1
|
Cogut V, Goris M, Jansma A, van der Staaij M, Henning RH. Hippocampal neuroimmune response in mice undergoing serial daily torpor induced by calorie restriction. Front Neuroanat 2024; 18:1334206. [PMID: 38686173 PMCID: PMC11056553 DOI: 10.3389/fnana.2024.1334206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024] Open
Abstract
Hibernating animals demonstrate a remarkable ability to withstand extreme physiological brain changes without triggering adverse neuroinflammatory responses. While hibernators may offer valuable insights into the neuroprotective mechanisms inherent to hibernation, studies using such species are constrained by the limited availability of molecular tools. Laboratory mice may serve as an alternative, entering states of hypometabolism and hypothermia similar to the torpor observed in hibernation when faced with energy shortage. Notably, prolonged calorie restriction (CR) induces serial daily torpor patterns in mice, comparable to species that utilize daily hibernation. Here, we examined the neuroinflammatory response in the hippocampus of male C57BL/6 mice undergoing serial daily torpor induced by a 30% CR for 4 weeks. During daily torpor episodes, CR mice exhibited transient increases in TNF-α mRNA expression, which normalized upon arousal. Concurrently, the CA1 region of the hippocampus showed persistent morphological changes in microglia, characterized by reduced cell branching, decreased cell complexity and altered shape. Importantly, these morphological changes were not accompanied by evident signs of astrogliosis or oxidative stress, typically associated with detrimental neuroinflammation. Collectively, the adaptive nature of the brain's inflammatory response to CR-induced torpor in mice parallels observations in hibernators, highlighting its value for studying the mechanisms of brain resilience during torpor. Such insights could pave the way for novel therapeutic interventions in stroke and neurodegenerative disorders in humans.
Collapse
Affiliation(s)
- Valeria Cogut
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
2
|
de Veij Mestdagh CF, Smit AB, Henning RH, van Kesteren RE. Mitochondrial Targeting against Alzheimer's Disease: Lessons from Hibernation. Cells 2023; 13:12. [PMID: 38201215 PMCID: PMC10778235 DOI: 10.3390/cells13010012] [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: 11/13/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide and yet remains without effective therapy. Amongst the many proposed causes of AD, the mitochondrial cascade hypothesis is gaining attention. Accumulating evidence shows that mitochondrial dysfunction is a driving force behind synaptic dysfunction and cognitive decline in AD patients. However, therapies targeting the mitochondria in AD have proven unsuccessful so far, and out-of-the-box options, such as hibernation-derived mitochondrial mechanisms, may provide valuable new insights. Hibernators uniquely and rapidly alternate between suppression and re-activation of the mitochondria while maintaining a sufficient energy supply and without acquiring ROS damage. Here, we briefly give an overview of mitochondrial dysfunction in AD, how it affects synaptic function, and why mitochondrial targeting in AD has remained unsuccessful so far. We then discuss mitochondria in hibernation and daily torpor in mice, covering current advancements in hibernation-derived mitochondrial targeting strategies. We conclude with new ideas on how hibernation-derived dual mitochondrial targeting of both the ATP and ROS pathways may boost mitochondrial health and induce local synaptic protein translation to increase synaptic function and plasticity. Further exploration of these mechanisms may provide more effective treatment options for AD in the future.
Collapse
Affiliation(s)
- Christina F. de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
- Alzheimer Center Amsterdam, Amsterdam UMC Location VUmc, 1081 HV Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
| | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, 9713 GZ Groningen, The Netherlands;
| | - Ronald E. van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands; (A.B.S.); (R.E.v.K.)
| |
Collapse
|
3
|
Alitalo O, González-Hernández G, Rosenholm M, Kohtala P, Matsui N, Müller HK, Theilmann W, Klein A, Kärkkäinen O, Rozov S, Rantamäki T, Kohtala S. Linking Hypothermia and Altered Metabolism with TrkB Activation. ACS Chem Neurosci 2023; 14:3212-3225. [PMID: 37551888 PMCID: PMC10485900 DOI: 10.1021/acschemneuro.3c00350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/13/2023] [Indexed: 08/09/2023] Open
Abstract
Many mechanisms have been proposed to explain acute antidepressant drug-induced activation of TrkB neurotrophin receptors, but several questions remain. In a series of pharmacological experiments, we observed that TrkB activation induced by antidepressants and several other drugs correlated with sedation, and most importantly, coinciding hypothermia. Untargeted metabolomics of pharmacologically dissimilar TrkB activating treatments revealed effects on shared bioenergetic targets involved in adenosine triphosphate (ATP) breakdown and synthesis, demonstrating a common perturbation in metabolic activity. Both activation of TrkB signaling and hypothermia were recapitulated by administration of inhibitors of glucose and lipid metabolism, supporting a close relationship between metabolic inhibition and neurotrophic signaling. Drug-induced TrkB phosphorylation was independent of electroencephalography slow-wave activity and remained unaltered in knock-in mice with the brain-derived neurotrophic factor (BDNF) Val66Met allele, which have impaired activity-dependent BDNF release, alluding to an activation mechanism independent from BDNF and neuronal activity. Instead, we demonstrated that the active maintenance of body temperature prevents activation of TrkB and other targets associated with antidepressants, including p70S6 kinase downstream of the mammalian target of rapamycin (mTOR) and glycogen synthase kinase 3β (GSK3β). Increased TrkB, GSK3β, and p70S6K phosphorylation was also observed during recovery sleep following sleep deprivation, when a physiological temperature drop is known to occur. Our results suggest that the changes in bioenergetics and thermoregulation are causally connected to TrkB activation and may act as physiological regulators of signaling processes involved in neuronal plasticity.
Collapse
Affiliation(s)
- Okko Alitalo
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Gemma González-Hernández
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Marko Rosenholm
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Center
for Translational Neuromedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Piia Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| | - Nobuaki Matsui
- Faculty
of Pharmacy, Gifu University of Medical
Science, 4-3-3 Nijigaoka,
Kani, Gifu 509-0293, Japan
| | - Heidi Kaastrup Müller
- Translational
Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus N 8200, Denmark
| | - Wiebke Theilmann
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
| | - Anders Klein
- Novo
Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen DK-2200, Denmark
- Department
of Drug Design & Pharmacology, University
of Copenhagen, Copenhagen DK-2100, Denmark
| | - Olli Kärkkäinen
- School
of Pharmacy, University of Eastern Finland, Kuopio 70210, Finland
- Afekta
Technologies Ltd., Kuopio 70210, Finland
| | - Stanislav Rozov
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Tomi Rantamäki
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
| | - Samuel Kohtala
- Laboratory
of Neurotherapeutics, Drug Research Program, Division of Pharmacology
and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki 00014, Finland
- SleepWell
Research Program, Faculty of Medicine, University
of Helsinki, Helsinki 00014, Finland
- Department
of Psychiatry, Weill Cornell Medicine, New York, New York 10021, United States
| |
Collapse
|
4
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Introduction: What Are Dendritic Spines? ADVANCES IN NEUROBIOLOGY 2023; 34:1-68. [PMID: 37962793 DOI: 10.1007/978-3-031-36159-3_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Dendritic spines are cellular specializations that greatly increase the connectivity of neurons and modulate the "weight" of most postsynaptic excitatory potentials. Spines are found in very diverse animal species providing neural networks with a high integrative and computational possibility and plasticity, enabling the perception of sensorial stimuli and the elaboration of a myriad of behavioral displays, including emotional processing, memory, and learning. Humans have trillions of spines in the cerebral cortex, and these spines in a continuum of shapes and sizes can integrate the features that differ our brain from other species. In this chapter, we describe (1) the discovery of these small neuronal protrusions and the search for the biological meaning of dendritic spines; (2) the heterogeneity of shapes and sizes of spines, whose structure and composition are associated with the fine-tuning of synaptic processing in each nervous area, as well as the findings that support the role of dendritic spines in increasing the wiring of neural circuits and their functions; and (3) within the intraspine microenvironment, the integration and activation of signaling biochemical pathways, the compartmentalization of molecules or their spreading outside the spine, and the biophysical properties that can affect parent dendrites. We also provide (4) examples of plasticity involving dendritic spines and neural circuits relevant to species survival and comment on (5) current research advancements and challenges in this exciting research field.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
5
|
Seasonal differences in the morphology and spine density of hippocampal neurons in wild ground squirrels. Brain Struct Funct 2022; 227:2349-2365. [DOI: 10.1007/s00429-022-02528-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/17/2022] [Indexed: 11/02/2022]
|
6
|
Hensleigh E, Murtishaw AS, Treat MD, Heaney CF, Bolton MM, Sabbagh JJ, Calvin KN, Kinney JW, Breukelen FV. Torpor does not influence spatial memory in hibernating golden-mantled ground squirrels (Spermophilus [Callospermophilus] lateralis). Physiol Biochem Zool 2022; 95:390-399. [DOI: 10.1086/721185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
7
|
Trujillo-Estrada L, Vanderklish PW, Nguyen MMT, Kuang RR, Nguyen C, Huynh E, da Cunha C, Javonillo DI, Forner S, Martini AC, Sarraf ST, Simmon VF, Baglietto-Vargas D, LaFerla FM. SPG302 Reverses Synaptic and Cognitive Deficits Without Altering Amyloid or Tau Pathology in a Transgenic Model of Alzheimer's Disease. Neurotherapeutics 2021; 18:2468-2483. [PMID: 34738197 PMCID: PMC8804111 DOI: 10.1007/s13311-021-01143-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2021] [Indexed: 12/04/2022] Open
Abstract
Alzheimer's disease (AD) is conceptualized as a synaptic failure disorder in which loss of glutamatergic synapses is a major driver of cognitive decline. Thus, novel therapeutic strategies aimed at regenerating synapses may represent a promising approach to mitigate cognitive deficits in AD patients. At present, no disease-modifying drugs exist for AD, and approved therapies are palliative at best, lacking in the ability to reverse the synaptic failure. Here, we tested the efficacy of a novel synaptogenic small molecule, SPG302 - a 3rd-generation benzothiazole derivative that increases the density of axospinous glutamatergic synapses - in 3xTg-AD mice. Daily dosing of 3xTg-AD mice with SPG302 at 3 and 30 mg/kg (i.p.) for 4 weeks restored hippocampal synaptic density and improved cognitive function in hippocampal-dependent tasks. Mushroom and stubby spine profiles were increased by SPG302, and associated with enhanced expression of key postsynaptic proteins - including postsynaptic density protein 95 (PSD95), drebrin, and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) - and increased colocalization of PSD95 with synaptophysin. Notably, SPG302 proved efficacious in this model without modifying Aβ and tau pathology. Thus, our study provides preclinical support for the idea that compounds capable of restoring synaptic density offer a viable strategy to reverse cognitive decline in AD.
