβ production and accumulation in the hippocampus of 5XFAD mice and protected the decline of long-term potentiation (LTP), resulting in the prevention of cognitive impairments, which was demonstrated using novel object recognition testing. These results indicate that Rheb(S16H) transduction of hippocampal neurons may have therapeutic potential in AD by inhibiting Aβ accumulation and preserving LTP associated with cognitive memory.
Collapse
Affiliation(s)
- Gyeong Joon Moon
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (G.J.M.); (S.K.); (M.-T.J.)
- BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Sehwan Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (G.J.M.); (S.K.); (M.-T.J.)
- BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Min-Tae Jeon
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (G.J.M.); (S.K.); (M.-T.J.)
- BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Kea Joo Lee
- Neural Circuits Research Group, Korea Brain Research Institute, Daegu 41062, Korea;
| | - Il-Sung Jang
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea;
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea;
| | - Michiko Nakamura
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 41940, Korea;
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea; (G.J.M.); (S.K.); (M.-T.J.)
- BK21 plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41566, Korea;
- Institute of Life Science & Biotechnology, Kyungpook National University, Daegu 41566, Korea
- Correspondence:
| |
Collapse
|
653
|
Kárpáti A, Yoshikawa T, Naganuma F, Matsuzawa T, Kitano H, Yamada Y, Yokoyama M, Futatsugi A, Mikoshiba K, Yanai K. Histamine H 1 receptor on astrocytes and neurons controls distinct aspects of mouse behaviour. Sci Rep 2019; 9:16451. [PMID: 31712580 PMCID: PMC6848115 DOI: 10.1038/s41598-019-52623-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 10/21/2019] [Indexed: 01/03/2023] Open
Abstract
Histamine is an important neurotransmitter that contributes to various processes, including the sleep-wake cycle, learning, memory, and stress responses. Its actions are mediated through histamine H1-H4 receptors. Gene knockout and pharmacological studies have revealed the importance of H1 receptors in learning and memory, regulation of aggression, and wakefulness. H1 receptors are abundantly expressed on neurons and astrocytes. However, to date, studies selectively investigating the roles of neuronal and astrocytic H1 receptors in behaviour are lacking. We generated novel astrocyte- and neuron-specific conditional knockout (cKO) mice to address this gap in knowledge. cKO mice showed cell-specific reduction of H1 receptor gene expression. Behavioural assessment revealed significant changes and highlighted the importance of H1 receptors on both astrocytes and neurons. H1 receptors on both cell types played a significant role in anxiety. Astrocytic H1 receptors were involved in regulating aggressive behaviour, circadian rhythms, and quality of wakefulness, but not sleep behaviour. Our results emphasise the roles of neuronal H1 receptors in recognition memory. In conclusion, this study highlights the novel roles of H1 receptors on astrocytes and neurons in various brain functions.
Collapse
Affiliation(s)
- Anikó Kárpáti
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan.
| | - Fumito Naganuma
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai, 983-8536, Japan
| | - Takuro Matsuzawa
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Haruna Kitano
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Yo Yamada
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Mariko Yokoyama
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| | - Akira Futatsugi
- Department of Basic Medical Sciences, Kobe City College of Nursing, 3-4 Gakuen-nishi-machi, Nishi-ku, Kobe, 651-2103, Japan
| | - Katsuhiko Mikoshiba
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, 201210, China
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai, 980-8575, Japan
| |
Collapse
|
654
|
Identity domains capture individual differences from across the behavioral repertoire. Nat Neurosci 2019; 22:2023-2028. [PMID: 31686022 DOI: 10.1038/s41593-019-0516-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 09/16/2019] [Indexed: 11/09/2022]
Abstract
Personality traits can offer considerable insight into the biological basis of individual differences. However, existing approaches toward understanding personality across species rely on subjective criteria and limited sets of behavioral readouts, which result in noisy and often inconsistent outcomes. Here we introduce a mathematical framework for describing individual differences along dimensions with maximum consistency and discriminative power. We validate this framework in mice, using data from a system for high-throughput longitudinal monitoring of group-housed male mice that yields a variety of readouts from across the behavioral repertoire of individual animals. We demonstrate a set of stable traits that capture variability in behavior and gene expression in the brain, allowing for better-informed mechanistic investigations into the biology of individual differences.
Collapse
|
655
|
Paternal valproic acid exposure in mice triggers behavioral alterations in offspring. Neurotoxicol Teratol 2019; 76:106837. [DOI: 10.1016/j.ntt.2019.106837] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/22/2019] [Accepted: 09/18/2019] [Indexed: 01/29/2023]
|
656
|
Gulinello M, Mitchell HA, Chang Q, Timothy O'Brien W, Zhou Z, Abel T, Wang L, Corbin JG, Veeraragavan S, Samaco RC, Andrews NA, Fagiolini M, Cole TB, Burbacher TM, Crawley JN. Rigor and reproducibility in rodent behavioral research. Neurobiol Learn Mem 2019; 165:106780. [PMID: 29307548 PMCID: PMC6034984 DOI: 10.1016/j.nlm.2018.01.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/22/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023]
Abstract
Behavioral neuroscience research incorporates the identical high level of meticulous methodologies and exacting attention to detail as all other scientific disciplines. To achieve maximal rigor and reproducibility of findings, well-trained investigators employ a variety of established best practices. Here we explicate some of the requirements for rigorous experimental design and accurate data analysis in conducting mouse and rat behavioral tests. Novel object recognition is used as an example of a cognitive assay which has been conducted successfully with a range of methods, all based on common principles of appropriate procedures, controls, and statistics. Directors of Rodent Core facilities within Intellectual and Developmental Disabilities Research Centers contribute key aspects of their own novel object recognition protocols, offering insights into essential similarities and less-critical differences. Literature cited in this review article will lead the interested reader to source papers that provide step-by-step protocols which illustrate optimized methods for many standard rodent behavioral assays. Adhering to best practices in behavioral neuroscience will enhance the value of animal models for the multiple goals of understanding biological mechanisms, evaluating consequences of genetic mutations, and discovering efficacious therapeutics.
Collapse
Affiliation(s)
- Maria Gulinello
- IDDRC Behavioral Core Facility, Neuroscience Department, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Heather A Mitchell
- IDD Models Core, Waisman Center, University of Wisconsin Madison, Madison, WI 53705, USA
| | - Qiang Chang
- IDD Models Core, Waisman Center, University of Wisconsin Madison, Madison, WI 53705, USA
| | - W Timothy O'Brien
- IDDRC Preclinical Models Core, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Zhaolan Zhou
- IDDRC Preclinical Models Core, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ted Abel
- IDDRC Preclinical Models Core, Children's Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Current affiliation: Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
| | - Li Wang
- IDDRC Neurobehavioral Core, Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA
| | - Joshua G Corbin
- IDDRC Neurobehavioral Core, Center for Neuroscience Research, Children's National Health System, Washington, DC 20010, USA
| | - Surabi Veeraragavan
- IDDRC Neurobehavioral Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Rodney C Samaco
- IDDRC Neurobehavioral Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nick A Andrews
- IDDRC Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA 02115, USA
| | - Michela Fagiolini
- IDDRC Neurodevelopmental Behavior Core, Boston Children's Hospital, Boston, MA 02115, USA
| | - Toby B Cole
- IDDRC Rodent Behavior Laboratory, Center on Human Development and Disability, University of Washington, Seattle, WA 98195, USA
| | - Thomas M Burbacher
- IDDRC Rodent Behavior Laboratory, Center on Human Development and Disability, University of Washington, Seattle, WA 98195, USA
| | - Jacqueline N Crawley
- IDDRC Rodent Behavior Core, MIND Institute, University of California Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
657
|
Planchez B, Surget A, Belzung C. Animal models of major depression: drawbacks and challenges. J Neural Transm (Vienna) 2019; 126:1383-1408. [PMID: 31584111 PMCID: PMC6815270 DOI: 10.1007/s00702-019-02084-y] [Citation(s) in RCA: 268] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Major depression is a leading contributor to the global burden of disease. This situation is mainly related to the chronicity and/or recurrence of the disorder, and to poor response to antidepressant therapy. Progress in this area requires valid animal models. Current models are based either on manipulating the environment to which rodents are exposed (during the developmental period or adulthood) or biological underpinnings (i.e. gene deletion or overexpression of candidate genes, targeted lesions of brain areas, optogenetic control of specific neuronal populations, etc.). These manipulations can alter specific behavioural and biological outcomes that can be related to different symptomatic and pathophysiological dimensions of major depression. However, animal models of major depression display substantial shortcomings that contribute to the lack of innovative pharmacological approaches in recent decades and which hamper our capabilities to investigate treatment-resistant depression. Here, we discuss the validity of these models, review putative models of treatment-resistant depression, major depression subtypes and recurrent depression. Furthermore, we identify future challenges regarding new paradigms such as those proposing dimensional rather than categorical approaches to depression.
Collapse
Affiliation(s)
| | | | - Catherine Belzung
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France.
- UMR 1253, iBrain, UFR Sciences et Techniques, Parc Grandmont, 37200, Tours, France.
| |
Collapse
|
658
|
Zheng ZV, Lam PK, Poon WS, Wong KCG. The Time Course of Cognitive Deficits in Experimental Subarachnoid Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2019; 127:121-125. [PMID: 31407072 DOI: 10.1007/978-3-030-04615-6_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating stroke type. Approximately 50% of survivors suffer from the permanent disability, caused by the cognitive deficits. To enrich the pre-clinical research on the neurological deficits after SAH, we investigate the temporal cognitive deficits and the longitudinal course of cognitive recovery in endovascular perforation SAH murine model. The SAH mice show reproducible body weakness and headache-symbolized moaning symptoms, which is closed to clinical patients. SAH mice exhibit significantly impaired cognitive function in domains of learning ability, short-term and long-term memory. The cognitive deficits occur mostly in the early phase and recover gradually till day 10 after SAH. The systematical assessments of cognitive function after experimental aneurysmal SAH elucidate the time course of cognitive deficits and provide the time window of potential interventions.
