51
|
Craeghs L, Callaerts-Vegh Z, Verslegers M, Van der Jeugd A, Govaerts K, Dresselaers T, Wogensen E, Verreet T, Moons L, Benotmane MA, Himmelreich U, D'Hooge R. Prenatal Radiation Exposure Leads to Higher-Order Telencephalic Dysfunctions in Adult Mice That Coincide with Reduced Synaptic Plasticity and Cerebral Hypersynchrony. Cereb Cortex 2021; 32:3525-3541. [PMID: 34902856 DOI: 10.1093/cercor/bhab431] [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: 04/06/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/14/2022] Open
Abstract
Higher-order telencephalic circuitry has been suggested to be especially vulnerable to irradiation or other developmentally toxic impact. This report details the adult effects of prenatal irradiation at a sensitive time point on clinically relevant brain functions controlled by telencephalic regions, hippocampus (HPC), and prefrontal cortex (PFC). Pregnant C57Bl6/J mice were whole-body irradiated at embryonic day 11 (start of neurogenesis) with X-ray intensities of 0.0, 0.5, or 1.0 Gy. Female offspring completed a broad test battery of HPC-/PFC-controlled tasks that included cognitive performance, fear extinction, exploratory, and depression-like behaviors. We examined neural functions that are mechanistically related to these behavioral and cognitive changes, such as hippocampal field potentials and long-term potentiation, functional brain connectivity (by resting-state functional magnetic resonance imaging), and expression of HPC vesicular neurotransmitter transporters (by immunohistochemical quantification). Prenatally exposed mice displayed several higher-order dysfunctions, such as decreased nychthemeral activity, working memory defects, delayed extinction of threat-evoked response suppression as well as indications of perseverative behavior. Electrophysiological examination indicated impaired hippocampal synaptic plasticity. Prenatal irradiation also induced cerebral hypersynchrony and increased the number of glutamatergic HPC terminals. These changes in brain connectivity and plasticity could mechanistically underlie the irradiation-induced defects in higher telencephalic functions.
Collapse
Affiliation(s)
- Livine Craeghs
- Department of Brain & Cognition, Research Group Biological Psychology, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Zsuzsanna Callaerts-Vegh
- Department of Brain & Cognition, Research Group Biological Psychology, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Mieke Verslegers
- Department of Radiobiology, Institute for Environmental Health and Safety, Nuclear Research Center (SCK CEN), Mol 2400, Belgium
| | - Ann Van der Jeugd
- Department of Brain & Cognition, Research Group Biological Psychology, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Kristof Govaerts
- Department of Imaging & Pathology, Research Group Biomedical MRI, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Tom Dresselaers
- Department of Imaging & Pathology, Research Group Biomedical MRI, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Elise Wogensen
- Department of Brain & Cognition, Research Group Biological Psychology, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Tine Verreet
- Department of Radiobiology, Institute for Environmental Health and Safety, Nuclear Research Center (SCK CEN), Mol 2400, Belgium
| | - Lieve Moons
- Department of Biology, Research Group Neural Circuit Development and Regeneration, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Mohammed A Benotmane
- Department of Radiobiology, Institute for Environmental Health and Safety, Nuclear Research Center (SCK CEN), Mol 2400, Belgium
| | - Uwe Himmelreich
- Department of Imaging & Pathology, Research Group Biomedical MRI, University of Leuven (KU Leuven), Leuven 3000, Belgium
| | - Rudi D'Hooge
- Department of Brain & Cognition, Research Group Biological Psychology, University of Leuven (KU Leuven), Leuven 3000, Belgium
| |
Collapse
|
52
|
Zhvania MG, Japaridze N, Tizabi Y, Lomidze N, Pochkhidze N, Lordkipanidze T. Age-related cognitive decline in rats is sex and context dependent. Neurosci Lett 2021; 765:136262. [PMID: 34560192 DOI: 10.1016/j.neulet.2021.136262] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 12/29/2022]
Abstract
Previously, we had observed age-related cognitive decline in male rats compared to adolescent and adult rats. This was shown in both a multi-branched maze test (MBM), as well as in the Morris water maze test (MWM). In the present study, we compared the behavior of similar age groups in both male and female rats using the same paradigms. The results confirmed the increase in errors and time spent in MBM in aged male rats compared to other age groups. However, no such differences were observed in female rats. In the acquisition phase of MWM, aged male rats did not differ significantly from the other two groups in terms of time spent in quadrants, whereas aged female rats spent significantly more time in quadrants compared to the other 2 age groups. Aged male rats also travelled significantly more than the other 2 age groups during the acquisition phase, whereas no such differences were observed in female rats. In both short term (30 min post acquisition) and long term (24 h after acquisition) retrieval phases of MWM, significant gender-related differences were also observed in all age groups. These findings suggest gender- and context-dependent alterations in cognitive functions during aging.
Collapse
Affiliation(s)
- Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, 3/5 K/Cholokashvili Avenue, 0162 Tbilisi, Georgia; Department of Neuron Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Street, 9160 Tbilisi, Georgia.
| | - Nadezhda Japaridze
- Department of Neuron Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Street, 9160 Tbilisi, Georgia; School of Medicine, New Vision University, 1A Evgeni Mikeladze Street, 0159 Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Nino Lomidze
- School of Natural Sciences and Medicine, Ilia State University, 3/5 K/Cholokashvili Avenue, 0162 Tbilisi, Georgia
| | - Nino Pochkhidze
- School of Natural Sciences and Medicine, Ilia State University, 3/5 K/Cholokashvili Avenue, 0162 Tbilisi, Georgia; Department of Neuron Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Street, 9160 Tbilisi, Georgia
| | - Tamar Lordkipanidze
- School of Natural Sciences and Medicine, Ilia State University, 3/5 K/Cholokashvili Avenue, 0162 Tbilisi, Georgia; Department of Neuron Ultrastructure and Nanoarchitecture, Ivane Beritashvili Center of Experimental Biomedicine, 14 Gotua Street, 9160 Tbilisi, Georgia
| |
Collapse
|
53
|
Tian T, Qin X, Wang Y, Shi Y, Yang X. 40 Hz Light Flicker Promotes Learning and Memory via Long Term Depression in Wild-Type Mice. J Alzheimers Dis 2021; 84:983-993. [PMID: 34602491 DOI: 10.3233/jad-215212] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND 40 Hz light flicker is a well-known non-invasive treatment that is thought to be effective in treating Alzheimer's disease. However, the effects of 40 Hz visual stimulation on neural networks, synaptic plasticity, and learning and memory in wild-type animals remain unclear. OBJECTIVE We aimed to explore the impact of 40 Hz visual stimulation on synaptic plasticity, place cell, and learning and memory in wild-type mice. METHODS c-Fos+ cell distribution and in vivo electrophysiology was used to explore the effects of 40 Hz chronic visual stimulation on neural networks and neuroplasticity in wild-type mice. The character of c-Fos+ distribution in the brain and the changes of corticosterone levels in the blood were used to investigate the state of animal. Place cell analysis and novel location test were utilized to examine the effects of 40 Hz chronic visual stimulation on learning and memory in wild-type mice. RESULTS We found that 40 Hz light flicker significantly affected many brain regions that are related to stress. Also, 40 Hz induced gamma enrichment within 15 min after light flickers and impaired the expression of long-term potentiation (LTP), while facilitated the expression of long-term depression (LTD) in the hippocampal CA1. Furthermore, 40 Hz light flicker enhanced the expression of corticosterone, rendered well-formed place cells unstable and improved animal's learning and memory in novel local recognition test, which could be blocked by pre-treatment with the LTD specific blocker Glu2A-3Y. CONCLUSION These finding suggested that 40 Hz chronic light flicker contains stress effects, promoting learning and memory in wild-type mice via LTD.
Collapse
Affiliation(s)
- Tian Tian
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Xin Qin
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Yali Wang
- Key Laboratory for the Brain Research of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Yan Shi
- Faculty of Laboratory Medicine, School of Medicine, Hunan Normal University, Changsha, China
| | - Xin Yang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| |
Collapse
|
54
|
Di Cicco G, Marzano E, Iacovelli L, Celli R, van Luijtelaar G, Nicoletti F, Ngomba RT, Wall MJ. Group I metabotropic glutamate receptor-mediated long term depression is disrupted in the hippocampus of WAG/Rij rats modelling absence epilepsy. Neuropharmacology 2021; 196:108686. [PMID: 34197893 DOI: 10.1016/j.neuropharm.2021.108686] [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: 02/15/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/19/2022]
Abstract
Absence epilepsy is frequently associated with cognitive dysfunction, although the underlying mechanisms are not well understood. Here we report that some forms of hippocampal synaptic plasticity are abnormal in symptomatic Wistar Albino Glaxo/Rijswijk (WAG/Rij) rats. Metabotropic Glu 1/5 receptor-mediated long term depression (LTD) at Schaffer collateral CA1 synapses is significantly reduced in symptomatic, 5-6 months old WAG/Rij rats compared to age-matched non epileptic control rats. There were no significant changes in mGlu1/5-dependent LTD in pre-symptomatic, 4-6 weeks old WAG/Rij rats compared to age matched controls. The changes in LTD found in symptomatic WAG/Rij forms are not indicative of general deficits in all forms of synaptic plasticity as long term potentiation (LTP) was unchanged. Immunoblot analysis of hippocampal tissue showed a significant reduction in mGlu5 receptor expression, a trend to an increase in pan Homer protein levels and a decrease in GluA1 receptor expression in the hippocampus of symptomatic WAG/Rij rats vs non-epileptic control rats. There were no changes in mGlu1α receptor or GluA2 protein levels. These findings suggest that abnormalities in hippocampal mGlu5 receptor-dependent synaptic plasticity are associated with the pathological phenotype of WAG/Rij rats. This lays the groundwork for the study of mGlu5 receptors as a candidate drug target for the treatment of cognitive dysfunction linked to absence epilepsy.
Collapse
Affiliation(s)
- Gabriele Di Cicco
- Departments of Physiology and Pharmacology, University Sapienza of Rome, Italy
| | - Emanuela Marzano
- Departments of Physiology and Pharmacology, University Sapienza of Rome, Italy
| | - Luisa Iacovelli
- Departments of Physiology and Pharmacology, University Sapienza of Rome, Italy
| | | | | | - Ferdinando Nicoletti
- Departments of Physiology and Pharmacology, University Sapienza of Rome, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Richard T Ngomba
- University of Lincoln, School of Pharmacy Lincoln, United Kingdom; and, Coventry, UK.
| | - Mark J Wall
- School of Life Sciences, University of Warwick, Coventry, UK.
| |
Collapse
|
55
|
Raven F, Aton SJ. The Engram's Dark Horse: How Interneurons Regulate State-Dependent Memory Processing and Plasticity. Front Neural Circuits 2021; 15:750541. [PMID: 34588960 PMCID: PMC8473837 DOI: 10.3389/fncir.2021.750541] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
Brain states such as arousal and sleep play critical roles in memory encoding, storage, and recall. Recent studies have highlighted the role of engram neurons-populations of neurons activated during learning-in subsequent memory consolidation and recall. These engram populations are generally assumed to be glutamatergic, and the vast majority of data regarding the function of engram neurons have focused on glutamatergic pyramidal or granule cell populations in either the hippocampus, amygdala, or neocortex. Recent data suggest that sleep and wake states differentially regulate the activity and temporal dynamics of engram neurons. Two potential mechanisms for this regulation are either via direct regulation of glutamatergic engram neuron excitability and firing, or via state-dependent effects on interneuron populations-which in turn modulate the activity of glutamatergic engram neurons. Here, we will discuss recent findings related to the roles of interneurons in state-regulated memory processes and synaptic plasticity, and the potential therapeutic implications of understanding these mechanisms.
Collapse
Affiliation(s)
| | - Sara J. Aton
- Department of Molecular, Cellular, and Developmental Biology, College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
56
|
Walker CD, Kuhn CM, Risher ML. The effects of peri-adolescent alcohol use on the developing hippocampus. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 160:251-280. [PMID: 34696875 DOI: 10.1016/bs.irn.2021.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adolescence is a period of continued brain development. Regions of the brain, such as the hippocampus, continue to undergo refinement and maturation throughout adolescence and into early adulthood. Adolescence is also a time of heightened sensitivity to novelty and reward, which contribute to an increase in risk-taking behaviors including the use of drugs and alcohol. Importantly, binge drinking is highly prevalent among adolescents and emerging adults. The hippocampus which is important for the integration of emotion, reward, homeostasis, and memory is particularly vulnerable to the neurotoxic effects of alcohol. In this chapter, we cover the fundamentals of hippocampal neuroanatomy and the current state of knowledge of the acute and chronic effects of ethanol in adolescent humans and adolescent rodent models. We focus on the hippocampal-dependent behavioral, structural, and neurochemical changes and identify knowledge gaps in our understanding of age-dependent neurobiological effects of alcohol use.
