1
|
Hanson JE, Yuan H, Perszyk RE, Banke TG, Xing H, Tsai MC, Menniti FS, Traynelis SF. Therapeutic potential of N-methyl-D-aspartate receptor modulators in psychiatry. Neuropsychopharmacology 2024; 49:51-66. [PMID: 37369776 PMCID: PMC10700609 DOI: 10.1038/s41386-023-01614-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 05/15/2023] [Indexed: 06/29/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate a slow component of excitatory synaptic transmission, are widely distributed throughout the central nervous system, and regulate synaptic plasticity. NMDA receptor modulators have long been considered as potential treatments for psychiatric disorders including depression and schizophrenia, neurodevelopmental disorders such as Rett Syndrome, and neurodegenerative conditions such as Alzheimer's disease. New interest in NMDA receptors as therapeutic targets has been spurred by the findings that certain inhibitors of NMDA receptors produce surprisingly rapid and robust antidepressant activity by a novel mechanism, the induction of changes in the brain that well outlast the presence of drug in the body. These findings are driving research into an entirely new paradigm for using NMDA receptor antagonists in a host of related conditions. At the same time positive allosteric modulators of NMDA receptors are being pursued for enhancing synaptic function in diseases that feature NMDA receptor hypofunction. While there is great promise, developing the therapeutic potential of NMDA receptor modulators must also navigate the potential significant risks posed by the use of such agents. We review here the emerging pharmacology of agents that target different NMDA receptor subtypes, offering new avenues for capturing the therapeutic potential of targeting this important receptor class.
Collapse
Affiliation(s)
- Jesse E Hanson
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hao Xing
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech Inc., South San Francisco, CA, 94080, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
2
|
Mollusky A, Reynolds-Lallement N, Lee D, Zhong JY, Magnusson KR. Investigating the effects of age and prior military service on fluid and crystallized cognitive functions using virtual morris water maze (vMWM) and NIH Toolbox tasks. Arch Gerontol Geriatr 2024; 116:105156. [PMID: 37604015 DOI: 10.1016/j.archger.2023.105156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/04/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023]
Abstract
Much of current knowledge of aging involves war veterans and research about age-related cognitive changes in veterans involves generalized or single function tests or health or neurological disorders. The current study examined military service within the context of comparisons of young and old humans involving generally healthy individuals to address normal age-associated cognitive changes. Adult participants included 11 young females (8 non-veterans; 3 veterans; 21-31 years), 5 young males (non-veterans, 21-24 years), 9 older females (non-veterans, 62-80 years), and 21 older males (11 non-veterans; 10 veterans; 60-86 years). They were tested in virtual Morris water maze (vMWM) tasks, which were designed to test spatial learning, cognitive flexibility and working memory, similar to rodent studies, and were validated by correlations with specific NIH Toolbox (NIH-TB) Cognitive Battery or Wechsler Memory Scale (WMS) Logical Memory I and II tests. Significant age-related deficits were seen on multiple vMWM tasks and NIH-TB fluid cognition tasks. Among older males, vMWM tasks appeared to be more sensitive, based on finding statistical differences, to prior military service than NIH Toolbox tasks. Compared with male non-veterans of comparable age and younger, older male veterans exhibited significant deficits in spatial learning, cognitive flexibility, and working memory on vMWM tasks. Our findings support continued development and characterization of vMWM tasks that are comparable between rodents and humans for translating aging interventions between species, and provide impetus for larger investigations examining the extent to which prior military service can serve as a "hidden" variable in normal biological declines of cognitive functions.
Collapse
Affiliation(s)
- Adina Mollusky
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, United States; Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States
| | - Nadjalisse Reynolds-Lallement
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, United States; Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States
| | - Dylan Lee
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, United States; Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States
| | - Jimmy Y Zhong
- Department of Psychology, School of Social and Health Sciences, James Cook University, Australia (Singapore campus), Singapore 387380, Singapore; College of Healthcare Sciences, James Cook University, Australia (Singapore campus), Singapore 387380, Singapore; Georgia State/Georgia Tech Center for Advanced Brain Imaging (CABI), Georgia Institute of Technology, Atlanta, GA 30318, United States
| | - Kathy R Magnusson
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, United States; Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, United States.
| |
Collapse
|
3
|
Ge Y, Wang YT. GluN2B-containing NMDARs in the mammalian brain: pharmacology, physiology, and pathology. Front Mol Neurosci 2023; 16:1190324. [PMID: 37324591 PMCID: PMC10264587 DOI: 10.3389/fnmol.2023.1190324] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) is critical for promoting physiological synaptic plasticity and neuronal viability. As a major subpopulation of the NMDAR, the GluN2B subunit-containing NMDARs have distinct pharmacological properties, physiological functions, and pathological relevance to neurological diseases compared with other NMDAR subtypes. In mature neurons, GluN2B-containing NMDARs are likely expressed as both diheteromeric and triheteromeric receptors, though the functional importance of each subpopulation has yet to be disentangled. Moreover, the C-terminal region of the GluN2B subunit forms structural complexes with multiple intracellular signaling proteins. These protein complexes play critical roles in both activity-dependent synaptic plasticity and neuronal survival and death signaling, thus serving as the molecular substrates underlying multiple physiological functions. Accordingly, dysregulation of GluN2B-containing NMDARs and/or their downstream signaling pathways has been implicated in neurological diseases, and various strategies to reverse these deficits have been investigated. In this article, we provide an overview of GluN2B-containing NMDAR pharmacology and its key physiological functions, highlighting the importance of this receptor subtype during both health and disease states.
Collapse
Affiliation(s)
- Yang Ge
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Yegla B, Rani A, Kumar A. Viral vector-mediated upregulation of serine racemase expression in medial prefrontal cortex improves learning and synaptic function in middle age rats. Aging (Albany NY) 2023; 15:2433-2449. [PMID: 37052995 PMCID: PMC10120901 DOI: 10.18632/aging.204652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
An age-associated decrease in N-methyl-D-aspartate receptor (NMDAR)-mediated synaptic function contributes to impaired synaptic plasticity and is associated with cognitive impairments. Levels of serine racemase (SR), an enzyme that synthesizes D-serine, an NMDAR co-agonist, decline with age. Thus, enhancing NMDAR function via increased SR expression in middle age, when subtle declines in cognition emerge, was predicted to enhance performance on a prefrontal cortex-mediated task sensitive to aging. Middle-aged (~12 mo) male Fischer-344 rats were injected bilaterally in the medial prefrontal cortex (mPFC) with viral vector (LV), SR (LV-SR) or control (LV-GFP). Rats were trained on the operant attentional set-shift task (AST) to examine cognitive flexibility and attentional function. LV-SR rats exhibited a faster rate of learning compared to controls during visual discrimination of the AST. Extradimensional set shifting and reversal were not impacted. Immunohistochemical analyses demonstrated that LV-SR significantly increased SR expression in the mPFC. Electrophysiological characterization of synaptic transmission in the mPFC slices obtained from LV-GFP and LV-SR animals indicated a significant increase in isolated NMDAR-mediated synaptic responses in LV-SR slices. Thus, results of the current study demonstrated that prefrontal SR upregulation in middle age rats can improve learning of task contingencies for visual discrimination and increase glutamatergic synaptic transmission, including NMDAR activity.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Asha Rani
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
5
|
El Mahmoudi N, Laurent C, Péricat D, Watabe I, Lapotre A, Jacob PY, Tonetto A, Tighilet B, Sargolini F. Long-lasting spatial memory deficits and impaired hippocampal plasticity following unilateral vestibular loss. Prog Neurobiol 2023; 223:102403. [PMID: 36821981 DOI: 10.1016/j.pneurobio.2023.102403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/25/2022] [Accepted: 01/04/2023] [Indexed: 02/23/2023]
Abstract
Unilateral vestibular loss (UVL) induces a characteristic vestibular syndrome composed of various posturo-locomotor, oculomotor, vegetative and perceptivo-cognitive symptoms. Functional deficits are progressively recovered over time during vestibular compensation, that is supported by the expression of multiscale plasticity mechanisms. While the dynamic of post-UVL posturo-locomotor and oculomotor deficits is well characterized, the expression over time of the cognitive deficits, and in particular spatial memory deficits, is still debated. In this study we aimed at investigating spatial memory deficits and their recovery in a rat model of unilateral vestibular neurectomy (UVN), using a wide spectrum of behavioral tasks. In parallel, we analyzed markers of hippocampal plasticity involved in learning and memory. Our results indicate the UVN affects all domains of spatial memory, from working memory to reference memory and object-in-place recognition. These deficits are associated with long-lasting impaired plasticity in the ipsilesional hippocampus. These results highlight the crucial role of symmetrical vestibular information in spatial memory and contribute to a better understanding of the cognitive disorders observed in vestibular patients.
Collapse
Affiliation(s)
- Nada El Mahmoudi
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France.
| | - Célia Laurent
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - David Péricat
- Université de Toulouse Paul Sabatier -CNRS, Institut de pharmacologie et de biologie structurale, Toulouse, France
| | - Isabelle Watabe
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Agnès Lapotre
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Pierre-Yves Jacob
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Alain Tonetto
- Aix Marseille Université-CNRS, Centrale Marseille, FSCM (FR 1739), PRATIM, F-13397 Marseille, France
| | - Brahim Tighilet
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France
| | - Francesca Sargolini
- Aix-Marseille Université -CNRS, Laboratoire de Neurosciences Cognitives, LNC UMR 7291, Centre Saint Charles, Case C; 3 Place Victor Hugo, 13331, Marseille Cedex 03, France.
| |
Collapse
|
6
|
Foster TC. Animal models for studies of alcohol effects on the trajectory of age-related cognitive decline. Alcohol 2023; 107:4-11. [PMID: 35504438 DOI: 10.1016/j.alcohol.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023]
Abstract
There is growing interest in understanding how ethanol use interacts with advancing age to influence the brain and cognition. Animal models are employed to investigate the cellular and molecular mechanisms of brain aging and age-related neurodegenerative diseases that underlie cognitive decline. However, all too often research on problems and diseases of the elderly are conducted in healthy young animals, providing little clinical relevance. The validity of animal models is discussed, and confounds due to age-related differences in anxiety, sensory-motor function, and procedural learning are highlighted in order to enhance the successful translation of preclinical results into clinical settings. The mechanism of action of ethanol on brain aging will depend on the dose, acute or chronic treatment, or withdrawal from treatment and the age examined. Due to the fact that humans experience alcohol use throughout life, important questions concern the effects of the dose and duration of ethanol treatment on the trajectory of cognitive function. Central to this research will be questions of the specificity of alcohol effects on cognitive functions and related brain regions that decline with age, as well as the interaction of alcohol with mechanisms or biomarkers of brain aging. Alternatively, moderate alcohol use may provide a source of reserve and resilience against brain aging. Longitudinal studies have the advantage of being sensitive to detecting the effects of treatment on the emergence of cognitive impairment in middle age and can minimize effects of stress/anxiety associated with the novelty of alcohol exposure and behavioral testing, which disproportionately influence aged animals. Finally, the effect of alcohol on senescent neurophysiology and biomarkers of brain aging are discussed. In particular, the interaction of age and effects of alcohol on inflammation, oxidative stress, N-methyl-d-aspartate receptor function, and the balance of excitatory and inhibitory synaptic transmission are highlighted.
Collapse
Affiliation(s)
- Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
7
|
Billard JM, Freret T. Improved NMDA Receptor Activation by the Secreted Amyloid-Protein Precursor-α in Healthy Aging: A Role for D-Serine? Int J Mol Sci 2022; 23:ijms232415542. [PMID: 36555191 PMCID: PMC9779005 DOI: 10.3390/ijms232415542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Impaired activation of the N-methyl-D-aspartate subtype of glutamate receptors (NMDAR) by D-serine is linked to cognitive aging. Whether this deregulation may be used to initiate pharmacological strategies has yet to be considered. To this end, we performed electrophysiological extracellular recordings at CA3/CA1 synapses in hippocampal slices from young and aged mice. We show that 0.1 nM of the soluble N-terminal recombinant fragment of the secreted amyloid-protein precursor-α (sAPPα) added in the bath significantly increased NMDAR activation in aged but not adult mice without impacting basal synaptic transmission. In addition, sAPPα rescued the age-related deficit of theta-burst-induced long-term potentiation. Significant NMDAR improvement occurred in adult mice when sAPPα was raised to 1 nM, and this effect was drastically reduced in transgenic mice deprived of D-serine through genetic deletion of the synthesizing enzyme serine racemase. Altogether, these results emphasize the interest to consider sAPPα treatment targeting D-serine-dependent NMDAR deregulation to alleviate cognitive aging.