Collapse
Affiliation(s)
- Zhiyuan Vera Zheng
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Kuen Lam
- Department of Surgery, Chow Tai Fook-Cheng Yu Tung Surgical Stem cell Research Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Kwok Chu George Wong
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
| |
Collapse
|
659
|
Muthiah MD, Huang DQ, Zhou L, Jumat NH, Choolani M, Chan JKY, Wee A, Lim SG, Dan YY. A murine model demonstrating reversal of structural and functional correlates of cirrhosis with progenitor cell transplantation. Sci Rep 2019; 9:15446. [PMID: 31659188 PMCID: PMC6817879 DOI: 10.1038/s41598-019-51189-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 09/03/2019] [Indexed: 01/07/2023] Open
Abstract
Development of cell transplantation for treating liver cirrhosis hinges critically on the availability of animal models for studying human stem cell transplantation. We report an immune-permissive murine model of liver cirrhosis with full clinical correlates of decompensated liver disease, and allows testing efficacy of stem cell transplantation. Liver cirrhosis was induced in Nod-scid gamma(NSG) mice with oral thioacetamide(TA) and compared to controls over 12 months. 4 month TA treated cirrhotic mice were then transplanted intrasplenically with 2million human fetal liver progenitor cells(HFH) and compared with cirrhotic controls 2 months after transplantation. NSG-TA mice developed shrunken and nodular livers with histological evidence of fibrosis as compared to controls. This was associated with evidence of worsening decompensated liver disease, with jaundice, hypoalbuminemia, coagulopathy, and encephalopathy in NSG-TA mice. Transplantation of HFH resulted in improvement in both fibrosis and markers of decompensated liver disease. We have demonstrated that NSG-TA mice can recapitulate the full clinical picture of structural and functional cirrhosis, both of which can be improved by transplantation of human fetal liver cells. This model serves as a valuable tool for validation of in vivo liver stem cell transplantation and opens up opportunities for studying the mechanism how stem cells reverse fibrosis.
Collapse
Affiliation(s)
- Mark D Muthiah
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel Q Huang
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lei Zhou
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nur Halisah Jumat
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mahesh Choolani
- Department of Obstetrics and Gynaecology, National University Health System, Singapore, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, Singapore
- Experimental Fetal Medicine Group, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Aileen Wee
- Department of Pathology, National University Health System, Singapore, Singapore
| | - Seng Gee Lim
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yock-Young Dan
- Department of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore.
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
660
|
Loftis JM, Taylor J, Hudson R, Firsick EJ. Neuroinvasion and cognitive impairment in comorbid alcohol dependence and chronic viral infection: An initial investigation. J Neuroimmunol 2019; 335:577006. [PMID: 31325774 DOI: 10.1016/j.jneuroim.2019.577006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022]
Abstract
Viruses that invade the central nervous system (CNS) can cause neuropsychiatric impairments. Similarly, chronic alcohol exposure can induce inflammatory responses that alter brain function. However, the effects of a chronic viral infection and comorbid alcohol use on neuroinflammation and behavior are not well-defined. We investigated the role of heavy alcohol intake in regulating inflammatory responses and behavioral signs of cognitive impairments in mice infected with lymphocytic choriomeningitis virus (LCMV) clone 13. LCMV-infected mice exposed to alcohol had increased peripheral inflammation and impaired cognitive function (as indicated by performance on the novel object recognition test). Initial findings suggest that brain region-specific dysregulation of microglial response to viral infection may contribute to cognitive impairments in the context of heavy alcohol use.
Collapse
Affiliation(s)
- Jennifer M Loftis
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA; Methamphetamine Abuse Research Center, Veterans Affairs Portland Health Care System, Oregon Health & Science University, Portland, OR, USA.
| | - Jonathan Taylor
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Psychiatry, Oregon Health & Science University, Portland, OR, USA
| | - Rebekah Hudson
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA; Department of Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Evan J Firsick
- Research & Development Service, Veterans Affairs Portland Health Care System, Portland, OR, USA
| |
Collapse
|
661
|
Christenson Wick Z, Tetzlaff MR, Krook-Magnuson E. Novel long-range inhibitory nNOS-expressing hippocampal cells. eLife 2019; 8:46816. [PMID: 31609204 PMCID: PMC6839902 DOI: 10.7554/elife.46816] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022] Open
Abstract
The hippocampus, a brain region that is important for spatial navigation and episodic memory, benefits from a rich diversity of neuronal cell-types. Through the use of an intersectional genetic viral vector approach in mice, we report novel hippocampal neurons which we refer to as LINCs, as they are long-range inhibitory neuronal nitric oxide synthase (nNOS)-expressing cells. LINCs project to several extrahippocampal regions including the tenia tecta, diagonal band, and retromammillary nucleus, but also broadly target local CA1 cells. LINCs are thus both interneurons and projection neurons. LINCs display regular spiking non-pyramidal firing patterns, are primarily located in the stratum oriens or pyramidale, have sparsely spiny dendrites, and do not typically express somatostatin, VIP, or the muscarinic acetylcholine receptor M2. We further demonstrate that LINCs can strongly influence hippocampal function and oscillations, including interregional coherence. The identification and characterization of these novel cells advances our basic understanding of both hippocampal circuitry and neuronal diversity.
Collapse
Affiliation(s)
- Zoé Christenson Wick
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, United States
| | - Madison R Tetzlaff
- Neuroscience Department, University of Minnesota, Minneapolis, United States
| | | |
Collapse
|
662
|
Consequences of VGluT3 deficiency on learning and memory in mice. Physiol Behav 2019; 212:112688. [PMID: 31622610 DOI: 10.1016/j.physbeh.2019.112688] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/22/2019] [Accepted: 09/22/2019] [Indexed: 01/06/2023]
Abstract
The aim of the present study was to test the hypothesis that vesicular glutamate transporter 3 (VGluT3) deficiency is associated with cognitive impairments. Male VGluT3 knockout (KO) and wild type (WT) mice were exposed to a behavioral test battery covering paradigms based on spontaneous exploratory behavior and reinforcement-based learning tests. Reversal learning was examined to test the cognitive flexibility. The VGluT3 KO mice clearly exhibited the ability to learn. The social recognition memory of KO mice was intact. The y-maze test revealed weaker working memory of VGluT3 KO mice. No significant learning impairments were noticed in operant conditioning or holeboard discrimination paradigm. In avoidance-based learning tests (Morris water maze and active avoidance), KO mice exhibited slightly slower learning process compared to WT mice, but not a complete learning impairment. In tests based on simple associations (operant conditioning, avoidance learning) an attenuation of cognitive flexibility was observed in KO mice. In conclusion, knocking out VGluT3 results in mild disturbances in working memory and learning flexibility. Apparently, this glutamate transporter is not a major player in learning and memory formation in general. Based on previous characteristics of VGluT3 KO mice we would have expected a stronger deficit. The observed hypolocomotion did not contribute to the mild cognitive disturbances herein reported, either.
Collapse
|
663
|
Krishna Chandran AM, Christina H, Das S, Mumbrekar KD, Satish Rao BS. Neuroprotective role of naringenin against methylmercury induced cognitive impairment and mitochondrial damage in a mouse model. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2019; 71:103224. [PMID: 31376681 DOI: 10.1016/j.etap.2019.103224] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 07/09/2019] [Accepted: 07/21/2019] [Indexed: 06/10/2023]
Abstract
Human exposure to organomercurials like methylmercury (MeHg) may occur by consumption of contaminated seafood, affecting various vital organs especially, brain contributing to neuro disorders. The citrus flavanone, naringenin (NAR) has shown strong antioxidant and anti-inflammatory effects and therefore may exert cytoprotective effect against xenobiotic agents. Herein, we investigated the neuroprotective role of NAR against MeHg induced functional changes in mitochondria, neuronal cell death and cognitive impairment in a mouse model. A neurotoxic dose of MeHg (4 mg/kg.b.wt.) was administered orally to mice for 15 days. This resulted in the reduction of GSH and GST, an increase in mitochondrial DNA damage and memory impairment. On the contrary, NAR pre-treatment (100 mg/kg.b.wt.), helped in lowering the oxidative burden which in turn maintained mitochondrial function and prevented induced neuronal cell death, ultimately improving the cognitive impairment. As MeHg intoxication occurs chronically, consumption of the dietary components rich in NAR may have its positive human health impact, ultimately improving the quality of life.
Collapse
Affiliation(s)
- Adwaid Manu Krishna Chandran
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Hannah Christina
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Shubhankar Das
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - Kamalesh D Mumbrekar
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India
| | - B S Satish Rao
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576 104, Karnataka, India.
| |
Collapse
|
664
|
Ni Y, Yang X, Zheng L, Wang Z, Wu L, Jiang J, Yang T, Ma L, Fu Z. Lactobacillus
and
Bifidobacterium
Improves Physiological Function and Cognitive Ability in Aged Mice by the Regulation of Gut Microbiota. Mol Nutr Food Res 2019; 63:e1900603. [DOI: 10.1002/mnfr.201900603] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/16/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Yinhua Ni
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Xin Yang
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Liujie Zheng
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Zhe Wang
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Lianxin Wu
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Jinlu Jiang
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Tianqi Yang
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Lingyan Ma
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| | - Zhengwei Fu
- College of Biotechnology and BioengineeringZhejiang University of Technology China
| |
Collapse
|
665
|
Commins S, Kirby BP. The complexities of behavioural assessment in neurodegenerative disorders: A focus on Alzheimer’s disease. Pharmacol Res 2019; 147:104363. [DOI: 10.1016/j.phrs.2019.104363] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/12/2019] [Accepted: 07/19/2019] [Indexed: 01/21/2023]
|
666
|
Leo A, Citraro R, Tallarico M, Iannone M, Fedosova E, Nesci V, De Sarro G, Sarkisova K, Russo E. Cognitive impairment in the WAG/Rij rat absence model is secondary to absence seizures and depressive-like behavior. Prog Neuropsychopharmacol Biol Psychiatry 2019; 94:109652. [PMID: 31095993 DOI: 10.1016/j.pnpbp.2019.109652] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 01/06/2023]
Abstract
Neuropsychiatric comorbidities are common in patients with epilepsy, remaining still an urgent unmet clinical need. Therefore, the management of epileptic disorders should not only be restricted to the achievement of seizure-freedom but must also be able to counteract its related comorbidities. Experimental animal models of epilepsy represent a valid tool not only to study epilepsy but also its associated comorbidities. The WAG/Rij rat is a well-established genetically-based model of absence epilepsy with depressive-like comorbidity, in which learning and memory impairment was also recently reported. Aim of this study was to clarify whether this cognitive decline is secondary or not to absence seizures and/or depressive-like behavior. The behavioral performance of untreated and ethosuximide-treated (300 mg/kg/day; 17 days) WAG/Rij rats at 6 and 12 months of age were assessed in several tests: forced swimming test, objects recognition test, social recognition test, Morris water maze and passive avoidance. According to our results, it seems that cognitive impairment in this strain, similarly to depressive-like behavior, is secondary to the occurrence of absence seizures, which might be necessary for the expression of cognitive impairment. Furthermore, our results suggest an age-dependent impairment of cognitive performance in WAG/Rij rats, which could be linked to the age-dependent increase of spike wave discharges. Consistently, it is possible that absence seizures, depressive-like behavior and cognitive deficit may arise independently and separately in lifetime from the same underlying network disease, as previously suggested for the behavioral features associated with other epileptic syndromes.