Collapse
Affiliation(s)
- Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Peter W Vanderklish
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - Marie Minh Thu Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Run Rong Kuang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Caroline Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Eric Huynh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Dominic Ibarra Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA
| | - Stella T Sarraf
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA
| | - Vincent F Simmon
- Spinogenix Inc, 10210 Campus Point Drive, Suite 150, San Diego, CA, 92121, USA.
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
- Departamento Biología Celular, Genetica y Fisiologia, Instituto de Investigacion Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Malaga, Spain.
- Centro de Investigacion Biomedica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, 92697, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, 92697-1450, USA.
| |
Collapse
|
8
|
Myrka A, Buck L. Cytoskeletal Arrest: An Anoxia Tolerance Mechanism. Metabolites 2021; 11:metabo11080561. [PMID: 34436502 PMCID: PMC8401981 DOI: 10.3390/metabo11080561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/14/2021] [Indexed: 12/16/2022] Open
Abstract
Polymerization of actin filaments and microtubules constitutes a ubiquitous demand for cellular adenosine-5′-triphosphate (ATP) and guanosine-5′-triphosphate (GTP). In anoxia-tolerant animals, ATP consumption is minimized during overwintering conditions, but little is known about the role of cell structure in anoxia tolerance. Studies of overwintering mammals have revealed that microtubule stability in neurites is reduced at low temperature, resulting in withdrawal of neurites and reduced abundance of excitatory synapses. Literature for turtles is consistent with a similar downregulation of peripheral cytoskeletal activity in brain and liver during anoxic overwintering. Downregulation of actin dynamics, as well as modification to microtubule organization, may play vital roles in facilitating anoxia tolerance. Mitochondrial calcium release occurs during anoxia in turtle neurons, and subsequent activation of calcium-binding proteins likely regulates cytoskeletal stability. Production of reactive oxygen species (ROS) formation can lead to catastrophic cytoskeletal damage during overwintering and ROS production can be regulated by the dynamics of mitochondrial interconnectivity. Therefore, suppression of ROS formation is likely an important aspect of cytoskeletal arrest. Furthermore, gasotransmitters can regulate ROS levels, as well as cytoskeletal contractility and rearrangement. In this review we will explore the energetic costs of cytoskeletal activity, the cellular mechanisms regulating it, and the potential for cytoskeletal arrest being an important mechanism permitting long-term anoxia survival in anoxia-tolerant species, such as the western painted turtle and goldfish.
Collapse
Affiliation(s)
- Alexander Myrka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
| | - Leslie Buck
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada;
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Correspondence: ; Tel.: +1-416-978-3506
| |
Collapse
|
9
|
Torpor enhances synaptic strength and restores memory performance in a mouse model of Alzheimer's disease. Sci Rep 2021; 11:15486. [PMID: 34326412 PMCID: PMC8322095 DOI: 10.1038/s41598-021-94992-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/13/2021] [Indexed: 12/22/2022] Open
Abstract
Hibernation induces neurodegeneration-like changes in the brain, which are completely reversed upon arousal. Hibernation-induced plasticity may therefore be of great relevance for the treatment of neurodegenerative diseases, but remains largely unexplored. Here we show that a single torpor and arousal sequence in mice does not induce dendrite retraction and synapse loss as observed in seasonal hibernators. Instead, it increases hippocampal long-term potentiation and contextual fear memory. This is accompanied by increased levels of key postsynaptic proteins and mitochondrial complex I and IV proteins, indicating mitochondrial reactivation and enhanced synaptic plasticity upon arousal. Interestingly, a single torpor and arousal sequence was also sufficient to restore contextual fear memory in an APP/PS1 mouse model of Alzheimer’s disease. Our study demonstrates that torpor in mice evokes an exceptional state of hippocampal plasticity and that naturally occurring plasticity mechanisms during torpor provide an opportunity to identify unique druggable targets for the treatment of cognitive impairment.
Collapse
|
10
|
Peretti D, Smith HL, Verity N, Humoud I, de Weerd L, Swinden DP, Hayes J, Mallucci GR. TrkB signaling regulates the cold-shock protein RBM3-mediated neuroprotection. Life Sci Alliance 2021; 4:4/4/e202000884. [PMID: 33563652 PMCID: PMC7893816 DOI: 10.26508/lsa.202000884] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 01/25/2021] [Accepted: 01/25/2021] [Indexed: 02/02/2023] Open
Abstract
Increasing levels of the cold-shock protein, RNA-binding motif 3 (RBM3), either through cooling or by ectopic over-expression, prevents synapse and neuronal loss in mouse models of neurodegeneration. To exploit this process therapeutically requires an understanding of mechanisms controlling cold-induced RBM3 expression. Here, we show that cooling increases RBM3 through activation of TrkB via PLCγ1 and pCREB signaling. RBM3, in turn, has a hitherto unrecognized negative feedback on TrkB-induced ERK activation through induction of its specific phosphatase, DUSP6. Thus, RBM3 mediates structural plasticity through a distinct, non-canonical activation of TrkB signaling, which is abolished in RBM3-null neurons. Both genetic reduction and pharmacological antagonism of TrkB and its downstream mediators abrogate cooling-induced RBM3 induction and prevent structural plasticity, whereas TrkB inhibition similarly prevents RBM3 induction and the neuroprotective effects of cooling in prion-diseased mice. Conversely, TrkB agonism induces RBM3 without cooling, preventing synapse loss and neurodegeneration. TrkB signaling is, therefore, necessary for the induction of RBM3 and related neuroprotective effects and provides a target by which RBM3-mediated synapse-regenerative therapies in neurodegenerative disorders can be used therapeutically without the need for inducing hypothermia.
Collapse
Affiliation(s)
- Diego Peretti
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Heather L Smith
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas Verity
- MRC Toxicology Unit at the University of Cambridge, Leicester, UK
| | - Ibrahim Humoud
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Lis de Weerd
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Dean P Swinden
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Joseph Hayes
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| | - Giovanna R Mallucci
- UK Dementia Research Institute at the University of Cambridge and Department of Clinical Neurosciences, Island Research Building, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
11
|
Duquette A, Pernègre C, Veilleux Carpentier A, Leclerc N. Similarities and Differences in the Pattern of Tau Hyperphosphorylation in Physiological and Pathological Conditions: Impacts on the Elaboration of Therapies to Prevent Tau Pathology. Front Neurol 2021; 11:607680. [PMID: 33488502 PMCID: PMC7817657 DOI: 10.3389/fneur.2020.607680] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/07/2020] [Indexed: 12/20/2022] Open
Abstract
Tau protein, a neuronal microtubule-associated protein, becomes hyperphosphorylated in several neurodegenerative diseases called tauopathies. Hyperphosphorylation of tau is correlated to its redistribution from the axon to the somato-dendritic compartment at early stages of tauopathies. Interestingly, tau hyperphosphorylation begins in different regions of the brain in each tauopathy. In some regions, both neurons and glial cells develop tau hyperphosphorylation. Tau hyperphosphorylation is also observed in physiological conditions such as hibernation and brain development. In the first section of present article, we will review the spatiotemporal and cellular distribution of hyperphosphorylated tau in the most frequent tauopathies. In the second section, we will compare the pattern of tau hyperphosphorylation in physiological and pathological conditions and discuss the sites that could play a pivotal role in the conversion of non-toxic to toxic forms of hyperphosphorylated tau. Furthermore, we will discuss the role of hyperphosphorylated tau in physiological and pathological conditions and the fact that tau hyperphosphorylation is reversible in physiological conditions but not in a pathological ones. In the third section, we will speculate how the differences and similarities between hyperphosphorylated tau in physiological and pathological conditions could impact the elaboration of therapies to prevent tau pathology. In the fourth section, the different therapeutic approaches using tau as a direct or indirect therapeutic target will be presented.