Collapse
|
8
|
He C, Ji J, Zhao X, Lei Y, Li H, Hao Y, Zhang S, Zhang J, Liu C, Nie J, Niu Q. The Role of PKC in Regulating NMDARs in Aluminum-Induced Learning and Memory Impairment in Rats. Neurotox Res 2021; 39:2042-2055. [PMID: 34499332 DOI: 10.1007/s12640-021-00407-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/28/2021] [Accepted: 08/30/2021] [Indexed: 01/27/2023]
Abstract
Aluminum is a widespread environmental neurotoxicant that can induce Alzheimer's disease (AD)-like damage, such as neuronal injury and impairment of learning and memory. Several studies have shown that aluminum could reduce the synaptic plasticity, but its molecular mechanism remains unclear. In this study, rats were treated with aluminum maltol (Al(mal)3) to establish a toxic animal model and PMA was used to interfere with the expression of PKC. The Morris water maze and open field test were used to investigate the behavioral changes of the rats. Western blotting and RT-PCR were used to detect the expression levels of NMDAR subunits, PKC and CaMKII. The results showed that Al(mal)3 damaged learning and memory function and reduced anxiety in rats. During this process, the expression of PKC was downregulated and it inhibited the expression of NMDARs through the phosphorylation of CaMKII.
Collapse
Affiliation(s)
- Chanting He
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Lab of Environmental Hazard & Health of Shanxi Province, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Anatomy, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jingjing Ji
- Fenyang College, Shanxi Medical University, Fenyang, 032200, Shanxi, China
| | - Xiaoyan Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yang Lei
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Huan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yanxia Hao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Shuhui Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jingsi Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chengjuan Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Lab of Environmental Hazard & Health of Shanxi Province, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Lab of Environmental Hazard & Health of Shanxi Province, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
- Key Lab of Cellular Physiology of Education Ministry, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
9
|
Arias-Cavieres A, Fonteh A, Castro-Rivera CI, Garcia AJ. Intermittent Hypoxia causes targeted disruption to NMDA receptor dependent synaptic plasticity in area CA1 of the hippocampus. Exp Neurol 2021; 344:113808. [PMID: 34256046 DOI: 10.1016/j.expneurol.2021.113808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/21/2021] [Accepted: 07/08/2021] [Indexed: 12/27/2022]
Abstract
Changed NMDA receptor (NMDAr) physiology is implicated with cognitive deficit resulting from conditions ranging from normal aging to neurological disease. Using intermittent hypoxia (IH) to experimentally model untreated sleep apnea, a clinical condition whose comorbidities include neurocognitive impairment, we recently demonstrated that IH causes a pro-oxidant condition that contributes to deficits in spatial memory and in NMDAr-dependent long-term potentiation (LTP). However, the impact of IH on additional forms of synaptic plasticity remains ill-defined. Here we show that IH prevents the induction of NMDAr-dependent LTP and long-term depression (LTD) in hippocampal brain slices from mice exposed to ten days of IH (IH10) yet spares NMDAr-independent forms of synaptic plasticity. Deficits in synaptic plasticity were accompanied by a reduction in hippocampal GluN1 expression. Acute manipulation of redox state using the reducing agent, Dithiothreitol (DTT) stimulated the NMDAr-dependent fEPSP following IH10. However, acute use of either DTT or MnTMPyP did not restore NMDAr-dependent synaptic plasticity after IH10 or prevent the IH-dependent reduction in GluN1, the obligatory subunit of the NMDAr. In contrast, MnTMPyP during IH10 (10-MnTMPyP), prevented the suppressive effects of IH on both NMDAr-dependent synaptic plasticity and GluN1 expression. These findings indicate that while the IH-dependent pro-oxidant state causes reversible oxidative neuromodulation of NMDAr activity, acute manipulation of redox state is ineffective in rescuing two key effects of IH related to the NMDAr within the hippocampus. These IH-dependent changes associated with the NMDAr may be a primary avenue by which IH enhances the vulnerability to impaired learning and memory when sleep apnea is left untreated in normal aging and in disease.
Collapse
Affiliation(s)
- Alejandra Arias-Cavieres
- Institute for Integrative Physiology, The University of Chicago, USA; Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA
| | - Ateh Fonteh
- Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA
| | - Carolina I Castro-Rivera
- Institute for Integrative Physiology, The University of Chicago, USA; Grossman Institute for Neuroscience, Quantitative Biology & Human Behavior, The University of Chicago, USA
| | - Alfredo J Garcia
- Institute for Integrative Physiology, The University of Chicago, USA; Grossman Institute for Neuroscience, Quantitative Biology & Human Behavior, The University of Chicago, USA; Department of Medicine, Section of Emergency Medicine, The University of Chicago, USA.
| |
Collapse
|
10
|
Jobson DD, Hase Y, Clarkson AN, Kalaria RN. The role of the medial prefrontal cortex in cognition, ageing and dementia. Brain Commun 2021; 3:fcab125. [PMID: 34222873 PMCID: PMC8249104 DOI: 10.1093/braincomms/fcab125] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/08/2021] [Accepted: 04/14/2021] [Indexed: 01/18/2023] Open
Abstract
Humans require a plethora of higher cognitive skills to perform executive functions, such as reasoning, planning, language and social interactions, which are regulated predominantly by the prefrontal cortex. The prefrontal cortex comprises the lateral, medial and orbitofrontal regions. In higher primates, the lateral prefrontal cortex is further separated into the respective dorsal and ventral subregions. However, all these regions have variably been implicated in several fronto-subcortical circuits. Dysfunction of these circuits has been highlighted in vascular and other neurocognitive disorders. Recent advances suggest the medial prefrontal cortex plays an important regulatory role in numerous cognitive functions, including attention, inhibitory control, habit formation and working, spatial or long-term memory. The medial prefrontal cortex appears highly interconnected with subcortical regions (thalamus, amygdala and hippocampus) and exerts top-down executive control over various cognitive domains and stimuli. Much of our knowledge comes from rodent models using precise lesions and electrophysiology readouts from specific medial prefrontal cortex locations. Although, anatomical disparities of the rodent medial prefrontal cortex compared to the primate homologue are apparent, current rodent models have effectively implicated the medial prefrontal cortex as a neural substrate of cognitive decline within ageing and dementia. Human brain connectivity-based neuroimaging has demonstrated that large-scale medial prefrontal cortex networks, such as the default mode network, are equally important for cognition. However, there is little consensus on how medial prefrontal cortex functional connectivity specifically changes during brain pathological states. In context with previous work in rodents and non-human primates, we attempt to convey a consensus on the current understanding of the role of predominantly the medial prefrontal cortex and its functional connectivity measured by resting-state functional MRI in ageing associated disorders, including prodromal dementia states, Alzheimer's disease, post-ischaemic stroke, Parkinsonism and frontotemporal dementia. Previous cross-sectional studies suggest that medial prefrontal cortex functional connectivity abnormalities are consistently found in the default mode network across both ageing and neurocognitive disorders such as Alzheimer's disease and vascular cognitive impairment. Distinct disease-specific patterns of medial prefrontal cortex functional connectivity alterations within specific large-scale networks appear to consistently feature in the default mode network, whilst detrimental connectivity alterations are associated with cognitive impairments independently from structural pathological aberrations, such as grey matter atrophy. These disease-specific patterns of medial prefrontal cortex functional connectivity also precede structural pathological changes and may be driven by ageing-related vascular mechanisms. The default mode network supports utility as a potential biomarker and therapeutic target for dementia-associated conditions. Yet, these associations still require validation in longitudinal studies using larger sample sizes.
Collapse
Affiliation(s)
- Dan D Jobson
- Translational and Clinical Research Institute,
Newcastle University, Campus for Ageing & Vitality,
Newcastle upon Tyne NE4 5PL, UK
| | - Yoshiki Hase
- Translational and Clinical Research Institute,
Newcastle University, Campus for Ageing & Vitality,
Newcastle upon Tyne NE4 5PL, UK
| | - Andrew N Clarkson
- Department of Anatomy, Brain Health Research Centre
and Brain Research New Zealand, University of Otago, Dunedin 9054,
New Zealand
| | - Rajesh N Kalaria
- Translational and Clinical Research Institute,
Newcastle University, Campus for Ageing & Vitality,
Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
11
|
Yegla B, Boles J, Kumar A, Foster TC. Partial microglial depletion is associated with impaired hippocampal synaptic and cognitive function in young and aged rats. Glia 2021; 69:1494-1514. [PMID: 33586813 DOI: 10.1002/glia.23975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
The role of microglia in mediating age-related changes in cognition and hippocampal synaptic function was examined by microglial depletion and replenishment using PLX3397. We observed age-related differences in microglial number and morphology, as well as increased Iba-1 expression, indicating microglial activation. PLX3397 treatment decreased microglial number, with aged rats exhibiting the lowest density. Young rats exhibited increased expression of pro-inflammatory cytokines during depletion and repopulation and maintenance of Iba-1 levels despite reduced microglial number. For aged rats, several cytokines increased with depletion and recovered during repopulation; however, aged rats did not fully recover microglial cell number or Iba-1 expression during repopulation, with a recovery comparable to young control levels rather than aged controls. Hippocampal CA3-CA1 synaptic transmission was impaired with age, and microglial depletion was associated with decreased total synaptic transmission in young and aged rats. A robust decline in N-methyl-d-aspartate-receptor-mediated synaptic transmission arose in young depleted rats specifically. Microglial replenishment normalized depletion-induced synaptic function to control levels; however, recovery of aged animals did not mirror young. Microglial depletion was associated with decreased context-object discrimination memory in both age groups, which recovered with microglial repopulation. Aged rats displayed impaired contextual and cued fear memory, and microglial replenishment did not recover their memory to the level of young. The current study indicates that cognitive function and synaptic transmission benefit from the support of aged microglia and are hindered by removal of these cells. Replenishment of microglia in aging did not ameliorate age-related cognitive impairments or senescent synaptic function.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Jake Boles
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics Program, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Strong KL, Epplin MP, Ogden KK, Burger PB, Kaiser TM, Wilding TJ, Kusumoto H, Camp CR, Shaulsky G, Bhattacharya S, Perszyk RE, Menaldino DS, McDaniel MJ, Zhang J, Le P, Banke TG, Hansen KB, Huettner JE, Liotta DC, Traynelis SF. Distinct GluN1 and GluN2 Structural Determinants for Subunit-Selective Positive Allosteric Modulation of N-Methyl-d-aspartate Receptors. ACS Chem Neurosci 2021; 12:79-98. [PMID: 33326224 DOI: 10.1021/acschemneuro.0c00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
N-Methyl-d-aspartate receptors (NMDARs) are ionotropic ligand-gated glutamate receptors that mediate fast excitatory synaptic transmission in the central nervous system (CNS). Several neurological disorders may involve NMDAR hypofunction, which has driven therapeutic interest in positive allosteric modulators (PAMs) of NMDAR function. Here we describe modest changes to the tetrahydroisoquinoline scaffold of GluN2C/GluN2D-selective PAMs that expands activity to include GluN2A- and GluN2B-containing recombinant and synaptic NMDARs. These new analogues are distinct from GluN2C/GluN2D-selective compounds like (+)-(3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone (CIQ) by virtue of their subunit selectivity, molecular determinants of action, and allosteric regulation of agonist potency. The (S)-enantiomers of two analogues (EU1180-55, EU1180-154) showed activity at NMDARs containing all subunits (GluN2A, GluN2B, GluN2C, GluN2D), whereas the (R)-enantiomers were primarily active at GluN2C- and GluN2D-containing NMDARs. Determination of the actions of enantiomers on triheteromeric receptors confirms their unique pharmacology, with greater activity of (S) enantiomers at GluN2A/GluN2D and GluN2B/GluN2D subunit combinations than (R) enantiomers. Evaluation of the (S)-EU1180-55 and EU1180-154 response of chimeric kainate/NMDA receptors revealed structural determinants of action within the pore-forming region and associated linkers. Scanning mutagenesis identified structural determinants within the GluN1 pre-M1 and M1 regions that alter the activity of (S)-EU1180-55 but not (R)-EU1180-55. By contrast, mutations in pre-M1 and M1 regions of GluN2D perturb the actions of only the (R)-EU1180-55 but not the (S) enantiomer. Molecular modeling supports the idea that the (S) and (R) enantiomers interact distinctly with GluN1 and GluN2 pre-M1 regions, suggesting that two distinct sites exist for these NMDAR PAMs, each of which has different functional effects.