Collapse
Affiliation(s)
- C D Walker
- Department of Biomedical Research, Joan C Edwards School of Medicine Marshall University, Huntington, WV, United States
| | - Cynthia M Kuhn
- Department of Pharmacology and Cancer Biology, School of Medicine, Duke University, Durham, NC, United States
| | - M-L Risher
- Department of Biomedical Research, Joan C Edwards School of Medicine Marshall University, Huntington, WV, United States; Neurobiology Research Laboratory, Hershel Woody Williams Veteran Affairs Medical Center, Huntington, WV, United States.
| |
Collapse
|
57
|
Cao Q, Wei Y, Deng J, Li J, Huang Y, Li Y, Zhang JC, Zhang Z, Lin S. NRG1 accelerates the forgetting of fear memories and facilitates the induction of long-term depression in adult mice. Psychopharmacology (Berl) 2021; 238:2535-2542. [PMID: 34189597 DOI: 10.1007/s00213-021-05877-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022]
Abstract
RATIONALE Forgetting of fear memory is a current medical therapy for posttraumatic stress disorder (PTSD), and hippocampal long-term depression (LTD) may be the underlying mechanism. Neuregulin 1 (NRG1), a trophic factor, reportedly modulates memory consolidation and synaptic plasticity. METHODS Fear memory was assessed using contextual fear conditioning. Electrophysiology was used to measure LTD and GABAergic transmission in the hippocampus. OBJECTIVES To determine the contribution of hippocampal NRG1 to fear memory forgetting and low-frequency stimulation (LFS)-induced LTD. RESULTS Administration of NRG1 in the hippocampus accelerated forgetting of contextual fear memories. Furthermore, NRG1 had no effect on low-frequency stimulation-induced LTD in young mice but significantly facilitated the induction of LTD and GABAergic transmission in adult animals. More importantly, NRG1-facilitated LTD induction in adult mice could be blocked by inhibition of GABAA receptors and NMDAR activation. CONCLUSION These findings suggest a role for NRG1 in fear memory forgetting and hippocampal LTD, providing a potential target for the development of drug-assisted PTSD therapy.
Collapse
Affiliation(s)
- Qianqian Cao
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yuan Wei
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sports University, Guangzhou, 510500, China
| | - Jialin Deng
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junfeng Li
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yanhua Huang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yuke Li
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Ji-Chun Zhang
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zili Zhang
- Department of Reproductive Medicine Center, The First People's Hospital of Foshan (Affiliated FoShan Hospital of Sun Yat-Sen University), Foshan, China.
| | - Song Lin
- Department of Physiology, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
58
|
Phillips TJ, Aldrich SJ. Peri-adolescent exposure to (meth)amphetamine in animal models. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:1-51. [PMID: 34801166 PMCID: PMC9134876 DOI: 10.1016/bs.irn.2021.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Experimentation with psychoactive drugs is often initiated in the peri-adolescent period, but knowledge of differences in the outcomes of peri-adolescent- vs adult-initiated exposure is incomplete. We consider the existing animal research in this area for (meth)amphetamines. Established for a number of phenotypes, is lower sensitivity of peri-adolescents than adults to acute effects of (meth)amphetamines, including neurotoxic effects of binge-level exposure. More variable are data for long-term consequences of peri-adolescent exposure on motivational and cognitive traits. Moreover, investigations often exclude an adult-initiated exposure group critical for answering questions about outcomes unique to peri-adolescent initiation. Despite this, it is clear from the animal research that (meth)amphetamine exposure during the peri-adolescent period, whether self- or other-administered, impacts brain motivational circuitry and cognitive function, and alters adult sensitivity to other drugs and natural rewards. Such consequences occurring in humans have the potential to predispose toward unfortunate and potentially disastrous family, social and livelihood outcomes.
Collapse
Affiliation(s)
- T J Phillips
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States; Veterans Affairs Portland Health Care System, Portland, OR, United States.
| | - S J Aldrich
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
59
|
Aagab acts as a novel regulator of NEDD4-1-mediated Pten nuclear translocation to promote neurological recovery following hypoxic-ischemic brain damage. Cell Death Differ 2021; 28:2367-2384. [PMID: 33712741 PMCID: PMC8328997 DOI: 10.1038/s41418-021-00757-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 01/31/2023] Open
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a main cause of mortality and severe neurologic impairment in the perinatal and neonatal period. However, few satisfactory therapeutic strategies are available. Here, we reported that a rapid nuclear translocation of phosphatase and tensin homolog deleted on chromosome TEN (PTEN) is an essential step in hypoxic-ischemic brain damage (HIBD)- and oxygen-glucose deprivation (OGD)-induced neuronal injures both in vivo and in vitro. In addition, we found that OGD-induced nuclear translocation of PTEN is dependent on PTEN mono-ubiquitination at the lysine 13 residue (K13) that is mediated by neural precursor cell expressed developmentally downregulated protein 4-1 (NEDD4-1). Importantly, we for the first time identified α- and γ-adaptin binding protein (Aagab) as a novel NEDD4-1 regulator to regulate the level of NEDD4-1, subsequently mediating Pten nuclear translocation. Finally, we demonstrated that genetic upregulation of Aagab or application of Tat-K13 peptide (a short interference peptide that flanks K13 residue of PTEN) not only reduced Pten nuclear translocation, but also significantly alleviated the deficits of myodynamia, motor and spatial learning and memory in HIBD model rats. These results suggest that Aagab may serve as a regulator of NEDD4-1-mediated Pten nuclear translocation to promote functional recovery following HIBD in neonatal rats, and provide a new potential therapeutic target to guide the clinical treatment for HIE.
Collapse
|
60
|
Ashby DM, Dias C, Aleksandrova LR, Lapish CC, Wang YT, Phillips AG. Disruption of Long-Term Depression Potentiates Latent Inhibition: Key Role for Central Nucleus of the Amygdala. Int J Neuropsychopharmacol 2021; 24:580-591. [PMID: 33693669 PMCID: PMC8299826 DOI: 10.1093/ijnp/pyab011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/19/2021] [Accepted: 03/05/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Latent inhibition (LI) reflects an adaptive form of learning impaired in certain forms of mental illness. Glutamate receptor activity is linked to LI, but the potential role of synaptic plasticity remains unspecified. METHODS Accordingly, the present study examined the possible role of long-term depression (LTD) in LI induced by prior exposure of rats to an auditory stimulus used subsequently as a conditional stimulus to signal a pending footshock. We employed 2 mechanistically distinct LTD inhibitors, the Tat-GluA23Y peptide that blocks endocytosis of the GluA2-containing glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor, or the selective glutamate n-methyl-d-aspartate receptor 2B antagonist, Ro25-6981, administered prior to the acquisition of 2-way conditioned avoidance with or without tone pre-exposure. RESULTS Systemic LTD blockade with the Tat-GluA23Y peptide strengthened the LI effect by further impairing acquisition of conditioned avoidance in conditional stimulus-preexposed rats compared with normal conditioning in non-preexposed controls. Systemic Ro25-6981 had no significant effects. Brain region-specific microinjections of the Tat-GluA23Y peptide into the nucleus accumbens, medial prefrontal cortex, or central or basolateral amygdala demonstrated that disruption of glutamate α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor endocytosis in the central amygdala also potentiated the LI effect. CONCLUSIONS These data revealed a previously unknown role for central amygdala LTD in LI as a key mediator of cognitive flexibility required to respond to previously irrelevant stimuli that acquire significance through reinforcement. The findings may have relevance both for our mechanistic understanding of LI and its alteration in disease states such as schizophrenia, while further elucidating the role of LTD in learning and memory.
Collapse
Affiliation(s)
- Donovan M Ashby
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carine Dias
- Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Lily R Aleksandrova
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Christopher C Lapish
- Department of Psychology, Indiana University - Purdue University Indianapolis, Indianapolis, IN, United States
| | - Yu Tian Wang
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| | - Anthony G Phillips
- Department of Psychiatry, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
61
|
Sun W, Cheng H, Yang Y, Tang D, Li X, An L. Requirements of Postnatal proBDNF in the Hippocampus for Spatial Memory Consolidation and Neural Function. Front Cell Dev Biol 2021; 9:678182. [PMID: 34336832 PMCID: PMC8319730 DOI: 10.3389/fcell.2021.678182] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/03/2021] [Indexed: 11/17/2022] Open
Abstract
Mature brain-derived neurotrophic factor (BDNF) and its downstream signaling pathways have been implicated in regulating postnatal development and functioning of rodent brain. However, the biological role of its precursor pro-brain-derived neurotrophic factor (proBDNF) in the postnatal brain remains unknown. The expression of hippocampal proBDNF was blocked in postnatal weeks, and multiple behavioral tests, Western blot and morphological techniques, and neural recordings were employed to investigate how proBDNF played a role in spatial cognition in adults. The peak expression and its crucial effects were found in the fourth but not in the second or eighth postnatal week. Blocking proBDNF expression disrupted spatial memory consolidation rather than learning or memory retrieval. Structurally, blocking proBDNF led to the reduction in spine density and proportion of mature spines. Although blocking proBDNF did not affect N-methyl-D-aspartate (NMDA) receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunits, the learning-induced phosphorylation of the GluN2B subunit level declined significantly. Functionally, paired-pulse facilitation, post-low-frequency stimulation (LFS) transiently enhanced depression, and GluN2B-dependent short-lasting long-term depression in the Schaffer collateral-CA1 pathway were weakened. The firing rate of pyramidal neurons was significantly suppressed around the target region during the memory test. Furthermore, the activation of GluN2B-mediated signaling could effectively facilitate neural function and mitigate memory impairment. The findings were consistent with the hypothesis that postnatal proBDNF played an essential role in synaptic and cognitive functions.
Collapse
Affiliation(s)
- Wei Sun
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Hong Cheng
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yang Yang
- Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongxin Tang
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiaolian Li
- Department of Neurology, Jinan Geriatric Hospital, Jinan, China
| | - Lei An
- Behavioral Neuroscience Laboratory, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Pediatric, The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Neurology, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
62
|
Lomidze N, Zhvania MG, Tizabi Y, Japaridze N, Pochkhidze N, Rzayev F, Lordkipanidze T. Aging affects cognition and hippocampal ultrastructure in male Wistar rats. Dev Neurobiol 2021; 81:833-846. [PMID: 34047044 DOI: 10.1002/dneu.22839] [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: 02/10/2021] [Revised: 04/11/2021] [Accepted: 05/16/2021] [Indexed: 12/18/2022]
Abstract
It is now well established that aging is associated with emotional and cognitive changes. Although the basis of such changes is not fully understood, ultrastructural alterations in key brain areas are likely contributing factors. Recently, we reported that aging-related anxiety in male Wistar rats is associated with ultrastructural changes in the central nucleus of amygdala, an area that plays important role in emotional regulation. In this study, we evaluated the cognitive performance of adolescent, adult, and aged male Wistar rats in multi-branch maze (MBM) as well as in Morris water maze (MWM). We also performed ultrastructural analysis of the CA1 region of the hippocampus, an area intimately involved in cognitive function. The behavioral data indicate significant impairments in few indices of cognitive functions in both tests in aged rats compared to the other two age groups. Concomitantly, a total number of presynaptic vesicles as well as vesicles in the resting pool were significantly lower, whereas postsynaptic mitochondrial area was significantly higher in aged rats compared to the other age groups. No significant differences in presynaptic terminal area or postsynaptic mitochondrial number were detected between the three age groups. These results indicate that selective ultrastructural changes in specific hippocampal region may accompany cognitive decline in aging rats.
