51
|
Ibrahim P, Almeida D, Nagy C, Turecki G. Molecular impacts of childhood abuse on the human brain. Neurobiol Stress 2021; 15:100343. [PMID: 34141833 PMCID: PMC8187840 DOI: 10.1016/j.ynstr.2021.100343] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/24/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Childhood abuse (CA) is a prevalent global health concern, increasing the risk of negative mental health outcomes later in life. In the literature, CA is commonly defined as physical, sexual, and emotional abuse, as well as neglect. Several mental disorders have been associated with CA, including depression, bipolar disorder, schizophrenia, and post-traumatic stress disorder, along with an increased risk of suicide. It is thought that traumatic life events occurring during childhood and adolescence may have a significant impact on essential brain functions, which may persist throughout adulthood. The interaction between the brain and the external environment can be mediated by epigenetic alterations in gene expression, and there is a growing body of evidence to show that such changes occur as a function of CA. Disruptions in the HPA axis, myelination, plasticity, and signaling have been identified in individuals with a history of CA. Understanding the molecular impact of CA on the brain is essential for the development of treatment and prevention measures. In this review, we will summarize studies that highlight the molecular changes associated with CA in the human brain, along with supporting evidence from peripheral studies and animal models. We will also discuss some of the limitations surrounding the study of CA and propose extracellular vesicles as a promising future approach in the field.
Collapse
Affiliation(s)
- Pascal Ibrahim
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Daniel Almeida
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
52
|
Postnatal Sox6 Regulates Synaptic Function of Cortical Parvalbumin-Expressing Neurons. J Neurosci 2021; 41:8876-8886. [PMID: 34503995 PMCID: PMC8549537 DOI: 10.1523/jneurosci.0021-21.2021] [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: 01/05/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 11/29/2022] Open
Abstract
Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood. SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.
Collapse
|
53
|
Learning-induced plasticity in the barrel cortex is disrupted by inhibition of layer 4 somatostatin-containing interneurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1869:119146. [PMID: 34599984 DOI: 10.1016/j.bbamcr.2021.119146] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/29/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022]
Abstract
Gaba-ergic neurons are a diverse cell class with extensive influence over cortical processing, but their role in experience-dependent plasticity is not completely understood. Here we addressed the role of cortical somatostatin- (SOM-INs) and vasoactive intestinal polypeptide- (VIP-INs) containing interneurons in a Pavlovian conditioning where stimulation of the vibrissae is used as a conditioned stimulus and tail shock as unconditioned one. This procedure induces a plastic change observed as an enlargement of the cortical functional representation of vibrissae activated during conditioning. Using layer-targeted, cell-selective DREADD transductions, we examined the involvement of SOM-INs and VIP-INs activity in learning-related plastic changes. Under optical recordings, we injected DREADD-expressing vectors into layer IV (L4) barrels or layer II/III (L2/3) areas corresponding to the activated vibrissae. The activity of the interneurons was modulated during all conditioning sessions, and functional 2-deoxyglucose (2DG) maps were obtained 24 h after the last session. In mice with L4 but not L2/3 SOM-INs suppressed during conditioning, the plastic change of whisker representation was absent. The behavioral effect of conditioning was disturbed. Both L4 SOM-INs excitation and L2/3 VIP-INs inhibition during conditioning did not affect the plasticity or the conditioned response. We found the activity of L4 SOM-INs is indispensable in the formation of learning-induced plastic change. We propose that L4 SOM-INs may provide disinhibition by blocking L4 parvalbumin interneurons, allowing a flow of information into upper cortical layers during learning.
Collapse
|
54
|
Wen Z, Chen ZS, Milad MR. Fear extinction learning modulates large-scale brain connectivity. Neuroimage 2021; 238:118261. [PMID: 34126211 PMCID: PMC8436785 DOI: 10.1016/j.neuroimage.2021.118261] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 11/22/2022] Open
Abstract
Exploring the neural circuits of the extinction of conditioned fear is critical to advance our understanding of fear- and anxiety-related disorders. The field has focused on examining the role of various regions of the medial prefrontal cortex, insular cortex, hippocampus, and amygdala in conditioned fear and its extinction. The contribution of this 'fear network' to the conscious awareness of fear has recently been questioned. And as such, there is a need to examine higher/multiple cortical systems that might contribute to the conscious feeling of fear and anxiety. Herein, we studied functional connectivity patterns across the entire brain to examine the contribution of multiple networks to the acquisition of fear extinction learning and its retrieval. We conducted trial-by-trial analyses on data from 137 healthy participants who underwent a two-day fear conditioning and extinction paradigm in a functional magnetic resonance imaging (fMRI) scanner. We found that functional connectivity across a broad range of brain regions, many of which are part of the default mode, frontoparietal, and ventral attention networks, increased from early to late extinction learning only to a conditioned cue. The increased connectivity during extinction learning predicted the magnitude of extinction memory tested 24 h later. Together, these findings provide evidence supporting recent studies implicating distributed brain regions in learning, consolidation and expression of fear extinction memory in the human brain.
Collapse
Affiliation(s)
- Zhenfu Wen
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States
| | - Zhe Sage Chen
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States; Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, United States; The Neuroscience Institute, New York University School of Medicine, New York, NY, United States
| | - Mohammed R Milad
- Department of Psychiatry, New York University School of Medicine, New York, NY, United States; The Neuroscience Institute, New York University School of Medicine, New York, NY, United States; Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, United States.
| |
Collapse
|
55
|
Mafi AM, Russ MG, Hofer LN, Pham VQ, Young JW, Mellott JG. Inferior collicular cells that project to the auditory thalamus are increasingly surrounded by perineuronal nets with age. Neurobiol Aging 2021; 105:1-15. [PMID: 34004491 PMCID: PMC8338758 DOI: 10.1016/j.neurobiolaging.2021.04.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/30/2021] [Accepted: 04/03/2021] [Indexed: 12/20/2022]
Abstract
The age-related loss of GABA in the inferior colliculus (IC) likely plays a role in the development of age-related hearing loss. Perineuronal nets (PNs), specialized aggregates of extracellular matrix, increase with age in the IC. PNs, associated with GABAergic neurotransmission, can stabilize synapses and inhibit structural plasticity. We sought to determine whether PN expression increased on GABAergic and non-GABAergic IC cells that project to the medial geniculate body (MG). We used retrograde tract-tracing in combination with immunohistochemistry for glutamic acid decarboxylase and Wisteria floribunda agglutinin across three age groups of Fischer Brown Norway rats. Results demonstrate that PNs increase with age on lemniscal and non-lemniscal IC-MG cells, however two key differences exist. First, PNs increased on non-lemniscal IC-MG cells during middle-age, but not until old age on lemniscal IC-MG cells. Second, increases of PNs on lemniscal IC-MG cells occurred on non-GABAergic cells rather than on GABAergic cells. These results suggest that synaptic stabilization and reduced plasticity likely occur at different ages on a subset of the IC-MG pathway.
Collapse
Affiliation(s)
- Amir M Mafi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Matthew G Russ
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Lindsay N Hofer
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Vincent Q Pham
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Jesse W Young
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA
| | - Jeffrey G Mellott
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University, Rootstown, OH USA.
| |
Collapse
|
56
|
Miyata S. Structural and Functional Remodeling of the Extracellular Matrix during Brain Development and Aging. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2003.1e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Shinji Miyata
- Faculty of Agriculture, Tokyo University of Agriculture and Technology
| |
Collapse
|
57
|
Miyata S. Structural and Functional Remodeling of the Extracellular Matrix during Brain Development and Aging. TRENDS GLYCOSCI GLYC 2021. [DOI: 10.4052/tigg.2003.1j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Shinji Miyata
- Faculty of Agriculture, Tokyo University of Agriculture and Technology
| |
Collapse
|
58
|
Lesnikova A, Casarotto P, Moliner R, Fred SM, Biojone C, Castrén E. Perineuronal Net Receptor PTPσ Regulates Retention of Memories. Front Synaptic Neurosci 2021; 13:672475. [PMID: 34366821 PMCID: PMC8339997 DOI: 10.3389/fnsyn.2021.672475] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/25/2021] [Indexed: 12/29/2022] Open
Abstract
Perineuronal nets (PNNs) have an important physiological role in the retention of learning by restricting cognitive flexibility. Their deposition peaks after developmental periods of intensive learning, usually in late childhood, and they help in long-term preservation of newly acquired skills and information. Modulation of PNN function by various techniques enhances plasticity and regulates the retention of memories, which may be beneficial when memory persistence entails negative symptoms such as post-traumatic stress disorder (PTSD). In this study, we investigated the role of PTPσ [receptor-type tyrosine-protein phosphatase S, a phosphatase that is activated by binding of chondroitin sulfate proteoglycans (CSPGs) from PNNs] in retention of memories using Novel Object Recognition and Fear Conditioning models. We observed that mice haploinsufficient for PTPRS gene (PTPσ+/–), although having improved short-term object recognition memory, display impaired long-term memory in both Novel Object Recognition and Fear Conditioning paradigm, as compared to WT littermates. However, PTPσ+/– mice did not show any differences in behavioral tests that do not heavily rely on cognitive flexibility, such as Elevated Plus Maze, Open Field, Marble Burying, and Forced Swimming Test. Since PTPσ has been shown to interact with and dephosphorylate TRKB, we investigated activation of this receptor and its downstream pathways in limbic areas known to be associated with memory. We found that phosphorylation of TRKB and PLCγ are increased in the hippocampus, prefrontal cortex, and amygdaloid complex of PTPσ+/– mice, but other TRKB-mediated signaling pathways are not affected. Our data suggest that PTPσ downregulation promotes TRKB phosphorylation in different brain areas, improves short-term memory performance but disrupts long-term memory retention in the tested animal models. Inhibition of PTPσ or disruption of PNN-PTPσ-TRKB complex might be a potential target for disorders where negative modulation of the acquired memories can be beneficial.
Collapse
Affiliation(s)
| | - Plinio Casarotto
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Rafael Moliner
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Senem Merve Fred
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Caroline Biojone
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
59
|
Hart CG, Karimi-Abdolrezaee S. Recent insights on astrocyte mechanisms in CNS homeostasis, pathology, and repair. J Neurosci Res 2021; 99:2427-2462. [PMID: 34259342 DOI: 10.1002/jnr.24922] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
Astrocytes play essential roles in development, homeostasis, injury, and repair of the central nervous system (CNS). Their development is tightly regulated by distinct spatial and temporal cues during embryogenesis and into adulthood throughout the CNS. Astrocytes have several important responsibilities such as regulating blood flow and permeability of the blood-CNS barrier, glucose metabolism and storage, synapse formation and function, and axon myelination. In CNS pathologies, astrocytes also play critical parts in both injury and repair mechanisms. Upon injury, they undergo a robust phenotypic shift known as "reactive astrogliosis," which results in both constructive and deleterious outcomes. Astrocyte activation and migration at the site of injury provides an early defense mechanism to minimize the extent of injury by enveloping the lesion area. However, astrogliosis also contributes to the inhibitory microenvironment of CNS injury and potentiate secondary injury mechanisms, such as inflammation, oxidative stress, and glutamate excitotoxicity, which facilitate neurodegeneration in CNS pathologies. Intriguingly, reactive astrocytes are increasingly a focus in current therapeutic strategies as their activation can be modulated toward a neuroprotective and reparative phenotype. This review will discuss recent advancements in knowledge regarding the development and role of astrocytes in the healthy and pathological CNS. We will also review how astrocytes have been genetically modified to optimize their reparative potential after injury, and how they may be transdifferentiated into neurons and oligodendrocytes to promote repair after CNS injury and neurodegeneration.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
60
|
The Impact of Stress Within and Across Generations: Neuroscientific and Epigenetic Considerations. Harv Rev Psychiatry 2021; 29:303-317. [PMID: 34049337 DOI: 10.1097/hrp.0000000000000300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The impact of stress and trauma on biological systems in humans can be substantial. They can result in epigenetic changes, accelerated brain development and sexual maturation, and predisposition to psychopathology. Such modifications may be accompanied by behavioral, emotional, and cognitive overtones during one's lifetime. Exposure during sensitive periods of neural development may lead to long-lasting effects that may not be affected by subsequent environmental interventions. The cumulative effects of life stressors in an individual may affect offspring's methylome makeup and epigenetic clocks, neurohormonal modulation and stress reactivity, and physiological and reproductive development. While offspring may suffer deleterious effects from parental stress and their own early-life adversity, these factors may also confer traits that prove beneficial and enhance fitness to their own environment. This article synthesizes the data on how stress shapes biological and behavioral dimensions, drawing from preclinical and human models. Advances in this field of knowledge should potentially allow for an improved understanding of how interventions may be increasingly tailored according to individual biomarkers and developmental history.