Collapse
Affiliation(s)
- Katie L. Strong
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Matthew P. Epplin
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Kevin K. Ogden
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Pieter B. Burger
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Thomas M. Kaiser
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Timothy J. Wilding
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Hiro Kusumoto
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chad R. Camp
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Gil Shaulsky
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama 36849, United States
| | - Riley E. Perszyk
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - David S. Menaldino
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Miranda J. McDaniel
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jing Zhang
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Phuong Le
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Tue G. Banke
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Kasper B. Hansen
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Division for Biological Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - James E. Huettner
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
13
|
Dahan L, Rampon C, Florian C. Age-related memory decline, dysfunction of the hippocampus and therapeutic opportunities. Prog Neuropsychopharmacol Biol Psychiatry 2020; 102:109943. [PMID: 32298784 DOI: 10.1016/j.pnpbp.2020.109943] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 04/01/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
While the aging of the population is a sign of progress for societies, it also carries its load of negative aspects. Among them, cognitive decline and in particular memory loss is a common feature of non-pathological aging. Autobiographical memories, which rely on the hippocampus, are a primary target of age-related cognitive decline. Here, focusing on the neurobiological mechanisms of memory formation and storage, we describe how hippocampal functions are altered across time in non-pathological mammalian brains. Several hallmarks of aging have been well described over the last decades; among them, we consider altered synaptic communication and plasticity, reduction of adult neurogenesis and epigenetic alterations. Building on the neurobiological processes of cognitive aging that have been identified to date, we review some of the strategies based on lifestyle manupulation allowing to address age-related cognitive deficits.
Collapse
Affiliation(s)
- Lionel Dahan
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Claire Rampon
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France
| | - Cédrick Florian
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse; CNRS, UPS, Toulouse Cedex 9, France.
| |
Collapse
|
14
|
Alpha-synuclein differentially reduces surface expression of N-methyl-d-aspartate receptors in the aging human brain. Neurobiol Aging 2020; 90:24-32. [PMID: 32171588 DOI: 10.1016/j.neurobiolaging.2020.02.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 01/22/2023]
Abstract
The aging brain is associated with reduced cell surface expression of N-methyl-d-aspartate receptors (NMDARs), but the mechanism remains poorly understood. In the present study, we showed that in the striatum and hippocampus but not the cerebellum and parietal cortex, levels of α-synuclein monomers and oligomers increased with age, which correlated negatively with the expression of GluN1, and positively with the expression of total Rab5B. The oligomer-α-synuclein exhibited a stronger correlation with the expression of surface GluN1 and total Rab5B. In MES23.5 cells, the monomer- or oligomer-α-synuclein were shown to increase in a manner dependent on the concentrations of the added monomers and oligomers. Again, the oligomer-α-synuclein showed more potent effects than the monomer-α-synuclein on surface GluN1 and total Rab5B expression. Accordingly, the oligomer-treated cells showed a greater reduction in NMDA-evoked Ca2+ influx than the monomer-treated cells, which was largely inhibited by pistop2, a clathrin inhibitor. These results suggest that the age-dependent accumulation of α-synuclein monomers and oligomers differentially contributes to the reduction in surface NMDAR expression in selective brain regions.
Collapse
|
15
|
Kohlmeier KA, Polli FS. Plasticity in the Brainstem: Prenatal and Postnatal Experience Can Alter Laterodorsal Tegmental (LDT) Structure and Function. Front Synaptic Neurosci 2020; 12:3. [PMID: 32116639 PMCID: PMC7019863 DOI: 10.3389/fnsyn.2020.00003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/14/2020] [Indexed: 12/16/2022] Open
Abstract
The brainstem has traditionally been considered an area of the brain with autonomous control of mostly homeostatic functions such as heart rate, respiration, and the sleep and wakefulness state, which would preclude the necessity to exhibit the high degree of synaptic or cellular mechanisms of plasticity typical of regions of the brain responsible for flexible, executive control, such as the medial prefrontal cortex or the hippocampus. The perception that the brainstem does not share the same degree of flexibility to alter synaptic strength and/or wiring within local circuits makes intuitive sense, as it is not easy to understand how "soft wiring" would be an advantage when considering the importance of faithful and consistent performance of the homeostatic, autonomic functions that are controlled by the brainstem. However, many of the molecular and cellular requirements which underlie strengthening of synapses seen in brain regions involved in higher-level processing are present in brainstem nuclei, and recent research suggest that the view of the brainstem as "hard wired," with rigid and static connectivity and with unchanging synaptic strength, is outdated. In fact, information from studies within the last decades, including work conducted in our group, leads us to propose that the brainstem can dynamically alter synaptic proteins, and change synaptic connections in response to prenatal or postnatal stimuli, and this would likely alter functionality and output. This article reviews recent research that has provided information resulting in our revision of the view of the brainstem as static and non-changing by using as example recent information gleaned from a brainstem pontine nucleus, the laterodorsal tegmentum (LDT). The LDT has demonstrated mechanisms underlying synaptic plasticity, and plasticity has been exhibited in the postnatal LDT following exposure to drugs of abuse. Further, exposure of the brain during gestation to drugs of abuse results in alterations in development of signaling pathways in the LDT. As the LDT provides a high degree of innervation of mesoaccumbal and mesocortical circuits involved in salience, as well as thalamocortical circuits involved in control of arousal and orientation, changes in synaptic strength would be expected to alter output, which would significantly impact behavioral state, motivated behavior and directed attention. Further, alterations in developmental trajectory within the LDT following prenatal exposure to drugs of abuse would be expected to impact on later life expression of motivation and arousal.
Collapse
Affiliation(s)
- Kristi A. Kohlmeier
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
16
|
Kumar A. Calcium Signaling During Brain Aging and Its Influence on the Hippocampal Synaptic Plasticity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:985-1012. [PMID: 31646542 DOI: 10.1007/978-3-030-12457-1_39] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Calcium (Ca2+) ions are highly versatile intracellular signaling molecules and are universal second messenger for regulating a variety of cellular and physiological functions including synaptic plasticity. Ca2+ homeostasis in the central nervous system endures subtle dysregulation with advancing age. Research has provided abundant evidence that brain aging is associated with altered neuronal Ca2+ regulation and synaptic plasticity mechanisms. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during aging. The current chapter takes a specific perspective, assessing various Ca2+ sources and the influence of aging on Ca2+ sources and synaptic plasticity in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in neuronal Ca2+ signaling and synaptic plasticity mechanisms will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment associated with aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Early sirtuin 2 inhibition prevents age-related cognitive decline in a senescence-accelerated mouse model. Neuropsychopharmacology 2020; 45:347-357. [PMID: 31471557 PMCID: PMC6901465 DOI: 10.1038/s41386-019-0503-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/02/2019] [Accepted: 08/22/2019] [Indexed: 02/07/2023]
Abstract
The senescence-accelerated mouse prone-8 (SAMP8) model has been considered as a good model for aged-related cognitive decline and Alzheimer's disease (AD). Since epigenetic alterations represent a crucial mechanism during aging, in the present study we tested whether the inhibition of the histone deacetylase sirtuin 2 (SIRT2) could ameliorate the age-dependent cognitive impairments and associated neuropathology shown by SAMP8 mice. To this end, the potent SIRT2-selective inhibitor, 33i (5 mg/kg i.p. 8 weeks) was administered to 5-month-old (early treatment) and 8-month-old (late treatment) SAMP8 and aged matched control, senescence-accelerated mouse resistant-1 (SAMR1) mice. 33i administration to 5-month-old SAMP8 mice improved spatial learning and memory impairments shown by this strain in the Morris water maze. SAMP8 showed hyperphosphorylation of tau protein and decrease levels of SIRT1 in the hippocampus, which were not altered by 33i treatment. However, this treatment upregulated the glutamate receptor subunits GluN2A, GluN2B, and GluA1 in both SAMR1 and SAMP8. Moreover, early SIRT2 inhibition prevented neuroinflammation evidenced by reduced levels of GFAP, IL-1β, Il-6, and Tnf-α, providing a plausible explanation for the improvement of cognitive deficits shown by 33i-treated SAMP8 mice. When 33i was administered to 8-month-old SAMP8 with a severe established pathology, increases in GluN2A, GluN2B, and GluA1 were observed; however, it was not able to reverse the cognitive decline or the neuroinflammation. These results suggest that early SIRT2 inhibition might be beneficial in preventing age-related cognitive deficits, neuroinflammation, and AD progression and could be an emerging candidate for the treatment of other diseases linked to dementia.
Collapse
|
18
|
Age-related differences in brain activations during spatial memory formation in a well-learned virtual Morris water maze (vMWM) task. Neuroimage 2019; 202:116069. [PMID: 31382044 DOI: 10.1016/j.neuroimage.2019.116069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/03/2019] [Accepted: 08/02/2019] [Indexed: 11/21/2022] Open
Abstract
The current study applied a rodent-based virtual Morris water maze (vMWM) protocol to an investigation of differences in search performance and brain activations between young and older male human adults. All participants completed in-lab practice and testing before performing the task in the fMRI scanner. Behavioral performance during fMRI scanning - measured in terms of corrected cumulative proximity (CCProx) to the goal - showed that a subgroup of older good performers attained comparable levels of search accuracy to the young while another subgroup of older poor performers exhibited consistently lower levels of search accuracy than both older good performers and the young. With regard to brain activations, young adults exhibited greater activations in the cerebellum and cuneus than all older adults, as well as older poor performers. Older good performers exhibited higher activation than older poor performers in the orbitofrontal cortex (BA 10/11), as well as in the cuneus and cerebellum. Brain-behavior correlations further showed that activations in regions involved in visuomotor control (cerebellum, lingual gyrus) and egocentric spatial processing (premotor cortex, precuneus) correlated positively with search accuracy (i.e., closer proximity to goal) in all participants. Notably, activations in the anterior hippocampus correlated positively with search accuracy (CCProx inversed) in the young but not in the old. Taken together, these findings implicated the orbitofrontal cortex and the cerebellum as playing crucial roles in executive and visuospatial processing in older adults, supporting the proposal of an age-related compensatory shift in spatial memory functions away from the hippocampus toward the prefrontal cortex.
Collapse
|
19
|
Lee G, Zhou Y. NMDAR Hypofunction Animal Models of Schizophrenia. Front Mol Neurosci 2019; 12:185. [PMID: 31417356 PMCID: PMC6685005 DOI: 10.3389/fnmol.2019.00185] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) hypofunction hypothesis has been proposed to help understand the etiology and pathophysiology of schizophrenia. This hypothesis was based on early observations that NMDAR antagonists could induce a full range of symptoms of schizophrenia in normal human subjects. Accumulating evidence in humans and animal studies points to NMDAR hypofunctionality as a convergence point for various symptoms of schizophrenia. Here we review animal models of NMDAR hypofunction generated by pharmacological and genetic approaches, and how they relate to the pathophysiology of schizophrenia. In addition, we discuss the limitations of animal models of NMDAR hypofunction and their potential utility for therapeutic applications.
Collapse
Affiliation(s)
| | - Yi Zhou
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
20
|
Migues PV, Wong J, Lyu J, Hardt O. NMDA receptor activity bidirectionally controls active decay of long-term spatial memory in the dorsal hippocampus. Hippocampus 2019; 29:883-888. [PMID: 31058409 DOI: 10.1002/hipo.23096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/17/2019] [Indexed: 12/22/2022]
Abstract
The time-dependent forgetting of long-term spatial memories involves activation of NMDA receptors (NMDARs) in the hippocampus. Here, we tested whether NMDARs regulate memory persistence bidirectionally, decreasing or increasing the rate of forgetting. We found that blocking NMDAR activation with AP5 or the GluN2B-selective antagonist Ro25-6981 in the dorsal hippocampus (dHPC) prevented the natural forgetting of long-term memory for the locations of objects in an open field arena. In contrast, while enhancing NMDAR function with the partial agonist D-Cycloserine did not affect the speed of forgetting for these types of memories, infusing the NMDAR co-agonist D-Serine significantly shortened their persistence. These results suggest that NMDAR activity can modulate the speed of constitutive long-term memory decay in the dHPC and that regulating NMDAR expression and D-Serine availability could provide a mechanism to control the duration of long-term memory.