Collapse
Affiliation(s)
- Antonio Leo
- University of Catanzaro, School of Medicine, Science of Health Dept., Catanzaro, Italy
| | - Rita Citraro
- University of Catanzaro, School of Medicine, Science of Health Dept., Catanzaro, Italy.
| | - Martina Tallarico
- University of Catanzaro, School of Medicine, Science of Health Dept., Catanzaro, Italy; CNR, Institute of Neurological Sciences, Pharmacology Section, Roccelletta di Borgia, Catanzaro, Italy
| | - Michelangelo Iannone
- CNR, Institute of Neurological Sciences, Pharmacology Section, Roccelletta di Borgia, Catanzaro, Italy
| | - Ekaterina Fedosova
- Institute of Higher Nervous Activity and Neurophysiology RAS, Moscow, Russia
| | - Valentina Nesci
- University of Catanzaro, School of Medicine, Science of Health Dept., Catanzaro, Italy
| | | | - Karine Sarkisova
- Institute of Higher Nervous Activity and Neurophysiology RAS, Moscow, Russia
| | - Emilio Russo
- University of Catanzaro, School of Medicine, Science of Health Dept., Catanzaro, Italy
| |
Collapse
|
667
|
Sen MK, Mahns DA, Coorssen JR, Shortland PJ. Behavioural phenotypes in the cuprizone model of central nervous system demyelination. Neurosci Biobehav Rev 2019; 107:23-46. [PMID: 31442519 DOI: 10.1016/j.neubiorev.2019.08.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/01/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022]
Abstract
The feeding of cuprizone (CPZ) to animals has been extensively used to model the processes of demyelination and remyelination, with many papers adopting a narrative linked to demyelinating conditions like multiple sclerosis (MS), the aetiology of which is unknown. However, no current animal model faithfully replicates the myriad of symptoms seen in the clinical condition of MS. CPZ ingestion causes mitochondrial and endoplasmic reticulum stress and subsequent apoptosis of oligodendrocytes leads to central nervous system demyelination and glial cell activation. Although there are a wide variety of behavioural tests available for characterizing the functional deficits in animal models of disease, including that of CPZ-induced deficits, they have focused on a narrow subset of outcomes such as motor performance, cognition, and anxiety. The literature has not been systematically reviewed in relation to these or other symptoms associated with clinical MS. This paper reviews these tests and makes recommendations as to which are the most important in order to better understand the role of this model in examining aspects of demyelinating diseases like MS.
Collapse
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - David A Mahns
- School of Medicine, Western Sydney University, New South Wales, Australia
| | - Jens R Coorssen
- Departments of Health Sciences and Biological Sciences, Faculties of Applied Health Sciences and Mathematics & Science, Brock University, Ontario, Canada.
| | - Peter J Shortland
- Science and Health, Western Sydney University, New South Wales, Australia.
| |
Collapse
|
668
|
Gorgun MF, Zhuo M, Dineley KT, Englander EW. Elevated Neuroglobin Lessens Neuroinflammation and Alleviates Neurobehavioral Deficits Induced by Acute Inhalation of Combustion Smoke in the Mouse. Neurochem Res 2019; 44:2170-2181. [PMID: 31420834 DOI: 10.1007/s11064-019-02856-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/03/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022]
Abstract
Acute inhalation of combustion smoke produces long-term neurologic deficits in survivors. To study the mechanisms that contribute to the development of neurologic deficits and identify targets for prevention, we developed a mouse model of acute inhalation of combustion smoke, which supports longitudinal investigation of mechanisms that underlie the smoke induced inimical sequelae in the brain. Using a transgenic mouse engineered to overexpress neuroglobin, a neuroprotective oxygen-binding globin protein, we previously demonstrated that elevated neuroglobin preserves mitochondrial respiration and attenuates formation of oxidative DNA damage in the mouse brain after smoke exposure. In the current study, we show that elevated neuronal neuroglobin attenuates the persistent inflammatory changes induced by smoke exposure in the mouse brain and mitigates concordant smoke-induced long-term neurobehavioral deficits. Specifically, we found that increases in hippocampal density of GFAP and Iba-1 positive cells that are detected post-smoke in wild-type mice are absent in the neuroglobin overexpressing transgenic (Ngb-tg) mice. Similarly, the smoke induced hippocampal myelin depletion is not observed in the Ngb-tg mice. Importantly, elevated neuroglobin alleviates behavioral and memory deficits that develop after acute smoke inhalation in the wild-type mice. Taken together, our findings suggest that the protective effects exerted by neuroglobin in the brains of smoke exposed mice afford protection from long-term neurologic sequelae of acute inhalation of combustion smoke. Our transgenic mouse provides a tool for assessing the potential of elevated neuroglobin as possible strategy for management of smoke inhalation injury.
Collapse
Affiliation(s)
- Murat F Gorgun
- Department of Surgery, Medical Branch, University of Texas, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Ming Zhuo
- Department of Surgery, Medical Branch, University of Texas, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Kelly T Dineley
- Department of Neurology, University of Texas Medical Branch, Galveston, TX, USA
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA
- Center for Addiction Research, University of Texas Medical Branch, Galveston, TX, USA
| | - Ella W Englander
- Department of Surgery, Medical Branch, University of Texas, 301 University Boulevard, Galveston, TX, 77555, USA.
- Shriners Hospitals for Children, Galveston, TX, USA.
| |
Collapse
|
669
|
Stefanello FV, Fontana BD, Ziani PR, Müller TE, Mezzomo NJ, Rosemberg DB. Exploring Object Discrimination in Zebrafish: Behavioral Performance and Scopolamine-Induced Cognitive Deficits at Different Retention Intervals. Zebrafish 2019; 16:370-378. [DOI: 10.1089/zeb.2018.1703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- Flavia V. Stefanello
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | - Barbara D. Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Paola R. Ziani
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Talise E. Müller
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
| | - Nathana J. Mezzomo
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Pharmacology, Federal University of Santa Maria, Santa Maria, Brazil
| | - Denis B. Rosemberg
- Laboratory of Experimental Neuropsychobiology, Department of Biochemistry and Molecular Biology, Natural and Exact Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
- Graduate Program in Biological Sciences, Toxicological Biochemistry, Federal University of Santa Maria, Santa Maria, Brazil
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, Louisiana
| |
Collapse
|
670
|
Balbi M, Vanni MP, Vega MJ, Silasi G, Sekino Y, Boyd JD, LeDue JM, Murphy TH. Longitudinal monitoring of mesoscopic cortical activity in a mouse model of microinfarcts reveals dissociations with behavioral and motor function. J Cereb Blood Flow Metab 2019; 39:1486-1500. [PMID: 29521138 PMCID: PMC6681536 DOI: 10.1177/0271678x18763428] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 12/14/2022]
Abstract
Small vessel disease is characterized by sporadic obstruction of small vessels leading to neuronal cell death. These microinfarcts often escape detection by conventional magnetic resonance imaging and are identified only upon postmortem examination. Our work explores a brain-wide microinfarct model in awake head-fixed mice, where occlusions of small penetrating arterioles are reproduced by endovascular injection of fluorescent microspheres. Mesoscopic functional connectivity was mapped longitudinally in awake GCaMP6 mice using genetically encoded calcium indicators for transcranial wide-field calcium imaging. Microsphere occlusions were quantified and changes in cerebral blood flow were measured with laser speckle imaging. The neurodeficit score in microinfarct mice was significantly higher than in sham, indicating impairment in motor function. The novel object recognition test showed a reduction in the discrimination index in microinfarct mice compared to sham. Graph-theoretic analysis of functional connectivity did not reveal significant differences in functional connectivity between sham and microinfarct mice. While behavioral tasks revealed impairments following microinfarct induction, the absence of measurable functional alterations in cortical activity has a less straightforward interpretation. The behavioral alterations produced by this model are consistent with alterations observed in human patients suffering from microinfarcts and support the validity of microsphere injection as a microinfarct model.