Collapse
|
61
|
Jovasevic V, Zhang H, Sananbenesi F, Guedea AL, Soman KV, Wiktorowicz JE, Fischer A, Radulovic J. Primary cilia are required for the persistence of memory and stabilization of perineuronal nets. iScience 2021; 24:102617. [PMID: 34142063 PMCID: PMC8185192 DOI: 10.1016/j.isci.2021.102617] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/02/2021] [Accepted: 05/19/2021] [Indexed: 01/11/2023] Open
Abstract
It is well established that the formation of episodic memories requires multiple hippocampal mechanisms operating on different time scales. Early mechanisms of memory formation (synaptic consolidation) have been extensively characterized. However, delayed mechanisms, which maintain hippocampal activity as memories stabilize in cortical circuits, are not well understood. Here we demonstrate that contrary to the transient expression of early- and delayed-response genes, the expression of cytoskeleton- and extracellular matrix-associated genes remains dynamic even at remote time points. The most profound expression changes clustered around primary cilium-associated and collagen genes. These genes most likely contribute to memory by stabilizing perineuronal nets in the dorsohippocampal CA1 subfield, as revealed by targeted disruptions of the primary cilium or perineuronal nets. The findings show that nonsynaptic, primary cilium-mediated mechanisms are required for the persistence of context memory.
Collapse
Affiliation(s)
- Vladimir Jovasevic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Room 13-100, Montgomery Ward Memorial Building, Chicago, IL 60611, USA
| | - Hui Zhang
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Rose F. Kennedy Center, 1410 Pelham Parkway South, Room 115, Bronx, NY 10461, USA
| | | | - Anita L. Guedea
- Department of Psychiatry and Behavioral Sciences, Northwestern University, Chicago, IL 60611, USA
| | - Kizhake V. Soman
- Division of Infectious Disease, Department of Internal Medicine, UTMB – Galveston, Galveston, TX 77555, USA
| | | | - Andre Fischer
- German Center for Neurodegenerative Diseases, Göttingen 37075, Germany
| | - Jelena Radulovic
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Room 13-100, Montgomery Ward Memorial Building, Chicago, IL 60611, USA
- Department of Neuroscience and Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Rose F. Kennedy Center, 1410 Pelham Parkway South, Room 115, Bronx, NY 10461, USA
| |
Collapse
|
62
|
Hirono M, Karube F, Yanagawa Y. Modulatory Effects of Monoamines and Perineuronal Nets on Output of Cerebellar Purkinje Cells. Front Neural Circuits 2021; 15:661899. [PMID: 34194302 PMCID: PMC8236809 DOI: 10.3389/fncir.2021.661899] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
Classically, the cerebellum has been thought to play a significant role in motor coordination. However, a growing body of evidence for novel neural connections between the cerebellum and various brain regions indicates that the cerebellum also contributes to other brain functions implicated in reward, language, and social behavior. Cerebellar Purkinje cells (PCs) make inhibitory GABAergic synapses with their target neurons: other PCs and Lugaro/globular cells via PC axon collaterals, and neurons in the deep cerebellar nuclei (DCN) via PC primary axons. PC-Lugaro/globular cell connections form a cerebellar cortical microcircuit, which is driven by serotonin and noradrenaline. PCs' primary outputs control not only firing but also synaptic plasticity of DCN neurons following the integration of excitatory and inhibitory inputs in the cerebellar cortex. Thus, strong PC-mediated inhibition is involved in cerebellar functions as a key regulator of cerebellar neural networks. In this review, we focus on physiological characteristics of GABAergic transmission from PCs. First, we introduce monoaminergic modulation of GABAergic transmission at synapses of PC-Lugaro/globular cell as well as PC-large glutamatergic DCN neuron, and a Lugaro/globular cell-incorporated microcircuit. Second, we review the physiological roles of perineuronal nets (PNNs), which are organized components of the extracellular matrix and enwrap the cell bodies and proximal processes, in GABA release from PCs to large glutamatergic DCN neurons and in cerebellar motor learning. Recent evidence suggests that alterations in PNN density in the DCN can regulate cerebellar functions.
Collapse
Affiliation(s)
- Moritoshi Hirono
- Department of Physiology, Wakayama Medical University, Wakayama, Japan
| | - Fuyuki Karube
- Lab of Histology and Cytology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
63
|
Ressler KJ. Translating Across Circuits and Genetics Toward Progress in Fear- and Anxiety-Related Disorders. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2021; 19:247-255. [PMID: 34690590 PMCID: PMC8475910 DOI: 10.1176/appi.focus.19205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/15/2020] [Indexed: 06/13/2023]
Abstract
(Reprinted with permission from Am J Psychiatry 2020; 177:214-222).
Collapse
|
64
|
Gisabella B, Babu J, Valeri J, Rexrode L, Pantazopoulos H. Sleep and Memory Consolidation Dysfunction in Psychiatric Disorders: Evidence for the Involvement of Extracellular Matrix Molecules. Front Neurosci 2021; 15:646678. [PMID: 34054408 PMCID: PMC8160443 DOI: 10.3389/fnins.2021.646678] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/22/2021] [Indexed: 12/13/2022] Open
Abstract
Sleep disturbances and memory dysfunction are key characteristics across psychiatric disorders. Recent advances have revealed insight into the role of sleep in memory consolidation, pointing to key overlap between memory consolidation processes and structural and molecular abnormalities in psychiatric disorders. Ongoing research regarding the molecular mechanisms involved in memory consolidation has the potential to identify therapeutic targets for memory dysfunction in psychiatric disorders and aging. Recent evidence from our group and others points to extracellular matrix molecules, including chondroitin sulfate proteoglycans and their endogenous proteases, as molecules that may underlie synaptic dysfunction in psychiatric disorders and memory consolidation during sleep. These molecules may provide a therapeutic targets for decreasing strength of reward memories in addiction and traumatic memories in PTSD, as well as restoring deficits in memory consolidation in schizophrenia and aging. We review the evidence for sleep and memory consolidation dysfunction in psychiatric disorders and aging in the context of current evidence pointing to the involvement of extracellular matrix molecules in these processes.
Collapse
Affiliation(s)
| | | | | | | | - Harry Pantazopoulos
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
65
|
Ciccarelli A, Weijers D, Kwan W, Warner C, Bourne J, Gross CT. Sexually dimorphic perineuronal nets in the rodent and primate reproductive circuit. J Comp Neurol 2021; 529:3274-3291. [PMID: 33950531 DOI: 10.1002/cne.25167] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022]
Abstract
Perineuronal nets are extracellular glycoprotein structures that have been found on some neurons in the central nervous system and that have been shown to regulate their structural plasticity. Until now work on perineuronal nets has been focused on their role in cortical structures where they are selectively expressed on parvalbumin-positive neurons and are reported to restrict the experience-dependent plasticity of inhibitory afferents. Here, we examined the expression of perineuronal nets subcortically, showing that they are expressed in several discrete structures, including nuclei that comprise the brain network controlling reproductive behaviors (e.g., mounting, lordosis, aggression, and social defense). In particular, perineuronal nets were found in the posterior dorsal division of the medial amygdala, the medial preoptic nucleus, the posterior medial bed nucleus of the stria terminalis, the ventrolateral ventromedial hypothalamus and adjacent tuberal nucleus, and the ventral premammillary nucleus in both the mouse and primate brain. Comparison of perineuronal nets in male and female mice revealed a significant sexually dimorphic expression, with expression found prominently on estrogen receptor expressing neurons in the medial amygdala. These findings suggest that perineuronal nets may be involved in regulating neural plasticity in the mammalian reproductive system.
Collapse
Affiliation(s)
- Alessandro Ciccarelli
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| | - Dilys Weijers
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| | - William Kwan
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Claire Warner
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - James Bourne
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Cornelius T Gross
- Epigenetics & Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome
| |
Collapse
|
66
|
Xia D, Li L, Yang B, Zhou Q. Altered Relationship Between Parvalbumin and Perineuronal Nets in an Autism Model. Front Mol Neurosci 2021; 14:597812. [PMID: 33912009 PMCID: PMC8072465 DOI: 10.3389/fnmol.2021.597812] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 03/16/2021] [Indexed: 12/26/2022] Open
Abstract
Altered function or presence of inhibitory neurons is documented in autism spectrum disorders (ASD), but the mechanism underlying this alternation is poorly understood. One major subtype of inhibitory neurons altered is the parvalbumin (PV)-containing neurons with reduced density and intensity in ASD patients and model mice. A subpopulation of PV+ neurons expresses perineuronal nets (PNN). To better understand whether the relationship between PV and PNN is altered in ASD, we measured quantitatively the intensities of PV and PNN in single PV+ neurons in the prelimbic prefrontal cortex (PrL-PFC) of a valproic acid (VPA) model of ASD at different ages. We found a decreased PV intensity but increased PNN intensity in VPA mice. The relationship between PV and PNN intensities is altered in VPA mice, likely due to an "abnormal" subpopulation of neurons with an altered PV-PNN relationship. Furthermore, reducing PNN level using in vivo injection of chondroitinase ABC corrects the PV expression in adult VPA mice. We suggest that the interaction between PV and PNN is disrupted in PV+ neurons in VPA mice which may contribute to the pathology in ASD.
Collapse
Affiliation(s)
- Dan Xia
- Key Laboratory of Chemical Genome, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, China.,Center for Child Care and Mental Health, Shenzhen Children's Hospital, Shenzhen, China
| | - Li Li
- State Key Laboratory of Organ Failure Research, Department of Biostatistics, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Binrang Yang
- Center for Child Care and Mental Health, Shenzhen Children's Hospital, Shenzhen, China
| | - Qiang Zhou
- Key Laboratory of Chemical Genome, State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Shenzhen Graduate School, Peking University, Shenzhen, China
| |
Collapse
|
67
|
The extracellular matrix regulates cortical layer dynamics and cross-columnar frequency integration in the auditory cortex. Commun Biol 2021; 4:322. [PMID: 33692502 PMCID: PMC7946889 DOI: 10.1038/s42003-021-01837-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 02/11/2021] [Indexed: 01/31/2023] Open
Abstract
In the adult vertebrate brain, enzymatic removal of the extracellular matrix (ECM) is increasingly recognized to promote learning, memory recall, and restorative plasticity. The impact of the ECM on translaminar dynamics during cortical circuit processing is still not understood. Here, we removed the ECM in the primary auditory cortex (ACx) of adult Mongolian gerbils using local injections of hyaluronidase (HYase). Using laminar current-source density (CSD) analysis, we found layer-specific changes of the spatiotemporal synaptic patterns with increased cross-columnar integration and simultaneous weakening of early local sensory input processing within infragranular layers Vb. These changes had an oscillatory fingerprint within beta-band (25-36 Hz) selectively within infragranular layers Vb. To understand the laminar interaction dynamics after ECM digestion, we used time-domain conditional Granger causality (GC) measures to identify the increased drive of supragranular layers towards deeper infragranular layers. These results showed that ECM degradation altered translaminar cortical network dynamics with a stronger supragranular lead of the columnar response profile.