Collapse
Affiliation(s)
- Paola Virginia Migues
- Department of Psychology, Behavioural Neuroscience, McGill University, Montréal, Québec, Canada
| | - Jacinda Wong
- Department of Psychology, Behavioural Neuroscience, McGill University, Montréal, Québec, Canada
| | - Jeongho Lyu
- Department of Psychology, Behavioural Neuroscience, McGill University, Montréal, Québec, Canada
| | - Oliver Hardt
- Department of Psychology, Behavioural Neuroscience, McGill University, Montréal, Québec, Canada.,Centre for Discovery Brain Sciences, Simons Initiative for the Developing Brain, The University of Edinburgh, Scotland, UK
| |
Collapse
|
21
|
Stroke Induces a BDNF-Dependent Improvement in Cognitive Flexibility in Aged Mice. Neural Plast 2019; 2019:1460890. [PMID: 31191635 PMCID: PMC6525942 DOI: 10.1155/2019/1460890] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/10/2019] [Accepted: 04/04/2019] [Indexed: 11/17/2022] Open
Abstract
Stroke remains a leading cause of disability worldwide. Recently, we have established an animal model of stroke that results in delayed impairment in spatial memory, allowing us to better investigate cognitive deficits. Young and aged brains show different recovery profiles after stroke; therefore, we assessed aged-related differences in poststroke cognition. As neurotrophic support diminishes with age, we also investigated the involvement of brain-derived neurotrophic factor (BDNF) in these differences. Young (3-6 months old) and aged (16-21 months old) mice were trained in operant touchscreen chambers to complete a visual pairwise discrimination (VD) task. Stroke or sham surgery was induced using the photothrombotic model to induce a bilateral prefrontal cortex stroke. Five days poststroke, an additional cohort of aged stroke animals were treated with intracerebral hydrogels loaded with the BDNF decoy, TrkB-Fc. Following treatment, animals underwent the reversal and rereversal task to identify stroke-induced cognitive deficits at days 17 and 37 poststroke, respectively. Assessment of sham animals using Cox regression and log-rank analyses showed aged mice exhibit an increased impairment on VD reversal and rereversal learning compared to young controls. Stroke to young mice revealed no impairment on either task. In contrast, stroke to aged mice facilitated a significant improvement in reversal learning, which was dampened in the presence of the BDNF decoy, TrkB-Fc. In addition, aged stroke control animals required significantly less consecutive days and correction trials to master the reversal task, relative to aged shams, an effect dampened by TrkB-Fc. Our findings support age-related differences in recovery of cognitive function after stroke. Interestingly, aged stroke animals outperformed their sham counterparts, suggesting reopening of a critical window for recovery that is being mediated by BDNF.
Collapse
|
22
|
Wang X, Shan Y, Tang Z, Gao L, Liu H. Neuroprotective effects of dexmedetomidine against isoflurane-induced neuronal injury via glutamate regulation in neonatal rats. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 13:153-160. [PMID: 30613136 PMCID: PMC6306062 DOI: 10.2147/dddt.s163197] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Considerable evidences support the finding that the anesthesia reagent isoflurane increases neuronal cell death in young rats. Recent studies have shown that dexmedetomidine can reduce isoflurane-induced neuronal injury, but the mechanism remains unclear. We investigated whether isoflurane cause neurotoxicity to the central nervous system by regulating the N-methyl-D-aspartate receptor (NMDAR) and excitatory amino acid transporter1 (EAAT1) in young rats. Furthermore, we examined if dexmedetomidine could decrease isoflurane-induced neurotoxicity. Methods Neonatal rats (postnatal day 7, n=144) were randomly divided into four groups of 36 animals each: control (saline injection without isoflurane); isoflurane (2% for 4 h); isoflurane + single dose of dexmedetomidine (75 µg/kg, 20 min before the start of 2% isoflurane for 4 h); and isoflurane + dual doses of dexmedetomidine (25 µg/kg, 20 min before and 2 h after start of isoflurane at 2% for 4 h). Six neonates from each group were euthanatized at 2 h, 12 h, 24 h, 3 days, 7 days and 28 days post-anesthesia. Hippocampi were collected and processed for protein extraction. Expression levels of the NMDAR subunits NR2A and NR2B, EAAT1 and caspase-3 were measured by western blot analysis. Results Protein levels of NR2A, EAAT1 and caspase-3 were significantly increased in hippocampus of the isoflurane group from 2 h to 3 days, while NR2B levels were decreased. However, the -induced increase in NR2A, EAAT1 and caspase-3 and the decrease in NR2B in isoflurane-exposed rats were ameliorated in the rats treated with single or dual doses of dexmedetomidine. Isoflurane-induced neuronal damage in neonatal rats is due in part to the increase in NR2A and EAAT1 and the decrease in NR2B in the hippocampus. Conclusion Dexmedetomidine protects the brain against the use of isoflurane through the regulation of NR2A, NR2B and EAAT1. However, using the same amount of dexmedetomidine, the trend of protection is basically the same.
Collapse
Affiliation(s)
- Xue Wang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, China, .,Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang 441000, China
| | - Yangyang Shan
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, China,
| | - Zhiyin Tang
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, China,
| | - Linlin Gao
- Department of Medical Research, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Hongtao Liu
- Department of Anesthesiology, Shengjing Hospital, China Medical University, Shenyang 110004, China,
| |
Collapse
|
23
|
Billard JM. Changes in Serine Racemase-Dependent Modulation of NMDA Receptor: Impact on Physiological and Pathological Brain Aging. Front Mol Biosci 2018; 5:106. [PMID: 30555832 PMCID: PMC6282039 DOI: 10.3389/fmolb.2018.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/09/2018] [Indexed: 02/02/2023] Open
Abstract
The N-methyl-D-Aspartate glutamate receptors (NMDARs) are pivotal for the functional and morphological plasticity that are required in neuronal networks for efficient brain activities and notably for cognitive-related abilities. Because NMDARs are heterogeneous in subunit composition and associated with multiple functional regulatory sites, their efficacy is under the tonic influence of numerous allosteric modulations, whose dysfunction generally represents the first step generating pathological states. Among the enzymatic candidates, serine racemase (SR) has recently gathered an increasing interest considering that it tightly regulates the production of d-serine, an amino acid now viewed as the main endogenous co-agonist necessary for NMDAR activation. Nowadays, SR deregulation is associated with a wide range of neurological and psychiatric diseases including schizophrenia, amyotrophic lateral sclerosis, and depression. This review aims at compelling the most recent experimental evidences indicating that changes in SR-related modulation of NMDARs also govern opposite functional dysfunctions in physiological and pathological (Alzheimer's disease) aging that finally results in memory disabilities in both cases. It also highlights SR as a relevant alternative target for new pharmacological strategies aimed at preventing functional alterations and cognitive impairments linked to the aging process.
Collapse
|
24
|
Kumar A, Foster TC. Alteration in NMDA Receptor Mediated Glutamatergic Neurotransmission in the Hippocampus During Senescence. Neurochem Res 2018; 44:38-48. [PMID: 30209673 DOI: 10.1007/s11064-018-2634-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/07/2018] [Accepted: 09/08/2018] [Indexed: 12/17/2022]
Abstract
Glutamate is the primary excitatory neurotransmitter in neurons and glia. N-methyl-D-aspartate (NMDA), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), and kainate receptors are major ionotropic glutamate receptors. Glutamatergic neurotransmission is strongly linked with Ca2+ homeostasis. Research has provided ample evidence that brain aging is associated with altered glutamatergic neurotransmission and Ca2+ dysregulation. Much of the work has focused on the hippocampus, a brain region critically involved in learning and memory, which is particularly susceptible to dysfunction during senescence. The current review examines Ca2+ regulation with a focus on the NMDA receptors in the hippocampus. Integrating the knowledge of the complexity of age-related alterations in Ca2+ homeostasis and NMDA receptor-mediated glutamatergic neurotransmission will positively shape the development of highly effective therapeutics to treat brain disorders including cognitive impairment.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, PO Box 100244, Gainesville, FL, 32610-0244, USA.
- Genetics and Genomics Program, University of Florida, Gainesville, FL, 32611, USA.
| |
Collapse
|
25
|
Zhan JQ, Zheng LL, Chen HB, Yu B, Wang W, Wang T, Ruan B, Pan BX, Chen JR, Li XF, Wei B, Yang YJ. Hydrogen Sulfide Reverses Aging-Associated Amygdalar Synaptic Plasticity and Fear Memory Deficits in Rats. Front Neurosci 2018; 12:390. [PMID: 29930496 PMCID: PMC5999728 DOI: 10.3389/fnins.2018.00390] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/22/2018] [Indexed: 01/05/2023] Open
Abstract
As an endogenous neuromodulator, hydrogen sulfide (H2S) exerts multiple biological effects in the brain. Previous studies have shown that H2S is involved in the regulation of neural synaptic plasticity and cognition in healthy rodents. It is well known that there is a progressive decline of cognitive function that occurs with increased age. The purpose of this study was to investigate the role of H2S in aging-associated amygdalar synaptic plasticity and cued fear memory deficits as well as to explore the underlying mechanisms. We found that H2S levels in the amygdala were significantly lower in aged rats when compared with healthy adult rates, which displayed significant deficits in long-term potentiation (LTP) in the thalamo-lateral amygdala (LA) pathway and amygdala-dependent cued fear memory. Bath application of an H2S donor, sodium hydrogen sulfide (NaHS), significantly reversed the impaired LTP in brain slices from aged rats, and intra-LA infusion of NaHS restored the cued fear memory in aged rats. Mechanismly, we found that H2S treatment significantly enhanced NMDAR-mediated synaptic responses in the thalamo-LA pathway of aged rats. Notably, GluN2B-containing NMDARs, but not GluN2A-containing NMDARs, contributed to the effects of H2S on aging-associated impairments of amygdalar LTP and fear memory, because applying GluN2B antagonist could abolish the beneficial effects of NaHS treatment on amygdalar LTP and cognitive performance in aged rats. Collectively, these results show that H2S can reverse aging-associated amygdalar synaptic plasticity and fear memory deficits by restoring the function of GluN2B-containing NMDARs, suggesting that supplement of H2S might be a therapeutic approach for aging-related cognitive disorders.
Collapse
Affiliation(s)
- Jin-Qiong Zhan
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Li-Li Zheng
- Department of Pharmacy, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Hai-Bo Chen
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Bin Yu
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Wei Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ting Wang
- Department of Pharmacology, College of Medical Science, China Three Gorges University, Yichang, China
| | - Bo Ruan
- Department of Pharmacology, College of Medical Science, China Three Gorges University, Yichang, China
| | - Bin-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, Nanchang, China
| | - Juan-Ru Chen
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Xue-Fen Li
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bo Wei
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| | - Yuan-Jian Yang
- Biological Psychiatry Laboratory, Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Ahn JH, Lee JS, Cho JH, Park JH, Lee TK, Song M, Kim H, Kang SH, Won MH, Lee CH. Age-dependent decrease of Nurr1 protein expression in the gerbil hippocampus. Biomed Rep 2018; 8:517-522. [PMID: 29904610 PMCID: PMC5996841 DOI: 10.3892/br.2018.1094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023] Open
Abstract
Nuclear receptor related-1 protein (Nurr1) serves important roles in hippocampal-dependent cognitive process. In the present study, the protein expression of Nurr1 was compared in the hippocampi of young [postnatal month 3 (PM 3)], adult (PM 12) and aged (PM 24) gerbils using western blot analysis and immunohistochemistry. Results indicated that the protein level of Nurr1 was significantly and gradually decreased in the gerbil hippocampus with increasing age. In addition, strong Nurr1 immunoreactivity was primarily observed in pyramidal neurons and granule cells of the hippocampus in the young group, which was determined to be reduced in the adult group and to a greater extent in the aged group. Collectively the data demonstrated that Nurr1 immunoreactivity was gradually and markedly decreased during normal aging. These results indicate that gradual decrease of Nurr1 expression in the hippocampus may be associated with the normal aging process and a decline in hippocampus-dependent cognitive function.