Collapse
Affiliation(s)
- Matilde Balbi
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
| | - Matthieu P Vanni
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
| | - Max J Vega
- Department of Psychology, Motivated
Cognition Lab, University of British Columbia, Vancouver, British Columbia,
Canada
| | - Gergely Silasi
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
| | - Yuki Sekino
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
| | - Jamie D Boyd
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
| | - Jeffrey M LeDue
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
- Djavad Mowafaghian Center for Brain
Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Timothy H Murphy
- Department of Psychiatry, Kinsmen
Laboratory of Neurological Research, University of British Columbia, Vancouver,
British Columbia, Canada
- Djavad Mowafaghian Center for Brain
Health, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
671
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
672
|
Chen YJ, Tang ZZ, Du L, Liu Y, Lu Q, Ma TF, Liu YW. A novel compound AB-38b improves diabetes-associated cognitive decline in mice via activation of Nrf2/ARE pathway. Brain Res Bull 2019; 150:160-167. [DOI: 10.1016/j.brainresbull.2019.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 04/30/2019] [Accepted: 05/14/2019] [Indexed: 12/14/2022]
|
673
|
Zhang R, Liu Y, Chen Y, Li Q, Marshall C, Wu T, Hu G, Xiao M. Aquaporin 4 deletion exacerbates brain impairments in a mouse model of chronic sleep disruption. CNS Neurosci Ther 2019; 26:228-239. [PMID: 31364823 PMCID: PMC6978250 DOI: 10.1111/cns.13194] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 06/20/2019] [Accepted: 06/26/2019] [Indexed: 12/24/2022] Open
Abstract
AIMS As a normal physiological process, sleep has recently been shown to facilitate clearance of macromolecular metabolic wastes from the brain via the glymphatic system. The aim of the present study was to investigate pathophysiological roles of astroglial aquaporin 4 (AQP4), a functional regulator of glymphatic clearance, in a mouse model of chronic sleep disruption (SD). METHODS Adult AQP4 null mice and wild-type (WT) mice were given 7 days of SD using the improved rotating rod method, and then received behavioral, neuropathological, and neurochemical analyses. RESULTS Aquaporin 4 deletion resulted in an impairment of glymphatic transport and accumulation of β-amyloid and Tau proteins in the brain following SD. AQP4 null SD mice exhibited severe activation of microglia, neuroinflammation, and synaptic protein loss in the hippocampus, as well as decreased working memory, compared with WT-SD mice. CONCLUSION These results demonstrate that AQP4-mediated glymphatic clearance ameliorates brain impairments caused by abnormal accumulation of metabolic wastes following chronic SD, thus serving as a potential target for sleep-related disorders.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yun Liu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Li
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Charles Marshall
- Department of Rehabilitation Sciences, University of Kentucky Center of Excellence in Rural Health, Hazard, KY, USA
| | - Ting Wu
- Department of Neurology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ming Xiao
- Jiangsu Province, Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.,Brain Institute, the Affiliated Nanjing Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
674
|
Aerobic Physical Exercise as a Neuroprotector Strategy for Ethanol Binge-Drinking Effects in the Hippocampus and Systemic Redox Status in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2415243. [PMID: 31354903 PMCID: PMC6637690 DOI: 10.1155/2019/2415243] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 05/06/2019] [Accepted: 05/27/2019] [Indexed: 12/22/2022]
Abstract
The heavy and episodic EtOH drinking pattern, equivalent to weekend consumption, characterizes the binge-drinking pattern and promotes a misbalance of encephalic metabolic functions, concurring to neurodegeneration and cerebral dysfunction. And for being a legal drug, it has global public health and social relevance. In this way, we aimed to investigate the effects of physical training, in a treadmill, on the deleterious effects of EtOH on hippocampal functions, related to memory and learning. For this, we used 40 Wistar rats, divided into four groups: Control group, Trained group (trained animals with doses of distilled water), EtOH group (nontrained animals with doses of 3 g/kg/day of EtOH, 20% w/v), and Trained+EtOH group (trained animals exposed to EtOH). The physical exercise was performed by running on a treadmill for 5 days a week for 4 weeks, and all doses of EtOH were administered through intragastric gavage in four repeated cycles of EtOH in binge. After the experimental period, the animals were submitted to the object recognition task and Morris water maze test, and after being euthanized, the blood and hippocampus were collected for Trolox Equivalent Antioxidant Capacity (TEAC), Reduced Glutathione Content (GSH), and Nitrite and Lipid Peroxidation (LPO) level measurements. Our results showed that EtOH caused marked oxidative stress and mnemonic damage, and the physical exercise promoted neuroprotective effects, among them, the modulation of oxidative biochemistry in plasma (by restoring GSH levels) and in the hippocampus (by reducing LPO levels and increasing antioxidant parameters) and cognitive function improvement. Therefore, physical exercise can be an important prophylactic and therapeutic tool in order to ameliorate and even prevent the deleterious effects of EtOH on cognitive functions.
Collapse
|
675
|
Lee JS, Lee Y, André EA, Lee KJ, Nguyen T, Feng Y, Jia N, Harris BT, Burns MP, Pak DTS. Inhibition of Polo-like kinase 2 ameliorates pathogenesis in Alzheimer's disease model mice. PLoS One 2019; 14:e0219691. [PMID: 31306446 PMCID: PMC6629081 DOI: 10.1371/journal.pone.0219691] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 06/28/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer disease (AD) is a neurodegenerative disorder characterized by pathological hallmarks of neurofibrillary tangles and amyloid plaques. The plaques are formed by aggregation and accumulation of amyloid β (Aβ), a cleavage fragment of amyloid precursor protein (APP). Enhanced neuronal activity and seizure events are frequently observed in AD, and elevated synaptic activity promotes Aβ production. However, the mechanisms that link synaptic hyperactivity to APP processing and AD pathogenesis are not well understood. We previously found that Polo-like kinase 2 (Plk2), a homeostatic repressor of neuronal overexcitation, promotes APP β-processing in vitro. Here, we report that Plk2 stimulates Aβ production in vivo, and that Plk2 levels are elevated in a spatiotemporally regulated manner in brains of AD mouse models and human AD patients. Genetic disruption of Plk2 kinase function reduces plaque deposits and activity-dependent Aβ production. Furthermore, pharmacological Plk2 inhibition hinders Aβ formation, synapse loss, and memory decline in an AD mouse model. Thus, Plk2 links synaptic overactivity to APP β-processing, Aβ production, and disease-relevant phenotypes in vivo, suggesting that Plk2 may be a potential target for AD therapeutics.
Collapse
Affiliation(s)
- Ji Soo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yeunkum Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Emily A. André
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Kea Joo Lee
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Thien Nguyen
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Yang Feng
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Nuo Jia
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Brent T. Harris
- Department of Neurology, Georgetown University Medical Center, Washington, DC, United States of America
| | - Mark P. Burns
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, United States of America
| | - Daniel T. S. Pak
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States of America
| |
Collapse
|
676
|
Tang Q, Ke H, Wu C, Zeng J, Li Z, Liu Y, Feng S, Xue Q, Xu X. Aqueous extract from You-Gui-Yin ameliorates cognitive impairment of chronic renal failure mice through targeting hippocampal CaMKIIα/CREB/BDNF and EPO/EPOR pathways. JOURNAL OF ETHNOPHARMACOLOGY 2019; 239:111925. [PMID: 31055001 DOI: 10.1016/j.jep.2019.111925] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 03/27/2019] [Accepted: 04/27/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE You-Gui-Yin (YGY) is a traditional Chinese recipe used for reinforcing kidney essence which is recorded in Jingyue Quanshu written by Zhang Jingyue in Ming dynasty. According to traditional Chinese medicine theory, kidney essence is associated with brain and without sufficient kidney essence, cognitive impairment may occur. AIM OF THE STUDY In this study, we aimed to investigate the effect of YGY extract on cognitive impairment of chronic renal failure (CRF) mice and explore the mechanisms involved. MATERIALS AND METHODS Aqueous extract of YGY was prepared from crude drugs and was quality controlled by high-performance liquid chromatography (HPLC). CRF was induced by 0.2% adenine in mice and CRF mice were intragastrically administered with 1.5 g kg-1, 3.0 g kg-1, and 6.0 g kg-1 of YGY extract. Mice were identified with CRF by determining several biochemical and physiological indexes, including creatinine clearance rate, serum creatinine, serum urea nitrogen, serum Ca, serum P, serum Mg, body weight and body temperature. Morris water maze and novel object recognition tests were conducted for evaluation of cognitive function. In addition, changes of CaMKIIα/CREB/BDNF and EPO/EPOR pathways in hippocampus were examined by detecting the protein expressions of CaMKIIα, p-CaMKIIα (Thr286), CREB1, p-CREB1 (Ser133), BDNF, EPO, EPOR, p-EPOR (Tyr485), STAT5, and AKT1 using western blotting assays. Also, the primary EPO-producing cells in brain (i.e. astrocytes) and EPO expression regulator HIF-2α were checked by fluorescence microscopy and western blotting assay, respectively. RESULTS Nine components in YGY extract were figured out and monitored with their contents by HPLC for the quality control of YGY extract. Biochemical and physiological measurements validated the success of induction of CRF in mice, and YGY extract significantly retarded the CRF progression and ameliorated the CRF-induced cognitive impairment. The behavioral tests showed that compared with normal control mice, CRF mice had impaired cognitive function. However, treatment of YGY extract significantly ameliorated the cognitive impairment of CRF mice. Additionally, decreased expressions of hippocampal CaMKIIα, p-CaMKIIα (Thr286), CREB1, p-CREB1 (Ser133), and BDNF were observed in the hippocampus of CRF mice, but YGY extract significantly restored these protein expressions. Moreover, hippocampal EPO, EPOR, p-EPOR (Tyr485), STAT5, AKT1, and HIF-2α, as well as the number of astrocytes in CA1 zone of hippocampus were also decreased in CRF mice, while YGY extract prominently promoted the expressions of these proteins and increased the number of astrocytes. CONCLUSIONS All the data in this study suggested that YGY extract ameliorated the cognitive impairment of CRF mice, and this amelioration was related to up-regulating the CaMKIIα/CREB/BDNF and EPO/EPOR pathways.
Collapse
Affiliation(s)
- Qing Tang
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Hui Ke
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Chao Wu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Jie Zeng
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Zhuohen Li
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Yang Liu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China
| | - Shan Feng
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China
| | - Qiang Xue
- Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of New Drug Screening from Traditional Chinese Medicine, Chongqing 400715, China; Pharmacology of Chinese Materia Medica - the Key Discipline Constructed by the State Administration of Traditional Chinese Medicine, Chongqing 400715, China.
| |
Collapse
|
677
|
Gondard E, Soto-Montenegro ML, Cassol A, Lozano AM, Hamani C. Transcranial direct current stimulation does not improve memory deficits or alter pathological hallmarks in a rodent model of Alzheimer's disease. J Psychiatr Res 2019; 114:93-98. [PMID: 31054455 DOI: 10.1016/j.jpsychires.2019.04.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022]
Abstract
Alzheimer's disease (AD) is a progressive and debilitating degenerative disorder for which there are currently no effective therapeutic options. Non-invasive neuromodulation, including transcranial direct current stimulation (tDCS), has been investigated for the treatment of cognitive symptoms in AD. Results from clinical and preclinical studies, however, have been somewhat controversial. We investigate whether tDCS delivered to triple transgenic (3xTg) AD mice improves memory deficits and mitigates the development of AD-type neuropathology. 3xTg AD mice and controls were implanted with paddle electrodes over the skull. The cathode was anterior to bregma and the anode anterior to lamda. tDCS was delivered for 20 min/day, 5 days/week over three weeks at 50 μA. Though this amplitude was lower than the one used in the preclinical literature, it generated a high current density compared to the clinical scenario. Memory testing was conducted during treatment weeks 2 and 3. Post-mortem pathological AD markers were studied. Our results show that performance of 3xTg mice in the novel object recognition and Morris water maze tests was significantly impaired compared to that of controls. In addition, AD transgenics had an increased expression of tau, phosphorylated-tau and amyloid precursor protein in the hippocampus. tDCS did not improve behavioural deficits or mitigated the development of AD neuropathology in 3xTg animals. In summary, we found that tDCS at the settings selected in our study was largely ineffective in improving memory performance or altering the expression of AD pathological hallmarks in a validated mouse model.