Collapse
Affiliation(s)
- Nino Lomidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Mzia G Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Yousef Tizabi
- Department of Pharmacology Howard, University College of Medicine, Washington, District of Columbia, USA
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia.,Medical School, New Vision University, Tbilisi, Georgia
| | - Nino Pochkhidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, Ivane Beritashviloi Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Fuad Rzayev
- Department of Histology, Embryology and Cytology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Tamar Lordkipanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| |
Collapse
|
63
|
Nakauchi S, Su H, Trang I, Sumikawa K. Long-term effects of early postnatal nicotine exposure on cholinergic function in the mouse hippocampal CA1 region. Neurobiol Learn Mem 2021; 181:107445. [PMID: 33895349 PMCID: PMC9836228 DOI: 10.1016/j.nlm.2021.107445] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 03/30/2021] [Accepted: 04/20/2021] [Indexed: 01/14/2023]
Abstract
In rodent models of smoking during pregnancy, early postnatal nicotine exposure results in impaired hippocampus-dependent memory, but the underlying mechanism remains elusive. Given that hippocampal cholinergic systems modulate memory and rapid development of hippocampal cholinergic systems occurs during nicotine exposure, here we investigated its impacts on cholinergic function. Both nicotinic and muscarinic activation produce transient or long-lasting depression of excitatory synaptic transmission in the hippocampal CA1 region. We found that postnatal nicotine exposure impairs both the induction and nicotinic modulation of NMDAR-dependent long-term depression (LTD). Activation of muscarinic receptors decreases excitatory synaptic transmission and CA1 network activity in both wild-type and α2 knockout mice. These muscarinic effects are still observed in nicotine-exposed mice. M1 muscarinic receptor activity is required for mGluR-dependent LTD. Early postnatal nicotine exposure has no effect on mGluR-dependent LTD induction, suggesting that it has no effect on the function of m1 muscarinic receptors involved in this form of LTD. Our results demonstrate that early postnatal nicotine exposure has more pronounced effects on nicotinic function than muscarinic function in the hippocampal CA1 region. Thus, impaired hippocampus-dependent memory may arise from the developmental disruption of nicotinic cholinergic systems in the hippocampal CA1 region.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/growth & development
- CA1 Region, Hippocampal/metabolism
- Cigarette Smoking
- Excitatory Postsynaptic Potentials/drug effects
- Excitatory Postsynaptic Potentials/physiology
- Female
- Lactation
- Long-Term Synaptic Depression/drug effects
- Long-Term Synaptic Depression/physiology
- Male
- Maternal Exposure
- Memory/drug effects
- Memory/physiology
- Mice
- Mice, Knockout
- Nicotine/pharmacology
- Nicotinic Agonists/pharmacology
- Receptor, Muscarinic M1/drug effects
- Receptor, Muscarinic M1/metabolism
- Receptors, Metabotropic Glutamate/drug effects
- Receptors, Metabotropic Glutamate/metabolism
- Receptors, Muscarinic/drug effects
- Receptors, Muscarinic/metabolism
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Nicotinic/drug effects
- Receptors, Nicotinic/metabolism
Collapse
Affiliation(s)
- Sakura Nakauchi
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA
| | - Hailing Su
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA
| | - Ivan Trang
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA
| | - Katumi Sumikawa
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697-4550, USA.
| |
Collapse
|
64
|
Fleming SA, Hauser J, Yan J, Donovan SM, Wang M, Dilger RN. A Mediation Analysis to Identify Links between Gut Bacteria and Memory in Context of Human Milk Oligosaccharides. Microorganisms 2021; 9:846. [PMID: 33920826 PMCID: PMC8071191 DOI: 10.3390/microorganisms9040846] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
Elucidating relationships between the gut and brain is of intense research focus. Multiple studies have demonstrated that modulation of the intestinal environment via prebiotics or probiotics can induce cognitively beneficial effects, such as improved memory or reduced anxiety. However, the mechanisms by which either act remain largely unknown. We previously demonstrated that different types of oligosaccharides affected short- and long-term memory in distinct ways. Given that the oligosaccharide content of human milk is highly variable, and that formula-fed infants typically do not consume similar amounts or types of oligosaccharides, their potential effects on brain development warrant investigation. Herein, a mediation analysis was performed on existing datasets, including relative abundance of bacterial genera, gene expression, brain volume, and cognition in young pigs. Analyses revealed that numerous bacterial genera in both the colon and feces were related to short- and/or long-term memory. Relationships between genera and memory appeared to differ between diets. Mediating variables frequently included GABAergic and glutamatergic hippocampal gene expression. Other mediating variables included genes related to myelination, transcription factors, brain volume, and exploratory behavior. Overall, this analysis identified multiple pathways between the gut and brain, with a focus on genes related to excitatory/inhibitory neurotransmission.
Collapse
Affiliation(s)
| | - Jonas Hauser
- Société des Produits Nestlé SA, 1000 Lausanne, Switzerland;
| | - Jian Yan
- Nestlé Product Technology Center Nutrition, CH-1800 Vevey, Switzerland;
| | - Sharon M. Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA; (S.M.D.); (M.W.)
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Mei Wang
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL 61801, USA; (S.M.D.); (M.W.)
| | - Ryan N. Dilger
- Traverse Science, Inc., Champaign, IL 61820, USA;
- Division of Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
- Piglet Nutrition and Cognition Laboratory, Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
- Neuroscience Program, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
65
|
Zheng K, Hu F, Zhou Y, Zhang J, Zheng J, Lai C, Xiong W, Cui K, Hu YZ, Han ZT, Zhang HH, Chen JG, Man HY, Liu D, Lu Y, Zhu LQ. miR-135a-5p mediates memory and synaptic impairments via the Rock2/Adducin1 signaling pathway in a mouse model of Alzheimer's disease. Nat Commun 2021; 12:1903. [PMID: 33771994 PMCID: PMC7998005 DOI: 10.1038/s41467-021-22196-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Aberrant regulation of microRNAs (miRNAs) has been implicated in the pathogenesis of Alzheimer's disease (AD), but most abnormally expressed miRNAs found in AD are not regulated by synaptic activity. Here we report that dysfunction of miR-135a-5p/Rock2/Add1 results in memory/synaptic disorder in a mouse model of AD. miR-135a-5p levels are significantly reduced in excitatory hippocampal neurons of AD model mice. This decrease is tau dependent and mediated by Foxd3. Inhibition of miR-135a-5p leads to synaptic disorder and memory impairments. Furthermore, excess Rock2 levels caused by loss of miR-135a-5p plays an important role in the synaptic disorder of AD via phosphorylation of Ser726 on adducin 1 (Add1). Blocking the phosphorylation of Ser726 on Add1 with a membrane-permeable peptide effectively rescues the memory impairments in AD mice. Taken together, these findings demonstrate that synaptic-related miR-135a-5p mediates synaptic/memory deficits in AD via the Rock2/Add1 signaling pathway, illuminating a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Kai Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Yang Zhou
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Juan Zhang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jie Zheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuan Lai
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Wan Xiong
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ke Cui
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Ya-Zhuo Hu
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Zhi-Tao Han
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Hong-Hong Zhang
- Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Disease, Institute of Geriatrics, Chinese PLA General Hospital and Chinese PLA Medical Academy, Beijing, P. R. China
| | - Jian-Guo Chen
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA, USA
| | - Dan Liu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Youming Lu
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China.
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P. R. China.
- The Institute of Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, P. R. China.
| |
Collapse
|
66
|
Greer CB, Wright J, Weiss JD, Lazarenko RM, Moran SP, Zhu J, Chronister KS, Jin AY, Kennedy AJ, Sweatt JD, Kaas GA. Tet1 Isoforms Differentially Regulate Gene Expression, Synaptic Transmission, and Memory in the Mammalian Brain. J Neurosci 2021; 41:578-593. [PMID: 33262245 PMCID: PMC7842754 DOI: 10.1523/jneurosci.1821-20.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 11/02/2020] [Indexed: 12/22/2022] Open
Abstract
The dynamic regulation of DNA methylation in postmitotic neurons is necessary for memory formation and other adaptive behaviors. Ten-eleven translocation 1 (TET1) plays a part in these processes by oxidizing 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), thereby initiating active DNA demethylation. However, attempts to pinpoint its exact role in the nervous system have been hindered by contradictory findings, perhaps due in part, to a recent discovery that two isoforms of the Tet1 gene are differentially expressed from early development into adulthood. Here, we demonstrate that both the shorter transcript (Tet1S ) encoding an N-terminally truncated TET1 protein and a full-length Tet1 (Tet1FL ) transcript encoding canonical TET1 are co-expressed in the adult mouse brain. We show that Tet1S is the predominantly expressed isoform and is highly enriched in neurons, whereas Tet1FL is generally expressed at lower levels and more abundant in glia, suggesting their roles are at least partially cell type-specific. Using viral-mediated, isoform and neuron-specific molecular tools, we find that the individual repression of each transcript leads to the dysregulation of unique gene ensembles and contrasting changes in basal synaptic transmission. In addition, Tet1S repression enhances, while Tet1FL impairs, hippocampal-dependent memory in male mice. Together, our findings demonstrate that each Tet1 isoform serves a distinct role in the mammalian brain.SIGNIFICANCE STATEMENT In the brain, activity-dependent changes in gene expression are required for the formation of long-term memories. DNA methylation plays an essential role in orchestrating these learning-induced transcriptional programs by influencing chromatin accessibility and transcription factor binding. Once thought of as a stable epigenetic mark, DNA methylation is now known to be impermanent and dynamically regulated, driving neuroplasticity in the brain. We found that Tet1, a member of the ten-eleven translocation (TET) family of enzymes that mediates removal of DNA methyl marks, is expressed as two separate isoforms in the adult mouse brain and that each differentially regulates gene expression, synaptic transmission and memory formation. Together, our findings demonstrate that each Tet1 isoform serves a distinct role in the CNS.
Collapse
Affiliation(s)
- C B Greer
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - J Wright
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - J D Weiss
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - R M Lazarenko
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - S P Moran
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - J Zhu
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - K S Chronister
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - A Y Jin
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - A J Kennedy
- Department of Chemistry, Bates College, Lewiston, Maine 04240
| | - J D Sweatt
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - G A Kaas
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| |
Collapse
|
67
|
Tan SZK, Kim JH. mGlu5: A double-edged sword for aversive learning related therapeutics. NEUROANATOMY AND BEHAVIOUR 2021. [DOI: 10.35430/nab.2021.e16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aversive memories underlie many types of anxiety disorders. One area of research to more effectively treat anxiety disorders has therefore been identifying pharmacological targets to affect memory processes. Among these targets, the metabotropic glutamate 5 receptor (mGlu5) has received attention due to the availability of drugs to utilize its role in learning and memory. In this review, we highlight preclinical studies examining the role of mGlu5 at various stages of aversive learning and its inhibition via extinction in order to gain a better understanding of its therapeutic potential. We suggest that mGlu5 has distinct roles at different stages of memory that not only makes it a tricky target, but a double-edged sword as a therapeutic. However, the selective involvement of mGlu5 in different memory stages allows for certain precision that could be harnessed clinically. We therefore suggest potential applications, limitations, and pitfalls when considering use of mGlu5 modulators as therapeutics. In addition, we recommend future studies to address important gaps in this literature, such as sex and age factors in light of anxiety disorders being more prevalent in those demographics.
Collapse
Affiliation(s)
- Shawn Zheng Kai Tan
- School of Biomedical Science, Li Ka Shing Faculty of Medicine, The University of Hong Kong
- European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Cambridgeshire, United Kingdom
| | - Jee Hyun Kim
- IMPACT – the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC, Australia
| |
Collapse
|
68
|
Ashby DM, Floresco SB, Phillips AG, McGirr A, Seamans JK, Wang YT. LTD is involved in the formation and maintenance of rat hippocampal CA1 place-cell fields. Nat Commun 2021; 12:100. [PMID: 33397954 PMCID: PMC7782827 DOI: 10.1038/s41467-020-20317-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023] Open
Abstract
Hippocampal synaptic plasticity includes both long-term potentiation (LTP) and long-term depression (LTD) of synaptic strength, and has been implicated in shaping place field representations that form upon initial exposure to a novel environment. However, direct evidence causally linking either LTP or LTD to place fields remains limited. Here, we show that hippocampal LTD regulates the acute formation and maintenance of place fields using electrophysiology and blocking specifically LTD in freely-moving rats. We also show that exploration of a novel environment produces a widespread and pathway specific de novo synaptic depression in the dorsal hippocampus. Furthermore, disruption of this pathway-specific synaptic depression alters both the dynamics of place field formation and the stability of the newly formed place fields, affecting spatial memory in rats. These results suggest that activity-dependent synaptic depression is required for the acquisition and maintenance of novel spatial information.
Collapse
Affiliation(s)
- Donovan M Ashby
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, AB, Canada
| | - Stan B Floresco
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z7, BC, Canada
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, V6T 1Z4, BC, Canada
| | - Anthony G Phillips
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z7, BC, Canada
- Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, V6T 2A1, BC, Canada
| | - Alexander McGirr
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, T2N 4N1, AB, Canada
- Department of Psychiatry, University of Calgary, 3330 Hospital Dr NW, Calgary, T2N 4N1, AB, Canada
| | - Jeremy K Seamans
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z7, BC, Canada
- Department of Psychiatry, University of British Columbia, 2255 Wesbrook Mall, Vancouver, V6T 2A1, BC, Canada
| | - Yu Tian Wang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, V6T 1Z7, BC, Canada.
- Department of Medicine, University of British Columbia, 2775 Laurel Street, 10th Floor, Vancouver, V5Z 1M9, BC, Canada.
| |
Collapse
|
69
|
Glushchak K, Ficarro A, Schoenfeld TJ. High-fat diet and acute stress have different effects on object preference tests in rats during adolescence and adulthood. Behav Brain Res 2020; 399:112993. [PMID: 33152318 DOI: 10.1016/j.bbr.2020.112993] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/30/2020] [Accepted: 10/28/2020] [Indexed: 12/16/2022]
Abstract
Meals of high-fat diet (HFD) during adolescence produce stronger impairments to memory during adolescence than adulthood, however recovery of memory from adolescent HFD is underexplored. In addition, many tests of rodent memory are confounded by aversive or food-based stimuli, making it difficult to determine baseline memory processing affected by HFD. Thus, we utilized three cohorts of rats (adolescent HFD, adult HFD, and adolescent HFD with recovery) to explore the effects of HFD at different ages on two traditional tests of memory based strictly on object exploration, novel object recognition and novel object location tests. To isolate stress as a variable, rats were tested either at baseline or with cold water swim occurring directly after object acquisition. Results show that preference for novel objects is impaired by stress across all groups, but HFD alone only impairs preference for novel objects during adolescence, although this recovers after switching to a control diet. Additionally, preference for an object in a new location is impaired by HFD in all age groups and fails to recover following diet change. Together the data suggest that stress and HFD differentially affect object preference, based on test type, except during the adolescent period. Because these tests are traditionally interpreted as memory processes dependent on two distinct brain regions, the hippocampus and perirhinal cortex, these results support that stress and HFD affect the hippocampus and perirhinal cortex differently. The data affirm that while perirhinal cortex-dependent behavior recovers, the adolescent period is susceptible to long-lasting dysfunctions of hippocampal behavior by HFD.