Collapse
|
68
|
An Extracellular Perspective on CNS Maturation: Perineuronal Nets and the Control of Plasticity. Int J Mol Sci 2021; 22:ijms22052434. [PMID: 33670945 PMCID: PMC7957817 DOI: 10.3390/ijms22052434] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
During restricted time windows of postnatal life, called critical periods, neural circuits are highly plastic and are shaped by environmental stimuli. In several mammalian brain areas, from the cerebral cortex to the hippocampus and amygdala, the closure of the critical period is dependent on the formation of perineuronal nets. Perineuronal nets are a condensed form of an extracellular matrix, which surrounds the soma and proximal dendrites of subsets of neurons, enwrapping synaptic terminals. Experimentally disrupting perineuronal nets in adult animals induces the reactivation of critical period plasticity, pointing to a role of the perineuronal net as a molecular brake on plasticity as the critical period closes. Interestingly, in the adult brain, the expression of perineuronal nets is remarkably dynamic, changing its plasticity-associated conditions, including memory processes. In this review, we aimed to address how perineuronal nets contribute to the maturation of brain circuits and the regulation of adult brain plasticity and memory processes in physiological and pathological conditions.
Collapse
|
69
|
Bertocchi I, Mele P, Ferrero G, Oberto A, Carulli D, Eva C. NPY-Y1 receptor signaling controls spatial learning and perineuronal net expression. Neuropharmacology 2020; 184:108425. [PMID: 33285203 DOI: 10.1016/j.neuropharm.2020.108425] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/24/2020] [Accepted: 12/01/2020] [Indexed: 12/17/2022]
Abstract
Perineuronal nets (PNNs) are extracellular matrix structures that form around some types of neurons at the end of critical periods, limiting neuronal plasticity. In the adult brain, PNNs play a crucial role in the regulation of learning and cognitive processes. Neuropeptide Y (NPY) is involved in the regulation of many physiological functions, including learning and memory abilities, via activation of Y1 receptors (Y1Rs). Here we demonstrated that the conditional depletion of the gene encoding the Y1R for NPY in adult forebrain excitatory neurons (Npy1rrfb mutant mice), induces a significant slowdown in spatial learning, which is associated with a robust intensification of PNN expression and an increase in the number of c-Fos expressing cells in the cornus ammonis 1 (CA1) of the dorsal hippocampus. Importantly, the enzymatic digestion of PNNs in CA1 normalizes c-Fos activity and completely rescues learning abilities of Npy1rrfb mice. These data highlight a previously unknown functional link between NPY-Y1R transmission and PNNs, which may play a role in the control of dorsal hippocampal excitability and related cognitive functions.
Collapse
Affiliation(s)
- Ilaria Bertocchi
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy
| | - Paolo Mele
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy
| | - Giuliano Ferrero
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy
| | - Alessandra Oberto
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy
| | - Daniela Carulli
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy; Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, Netherlands
| | - Carola Eva
- Neuroscience Institute of the Cavalieri-Ottolenghi Foundation, 10043, Orbassano, Turin, Italy; Department of Neuroscience, University of Turin, 10126, Turin, Italy; Neuroscience Institute of Turin (NIT), Italy.
| |
Collapse
|
70
|
Gomes FV, Zhu X, Grace AA. The pathophysiological impact of stress on the dopamine system is dependent on the state of the critical period of vulnerability. Mol Psychiatry 2020; 25:3278-3291. [PMID: 31488866 PMCID: PMC7056584 DOI: 10.1038/s41380-019-0514-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/02/2019] [Accepted: 07/18/2019] [Indexed: 12/18/2022]
Abstract
Unregulated stress during critical periods of development is proposed to drive deficits consistent with schizophrenia in adults. If accurate, reopening the critical period could make the adult susceptible to pathology. We evaluated the impact of early adolescent and adult stress exposure (combination of daily footshock for 10 days and 3 restraint sessions) on (1) midbrain dopamine (DA) neuron activity, (2) ventral hippocampal (vHipp) pyramidal neuron activity, and (3) the number of parvalbumin (PV) interneurons in the vHipp and their associated perineuronal nets (PNNs). Ventral tegmental area (VTA) DA neuron population activity and vHipp activity was increased 1-2 and 5-6 weeks post-adolescent stress, along with a decrease in the number of PV+, PNN+, PV + /PNN + cells in the vHipp, which are consistent with the MAM model of schizophrenia. In contrast, adult stress decreased VTA DA neuron population activity only at 1-2 weeks post stress, which is consistent with what has been observed in animal models of depression, without impacting vHipp activity and PV/PNN expression. Administration of valproate (VPA), which can re-instate the critical period of plasticity via histone deacetylase (HDAC) inhibition, caused adult stress to produce changes similar to those induced by adolescent stress, presumably by increasing stress vulnerability to early adolescent levels. Our findings indicate that timing of stress is a critical determinant of the pathology produced in the adult: adolescent stress led to circuit deficits that recapitulates schizophrenia, whereas adult stress induced a depression-like hypodopaminergic state. Reopening the critical period in the adult restores vulnerability to stress-induced pathology resembling schizophrenia.
Collapse
Affiliation(s)
- Felipe V. Gomes
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| | - Xiyu Zhu
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| | - Anthony A. Grace
- Departments of Neuroscience, Psychiatry and Psychology, University of Pittsburgh, PA, USA
| |
Collapse
|
71
|
Sugitani K, Egorova D, Mizumoto S, Nishio S, Yamada S, Kitagawa H, Oshima K, Nadano D, Matsuda T, Miyata S. Hyaluronan degradation and release of a hyaluronan-aggrecan complex from perineuronal nets in the aged mouse brain. Biochim Biophys Acta Gen Subj 2020; 1865:129804. [PMID: 33253804 DOI: 10.1016/j.bbagen.2020.129804] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Perineuronal nets (PNNs) are insoluble aggregates of extracellular matrix molecules in the brain that consist of hyaluronan (HA) and chondroitin sulfate proteoglycans (CSPGs). PNNs promote the acquisition and storage of memories by stabilizing the formation of synapses in the adult brain. Although the deterioration of PNNs has been suggested to contribute to the age-dependent decline in brain function, the molecular mechanisms underlying age-related changes in PNNs remain unclear. METHODS The amount and solubility of PNN components were investigated by sequential extraction followed by a disaccharide analysis and immunoblotting. We examined the interaction between HA and aggrecan, a major HA-binding CSPG, by combining mass spectrometry and pull-down assays. RESULTS The solubility and amount of HA increased in the brain with age. Among several CSPGs, the solubility of aggrecan was selectively elevated during aging. In contrast to alternations in biochemical properties, the expression of PNN components at the transcript level was not markedly changed by aging. The increased solubility of aggrecan was not due to the loss of HA-binding properties. Our results indicated that the degradation of high-molecular-mass HA induced the release of the HA-aggrecan complex from PNNs in the aged brain. CONCLUSION The present study revealed a novel mechanism underlying the age-related deterioration of PNNs in the brain.
Collapse
Affiliation(s)
- Kei Sugitani
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Diana Egorova
- Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan
| | - Shuji Mizumoto
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Shunsuke Nishio
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-Ku, Nagoya 468-8503, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakitamachi, Higashinada-Ku, Kobe 658-8558, Japan
| | - Kenzi Oshima
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Daita Nadano
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan
| | - Tsukasa Matsuda
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Food and Agricultural Sciences, Fukushima University, Kanayagawa 1, Fukushima 960-1296, Japan
| | - Shinji Miyata
- Graduate School of Bioagricultural Sciences, Nagoya University, Furocho, Chikusa-Ku, Nagoya 464-8601, Japan; Faculty of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwaicho, Fuchu, Tokyo 183-8509, Japan.
| |
Collapse
|
72
|
Spijker S, Koskinen MK, Riga D. Incubation of depression: ECM assembly and parvalbumin interneurons after stress. Neurosci Biobehav Rev 2020; 118:65-79. [DOI: 10.1016/j.neubiorev.2020.07.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
|
73
|
Guadagno A, Verlezza S, Long H, Wong TP, Walker CD. It Is All in the Right Amygdala: Increased Synaptic Plasticity and Perineuronal Nets in Male, But Not Female, Juvenile Rat Pups after Exposure to Early-Life Stress. J Neurosci 2020; 40:8276-8291. [PMID: 32978287 PMCID: PMC7577595 DOI: 10.1523/jneurosci.1029-20.2020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 01/09/2023] Open
Abstract
Early-life stress (ELS) is associated with increased vulnerability to mental disorders. The basolateral amygdala (BLA) plays a critical role in fear conditioning and is extremely sensitive to ELS. Using a naturalistic rodent model of ELS, the limited bedding paradigm (LB) between postnatal days 1-10, we previously documented that LB male, but not female preweaning rat pups display increased BLA neuron spine density paralleled with enhanced evoked synaptic responses and altered BLA functional connectivity. Since ELS effects are often sexually dimorphic and amygdala processes exhibit hemispheric asymmetry, we investigated changes in synaptic plasticity and neuronal excitability of BLA neurons in vitro in the left and right amygdala of postnatal days 22-28 male and female offspring from normal bedding or LB mothers. We report that LB conditions enhanced synaptic plasticity in the right, but not the left BLA of males exclusively. LB males also showed increased perineuronal net density, particularly around parvalbumin (PV) cells, and impaired fear-induced activity of PV interneurons only in the right BLA. Action potentials fired from right BLA neurons of LB females displayed slower maximal depolarization rates and decreased amplitudes compared with normal bedding females, concomitant with reduced NMDAR GluN1 subunit expression in the right BLA. In LB males, reduced GluA2 expression in the right BLA might contribute to the enhanced LTP. These findings suggest that LB differentially programs synaptic plasticity and PV/perineuronal net development in the left and right BLA. Furthermore, our study demonstrates that the effects of ELS exposure on BLA synaptic function are sexually dimorphic and possibly recruiting different mechanisms.SIGNIFICANCE STATEMENT Early-life stress (ELS) induces long-lasting consequences on stress responses and emotional regulation in humans, increasing vulnerability to the development of psychopathologies. The effects of ELS in a number of brain regions, including the amygdala, are often sexually dimorphic, and have been reproduced using the rodent limited bedding paradigm of early adversity. The present study examines sex differences in synaptic plasticity and cellular activation occurring in the developing left and right amygdala after limited bedding exposure, a phenomenon that could shape long-term emotional behavioral outcomes. Studying how ELS selectively produces effects in one amygdala hemisphere during a critical period of brain development could guide further investigation into sex-dependent mechanisms and allow for more targeted and improved treatment of stress-and emotionality-related disorders.