Collapse
Affiliation(s)
- Elise Gondard
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON, Canada
| | | | - Amanda Cassol
- Medical School, University of Passo Fundo, Passo Fundo, RS, Brazil
| | - Andres M Lozano
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON, Canada; Department of Surgery, Division of Neurosurgery, University of Toronto, Toronto, Canada
| | - Clement Hamani
- Krembil Research Institute, Toronto Western Hospital, Toronto, ON, Canada; Department of Surgery, Division of Neurosurgery, University of Toronto, Toronto, Canada; Neuroimaging Research Section, Centre for Addictions and Mental Health, Toronto, ON, Canada; Harquail Centre for Neuromodulation, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada.
| |
Collapse
|
678
|
Anderson CJ, Bredvik K, Burstein SR, Davis C, Meadows SM, Dash J, Case L, Milner TA, Kawamata H, Zuberi A, Piersigilli A, Lutz C, Manfredi G. ALS/FTD mutant CHCHD10 mice reveal a tissue-specific toxic gain-of-function and mitochondrial stress response. Acta Neuropathol 2019; 138:103-121. [PMID: 30877432 DOI: 10.1007/s00401-019-01989-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/25/2019] [Accepted: 03/08/2019] [Indexed: 12/12/2022]
Abstract
Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10), a mitochondrial protein of unknown function, cause a disease spectrum with clinical features of motor neuron disease, dementia, myopathy and cardiomyopathy. To investigate the pathogenic mechanisms of CHCHD10, we generated mutant knock-in mice harboring the mouse-equivalent of a disease-associated human S59L mutation, S55L in the endogenous mouse gene. CHCHD10S55L mice develop progressive motor deficits, myopathy, cardiomyopathy and accelerated mortality. Critically, CHCHD10 accumulates in aggregates with its paralog CHCHD2 specifically in affected tissues of CHCHD10S55L mice, leading to aberrant organelle morphology and function. Aggregates induce a potent mitochondrial integrated stress response (mtISR) through mTORC1 activation, with elevation of stress-induced transcription factors, secretion of myokines, upregulated serine and one-carbon metabolism, and downregulation of respiratory chain enzymes. Conversely, CHCHD10 ablation does not induce disease pathology or activate the mtISR, indicating that CHCHD10S55L-dependent disease pathology is not caused by loss-of-function. Overall, CHCHD10S55L mice recapitulate crucial aspects of human disease and reveal a novel toxic gain-of-function mechanism through maladaptive mtISR and metabolic dysregulation.
Collapse
|
679
|
Li F, Liu L. Comparison of kainate-induced seizures, cognitive impairment and hippocampal damage in male and female mice. Life Sci 2019; 232:116621. [PMID: 31269415 DOI: 10.1016/j.lfs.2019.116621] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/11/2019] [Accepted: 06/29/2019] [Indexed: 12/22/2022]
Abstract
Kainate (KA) mouse model induced by intraperitoneal injection has been widely used for epilepsy and neurodegeneration studies. KA elicits sustained epileptic activity in mouse brain revealed by recurrent behavioral seizures, deteriorative neurodegeneration and various neurological deficits. However, to date, the vast majority of the studies used male mice only, and few studies on the comparison of brain injury between male and female mice in this model were reported. Epidemiological studies indicate that sex may affect the susceptibility to seizure response and neurodegeneration process. Therefore, this study focused on the effect of sex difference on KA-induced recurrent seizures and mortality, locomotor activity and cognitive impairment, and hippocampal neurodegeneration and reactive gliosis in mice. Our results showed that, compared to females, adult male mice exhibited worse performance in mortality rate, severity of epileptic seizures, and cognitive impairment indicated by novel object recognition task. Unexpectedly, post-KA male and female mice underwent similar decline and recovery of locomotor activity. KA-induced neurodegeneration in the whole hippocampus, particularly in CA1 and CA3 subregions, along with the deteriorative reactive gliosis in astrocytes and microglia, was more severe in males than that in females. These data provided the direct in vivo evidence that indicates the key role of sex difference in studies with KA mouse model, and this could be beneficial for optimizing the design of future studies.
Collapse
Affiliation(s)
- Fengling Li
- Department of Pharmacy, Linyi Tumor Hospital, Linyi, Shandong 276001, China
| | - Lei Liu
- Department of Anesthesiology, Center for Translational Research in Neurodegenerative Disease, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
680
|
Shen J, Wang D, Wang X, Gupta S, Ayloo B, Wu S, Prasad P, Xiong Q, Xia J, Ge S. Neurovascular Coupling in the Dentate Gyrus Regulates Adult Hippocampal Neurogenesis. Neuron 2019; 103:878-890.e3. [PMID: 31257104 DOI: 10.1016/j.neuron.2019.05.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Newborn dentate granule cells (DGCs) are continuously generated in the adult brain. The mechanism underlying how the adult brain governs hippocampal neurogenesis remains poorly understood. In this study, we investigated how coupling of pre-existing neurons to the cerebrovascular system regulates hippocampal neurogenesis. Using a new in vivo imaging method in freely moving mice, we found that hippocampus-engaged behaviors, such as exploration in a novel environment, rapidly increased microvascular blood-flow velocity in the dentate gyrus. Importantly, blocking this exploration-elevated blood flow dampened experience-induced hippocampal neurogenesis. By imaging the neurovascular niche in combination with chemogenetic manipulation, we revealed that pre-existing DGCs actively regulated microvascular blood flow. This neurovascular coupling was linked by parvalbumin-expressing interneurons, primarily through nitric-oxide signaling. Further, we showed that insulin growth factor 1 signaling participated in functional hyperemia-induced neurogenesis. Together, our findings revealed a neurovascular coupling network that regulates experience-induced neurogenesis in the adult brain.
Collapse
Affiliation(s)
- Jia Shen
- The Program of Genetics, SUNY at Stony Brook, Stony Brook, NY 11794, USA; Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Depeng Wang
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Xinxing Wang
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Shashank Gupta
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Bhargav Ayloo
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Song Wu
- Department of Applied Mathematics and Statistics, SUNY at Stony Brook, Stony Brook, NY 11794, USA
| | - Paras Prasad
- Institute for Lasers, Photonics and Biophotonics and the Department of Chemistry, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | - Qiaojie Xiong
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA.
| | - Jun Xia
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY 14260, USA.
| | - Shaoyu Ge
- Department of Neurobiology & Behavior, SUNY at Stony Brook, Stony Brook, NY 11794, USA.
| |
Collapse
|
681
|
Ganglioside GQ1b ameliorates cognitive impairments in an Alzheimer's disease mouse model, and causes reduction of amyloid precursor protein. Sci Rep 2019; 9:8512. [PMID: 31186474 PMCID: PMC6560179 DOI: 10.1038/s41598-019-44739-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/20/2019] [Indexed: 01/01/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays crucial roles in memory impairments including Alzheimer’s disease (AD). Previous studies have reported that tetrasialoganglioside GQ1b is involved in long-term potentiation and cognitive functions as well as BDNF expression. However, in vitro and in vivo functions of GQ1b against AD has not investigated yet. Consequently, treatment of oligomeric Aβ followed by GQ1b significantly restores Aβ1–42-induced cell death through BDNF up-regulation in primary cortical neurons. Bilateral infusion of GQ1b into the hippocampus ameliorates cognitive deficits in the triple-transgenic AD mouse model (3xTg-AD). GQ1b-infused 3xTg-AD mice had substantially increased BDNF levels compared with artificial cerebrospinal fluid (aCSF)-treated 3xTg-AD mice. Interestingly, we also found that GQ1b administration into hippocampus of 3xTg-AD mice reduces Aβ plaque deposition and tau phosphorylation, which correlate with APP protein reduction and phospho-GSK3β level increase, respectively. These findings demonstrate that the tetrasialoganglioside GQ1b may contribute to a potential strategy of AD treatment.
Collapse
|
682
|
Lauterborn JC, Schultz MN, Le AA, Amani M, Friedman AE, Leach PT, Gall CM, Lynch GS, Crawley JN. Spaced training improves learning in Ts65Dn and Ube3a mouse models of intellectual disabilities. Transl Psychiatry 2019; 9:166. [PMID: 31182707 PMCID: PMC6557858 DOI: 10.1038/s41398-019-0495-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 12/29/2022] Open
Abstract
Benefits of distributed learning strategies have been extensively described in the human literature, but minimally investigated in intellectual disability syndromes. We tested the hypothesis that training trials spaced apart in time could improve learning in two distinct genetic mouse models of neurodevelopmental disorders characterized by intellectual impairments. As compared to training with massed trials, spaced training significantly improved learning in both the Ts65Dn trisomy mouse model of Down syndrome and the maternally inherited Ube3a mutant mouse model of Angelman syndrome. Spacing the training trials at 1 h intervals accelerated acquisition of three cognitive tasks by Ts65Dn mice: (1) object location memory, (2) novel object recognition, (3) water maze spatial learning. Further, (4) spaced training improved water maze spatial learning by Ube3a mice. In contrast, (5) cerebellar-mediated rotarod motor learning was not improved by spaced training. Corroborations in three assays, conducted in two model systems, replicated within and across two laboratories, confirm the strength of the findings. Our results indicate strong translational relevance of a behavioral intervention strategy for improving the standard of care in treating the learning difficulties that are characteristic and clinically intractable features of many neurodevelopmental disorders.
Collapse
Affiliation(s)
- J C Lauterborn
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M N Schultz
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - A A Le
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - M Amani
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - A E Friedman
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Harvard University, Cambridge, MA, USA
| | - P T Leach
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA
- Biogen Inc., Cambridge, MA, USA
| | - C M Gall
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - G S Lynch
- Department of Anatomy & Neurobiology, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
- Department of Psychiatry and Human Behavior, School of Medicine, University of California Irvine, Irvine, CA, 92697, USA
| | - J N Crawley
- MIND Institute, Department of Psychiatry and Behavioral Sciences, University of California Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
683
|
R von Collenberg C, Schmitt D, Rülicke T, Sendtner M, Blum R, Buchner E. An essential role of the mouse synapse-associated protein Syap1 in circuits for spontaneous motor activity and rotarod balance. Biol Open 2019; 8:bio.042366. [PMID: 31118165 PMCID: PMC6602322 DOI: 10.1242/bio.042366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Synapse-associated protein 1 (Syap1) is the mammalian homologue of synapse-associated protein of 47 kDa (Sap47) in Drosophila. Genetic deletion of Sap47 leads to deficiencies in short-term plasticity and associative memory processing in flies. In mice, Syap1 is prominently expressed in the nervous system, but its function is still unclear. We have generated Syap1 knockout mice and tested motor behaviour and memory. These mice are viable and fertile but display distinct deficiencies in motor behaviour. Locomotor activity specifically appears to be reduced in early phases when voluntary movement is initiated. On the rotarod, a more demanding motor test involving control by sensory feedback, Syap1-deficient mice dramatically fail to adapt to accelerated speed or to a change in rotation direction. Syap1 is highly expressed in cerebellar Purkinje cells and cerebellar nuclei. Thus, this distinct motor phenotype could be due to a so-far unknown function of Syap1 in cerebellar sensorimotor control. The observed motor defects are highly specific since other tests in the modified SHIRPA exam, as well as cognitive tasks like novel object recognition, Pavlovian fear conditioning, anxiety-like behaviour in open field dark-light transition and elevated plus maze do not appear to be affected in Syap1 knockout mice. Summary: Knockout of the Syap1 gene in mice causes a distinct motor behaviour phenotype characterised by reduced initial locomotor activity and impaired rotarod performance.