Collapse
Affiliation(s)
- Antoine Duquette
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Camille Pernègre
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Ariane Veilleux Carpentier
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Nicole Leclerc
- Research Center of the University of Montreal Hospital (CRCHUM), Montréal, QC, Canada.,Département de Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
12
|
Shi Z, Qin M, Huang L, Xu T, Chen Y, Hu Q, Peng S, Peng Z, Qu LN, Chen SG, Tuo QH, Liao DF, Wang XP, Wu RR, Yuan TF, Li YH, Liu XM. Human torpor: translating insights from nature into manned deep space expedition. Biol Rev Camb Philos Soc 2020; 96:642-672. [PMID: 33314677 DOI: 10.1111/brv.12671] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
During a long-duration manned spaceflight mission, such as flying to Mars and beyond, all crew members will spend a long period in an independent spacecraft with closed-loop bioregenerative life-support systems. Saving resources and reducing medical risks, particularly in mental heath, are key technology gaps hampering human expedition into deep space. In the 1960s, several scientists proposed that an induced state of suppressed metabolism in humans, which mimics 'hibernation', could be an ideal solution to cope with many issues during spaceflight. In recent years, with the introduction of specific methods, it is becoming more feasible to induce an artificial hibernation-like state (synthetic torpor) in non-hibernating species. Natural torpor is a fascinating, yet enigmatic, physiological process in which metabolic rate (MR), body core temperature (Tb ) and behavioural activity are reduced to save energy during harsh seasonal conditions. It employs a complex central neural network to orchestrate a homeostatic state of hypometabolism, hypothermia and hypoactivity in response to environmental challenges. The anatomical and functional connections within the central nervous system (CNS) lie at the heart of controlling synthetic torpor. Although progress has been made, the precise mechanisms underlying the active regulation of the torpor-arousal transition, and their profound influence on neural function and behaviour, which are critical concerns for safe and reversible human torpor, remain poorly understood. In this review, we place particular emphasis on elaborating the central nervous mechanism orchestrating the torpor-arousal transition in both non-flying hibernating mammals and non-hibernating species, and aim to provide translational insights into long-duration manned spaceflight. In addition, identifying difficulties and challenges ahead will underscore important concerns in engineering synthetic torpor in humans. We believe that synthetic torpor may not be the only option for manned long-duration spaceflight, but it is the most achievable solution in the foreseeable future. Translating the available knowledge from natural torpor research will not only benefit manned spaceflight, but also many clinical settings attempting to manipulate energy metabolism and neurobehavioural functions.
Collapse
Affiliation(s)
- Zhe Shi
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.,Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China
| | - Meng Qin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Lu Huang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, 510632, China
| | - Tao Xu
- Department of Anesthesiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qin Hu
- College of Life Sciences and Bio-Engineering, Beijing University of Technology, Beijing, 100024, China
| | - Sha Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhuang Peng
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Li-Na Qu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Shan-Guang Chen
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Qin-Hui Tuo
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Duan-Fang Liao
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Xiao-Ping Wang
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ren-Rong Wu
- National Clinical Research Center for Mental Disorders, and Department of Psychaitry, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Ti-Fei Yuan
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200030, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226000, China
| | - Ying-Hui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Xin-Min Liu
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.,Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| |
Collapse
|
13
|
Mohr SM, Bagriantsev SN, Gracheva EO. Cellular, Molecular, and Physiological Adaptations of Hibernation: The Solution to Environmental Challenges. Annu Rev Cell Dev Biol 2020; 36:315-338. [PMID: 32897760 DOI: 10.1146/annurev-cellbio-012820-095945] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Thriving in times of resource scarcity requires an incredible flexibility of behavioral, physiological, cellular, and molecular functions that must change within a relatively short time. Hibernation is a collection of physiological strategies that allows animals to inhabit inhospitable environments, where they experience extreme thermal challenges and scarcity of food and water. Many different kinds of animals employ hibernation, and there is a spectrum of hibernation phenotypes. Here, we focus on obligatory mammalian hibernators to identify the unique challenges they face and the adaptations that allow hibernators to overcome them. This includes the cellular and molecular strategies used to combat low environmental and body temperatures and lack of food and water. We discuss metabolic, neuronal, and hormonal cues that regulate hibernation and how they are thought to be coordinated by internal clocks. Last, we touch on questions that are left to be addressed in the field of hibernation research. Studies from the last century and more recent work reveal that hibernation is not simply a passive reduction in body temperature and vital parameters but rather an active process seasonally regulated at the molecular, cellular, and organismal levels.
Collapse
Affiliation(s)
- Sarah M Mohr
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA;
| | - Sviatoslav N Bagriantsev
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA;
| | - Elena O Gracheva
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06510, USA; .,Department of Neuroscience and Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, Connecticut 06510, USA;
| |
Collapse
|
14
|
Regalado-Reyes M, Benavides-Piccione R, Fernaud-Espinosa I, DeFelipe J, León-Espinosa G. Effect of Phosphorylated Tau on Cortical Pyramidal Neuron Morphology during Hibernation. Cereb Cortex Commun 2020; 1:tgaa018. [PMID: 34296096 PMCID: PMC8152943 DOI: 10.1093/texcom/tgaa018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/17/2022] Open
Abstract
The dendritic spines of pyramidal cells are the main postsynaptic target of excitatory glutamatergic synapses. Morphological alterations have been described in hippocampal dendritic spines during hibernation-a state of inactivity and metabolic depression that occurs via a transient neuronal tau hyperphosphorylation. Here, we have used the hibernating Syrian hamster to investigate the effect of hyperphosphorylated tau regarding neocortical neuronal structure. In particular, we examined layer Va pyramidal neurons. Our results indicate that hibernation does not promote significant changes in dendritic spine density. However, tau hyperphosphorylated neurons show a decrease in complexity, an increase in the tortuosity of the apical dendrites, and an increase in the diameter of the basal dendrites. Tau protein hyperphosphorylation and aggregation have been associated with loss or alterations of dendritic spines in neurodegenerative diseases, such as Alzheimer's disease (AD). Our results may shed light on the correlation between tau hyperphosphorylation and the neuropathological processes in AD. Moreover, we observed changes in the length and area of the apical and basal dendritic spines during hibernation regardless of tau hyperphosphorylation. The morphological changes observed here also suggest region specificity, opening up debate about a possible relationship with the differential brain activity registered in these regions in previous studies.
Collapse
Affiliation(s)
- Mamen Regalado-Reyes
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28002, Spain
| | - Ruth Benavides-Piccione
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28002, Spain
| | - Isabel Fernaud-Espinosa
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28002, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28002, Spain
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid 28002, Spain
| |
Collapse
|
15
|
León-Espinosa G, DeFelipe J, Muñoz A. The Golgi Apparatus of Neocortical Glial Cells During Hibernation in the Syrian Hamster. Front Neuroanat 2019; 13:92. [PMID: 31824270 PMCID: PMC6882278 DOI: 10.3389/fnana.2019.00092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 10/28/2019] [Indexed: 12/15/2022] Open
Abstract
Hibernating mammals undergo torpor periods characterized by a general decrease in body temperature, metabolic rate, and brain activity accompanied by complex adaptive brain changes that appear to protect the brain from extreme conditions of hypoxia and low temperatures. These processes are accompanied by morphological and neurochemical changes in the brain including those in cortical neurons such as the fragmentation and reduction of the Golgi apparatus (GA), which both reverse a few hours after arousal from the torpor state. In the present study, we characterized – by immunofluorescence and confocal microscopy – the GA of cortical astrocytes, oligodendrocytes, and microglial cells in the Syrian hamster, which is a facultative hibernator. We also show that after artificial induction of hibernation, in addition to neurons, the GA of glia in the Syrian hamster undergoes important structural changes, as well as modifications in the intensity of immunostaining and distribution patterns of Golgi structural proteins at different stages of the hibernation cycle.