Collapse
Affiliation(s)
- Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Joon Seok Lee
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine and Institute of Medical Sciences, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Seok Hoon Kang
- Department of Medical Education, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Choong Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan, Chungcheongnam 31116, Republic of Korea
| |
Collapse
|
27
|
Portero-Tresserra M, Martí-Nicolovius M, Tarrés-Gatius M, Candalija A, Guillazo-Blanch G, Vale-Martínez A. Intra-hippocampal D-cycloserine rescues decreased social memory, spatial learning reversal, and synaptophysin levels in aged rats. Psychopharmacology (Berl) 2018; 235:1463-1477. [PMID: 29492616 DOI: 10.1007/s00213-018-4858-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 02/15/2018] [Indexed: 01/08/2023]
Abstract
RATIONALE Aging is characterized by a decrease in N-methyl-D-aspartate receptors (NMDARs) in the hippocampus, which might be one of the factors involved in the age-dependent cognitive decline. D-Cycloserine (DCS), a partial agonist of the NMDAR glycine recognition site, could improve memory deficits associated to neurodegenerative disorders and cognitive deficits observed in normal aging. OBJECTIVES AND METHODS The aim of the present study was to explore whether DCS would reverse age-dependent memory deficits and decreases in NMDA receptor subunits (GluN1, GluN2A, and GluN2B) and the presynaptic protein synaptophysin in Wistar rats. We investigated the effects of pre-training infusions of DCS (10 μg/hemisphere) in the ventral hippocampus on two hippocampal-dependent learning tasks, the social transmission of food preference (STFP), and the Morris water maze (MWM). RESULTS The results revealed that infusions of DCS administered before the acquisition sessions rescued deficits in the STFP retention and MWM reversal learning in old rats. DCS also significantly increased the hippocampal levels of synaptophysin in old rats, which correlated with STFP and MWM performance in all tests. Moreover, although the levels of the GluN1 subunit correlated with the MWM acquisition and reversal, DCS did not enhance the expression of such synaptic protein. CONCLUSIONS The present behavioral results support the role of DCS as a cognitive enhancer and suggest that enhancing the function of NMDARs and synaptic plasticity in the hippocampus may be related to improvement in social memory and spatial learning reversal in aged animals.
Collapse
Affiliation(s)
- Marta Portero-Tresserra
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Margarita Martí-Nicolovius
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Mireia Tarrés-Gatius
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Candalija
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Gemma Guillazo-Blanch
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Vale-Martínez
- Departament de Psicobiologia i Metodologia de les Ciències de la Salut, Institut de Neurociències, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
28
|
Wu B, Liu J, Zhao R, Li Y, Peer J, Braun AL, Zhao L, Wang Y, Tong Z, Huang Y, Zheng JC. Glutaminase 1 regulates the release of extracellular vesicles during neuroinflammation through key metabolic intermediate alpha-ketoglutarate. J Neuroinflammation 2018. [PMID: 29540215 PMCID: PMC5853116 DOI: 10.1186/s12974-018-1120-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Extracellular vesicles (EVs) are important in the intercellular communication of the central nervous system, and their release is increased during neuroinflammation. Our previous data demonstrated an increased release of EVs during HIV-1 infection and immune activation in glial cells. However, the molecular mechanism by which infection and inflammation increase EV release remains unknown. In the current study, we investigated the role of glutaminase 1 (GLS1)-mediated glutaminolysis and the production of a key metabolic intermediate α-ketoglutarate on EV release. Methods Human monocyte-derived macrophage primary cultures and a BV2 microglia cell line were used to represent the innate immune cells in the CNS. Transmission electron microscopy, nanoparticle tracking analysis, and Western blots were used to determine the EV regulation. GLS1 overexpression was performed using an adenovirus vector in vitro and transgenic mouse models in vivo. Data were evaluated statistically by ANOVA, followed by the Bonferroni post-test for paired observations. Results Our data revealed an increased release of EVs in GLS1-overexpressing HeLa cells. In HIV-1-infected macrophages and immune-activated microglia BV2 cells, treatment with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) or CB839, two specific GLS inhibitors, significantly decreased EV release, suggesting a critical role of GLS1 in EV release. Furthermore, addition of α-ketoglutarate or ceramide rescued EV release during BPTES treatment, implicating α-ketoglutarate and ceramide as critical downstream effectors for GLS inhibitors. These findings were further corroborated with the investigation of brain tissues in GLS1-transgenic mice. The EV levels were significantly higher in GLS1 transgenic mice than those in control mice, suggesting that GLS1 increases EV release in vivo. Conclusions These findings suggest that GLS1-mediated glutaminolysis and its downstream production of α-ketoglutarate are essential in regulating EV release during HIV-1 infection and immune activation. These new mechanistic regulations may help understand how glutamine metabolism shapes EV biogenesis and release during neuroinflammation. Electronic supplementary material The online version of this article (10.1186/s12974-018-1120-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Beiqing Wu
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jianhui Liu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Runze Zhao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yuju Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Justin Peer
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Alexander L Braun
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Lixia Zhao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yi Wang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Zenghan Tong
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yunlong Huang
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China. .,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Jialin C Zheng
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China. .,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-5930, USA.
| |
Collapse
|
29
|
Jiang Y, Lin MK, Jicha GA, Ding X, McIlwrath SL, Fardo DW, Broster LS, Schmitt FA, Kryscio R, Lipsky RH. Functional human GRIN2B promoter polymorphism and variation of mental processing speed in older adults. Aging (Albany NY) 2018; 9:1293-1306. [PMID: 28439047 PMCID: PMC5425128 DOI: 10.18632/aging.101228] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 04/17/2017] [Indexed: 02/07/2023]
Abstract
We investigated the role of a single nucleotide polymorphism rs3764030 (G>A) within the human GRIN2B promoter in mental processing speed in healthy, cognitively intact, older adults. In vitro DNA-binding and reporter gene assays of different allele combinations in transfected cells showed that the A allele was a gain-of-function variant associated with increasing GRIN2B mRNA levels. We tested the hypothesis that individuals with A allele will have better memory performance (i.e. faster reaction times) in older age. Twenty-eight older adults (ages 65-86) from a well-characterized longitudinal cohort were recruited and performed a modified delayed match-to-sample task. The rs3764030 polymorphism was genotyped and participants were grouped based on the presence of the A allele into GG and AA/AG. Carriers of the A allele maintained their speed of memory retrieval over age compared to GG carriers (p = 0.026 slope of the regression line between AA and AG versus GG groups). To validate the results, 12 older adults from the same cohort participated in a different version of the short-term memory task. Reaction times were significantly slower with age in older adults with G allele (p < 0.001). These findings support a role for rs3764030 in maintaining faster mental processing speed over aging.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Behavioral Science, University of Kentucky, Lexington, KY 40536, USA.,Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA
| | - Ming Kuan Lin
- Department of Molecular Neuroscience, the Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA
| | - Gregory A Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.,Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Xiuhua Ding
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.,Departments of Statistics and Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Sabrina L McIlwrath
- Department of Behavioral Science, University of Kentucky, Lexington, KY 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.,Departments of Statistics and Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Lucas S Broster
- Department of Behavioral Science, University of Kentucky, Lexington, KY 40536, USA
| | - Frederick A Schmitt
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.,Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Richard Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536, USA.,Departments of Statistics and Biostatistics, University of Kentucky, Lexington, KY 40536, USA
| | - Robert H Lipsky
- Department of Molecular Neuroscience, the Krasnow Institute for Advanced Study, George Mason University, Fairfax, VA 22030, USA.,Department of Neurosciences, Inova Neuroscience Institute, Inova Health System, Falls Church, VA 22042, USA
| |
Collapse
|
30
|
Glutaminase C overexpression in the brain induces learning deficits, synaptic dysfunctions, and neuroinflammation in mice. Brain Behav Immun 2017. [PMID: 28624534 PMCID: PMC5650935 DOI: 10.1016/j.bbi.2017.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glutaminolysis, a metabolic process that converts glutamine to glutamate, is particularly important for the central nervous system since glutamate is the major transmitter of excitatory synapses. Glutaminase is the mitochondrial enzyme that catalyzes the first step of glutaminolysis. Two genes encode at least four isoforms of glutaminase in humans. Gls1 gene encodes isoforms kidney-type glutaminase (KGA) and glutaminase C (GAC) through alternative splicing, whereas Gls2 gene encodes liver-type glutaminase isoforms. KGA and GAC have been associated with several neurological diseases. However, it remains unclear whether changes in their expressions can directly cause brain abnormalities. Using a transgenic approach, we generated mice that overexpressed GAC in the brain. The resulting transgenic mice had severe impairments in spatial and fear learning compared with littermate controls. The learning deficits were consistent with diminished hippocampal long-term potentiation in the hippocampal slices of the GAC transgenic mice. Furthermore, we found increases in astrocyte and microglia markers, inflammatory factors, and a decrease in synapse marker synaptophysin, suggesting neuroinflammation and synaptic changes in the GAC transgenic mouse brains. In conclusion, these findings provide the first evidence that GAC overexpression in the brain has deleterious effects on learning and synaptic integrity in vivo.
Collapse
|
31
|
Boksha IS, Prokhorova TA, Savushkina OK, Tereshkina EB. Klotho protein: Its role in aging and central nervous system pathology. BIOCHEMISTRY (MOSCOW) 2017; 82:990-1005. [DOI: 10.1134/s0006297917090024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Shivarama Shetty M, Sajikumar S. 'Tagging' along memories in aging: Synaptic tagging and capture mechanisms in the aged hippocampus. Ageing Res Rev 2017; 35:22-35. [PMID: 28065806 DOI: 10.1016/j.arr.2016.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/12/2016] [Accepted: 12/30/2016] [Indexed: 02/06/2023]
Abstract
Aging is accompanied by a general decline in the physiological functions of the body with the deteriorating organ systems. Brain is no exception to this and deficits in cognitive functions are quite common in advanced aging. Though a variety of age-related alterations are observed in the structure and function throughout the brain, certain regions show selective vulnerability. Medial temporal lobe, especially the hippocampus, is one such preferentially vulnerable region and is a crucial structure involved in the learning and long-term memory functions. Hippocampal synaptic plasticity, such as long-term potentiation (LTP) and depression (LTD), are candidate cellular correlates of learning and memory and alterations in these properties have been well documented in aging. A related phenomenon called synaptic tagging and capture (STC) has been proposed as a mechanism for cellular memory consolidation and to account for temporal association of memories. Mounting evidences from behavioral settings suggest that STC could be a physiological phenomenon. In this article, we review the recent data concerning STC and provide a framework for how alterations in STC-related mechanisms could contribute to the age-associated memory impairments. The enormity of impairment in learning and memory functions demands an understanding of age-associated memory deficits at the fundamental level given its impact in the everyday tasks, thereby in the quality of life. Such an understanding is also crucial for designing interventions and preventive measures for successful brain aging.
Collapse
|
33
|
Márquez Loza A, Elias V, Wong CP, Ho E, Bermudez M, Magnusson KR. Effects of ibuprofen on cognition and NMDA receptor subunit expression across aging. Neuroscience 2017; 344:276-292. [PMID: 28057539 DOI: 10.1016/j.neuroscience.2016.12.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 12/13/2016] [Accepted: 12/22/2016] [Indexed: 11/28/2022]
Abstract
Age-related declines in long- and short-term memory show relationships to decreases in N-methyl-d-aspartate (NMDA) receptor expression, which may involve inflammation. This study was designed to determine effects of an anti-inflammatory drug, ibuprofen, on cognitive function and NMDA receptor expression across aging. Male C57BL/6 mice (ages 5, 14, 20, and 26months) were fed ibuprofen (375ppm) in NIH31 diet or diet alone for 6weeks prior to testing. Behavioral testing using the Morris water maze showed that older mice performed significantly worse than younger in spatial long-term memory, reversal, and short-term memory tasks. Ibuprofen enhanced overall performance in the short-term memory task, but this appeared to be more related to improved executive function than memory. Ibuprofen induced significant decreases over all ages in the mRNA densities for GluN2B subunit, all GluN1 splice variants, and GluN1-1 splice forms in the frontal cortex and in protein expression of GluN2A, GluN2B and GluN1 C2' cassettes in the hippocampus. GluN1-3 splice form mRNA and C2' cassette protein were significantly increased across ages in frontal lobes of ibuprofen-treated mice. Ibuprofen did not alter expression of pro-inflammatory cytokines IL-1β and TNFα, but did reduce the area of reactive astrocyte immunostaining in frontal cortex of aged mice. Enhancement in executive function showed a relationship to increased GluN1-3 mRNA and decreased gliosis. These findings suggest that inflammation may play a role in executive function declines in aged animals, but other effects of ibuprofen on NMDA receptors appeared to be unrelated to aging or inflammation.