Collapse
Affiliation(s)
- Karina Glushchak
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Alexandria Ficarro
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA
| | - Timothy J Schoenfeld
- Department of Psychological Science and Neuroscience, Belmont University, Nashville, TN, 37212, USA.
| |
Collapse
|
70
|
Ashourpour F, Jafari A, Babaei P. Co-treatment of AMPA endocytosis inhibitor and GluN2B antagonist facilitate consolidation and retrieval of memory impaired by β amyloid peptide. Int J Neurosci 2020; 132:714-723. [PMID: 33115292 DOI: 10.1080/00207454.2020.1837800] [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] [Indexed: 01/30/2023]
Abstract
BACKGROUND Glutamate neurotransmission stands as an important issue to minimize memory impairment. We investigated the effects of an inhibitor of α-amino-3-hydroxy-5-methyl-4-isozazole propionic acid receptors (AMPA) endocytosis and GluN2B subunit of N-methyl-d-aspartate receptors (NMDA), either isolated or combined, on memory impairments induced by Amyloid beta1-42 (Aβ). METHODS Eighty male Wistar rats were used for two experiments of consolidation and retrieval of memory. Memory impairment was induced by intracerebroventricular (ICV) injection of Aβ1-42 (2 μg/μl), and evaluated using Morris Water Maze (MWM). Each experiment consisted of 5 groups: Saline + Saline, Aβ + Saline, Aβ + Ifenprodil (Ifen, 3 nmol/ICV), Aβ +Tat-GluR23Y (3 µmol/kg/IP), and Aβ1 +Ifen + Tat-GluR23Y. Then, hippocampal cAMP-response element-binding protein (CREB) was measured by western blotting. Data were analyzed by Analysis of variance (ANOVA) repeated measure, and one-way Anova followed by Tukey's post hoc test. RESULTS During retrieval, Aβ+ Tat-GluR23Y showed significant improvement in total time spent (TTS) in the target quadrant (p = 0.009), escape latency to a platform (p = 0.008) and hippocampal level of CREB (p = 0.006) compared with Aβ + saline. Also, coadministration of Tat-GluR23Yand Ifen similar to Tat-GluR23Y alone caused significant improvement in TTS (p = 0.014) and latency to platform (p = 0.013). During consolidation, shorter escape latency (p = 0.001), longer TTS (p = 0.002) and higher level of hippocampal CREB were observed in the Aβ + Tat-GluR23Y (p = 0.001) and Aβ+ Tat-GluR23Y + Ifen (p = 0.017), respectively. CONCLUSION The present study provides pieces of evidence that inhibition of AMPARs endocytosis using Tat-GluR23Y facilitates memory consolidation and retrieval in Aβ induced memory impairment via the CREB signaling pathway.[Formula: see text].
Collapse
Affiliation(s)
- Fatemeh Ashourpour
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Adele Jafari
- Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvin Babaei
- Cellular & Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.,Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
71
|
Dashniani MG, Burjanadze MA, Chkhikvishvili NC, Solomonia RO, Kandashvili M, Naneishvili TL, Beselia GV, Kruashvili LB, Chighladze MR. Modulation of spatial memory and expression of hippocampal neurotransmitter receptors by selective lesion of medial septal cholinergic and GABAergic neurons. Exp Brain Res 2020; 238:2385-2397. [PMID: 32770352 DOI: 10.1007/s00221-020-05889-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/20/2020] [Indexed: 11/30/2022]
Abstract
The medial septum (MS) is an important modulator of hippocampal function. The degree of damage in which the particular set of septo-hippocampal projections contributes to the deficits of spatial memory with concomitant changes of hippocampal receptors expression has not been studied till present. Therefore, we investigated spatial memory and the expression level of cholinergic (α7 nACh and M1), GABAergic (α1 subunit of GABAA) and glutamatergic (NR2B subunit of NMDA and GluR 1 subunit of AMPA) receptors in the hippocampus following selective lesions of cholinergic and GABAergic septo-hippocampal projection. Learning process and long-term spatial memory were assessed using a Morris water maze. The obtained results revealed that in contrast to cholinergic lesions, rats with MS GABAergic lesions exhibit a retention deficit in 3 days after training. Western blot analyses revealed the MS cholinergic lesions have significant effect on the expression level of the M1 mACh receptors, while MS GABAergic lesions induce dramatic modulations of hippocampal glutamatergic, cholinergic and GABAergic receptors expression. These results for the first time demonstrated that selective lesions of MS cholinergic and GABAergic neurons differentially affect long-term spatial memory and the memory deficit after MS GABAergic lesion is paralleled with significant changes of hippocampal glutamate, GABA and acetylcholine receptors expression.
Collapse
Affiliation(s)
- Manana G Dashniani
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia.
| | - Maia A Burjanadze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Nino C Chkhikvishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Revaz O Solomonia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
- Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Manana Kandashvili
- Institute of Chemical Biology, Ilia State University, 0162, Tbilisi, Georgia
| | - Temur L Naneishvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Gela V Beselia
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
- Department of Physiology and Pharmacology, Petre Shotadze Tbilisi Medical Academy, 0144, Tbilisi, Georgia
| | - Lali B Kruashvili
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| | - Mariam R Chighladze
- Department of Behavior and Cognitive Function, I. Beritashvili Center of Experimental Biomedicine, 0160, Tbilisi, Georgia
| |
Collapse
|
72
|
Clinical Evidence of Antidepressant Effects of Insulin and Anti-Hyperglycemic Agents and Implications for the Pathophysiology of Depression-A Literature Review. Int J Mol Sci 2020; 21:ijms21186969. [PMID: 32971941 PMCID: PMC7554794 DOI: 10.3390/ijms21186969] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/21/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
Close connections between depression and type 2 diabetes (T2DM) have been suggested by many epidemiological and experimental studies. Disturbances in insulin sensitivity due to the disruption of various molecular pathways cause insulin resistance, which underpins many metabolic disorders, including diabetes, as well as depression. Several anti-hyperglycemic agents have demonstrated antidepressant properties in clinical trials, probably due to their action on brain targets based on the shared pathophysiology of depression and T2DM. In this article, we review reports of clinical trials examining the antidepressant effect of these medications, including insulin, metformin, glucagon like peptide-1 receptor agonists (GLP-1RA), and peroxisome proliferator-activated receptor (PPAR)-γ agonists, and briefly consider possible molecular mechanisms underlying the associations between amelioration of insulin resistance and improvement of depressive symptoms. In doing so, we intend to suggest an integrative perspective for understanding the pathophysiology of depression.
Collapse
|
73
|
Hwang H, Hur YN, Sohn H, Seo J, Hong JH, Cho E, Choi Y, Lee S, Song S, Lee AR, Kim S, Jo DG, Rhim H, Park M. Cyclin Y, a novel actin-binding protein, regulates spine plasticity through the cofilin-actin pathway. Prog Neurobiol 2020; 198:101915. [PMID: 32966834 DOI: 10.1016/j.pneurobio.2020.101915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 09/05/2020] [Accepted: 09/15/2020] [Indexed: 11/16/2022]
Abstract
While positive regulators of hippocampal long-term potentiation (LTP) have extensively been investigated, relatively little is known about the inhibitory regulators of LTP. We previously reported that Cyclin Y (CCNY), a member of cyclin family generally known to function in proliferating cells, is a novel postsynaptic protein that serves as a negative regulator of functional LTP. However, whether CCNY plays a role in structural LTP, which is mechanistically linked to functional LTP, and which mechanisms are involved in the CCNY-mediated suppression of LTP at the molecular level remain elusive. Here, we report that CCNY negatively regulates the plasticity-induced changes in spine morphology through the control of actin dynamics. We observed that CCNY directly binds to filamentous actin and interferes with LTP-induced actin polymerization as well as depolymerization by blocking the activation of cofilin, an actin-depolymerizing factor, thus resulting in less plastic spines and the impairment of structural LTP. These data suggest that CCNY acts as an inhibitory regulator for both structural and functional LTP by modulating actin dynamics through the cofilin-actin pathway. Collectively, our findings provide a mechanistic insight into the inhibitory modulation of hippocampal LTP by CCNY, highlighting a novel function of a cyclin family protein in non-proliferating neuronal cells.
Collapse
Affiliation(s)
- Hongik Hwang
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Young-Na Hur
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Heesung Sohn
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of Life Sciences, School of Natural Science, Hanyang University, Seoul 04763, South Korea
| | - Jiyeon Seo
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Jung-Hwa Hong
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Eunsil Cho
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Yuri Choi
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Saebom Lee
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Seongeun Song
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - A-Ram Lee
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Suyeon Kim
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hyewhon Rhim
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of Neuroscience, Korea University of Science and Technology, Daejeon 34113, South Korea
| | - Mikyoung Park
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, South Korea; Department of Neuroscience, Korea University of Science and Technology, Daejeon 34113, South Korea.
| |
Collapse
|
74
|
Sumi T, Harada K. Mechanism underlying hippocampal long-term potentiation and depression based on competition between endocytosis and exocytosis of AMPA receptors. Sci Rep 2020; 10:14711. [PMID: 32895399 PMCID: PMC7477194 DOI: 10.1038/s41598-020-71528-3] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/17/2020] [Indexed: 02/08/2023] Open
Abstract
N-methyl-D-aspartate (NMDA) receptor-dependent long-term potentiation (LTP) and long-term depression (LTD) of signal transmission form neural circuits and thus are thought to underlie learning and memory. These mechanisms are mediated by AMPA receptor (AMPAR) trafficking in postsynaptic neurons. However, the regulatory mechanism of bidirectional plasticity at excitatory synapses remains unclear. We present a network model of AMPAR trafficking for adult hippocampal pyramidal neurons, which reproduces both LTP and LTD. We show that the induction of both LTP and LTD is regulated by the competition between exocytosis and endocytosis of AMPARs, which are mediated by the calcium-sensors synaptotagmin 1/7 (Syt1/7) and protein interacting with C-kinase 1 (PICK1), respectively. Our result indicates that recycling endosomes containing AMPAR are always ready for Syt1/7-dependent exocytosis of AMPAR at peri-synaptic/synaptic membranes. This is because molecular motor myosin Vb constitutively transports the recycling endosome toward the membrane in a Ca2+-independent manner.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan. .,Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama, 700-8530, Japan.
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, 1-1 Hibarigaoka, Tempaku-cho, Toyohashi, Aichi, 441-8580, Japan
| |
Collapse
|
75
|
Beckmann ND, Lin WJ, Wang M, Cohain AT, Charney AW, Wang P, Ma W, Wang YC, Jiang C, Audrain M, Comella PH, Fakira AK, Hariharan SP, Belbin GM, Girdhar K, Levey AI, Seyfried NT, Dammer EB, Duong D, Lah JJ, Haure-Mirande JV, Shackleton B, Fanutza T, Blitzer R, Kenny E, Zhu J, Haroutunian V, Katsel P, Gandy S, Tu Z, Ehrlich ME, Zhang B, Salton SR, Schadt EE. Multiscale causal networks identify VGF as a key regulator of Alzheimer's disease. Nat Commun 2020; 11:3942. [PMID: 32770063 PMCID: PMC7414858 DOI: 10.1038/s41467-020-17405-z] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 06/15/2020] [Indexed: 12/31/2022] Open
Abstract
Though discovered over 100 years ago, the molecular foundation of sporadic Alzheimer's disease (AD) remains elusive. To better characterize the complex nature of AD, we constructed multiscale causal networks on a large human AD multi-omics dataset, integrating clinical features of AD, DNA variation, and gene- and protein-expression. These probabilistic causal models enabled detection, prioritization and replication of high-confidence master regulators of AD-associated networks, including the top predicted regulator, VGF. Overexpression of neuropeptide precursor VGF in 5xFAD mice partially rescued beta-amyloid-mediated memory impairment and neuropathology. Molecular validation of network predictions downstream of VGF was also achieved in this AD model, with significant enrichment for homologous genes identified as differentially expressed in 5xFAD brains overexpressing VGF. Our findings support a causal role for VGF in protecting against AD pathogenesis and progression.
Collapse
Affiliation(s)
- Noam D Beckmann
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 510120, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ariella T Cohain
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander W Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Center for Statistical Genetics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Ying-Chih Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Cheng Jiang
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Mickael Audrain
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Phillip H Comella
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amanda K Fakira
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Siddharth P Hariharan
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Gillian M Belbin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kiran Girdhar
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Allan I Levey
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Duc Duong
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - James J Lah
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Jean-Vianney Haure-Mirande
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Ben Shackleton
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Tomas Fanutza
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Eimear Kenny
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Jun Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Sema4, Stamford, CT, 06902, USA
| | - Vahram Haroutunian
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Pavel Katsel
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, JJ Peters VA Medical Center, 130 West Kingsbridge Road, Bronx, NY, 10468, USA
| | - Zhidong Tu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Michelle E Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Stephen R Salton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
| | - Eric E Schadt
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA.