Collapse
Affiliation(s)
- Cora R von Collenberg
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Dominique Schmitt
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Thomas Rülicke
- Institute of Laboratory Animal Science, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Robert Blum
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| | - Erich Buchner
- Institute of Clinical Neurobiology, University Hospital Würzburg, Versbacher Str. 5, 97078 Würzburg, Germany
| |
Collapse
|
684
|
Wang J, Xie R, Kou X, Liu Y, Qi C, Liu R, You W, Gao J, Gao X. A protein phosphatase 2A deficit in the hippocampal CA1 area impairs memory extinction. Mol Brain 2019; 12:51. [PMID: 31113458 PMCID: PMC6528246 DOI: 10.1186/s13041-019-0469-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 04/30/2019] [Indexed: 01/17/2023] Open
Abstract
Protein phosphorylation plays an important role in learning and memory. Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase involved in the regulation of neural synaptic plasticity. Here, to determine if PP2A is necessary for successful learning and memory, we have utilized a Tg (Camk2a-cre) T29–2Stl mice to specific knock down the expression of hippocampal PP2A in mice. By analysing behavioural, we observed that loss of PP2A in the hippocampal CA1 area did not affect the formation of memory but impaired contextual fear memory extinction. We use the electrophysiological recording to find the synaptic mechanisms. The results showed that the basic synapse transmission and synaptic plasticity of PP2A conditional knockout (CKO) mice were impaired. Moreover, PP2A CKO mice exhibited a saturating long-term potentiation inducted by strong theta burst stimulation but no depotentiation after low-frequency stimulation. Taken together, our results provide the evidence that PP2A is involved in synaptic transmission and hippocampus-dependent memory extinction.
Collapse
Affiliation(s)
- Jing Wang
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Ran Xie
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Xiaolin Kou
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Yu Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Cui Qi
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Rui Liu
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Weiyan You
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China
| | - Jun Gao
- Department of Neurobiology, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Xiang Gao
- Model Animal Research Center and MOE Key Laboratory of Model Animals for Disease Study, Nanjing University, Nanjing, 210093, Jiangsu, China.
| |
Collapse
|
685
|
Geraghty AC, Gibson EM, Ghanem RA, Greene JJ, Ocampo A, Goldstein AK, Ni L, Yang T, Marton RM, Paşca SP, Greenberg ME, Longo FM, Monje M. Loss of Adaptive Myelination Contributes to Methotrexate Chemotherapy-Related Cognitive Impairment. Neuron 2019; 103:250-265.e8. [PMID: 31122677 DOI: 10.1016/j.neuron.2019.04.032] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 01/29/2019] [Accepted: 04/22/2019] [Indexed: 01/05/2023]
Abstract
Activity-dependent myelination is thought to contribute to adaptive neurological function. However, the mechanisms by which activity regulates myelination and the extent to which myelin plasticity contributes to non-motor cognitive functions remain incompletely understood. Using a mouse model of chemotherapy-related cognitive impairment (CRCI), we recently demonstrated that methotrexate (MTX) chemotherapy induces complex glial dysfunction for which microglial activation is central. Here, we demonstrate that remote MTX exposure blocks activity-regulated myelination. MTX decreases cortical Bdnf expression, which is restored by microglial depletion. Bdnf-TrkB signaling is a required component of activity-dependent myelination. Oligodendrocyte precursor cell (OPC)-specific TrkB deletion in chemotherapy-naive mice results in impaired cognitive behavioral performance. A small-molecule TrkB agonist rescues both myelination and cognitive impairment after MTX chemotherapy. This rescue after MTX depends on intact TrkB expression in OPCs. Taken together, these findings demonstrate a molecular mechanism required for adaptive myelination that is aberrant in CRCI due to microglial activation.
Collapse
Affiliation(s)
- Anna C Geraghty
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Erin M Gibson
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Reem A Ghanem
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Jacob J Greene
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Alfonso Ocampo
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Andrea K Goldstein
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Lijun Ni
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Tao Yang
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Rebecca M Marton
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Sergiu P Paşca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | | | - Frank M Longo
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Stanford University, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
686
|
Soria B, Martin-Montalvo A, Aguilera Y, Mellado-Damas N, López-Beas J, Herrera-Herrera I, López E, Barcia JA, Alvarez-Dolado M, Hmadcha A, Capilla-González V. Human Mesenchymal Stem Cells Prevent Neurological Complications of Radiotherapy. Front Cell Neurosci 2019; 13:204. [PMID: 31156392 PMCID: PMC6532528 DOI: 10.3389/fncel.2019.00204] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/24/2019] [Indexed: 12/27/2022] Open
Abstract
Radiotherapy is a highly effective tool for the treatment of brain cancer. However, radiation also causes detrimental effects in the healthy tissue, leading to neurocognitive sequelae that compromise the quality of life of brain cancer patients. Despite the recognition of this serious complication, no satisfactory solutions exist at present. Here we investigated the effects of intranasal administration of human mesenchymal stem cells (hMSCs) as a neuroprotective strategy for cranial radiation in mice. Our results demonstrated that intranasally delivered hMSCs promote radiation-induced brain injury repair, improving neurological function. This intervention confers protection against inflammation, oxidative stress, and neuronal loss. hMSC administration reduces persistent activation of damage-induced c-AMP response element-binding signaling in irradiated brains. Furthermore, hMSC treatment did not compromise the survival of glioma-bearing mice. Our findings encourage the therapeutic use of hMSCs as a non-invasive approach to prevent neurological complications of radiotherapy, improving the quality of life of brain tumor patients.
Collapse
Affiliation(s)
- Bernat Soria
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Alejandro Martin-Montalvo
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| | - Yolanda Aguilera
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| | - Nuria Mellado-Damas
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| | - Javier López-Beas
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| | - Isabel Herrera-Herrera
- Department of Neuroradiology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Escarlata López
- Department of Radiation Oncology, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Juan A Barcia
- Service of Neurosurgery, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Department of Surgery, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Alvarez-Dolado
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| | - Abdelkrim Hmadcha
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Vivian Capilla-González
- Department of Regeneration and Cell Therapy, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide - University of Seville, CSIC, Seville, Spain
| |
Collapse
|
687
|
Fronza MG, Baldinotti R, Martins MC, Goldani B, Dalberto BT, Kremer FS, Begnini K, Pinto LDS, Lenardão EJ, Seixas FK, Collares T, Alves D, Savegnago L. Rational design, cognition and neuropathology evaluation of QTC-4-MeOBnE in a streptozotocin-induced mouse model of sporadic Alzheimer's disease. Sci Rep 2019; 9:7276. [PMID: 31086208 PMCID: PMC6513848 DOI: 10.1038/s41598-019-43532-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 04/23/2019] [Indexed: 11/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial pathology characterized by amyloid deposits, neurofibrillary formation, oxidative stress and cholinergic system dysfunction. In this sense, here we report the rational design of a multi-target directed ligand (MTDL) for AD based on virtual screening and bioinformatic analyses, exploring the molecular targets β-secretase (BACE-1), glycogen synthase kinase-3β (GSK-3β) and acetylcholinesterase (AChE). After this screening, the compound with higher molecular docking affinity was selected, the 1-(7-chloroquinolin-4-yl)-N-(4-methoxybenzyl)-5-methyl-1H-1,2,3-triazole-4 carboxamide(QTC-4-MeOBnE). To further our studies, the protective effect of QTC-4-MeOBnE (0.1 and 1 mg/kg for 20 days) on STZ-induced sporadic AD mice was determined. QTC-4-MeOBnE pretreatment attenuated cognitive and memory deficit induced by STZ in an object recognition test, Y-maze, social recognition test and step-down passive avoidance. The mechanisms underlying this action might be attributed to the reduction of lipid peroxidation and reactive species formation in the prefrontal cortex and hippocampus of mice submitted to STZ. In addition, QTC-4-MeOBnE pretreatment abolished the up-regulation of AChE activity and the overexpression of GSK 3β and genes involved in amyloid cascade such as BACE-1, protein precursor amyloid, у-secretase, induced by STZ. Moreover, toxicological parameters were not modified by QTC-4-MeOBnE chronic treatment. This evidence suggests that QTC-4-MeOBnE exerts its therapeutic effect through multiple pathways involved in AD.
Collapse
Affiliation(s)
- Mariana G Fronza
- Research Group on Neurobiotechnology - GPN, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Rodolfo Baldinotti
- Research Group on Neurobiotechnology - GPN, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Maria Clara Martins
- Research Group on Neurobiotechnology - GPN, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Bruna Goldani
- Laboratory of Clean Organic Synthesis - LASOL, CCQFA, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Bianca Thaís Dalberto
- Laboratory of Clean Organic Synthesis - LASOL, CCQFA, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Frederico Schmitt Kremer
- Laboratory of Bioinformatics and Proteomics - BIOPRO-LAB, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Karine Begnini
- Oncology Research Group - GPO, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Luciano da Silva Pinto
- Laboratory of Bioinformatics and Proteomics - BIOPRO-LAB, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Eder João Lenardão
- Laboratory of Clean Organic Synthesis - LASOL, CCQFA, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Fabiana K Seixas
- Oncology Research Group - GPO, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Tiago Collares
- Oncology Research Group - GPO, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis - LASOL, CCQFA, Federal University of Pelotas, Pelotas, RS, Brazil
| | - Lucielli Savegnago
- Research Group on Neurobiotechnology - GPN, CDTec, Federal University of Pelotas, UFPel, Pelotas, RS, Brazil.
| |
Collapse
|
688
|
Yousef H, Czupalla CJ, Lee D, Chen MB, Burke AN, Zera KA, Zandstra J, Berber E, Lehallier B, Mathur V, Nair RV, Bonanno LN, Yang AC, Peterson T, Hadeiba H, Merkel T, Körbelin J, Schwaninger M, Buckwalter MS, Quake SR, Butcher EC, Wyss-Coray T. Aged blood impairs hippocampal neural precursor activity and activates microglia via brain endothelial cell VCAM1. Nat Med 2019; 25:988-1000. [PMID: 31086348 PMCID: PMC6642642 DOI: 10.1038/s41591-019-0440-4] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/11/2019] [Indexed: 01/25/2023]
Abstract
An aged circulatory environment can activate microglia, reduce neural precursor cell activity, and impair cognition in mice. We hypothesized that brain endothelial cells (BECs) mediate at least some of these effects. We observe BECs in the aged mouse hippocampus express an inflammatory transcriptional profile with focal upregulation of Vascular Cell Adhesion Molecule 1 (VCAM1), a protein that facilitates vascular-immune cell interactions. Concomitantly, the shed, soluble form of VCAM1 is prominently increased in plasma of aged humans and mice, and their plasma is sufficient to increase VCAM1 expression in cultured BECs and young mouse hippocampi. Systemic anti-VCAM1 antibody or genetic ablation of VCAM1 in BECs counteracts the detrimental effects of aged plasma on young brains and reverses aging aspects including microglial reactivity and cognitive deficits in old mouse brains. Together, these findings establish brain endothelial VCAM1 at the blood-brain barrier (BBB) as a possible target to treat age-related neurodegeneration.