Collapse
Affiliation(s)
- Alejandra Márquez Loza
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Valerie Elias
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Carmen P Wong
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA; School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA.
| | - Michelle Bermudez
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| | - Kathy R Magnusson
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR 97331, USA; Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
34
|
Tian D, Tian M, Ma Z, Zhang L, Cui Y, Li J. Voluntary exercise rescues sevoflurane-induced memory impairment in aged male mice. Exp Brain Res 2016; 234:3613-3624. [PMID: 27540727 DOI: 10.1007/s00221-016-4756-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 08/10/2016] [Indexed: 12/16/2022]
Abstract
Postoperative cognitive impairment is especially common in older patients following major surgery. Although exposure to sevoflurane is known to cause memory deficits, few studies have examined the putative approaches to reduce such impairments. This study tested the hypotheses that sevoflurane exposure can decrease NR2B subunit-containing NMDA receptor activity in hippocampus of aged mice, and voluntary exercise may counteract the declining hippocampal functions. We found that long exposure (3 h/day for 3 days), but not short exposure (1 h/day for 3 days), to 3 % sevoflurane produced a long-lasting spatial memory deficits up to 3 weeks in aged mice, and such an effect was not due to the neuronal loss in the hippocampus, but was correlated with a long-term decrease in Fyn kinase expression and NR2B subunit phosphorylation in the hippocampus. Furthermore, voluntary exercise rescued sevoflurane-induced spatial memory deficits in aged mice and restored Fyn kinase expression and NR2B subunit phosphorylation in the hippocampus to a level comparable to control animals. Generally, our results suggested that Fyn-mediated NR2B subunit phosphorylation may play a critical role in sevoflurane-induced impairment in cognitive functions in aged animals, and voluntary exercise might be an important non-pharmacological approach to treatment of inhaled anesthetics-induced postoperative cognitive impairment in clinical settings.
Collapse
Affiliation(s)
- Dan Tian
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Miao Tian
- Department of Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Zhiming Ma
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China
| | - Leilei Zhang
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Yunfeng Cui
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Jinlong Li
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
35
|
Yang YJ, Zhao Y, Yu B, Xu GG, Wang W, Zhan JQ, Tang ZY, Wang T, Wei B. GluN2B-containing NMDA receptors contribute to the beneficial effects of hydrogen sulfide on cognitive and synaptic plasticity deficits in APP/PS1 transgenic mice. Neuroscience 2016; 335:170-83. [PMID: 27581687 DOI: 10.1016/j.neuroscience.2016.08.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 08/19/2016] [Accepted: 08/19/2016] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most common type of clinical dementia. Previous studies have demonstrated that hydrogen sulfide (H2S) is implicated with the pathology of AD, and exogenous H2S attenuates spatial memory impairments in AD animal models. However, the molecular mechanism by which H2S improves cognition in AD has not been fully explored. Here, we report that chronic administration of sodium hydrosulfide (NaHS, a H2S donor) elevated hippocampal H2S levels and enhanced hippocampus-dependent contextual fear memory and novel object recognition in amyloid precursor protein (APP)/presenilin-1 (PS1) transgenic mice. In parallel with these behavioral results, treating transgenic mice with NaHS reversed impaired hippocampal long-term potentiation (LTP), which is deemed as the neurobiological basis of learning and memory. At the molecular level, we found that treatment with NaHS did not affect the expression of the GluN1 and GluN2A subunits of NMDA receptor (NMDAR), but did prevent the downregulation of GluN2B subunit and restored its synaptic abundance, response and downstream signaling in the hippocampus in transgenic mice. Moreover, applying Ro 25-6981, a specific GluN2B antagonist, abolished the beneficial effects of NaHS on cognitive performance and hippocampal LTP in transgenic mice. Collectively, our results indicate that H2S can reverse cognitive and synaptic plasticity deficits in AD model mice by restoring surface GluN2B expression and the function of GluN2B-containing NMDARs.
Collapse
Affiliation(s)
- Yuan-Jian Yang
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang 330029, PR China; Medical Experimental Center, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang 330029, PR China
| | - Ying Zhao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Bin Yu
- Medical Experimental Center, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang 330029, PR China
| | - Guo-Gang Xu
- Nanlou Respiratory Department, Chinese PLA General Hospital, Beijing 100853, PR China
| | - Wei Wang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Jin-Qiong Zhan
- Medical Experimental Center, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang 330029, PR China
| | - Zhen-Yu Tang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Ting Wang
- Department of Pharmacology, College of Medical Science, Three Gorges University, Yichang 443002, PR China.
| | - Bo Wei
- Department of Psychiatry, Jiangxi Mental Hospital/Affiliated Mental Hospital of Nanchang University, Nanchang 330029, PR China.
| |
Collapse
|
36
|
Ohizumi Y. [A new strategy for preventive and functional therapeutic methods for dementia--approach using natural products]. YAKUGAKU ZASSHI 2016; 135:449-64. [PMID: 25759053 DOI: 10.1248/yakushi.14-00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) has become a serious social problem in Japan. However, effective preventive and fundamental therapeutic methods for AD have not yet been developed. Using a new strategy in the course of our survey of numerous natural resouces having neurotrophic activity, we isolated a variety of active constituents and proved their pharmacological properties. As a result, we successfully found nobiletin, a compound with anti-dementia activity that comes from citrus peels. Also, we have demonstrated that nobiletin ameliorates cognitive impairment in several dementia model animals such as chronically amyloid β(Aβ) infused rats, amyloid precursor protein transgenic (APPTg) mice, olfactory-bulbectomized (OBX) mice, N-methyl-D-aspartate (NMDA) receptor antagonist (MK-801)-treated mice, senescence-accelated mice and bilaterial common carotid arteries occlusion mice. In a APPTg mouse of AD, nobiletin greatly improved memory impairment, and this was accompanied by a marked decrease in Aβ deposition. Also, in OBX mice memory impairment was markedly recoverd by nobiletin, accompanied by improvement of a decrease indensity of cholinergic neurons. Interestingly, nobiletin improves age-related congnitive impairment and decreased hyperphosphorylation of tau as well as oxidative stress in senescence-accelerated mice. In cultured cells, nobiletin reversed the Aβ-induced inhibition of glutamate-induced increases in cAMP response element binding protein (CREB) phosphorylation and modulated gen expression of thioredoxin-interacting protein and NMDA resceptor subunits. These results suggest that nobiletin prevents memory impairment and exhibits a protecting action against neurodgeneration in AD model animals. Nobiletin and citrus peels thus have potential as functional foods for prevention of dementia.
Collapse
Affiliation(s)
- Yasushi Ohizumi
- Graduate School of Pharmaceutical Sciences, Tohoku University; 6-3 Aoba, Aramaki, Aoba-ku, Sendai 980-8578, Japan; Graduate School of Engineering, Tohoku University; 6-6-07 Aoba, Aramaki, Aoba-ku, Sendai 980-8579; Faculty of Pharmaceutical Sciences, University of Shizuoka; 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan; Kansei Research Institute, Tohoku Fukushi University; 1-19-1 Kunimi, Aoba-ku, Sendai 989-3201, Japan; Yokohama College of Pharmacy; 601 Matano-cho, Totsuka-ku, Yokohama 245-0066, Japan
| |
Collapse
|
37
|
Zamzow DR, Elias V, Acosta VA, Escobedo E, Magnusson KR. Higher levels of phosphorylated Y1472 on GluN2B subunits in the frontal cortex of aged mice are associated with good spatial reference memory, but not cognitive flexibility. AGE (DORDRECHT, NETHERLANDS) 2016; 38:50. [PMID: 27094400 PMCID: PMC5005925 DOI: 10.1007/s11357-016-9913-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 04/06/2016] [Indexed: 06/05/2023]
Abstract
The N-methyl-D-aspartate receptor (NMDAr) is particularly vulnerable to aging. The GluN2B subunit of the NMDAr, compared to other NMDAr subunits, suffers the greatest losses of expression in the aging brain, especially in the frontal cortex. While expression levels of GluN2B mRNA and protein in the aged brain are well documented, there has been little investigation into age-related posttranslational modifications of the subunit. In this study, we explored some of the mechanisms that may promote differences in the NMDAr complex in the frontal cortex of aged animals. Two ages of mice, 3 and 24 months, were behaviorally tested in the Morris water maze. The frontal cortex and hippocampus from each mouse were subjected to differential centrifugation followed by solubilization in Triton X-100. Proteins from Triton-insoluble membranes, Triton-soluble membranes, and intracellular membranes/cytosol were examined by Western blot. Higher levels of GluN2B tyrosine 1472 phosphorylation in frontal cortex synaptic fractions of old mice were associated with better reference learning but poorer cognitive flexibility. Levels of GluN2B phosphotyrosine 1336 remained steady, but there were greater levels of the calpain-induced 115 kDa GluN2B cleavage product on extrasynaptic membranes in these old good learners. There was an age-related increase in calpain activity, but it was not associated with better learning. These data highlight a unique aging change for aged mice with good spatial learning that might be detrimental to cognitive flexibility. This study also suggests that higher levels of truncated GluN2B on extrasynaptic membranes are not deleterious to spatial memory in aged mice.
Collapse
Affiliation(s)
| | - Val Elias
- Oregon State University, Corvallis, OR, USA
| | | | | | | |
Collapse
|
38
|
Lazarczyk MJ, Kemmler JE, Eyford BA, Short JA, Varghese M, Sowa A, Dickstein DR, Yuk FJ, Puri R, Biron KE, Leist M, Jefferies WA, Dickstein DL. Major Histocompatibility Complex class I proteins are critical for maintaining neuronal structural complexity in the aging brain. Sci Rep 2016; 6:26199. [PMID: 27229916 PMCID: PMC4882527 DOI: 10.1038/srep26199] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 04/27/2016] [Indexed: 12/19/2022] Open
Abstract
Major histocompatibility complex class I (MHCI) proteins have been implicated in neuronal function through the modulation of neuritogenesis, synaptogenesis, synaptic plasticity, and memory consolidation during development. However, the involvement of MHCI in the aged brain is unclear. Here we demonstrate that MHCI deficiency results in significant dendritic atrophy along with an increase in thin dendritic spines and a reduction in stubby spines in the hippocampus of aged (12 month old) mice. Ultrastructural analyses revealed a decrease in spine head diameter and post synaptic density (PSD) area, as well as an increase in overall synapse density, and non-perforated, small spines. Interestingly, we found that the changes in synapse density and morphology appear relatively late (after the age of 6 months). Finally, we found a significant age dependent increase in the levels of the glutamate receptor, GluN2B in aged MHCI knockout mice, with no change in GluA2/3, VGluT1, PSD95 or synaptophysin. These results indicate that MHCI may be also be involved in maintaining brain integrity at post-developmental stages notably in the modulation of neuronal and spine morphology and synaptic function during non-pathological aging which could have significant implications for cognitive function.