Collapse
Affiliation(s)
- Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Madrid, Spain.,Departamento de Química y Bioquímica, Facultad de Farmacia, CEU San Pablo University, CEU Universities, Madrid, Spain
| | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Madrid, Spain.,Instituto Cajal, CSIC, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain
| | - Alberto Muñoz
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Madrid, Spain.,Instituto Cajal, CSIC, Madrid, Spain.,Departamento de Biología Celular, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
16
|
Bullmann T, Feneberg E, Kretzschmann TP, Ogunlade V, Holzer M, Arendt T. Hibernation Impairs Odor Discrimination - Implications for Alzheimer's Disease. Front Neuroanat 2019; 13:69. [PMID: 31379517 PMCID: PMC6646461 DOI: 10.3389/fnana.2019.00069] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/21/2019] [Indexed: 11/13/2022] Open
Abstract
Reversible formation of PHF-like phosphorylated tau, an early feature of Alzheimer's disease (AD) was previously shown to occur in torpor during hibernation in the Golden hamster (Syrian hamster, Mesocricetus auratus). Here, we tackled the question to what extent hibernating Golden hamsters can serve as a model for the early stage of AD. During early AD, anosmia, the loss of olfactory function, is a common and typical feature. We, thus, investigated tau phosphorylation, synaptic plasticity and behavioral physiology of the olfactory system during hibernation. Tau was phosphorylated on several AD-relevant epitopes, and distribution of PHF-like phosphorylated tau in the olfactory bulb was quite similar to what is seen in AD. Tau phosphorylation was not associated with a destabilization of microtubules and did not lead to fibril formation. Previously, we observed a transient spine reduction in pyramidal cells in the hippocampus, which is correlated with the distribution of phosphorylated tau. Here we show that granule cells in the olfactory bulb are devoid of phosphorylated tau and maintain their spines number during torpor. No reduction of synaptic proteins was observed. However, hibernation did impair the recall performance in a two-odor discrimination task. We conclude that hibernation is associated with a specific olfactory memory deficit, which might not be attributed to the formation of PHF-like phosphorylated tau within the olfactory bulb. We discuss a possible involvement of modulatory input provided by cholinergic neurons in the basal forebrain, which are affected by hibernation.
Collapse
Affiliation(s)
- Torsten Bullmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Emily Feneberg
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Tanja Petra Kretzschmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Vera Ogunlade
- Department of Neuropathology, University of Leipzig, Leipzig, Germany
| | - Max Holzer
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration, Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
17
|
Harding EC, Franks NP, Wisden W. The Temperature Dependence of Sleep. Front Neurosci 2019; 13:336. [PMID: 31105512 PMCID: PMC6491889 DOI: 10.3389/fnins.2019.00336] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 03/22/2019] [Indexed: 12/18/2022] Open
Abstract
Mammals have evolved a range of behavioural and neurological mechanisms that coordinate cycles of thermoregulation and sleep. Whether diurnal or nocturnal, sleep onset and a reduction in core temperature occur together. Non-rapid eye movement (NREM) sleep episodes are also accompanied by core and brain cooling. Thermoregulatory behaviours, like nest building and curling up, accompany this circadian temperature decline in preparation for sleeping. This could be a matter of simply comfort as animals seek warmth to compensate for lower temperatures. However, in both humans and other mammals, direct skin warming can shorten sleep-latency and promote NREM sleep. We discuss the evidence that body cooling and sleep are more fundamentally connected and that thermoregulatory behaviours, prior to sleep, form warm microclimates that accelerate NREM directly through neuronal circuits. Paradoxically, this warmth might also induce vasodilation and body cooling. In this way, warmth seeking and nesting behaviour might enhance the circadian cycle by activating specific circuits that link NREM initiation to body cooling. We suggest that these circuits explain why NREM onset is most likely when core temperature is at its steepest rate of decline and why transitions to NREM are accompanied by a decrease in brain temperature. This connection may have implications for energy homeostasis and the function of sleep.
Collapse
Affiliation(s)
- Edward C Harding
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Nicholas P Franks
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - William Wisden
- Department of Life Sciences, Imperial College London, London, United Kingdom.,Centre for Neurotechnology, Imperial College London, London, United Kingdom.,UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Gonzalez-Riano C, León-Espinosa G, Regalado-Reyes M, García A, DeFelipe J, Barbas C. Metabolomic Study of Hibernating Syrian Hamster Brains: In Search of Neuroprotective Agents. J Proteome Res 2019; 18:1175-1190. [DOI: 10.1021/acs.jproteome.8b00816] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Gonzalo León-Espinosa
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
| | - Mamen Regalado-Reyes
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
| | | | - Javier DeFelipe
- Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid, Campus Montegancedo, 28223 Pozuelo de Alarcón, Madrid, Spain
- Instituto Cajal (CSIC), Avenida Doctor Arce 37, 28002 Madrid, Spain
- CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Calle de Valderrebollo, 5, 28031 Madrid, Spain
| | | |
Collapse
|
19
|
León-Espinosa G, Antón-Fernández A, Tapia-González S, DeFelipe J, Muñoz A. Modifications of the axon initial segment during the hibernation of the Syrian hamster. Brain Struct Funct 2018; 223:4307-4321. [PMID: 30219944 DOI: 10.1007/s00429-018-1753-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/09/2018] [Indexed: 02/07/2023]
Abstract
Mammalian hibernation is a natural process in which the brain undergoes profound adaptive changes that appear to protect the brain from extreme hypoxia and hypothermia. In addition to a virtual cessation of neural and metabolic activity, these changes include a decrease in adult neurogenesis; the retraction of neuronal dendritic trees; changes in dendritic spines and synaptic connections; fragmentation of the Golgi apparatus; and the phosphorylation of the microtubule-associated protein tau. Furthermore, alterations of microglial cells also occur in torpor. Importantly, all of these changes are rapidly and fully reversed when the animals arouse from torpor state, with no apparent brain damage occurring. Thus, hibernating animals are excellent natural models to study different aspects of brain plasticity. The axon initial segment (AIS) is critical for the initiation of action potentials in neurons and is an efficient site for the regulation of neural activity. This specialized structure-characterized by the expression of different types of ion channels and adhesion, scaffolding and cytoskeleton proteins-is subjected to morpho-functional plastic changes upon variations in neural activity or in pathological conditions. Here, we used immunocytochemistry and 3D confocal microscopy reconstruction techniques to measure the possible morphological differences in the AIS of neocortical (layers II-III and V) and hippocampal (CA1) neurons during the hibernation of the Syrian hamster. Our results indicate that the general integrity of the AIS is resistant to the ischemia/hypoxia conditions that are characteristic of the torpor phase of hibernation. In addition, the length of the AIS significantly increased in all the regions studied-by about 16-20% in torpor animals compared to controls, suggesting the existence of compensatory mechanisms in response to a decrease in neuronal activity during the torpor phase of hibernation. Furthermore, in double-labeling experiment, we found that the AIS in layer V of torpid animals was longer in neurons expressing phospho-tau than in those not labeled for phospho-tau. This suggests that AIS plastic changes were more marked in phospho-tau accumulating neurons. Overall, the results further emphasize that mammalian hibernation is a good physiological model to study AIS plasticity mechanisms in non-pathological conditions.
Collapse
Affiliation(s)
- Gonzalo León-Espinosa
- Instituto Cajal, CSIC, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223, Madrid, Spain.,Facultad de Farmacia, Universidad San Pablo CEU, Madrid, Spain
| | - Alejandro Antón-Fernández
- Instituto Cajal, CSIC, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223, Madrid, Spain
| | - Silvia Tapia-González
- Instituto Cajal, CSIC, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223, Madrid, Spain
| | - Javier DeFelipe
- Instituto Cajal, CSIC, Madrid, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223, Madrid, Spain.,CIBERNED, Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Madrid, Spain
| | - Alberto Muñoz
- Instituto Cajal, CSIC, Madrid, Spain. .,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica (CTB), Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223, Madrid, Spain. .,Departamento de Biología Celular, Universidad Complutense, Madrid, Spain.
| |
Collapse
|
20
|
Changes in neocortical and hippocampal microglial cells during hibernation. Brain Struct Funct 2017; 223:1881-1895. [PMID: 29260372 DOI: 10.1007/s00429-017-1596-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/15/2017] [Indexed: 12/29/2022]
Abstract
Mammalian hibernation proceeds alongside a wide range of complex brain adaptive changes that appear to protect the brain from extreme hypoxia and hypothermia. Using immunofluorescence, confocal microscopy, quantitative analysis methods and intracellular injections, we have characterized microglia morphological changes that occur in the neocortex and hippocampus of the Syrian hamster during hibernation. In euthermic hamsters, microglial cells showed the typical ramified/resting morphology with multiple long, thin and highly-branched processes homogeneously immunostained for Iba-1. However, during torpor, microglial cell process numbers increase significantly accompanied by a shortening of the Iba-1 immunoreactive processes, which show a fragmented appearance. Adaptative changes of microglial cells during torpor coursed with no expression of microglial cell activation markers. We discuss the possibility that these morphological changes may contribute to neuronal damage prevention during hibernation.