Collapse
Affiliation(s)
- Hanadie Yousef
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Cathrin J Czupalla
- VA Palo Alto Health Care System, Palo Alto, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Davis Lee
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Michelle B Chen
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford, CA, USA
| | - Ashley N Burke
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Judith Zandstra
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Elisabeth Berber
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Vidhu Mathur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ramesh V Nair
- Stanford Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Liana N Bonanno
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Andrew C Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Departments of Bioengineering and Applied Physics, Stanford University, Stanford, CA, USA
| | - Todd Peterson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Husein Hadeiba
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Taylor Merkel
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Jakob Körbelin
- Section of Pneumology, Department of Oncology, Hematology and Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lubeck, Lubeck, Germany
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Stephen R Quake
- Departments of Bioengineering and Applied Physics, Stanford University, Stanford, CA, USA.,Chan Zuckerberg Biohub, Stanford, CA, USA
| | - Eugene C Butcher
- VA Palo Alto Health Care System, Palo Alto, CA, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,VA Palo Alto Health Care System, Palo Alto, CA, USA. .,Palo Alto Veterans Institute for Research, Palo Alto, CA, USA. .,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
689
|
High dose of dexamethasone protects against EAE-induced motor deficits but impairs learning/memory in C57BL/6 mice. Sci Rep 2019; 9:6673. [PMID: 31040362 PMCID: PMC6491620 DOI: 10.1038/s41598-019-43217-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/17/2019] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and neuroinflammatory disease characterized by demyelination of the Central Nervous System. Immune cells activation and release of pro-inflammatory cytokines play a crucial role in the disease modulation, decisively contributing to the neurodegeneration observed in MS and the experimental autoimmune encephalomyelitis (EAE), the widely used MS animal model. Synthetic glucocorticoids, commonly used to treat the MS attacks, have controversial effects on neuroinflammation and cognition. We sought to verify the influence of dexamethasone (DEX) on the EAE progression and on EAE-induced cognitive deficits. In myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE female mice, treated once with DEX (50 mg/kg) or not, on the day of immunization, DEX decreased EAE-induced motor clinical scores, infiltrating cells in the spinal cord and delayed serum corticosterone peak. At the asymptomatic phase (8-day post-immunization), DEX did not protected from the EAE-induced memory consolidation deficits, which were accompanied by increased glucocorticoid receptor (GR) activity and decreased EGR-1 expression in the hippocampus. Blunting hippocampal GR genomic activation with DnGR vectors prevented DEX effects on EAE-induced memory impairment. These data suggest that, although DEX improves clinical signs, it decreases cognitive and memory capacity by diminishing neuronal activity and potentiating some aspects of neuroinflammation in EAE.
Collapse
|
690
|
Ge X, Li W, Huang S, Yin Z, Yang M, Han Z, Han Z, Chen F, Wang H, Lei P, Zhang J. Increased miR-21-3p in Injured Brain Microvascular Endothelial Cells after Traumatic Brain Injury Aggravates Blood–Brain Barrier Damage by Promoting Cellular Apoptosis and Inflammation through Targeting MAT2B. J Neurotrauma 2019; 36:1291-1305. [PMID: 29695199 DOI: 10.1089/neu.2018.5728] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Xintong Ge
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenzhu Li
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shan Huang
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyu Yin
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengchen Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Zhenying Han
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Zhaoli Han
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fanglian Chen
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| | - Haichen Wang
- Department of Neurology, Duke University Medical Center, Durham, North Carolina
| | - Ping Lei
- Laboratory of Neuro-Trauma and Neurodegenerative Disorders, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education, Tianjin, China
| |
Collapse
|
691
|
Fragopoulou AF, Qian Y, Heijtz RD, Forssberg H. Can Neonatal Systemic Inflammation and Hypoxia Yield a Cerebral Palsy-Like Phenotype in Periadolescent Mice? Mol Neurobiol 2019; 56:6883-6900. [PMID: 30941732 PMCID: PMC6728419 DOI: 10.1007/s12035-019-1548-8] [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: 08/07/2018] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
Cerebral palsy (CP) is one of the most common childhood-onset motor disabilities, attributed to injuries of the immature brain in the foetal or early postnatal period. The underlying mechanisms are poorly understood, rendering prevention and treatment strategies challenging. The aim of the present study was to establish a mouse model of CP for preclinical assessment of new interventions. For this purpose, we explored the impact of a double neonatal insult (i.e. systemic inflammation combined with hypoxia) on behavioural and cellular outcomes relevant to CP during the prepubertal to adolescent period of mice. Pups were subjected to intraperitoneal lipopolysaccharide (LPS) injections from postnatal day (P) 3 to P6 followed by hypoxia at P7. Gene expression analysis at P6 revealed a strong inflammatory response in a brain region-dependent manner. A comprehensive battery of behavioural assessments performed between P24 and P47 showed impaired limb placement and coordination when walking on a horizontal ladder in both males and females. Exposed males also displayed impaired performance on a forelimb skilled reaching task, altered gait pattern and increased exploratory activity. Exposed females showed a reduction in grip strength and traits of anxiety-like behaviour. These behavioural alterations were not associated with gross morphological changes, white matter lesions or chronic inflammation in the brain. Our results indicate that the neonatal double-hit with LPS and hypoxia can induce subtle long-lasting deficits in motor learning and fine motor skills, which partly reflect the symptoms of children with CP who have mild gross and fine motor impairments.
Collapse
Affiliation(s)
- Adamantia F Fragopoulou
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden. .,Department of Women's and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.
| | - Yu Qian
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rochellys Diaz Heijtz
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 171 77, Stockholm, Sweden.,INSERM U1239, University of Rouen Normandy, 76130, Mont-Saint-Aignan, France
| | - Hans Forssberg
- Department of Women's and Children's Health, Karolinska Institutet, 171 76, Stockholm, Sweden.
| |
Collapse
|
692
|
Cognitive dysfunction in mice with passively induced MuSK antibody seropositive myasthenia gravis. J Neurol Sci 2019; 399:15-21. [DOI: 10.1016/j.jns.2019.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/23/2019] [Accepted: 02/01/2019] [Indexed: 11/19/2022]
|
693
|
Shan W, Li J, Xu W, Li H, Zuo Z. Critical role of UQCRC1 in embryo survival, brain ischemic tolerance and normal cognition in mice. Cell Mol Life Sci 2019; 76:1381-1396. [PMID: 30666338 PMCID: PMC6421091 DOI: 10.1007/s00018-019-03007-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022]
Abstract
Ubiquinol cytochrome c reductase core protein I (UQCRC1) is a component of the complex III in the respiratory chain. Its biological functions are unknown. Here, we showed that knockout of UQCRC1 led to embryonic lethality. Disrupting one UQCRC1 allele in mice (heterozygous mice) of both sexes did not affect their growth but reduced UQCRC1 mRNA and protein in the brain. These mice had decreased complex III formation, complex III activity and ATP content in the brain at baseline. They developed worsened neurological outcome after brain ischemia/hypoxia or focal brain ischemia compared with wild-type mice. The ischemic cerebral cortex of the heterozygous mice had decreased mitochondrial membrane potential and ATP content as well as increased free radicals. Also, the heterozygous mice performed poorly in the Barnes maze and novel object recognition tests. Finally, UQCRC1 was expressed abundantly in neurons and astrocytes. These results suggest a critical role of UQCRC1 in embryo survival. UQCRC1 may also be important by forming the complex III to maintain normal brain ischemic tolerance, learning and memory.
Collapse
Affiliation(s)
- Weiran Shan
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA
| | - Jun Li
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA
| | - Wenhao Xu
- Genetically Engineered Murine Model Core, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Hong Li
- Department of Anesthesiology, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia Health System, 1 Hospital Drive, PO Box 800710, Charlottesville, VA, 22908-0710, USA.
- Department of Neuroscience and Neurological Surgery, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
694
|
γ-Oryzanol Improves Cognitive Function and Modulates Hippocampal Proteome in Mice. Nutrients 2019; 11:nu11040753. [PMID: 30935111 PMCID: PMC6520752 DOI: 10.3390/nu11040753] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/18/2023] Open
Abstract
Rice (Oryza sativa L.) is the richest source of γ-oryzanol, a compound endowed with antioxidant and anti-inflammatory properties. γ-Oryzanol has been demonstrated to cross the blood-brain barrier in intact form and exert beneficial effects on brain function. This study aimed to clarify the effects of γ-oryzanol in the hippocampus in terms of cognitive function and protein expression. Adult mice were administered with γ-oryzanol 100 mg/kg or vehicle (control) once a day for 21 consecutive days following which cognitive behavior and hippocampal proteome were investigated. Cognitive tests using novel object recognition and Y-maze showed that long-term consumption of γ-oryzanol improves cognitive function in mice. To investigate the hippocampal proteome modulated by γ-oryzanol, 2D-difference gel electrophoresis (2D-DIGE) was performed. Interestingly, we found that γ-oryzanol modulates quantitative changes of proteins involved in synaptic plasticity and neuronal trafficking, neuroprotection and antioxidant activity, and mitochondria and energy metabolism. These findings suggested γ-oryzanol as a natural compound able to maintain and reinforce brain function. Although more intensive studies are needed, we propose γ-oryzanol as a putative dietary phytochemical for preserving brain reserve, the ability to tolerate age-related changes, thereby preventing clinical symptoms or signs of neurodegenerative diseases.