Collapse
Affiliation(s)
- Angela Guadagno
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, H3A 0G4, Canada
| | - Silvanna Verlezza
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Hong Long
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
| | - Tak Pan Wong
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 0G4, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, Quebec, H4H 1R3, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, H3A 0G4, Canada
| |
Collapse
|
74
|
Premachandran H, Zhao M, Arruda-Carvalho M. Sex Differences in the Development of the Rodent Corticolimbic System. Front Neurosci 2020; 14:583477. [PMID: 33100964 PMCID: PMC7554619 DOI: 10.3389/fnins.2020.583477] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/01/2020] [Indexed: 12/18/2022] Open
Abstract
In recent years, a growing body of research has shown sex differences in the prevalence and symptomatology of psychopathologies, such as depression, anxiety, and fear-related disorders, all of which show high incidence rates in early life. This has highlighted the importance of including female subjects in animal studies, as well as delineating sex differences in neural processing across development. Of particular interest is the corticolimbic system, comprising the hippocampus, amygdala, and medial prefrontal cortex. In rodents, these corticolimbic regions undergo dynamic changes in early life, and disruption to their normative development is believed to underlie the age and sex-dependent effects of stress on affective processing. In this review, we consolidate research on sex differences in the hippocampus, amygdala, and medial prefrontal cortex across early development. First, we briefly introduce current principles on sexual differentiation of the rodent brain. We then showcase corticolimbic regional sex differences in volume, morphology, synaptic organization, cell proliferation, microglia, and GABAergic signaling, and explain how these differences are influenced by perinatal and pubertal gonadal hormones. In compiling this research, we outline evidence of what and when sex differences emerge in the developing corticolimbic system, and illustrate how temporal dynamics of its maturational trajectory may differ in male and female rodents. This will help provide insight into potential neural mechanisms underlying sex-specific critical windows for stress susceptibility and behavioral emergence.
Collapse
Affiliation(s)
| | - Mudi Zhao
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada
| | - Maithe Arruda-Carvalho
- Department of Psychology, University of Toronto Scarborough, Toronto, ON, Canada.,Department of Cell and Systems Biology, University of Toronto Scarborough, Toronto, ON, Canada
| |
Collapse
|
75
|
Blanco I, Conant K. Extracellular matrix remodeling with stress and depression: Studies in human, rodent and zebrafish models. Eur J Neurosci 2020; 53:3879-3888. [PMID: 32673433 DOI: 10.1111/ejn.14910] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 06/17/2020] [Accepted: 07/09/2020] [Indexed: 01/17/2023]
Abstract
Emerging evidence suggests that extracellular matrix (ECM) alterations occur with stress. Specifically, increases in perineuronal net (PNN) deposition have been observed in rodents exposed to chronic corticosterone or persistent social defeat stress. The PNN is a specific form of ECM that is predominantly localized to parvalbumin (PV)-expressing inhibitory interneurons where it modulates neuronal excitability and brain oscillations that are influenced by the same. Consistent with a role for ECM changes in contributing to the depressive phenotype, recent studies have demonstrated that monoamine reuptake inhibitor type antidepressants can reduce PNN deposition, improve behavior and stimulate changes in gamma oscillatory power that may be important to mood and memory. The present review will highlight studies in humans, rodents and zebrafish that have examined stress, PNN deposition and/or gamma oscillations with a focus on potential cellular and molecular underpinnings.
Collapse
Affiliation(s)
- Ismary Blanco
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA.,Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
76
|
Crapser JD, Ochaba J, Soni N, Reidling JC, Thompson LM, Green KN. Microglial depletion prevents extracellular matrix changes and striatal volume reduction in a model of Huntington's disease. Brain 2020; 143:266-288. [PMID: 31848580 DOI: 10.1093/brain/awz363] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/22/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
Huntington's disease is associated with a reactive microglial response and consequent inflammation. To address the role of these cells in disease pathogenesis, we depleted microglia from R6/2 mice, a rapidly progressing model of Huntington's disease marked by behavioural impairment, mutant huntingtin (mHTT) accumulation, and early death, through colony-stimulating factor 1 receptor inhibition (CSF1Ri) with pexidartinib (PLX3397) for the duration of disease. Although we observed an interferon gene signature in addition to downregulated neuritogenic and synaptic gene pathways with disease, overt inflammation was not evident by microglial morphology or cytokine transcript levels in R6/2 mice. Nonetheless, CSF1Ri-induced microglial elimination reduced or prevented disease-related grip strength and object recognition deficits, mHTT accumulation, astrogliosis, and striatal volume loss, the latter of which was not associated with reductions in cell number but with the extracellular accumulation of chondroitin sulphate proteoglycans (CSPGs)-a primary component of glial scars. A concurrent loss of proteoglycan-containing perineuronal nets was also evident in R6/2 mice, and microglial elimination not only prevented this but also strikingly increased perineuronal nets in the brains of naïve littermates, suggesting a new role for microglia as homeostatic regulators of perineuronal net formation and integrity.
Collapse
Affiliation(s)
- Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Joseph Ochaba
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Neelakshi Soni
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jack C Reidling
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.,Department of Psychiatry and Human Behavior, University of California, Irvine, CA, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine (UCI), Irvine, CA, USA.,Institute of Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| |
Collapse
|
77
|
Crapser JD, Spangenberg EE, Barahona RA, Arreola MA, Hohsfield LA, Green KN. Microglia facilitate loss of perineuronal nets in the Alzheimer's disease brain. EBioMedicine 2020; 58:102919. [PMID: 32745992 PMCID: PMC7399129 DOI: 10.1016/j.ebiom.2020.102919] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Microglia, the brain's principal immune cell, are increasingly implicated in Alzheimer's disease (AD), but the molecular interfaces through which these cells contribute to amyloid beta (Aβ)-related neurodegeneration are unclear. We recently identified microglial contributions to the homeostatic and disease-associated modulation of perineuronal nets (PNNs), extracellular matrix structures that enwrap and stabilize neuronal synapses, but whether PNNs are altered in AD remains controversial. METHODS Extensive histological analysis was performed on male and female 5xFAD mice at 4, 8, 12, and 18 months of age to assess plaque burden, microgliosis, and PNNs. Findings were validated in postmortem AD tissue. The role of neuroinflammation in PNN loss was investigated via LPS treatment, and the ability to prevent or rescue disease-related reductions in PNNs was assessed by treating 5xFAD and 3xTg-AD model mice with colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 to deplete microglia. FINDINGS Utilizing the 5xFAD mouse model and human cortical tissue, we report that PNNs are extensively lost in AD in proportion to plaque burden. Activated microglia closely associate with and engulf damaged nets in the 5xFAD brain, and inclusions of PNN material are evident in mouse and human microglia, while aggrecan, a critical PNN component, deposits within human dense-core plaques. Disease-associated reductions in parvalbumin (PV)+ interneurons, frequently coated by PNNs, are preceded by PNN coverage and integrity impairments, and similar phenotypes are elicited in wild-type mice following microglial activation with LPS. Chronic pharmacological depletion of microglia prevents 5xFAD PNN loss, with similar results observed following depletion in aged 3xTg-AD mice, and this occurs despite plaque persistence. INTERPRETATION We conclude that phenotypically altered microglia facilitate plaque-dependent PNN loss in the AD brain. FUNDING The NIH (NIA, NINDS) and the Alzheimer's Association.
Collapse
Affiliation(s)
- Joshua D Crapser
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | | | - Rocio A Barahona
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Miguel A Arreola
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Lindsay A Hohsfield
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA.
| |
Collapse
|
78
|
Circadian Rhythms of Perineuronal Net Composition. eNeuro 2020; 7:ENEURO.0034-19.2020. [PMID: 32719104 PMCID: PMC7405073 DOI: 10.1523/eneuro.0034-19.2020] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/19/2020] [Accepted: 05/22/2020] [Indexed: 12/23/2022] Open
Abstract
Perineuronal nets (PNNs) are extracellular matrix (ECM) structures that envelop neurons and regulate synaptic functions. Long thought to be stable structures, PNNs have been recently shown to respond dynamically during learning, potentially regulating the formation of new synapses. We postulated that PNNs vary during sleep, a period of active synaptic modification. Notably, PNN components are cleaved by matrix proteases such as the protease cathepsin-S. This protease is diurnally expressed in the mouse cortex, coinciding with dendritic spine density rhythms. Thus, cathepsin-S may contribute to PNN remodeling during sleep, mediating synaptic reorganization. These studies were designed to test the hypothesis that PNN numbers vary in a diurnal manner in the rodent and human brain, as well as in a circadian manner in the rodent brain, and that these rhythms are disrupted by sleep deprivation. In mice, we observed diurnal and circadian rhythms of PNNs labeled with the lectin Wisteria floribunda agglutinin (WFA+ PNNs) in several brain regions involved in emotional memory processing. Sleep deprivation prevented the daytime decrease of WFA+ PNNs and enhances fear memory extinction. Diurnal rhythms of cathepsin-S expression in microglia were observed in the same brain regions, opposite to PNN rhythms. Finally, incubation of mouse sections with cathepsin-S eliminated PNN labeling. In humans, WFA+ PNNs showed a diurnal rhythm in the amygdala and thalamic reticular nucleus (TRN). Our results demonstrate that PNNs vary in a circadian manner and this is disrupted by sleep deprivation. We suggest that rhythmic modification of PNNs may contribute to memory consolidation during sleep.
Collapse
|
79
|
Yang X. Chondroitin sulfate proteoglycans: key modulators of neuronal plasticity, long-term memory, neurodegenerative, and psychiatric disorders. Rev Neurosci 2020; 31:555-568. [PMID: 32126020 DOI: 10.1515/revneuro-2019-0117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/02/2020] [Indexed: 12/19/2022]
Abstract
The chondroitin sulfate proteoglycans (CSPGs) are large groups of heterogenous proteoglycans that are mainly expressed by reactive astrocytes in the central nervous system (CNS). They share similar core proteins and are post-transcriptionally modified by chondroitin sulfate glycosaminoglycans. CSPGs are the major components of the perineuronal nets (PNN) that regulate the opening and closure of the critical period. Mounting reports have documented the crucial roles of CSPGs in restricting neuronal plasticity, axonal growth, and pathfinding during development as well as axonal regeneration after CNS injury. Moreover, CSPGs and PNNs modulate long-term memory, which impairments frequently happened in several neurodegenerative and psychiatric disorders. This review will shortly introduce the expression patterns of CSPGs during development and after injury, the PNNs constitutions, the roles of CSPGs and PNNs in axonal regrowth, discuss the most recently identified roles of CSPGs and PNNs in mediating long-term memory and their correlation with brain disorders, and finally, propose a short perspective of future investigations. Hopefully, further explorations may validate the therapeutic potentials of PNNs and CSPGs.