Collapse
|
21
|
Roda E, Bottone MG, Insolia V, Barni S, Bernocchi G. Changes in the cerebellar cytoarchitecture of hibernating hedgehog Erinaceus europaeus L. (Mammalia): an immunocytochemical approach. EUROPEAN ZOOLOGICAL JOURNAL 2017. [DOI: 10.1080/24750263.2017.1380722] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- E. Roda
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
- Laboratory of Clinical & Experimental Toxicology and Poison Control Centre and National Toxicology Information Centre, Toxicology Unit, ICS Maugeri Spa Benefit Corporation, IRCCS of Pavia, Pavia, Italy
| | - M. G. Bottone
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - V. Insolia
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - S. Barni
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| | - G. Bernocchi
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratory of Cell Biology and Neurobiology, University of Pavia, Pavia, Italy
| |
Collapse
|
22
|
Onufriev MV, Semenova TP, Sergun’kina MA, Volkova EP, Yakovlev AA, Zakharova NM, Gulyaeva NV. Changes in cyclin and cyclin-dependent protein kinase expression in the long-tailed ground squirrel (Spermophilus undulatus) brain during hibernation and awakening. Biophysics (Nagoya-shi) 2016. [DOI: 10.1134/s0006350916050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
23
|
León-Espinosa G, García E, Gómez-Pinedo U, Hernández F, DeFelipe J, Ávila J. Decreased adult neurogenesis in hibernating Syrian hamster. Neuroscience 2016; 333:181-92. [DOI: 10.1016/j.neuroscience.2016.07.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/08/2016] [Accepted: 07/11/2016] [Indexed: 12/29/2022]
|
24
|
Onufriev MV, Semenova TP, Volkova EP, Sergun’kina MA, Yakovlev AA, Zakharova NM, Gulyaeva NV. Seasonal changes in actin and Cdk5 expression in different brain regions of the Yakut ground squirrel (Spermophilus undulatus). NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416020070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Onufriev MV, Semenova TP, Volkova EP, Sergun’kina MA, Yakovlev AA, Zakharova NM, Gulyaeva NV. The characteristics of the expression of the Cdk1 and Cyclin B1 Proteins in the brain of the Yakut ground squirrel (Spermophilus undulatus) at different stages of the hibernation cycle. NEUROCHEM J+ 2016. [DOI: 10.1134/s1819712416020082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Seasonal and post-trauma remodeling in cone-dominant ground squirrel retina. Exp Eye Res 2016; 150:90-105. [PMID: 26808487 DOI: 10.1016/j.exer.2016.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 12/14/2015] [Accepted: 01/14/2016] [Indexed: 02/02/2023]
Abstract
With a photoreceptor mosaic containing ∼85% cones, the ground squirrel is one of the richest known mammalian sources of these important retinal cells. It also has a visual ecology much like the human's. While the ground squirrel retina is understandably prominent in the cone biochemistry, physiology, and circuitry literature, far less is known about the remodeling potential of its retinal pigment epithelium, neurons, macroglia, or microglia. This review aims to summarize the data from ground squirrel retina to this point in time, and to relate them to data from other brain areas where appropriate. We begin with a survey of the ground squirrel visual system, making comparisons with traditional rodent models and with human. Because this animal's status as a hibernator often goes unnoticed in the vision literature, we then present a brief primer on hibernation biology. Next we review what is known about ground squirrel retinal remodeling concurrent with deep torpor and with rapid recovery upon re-warming. Notable here is rapidly-reversible, temperature-dependent structural plasticity of cone ribbon synapses, as well as pre- and post-synaptic plasticity throughout diverse brain regions. It is not yet clear if retinal cell types other than cones engage in torpor-associated synaptic remodeling. We end with the small but intriguing literature on the ground squirrel retina's remodeling responses to insult by retinal detachment. Notable for widespread loss of (cone) photoreceptors, there is surprisingly little remodeling of the RPE or Müller cells. Microglial activation appears minimal, and remodeling of surviving second- and third-order neurons seems absent, but both require further study. In contrast, traumatic brain injury in the ground squirrel elicits typical macroglial and microglial responses. Overall, the data to date strongly suggest a heretofore unrecognized, natural checkpoint between retinal deafferentiation and RPE and Müller cell remodeling events. As we continue to discover them, the unique ways by which ground squirrel retina responds to hibernation or injury may be adaptable to therapeutic use.
Collapse
|
27
|
Bullmann T, Seeger G, Stieler J, Hanics J, Reimann K, Kretzschmann TP, Hilbrich I, Holzer M, Alpár A, Arendt T. Tau phosphorylation-associated spine regression does not impair hippocampal-dependent memory in hibernating golden hamsters. Hippocampus 2015; 26:301-18. [DOI: 10.1002/hipo.22522] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/12/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023]
Affiliation(s)
- Torsten Bullmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
- Frey Initiative Research Unit, RIKEN Quantitative Biology Center; Japan
| | - Gudrun Seeger
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Jens Stieler
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - János Hanics
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Katja Reimann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Tanja Petra Kretzschmann
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Isabel Hilbrich
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Max Holzer
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| | - Alán Alpár
- MTA-SE NAP B Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences; Budapest Hungary
- Department of Anatomy; Semmelweis University; Budapest Hungary
| | - Thomas Arendt
- Department of Molecular and Cellular Mechanisms of Neurodegeneration; Paul Flechsig Institute of Brain Research, University of Leipzig; Leipzig Germany
| |
Collapse
|
28
|
Blackiston DJ, Shomrat T, Levin M. The stability of memories during brain remodeling: A perspective. Commun Integr Biol 2015; 8:e1073424. [PMID: 27066165 PMCID: PMC4802789 DOI: 10.1080/19420889.2015.1073424] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 07/13/2015] [Indexed: 01/10/2023] Open
Abstract
One of the most important features of the nervous system is memory: the ability to represent and store experiences, in a manner that alters behavior and cognition at future times when the original stimulus is no longer present. However, the brain is not always an anatomically stable structure: many animal species regenerate all or part of the brain after severe injury, or remodel their CNS toward a new configuration as part of their life cycle. This raises a fascinating question: what are the dynamics of memories during brain regeneration? Can stable memories remain intact when cellular turnover and spatial rearrangement modify the biological hardware within which experiences are stored? What can we learn from model species that exhibit both, regeneration and memory, with respect to robustness and stability requirements for long-term memories encoded in living tissues? In this Perspective, we discuss relevant data in regenerating planaria, metamorphosing insects, and hibernating ground squirrels. While much remains to be done to understand this remarkable process, molecular-level insight will have important implications for cognitive science, regenerative medicine of the brain, and the development of non-traditional computational media in synthetic bioengineering.
Collapse
Affiliation(s)
- Douglas J Blackiston
- Center for Regenerative and Developmental Biology and Department of Biology; Tufts University ; Medford, MA USA
| | - Tal Shomrat
- Department of Neurobiology; Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus; Jerusalem, Israel; School of Marine Sciences, Ruppin Academic Center; Michmoret, Israel
| | - Michael Levin
- Center for Regenerative and Developmental Biology and Department of Biology; Tufts University ; Medford, MA USA
| |
Collapse
|
29
|
RBM3 mediates structural plasticity and protective effects of cooling in neurodegeneration. Nature 2015; 518:236-9. [PMID: 25607368 PMCID: PMC4338605 DOI: 10.1038/nature14142] [Citation(s) in RCA: 160] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 12/05/2014] [Indexed: 12/18/2022]
Abstract
In the healthy adult brain synapses are continuously remodelled through a process of elimination and formation known as structural plasticity1. Reduction in synapse number is a consistent early feature of neurodegenerative diseases2, 3, suggesting deficient compensatory mechanisms. While much is known about toxic processes leading to synaptic dysfunction and loss in these disorders2,3, how synaptic regeneration is affected is unknown. In hibernating mammals, cooling induces loss of synaptic contacts, which are reformed on rewarming, a form of structural plasticity4, 5. We have found that similar changes occur in artificially cooled laboratory rodents. Cooling and hibernation also induce a number cold-shock proteins in the brain, including the RNA binding protein, RBM36. The relationship of such proteins to structural plasticity is unknown. Here we show that synapse regeneration is impaired in mouse models of neurodegenerative disease, in association with the failure to induce RBM3. In both prion-infected and 5×FAD (Alzheimer-type) mice7, the capacity to regenerate synapses after cooling declined in parallel with the loss of induction of RBM3. Enhanced expression of RBM3 in the hippocampus prevented this deficit and restored the capacity for synapse reassembly after cooling. Further, RBM3 over-expression, achieved either by boosting endogenous levels through hypothermia prior to the loss of the RBM3 response, or by lentiviral delivery, resulted in sustained synaptic protection in 5×FAD mice and throughout the course of prion disease, preventing behavioural deficits and neuronal loss and significantly prolonging survival. In contrast, knockdown of RBM3 exacerbated synapse loss in both models and accelerated disease and prevented the neuroprotective effects of cooling. Thus, deficient synapse regeneration, mediated at least in part by failure of the RBM3 stress response, contributes to synapse loss throughout the course of neurodegenerative disease. The data support enhancing cold shock pathways as potential protective therapies in neurodegenerative disorders.