Collapse
Affiliation(s)
- Maciej J Lazarczyk
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Department of Mental Health and Psychiatry, Division of General Psychiatry, University Hospitals of Geneva, Faculty of Medicine of the University of Geneva, Geneva, Switzerland
| | - Julia E Kemmler
- University of Konstanz, Doerenkamp-Zbinden, Universitätsstrasse. 10, 78457 Konstanz, Germany
| | - Brett A Eyford
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada
| | - Jennifer A Short
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Merina Varghese
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Allison Sowa
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Daniel R Dickstein
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Frank J Yuk
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rishi Puri
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kaan E Biron
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada.,Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Marcel Leist
- University of Konstanz, Doerenkamp-Zbinden, Universitätsstrasse. 10, 78457 Konstanz, Germany
| | - Wilfred A Jefferies
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, British Columbia, V6T 1Z4, Canada.,Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.,Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.,Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada.,Department of Medical Genetics, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Dara L Dickstein
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.,Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
39
|
Foster TC, Kyritsopoulos C, Kumar A. Central role for NMDA receptors in redox mediated impairment of synaptic function during aging and Alzheimer's disease. Behav Brain Res 2016; 322:223-232. [PMID: 27180169 DOI: 10.1016/j.bbr.2016.05.012] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/15/2016] [Accepted: 05/05/2016] [Indexed: 01/07/2023]
Abstract
Increased human longevity has magnified the negative impact that aging can have on cognitive integrity of older individuals experiencing some decline in cognitive function. Approximately 30% of the elderly will have cognitive problems that influence their independence. Impaired executive function and memory performance are observed in normal aging and yet can be an early sign of a progressive cognitive impairment of Alzheimer's disease (AD), the most common form of dementia. Brain regions that are vulnerable to aging exhibit the earliest pathology of AD. Senescent synaptic function is observed as a shift in Ca2+-dependent synaptic plasticity and similar mechanisms are thought to contribute to the early cognitive deficits associated with AD. In the case of aging, intracellular redox state mediates a shift in Ca2+ regulation including N-methyl-d-aspartate (NMDA) receptor hypofunction and increased Ca2+ release from intracellular stores to alter synaptic plasticity. AD can interact with these aging processes such that molecules linked to AD, β-amyloid (Aβ) and mutated presenilin 1 (PS1), can also degrade NMDA receptor function, promote Ca2+ release from intracellular stores, and may increase oxidative stress. Thus, age is one of the most important predictors of AD and brain aging likely contributes to the onset of AD. The focus of this review article is to provide an update on mechanisms that contribute to the senescent synapse and possible interactions with AD-related molecules, with special emphasis on regulation of NMDA receptors.
Collapse
Affiliation(s)
- T C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America.
| | - C Kyritsopoulos
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America
| | - A Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, United States of America.
| |
Collapse
|
40
|
Yang P, Leu D, Ye K, Srinivasan C, Fike JR, Huang TT. Cognitive impairments following cranial irradiation can be mitigated by treatment with a tropomyosin receptor kinase B agonist. Exp Neurol 2016; 279:178-186. [PMID: 26946222 PMCID: PMC4933026 DOI: 10.1016/j.expneurol.2016.02.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 02/10/2016] [Accepted: 02/26/2016] [Indexed: 12/18/2022]
Abstract
Brain radiotherapy is frequently used successfully to treat brain tumors. However, radiotherapy is often associated with declines in short-term and long-term memory, learning ability, and verbal fluency. We previously identified a downregulation of the brain-derived neurotrophic factor (BDNF) following cranial irradiation in experimental animals. In the present study, we investigated whether targeting the BDNF high affinity receptor, tropomysin receptor kinase B (TrkB), could mitigate radiation-induced cognitive deficits. After irradiation, chronic treatment with a small molecule TrkB agonist, 7,8-dihydroxyflavone (DHF) in mice led to enhanced activation of TrkB and its downstream targets ERK and AKT, both important factors in neuronal development. DHF treatment significantly restored spatial, contextual, and working memory, and the positive effects persisted for at least 3months after completion of the treatment. Consistent with preservation of cognitive functions, chronic DHF treatment mitigated radiation-induced suppression of hippocampal neurogenesis. Spine density and major components of the excitatory synapses, including glutamate receptors and postsynaptic density protein 95 (PSD-95), were also maintained at normal levels by DHF treatment after irradiation. Taken together, our results show that chronic treatment with DHF after irradiation significantly mitigates radiation-induced cognitive defects. This is achieved most likely by preservation of hippocampal neurogenesis and synaptic plasticity.
Collapse
Affiliation(s)
- Phillip Yang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - David Leu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Palo Alto Veterans Institute for Research, Palo Alto, CA 94304, USA
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Chandra Srinivasan
- Department of Chemistry and Biochemistry, California State University Fullerton, CA 92831, USA
| | - John R Fike
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, CA 94143, USA
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA; Geriatric Research, Education, and Clinical Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
41
|
Abstract
UNLABELLED A decline in estradiol (E2)-mediated cognitive benefits denotes a critical window for the therapeutic effects of E2, but the mechanism for closing of the critical window is unknown. We hypothesized that upregulating the expression of estrogen receptor α (ERα) or estrogen receptor β (ERβ) in the hippocampus of aged animals would restore the therapeutic potential of E2 treatments and rejuvenate E2-induced hippocampal plasticity. Female rats (15 months) were ovariectomized, and, 14 weeks later, adeno-associated viral vectors were used to express ERα, ERβ, or green fluorescent protein (GFP) in the CA1 region of the dorsal hippocampus. Animals were subsequently treated for 5 weeks with cyclic injections of 17β-estradiol-3-benzoate (EB, 10 μg) or oil vehicle. Spatial memory was examined 48 h after EB/oil treatment. EB treatment in the GFP (GFP + EB) and ERβ (ERβ + EB) groups failed to improve episodic spatial memory relative to oil-treated animals, indicating closing of the critical window. Expression of ERβ failed to improve cognition and was associated with a modest learning impairment. Cognitive benefits were specific to animals expressing ERα that received EB treatment (ERα + EB), such that memory was improved relative to ERα + oil and GFP + EB. Similarly, ERα + EB animals exhibited enhanced NMDAR-mediated synaptic transmission compared with the ERα + oil and GFP + EB groups. This is the first demonstration that the window for E2-mediated benefits on cognition and hippocampal E2 responsiveness can be reinstated by increased expression of ERα. SIGNIFICANCE STATEMENT Estradiol is neuroprotective, promotes synaptic plasticity in the hippocampus, and protects against cognitive decline associated with aging and neurodegenerative diseases. However, animal models and clinical studies indicate a critical window for the therapeutic treatment such that the beneficial effects are lost with advanced age and/or with extended hormone deprivation. We used gene therapy to upregulate expression of the estrogen receptors ERα and ERβ and demonstrate that the window for estradiol's beneficial effects on memory and hippocampal synaptic function can be reinstated by enhancing the expression of ERα. Our findings suggest that the activity of ERα controls the therapeutic window by regulating synaptic plasticity mechanisms involved in memory.
Collapse
|
42
|
Montgomery KS, Edwards G, Levites Y, Kumar A, Myers CE, Gluck MA, Setlow B, Bizon JL. Deficits in hippocampal-dependent transfer generalization learning accompany synaptic dysfunction in a mouse model of amyloidosis. Hippocampus 2016; 26:455-71. [PMID: 26418152 PMCID: PMC4803574 DOI: 10.1002/hipo.22535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 11/08/2022]
Abstract
Elevated β-amyloid and impaired synaptic function in hippocampus are among the earliest manifestations of Alzheimer's disease (AD). Most cognitive assessments employed in both humans and animal models, however, are insensitive to this early disease pathology. One critical aspect of hippocampal function is its role in episodic memory, which involves the binding of temporally coincident sensory information (e.g., sights, smells, and sounds) to create a representation of a specific learning epoch. Flexible associations can be formed among these distinct sensory stimuli that enable the "transfer" of new learning across a wide variety of contexts. The current studies employed a mouse analog of an associative "transfer learning" task that has previously been used to identify risk for prodromal AD in humans. The rodent version of the task assesses the transfer of learning about stimulus features relevant to a food reward across a series of compound discrimination problems. The relevant feature that predicts the food reward is unchanged across problems, but an irrelevant feature (i.e., the context) is altered. Experiment 1 demonstrated that C57BL6/J mice with bilateral ibotenic acid lesions of hippocampus were able to discriminate between two stimuli on par with control mice; however, lesioned mice were unable to transfer or apply this learning to new problem configurations. Experiment 2 used the APPswe PS1 mouse model of amyloidosis to show that robust impairments in transfer learning are evident in mice with subtle β-amyloid-induced synaptic deficits in the hippocampus. Finally, Experiment 3 confirmed that the same transfer learning impairments observed in APPswePS1 mice were also evident in the Tg-SwDI mouse, a second model of amyloidosis. Together, these data show that the ability to generalize learned associations to new contexts is disrupted even in the presence of subtle hippocampal dysfunction and suggest that, across species, this aspect of hippocampal-dependent learning may be useful for early identification of AD-like pathology.
Collapse
Affiliation(s)
- Karienn S. Montgomery
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX
| | - George Edwards
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, University of Texas Health Science Center in Houston, Houston, TX
| | - Yona Levites
- Department of Neuroscience, University of Florida, Gainesville, FL
| | - Ashok Kumar
- Department of Neuroscience, University of Florida, Gainesville, FL
| | - Catherine E. Myers
- VA New Jersey Health Care System, East Orange, NJ 07018
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Mark A. Gluck
- Center for Molecular & Behavioral Neuroscience, Rutgers University, Newark, NJ
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL
| | | |
Collapse
|
43
|
Abraham CR, Mullen PC, Tucker-Zhou T, Chen CD, Zeldich E. Klotho Is a Neuroprotective and Cognition-Enhancing Protein. VITAMINS AND HORMONES 2016; 101:215-38. [PMID: 27125744 DOI: 10.1016/bs.vh.2016.02.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this chapter, we will describe what has been learned about Klotho and its potential functions in the brain. Klotho is localized in the choroid plexus and, to a lesser extent, in hippocampal neurons. Cognitive decline is a common issue in human aging affecting over 50% of the population. This cognitive decline can also be seen in animal models such as the Rhesus monkey. A long-term study undertaken by our lab demonstrated that normal brain aging in rhesus monkeys and other animal models is associated with a significant downregulation of Klotho expression. This observation substantiates data from other laboratories that have reported that loss of Klotho accelerates the development of aging-like phenotypes, including cognitive deficits, whereas Klotho overexpression extends life span and enhances cognition in mice and humans. Klotho is a type 1 transmembrane pleiotropic protein predominantly expressed in kidney and brain and shed by ADAM 10 and 17 into the blood and cerebral spinal fluid, respectively. While the renal functions of Klotho are well known, its roles in the brain remain to be fully elucidated. We recently demonstrated that Klotho protects hippocampal neurons from amyloid and glutamate toxicity via the activation of an antioxidant enzymatic system suggesting Klotho is a neuroprotective protein. Furthermore, Klotho is necessary for oligodendrocyte maturation and myelin integrity. Through its diverse roles in the brain, Klotho has become a new therapeutic target for neurodegenerative diseases such as Alzheimer's disease and demyelinating diseases like multiple sclerosis. Discovery of small molecule Klotho enhancers may lead to novel treatments for these incurable disorders.
Collapse
Affiliation(s)
- C R Abraham
- Boston University School of Medicine, Boston, MA, United States.
| | - P C Mullen
- Boston University School of Medicine, Boston, MA, United States
| | - T Tucker-Zhou
- Boston University School of Medicine, Boston, MA, United States
| | - C D Chen
- Boston University School of Medicine, Boston, MA, United States
| | - E Zeldich
- Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
44
|
Kumar A. NMDA Receptor Function During Senescence: Implication on Cognitive Performance. Front Neurosci 2015; 9:473. [PMID: 26732087 PMCID: PMC4679982 DOI: 10.3389/fnins.2015.00473] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 11/25/2015] [Indexed: 12/13/2022] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors, a family of L-glutamate receptors, play an important role in learning and memory, and are critical for spatial memory. These receptors are tetrameric ion channels composed of a family of related subunits. One of the hallmarks of the aging human population is a decline in cognitive function; studies in the past couple of years have demonstrated deterioration in NMDA receptor subunit expression and function with advancing age. However, a direct relationship between impaired memory function and a decline in NMDA receptors is still ambiguous. Recent studies indicate a link between an age-associated NMDA receptor hypofunction and memory impairment and provide evidence that age-associated enhanced oxidative stress might be contributing to the alterations associated with senescence. However, clear evidence is still deficient in demonstrating the underlying mechanisms and a relationship between age-associated impaired cognitive faculties and NMDA receptor hypofunction. The current review intends to present an overview of the research findings regarding changes in expression of various NMDA receptor subunits and deficits in NMDA receptor function during senescence and its implication in age-associated impaired hippocampal-dependent memory function.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Neuroscience, Evelyn F. and William L. McKnight Brain Institute, University of Florida Gainesville, FL, USA
| |
Collapse
|
45
|
Kempsell AT, Fieber LA. Age-related deficits in synaptic plasticity rescued by activating PKA or PKC in sensory neurons of Aplysia californica. Front Aging Neurosci 2015; 7:173. [PMID: 26388769 PMCID: PMC4558425 DOI: 10.3389/fnagi.2015.00173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/19/2015] [Indexed: 01/31/2023] Open
Abstract
Brain aging is associated with declines in synaptic function that contribute to memory loss, including reduced postsynaptic response to neurotransmitters and decreased neuronal excitability. To understand how aging affects memory in a simple neural circuit, we studied neuronal proxies of memory for sensitization in mature vs. advanced age Aplysia californica (Aplysia). L-Glutamate- (L-Glu-) evoked excitatory currents were facilitated by the neuromodulator serotonin (5-HT) in sensory neurons (SN) isolated from mature but not aged animals. Activation of protein kinase A (PKA) and protein kinase C (PKC) signaling rescued facilitation of L-Glu currents in aged SN. Similarly, PKA and PKC activators restored increased excitability in aged tail SN. These results suggest that altered synaptic plasticity during aging involves defects in second messenger systems.