- Sema4, Stamford, CT, 06902, USA.
| |
Collapse
|
76
|
Temido-Ferreira M, Ferreira DG, Batalha VL, Marques-Morgado I, Coelho JE, Pereira P, Gomes R, Pinto A, Carvalho S, Canas PM, Cuvelier L, Buée-Scherrer V, Faivre E, Baqi Y, Müller CE, Pimentel J, Schiffmann SN, Buée L, Bader M, Outeiro TF, Blum D, Cunha RA, Marie H, Pousinha PA, Lopes LV. Age-related shift in LTD is dependent on neuronal adenosine A 2A receptors interplay with mGluR5 and NMDA receptors. Mol Psychiatry 2020; 25:1876-1900. [PMID: 29950682 PMCID: PMC7387321 DOI: 10.1038/s41380-018-0110-9] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 05/02/2018] [Accepted: 05/14/2018] [Indexed: 01/31/2023]
Abstract
Synaptic dysfunction plays a central role in Alzheimer's disease (AD), since it drives the cognitive decline. An association between a polymorphism of the adenosine A2A receptor (A2AR) encoding gene-ADORA2A, and hippocampal volume in AD patients was recently described. In this study, we explore the synaptic function of A2AR in age-related conditions. We report, for the first time, a significant overexpression of A2AR in hippocampal neurons of aged humans, which is aggravated in AD patients. A similar profile of A2AR overexpression in rats was sufficient to drive age-like memory impairments in young animals and to uncover a hippocampal LTD-to-LTP shift. This was accompanied by increased NMDA receptor gating, dependent on mGluR5 and linked to enhanced Ca2+ influx. We confirmed the same plasticity shift in memory-impaired aged rats and APP/PS1 mice modeling AD, which was rescued upon A2AR blockade. This A2AR/mGluR5/NMDAR interaction might prove a suitable alternative for regulating aberrant mGluR5/NMDAR signaling in AD without disrupting their constitutive activity.
Collapse
Grants
- FCT - Fundação para a Ciência e Tecnologia
- Région Hauts de France (PARTNAIRR COGNADORA), ANR (ADORATAU and SPREADTAU), LECMA/Alzheimer Forschung Initiative, Programmes d’Investissements d’Avenir LabEx (excellence laboratory) DISTALZ (Development of Innovative Strategies for a Transdisciplinary approach to ALZheimer’s disease), France Alzheimer/Fondation de France, the FHU VasCog research network (Lille, France), Fondation pour la Recherche Médicale, Fondation Plan Alzheimer, INSERM, CNRS, Université Lille 2, Lille Métropole Communauté Urbaine, FEDER, DN2M, LICEND and CoEN.
- DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Goettingen
- ATIP/AVENIR program (Centre National de la Recherche Scientifique - CNRS)
- ATIP/AVENIR program (Centre National de la Recherche Scientifique - CNRS), by the Foundation Plan Alzheimer (Senior Innovative Grant 2010)
Collapse
Affiliation(s)
- Mariana Temido-Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Diana G Ferreira
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, Porto, Portugal
- MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, 4200-450, Porto, Portugal
| | - Vânia L Batalha
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Inês Marques-Morgado
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Joana E Coelho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Pedro Pereira
- Laboratory of Neuropathology, Department of Neurosciences, Hospital de Santa Maria, CHLN, EPE, 1649-035, Lisbon, Portugal
| | - Rui Gomes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
- Faculdade de Ciências da Universidade de Lisboa, 1749-016, Lisbon, Portugal
| | - Andreia Pinto
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Sara Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal
| | - Paula M Canas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Laetitia Cuvelier
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Valerie Buée-Scherrer
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Emilie Faivre
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Younis Baqi
- PharmaCenter Bonn, Pharmazeutische Chemie I, Pharmazeutisches Institut, University of Bonn, Bonn, Germany
- Department of Chemistry, Faculty of Science, Sultan Qaboos University, PO Box 36, Postal Code 123, Muscat, Oman
| | - Christa E Müller
- PharmaCenter Bonn, Pharmazeutische Chemie I, Pharmazeutisches Institut, University of Bonn, Bonn, Germany
| | - José Pimentel
- Laboratory of Neuropathology, Department of Neurosciences, Hospital de Santa Maria, CHLN, EPE, 1649-035, Lisbon, Portugal
| | - Serge N Schiffmann
- Laboratory of Neurophysiology, ULB Neuroscience Institute, Université Libre de Bruxelles (ULB), 1070, Brussels, Belgium
| | - Luc Buée
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Michael Bader
- Max-Delbrück-Center for Molecular Medicine (MDC), 13125, Berlin, Germany
- Charité-University Medicine, 10117, Berlin, Germany
- Institute of Biology, University of Lübeck, 23652, Lübeck, Germany
| | - Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Waldweg 33, 37073, Göttingen, Germany
- Max Planck Institute for Experimental Medicine, 37075, Göttingen, Germany
- CEDOC, Chronic Diseases Research Center, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, 1150-082, Lisbon, Portugal
- Institute of Neuroscience, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, United Kingdom
| | - David Blum
- Université de Lille, Institut National de la Santé et de la Recherche Medicale (INSERM), CHU Lille, UMR-S 1172 JPArc, "Alzheimer & Tauopathie", LabEx DISTALZ, Lille, France
| | - Rodrigo A Cunha
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Hélène Marie
- Université Côte d'Azur, CNRS UMR7276, IPMC, 06560, Valbonne, France
| | - Paula A Pousinha
- Université Côte d'Azur, CNRS UMR7276, IPMC, 06560, Valbonne, France
| | - Luísa V Lopes
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, Universidade de Lisboa, 1649-028, Lisbon, Portugal.
| |
Collapse
|
77
|
Ladjimi MH, Barbouche R, Ben Rhouma K, Sakly M, Tebourbi O, Save E. Effects of PACAP-38 and an analog, acetyl-[Ala15, Ala20] PACAP-38-propylamide, on memory consolidation in the detection of spatial novelty task in rats. Brain Res 2020; 1739:146858. [DOI: 10.1016/j.brainres.2020.146858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/17/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022]
|
78
|
Spinelli M, Fusco S, Grassi C. Brain insulin resistance impairs hippocampal plasticity. VITAMINS AND HORMONES 2020; 114:281-306. [PMID: 32723548 DOI: 10.1016/bs.vh.2020.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nutrient-related signals have been demonstrated to influence brain development and cognitive functions. In particular, insulin signaling has been shown to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Alteration of brain insulin signaling interferes with the maintenance of neural stem cell niche and neuronal activity in the hippocampus. Brain insulin resistance is also emerging as key factor causing the cognitive impairment observed in metabolic and neurodegenerative diseases. Here, we review the molecular mechanisms involved in the insulin modulation of both adult neurogenesis and synaptic activity in the hippocampus. We also summarize the effects of altered insulin sensitivity on hippocampal plasticity. Finally, we reassume the experimental and epidemiological evidence highlighting the critical role of brain insulin resistance at the crossroad between type 2 diabetes and Alzheimer's disease.
Collapse
Affiliation(s)
- Matteo Spinelli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
79
|
Shen H, Zhu H, Panja D, Gu Q, Li Z. Autophagy controls the induction and developmental decline of NMDAR-LTD through endocytic recycling. Nat Commun 2020; 11:2979. [PMID: 32532981 PMCID: PMC7293213 DOI: 10.1038/s41467-020-16794-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 05/18/2020] [Indexed: 01/08/2023] Open
Abstract
NMDA receptor-dependent long-term depression (NMDAR-LTD) is a long-lasting form of synaptic plasticity. Its expression is mediated by the removal of AMPA receptors from postsynaptic membranes. Under basal conditions, endocytosed AMPA receptors are rapidly recycled back to the plasma membrane. In NMDAR-LTD, however, they are diverted to late endosomes for degradation. The mechanism for this switch is largely unclear. Additionally, the inducibility of NMDAR-LTD is greatly reduced in adulthood. The underlying mechanism and physiological significance of this phenomenon are elusive. Here, we report that autophagy inhibition is essential for the induction and developmental dampening of NMDAR-LTD. Autophagy is inhibited during NMDAR-LTD to decrease endocytic recycling. Autophagy inhibition is both necessary and sufficient for LTD induction. In adulthood, autophagy is up-regulated to make LTD induction harder, thereby preventing the adverse effect of excessive LTD on memory consolidation. These findings reveal the unrecognized functions of autophagy in synaptic plasticity, endocytic recycling, and memory. NMDA receptor-dependent long-term depression (NMDAR-LTD) is a form of synaptic plasticity mediated by reduced recycling of AMPA receptors to the plasma membrane. Here the authors show that autophagy is a regulator of this endocytic recycling and autophagy upregulation dampens NMDAR-LTD in adulthood.
Collapse
Affiliation(s)
- Hongmei Shen
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education & Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.,Nantong Brain Hospital & Mental Health Center Affiliated to Nantong University, Nantong University, Nantong, 226005, China
| | - Huiwen Zhu
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Debabrata Panja
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Qinhua Gu
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zheng Li
- Section on Synapse Development and Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
80
|
da Costa ER, Henrique EP, da Silva JB, Pereira PDC, de Abreu CC, Fernandes TN, Magalhães NGM, de Jesus Falcão da Silva A, Guerreiro LCF, Diniz CG, Diniz CWP, Diniz DG. Changes in hippocampal astrocyte morphology of Ruddy turnstone (Arenaria interpres) during the wintering period at the mangroves of Amazon River estuary. J Chem Neuroanat 2020; 108:101805. [PMID: 32505650 DOI: 10.1016/j.jchemneu.2020.101805] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 11/15/2022]
Abstract
Astrocytes are essential for lipid neuronal metabolism in long-distance uninterrupted migratory flights, when glucose is not available as the main source of energy. We previously demonstrated in Calidris pusilla that after uninterrupted 5 days transatlantic flight, astrocytes shrink and reduce its number in the hippocampal formation. Here we shifted our attention to the wintering period and tested the hypothesis that hippocampal astrocyte morphology of A interpres will change as the wintering period progresses towards the premigration window. To that end we used Arenaria interpres, which also crosses the Atlantic Ocean and reaches the mangroves of the Amazon River estuary for wintering. Birds were captured in September/October (closer to the arrival in the coast of Bragança, Para, Brazil for wintering) and in April/May (closer to the departure towards the breeding sites) and had their brains processed for selective GFAP-astrocyte immunolabeling. Three-dimensional reconstructions of the immunostained astrocytes were performed and morphological classification was done based on hierarchical cluster and discriminant analysis of multimodal morphometric features. We found two morphological phenotypes of astrocytes in the newcomers which differentially increased its morphological complexities as wintering period progresses towards the pre-migration window. Taken together, our findings demonstrate that the long-distance non-stop flight and wintering period differentially affected the two astrocytes morphotypes, suggesting distinct physiological roles for these cells. We suggest that morphological changes during the wintering period, may be part of the adaptive plasticity of the local hippocampal circuits of A. interpres in preparation for the long journey back to their breeding sites in the north hemisphere.
Collapse
Affiliation(s)
- Emanuel Ramos da Costa
- Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brazil
| | - Ediely Pereira Henrique
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - João Batista da Silva
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Patrick Douglas Corrêa Pereira
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Cintya Castro de Abreu
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Taiany Nogueira Fernandes
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Nara Gyzely Morais Magalhães
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Anderson de Jesus Falcão da Silva
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Luma Cristina Ferreira Guerreiro
- Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brazil
| | - Cristovam Guerreiro Diniz
- Instituto Federal de Educação, Ciência e Tecnologia do Pará, Laboratório de Biologia Molecular e Neuroecologia, Rua da Escola Agrícola S/N, Vila Sinhá, CEP: 68.600-000, Bragança, Pará, Brazil
| | - Cristovam Wanderley Picanço Diniz
- Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brazil.
| | - Daniel Guerreiro Diniz
- Universidade Federal do Pará, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Laboratório de Investigações em Neurodegeneração e Infecção, Rua dos Mundurucus, 4487, Guamá, CEP: 66.073-005, Belém, Pará, Brazil; Instituto Evandro Chagas, Laboratório de Microscopia Eletrônica, Avenida Almirante Barroso, 492, Marco, CEP: 66.093-020, Belém, Pará, Brazil
| |
Collapse
|
81
|
Tyrosine phosphorylation of the AMPA receptor subunit GluA2 gates homeostatic synaptic plasticity. Proc Natl Acad Sci U S A 2020; 117:4948-4958. [PMID: 32071234 PMCID: PMC7060742 DOI: 10.1073/pnas.1918436117] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hebbian plasticity, comprised of long-term potentiation (LTP) and depression (LTD), allows neurons to encode and respond to specific stimuli; while homeostatic synaptic scaling is a counterbalancing mechanism that enables the maintenance of stable neural circuits. Both types of synaptic plasticity involve the control of postsynaptic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor (AMPAR) abundance, which is modulated by AMPAR phosphorylation. To address the necessity of GluA2 phospho-Y876 in synaptic plasticity, we generated phospho-deficient GluA2 Y876F knock-in mice. We show that, while GluA2 phospho-Y876 is not necessary for Hebbian plasticity, it is essential for both in vivo and in vitro homeostatic upscaling. Bidirectional changes in GluA2 phospho-Y876 were observed during homeostatic scaling, with a decrease during downscaling and an increase during upscaling. GluA2 phospho-Y876 is necessary for synaptic accumulation of glutamate receptor interacting protein 1 (GRIP1), a crucial scaffold protein that delivers AMPARs to synapses, during upscaling. Furthermore, increased phosphorylation at GluA2 Y876 increases GluA2 binding to GRIP1. These results demonstrate that AMPAR trafficking during homeostatic upscaling can be gated by a single phosphorylation site on the GluA2 subunit.