Collapse
|
30
|
Malanowski S, Craver CF. The spine problem: finding a function for dendritic spines. Front Neuroanat 2014; 8:95. [PMID: 25309340 PMCID: PMC4159972 DOI: 10.3389/fnana.2014.00095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 08/24/2014] [Indexed: 11/13/2022] Open
Abstract
Why do neurons have dendritic spines? This question-the heart of what Yuste calls "the spine problem"-presupposes that why-questions of this sort have scientific answers: that empirical findings can favor or count against claims about why neurons have spines. Here we show how such questions can receive empirical answers. We construe such why-questions as questions about how spines make a difference to the behavior of some mechanism that we take to be significant. Why-questions are driven fundamentally by the effort to understand how some item, such as the dendritic spine, is situated in the causal structure of the world (the causal nexus). They ask for a filter on that busy world that allows us to see a part's individual contribution to a mechanism, independent of everything else going on. So understood, answers to why-questions can be assessed by testing the claims these answers make about the causal structure of a mechanism. We distinguish four ways of making a difference to a mechanism (necessary, modulatory, component, background condition), and we sketch their evidential requirements. One consequence of our analysis is that there are many spine problems and that any given spine problem might have many acceptable answers.
Collapse
Affiliation(s)
- Sarah Malanowski
- Department of Philosophy, Washington University in St. Louis St. Louis, MO, USA
| | - Carl F Craver
- Department of Philosophy, Washington University in St. Louis St. Louis, MO, USA
| |
Collapse
|
31
|
van der Zee EA. Synapses, spines and kinases in mammalian learning and memory, and the impact of aging. Neurosci Biobehav Rev 2014; 50:77-85. [PMID: 24998408 DOI: 10.1016/j.neubiorev.2014.06.012] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/21/2014] [Accepted: 06/24/2014] [Indexed: 02/04/2023]
Abstract
Synapses are the building blocks of neuronal networks. Spines, the postsynaptic elements, are morphologically the most plastic part of the synapse. It is thought that spine plasticity underlies learning and memory processes, driven by kinases and cytoskeleton protein reorganization. Spine strength depends primarily on the number of incorporated glutamatergic receptors, which are more numerous in larger spines. Intrinsic and circadian fluctuations, occurring independently of presynaptic stimulation, demonstrate the native instability of spines. Despite innate spine instability some spines remain intact lifelong. Threats to spine survival are reduced by physical and mental activity, and declining sensory input, conditions characteristic for aging. Large spines are considered less vulnerable than thin spines, and in the older brain large spines are more abundant, whereas the thin spines are functionally weaker. It can be speculated that this shift towards memory spines contributes to enhanced retention of remote memories typically seen in the elderly. Gaining further insight in spine plasticity regulation, its homeostatic nature and how to maintain spine health will be important future research topics in Neuroscience.
Collapse
Affiliation(s)
- Eddy A van der Zee
- Department of Molecular Neurobiology, Centre for Behaviour and Neurosciences, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| |
Collapse
|
32
|
Sala C, Segal M. Dendritic spines: the locus of structural and functional plasticity. Physiol Rev 2014; 94:141-88. [PMID: 24382885 DOI: 10.1152/physrev.00012.2013] [Citation(s) in RCA: 338] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The introduction of high-resolution time lapse imaging and molecular biological tools has changed dramatically the rate of progress towards the understanding of the complex structure-function relations in synapses of central spiny neurons. Standing issues, including the sequence of molecular and structural processes leading to formation, morphological change, and longevity of dendritic spines, as well as the functions of dendritic spines in neurological/psychiatric diseases are being addressed in a growing number of recent studies. There are still unsettled issues with respect to spine formation and plasticity: Are spines formed first, followed by synapse formation, or are synapses formed first, followed by emergence of a spine? What are the immediate and long-lasting changes in spine properties following exposure to plasticity-producing stimulation? Is spine volume/shape indicative of its function? These and other issues are addressed in this review, which highlights the complexity of molecular pathways involved in regulation of spine structure and function, and which contributes to the understanding of central synaptic interactions in health and disease.
Collapse
|
33
|
Zhang Y, Meredith GE, Mendoza-Elias N, Rademacher DJ, Tseng KY, Steece-Collier K. Aberrant restoration of spines and their synapses in L-DOPA-induced dyskinesia: involvement of corticostriatal but not thalamostriatal synapses. J Neurosci 2013; 33:11655-67. [PMID: 23843533 PMCID: PMC3724545 DOI: 10.1523/jneurosci.0288-13.2013] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 05/07/2013] [Accepted: 06/05/2013] [Indexed: 11/21/2022] Open
Abstract
We examined the structural plasticity of excitatory synapses from corticostriatal and thalamostriatal pathways and their postsynaptic targets in adult Sprague-Dawley rats to understand how these striatal circuits change in l-DOPA-induced dyskinesias (LIDs). We present here detailed electron and light microscopic analyses that provide new insight into the nature of the structural and synaptic remodeling of medium spiny neurons in response to LIDs. Numerous studies have implicated enhanced glutamate signaling and persistent long-term potentiation as central to the behavioral sensitization phenomenon of LIDs. Moreover, experience-dependent alterations in behavior are thought to involve structural modifications, specifically alterations in patterns of synaptic connectivity. Thus, we hypothesized that in the striatum of rats with LIDs, one of two major glutamatergic pathways would form new or altered contacts, especially onto the spines of medium spiny neuron (MSNs). Our data provide compelling evidence for a dramatic rewiring of the striatum of dyskinetic rats and that this rewiring involves corticostriatal but not thalamostriatal contacts onto MSNs. There is a dramatic increase in corticostriatal contacts onto spines and dendrites that appear to be directly linked to dyskinetic behaviors, since they were not seen in the striatum of animals that did not develop dyskinesia. There is also an aberrant increase in spines receiving more than one excitatory contact(i.e., multisynaptic spines) in the dyskinetic animals compared with the 6-hydroxydopamine-treated and control rats. Such alterations could substantially impair the ability of striatal neurons to gate cortically driven signals and contribute to the loss of bidirectional synaptic plasticity.
Collapse
Affiliation(s)
- Yiyue Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, and
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, and
| | | | - Nasya Mendoza-Elias
- Department of Pharmaceutical Sciences, College of Pharmacy, and
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, and
| | - David J. Rademacher
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan 49503
| | - Kuei Y. Tseng
- Department of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, and
| | - Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, Michigan 49503
| |
Collapse
|
34
|
Arendt T, Bullmann T. Neuronal plasticity in hibernation and the proposed role of the microtubule-associated protein tau as a "master switch" regulating synaptic gain in neuronal networks. Am J Physiol Regul Integr Comp Physiol 2013; 305:R478-89. [PMID: 23824962 DOI: 10.1152/ajpregu.00117.2013] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The present paper provides an overview of adaptive changes in brain structure and learning abilities during hibernation as a behavioral strategy used by several mammalian species to minimize energy expenditure under current or anticipated inhospitable environmental conditions. One cellular mechanism that contributes to the regulated suppression of metabolism and thermogenesis during hibernation is reversible phosphorylation of enzymes and proteins, which limits rates of flux through metabolic pathways. Reversible phosphorylation during hibernation also affects synaptic membrane proteins, a process known to be involved in synaptic plasticity. This mechanism of reversible protein phosphorylation also affects the microtubule-associated protein tau, thereby generating a condition that in the adult human brain is associated with aggregation of tau protein to paired helical filaments (PHFs), as observed in Alzheimer's disease. Here, we put forward the concept that phosphorylation of tau is a neuroprotective mechanism to escape NMDA-mediated hyperexcitability of neurons that would otherwise occur during slow gradual cooling of the brain. Phosphorylation of tau and its subsequent targeting to subsynaptic sites might, thus, work as a kind of "master switch," regulating NMDA receptor-mediated synaptic gain in a wide array of neuronal networks, thereby enabling entry into torpor. If this condition lasts too long, however, it may eventually turn into a pathological trigger, driving a cascade of events leading to neurodegeneration, as in Alzheimer's disease or other "tauopathies".
Collapse
Affiliation(s)
- Thomas Arendt
- Paul Flechsig Institute of Brain Research, Universität Leipzig, Germany.
| | | |
Collapse
|
35
|
Moshkov DA, Shtanchaev RS, Mikheeva IB, Bezgina EN, Kokanova NA, Mikhailova GZ, Tiras NR, Pavlik LL. Visual input controls the functional activity of goldfish Mauthner neuron through the reciprocal synaptic mechanism. J Integr Neurosci 2013; 12:17-34. [DOI: 10.1142/s0219635213500039] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
36
|
Han HS, Park J, Kim JH, Suk K. Molecular and cellular pathways as a target of therapeutic hypothermia: pharmacological aspect. Curr Neuropharmacol 2012; 10:80-7. [PMID: 22942881 PMCID: PMC3286850 DOI: 10.2174/157015912799362751] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 08/08/2011] [Accepted: 09/08/2011] [Indexed: 11/22/2022] Open
Abstract
Induced therapeutic hypothermia is the one of the most effective tools against brain injury and inflammation. Even though its beneficial effects are well known, there are a lot of pitfalls to overcome, since the potential adverse effects of systemic hypothermia are still troublesome. Without the knowledge of the precise mechanisms of hypothermia, it will be difficult to tackle the application of hypothermia in clinical fields. Better understanding of the characteristics and modes of hypothermic actions may further extend the usage of hypothermia by developing novel drugs based on the hypothermic mechanisms or by combining hypothermia with other therapeutic modalities such as neuroprotective drugs. In this review, we describe the potential therapeutic targets for the development of new drugs, with a focus on signal pathways, gene expression, and structural changes of cells. Theapeutic hypothermia has been shown to attenuate neuroinflammation by reducing the production of reactive oxygen species and proinflammatory mediators in the central nervous system. Along with the mechanism-based drug targets, applications of therapeutic hypothermia in combination with drug treatment will also be discussed in this review.