Collapse
Affiliation(s)
- Xin Yang
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, P.R. China
| |
Collapse
|
80
|
Assmann A, Richter A, Schütze H, Soch J, Barman A, Behnisch G, Knopf L, Raschick M, Schult A, Wüstenberg T, Behr J, Düzel E, Seidenbecher CI, Schott BH. Neurocan genome-wide psychiatric risk variant affects explicit memory performance and hippocampal function in healthy humans. Eur J Neurosci 2020; 53:3942-3959. [PMID: 32583466 DOI: 10.1111/ejn.14872] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 12/26/2022]
Abstract
Alterations of the brain extracellular matrix (ECM) can perturb the structure and function of brain networks like the hippocampus, a key region in human memory that is commonly affected in psychiatric disorders. Here, we investigated the potential effects of a genome-wide psychiatric risk variant in the NCAN gene encoding the ECM proteoglycan neurocan (rs1064395) on memory performance, hippocampal function and cortical morphology in young, healthy volunteers. We assessed verbal memory performance in two cohorts (N = 572, 302) and found reduced recall performance in risk allele (A) carriers across both cohorts. In 117 participants, we performed functional magnetic resonance imaging using a novelty-encoding task with visual scenes. Risk allele carriers showed higher false alarm rates during recognition, accompanied by inefficiently increased left hippocampal activation. To assess effects of rs1064395 on brain morphology, we performed voxel-based morphometry in 420 participants from four independent cohorts and found lower grey matter density in the ventrolateral and rostral prefrontal cortex of risk allele carriers. In silico eQTL analysis revealed that rs1064395 SNP is linked not only to increased prefrontal expression of the NCAN gene itself, but also of the neighbouring HAPLN4 gene, suggesting a more complex effect of the SNP on ECM composition. Our results suggest that the NCAN rs1064395 A allele is associated with lower hippocampus-dependent memory function, variation of prefrontal cortex structure and ECM composition. Considering the well-documented hippocampal and prefrontal dysfunction in bipolar disorder and schizophrenia, our results may reflect an intermediate phenotype by which NCAN rs1064395 contributes to disease risk.
Collapse
Affiliation(s)
- Anne Assmann
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Neurology, Otto von Guericke University, Magdeburg, Germany.,German Center for Neurodegenerative Diseases, Magdeburg, Germany
| | - Anni Richter
- Leibniz Institute for Neurobiology, Magdeburg, Germany
| | - Hartmut Schütze
- German Center for Neurodegenerative Diseases, Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University, Magdeburg, Germany
| | - Joram Soch
- German Center for Neurodegenerative Diseases, Göttingen, Germany.,Bernstein Center for Computational Neuroscience, Humboldt University, Berlin, Germany
| | | | | | - Lea Knopf
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Matthias Raschick
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Annika Schult
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Department of Neurology, Otto von Guericke University, Magdeburg, Germany
| | - Torsten Wüstenberg
- Department of Psychiatry and Psychotherapy, Charité University Medicine, Berlin, Germany.,Department of Clinical Psychology and Psychotherapy, Institute of Psychology, University of Heidelberg, Heidelberg, Germany
| | - Joachim Behr
- Department of Psychiatry and Psychotherapy, Charité University Medicine, Berlin, Germany.,Department of Psychiatry and Psychotherapy, Medical School Brandenburg, Neuruppin, Germany
| | - Emrah Düzel
- German Center for Neurodegenerative Diseases, Magdeburg, Germany.,Institute of Cognitive Neurology and Dementia Research, Otto von Guericke University, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Constanze I Seidenbecher
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Björn H Schott
- Leibniz Institute for Neurobiology, Magdeburg, Germany.,German Center for Neurodegenerative Diseases, Göttingen, Germany.,Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Göttingen, Germany
| |
Collapse
|
81
|
Lateralized Expression of Cortical Perineuronal Nets during Maternal Experience is Dependent on MECP2. eNeuro 2020; 7:ENEURO.0500-19.2020. [PMID: 32332080 PMCID: PMC7294466 DOI: 10.1523/eneuro.0500-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023] Open
Abstract
Cortical neuronal circuits along the sensorimotor pathways are shaped by experience during critical periods of heightened plasticity in early postnatal development. After closure of critical periods, measured histologically by the formation and maintenance of extracellular matrix structures called perineuronal nets (PNNs), the adult mouse brain exhibits restricted plasticity and maturity. Mature PNNs are typically considered to be stable structures that restrict synaptic plasticity on cortical parvalbumin+ (PV+) GABAergic neurons. Changes in environment (i.e., novel behavioral training) or social contexts (i.e., motherhood) are known to elicit synaptic plasticity in relevant neural circuitry. However, little is known about concomitant changes in the PNNs surrounding the cortical PV+ GABAergic neurons. Here, we show novel changes in PNN density in the primary somatosensory cortex (SS1) of adult female mice after maternal experience [called surrogate (Sur)], using systematic microscopy analysis of a whole brain region. On average, PNNs were increased in the right barrel field and decreased in the left forelimb regions. Individual mice had left hemisphere dominance in PNN density. Using adult female mice deficient in methyl-CpG-binding protein 2 (MECP2), an epigenetic regulator involved in regulating experience-dependent plasticity, we found that MECP2 is critical for this precise and dynamic expression of PNN. Adult naive Mecp2-heterozygous (Het) females had increased PNN density in specific subregions in both hemispheres before maternal experience, compared with wild-type (WT) littermate controls. The laterality in PNN expression seen in naive Het (NH) was lost after maternal experience in Sur Het (SH) mice, suggesting possible intact mechanisms for plasticity. Together, our results identify subregion and hemisphere-specific alterations in PNN expression in adult females, suggesting extracellular matrix plasticity as a possible neurobiological mechanism for adult behaviors in rodents.
Collapse
|
82
|
Ribic A. Stability in the Face of Change: Lifelong Experience-Dependent Plasticity in the Sensory Cortex. Front Cell Neurosci 2020; 14:76. [PMID: 32372915 PMCID: PMC7186337 DOI: 10.3389/fncel.2020.00076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 03/17/2020] [Indexed: 11/13/2022] Open
Abstract
Plasticity is a fundamental property of the nervous system that enables its adaptations to the ever-changing environment. Heightened plasticity typical for developing circuits facilitates their robust experience-dependent functional maturation. This plasticity wanes during adolescence to permit the stabilization of mature brain function, but abundant evidence supports that adult circuits exhibit both transient and long-term experience-induced plasticity. Cortical plasticity has been extensively studied throughout the life span in sensory systems and the main distinction between development and adulthood arising from these studies is the concept that passive exposure to relevant information is sufficient to drive robust plasticity early in life, while higher-order attentional mechanisms are necessary to drive plastic changes in adults. Recent work in the primary visual and auditory cortices began to define the circuit mechanisms that govern these processes and enable continuous adaptation to the environment, with transient circuit disinhibition emerging as a common prerequisite for both developmental and adult plasticity. Drawing from studies in visual and auditory systems, this review article summarizes recent reports on the circuit and cellular mechanisms of experience-driven plasticity in the developing and adult brains and emphasizes the similarities and differences between them. The benefits of distinct plasticity mechanisms used at different ages are discussed in the context of sensory learning, as well as their relationship to maladaptive plasticity and neurodevelopmental brain disorders. Knowledge gaps and avenues for future work are highlighted, and these will hopefully motivate future research in these areas, particularly those about the learning of complex skills during development.
Collapse
Affiliation(s)
- Adema Ribic
- Department of Psychology, College and Graduate School of Arts and Sciences, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
83
|
Uriarte N, Ferreño M, Méndez D, Nogueira J. Reorganization of perineuronal nets in the medial Preoptic Area during the reproductive cycle in female rats. Sci Rep 2020; 10:5479. [PMID: 32214157 PMCID: PMC7096482 DOI: 10.1038/s41598-020-62163-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/09/2020] [Indexed: 01/01/2023] Open
Abstract
Perineuronal nets (PNNs) are aggregations of extracellular matrix associated with specific neuronal populations in the central nervous system, suggested to play key roles in neural development, synaptogenesis and experience-dependent synaptic plasticity. Pregnancy and lactation are characterized by a dramatic increase in neuroplasticity. However, dynamic changes in the extracellular matrix associated with maternal circuits have been mostly overlooked. We analyzed the structure of PNNs in an essential nucleus of the maternal circuit, the medial preoptic area (mPOA), during the reproductive cycle of rats, using the Wisteria floribunda (WFA) label. PNNs associated to neurons in the mPOA start to assemble halfway through gestation and become highly organized prior to parturition, fading through the postpartum period. This high expression of PNNs during pregnancy appears to be mediated by the influence of estrogen, progesterone and prolactin, since a hormonal simulated-gestation treatment induced the expression of PNNs in ovariectomized females. We found that PNNs associated neurons in the mPOA express estrogen receptor α and progesterone receptors, supporting a putative role of reproductive hormones in the signaling mechanisms that trigger the assembly of PNNs in the mPOA. This is the first report of PNNs presence and remodeling in mPOA during adulthood induced by physiological variables.
Collapse
Affiliation(s)
- Natalia Uriarte
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, 11400, Uruguay
| | - Marcela Ferreño
- Laboratorio de Neurociencias, Facultad de Ciencias, Universidad de la República, Montevideo, 11400, Uruguay
| | - Diego Méndez
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, 11800, Uruguay
| | - Javier Nogueira
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, 11800, Uruguay.
| |
Collapse
|
84
|
Logue MW, Miller MW, Wolf EJ, Huber BR, Morrison FG, Zhou Z, Zheng Y, Smith AK, Daskalakis NP, Ratanatharathorn A, Uddin M, Nievergelt CM, Ashley-Koch AE, Baker DG, Beckham JC, Garrett ME, Boks MP, Geuze E, Grant GA, Hauser MA, Kessler RC, Kimbrel NA, Maihofer AX, Marx CE, Qin XJ, Risbrough VB, Rutten BPF, Stein MB, Ursano RJ, Vermetten E, Vinkers CH, Ware EB, Stone A, Schichman SA, McGlinchey RE, Milberg WP, Hayes JP, Verfaellie M, the Traumatic Stress Brain Study Group FriedmanMatthew J.AlvarezVictor E.BenedekDavidBradyChristopherCruzDianneDavisDavid A.DumanRonald S.GirgentiMatthew J.HardegreeMelanieHoltzheimerPaul E.KeaneTerence M.KowellNeilKrystalJohn H.McKeeAnnMarxBrianMashDeborahScottWilliam K.SteinThorWilliamsonDouglas E.YoungKeith A.. An epigenome-wide association study of posttraumatic stress disorder in US veterans implicates several new DNA methylation loci. Clin Epigenetics 2020; 12:46. [PMID: 32171335 PMCID: PMC7071645 DOI: 10.1186/s13148-020-0820-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous studies using candidate gene and genome-wide approaches have identified epigenetic changes in DNA methylation (DNAm) associated with posttraumatic stress disorder (PTSD). METHODS In this study, we performed an EWAS of PTSD in a cohort of Veterans (n = 378 lifetime PTSD cases and 135 controls) from the Translational Research Center for TBI and Stress Disorders (TRACTS) cohort assessed using the Illumina EPIC Methylation BeadChip which assesses DNAm at more than 850,000 sites throughout the genome. Our model included covariates for ancestry, cell heterogeneity, sex, age, and a smoking score based on DNAm at 39 smoking-associated CpGs. We also examined in EPIC-based DNAm data generated from pre-frontal cortex (PFC) tissue from the National PTSD Brain Bank (n = 72). RESULTS The analysis of blood samples yielded one genome-wide significant association with PTSD at cg19534438 in the gene G0S2 (p = 1.19 × 10-7, padj = 0.048). This association was replicated in an independent PGC-PTSD-EWAS consortium meta-analysis of military cohorts (p = 0.0024). We also observed association with the smoking-related locus cg05575921 in AHRR despite inclusion of a methylation-based smoking score covariate (p = 9.16 × 10-6), which replicates a previously observed PGC-PTSD-EWAS association (Smith et al. 2019), and yields evidence consistent with a smoking-independent effect. The top 100 EWAS loci were then examined in the PFC data. One of the blood-based PTSD loci, cg04130728 in CHST11, which was in the top 10 loci in blood, but which was not genome-wide significant, was significantly associated with PTSD in brain tissue (in blood p = 1.19 × 10-5, padj = 0.60, in brain, p = 0.00032 with the same direction of effect). Gene set enrichment analysis of the top 500 EWAS loci yielded several significant overlapping GO terms involved in pathogen response, including "Response to lipopolysaccharide" (p = 6.97 × 10-6, padj = 0.042). CONCLUSIONS The cross replication observed in independent cohorts is evidence that DNA methylation in peripheral tissue can yield consistent and replicable PTSD associations, and our results also suggest that that some PTSD associations observed in peripheral tissue may mirror associations in the brain.