Collapse
|
82
|
Lobzhanidze G, Japaridze N, Lordkipanidze T, Rzayev F, MacFabe D, Zhvania M. Behavioural and brain ultrastructural changes following the systemic administration of propionic acid in adolescent male rats. Further development of a rodent model of autism. Int J Dev Neurosci 2020; 80:139-156. [PMID: 31997401 DOI: 10.1002/jdn.10011] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/08/2020] [Accepted: 01/19/2020] [Indexed: 12/18/2022] Open
Abstract
Short chain fatty acids, produced as gut microbiome metabolites but also present in the diet, exert broad effects in host physiology. Propionic acid (PPA), along with butyrate and acetate, plays a growing role in health, but also in neurological conditions. Increased PPA exposure in humans, animal models and cell lines elicit diverse behavioural and biochemical changes consistent with organic acidurias, mitochondrial disorders and autism spectrum disorders (ASD). ASD is considered a disorder of synaptic dysfunction and cell signalling, but also neuroinflammatory and neurometabolic components. We examined behaviour (Morris water and radial arm mazes) and the ultrastructure of the hippocampus and medial prefrontal cortex (electron microscopy) following a single intraperitoneal (i.p.) injection of PPA (175 mg/kg) in male adolescent rats. PPA treatment showed altered social and locomotor behaviour without changes in learning and memory. Both transient and enduring ultrastructural alterations in synapses, astro- and microglia were detected in the CA1 hippocampal area. Electron microscopic analysis showed the PPA treatment significantly decreased the total number of synaptic vesicles, presynaptic mitochondria and synapses with a symmetric active zone. Thus, brief systemic administration of this dietary and enteric short chain fatty acid produced behavioural and dynamic brain ultrastructural changes, providing further validation of the PPA model of ASD.
Collapse
Affiliation(s)
- Giorgi Lobzhanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia.,Medical School, New Vision University, Tbilisi, Georgia
| | - Tamar Lordkipanidze
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| | - Fuad Rzayev
- Laboratory of Electron Microscopy, Research Center of Azerbaijan Medical University, Baku, Azerbaijan
| | - Derrick MacFabe
- The Kilee Patchell-Evans Autism Research Group, London, ON, Canada.,Faculty of Medicine, Department of Microbiology, Center for Healthy Eating and Food Innovation, Maastricht University, Maastricht, the Netherlands
| | - Mzia Zhvania
- School of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.,Department of Brain Ultrastructure and Nanoarchitecture, I. Beritashvili Center of Experimental Biomedicine, Tbilisi, Georgia
| |
Collapse
|
83
|
Synaptic GluN2A-Containing NMDA Receptors: From Physiology to Pathological Synaptic Plasticity. Int J Mol Sci 2020; 21:ijms21041538. [PMID: 32102377 PMCID: PMC7073220 DOI: 10.3390/ijms21041538] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/21/2020] [Indexed: 12/16/2022] Open
Abstract
N-Methyl-d-Aspartate Receptors (NMDARs) are ionotropic glutamate-gated receptors. NMDARs are tetramers composed by several homologous subunits of GluN1-, GluN2-, or GluN3-type, leading to the existence in the central nervous system of a high variety of receptor subtypes with different pharmacological and signaling properties. NMDAR subunit composition is strictly regulated during development and by activity-dependent synaptic plasticity. Given the differences between GluN2 regulatory subunits of NMDAR in several functions, here we will focus on the synaptic pool of NMDARs containing the GluN2A subunit, addressing its role in both physiology and pathological synaptic plasticity as well as the contribution in these events of different types of GluN2A-interacting proteins.
Collapse
|
84
|
Harvey RE, Berkowitz LE, Hamilton DA, Clark BJ. The effects of developmental alcohol exposure on the neurobiology of spatial processing. Neurosci Biobehav Rev 2019; 107:775-794. [PMID: 31526818 PMCID: PMC6876993 DOI: 10.1016/j.neubiorev.2019.09.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/02/2019] [Accepted: 09/11/2019] [Indexed: 01/20/2023]
Abstract
The consumption of alcohol during gestation is detrimental to the developing central nervous system. One functional outcome of this exposure is impaired spatial processing, defined as sensing and integrating information pertaining to spatial navigation and spatial memory. The hippocampus, entorhinal cortex, and anterior thalamus are brain regions implicated in spatial processing and are highly susceptible to the effects of developmental alcohol exposure. Some of the observed effects of alcohol on spatial processing may be attributed to changes at the synaptic to circuit level. In this review, we first describe the impact of developmental alcohol exposure on spatial behavior followed by a summary of the development of brain areas involved in spatial processing. We then provide an examination of the consequences of prenatal and early postnatal alcohol exposure in rodents on hippocampal, anterior thalamus, and entorhinal cortex-dependent spatial processing from the cellular to behavioral level. We conclude by highlighting several unanswered questions which may provide a framework for future investigation.
Collapse
Affiliation(s)
- Ryan E Harvey
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | - Laura E Berkowitz
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | - Derek A Hamilton
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States
| | - Benjamin J Clark
- Department of Psychology, University of New Mexico, Albuquerque, NM, United States.
| |
Collapse
|
85
|
Mao Z, He S, Mesnard C, Synowicki P, Zhang Y, Chung L, Wiesman AI, Wilson TW, Monaghan DT. NMDA receptors containing GluN2C and GluN2D subunits have opposing roles in modulating neuronal oscillations; potential mechanism for bidirectional feedback. Brain Res 2019; 1727:146571. [PMID: 31786200 DOI: 10.1016/j.brainres.2019.146571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022]
Abstract
NMDA receptor (NMDAR) antagonists such as ketamine, can reproduce many of the symptoms of schizophrenia. A reliable indicator of NMDAR channel blocker action in vivo is the augmentation of neuronal oscillation power. Since the coordinated and rhythmic activation of neuronal assemblies (oscillations) is necessary for perception, cognition and working memory, their disruption (inappropriate augmentation or inhibition of oscillatory power or inter-regional coherence) both in psychiatric conditions and with NMDAR antagonists may reflect the underlying defects causing schizophrenia symptoms. NMDAR antagonists and knockout (KO) mice were used to evaluate the role of GluN2C and GluN2D NMDAR subunits in generating NMDAR antagonist-induced oscillations. We find that basal oscillatory power was elevated in GluN2C-KO mice, especially in the low gamma frequencies while there was no statistically significant difference in basal oscillations between WT and GluN2D-KO mice. Compared to wildtype (WT) mice, NMDAR channel blockers caused a greater increase in oscillatory power in GluN2C-KO mice and were relatively ineffective in inducing oscillations in GluN2D-KO mice. In contrast, preferential blockade of GluN2A- and GluN2B-containing receptors induced oscillations that did not appear to be changed in either KO animal. We propose a model wherein NMDARs containing GluN2C in astrocytes and GluN2D in interneurons serve to detect local cortical excitatory synaptic activity and provide excitatory and inhibitory feedback, respectively, to local populations of postsynaptic excitatory neurons and thereby bidirectionally modulate oscillatory power.
Collapse
Affiliation(s)
- Zhihao Mao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Shengxi He
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Christopher Mesnard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Paul Synowicki
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Yuning Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Lucy Chung
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Alex I Wiesman
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tony W Wilson
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| |
Collapse
|
86
|
Xie YC, Yao ZH, Yao XL, Pan JZ, Zhang SF, Zhang Y, Hu JC. Glucagon-Like Peptide-2 Receptor is Involved in Spatial Cognitive Dysfunction in Rats After Chronic Cerebral Hypoperfusion. J Alzheimers Dis 2019; 66:1559-1576. [PMID: 30452417 DOI: 10.3233/jad-180782] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) affects the aging population and especially patients with neurodegenerative diseases, such as Alzheimer's disease or Parkinson's disease. CCH is closely related to the cognitive dysfunction in these diseases. Glucagon-like peptide-2 receptor (GLP2R) mRNA and protein are highly expressed in the gut and in hippocampal neurons. This receptor is involved in the regulation of food intake and the control of energy balance and glucose homeostasis. The present study employed behavioral techniques, electrophysiology, western blotting, immunohistochemistry, quantitative real time polymerase chain reaction (qRT-PCR), and Golgi staining to investigate whether the expression of GLP2R changes after CCH and whether GLP2R is involved in cognitive impairment caused by CCH. Our findings show that CCH significantly decreased hippocampal GLP2R mRNA and protein levels. GLP2R upregulation could prevent CCH-induced cognitive impairment. It also improved the CCH-induced impairment of long-term potentiation and long-term depression. Additionally, GLP2R modulated after CCH the AKT-mTOR-p70S6K pathway in the hippocampus. Moreover, an upregulation of the GLP2R increased the neurogenesis in the dentate gyrus, neuronal activity, and density of dendritic spines and mushroom spines in hippocampal neurons. Our findings reveal the involvement of GLP2R via a modulation of the AKT-mTOR-p70S6K pathway in the mechanisms underlying CCH-induced impairments of spatial learning and memory. We suggest that the GLP2R and the AKT-mTOR-p70S6K pathway in the hippocampus are promising targets to treat cognition deficits in CCH.
Collapse
Affiliation(s)
- Yan-Chun Xie
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhao-Hui Yao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiao-Li Yao
- Department of Neurology, Central Hospital of Zhengzhou, Zhengzhou, China
| | - Jian-Zhen Pan
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shao-Feng Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ji-Chang Hu
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
87
|
Deng CK, Mu ZH, Miao YH, Liu YD, Zhou L, Huang YJ, Zhang F, Wang YY, Yang ZH, Qian ZY, Wang X, Guo JZ, Zhang MY, Liao XY, Wan Q, Lu D, Zou YY. Gastrodin Ameliorates Motor Learning Deficits Through Preserving Cerebellar Long-Term Depression Pathways in Diabetic Rats. Front Neurosci 2019; 13:1239. [PMID: 31824244 PMCID: PMC6883220 DOI: 10.3389/fnins.2019.01239] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 11/01/2019] [Indexed: 01/21/2023] Open
Abstract
Cognitive dysfunction is a very severe consequence of diabetes, but the underlying causes are still unclear. Recently, the cerebellum was reported to play an important role in learning and memory. Since long-term depression (LTD) is a primary cellular mechanism for cerebellar motor learning, we aimed to explore the role of cerebellar LTD pathways in diabetic rats and the therapeutic effect of gastrodin. Diabetes was induced by a single injection of streptozotocin into adult Sprague-Dawley rats. Motor learning ability was assessed by a beam walk test. Pathological changes of the cerebellum were assessed by Hematoxylin-Eosin (HE) and Nissl staining. Cellular apoptosis was assessed by anti-caspase-3 immunostaining. Protein expression levels of LTD pathway-related factors, including GluR2, protein kinase C (PKC), NR2A, and nNOS, in the cerebellar cortex were evaluated by western blotting and double immunofluorescence. The NO concentration was measured. The cellular degeneration and the apoptosis of Purkinje cells were evident in the cerebellum of diabetic rats. Protein expression levels of GluR2 (NC9W: 1.26 ± 0.12; DM9W + S: 0.81 ± 0.07), PKC (NC9W: 1.66 ± 0.10; DM9W + S: 0.58 ± 0.19), NR2A (NC9W: 1.40 ± 0.05; DM9W + S: 0.63 ± 0.06), nNOS (NC9W: 1.26 ± 0.12; DM9W + S: 0.68 ± 0.04), and NO (NC9W: 135.61 ± 31.91; DM9W + S: 64.06 ± 24.01) in the cerebellum were significantly decreased in diabetic rats. Following gastrodin intervention, the outcome of motor learning ability was significantly improved (NC9W: 6.70 ± 3.31; DM9W + S: 20.47 ± 9.43; DM9W + G: 16.04 ± 7.10). In addition, degeneration and apoptosis were ameliorated, and this was coupled with the elevation of the protein expression of the abovementioned biomarkers. Arising from the above, we concluded that gastrodin may contribute to the improvement of motor learning by protecting the LTD pathways in Purkinje cells.