Collapse
Affiliation(s)
- Hyung Soo Han
- Department of Physiology, Brain Science & Engineering Institute, Kyungpook National University School of Medicine, Daegu, 700-422, Korea
| | | | | | | |
Collapse
|
37
|
Seasonal and sex differences in the hippocampus of a wild rodent. Behav Brain Res 2012; 236:131-138. [PMID: 22974551 DOI: 10.1016/j.bbr.2012.08.044] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/29/2012] [Accepted: 08/28/2012] [Indexed: 01/14/2023]
Abstract
Studies across and within species suggest that hippocampus size is sexually dimorphic in polygamous species, but not in monogamous species. Although hippocampal volume varies with sex, season and mating system, few studies have simultaneously tested for sex and seasonal differences. Here, we test for sex and seasonal differences in the hippocampal volume of wild Richardson's ground squirrels (Urocitellus richardsonii), a polygamous species that lives in matrilineal, kin-based social groups and has profound sex differences in behavior. Based on the behavior and ecology of this species, we predicted that males would have a significantly larger hippocampus than females and that the hippocampus would be largest in males during the breeding season. Analyses of both absolute and relative volumes of the hippocampus yielded a significant difference between the sexes and seasons as well as an interaction between the two such that non-breeding males have significantly larger hippocampal volumes than breeding males or females from either season. Dentate gyrus, CA1 and CA3 subfield volumes were generally larger in the non-breeding season and in males, but no significant interaction effects were detected. This sex and seasonal variation in hippocampal volume is likely the result of their social organization and male-only food caching behavior during the non-breeding season. The demonstration of a sex and seasonal variation in hippocampal volume suggests that Richardson's ground squirrel may be a useful model for understanding hippocampal plasticity within a natural context.
Collapse
|
38
|
Velázquez-Zamora DA, González-Tapia D, González-Ramírez MM, Flores-Soto ME, Vázquez-Valls E, Cervantes M, González-Burgos I. Plastic changes in dendritic spines of hippocampal CA1 pyramidal neurons from ovariectomized rats after estradiol treatment. Brain Res 2012; 1470:1-10. [PMID: 22750586 DOI: 10.1016/j.brainres.2012.06.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 05/02/2012] [Accepted: 06/06/2012] [Indexed: 02/05/2023]
Abstract
Cognitive impairment or its recovery has been associated with the absence or reestablishment of estrogenic actions in the central nervous system of female experimental animals or women. It has been proposed that these cognitive phenomena are related to estrogen-mediated modulatory activity of synaptic transmission in brain structures involved in cognitive functions. In the present work a morphological study was conducted in adult female ovariectomized rats to evaluate estradiol-dependent dendritic spine sprouting in hippocampal pyramidal neurons, and changes in the presynaptic marker synaptophysin. Three or ten days after estradiol treatment (10 μg/day, twice) in the ovariectomized rats, a significant increase of synaptophysin was observed, which was coincident with a significant higher numerical density of thin (22%), stubby (36%), mushroom (47%) and double spines (125%), at day 3, without significant changes of spine density at day 10, after treatment. These results may be interpreted as evidence of pre- and postsynaptic plastic events that may be involved in the modulation of cognitive-related behavioral performance after estrogen replacement therapy.
Collapse
|
39
|
Harris KM, Weinberg RJ. Ultrastructure of synapses in the mammalian brain. Cold Spring Harb Perspect Biol 2012; 4:cshperspect.a005587. [PMID: 22357909 DOI: 10.1101/cshperspect.a005587] [Citation(s) in RCA: 279] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The morphology and molecular composition of synapses provide the structural basis for synaptic function. This article reviews the electron microscopy of excitatory synapses on dendritic spines, using data from rodent hippocampus, cerebral cortex, and cerebellar cortex. Excitatory synapses have a prominent postsynaptic density, in contrast with inhibitory synapses, which have less dense presynaptic or postsynaptic specializations and are usually found on the cell body or proximal dendritic shaft. Immunogold labeling shows that the presynaptic active zone provides a scaffold for key molecules involved in the release of neurotransmitter, whereas the postsynaptic density contains ligand-gated ionic channels, other receptors, and a complex network of signaling molecules. Delineating the structure and molecular organization of these axospinous synapses represents a crucial step toward understanding the mechanisms that underlie synaptic transmission and the dynamic modulation of neurotransmission associated with short- and long-term synaptic plasticity.
Collapse
Affiliation(s)
- Kristen M Harris
- Center for Learning and Memory, Neurobiology Section, University of Texas, Austin, 78712, USA.
| | | |
Collapse
|
40
|
Workman JL, Manny N, Walton JC, Nelson RJ. Short day lengths alter stress and depressive-like responses, and hippocampal morphology in Siberian hamsters. Horm Behav 2011; 60:520-8. [PMID: 21851822 DOI: 10.1016/j.yhbeh.2011.07.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 07/29/2011] [Accepted: 07/31/2011] [Indexed: 01/27/2023]
Abstract
Many psychological disorders comprise a seasonal component. For instance, seasonal affective disorder (SAD) is characterized by depression during autumn and winter. Because hippocampal atrophy may underlie the symptoms of depression and depressive-like behaviors, one goal of this study was to determine whether short days also induce structural changes in the hippocampus using photoperiod responsive rodents--Siberian hamsters. Exposure to short days increases depressive-like responses (increased immobility in the forced swim test) in hamsters. Male hamsters were housed in either short (LD 8:16) or long days (LD 16:8) for 10 weeks and tested in the forced swim test. Brains were removed and processed for Golgi impregnation. HPA axis function may account for photoperiod-related changes in depressive-like responses. Thus, stress reactivity was assessed in another cohort of photoperiod-manipulated animals. Short days reduced soma size and dendritic complexity in the CA1 region. Photoperiod did not induce gross changes in stress reactivity, but an acute stressor disrupted the typical nocturnal peak in cortisol concentrations. These data reveal that immobility induced by exposure to short days is correlated with reduced CA1 cell complexity (and perhaps connectivity). This study is the first to investigate hippocampal changes in the context of short-day induced immobility and may be relevant for understanding psychological disorders with a seasonal component.
Collapse
Affiliation(s)
- Joanna L Workman
- Department of Psychology, The Ohio State University, Columbus, OH 43201, USA.
| | | | | | | |
Collapse
|
41
|
Recovery from chronic monocular deprivation following reactivation of thalamocortical plasticity by dark exposure. Nat Commun 2011; 2:317. [PMID: 21587234 DOI: 10.1038/ncomms1312] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 04/13/2011] [Indexed: 11/08/2022] Open
Abstract
Chronic monocular deprivation induces severe amblyopia that is resistant to spontaneous reversal. However, dark exposure initiated in adulthood reactivates synaptic plasticity in the visual cortex and promotes recovery from chronic monocular deprivation in Long Evans rats. Here we show that chronic monocular deprivation induces a significant decrease in the density of dendritic spines on principal neurons throughout the deprived visual cortex. Nevertheless, dark exposure followed by reverse deprivation promotes the recovery of dendritic spine density of neurons in all laminae. Importantly, the ocular dominance of neurons in thalamo-recipient laminae of the cortex, and the amplitude of the thalamocortical visually evoked potential recover following dark exposure and reverse deprivation. Thus, dark exposure reactivates widespread synaptic plasticity in the adult visual cortex, including thalamocortical synapses, during the recovery from chronic monocular deprivation.
Collapse
|
42
|
Suspension of mitotic activity in dentate gyrus of the hibernating ground squirrel. Neural Plast 2011; 2011:867525. [PMID: 21773054 PMCID: PMC3136081 DOI: 10.1155/2011/867525] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/05/2011] [Accepted: 03/10/2011] [Indexed: 11/25/2022] Open
Abstract
Neurogenesis occurs in the adult mammalian hippocampus, a region of the brain important for learning and memory. Hibernation in Siberian ground squirrels provides a natural model to study mitosis as the rapid fall in body temperature in 24 h (from 35-36°C to +4–6°C) permits accumulation of mitotic cells at different stages of the cell cycle. Histological methods used to study adult neurogenesis are limited largely to fixed tissue, and the mitotic state elucidated depends on the specific phase of mitosis at the time of day. However, using an immunohistochemical study of doublecortin (DCX) and BrdU-labelled neurons, we demonstrate that the dentate gyrus of the ground squirrel hippocampus contains a population of immature cells which appear to possess mitotic activity. Our data suggest that doublecortin-labelled immature cells exist in a mitotic state and may represent a renewable pool for generation of new neurons within the dentate gyrus.