Collapse
Affiliation(s)
- Mark W. Logue
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.475010.70000 0004 0367 5222,Biomedical Genetics, Boston University School of Medicine, Boston, MA USA ,grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Mark W. Miller
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Erika J. Wolf
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Bertrand Russ Huber
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Filomene G. Morrison
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA
| | - Zhenwei Zhou
- grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Yuanchao Zheng
- grid.189504.10000 0004 1936 7558Department of Biostatistics, Boston University School of Public Health, Boston, MA USA
| | - Alicia K. Smith
- grid.189967.80000 0001 0941 6502Department of Gynecology and Obstetrics, Emory University, Atlanta, GA USA ,grid.189967.80000 0001 0941 6502Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA USA
| | - Nikolaos P. Daskalakis
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.240206.20000 0000 8795 072XMcLean Hospital, Belmont, MA USA ,Cohen Veterans Bioscience, Cambridge, MA USA ,grid.59734.3c0000 0001 0670 2351Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Andrew Ratanatharathorn
- grid.21729.3f0000000419368729Department of Epidemiology, Columbia University, New York, NY USA
| | - Monica Uddin
- grid.170693.a0000 0001 2353 285XGenomics Program, University of South Florida College of Public Health, Tampa, FL USA ,grid.170693.a0000 0001 2353 285X,Global Health and Infectious Disease Research Program, University of South Florida College of Public Health, Tampa, FL USA
| | - Caroline M. Nievergelt
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Allison E. Ashley-Koch
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Dewleen G. Baker
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Jean C. Beckham
- grid.26009.3d0000 0004 1936 7961Department of Psychiatry and Behavioral Sciences, Duke University, Durham, NC USA ,grid.410332.70000 0004 0419 9846Research, Durham VA Medical Center, Durham, NC USA ,grid.281208.10000 0004 0419 3073Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center (MIRECC), Durham, NC USA
| | - Melanie E. Garrett
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Marco P. Boks
- grid.7692.a0000000090126352Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, Utrecht Netherlands
| | - Elbert Geuze
- grid.7692.a0000000090126352Department of Psychiatry, UMC Utrecht Brain Center, Utrecht, Utrecht Netherlands ,Brain Research and Innovation Centre, Netherlands Ministry of Defence, Utrecht, Utrecht Netherlands
| | - Gerald A. Grant
- grid.240952.80000000087342732Department of Neurosurgery, Stanford University Medical Center, Stanford, CA USA
| | - Michael A. Hauser
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Ronald C. Kessler
- grid.38142.3c000000041936754XDepartment of Health Care Policy, Harvard Medical School, Boston, MA USA
| | - Nathan A. Kimbrel
- grid.410332.70000 0004 0419 9846Research, Durham VA Medical Center, Durham, NC USA ,grid.281208.10000 0004 0419 3073Genetics Research Laboratory, VA Mid-Atlantic Mental Illness Research, Education, and Clinical Center (MIRECC), Durham, NC USA ,grid.26009.3d0000 0004 1936 7961Duke Molecular Physiology Institute, Duke University, Durham, NC USA
| | - Adam X. Maihofer
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Christine E. Marx
- grid.21925.3d0000 0004 1936 9000Department of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, Pittsburgh, PA USA ,grid.189509.c0000000100241216Department of Psychiatry & Behavioral Sciences, Duke University Medical Center, Durham, NC USA
| | - Xue-Jun Qin
- grid.189509.c0000000100241216Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC USA
| | - Victoria B. Risbrough
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Center of Excellence for Stress and Mental Health, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Bart P. F. Rutten
- grid.412966.e0000 0004 0480 1382School for Mental Health and Neuroscience, Department of Psychiatry and Neuropsychology, Maastricht Universitair Medisch Centrum, Maastricht, Limburg Netherlands
| | - Murray B. Stein
- grid.266100.30000 0001 2107 4242Department of Psychiatry, University of California San Diego, La Jolla, CA USA ,grid.410371.00000 0004 0419 2708Psychiatry Service, Veterans Affairs San Diego Healthcare System, San Diego, CA USA ,grid.410371.00000 0004 0419 2708Million Veteran Program, Veterans Affairs San Diego Healthcare System, San Diego, CA USA
| | - Robert J. Ursano
- grid.265436.00000 0001 0421 5525Department of Psychiatry, Uniformed Services University, Bethesda, MD USA
| | - Eric Vermetten
- Arq, Psychotrauma Reseach Expert Group, Diemen, NH Netherlands ,grid.10419.3d0000000089452978Department of Psychiatry, Leiden University Medical Center, Leiden, ZH Netherlands ,Netherlands Defense Department, Research Center, Utrecht, UT Netherlands ,grid.137628.90000 0004 1936 8753Department of Psychiatry, New York University School of Medicine, New York, NY USA
| | - Christiaan H. Vinkers
- Department of Anatomy and Neurosciences, Amsterdam UMC (location VUmc), Amsterdam, Holland Netherlands ,Department of Psychiatry, Amsterdam UMC (location VUmc), Amsterdam, Holland Netherlands
| | - Erin B. Ware
- grid.214458.e0000000086837370Institute for Social Research, Survey Research Center, University of Michigan, Michigan, MI USA
| | - Annjanette Stone
- grid.413916.80000 0004 0419 1545Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR USA
| | - Steven A. Schichman
- grid.413916.80000 0004 0419 1545Pharmacogenomics Analysis Laboratory, Research Service, Central Arkansas Veterans Healthcare System, Little Rock, AR USA
| | - Regina E. McGlinchey
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Health Care System, Boston, MA USA
| | - William P. Milberg
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA USA ,grid.410370.10000 0004 4657 1992Geriatric Research Educational and Clinical Center and Translational Research Center for TBI and Stress Disorders, VA Boston Health Care System, Boston, MA USA
| | - Jasmeet P. Hayes
- grid.410370.10000 0004 4657 1992National Center for PTSD, VA Boston Healthcare System, Boston, MA USA ,grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.261331.40000 0001 2285 7943Department of Psychology and Chronic Brain Injury Program, The Ohio State University, Columbus, OH USA
| | - Mieke Verfaellie
- grid.475010.70000 0004 0367 5222Department of Psychiatry, Boston University School of Medicine, Boston, MA USA ,grid.475010.70000 0004 0367 5222Memory Disorders Research Center, VA Boston Healthcare System and Boston University School of Medicine, Boston, MA USA
| | | |
Collapse
|
85
|
Abstract
Understanding mechanisms underlying learning and memory is crucial in view of tackling cognitive decline occurring during aging or following neurological disorders. The cerebellum offers an ideal system to achieve this goal because of the well-characterized forms of motor learning that it controls. It is so far unclear whether cerebellar memory processes depend on changes in perineuronal nets (PNNs). PNNs are assemblies of extracellular matrix molecules around neurons, which regulate neural plasticity. Here we demonstrate that during eyeblink conditioning (EBC), which is a form of cerebellar motor learning, PNNs in the mouse deep cerebellar nuclei are dynamically modulated, and PNN changes are essential for the formation and storage of EBC memories. Together, these results unveil an important mechanism controlling motor associative memories. Perineuronal nets (PNNs) are assemblies of extracellular matrix molecules, which surround the cell body and dendrites of many types of neuron and regulate neural plasticity. PNNs are prominently expressed around neurons of the deep cerebellar nuclei (DCN), but their role in adult cerebellar plasticity and behavior is far from clear. Here we show that PNNs in the mouse DCN are diminished during eyeblink conditioning (EBC), a form of associative motor learning that depends on DCN plasticity. When memories are fully acquired, PNNs are restored. Enzymatic digestion of PNNs in the DCN improves EBC learning, but intact PNNs are necessary for memory retention. At the structural level, PNN removal induces significant synaptic rearrangements in vivo, resulting in increased inhibition of DCN baseline activity in awake behaving mice. Together, these results demonstrate that PNNs are critical players in the regulation of cerebellar circuitry and function.
Collapse
|
86
|
Tran AP, Warren PM, Silver J. Regulation of autophagy by inhibitory CSPG interactions with receptor PTPσ and its impact on plasticity and regeneration after spinal cord injury. Exp Neurol 2020; 328:113276. [PMID: 32145250 DOI: 10.1016/j.expneurol.2020.113276] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022]
Abstract
Chondroitin sulfate proteoglycans (CSPGs), extracellular matrix molecules that increase dramatically following a variety of CNS injuries or diseases, have long been known for their potent capacity to curtail cell migrations as well as axon regeneration and sprouting. The inhibition can be conferred through binding to their major cognate receptor, Protein Tyrosine Phosphatase Sigma (PTPσ). However, the precise mechanisms downstream of receptor binding that mediate growth inhibition have remained elusive. Recently, CSPGs/PTPσ interactions were found to regulate autophagic flux at the axon growth cone by dampening the autophagosome-lysosomal fusion step. Because of the intense interest in autophagic phenomena in the regulation of a wide variety of critical cellular functions, we summarize here what is currently known about dysregulation of autophagy following spinal cord injury, and highlight this critical new mechanism underlying axon regeneration failure. Furthermore, we review how CSPGs/PTPσ interactions influence plasticity through autophagic regulation and how PTPσ serves as a switch to execute either axon outgrowth or synaptogenesis. This has exciting implications for the role CSPGs play not only in axon regeneration failure after spinal cord injury, but also in neurodegenerative diseases where, again, inhibitory CSPGs are upregulated.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Seattle Children's Hospital Research Institute, Integrative Center for Brain Research, Seattle, Washington, USA
| | - Philippa Mary Warren
- King's College London, Regeneration Group, The Wolfson Centre for Age-Related Diseases, Guy's Campus, London Bridge, London, UK
| | - Jerry Silver
- Case Western Reserve University, School of Medicine, Department of Neurosciences, Cleveland, OH, USA.
| |
Collapse
|
87
|
Abstract
Anxiety and fear-related disorders are common and disabling, and they significantly increase risk for suicide and other causes of morbidity and mortality. However, there is tremendous potential for translational neuroscience to advance our understanding of these disorders, leading to novel and powerful interventions and even to preventing their initial development. This overview examines the general circuits and processes thought to underlie fear and anxiety, along with the promise of translational research. It then examines some of the data-driven "next-generation" approaches that are needed for discovery and understanding but that do not always fit neatly into established models. From one perspective, these disorders offer among the most tractable problems in psychiatry, with a great deal of accumulated understanding, across species, of neurocircuit, behavioral, and, increasingly, genetic mechanisms, of how dysregulation of fear and threat processes contributes to anxiety-related disorders. One example is the progressively sophisticated understanding of how extinction underlies the exposure therapy component of cognitive-behavioral therapy approaches, which are ubiquitously used across anxiety and fear-related disorders. However, it is also critical to examine gaps in our understanding between reasonably well-replicated examples of successful translation, areas of significant deficits in knowledge, and the role of large-scale data-driven approaches in future progress and discovery. Although a tremendous amount of progress is still needed, translational approaches to understanding, treating, and even preventing anxiety and fear-related disorders offer great opportunities for successfully bridging neuroscience discovery to clinical practice.