Collapse
Affiliation(s)
- Cheng-Kun Deng
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China.,Department of Thoracic Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhi-Hao Mu
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Yi-He Miao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China.,Department of Orthopedics, The Fifth Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yi-Dan Liu
- Institute of Drug Discovery and Development, Kunming Pharmaceutical Corporation, Kunming, China
| | - Lei Zhou
- The Key Laboratory of Stem Cell and Regenerative Medicine of Yunnan Province, Kunming Medical University, Kunming, China
| | - Yong-Jie Huang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China.,Emergency Department, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fan Zhang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China.,The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yao-Yi Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China.,The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhi-Hong Yang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Zhong-Yi Qian
- Department of Morphological Laboratory, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xie Wang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Jia-Zhi Guo
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China
| | - Mei-Yan Zhang
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Xin-Yu Liao
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| | - Qi Wan
- Institute of Neuroregeneration and Neurorehabilitation, Department of Neurosurgery of the Affiliated Hospital, Qingdao University, Qingdao, China
| | - Di Lu
- Biomedical Engineering Research Center, Kunming Medical University, Kunming, China
| | - Ying-Ying Zou
- Department of Pathology and Pathophysiology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming, China
| |
Collapse
|
88
|
Prenatal melamine exposure impairs cognitive flexibility and hippocampal synaptic plasticity in adolescent and adult female rats. Pharmacol Biochem Behav 2019; 186:172791. [DOI: 10.1016/j.pbb.2019.172791] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 08/20/2019] [Accepted: 09/09/2019] [Indexed: 11/24/2022]
|
89
|
Contreras A, Polín E, Miguéns M, Pérez-García C, Pérez V, Ruiz-Gayo M, Morales L, Del Olmo N. Intermittent-Excessive and Chronic-Moderate Ethanol Intake during Adolescence Impair Spatial Learning, Memory and Cognitive Flexibility in the Adulthood. Neuroscience 2019; 418:205-217. [PMID: 31491502 DOI: 10.1016/j.neuroscience.2019.08.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
Intermittent and excessive ethanol consumption over very short periods of time, known as binge drinking, is common in the adolescence, considered a vulnerable period to the effects of alcohol in terms of cognitive performance. One of the brain functions most drastically affected by ethanol in adolescent individuals seems to be spatial learning and memory dependent on the hippocampus. In the current study we have focused on the long-lasting effects on spatial learning and memory of intermittent and excessive alcohol consumption compared to chronic and moderate alcohol exposure during adolescence. Five-week old male Wistar rats consumed ethanol for 24 days following two different self-administration protocols that differed in the intake pattern. Spatial learning and memory were evaluated in the radial arm maze. Hippocampal synaptic plasticity was assessed by measuring field excitatory postsynaptic potentials. Hippocampal expression of AMPA and NMDA receptor subunits as well as levels of phosphorylated Ser9-GSK3β (the inactive form of GSK3β) were also quantified. Our results show that both patterns of ethanol intake during adolescence impair spatial learning, memory and cognitive flexibility in the adulthood in a dose-dependent way. Nevertheless, changes in synaptic plasticity, gene expression and levels of inactive GSK3β depended on the pattern of ethanol intake.
Collapse
Affiliation(s)
- Ana Contreras
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Eduardo Polín
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Miguel Miguéns
- Departamento de Psicología Básica I, Facultad de Psicología, UNED, Spain
| | - Carmen Pérez-García
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Vicente Pérez
- Departamento de Psicología Básica I, Facultad de Psicología, UNED, Spain
| | - Mariano Ruiz-Gayo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Lidia Morales
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Nuria Del Olmo
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain.
| |
Collapse
|
90
|
Temido-Ferreira M, Coelho JE, Pousinha PA, Lopes LV. Novel Players in the Aging Synapse: Impact on Cognition. J Caffeine Adenosine Res 2019; 9:104-127. [PMID: 31559391 PMCID: PMC6761599 DOI: 10.1089/caff.2019.0013] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
While neuronal loss has long been considered as the main contributor to age-related cognitive decline, these alterations are currently attributed to gradual synaptic dysfunction driven by calcium dyshomeostasis and alterations in ionotropic/metabotropic receptors. Given the key role of the hippocampus in encoding, storage, and retrieval of memory, the morpho- and electrophysiological alterations that occur in the major synapse of this network-the glutamatergic-deserve special attention. We guide you through the hippocampal anatomy, circuitry, and function in physiological context and focus on alterations in neuronal morphology, calcium dynamics, and plasticity induced by aging and Alzheimer's disease (AD). We provide state-of-the art knowledge on glutamatergic transmission and discuss implications of these novel players for intervention. A link between regular consumption of caffeine-an adenosine receptor blocker-to decreased risk of AD in humans is well established, while the mechanisms responsible have only now been uncovered. We review compelling evidence from humans and animal models that implicate adenosine A2A receptors (A2AR) upsurge as a crucial mediator of age-related synaptic dysfunction. The relevance of this mechanism in patients was very recently demonstrated in the form of a significant association of the A2AR-encoding gene with hippocampal volume (synaptic loss) in mild cognitive impairment and AD. Novel pathways implicate A2AR in the control of mGluR5-dependent NMDAR activation and subsequent Ca2+ dysfunction upon aging. The nature of this receptor makes it particularly suited for long-term therapies, as an alternative for regulating aberrant mGluR5/NMDAR signaling in aging and disease, without disrupting their crucial constitutive activity.
Collapse
Affiliation(s)
- Mariana Temido-Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joana E. Coelho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Paula A. Pousinha
- Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRS UMR7275, Université Côte d'Azur, Valbonne, France
| | - Luísa V. Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
91
|
Spinelli M, Fusco S, Grassi C. Brain Insulin Resistance and Hippocampal Plasticity: Mechanisms and Biomarkers of Cognitive Decline. Front Neurosci 2019; 13:788. [PMID: 31417349 PMCID: PMC6685093 DOI: 10.3389/fnins.2019.00788] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
In the last decade, much attention has been devoted to the effects of nutrient-related signals on brain development and cognitive functions. A turning point was the discovery that brain areas other than the hypothalamus expressed receptors for hormones related to metabolism. In particular, insulin signaling has been demonstrated to impact on molecular cascades underlying hippocampal plasticity, learning and memory. Here, we summarize the molecular evidence linking alteration of hippocampal insulin sensitivity with changes of both adult neurogenesis and synaptic plasticity. We also review the epidemiological studies and experimental models emphasizing the critical role of brain insulin resistance at the crossroad between metabolic and neurodegenerative disease. Finally, we brief novel findings suggesting how biomarkers of brain insulin resistance, involving the study of brain-derived extracellular vesicles and brain glucose metabolism, may predict the onset and/or the progression of cognitive decline.
Collapse
Affiliation(s)
- Matteo Spinelli
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Salvatore Fusco
- Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Claudio Grassi
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
92
|
Pinto-Duarte A, Roberts AJ, Ouyang K, Sejnowski TJ. Impairments in remote memory caused by the lack of Type 2 IP 3 receptors. Glia 2019; 67:1976-1989. [PMID: 31348567 DOI: 10.1002/glia.23679] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 11/10/2022]
Abstract
The second messenger inositol 1,4,5-trisphosphate (IP3 ) is paramount for signal transduction in biological cells, mediating Ca2+ release from the endoplasmic reticulum. Of the three isoforms of IP3 receptors identified in the nervous system, Type 2 (IP3 R2) is the main isoform expressed by astrocytes. The complete lack of IP3 R2 in transgenic mice was shown to significantly disrupt Ca2+ signaling in astrocytes, while leaving neuronal intracellular pathways virtually unperturbed. Whether and how this predominantly nonneuronal receptor might affect long-term memory function has been a matter of intense debate. In this work, we found that the absence of IP3 R2-mediated signaling did not disrupt normal learning or recent (24-48 h) memory. Contrary to expectations, however, mice lacking IP3 R2 exhibited remote (2-4 weeks) memory deficits. Not only did the lack of IP3 R2 impair remote recognition, fear, and spatial memories, but it also prevented naturally occurring post-encoding memory enhancements consequent to memory consolidation. Consistent with the key role played by the downscaling of synaptic transmission in memory consolidation, we found that NMDAR-dependent long-term depression was abnormal in ex vivo hippocampal slices acutely prepared from IP3 R2-deficient mice, a deficit that could be prevented upon supplementation with D-serine - an NMDA-receptor co-agonist whose synthesis depends upon astrocytes' activity. Our results reveal that IP3 R2 activation, which in the brain is paramount for Ca2+ signaling in astrocytes, but not in neurons, can help shape brain plasticity by enhancing the consolidation of newly acquired information into long-term memories that can guide remote cognitive behaviors.
Collapse
Affiliation(s)
- António Pinto-Duarte
- Computational Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California.,Institute for Neural Computation, University of California San Diego, La Jolla, California
| | - Amanda J Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, California
| | - Kunfu Ouyang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Terrence J Sejnowski
- Computational Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, California.,Institute for Neural Computation, University of California San Diego, La Jolla, California.,Division of Biological Sciences, University of California San Diego, La Jolla, California
| |
Collapse
|
93
|
Martín‐Segura A, Ahmed T, Casadomé‐Perales Á, Palomares‐Perez I, Palomer E, Kerstens A, Munck S, Balschun D, Dotti CG. Age-associated cholesterol reduction triggers brain insulin resistance by facilitating ligand-independent receptor activation and pathway desensitization. Aging Cell 2019; 18:e12932. [PMID: 30884121 PMCID: PMC6516156 DOI: 10.1111/acel.12932] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 01/08/2023] Open
Abstract
In the brain, insulin plays an important role in cognitive processes. During aging, these faculties decline, as does insulin signaling. The mechanism behind this last phenomenon is unclear. In recent studies, we reported that the mild and gradual loss of cholesterol in the synaptic fraction of hippocampal neurons during aging leads to a decrease in synaptic plasticity evoked by glutamate receptor activation and also by receptor tyrosine kinase (RTK) signaling. As insulin and insulin growth factor activity are dependent on tyrosine kinase receptors, we investigated whether the constitutive loss of brain cholesterol is also involved in the decay of insulin function with age. Using long‐term depression (LTD) induced by application of insulin to hippocampal slices as a read‐out, we found that the decline in insulin function during aging could be monitored as a progressive impairment of insulin‐LTD. The application of a cholesterol inclusion complex, which donates cholesterol to the membrane and increases membrane cholesterol levels, rescued the insulin signaling deficit and insulin‐LTD. In contrast, extraction of cholesterol from hippocampal neurons of adult mice produced the opposite effect. Furthermore, in vivo inhibition of Cyp46A1, an enzyme involved in brain cholesterol loss with age, improved insulin signaling. Fluorescence resonance energy transfer (FRET) experiments pointed to a change in receptor conformation by reduced membrane cholesterol, favoring ligand‐independent autophosphorylation. Together, these results indicate that changes in membrane fluidity of brain cells during aging play a key role in the decay of synaptic plasticity and cognition that occurs at this late stage of life.
Collapse
Affiliation(s)
- Adrián Martín‐Segura
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa CSIC/UAM Madrid Spain
- Department of Developmental and Molecular Biology Albert Einstein College of Medicine Bronx New York
| | - Tariq Ahmed
- Faculty of Psychology & Educational Sciences University of Leuven Leuven Belgium
- Neurological Disorders Research Center QBRI‐HBKU Doha Qatar
| | - Álvaro Casadomé‐Perales
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa CSIC/UAM Madrid Spain
| | - Irene Palomares‐Perez
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa CSIC/UAM Madrid Spain
| | - Ernest Palomer
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa CSIC/UAM Madrid Spain
- Cell & Developmental Biology Department University College London London UK
| | - Axelle Kerstens
- Department of Neuroscience, VIB Center for Brain and Disease Research University of Leuven Leuven Belgium
| | - Sebastian Munck
- Department of Neuroscience, VIB Center for Brain and Disease Research University of Leuven Leuven Belgium
| | - Detlef Balschun
- Faculty of Psychology & Educational Sciences University of Leuven Leuven Belgium
| | - Carlos G. Dotti
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa CSIC/UAM Madrid Spain
| |
Collapse
|
94
|
Matsuda S, Kakegawa W, Yuzaki M. PhotonSABER: new tool shedding light on endocytosis and learning mechanisms in vivo. Commun Integr Biol 2019; 12:34-37. [PMID: 31143361 PMCID: PMC6527187 DOI: 10.1080/19420889.2019.1586048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 02/15/2019] [Indexed: 11/01/2022] Open
Abstract
In the central nervous system, activity-dependent endocytosis of postsynaptic AMPA-type glutamate receptors (AMPA receptors) is thought to mediate long-term depression (LTD), which is a synaptic plasticity model in various neuronal circuits. However, whether and how AMPA receptor endocytosis and LTD at specific synapses are causally linked to learning and memory in vivo remains unclear. Recently, we developed a new optogenetic tool, PhotonSABER, which could control AMPA receptor endocytosis in temporal, spatial, and cell-type-specific manners at activated synapses. Using PhotonSABER, we found that AMPA receptor endocytosis and LTD at synapses between parallel fibers and Purkinje cells in the cerebellum mediate oculomotor learning. We also found that PhotonSABER could inhibit endocytosis of epidermal growth factor receptors in HeLa cells upon light stimulation. These results demonstrate that PhotonSABER is a powerful tool for analyzing the physiological functions of endocytosis in non-neuronal cells, as well as the roles of LTD in various brain regions.