Collapse
|
43
|
Hengen KB, Gomez TM, Stang KM, Johnson SM, Behan M. Changes in ventral respiratory column GABAaR ε- and δ-subunits during hibernation mediate resistance to depression by EtOH and pentobarbital. Am J Physiol Regul Integr Comp Physiol 2010; 300:R272-83. [PMID: 21084677 DOI: 10.1152/ajpregu.00607.2010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During hibernation in the 13-lined ground squirrel, Ictidomys tridecemlineatus, the cerebral cortex is electrically silent, yet the brainstem continues to regulate cardiorespiratory function. Previous work showed that neurons in slices through the medullary ventral respiratory column (VRC) but not the cortex are insensitive to high doses of pentobarbital during hibernation, leading to the hypothesis that GABA(A) receptors (GABA(A)R) in the VRC undergo a seasonal modification in subunit composition. To test whether alteration of GABA(A)R subunits are responsible for hibernation-associated pentobarbital insensitivity, we examined an array of subunits using RT-PCR and Western blots and identified changes in ε- and δ-subunits in the medulla but not the cortex. Using immunohistochemistry, we confirmed that during hibernation, the expression of ε-subunit-containing GABA(A)Rs nearly doubles in the VRC. We also identified a population of δ-subunit-containing GABA(A)Rs adjacent to the VRC that were differentially expressed during hibernation. As δ-subunit-containing GABA(A)Rs are particularly sensitive to ethanol (EtOH), multichannel electrodes were inserted in slices of medulla and cortex from hibernating squirrels and EtOH was applied. EtOH, which normally inhibits neuronal activity, excited VRC but not cortical neurons during hibernation. This excitation was prevented by bicuculline pretreatment, indicating the involvement of GABA(A)Rs. We propose that neuronal activity in the VRC during hibernation is unaffected by pentobarbital due to upregulation of ε-subunit-containing GABA(A)Rs on VRC neurons. Synaptic input from adjacent inhibitory interneurons that express δ-subunit-containing GABA(A)Rs is responsible for the excitatory effects of EtOH on VRC neurons during hibernation.
Collapse
Affiliation(s)
- K B Hengen
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin, USA
| | | | | | | | | |
Collapse
|
44
|
Workman JL, Nelson RJ. Potential animal models of seasonal affective disorder. Neurosci Biobehav Rev 2010; 35:669-79. [PMID: 20800614 DOI: 10.1016/j.neubiorev.2010.08.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 08/13/2010] [Accepted: 08/19/2010] [Indexed: 01/05/2023]
Abstract
Seasonal affective disorder (SAD) is characterized by depressive episodes during winter that are alleviated during summer and by morning bright light treatment. Currently, there is no animal model of SAD. However, it may be possible to use rodents that respond to day length (photoperiod) to understand how photoperiod can shape the brain and behavior in humans. As nights lengthen in the autumn, the duration of the nightly elevation of melatonin increase; seasonally breeding animals use this information to orchestrate seasonal changes in physiology and behavior. SAD may originate from the extended duration of nightly melatonin secretion during fall and winter. These similarities between humans and rodents in melatonin secretion allows for comparisons with rodents that express more depressive-like responses when exposed to short day lengths. For instance, Siberian hamsters, fat sand rats, Nile grass rats, and Wistar rats display a depressive-like phenotype when exposed to short days. Current research in depression and animal models of depression suggests that hippocampal plasticity may underlie the symptoms of depression and depressive-like behaviors, respectively. It is also possible that day length induces structural changes in human brains. Many seasonally breeding rodents undergo changes in whole brain and hippocampal volume in short days. Based on strict validity criteria, there is no animal model of SAD, but rodents that respond to reduced day lengths may be useful to approximate the neurobiological phenomena that occur in people with SAD, leading to greater understanding of the etiology of the disorder as well as novel therapeutic interventions.
Collapse
Affiliation(s)
- Joanna L Workman
- Department of Psychology, The Ohio State University, Columbus, OH 43201, USA.
| | | |
Collapse
|
45
|
Segal M. Dendritic spines, synaptic plasticity and neuronal survival: activity shapes dendritic spines to enhance neuronal viability. Eur J Neurosci 2010; 31:2178-84. [PMID: 20550565 DOI: 10.1111/j.1460-9568.2010.07270.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
An emerging view of structure-function relations of synapses in central spiny neurons asserts that larger spines produce large synaptic currents and that these large spines are persistent ('memory') compared to small spines which are transient. Furthermore, 'learning' involves enlargement of small spine heads and their conversion to being large and stable. It is also assumed that the number of spines, hence the number of synapses, is reflected in the frequency of miniature excitatory postsynaptic currents (mEPSCs). Consequently, there is an assumption that the size and number of mEPSCs are closely correlated with, respectively, the physical size of synapses and number of spines. However, several recent observations do not conform to these generalizations, necessitating a reassessment of the model: spine dimension and synaptic responses are not always correlated. It is proposed that spines are formed and shaped by ongoing network activity, not necessarily by a 'learning' event, to the extent that, in the absence of such activity, new spines are not formed and existing ones disappear or convert into thin filopodia. In the absence of spines, neurons can still maintain synapses with afferent fibers, which can now terminate on its dendritic shaft. Shaft synapses are likely to produce larger synaptic currents than spine synapses. Following loss of their spines, neurons are less able to cope with the large synaptic inputs impinging on their dendritic shafts, and these inputs may lead to their eventual death. Thus, dendritic spines protect neurons from synaptic activity-induced rises in intracellular calcium concentrations.
Collapse
Affiliation(s)
- Menahem Segal
- Department of Neurobiology, The Weizmann Institute, Rehovot, Israel.
| |
Collapse
|
46
|
Keep cool: Memory is retained during hibernation in Alpine marmots. Physiol Behav 2009; 98:78-84. [DOI: 10.1016/j.physbeh.2009.04.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Revised: 03/17/2009] [Accepted: 04/16/2009] [Indexed: 11/22/2022]
|
47
|
Popov VI, Kraev IV, Banks D, Davies HA, Morenkov ED, Stewart MG, Fesenko EE. Three-dimensional ultrastructural and immunohistochemical study of immature neurons in the subgranular zone of the rat dentate gyrus. Biophysics (Nagoya-shi) 2009. [DOI: 10.1134/s0006350909040174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
48
|
Popov VI, Stewart MG. Complexity of contacts between synaptic boutons and dendritic spines in adult rat hippocampus: Three-dimensional reconstructions from serial ultrathin sections in vivo. Synapse 2009; 63:369-77. [DOI: 10.1002/syn.20613] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Kolomiytseva IK, Perepelkina NI, Zharikova AD, Popov VI. Membrane lipids and morphology of brain cortex synaptosomes isolated from hibernating Yakutian ground squirrel. Comp Biochem Physiol B Biochem Mol Biol 2008; 151:386-91. [PMID: 18760375 DOI: 10.1016/j.cbpb.2008.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 08/06/2008] [Accepted: 08/06/2008] [Indexed: 11/18/2022]
Abstract
Synaptosomes were isolated from Yakutian ground squirrel brain cortex of summer and winter hibernating animals in active and torpor states. Synaptosomal membrane cholesterol and phospholipids were determined. The seasonal changes of synaptosomal lipid composition were found. Synaptosomes isolated from hibernating Yakutian ground squirrel brain cortex maintained the cholesterol sphingomyelin, phosphatidylethanolamine, lysophosphatidylcholine, cardiolipin, phosphatidylinositol and phosphatidylserine contents 2.5, 1.8, 2.6, 1.8, 1.6, and 1.3 times less, respectively, and the content of phosphatidylcholine twice as much as the one in summer season. The synaptosomal membrane lipid composition of summer animals was shown to be markedly different from that as hibernating ground squirrels and non-hibernating rodents. It is believed that phenotypic changes of synaptosomal membrane lipid composition in summer Yakutian ground squirrel are the important preparation step for hibernation. The phosphatidylethanolamine content was increased in torpor state compared with winter-active state and the molar ratio of cholesterol/phospholipids in synaptosomal membrane of winter torpor ground squirrels was lower than that in active winter and summer animals. These events were supposed to lead to increase of the synaptosomal membrane fluidity during torpor. Synaptosomes isolated from torpor animals have larger sizes and contain a greater number of synaptic vesicles on the synaptosomal profile area. The synaptosomal membrane lipid composition and synaptosome morphology were involved in phenotypic adaptation of Yakutian ground squirrel to hibernation.
Collapse
Affiliation(s)
- Iskra K Kolomiytseva
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow region, Russia.
| | | | | | | |
Collapse
|