Collapse
|
695
|
Mastinu A, Bonini SA, Rungratanawanich W, Aria F, Marziano M, Maccarinelli G, Abate G, Premoli M, Memo M, Uberti D. Gamma-oryzanol Prevents LPS-induced Brain Inflammation and Cognitive Impairment in Adult Mice. Nutrients 2019; 11:nu11040728. [PMID: 30934852 PMCID: PMC6520753 DOI: 10.3390/nu11040728] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Rice (Oryza sativa L.) is the main food source for more than half of humankind. Rice is rich in phytochemicals and antioxidants with several biological activities; among these compounds, the presence of γ-oryzanol is noteworthy. The present study aims to explore the effects of γ-oryzanol on cognitive performance in a mouse model of neuroinflammation and cognitive alterations. Methods: Mice received 100 mg/kg γ-oryzanol (ORY) or vehicle once daily for 21 consecutive days and were then exposed to an inflammatory stimulus elicited by lipopolysaccharide (LPS). A novel object recognition test and mRNA expression of antioxidant and neuroinflammatory markers in the hippocampus were evaluated. Results: ORY treatment was able to improve cognitive performance during the neuroinflammatory response. Furthermore, phase II antioxidant enzymes such as heme oxygenase-1 (HO-1) and NADPH-dehydrogenase-quinone-1 (NQO1) were upregulated in the hippocampi of ORY and ORY+LPS mice. Lastly, γ-oryzanol showed a strong anti-inflammatory action by downregulating inflammatory genes after LPS treatment. Conclusion: These results suggest that chronic consumption of γ-oryzanol can revert the LPS-induced cognitive and memory impairments by promoting hippocampal antioxidant and anti-inflammatory molecular responses.
Collapse
Affiliation(s)
- Andrea Mastinu
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Sara Anna Bonini
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Wiramon Rungratanawanich
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Francesca Aria
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Mariagrazia Marziano
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Giuseppina Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Giulia Abate
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Marika Premoli
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Maurizio Memo
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| | - Daniela Uberti
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy.
| |
Collapse
|
696
|
Edfawy M, Guedes JR, Pereira MI, Laranjo M, Carvalho MJ, Gao X, Ferreira PA, Caldeira G, Franco LO, Wang D, Cardoso AL, Feng G, Carvalho AL, Peça J. Abnormal mGluR-mediated synaptic plasticity and autism-like behaviours in Gprasp2 mutant mice. Nat Commun 2019; 10:1431. [PMID: 30926797 PMCID: PMC6440958 DOI: 10.1038/s41467-019-09382-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by dysfunction in social interactions, stereotypical behaviours and high co-morbidity with intellectual disability. A variety of syndromic and non-syndromic neurodevelopmental disorders have been connected to alterations in metabotropic glutamate receptor (mGluR) signalling. These receptors contribute to synaptic plasticity, spine maturation and circuit development. Here, we investigate the physiological role of Gprasp2, a gene linked to neurodevelopmental disabilities and involved in the postendocytic sorting of G-protein-coupled receptors. We show that Gprasp2 deletion leads to ASD-like behaviour in mice and alterations in synaptic communication. Manipulating the levels of Gprasp2 bidirectionally modulates the surface availability of mGluR5 and produces alterations in dendritic complexity, spine density and synaptic maturation. Loss of Gprasp2 leads to enhanced hippocampal long-term depression, consistent with facilitated mGluR-dependent activation. These findings demonstrate a role for Gprasp2 in glutamatergic synapses and suggest a possible mechanism by which this gene is linked to neurodevelopmental diseases.
Collapse
Affiliation(s)
- Mohamed Edfawy
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Joana R Guedes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Marta I Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mariana Laranjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mário J Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Xian Gao
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai, 200062, China.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Pedro A Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Gladys Caldeira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Lara O Franco
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ana Luisa Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai, 200062, China.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ana Luisa Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, 3004-517, Coimbra, Portugal
| | - João Peça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.
| |
Collapse
|
697
|
Leem YH, Park JS, Chang H, Park J, Kim HS. Exercise Prevents Memory Consolidation Defects Via Enhancing Prolactin Responsiveness of CA1 Neurons in Mice Under Chronic Stress. Mol Neurobiol 2019; 56:6609-6625. [DOI: 10.1007/s12035-019-1560-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 03/13/2019] [Indexed: 12/21/2022]
|
698
|
Serotonin receptor HTR6-mediated mTORC1 signaling regulates dietary restriction-induced memory enhancement. PLoS Biol 2019; 17:e2007097. [PMID: 30883547 PMCID: PMC6438579 DOI: 10.1371/journal.pbio.2007097] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 03/28/2019] [Accepted: 03/08/2019] [Indexed: 12/21/2022] Open
Abstract
Dietary restriction (DR; sometimes called calorie restriction) has profound beneficial effects on physiological, psychological, and behavioral outcomes in animals and in humans. We have explored the molecular mechanism of DR-induced memory enhancement and demonstrate that dietary tryptophan-a precursor amino acid for serotonin biosynthesis in the brain-and serotonin receptor 5-hydroxytryptamine receptor 6 (HTR6) are crucial in mediating this process. We show that HTR6 inactivation diminishes DR-induced neurological alterations, including reduced dendritic complexity, increased spine density, and enhanced long-term potentiation (LTP) in hippocampal neurons. Moreover, we find that HTR6-mediated mechanistic target of rapamycin complex 1 (mTORC1) signaling is involved in DR-induced memory improvement. Our results suggest that the HTR6-mediated mTORC1 pathway may function as a nutrient sensor in hippocampal neurons to couple memory performance to dietary intake.
Collapse
|
699
|
Martorell AJ, Paulson AL, Suk HJ, Abdurrob F, Drummond GT, Guan W, Young JZ, Kim DNW, Kritskiy O, Barker SJ, Mangena V, Prince SM, Brown EN, Chung K, Boyden ES, Singer AC, Tsai LH. Multi-sensory Gamma Stimulation Ameliorates Alzheimer's-Associated Pathology and Improves Cognition. Cell 2019; 177:256-271.e22. [PMID: 30879788 DOI: 10.1016/j.cell.2019.02.014] [Citation(s) in RCA: 442] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 06/30/2018] [Accepted: 02/11/2019] [Indexed: 01/06/2023]
Abstract
We previously reported that inducing gamma oscillations with a non-invasive light flicker (gamma entrainment using sensory stimulus or GENUS) impacted pathology in the visual cortex of Alzheimer's disease mouse models. Here, we designed auditory tone stimulation that drove gamma frequency neural activity in auditory cortex (AC) and hippocampal CA1. Seven days of auditory GENUS improved spatial and recognition memory and reduced amyloid in AC and hippocampus of 5XFAD mice. Changes in activation responses were evident in microglia, astrocytes, and vasculature. Auditory GENUS also reduced phosphorylated tau in the P301S tauopathy model. Furthermore, combined auditory and visual GENUS, but not either alone, produced microglial-clustering responses, and decreased amyloid in medial prefrontal cortex. Whole brain analysis using SHIELD revealed widespread reduction of amyloid plaques throughout neocortex after multi-sensory GENUS. Thus, GENUS can be achieved through multiple sensory modalities with wide-ranging effects across multiple brain areas to improve cognitive function.
Collapse
Affiliation(s)
- Anthony J Martorell
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Abigail L Paulson
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Ho-Jun Suk
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; MIT Media Lab, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Harvard-MIT Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Fatema Abdurrob
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gabrielle T Drummond
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Webster Guan
- Department of Chemical Engineering, MIT, Cambridge, MA, USA
| | - Jennie Z Young
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David Nam-Woo Kim
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Oleg Kritskiy
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Scarlett J Barker
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Vamsi Mangena
- Harvard-MIT Health Sciences and Technology, Cambridge, MA 02139, USA
| | - Stephanie M Prince
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Emery N Brown
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Kwanghun Chung
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, MIT, Cambridge, MA, USA; Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA
| | - Edward S Boyden
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; MIT Media Lab, Departments of Biological Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Annabelle C Singer
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02139, USA.
| |
Collapse
|
700
|
Datta S, Samanta D, Tiwary B, Chaudhuri AG, Chakrabarti N. Sex and estrous cycle dependent changes in locomotor activity, anxiety and memory performance in aged mice after exposure of light at night. Behav Brain Res 2019; 365:198-209. [PMID: 30853396 DOI: 10.1016/j.bbr.2019.03.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 03/06/2019] [Accepted: 03/06/2019] [Indexed: 12/12/2022]
Abstract
Light-at-night (LAN) can affect mammalian behaviour. But, the effects of LAN on aged rodents remain undefined yet. In the present investigation, aged Swiss Albino mice, habituated in regular light-dark cycle, were exposed to bright-light-pulse (1-h) at night on the day of study followed by experimentations for assessment of locomotor activities in the open field, anxiety in the elevated plus maze and short-term memory for novel object recognition (NOR) in the habituated field. Under without-bright-light exposure, (a) aged proestrous females showed greater locomotor activities and less anxiety than in aged diestrous females, (b) aged males showed locomotor activities and anxiety level similar to aged diestrous females and aged proestrous females respectively and (c) all animals failed to retain in object discrimination memory. LAN exposure exhibited the continual failure of such retention of memory while animals showed free and spontaneous exploration with thigmotactic behaviour having no object bias and/or phobia, but time stay in objects by animals altered variably among sexes and stages of estrous cycle. Overall, the LAN caused (a) diminution in locomotor activities, rise in anxiety and failure of memory for recognition of both familiar and novel objects in aged proestrous females, (b) hyperlocomotor activities and reduction in anxiety in both males and diestrous females with the failure of memory for recognition of novel objects only in aged males while diestrous females showed enhanced exploration time to both objects during NOR. Thus, nocturnal behaviour of aged mice varies with sex and estrous cycle and light acts differentially on them.
Collapse
Affiliation(s)
- Siddhartha Datta
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India; UGC-CPEPA Centre for "Electro-physiological and Neuro-imaging studies including Mathematical Modelling", University of Calcutta, Kolkata, West Bengal, India.
| | - Diptaman Samanta
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India.
| | - Basant Tiwary
- Centre for Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, India.
| | | | - Nilkanta Chakrabarti
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India; UGC-CPEPA Centre for "Electro-physiological and Neuro-imaging studies including Mathematical Modelling", University of Calcutta, Kolkata, West Bengal, India; S.N. Pradhan Centre for Neurosciences, University of Calcutta, Kolkata, West Bengal, India.
| |
Collapse
|