Collapse
Affiliation(s)
- Shinji Matsuda
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.,Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo, Japan.,Brain Science Inspired Life Support Research Center (BLSC), The University of Electro-Communications, Tokyo, Japan
| | - Wataru Kakegawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Michisuke Yuzaki
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
95
|
Effects of early postnatal MK-801 treatment on behavioral properties in rats: Differences according to treatment schedule. Behav Brain Res 2019; 370:111926. [PMID: 31029708 DOI: 10.1016/j.bbr.2019.111926] [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: 01/31/2019] [Revised: 03/24/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022]
Abstract
It has been proposed that animals administered early postnatal NMDA (N-methyl-d-aspartate) glutamate receptor antagonists represent a model of schizophrenia; however, drug treatment schedules remain quite different among these animal studies. In this study, we compared the behavioral effects of long-term (14-day) and short-term (5-day) early postnatal treatment of the NMDA receptor antagonist MK-801 (dizocilpine; 5-methyl-10,11-dihydro-5H-dibenzo[a,d]-cyclohepten-5,10-imine). In addition, different drug treatment periods were applied to the short-term treatment study in order to determine the critical developmental period of drug effects. For experiment 1, rats were treated with MK-801 (0.2 or 0.4 mg/kg, twice daily) during postnatal days (PNDs) 7-20. For experiment 2, MK-801 (0.2 mg/kg, twice daily) was administered during the periods of PNDs 7-11, 12-16, and 17-21. In adulthood, several behavioral tests, including prepulse inhibition, open-field, and spontaneous alternation tests, were performed in experiments 1 and 2. The delayed nonmatching-to-position task was also conducted in experiment 2 on separate rats treated for 5 days in the same manner. Our results indicated that the 14-day MK-801 treatment inhibited the prepulse inhibition and decreased immobility in the forced-swim test, whereas the 5-day MK-801 treatment induced only slight behavioral effects. Collectively, our findings suggest that long-term early postnatal treatment with an NMDA receptor antagonist may be detrimental to some behavioral functions, such as sensorimotor gating and stress coping; however, treatment for longer periods is needed to elicit detrimental effects.
Collapse
|
96
|
Fan KM, Qiu LJ, Ma N, Du YN, Qian ZQ, Wei CL, Han J, Ren W, Shi MM, Liu ZQ. Acute Stress Facilitates LTD Induction at Glutamatergic Synapses in the Hippocampal CA1 Region by Activating μ-Opioid Receptors on GABAergic Neurons. Front Neurosci 2019; 13:71. [PMID: 30800053 PMCID: PMC6375894 DOI: 10.3389/fnins.2019.00071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/23/2019] [Indexed: 12/15/2022] Open
Abstract
Acute stress impairs recall memory through the facilitation of long-term depression (LTD) of hippocampal synaptic transmission. The endogenous opioid system (EOS) plays essential roles in stress-related emotional and physiological responses. Specifically, behavioral studies have shown that the impairment of memory retrieval induced by stressful events involves the activation of opioid receptors. However, it is unclear whether signaling mediated by μ-opioid receptors (μRs), one of the three major opioid receptors, participates in acute stress-related hippocampal LTD facilitation. Here, we examined the effects of a single elevated platform (EP) stress exposure on excitatory synaptic transmission and plasticity at the Schaffer collateral-commissural (SC) to CA1 synapses by recording electrically evoked field excitatory postsynaptic potentials and population spikes of hippocampal pyramidal neurons in anesthetized adult mice. EP stress exposure attenuated GABAergic feedforward and feedback inhibition of CA1 pyramidal neurons and facilitated low-frequency stimulation (LFS)-induced long-term depression (LTD) at SC-CA1 glutamatergic synapses. These effects were reproduced by exogenously activating μRs in unstressed mice. The specific deletion of μRs on GABAergic neurons (μRGABA) not only prevented the EP stress-induced memory impairment but also reversed the EP stress-induced attenuation of GABAergic inhibition and facilitation of LFS-LTD. Our results suggest that acute stress endogenously activates μRGABA to attenuate hippocampal GABAergic signaling, thereby facilitating LTD induction at excitatory synapses and eliciting memory impairments.
Collapse
Affiliation(s)
- Ka-Min Fan
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Li-Juan Qiu
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Ning Ma
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Yi-Nan Du
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Zhao-Qiang Qian
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Chun-Ling Wei
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Jing Han
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Wei Ren
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Mei-Mei Shi
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| | - Zhi-Qiang Liu
- MOE Key Laboratory of Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
97
|
Carvalho C, Cardoso SM, Correia SC, Moreira PI. Tortuous Paths of Insulin Signaling and Mitochondria in Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1128:161-183. [PMID: 31062330 DOI: 10.1007/978-981-13-3540-2_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Due to the exponential growth of aging population worldwide, neurodegenerative diseases became a major public health concern. Among them, Alzheimer's disease (AD) prevails as the most common in the elderly, rendering it a research priority. After several decades considering the brain as an insulin-insensitive organ, recent advances proved a central role for this hormone in learning and memory processes and showed that AD shares a high number of features with systemic conditions characterized by insulin resistance. Mitochondrial dysfunction has also been widely demonstrated to play a major role in AD development supporting the idea that this neurodegenerative disease is characterized by a pronounced metabolic dysregulation. This chapter is intended to discuss evidence demonstrating the key role of insulin signaling and mitochondrial anomalies in AD.
Collapse
Affiliation(s)
- Cristina Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Susana M Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Sónia C Correia
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal. .,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
98
|
Sun W, Li X, Tang C, An L. Acute Low Alcohol Disrupts Hippocampus-Striatum Neural Correlate of Learning Strategy by Inhibition of PKA/CREB Pathway in Rats. Front Pharmacol 2018; 9:1439. [PMID: 30574089 PMCID: PMC6291496 DOI: 10.3389/fphar.2018.01439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 11/22/2018] [Indexed: 11/30/2022] Open
Abstract
The hippocampus and striatum guide place-strategy and response-strategy learning, respectively, and they have dissociable roles in memory systems, which could compensate in case of temporary or permanent damage. Although acute alcohol (AA) treatment had been shown to have adverse effects on hippocampal function, whether it causes the functional compensation and the underlying mechanisms is unknown. In this study, rats treated with a low dose of AA avoided a hippocampus-dependent spatial strategy, instead preferring a striatum-dependent response strategy. Consistently, the learning-induced increase in hippocampal, but not striatal, pCREB was rendered less pronounced due to diminished activity of pPKA, but not pERK or pCaMKII. As rats approached the turn-decision area, Sp-cAMP, a PKA activator, was found to mitigate the inhibitory effect of AA on intra- and cross-structure synchronized neuronal oscillations, and rescue response-strategy bias and spatial learning deficits. Our study provides strong evidence of the critical link between neural couplings and strategy selection. Moreover, the PKA/CREB-signaling pathway is involved in the suppressive effect of AA on neural correlates of place-learning strategy. The novel important evidence provided here shows the functional couplings between the hippocampus and striatum in spatial learning processing and suggests possible avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Sun
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoliang Li
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunzhi Tang
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei An
- Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.,College of Acupuncture-Moxibustion and Orthopedics, Guiyang University of Chinese Medicine, Guiyang, China.,Department of Physiology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
99
|
Hilal ML, Moreau MM, Racca C, Pinheiro VL, Piguel NH, Santoni MJ, Dos Santos Carvalho S, Blanc JM, Abada YSK, Peyroutou R, Medina C, Doat H, Papouin T, Vuillard L, Borg JP, Rachel R, Panatier A, Montcouquiol M, Oliet SHR, Sans N. Activity-Dependent Neuroplasticity Induced by an Enriched Environment Reverses Cognitive Deficits in Scribble Deficient Mouse. Cereb Cortex 2018; 27:5635-5651. [PMID: 28968740 DOI: 10.1093/cercor/bhw333] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Indexed: 12/31/2022] Open
Abstract
Planar cell polarity (PCP) signaling is well known to play a critical role during prenatal brain development; whether it plays specific roles at postnatal stages remains rather unknown. Here, we investigated the role of a key PCP-associated gene scrib in CA1 hippocampal structure and function at postnatal stages. We found that Scrib is required for learning and memory consolidation in the Morris water maze as well as synaptic maturation and NMDAR-dependent bidirectional plasticity. Furthermore, we unveiled a direct molecular interaction between Scrib and PP1/PP2A phosphatases whose levels were decreased in postsynaptic density of conditional knock-out mice. Remarkably, exposure to enriched environment (EE) preserved memory formation in CaMK-Scrib-/- mice by recovering synaptic plasticity and maturation. Thus, Scrib is required for synaptic function involved in memory formation and EE has beneficiary therapeutic effects. Our results demonstrate a distinct new role for a PCP-associated protein, beyond embryonic development, in cognitive functions during adulthood.
Collapse
Affiliation(s)
- Muna L Hilal
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Maité M Moreau
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Claudia Racca
- Institute of Neuroscience, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Vera L Pinheiro
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Nicolas H Piguel
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Marie-Josée Santoni
- CRCM, INSERM U1068, F-13009 Marseille, France.,CRCM, CNRS UMR7258, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13007 Marseille, France
| | - Steve Dos Santos Carvalho
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Jean-Michel Blanc
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,BioXtal Structural Biology Unit, Campus de Luminy, F-13288 Marseille, France.,University of Bordeaux, Plateforme de Biochimie et de Biophysique des protéines, FR Bordeaux Neurocampus, F-33000 Bordeaux, France
| | - Yah-Se K Abada
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Ronan Peyroutou
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Chantal Medina
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Hélène Doat
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Thomas Papouin
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Laurent Vuillard
- BioXtal Structural Biology Unit, Campus de Luminy, F-13288 Marseille, France
| | - Jean-Paul Borg
- CRCM, INSERM U1068, F-13009 Marseille, France.,CRCM, CNRS UMR7258, F-13009 Marseille, France.,Institut Paoli-Calmettes, F-13009 Marseille, France.,Aix-Marseille Université, F-13007 Marseille, France
| | - Rivka Rachel
- Mouse Cancer Genetics Program, National Cancer Institute-Frederick, Frederick, Maryland 21702, USA
| | - Aude Panatier
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Mireille Montcouquiol
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Stéphane H R Oliet
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| | - Nathalie Sans
- INSERM, Neurocentre Magendie, Unité U1215, F-33000 Bordeaux, France.,University of Bordeaux, Neurocentre Magendie, U1215, F-33000 Bordeaux, France
| |
Collapse
|
100
|
Gajardo I, Salazar CS, Lopez-Espíndola D, Estay C, Flores-Muñoz C, Elgueta C, Gonzalez-Jamett AM, Martínez AD, Muñoz P, Ardiles ÁO. Lack of Pannexin 1 Alters Synaptic GluN2 Subunit Composition and Spatial Reversal Learning in Mice. Front Mol Neurosci 2018; 11:114. [PMID: 29692709 PMCID: PMC5902501 DOI: 10.3389/fnmol.2018.00114] [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: 11/15/2017] [Accepted: 03/22/2018] [Indexed: 01/24/2023] Open
Abstract
Long-term potentiation (LTP) and long-term depression (LTD) are two forms of synaptic plasticity that have been considered as the cellular substrate of memory formation. Although LTP has received considerable more attention, recent evidences indicate that LTD plays also important roles in the acquisition and storage of novel information in the brain. Pannexin 1 (Panx1) is a membrane protein that forms non-selective channels which have been shown to modulate the induction of hippocampal synaptic plasticity. Animals lacking Panx1 or blockade of Pannexin 1 channels precludes the induction of LTD and facilitates LTP. To evaluate if the absence of Panx1 also affects the acquisition of rapidly changing information we trained Panx1 knockout (KO) mice and wild type (WT) littermates in a visual and hidden version of the Morris water maze (MWM). We found that KO mice find the hidden platform similarly although slightly quicker than WT animals, nonetheless, when the hidden platform was located in the opposite quadrant (OQ) to the previous learned location, KO mice spent significantly more time in the previous quadrant than in the new location indicating that the absence of Panx1 affects the reversion of a previously acquired spatial memory. Consistently, we observed changes in the content of synaptic proteins critical to LTD, such as GluN2 subunits of N-methyl-D-aspartate receptors (NMDARs), which changed their contribution to synaptic plasticity in conditions of Panx1 ablation. Our findings give further support to the role of Panx1 channels on the modulation of synaptic plasticity induction, learning and memory processes.
Collapse
Affiliation(s)
- Ivana Gajardo
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudia S Salazar
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Daniela Lopez-Espíndola
- Escuela de Tecnología Médica, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Estay
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Elgueta
- Institute for Physiology I, University of Freiburg, Freiburg, Germany
| | - Arlek M Gonzalez-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Programa de Farmacología Molecular y Clínica, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Agustín D Martínez
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Pablo Muñoz
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro de Investigaciones Biomédicas, Escuela de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Center for Applied Neurological Sciences, Faculty of Medicine, Universidad de Valparaíso, Valparaíso, Chile
| | - Álvaro O Ardiles
- Departamento de Patología y Fisiología, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.,Centro Interdisciplinario de Estudios en Salud, Facultad de Medicina, Universidad de Valparaíso, Viña del Mar, Chile
| |
Collapse
|