Collapse
|
88
|
Souter L, Kwok JCF. Visualization of Perineuronal Nets in Central Nervous System Tissue Sections. Methods Mol Biol 2020; 2043:251-260. [PMID: 31463917 DOI: 10.1007/978-1-4939-9698-8_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The perineuronal net (PNN) is a specialized extracellular matrix structure that surrounds subpopulations of neurons in the central nervous system (CNS). The appearance of PNNs on the cell surface marks the closure of the critical period during development and has been observed to reduce synaptic plasticity. Perineuronal nets comprise hyaluronan, chondroitin sulfate proteoglycans (CSPGs), link proteins, tenascin-R, and other components, some of which are substrates for a disintegrin-like and metalloprotease domain with thrombospondin type 1 motifs (ADAMTS) proteases. There is a high heterogeneity of PNNs in the CNS. Depending on which part of the CNS is studied, the PNNs may be observed surrounding the soma, or both the soma and proximal dendrites. The most robust marker for PNN is a lectin called Wisteria floribunda agglutinin. Here, we describe a method for preparing tissue for visualization of PNNs in CNS.
Collapse
Affiliation(s)
- Luke Souter
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK.,School of Mechanical Engineering, Faculty of Engineering,, University of Leeds, Leeds, UK
| | - Jessica C F Kwok
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK. .,Institute of Experimental Medicine, Czech Academy of Science, Prague 4, Czech Republic.
| |
Collapse
|
89
|
Perineuronal nets protect long-term memory by limiting activity-dependent inhibition from parvalbumin interneurons. Proc Natl Acad Sci U S A 2019; 116:27063-27073. [PMID: 31843906 DOI: 10.1073/pnas.1902680116] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Perineuronal nets (PNNs), a complex of extracellular matrix molecules that mostly surround GABAergic neurons in various brain regions, play a critical role in synaptic plasticity. The function and cellular mechanisms of PNNs in memory consolidation and reconsolidation processes are still not well understood. We hypothesized that PNNs protect long-term memory by limiting feedback inhibition from parvalbumin (PV) interneurons to projection neurons. Using behavioral, electrophysiological, and optogenetic approaches, we investigated the role of PNNs in fear memory consolidation and reconsolidation and GABAergic long-term potentiation (LTP). We made the discovery that the formation of PNNs was promoted by memory events in the hippocampus (HP), and we also demonstrated that PNN formation in both the HP and the anterior cingulate cortex (ACC) is essential for memory consolidation and reconsolidation of recent and remote memories. Removal of PNNs resulted in evident LTP impairments, which were rescued by acute application of picrotoxin, a GABAA receptor blocker, indicating that enhanced inhibition was the cause of the LTP impairments induced by PNN removal. Moreover, removal of PNNs switched GABAA receptor-mediated long-term depression to LTP through a presynaptic mechanism. Furthermore, the reduced activity of PV interneurons surrounded by PNNs regulated theta oscillations during fear memory consolidation. Finally, optogenetically suppressing PV interneurons rescued the memory impairment caused by removal of PNNs. Altogether, these results unveil the function of PV interneurons surrounding PNNs in protecting recent and remote contextual memory through the regulation of PV neuron GABA release.
Collapse
|
90
|
Dalmay T, Abs E, Poorthuis RB, Hartung J, Pu DL, Onasch S, Lozano YR, Signoret-Genest J, Tovote P, Gjorgjieva J, Letzkus JJ. A Critical Role for Neocortical Processing of Threat Memory. Neuron 2019; 104:1180-1194.e7. [PMID: 31727549 DOI: 10.1016/j.neuron.2019.09.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/10/2019] [Accepted: 09/17/2019] [Indexed: 01/10/2023]
Abstract
Memory of cues associated with threat is critical for survival and a leading model for elucidating how sensory information is linked to adaptive behavior by learning. Although the brain-wide circuits mediating auditory threat memory have been intensely investigated, it remains unclear whether the auditory cortex is critically involved. Here we use optogenetic activity manipulations in defined cortical areas and output pathways, viral tracing, pathway-specific in vivo 2-photon calcium imaging, and computational analyses of population plasticity to reveal that the auditory cortex is selectively required for conditioning to complex stimuli, whereas the adjacent temporal association cortex controls all forms of auditory threat memory. More temporal areas have a stronger effect on memory and more neurons projecting to the lateral amygdala, which control memory to complex stimuli through a balanced form of population plasticity that selectively supports discrimination of significant sensory stimuli. Thus, neocortical processing plays a critical role in cued threat memory.
Collapse
Affiliation(s)
- Tamas Dalmay
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - Elisabeth Abs
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | | | - Jan Hartung
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - De-Lin Pu
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - Sebastian Onasch
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany
| | - Yave R Lozano
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Jérémy Signoret-Genest
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany; Department of Psychiatry, Center of Mental Health, 97078 Würzburg, Germany
| | - Philip Tovote
- Institute of Clinical Neurobiology, University Hospital Würzburg, 97078 Würzburg, Germany
| | - Julijana Gjorgjieva
- Max Planck Institute for Brain Research, 60438 Frankfurt, Germany; School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | | |
Collapse
|
91
|
Maddox SA, Hartmann J, Ross RA, Ressler KJ. Deconstructing the Gestalt: Mechanisms of Fear, Threat, and Trauma Memory Encoding. Neuron 2019; 102:60-74. [PMID: 30946827 DOI: 10.1016/j.neuron.2019.03.017] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/03/2019] [Accepted: 03/12/2019] [Indexed: 01/14/2023]
Abstract
Threat processing is central to understanding debilitating fear- and trauma-related disorders such as posttraumatic stress disorder (PTSD). Progress has been made in understanding the neural circuits underlying the "engram" of threat or fear memory formation that complements a decades-old appreciation of the neurobiology of fear and threat involving hub structures such as the amygdala. In this review, we examine key recent findings, as well as integrate the importance of hormonal and physiological approaches, to provide a broader perspective of how bodily systems engaged in threat responses may interact with amygdala-based circuits in the encoding and updating of threat-related memory. Understanding how trauma-related memories are encoded and updated throughout the brain and the body will ultimately lead to novel biologically-driven approaches for treatment and prevention.
Collapse
Affiliation(s)
- Stephanie A Maddox
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Jakob Hartmann
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Rachel A Ross
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA
| | - Kerry J Ressler
- Neurobiology of Fear Laboratory, Division of Depression and Anxiety Disorders, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA; Department of Psychiatry, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
92
|
Threat Memory Reminder Under Matrix Metalloproteinase 9 Inhibitor Doxycycline Globally Reduces Subsequent Memory Plasticity. J Neurosci 2019; 39:9424-9434. [PMID: 31615840 PMCID: PMC6867817 DOI: 10.1523/jneurosci.1285-19.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/16/2019] [Accepted: 09/11/2019] [Indexed: 11/21/2022] Open
Abstract
Associative memory can be rendered malleable by a reminder. Blocking the ensuing reconsolidation process is suggested as a therapeutic target for unwanted aversive memories. Matrix metalloproteinase-9 (MMP-9) is required for structural synapse remodeling involved in memory consolidation. Inhibiting MMP-9 with doxycycline is suggested to attenuate human threat conditioning. Here, we investigated whether MMP-9 inhibition also interferes with threat memory reconsolidation. Male and female human participants (N = 78) learned the association between two visual conditioned stimuli (CS+) and a 50% chance of an unconditioned nociceptive stimulus (US), and between CS- and the absence of US. On day 7, one CS+ was reminded without reinforcement 3.5 h after ingesting either 200 mg of doxycycline or placebo. On day 14, retention of CS memory was assessed under extinction by fear-potentiated startle. Contrary to our expectations, we observed a greater CS+/CS- difference in participants who were reminded under doxycycline compared with placebo. Participants who were reminded under placebo showed extinction learning during the retention test, which was not observed in the doxycycline group. There was no difference between the reminded and the nonreminded CS+ in either group. In contrast, during relearning after the retention test, the CS+/CS- difference was more pronounced in the placebo group than in the doxycycline group. To summarize, a single dose of doxycycline before threat memory reminder appeared to have no specific impact on reconsolidation, but to globally impair extinction learning, and threat relearning, beyond drug clearance.SIGNIFICANCE STATEMENT Matrix metalloproteinase-9 inhibition appears to attenuate memory consolidation. It could also be a target for blocking reconsolidation. Here, we test this hypothesis in human threat conditioning. We find that doxycycline has no specific impact on a reminded cue, but confers a global reduction in extinction learning and threat learning beyond the clearance of the drug. This may point toward a more long-lasting impact of doxycycline treatment on memory plasticity.
Collapse
|
93
|
Reinhard SM, Rais M, Afroz S, Hanania Y, Pendi K, Espinoza K, Rosenthal R, Binder DK, Ethell IM, Razak KA. Reduced perineuronal net expression in Fmr1 KO mice auditory cortex and amygdala is linked to impaired fear-associated memory. Neurobiol Learn Mem 2019; 164:107042. [PMID: 31326533 PMCID: PMC7519848 DOI: 10.1016/j.nlm.2019.107042] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/20/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Fragile X Syndrome (FXS) is a leading cause of heritable intellectual disability and autism. Humans with FXS show anxiety, sensory hypersensitivity and impaired learning. The mechanisms of learning impairments can be studied in the mouse model of FXS, the Fmr1 KO mouse, using tone-associated fear memory paradigms. Our previous study reported impaired development of parvalbumin (PV) positive interneurons and perineuronal nets (PNN) in the auditory cortex of Fmr1 KO mice. A recent study suggested PNN dynamics in the auditory cortex following tone-shock association is necessary for fear expression. Together these data suggest that abnormal PNN regulation may underlie tone-fear association learning deficits in Fmr1 KO mice. We tested this hypothesis by quantifying PV and PNN expression in the amygdala, hippocampus and auditory cortex of Fmr1 KO mice following fear conditioning. We found impaired tone-associated memory formation in Fmr1 KO mice. This was paralleled by impaired learning-associated regulation of PNNs in the superficial layers of auditory cortex in Fmr1 KO mice. PV cell density decreased in the auditory cortex in response to fear conditioning in both WT and Fmr1 KO mice. Learning-induced increase of PV expression in the CA3 hippocampus was only observed in WT mice. We also found reduced PNN density in the amygdala and auditory cortex of Fmr1 KO mice in all conditions, as well as reduced PNN intensity in CA2 hippocampus. There was a positive correlation between tone-associated memory and PNN density in the amygdala and auditory cortex, consistent with a tone-association deficit. Altogether our studies suggest a link between impaired PV and PNN regulation within specific regions of the fear conditioning circuit and impaired tone memory formation in Fmr1 KO mice.