Collapse
Affiliation(s)
- Andrew T Kempsell
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami Miami, FL, USA
| | - Lynne A Fieber
- Department of Marine Biology and Ecology, Rosenstiel School of Marine and Atmospheric Science, University of Miami Miami, FL, USA
| |
Collapse
|
46
|
Rai R, Singh HK, Prasad S. A Special Extract of Bacopa monnieri (CDRI-08) Restores Learning and Memory by Upregulating Expression of the NMDA Receptor Subunit GluN2B in the Brain of Scopolamine-Induced Amnesic Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2015; 2015:254303. [PMID: 26413117 PMCID: PMC4564605 DOI: 10.1155/2015/254303] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 02/07/2015] [Accepted: 02/16/2015] [Indexed: 12/23/2022]
Abstract
In the present communication, we have investigated effects of the CDRI-08, a well characterized extract of Bacopa monnieri, on expression of the GluN2B subunit of NMDAR in various brain regions of the scopolamine-induced amnesic mice. Our behavioral data reveal that scopolamine-treated amnesic mice exhibit significant decline in the spatial memory compared to the normal control mice. Our RT-PCR and immunoblotting data revealed that the scopolamine treatment resulted in a significant downregulation of the NMDAR GluN2B subunit expression in prefrontal cortex and hippocampus. Our enzyme assay data revealed that scopolamine caused a significant increase in the acetylcholinesterase activity in both the brain regions. Further, oral administration of the CDRI-08 to scopolamine-treated amnesic mice restored the spatial memory which was found to be associated with significant upregulation of the GluN2B subunit expression and decline in the acetylcholinesterase activity in prefrontal cortex as well as hippocampus towards their levels in the normal control mice. Our study provides the evidence for the mechanism underlying role of the Bacopa monnieri extract (CDRI-08) in restoring spatial memory in amnesic mice, which may have therapeutic implications.
Collapse
Affiliation(s)
- Rakesh Rai
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh 221 005, India
| | - Hemant K. Singh
- Lumen Research Foundation, Ashok Nagar, Chennai 600083, India
| | - S. Prasad
- Biochemistry and Molecular Biology Laboratory, Brain Research Centre, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh 221 005, India
| |
Collapse
|
47
|
Navarria L, Zaltieri M, Longhena F, Spillantini MG, Missale C, Spano P, Bellucci A. Alpha-synuclein modulates NR2B-containing NMDA receptors and decreases their levels after rotenone exposure. Neurochem Int 2015; 85-86:14-23. [PMID: 25846226 DOI: 10.1016/j.neuint.2015.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/13/2015] [Accepted: 03/31/2015] [Indexed: 02/08/2023]
Abstract
Alpha-synuclein (α-syn) is the main protein component of Lewy bodies (LBs), that together with nigrostriatal dopamine neuron loss constitute typical pathological hallmarks of Parkinson's disease (PD). Glutamate N-methyl-d-aspartate receptor (NMDAR) abnormalities, peculiarly involving NR2B-containing NMDAR, have been observed in the brain of PD patients and in several experimental models of the disease. Recent findings, indicating that α-syn can modulate NMDAR trafficking and function, suggest that this protein may be a pivotal regulator of NMDAR activity. Prompted by these evidences, we used fluorescence immunocytochemistry, western blotting and ratiometric Ca(2+) measurements to investigate whether wild type (wt) or C-terminally truncated α-syn can specifically modulate NR2B-containing NMDAR levels, subcellular trafficking and function. In addition, we evaluated whether the exposure of primary cortical neurons to increasing concentrations of rotenone could differentially regulate NR2B levels and cell viability in the presence or in the absence of α-syn. Our results indicate that both wt and C-terminally truncated α-syn negatively modulate NR2B-containing NMDAR levels, membrane translocation and function. Moreover, we found that absence of α-syn abolishes the rotenone-dependent decrease of NR2B levels and reduces neuronal vulnerability in primary cortical neurons. These findings suggest that α-syn can modulate neuronal resilience by regulating NR2B-containing NMDAR, whose specific alterations could connect α-syn pathology to neuronal degeneration in PD.
Collapse
Affiliation(s)
- Laura Navarria
- Department of Molecular and Translation Medicine, University of Brescia, Italy
| | - Michela Zaltieri
- Department of Molecular and Translation Medicine, University of Brescia, Italy
| | - Francesca Longhena
- Department of Molecular and Translation Medicine, University of Brescia, Italy
| | - Maria Grazia Spillantini
- Department of Clinical Neuroscience, The Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - Cristina Missale
- Department of Molecular and Translation Medicine, University of Brescia, Italy
| | - PierFranco Spano
- Department of Molecular and Translation Medicine, University of Brescia, Italy; IRCCS San Camillo, Venice, Italy
| | - Arianna Bellucci
- Department of Molecular and Translation Medicine, University of Brescia, Italy.
| |
Collapse
|
48
|
Hill M, Dušková M, Stárka L. Dehydroepiandrosterone, its metabolites and ion channels. J Steroid Biochem Mol Biol 2015; 145:293-314. [PMID: 24846830 DOI: 10.1016/j.jsbmb.2014.05.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Revised: 05/06/2014] [Accepted: 05/11/2014] [Indexed: 11/20/2022]
Abstract
This review is focused on the physiological and pathophysiological relevance of steroids influencing the activities of the central and peripheral nervous systems with regard to their concentrations in body fluids and tissues in various stages of human life like the fetal development or pregnancy. The data summarized in this review shows that DHEA and its unconjugated and sulfated metabolites are physiologically and pathophysiologically relevant in modulating numerous ion channels and participate in vital functions of the human organism. DHEA and its unconjugated and sulfated metabolites including 5α/β-reduced androstane steroids participate in various physiological and pathophysiological processes like the management of GnRH cyclic release, regulation of glandular and neurotransmitter secretions, maintenance of glucose homeostasis on one hand and insulin insensitivity on the other hand, control of skeletal muscle and smooth muscle activities including vasoregulation, promotion of tolerance to ischemia and other neuroprotective effects. In respect of prevalence of steroid sulfates over unconjugated steroids in the periphery and the opposite situation in the CNS, the sulfated androgens and androgen metabolites reach relevance in peripheral organs. The unconjugated androgens and estrogens are relevant in periphery and so much the more in the CNS due to higher concentrations of most unconjugated steroids in the CNS tissues than in circulation and peripheral organs. This article is part of a Special Issue entitled "Essential role of DHEA".
Collapse
Affiliation(s)
- M Hill
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - M Dušková
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| | - L Stárka
- Steroid Hormone Unit, Institute of Endocrinology, Národní třída 8, Prague 116 94, Praha 1, CZ 116 94, Czech Republic.
| |
Collapse
|
49
|
Ménard C, Quirion R, Vigneault E, Bouchard S, Ferland G, El Mestikawy S, Gaudreau P. Glutamate presynaptic vesicular transporter and postsynaptic receptor levels correlate with spatial memory status in aging rat models. Neurobiol Aging 2014; 36:1471-82. [PMID: 25556161 DOI: 10.1016/j.neurobiolaging.2014.11.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/01/2014] [Accepted: 11/21/2014] [Indexed: 11/28/2022]
Abstract
In humans, memory capacities are generally affected with aging, even without any reported neurologic disorders. The mechanisms behind cognitive decline are not well understood. We studied here whether postsynaptic glutamate receptor and presynaptic vesicular glutamate transporters (VGLUTs) levels may change in the course of aging and be related to cognitive abilities using various age-impaired (AI) or age-unimpaired rat strains. Twenty-four-month-old Long-Evans (LE) rats with intact spatial memory maintained postsynaptic ionotropic glutamate receptor levels in the hippocampal-adjacent cortex similar to those of young animals. In contrast, AI rats showed significantly reduced expression of ionotropic glutamate receptor GluR2, NR2A and NR2B subunits. In AI LE rats, VGLUT1 and VGLUT2 levels were increased and negatively correlated with receptor levels as shown by principal component analysis and correlation matrices. We also investigated whether glutamatergic receptors and VGLUT levels were altered in the obesity-resistant LOU/C/Jall (LOU) rat strain which is characterized by intact memory despite aging. No difference was observed between 24-month-old LOU rats and their young counterparts. Taken together, the unaltered spatial memory performance of 24-month-old age-unimpaired LE and LOU rats suggests that intact coordination of the presynaptic and postsynaptic hippocampal-adjacent cortex glutamatergic networks may be important for successful cognitive aging. Accordingly, altered expression of presynaptic and postsynaptic glutamatergic components, such as in AI LE rats, could be considered a marker of age-related cognitive deficits.
Collapse
Affiliation(s)
- Caroline Ménard
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center, Montreal, Quebec, Canada; Department of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Rémi Quirion
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Erika Vigneault
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada
| | - Sylvain Bouchard
- Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guylaine Ferland
- Institut Universitaire de Gériatrie de Montréal Research Center, University of Montreal, Montreal, Quebec, Canada; Department of Nutrition, University of Montreal, Montreal, Quebec, Canada
| | - Salah El Mestikawy
- Douglas Mental Health University Institute, McGill University, Montreal, Quebec, Canada; INSERM U952, CNRS UMR7224, Université Pierre et Marie Curie, Paris, France
| | - Pierrette Gaudreau
- Laboratory of Neuroendocrinology of Aging, Centre Hospitalier de l'Université de Montréal Research Center, Montreal, Quebec, Canada; Department of Medicine, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
50
|
Strong KL, Jing Y, Prosser AR, Traynelis SF, Liotta DC. NMDA receptor modulators: an updated patent review (2013-2014). Expert Opin Ther Pat 2014; 24:1349-66. [PMID: 25351527 DOI: 10.1517/13543776.2014.972938] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION The NMDA receptor mediates a slow component of excitatory synaptic transmission, and NMDA receptor dysfunction has been implicated in numerous neurological disorders. Thus, interest in developing modulators that are capable of regulating the channel continues to be strong. Recent research has led to the discovery of a number of compounds that hold therapeutic and clinical value. Deeper insight into the NMDA intersubunit interactions and structural motifs gleaned from the recently solved crystal structures of the NMDA receptor should facilitate a deeper understanding of how these compounds modulate the receptor. AREAS COVERED This article discusses the known pharmacology of NMDA receptors. A discussion of the patent literature since 2012 is also included, with an emphasis on those that claimed new chemical entities as regulators of the NMDA receptor. EXPERT OPINION The number of patents involving novel NMDA receptor modulators suggests a renewed interest in the NMDA receptor as a therapeutic target. Subunit-selective modulators continue to show promise, and the development of new subunit-selective NMDA receptor modulators appears poised for continued growth. Although a modest number of channel blocker patents were published, successful clinical outcomes involving ketamine have led to a resurgent interest in low-affinity channel blockers as therapeutics.
Collapse
Affiliation(s)
- Katie L Strong
- Emory University, Department of Chemistry , 1521 Dickey Drive, Atlanta, GA 30322 , USA
| | | | | | | | | |
Collapse
|