Collapse
Affiliation(s)
- Sarah M Reinhard
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Sonia Afroz
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Yasmien Hanania
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Kasim Pendi
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Katherine Espinoza
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Robert Rosenthal
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA
| | - Devin K Binder
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| | - Iryna M Ethell
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| | - Khaleel A Razak
- Psychology Department and Psychology Graduate Program, University of California, Riverside, CA 92521, USA; Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
94
|
Quraishe S, Newman T, Anderson L. Auditory temporal acuity improves with age in the male mouse auditory thalamus: A role for perineuronal nets? J Neurosci Res 2019; 98:1780-1799. [PMID: 31562661 DOI: 10.1002/jnr.24537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/13/2019] [Accepted: 09/15/2019] [Indexed: 11/09/2022]
Abstract
The ability to perceive and interpret environmental sound accurately is conserved across many species and is fundamental for understanding communication via vocalizations. Auditory acuity and temporally controlled neuronal firing underpin this ability. Deterioration in neuronal firing precision likely contributes to poorer hearing performance, yet the role of neural processing by key nuclei in the central auditory pathways is not fully understood. Here, we record from the auditory thalamus (medial geniculate body [MGB]) of young and middle-aged, normally hearing male CBA/Ca mice. We report changes in temporal processing of auditory stimuli, with neurons recorded from ventral and medial MGB subdivisions of older animals more likely to synchronize to rapid temporally varying stimuli. MGB subdivisions also showed increased probability of neuronal firing and shorter response latencies to clicks in older animals. Histological investigation of neuronal extracellular specializations, perineuronal nets (PNNs) and axonal coats, in the MGB identified greater organization of PNNs around MGB neurons and the presence of axonal coats within older animals. This supports the observation that neural responses recorded from ventral and medial MGB of older mice were more likely to synchronize to temporally varying stimuli presented at faster repetition rates than those recorded from young adult animals. These changes are observed in animals with normal hearing thresholds, confirming that neural processing differs between the MGB subdivisions and such processing is associated with age-related changes to PNNs. Understanding these age-related changes and how they occur have important implications for the design of effective therapeutic interventions to improve speech intelligibility into later life.
Collapse
Affiliation(s)
- Shmma Quraishe
- School of Biological Sciences, B85, University of Southampton, Southampton, UK
| | - Tracey Newman
- Clinical and Experimental Sciences, B85, University of Southampton, Southampton, UK
| | | |
Collapse
|
95
|
Alexander GM, Riddick NV, McCann KE, Lustberg D, Moy SS, Dudek SM. Modulation of CA2 neuronal activity increases behavioral responses to fear conditioning in female mice. Neurobiol Learn Mem 2019; 163:107044. [PMID: 31319167 PMCID: PMC6689262 DOI: 10.1016/j.nlm.2019.107044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/13/2019] [Accepted: 07/14/2019] [Indexed: 01/04/2023]
Abstract
Activity of hippocampal pyramidal cells is critical for certain forms of learning and memory, and work from our lab and others has shown that CA2 neuronal activity is required for social cognition and behavior. Silencing of CA2 neurons in mice impairs social memory, and mice lacking Regulator of G-Protein Signaling 14 (RGS14), a protein that is highly enriched in CA2 neurons, learn faster than wild types in the Morris water maze spatial memory test. Although the enhanced spatial learning abilities of the RGS14 KO mice suggest a role for CA2 neurons in at least one hippocampus-dependent behavior, the role of CA2 neurons in fear conditioning, which requires activity of hippocampus, amygdala, and possibly prefrontal cortex is unknown. In this study, we expressed excitatory or inhibitory DREADDs in CA2 neurons and administered CNO before the shock-tone-context pairing. On subsequent days, we measured freezing behavior in the same context but without the tone (contextual fear) or in a new context but in the presence of the tone (cued fear). We found that increasing CA2 neuronal activity with excitatory DREADDs during training resulted in increased freezing during the cued fear tests in males and females. Surprisingly, we found that only females showed increased freezing during the contextual fear memory tests. Using inhibitory DREADDs, we found that inhibiting CA2 neuronal activity during the training phase also resulted in increased freezing in females during the subsequent contextual fear memory test. Finally, we tested fear conditioning in RGS14 KO mice and found that female KO mice had increased freezing on the cued fear memory test. These three separate lines of evidence suggest that CA2 neurons are actively involved in both intra- and extra-hippocampal brain processes and function to influence fear memory. Finally, the intriguing and consistent findings of enhanced fear conditioning only among females is strongly suggestive of a sexual dimorphism in CA2-linked circuits.
Collapse
Affiliation(s)
- Georgia M Alexander
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Natallia V Riddick
- Carolina Institute for Developmental Disabilities and Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, United States
| | - Katharine E McCann
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Daniel Lustberg
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States
| | - Sheryl S Moy
- Carolina Institute for Developmental Disabilities and Department of Psychiatry, University of North Carolina School of Medicine, Chapel Hill, NC 27599, United States
| | - Serena M Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, United States.
| |
Collapse
|
96
|
Reichelt AC, Hare DJ, Bussey TJ, Saksida LM. Perineuronal Nets: Plasticity, Protection, and Therapeutic Potential. Trends Neurosci 2019; 42:458-470. [DOI: 10.1016/j.tins.2019.04.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/16/2019] [Accepted: 04/22/2019] [Indexed: 12/18/2022]
|
97
|
The roles of perineuronal nets and the perinodal extracellular matrix in neuronal function. Nat Rev Neurosci 2019; 20:451-465. [PMID: 31263252 DOI: 10.1038/s41583-019-0196-3] [Citation(s) in RCA: 350] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2019] [Indexed: 01/09/2023]
Abstract
Perineuronal nets (PNNs) are extracellular matrix (ECM) chondroitin sulfate proteoglycan (CSPG)-containing structures that surround the soma and dendrites of various mammalian neuronal cell types. PNNs appear during development around the time that the critical periods for developmental plasticity end and are important for both their onset and closure. A similar structure - the perinodal ECM - surrounds the axonal nodes of Ranvier and appears as myelination is completed, acting as an ion-diffusion barrier that affects axonal conduction speed. Recent work has revealed the importance of PNNs in controlling plasticity in the CNS. Digestion, blocking or removal of PNNs influences functional recovery after a variety of CNS lesions. PNNs have further been shown to be involved in the regulation of memory and have been implicated in a number of psychiatric disorders.
Collapse
|
98
|
Learning Induces Transient Upregulation of Brevican in the Auditory Cortex during Consolidation of Long-Term Memories. J Neurosci 2019; 39:7049-7060. [PMID: 31217331 DOI: 10.1523/jneurosci.2499-18.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 05/14/2019] [Accepted: 05/14/2019] [Indexed: 12/25/2022] Open
Abstract
It is a daily challenge for our brains to establish new memories via learning while providing stable storage of remote memories. In the adult vertebrate brain, bimodal regulation of the extracellular matrix (ECM) may regulate the delicate balance of learning-dependent plasticity and stable memory formation. Here, we trained adult male mice in a cortex-dependent auditory discrimination task and measured the abundance of ECM proteins brevican (BCN) and tenascin-R over the course of acquisition learning, consolidation, and long-term recall in two learning-relevant brain regions; the auditory cortex and hippocampus. Although early training led to a general downregulation of total ECM proteins, successful retrieval correlated with a region-specific and transient upregulation of BCN levels in the auditory cortex. No other parameter such as arousal or stress could account for the transient and region-specific BCN upregulation. This performance-dependent biphasic regulation of the ECM may assist transient plasticity to facilitate initial learning and subsequently promote the long-term consolidation of memory.SIGNIFICANCE STATEMENT The capacity to learn throughout life and at the same time guarantee lifelong storage and remote recall of established memories is a daily challenge. Emerging evidence suggests an important function of the extracellular matrix (ECM), a conglomerate of secreted proteins and polysaccharides in the adult vertebrate brain. We trained mice in an auditory long-term memory task and measured learning-related dynamic changes of the ECM protein brevican. Specifically, in the auditory cortex brevican is downregulated during initial learning and subsequently upregulated in exclusively those animals that have learned the task, suggesting a performance-dependent regulation in the service of memory consolidation and storage. Our data may provide novel therapeutic implications for several neuropsychiatric diseases involving dysregulation of the ECM.
Collapse
|
99
|
Pesarico AP, Bueno-Fernandez C, Guirado R, Gómez-Climent MÁ, Curto Y, Carceller H, Nacher J. Chronic Stress Modulates Interneuronal Plasticity: Effects on PSA-NCAM and Perineuronal Nets in Cortical and Extracortical Regions. Front Cell Neurosci 2019; 13:197. [PMID: 31133813 PMCID: PMC6524695 DOI: 10.3389/fncel.2019.00197] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/18/2019] [Indexed: 01/31/2023] Open
Abstract
Chronic stress has an important impact on the adult brain. However, most of the knowledge on its effects is focused on principal neurons and less on inhibitory neurons. Consequently, recent reports have begun to describe stress-induced alterations in the structure, connectivity and neurochemistry of interneurons. Some of these changes appear to be mediated by certain molecules particularly associated to interneurons, such as the polysialylated form of the neural cell adhesion molecule (PSA-NCAM) and components of the perineuronal nets (PNN), specialized regions of the extracellular matrix. These plasticity-related molecules modulate interneuronal structure and connectivity, particularly of parvalbumin expressing basket interneurons, both during development and adult life. These inhibitory neurons are specially affected after chronic stress and in some stress-related disorders, in which the expression of PSA-NCAM and certain components of PNN are also altered. For these reasons we have decided to study PSA-NCAM, PNN and parvalbumin expressing interneurons after 10 days of chronic restraint stress, a time point in which its behavioral consequences are starting to appear. We have focused initially on the medial prefrontal cortex (mPFC), basolateral amygdala (BLA) and hippocampus, regions affected by stress and stress-related psychiatric diseases, but we have also explored the habenula and the thalamic reticular nucleus (TRN) due to the important presence of PNN and their relationship with certain disorders. PSA-NCAM expression was increased by stress in the stratum lacunosum-moleculare of CA1. Increases in parvalbumin immunoreactive cells were detected in the mPFC and the BLA, but were not accompanied by increases in the number of parvalbumin expressing perisomatic puncta on the somata of principal neurons. The number of PNN was also increased in the mPFC and the habenula, although habenular PNN were not associated to parvalbumin cells. Increased expression of parvalbumin and components of PNN were also detected in the TRN after chronic restraint stress, revealing for the first time substantial effects on this region. Our study shows that, even a short chronic stress protocol, can induce consistent changes in interneuronal plasticity-related molecules in cortical and extracortical regions, which may represent initial responses of inhibitory circuits to counteract the effects of this aversive experience.
Collapse
Affiliation(s)
- Ana Paula Pesarico
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Ramón Guirado
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - María Ángeles Gómez-Climent
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Yasmina Curto
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Hector Carceller
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain.,Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain
| |
Collapse
|
100
|
Abstract
The acquisition of associated signals is commonly seen in life. The integrative storage of these exogenous and endogenous signals is essential for cognition, emotion and behaviors. In terms of basic units of memory traces or engrams, associative memory cells are recruited in the brain during learning, cognition and emotional reactions. The recruitment and refinement of associative memory cells facilitate the retrieval of memory-relevant events and the learning of reorganized unitary signals that have been acquired. The recruitment of associative memory cells is fulfilled by generating mutual synapse innervations among them in coactivated brain regions. Their axons innervate downstream neurons convergently and divergently to recruit secondary associative memory cells. Mutual synapse innervations among associative memory cells confer the integrative storage and reciprocal retrieval of associated signals. Their convergent synapse innervations to secondary associative memory cells endorse integrative cognition. Their divergent innervations to secondary associative memory cells grant multiple applications of associated signals. Associative memory cells in memory traces are defined to be nerve cells that are able to encode multiple learned signals and receive synapse innervations carrying these signals. An impairment in the recruitment and refinement of associative memory cells will lead to the memory deficit associated with neurological diseases and psychological disorders. This review presents a comprehensive diagram for the recruitment and refinement of associative memory cells for memory-relevant events in a lifetime.
Collapse
Affiliation(s)
- Jin-Hui Wang
- College of Life Sciences, Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|