51
|
Svalina MN, Sullivan R, Restrepo D, Huntsman MM. From circuits to behavior: Amygdala dysfunction in fragile X syndrome. Front Integr Neurosci 2023; 17:1128529. [PMID: 36969493 PMCID: PMC10034113 DOI: 10.3389/fnint.2023.1128529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by a repeat expansion mutation in the promotor region of the FMR1 gene resulting in transcriptional silencing and loss of function of fragile X messenger ribonucleoprotein 1 protein (FMRP). FMRP has a well-defined role in the early development of the brain. Thus, loss of the FMRP has well-known consequences for normal cellular and synaptic development leading to a variety of neuropsychiatric disorders including an increased prevalence of amygdala-based disorders. Despite our detailed understanding of the pathophysiology of FXS, the precise cellular and circuit-level underpinnings of amygdala-based disorders is incompletely understood. In this review, we discuss the development of the amygdala, the role of neuromodulation in the critical period plasticity, and recent advances in our understanding of how synaptic and circuit-level changes in the basolateral amygdala contribute to the behavioral manifestations seen in FXS.
Collapse
Affiliation(s)
- Matthew N. Svalina
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Regina Sullivan
- Brain Institute, Nathan Kline Institute, Orangeburg, NY, United States
- Child and Adolescent Psychiatry, Child Study Center, New York University School of Medicine, New York, NY, United States
| | - Diego Restrepo
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Molly M. Huntsman
- Neuroscience Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- *Correspondence: Molly M. Huntsman,
| |
Collapse
|
52
|
Jeon YS, Jeong D, Kweon H, Kim JH, Kim CY, Oh Y, Lee YH, Kim CH, Kim SG, Jeong JW, Kim E, Lee SH. Adolescent Parvalbumin Expression in the Left Orbitofrontal Cortex Shapes Sociability in Female Mice. J Neurosci 2023; 43:1555-1571. [PMID: 36717231 PMCID: PMC10008055 DOI: 10.1523/jneurosci.0918-22.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 12/27/2022] [Accepted: 01/05/2023] [Indexed: 01/31/2023] Open
Abstract
The adolescent social experience is essential for the maturation of the prefrontal cortex in mammalian species. However, it still needs to be determined which cortical circuits mature with such experience and how it shapes adult social behaviors in a sex-specific manner. Here, we examined social-approaching behaviors in male and female mice after postweaning social isolation (PWSI), which deprives social experience during adolescence. We found that the PWSI, particularly isolation during late adolescence, caused an abnormal increase in social approaches (hypersociability) only in female mice. We further found that the PWSI female mice showed reduced parvalbumin (PV) expression in the left orbitofrontal cortex (OFCL). When we measured neural activity in the female OFCL, a substantial number of neurons showed higher activity when mice sniffed other mice (social sniffing) than when they sniffed an object (object sniffing). Interestingly, the PWSI significantly reduced both the number of activated neurons and the activity level during social sniffing in female mice. Similarly, the CRISPR/Cas9-mediated knockdown of PV in the OFCL during late adolescence enhanced sociability and reduced the social sniffing-induced activity in adult female mice via decreased excitability of PV+ neurons and reduced synaptic inhibition in the OFCL Moreover, optogenetic activation of excitatory neurons or optogenetic inhibition of PV+ neurons in the OFCL enhanced sociability in female mice. Our data demonstrate that the adolescent social experience is critical for the maturation of PV+ inhibitory circuits in the OFCL; this maturation shapes female social behavior via enhancing social representation in the OFCL SIGNIFICANCE STATEMENT Adolescent social isolation often changes adult social behaviors in mammals. Yet, we do not fully understand the sex-specific effects of social isolation and the brain areas and circuits that mediate such changes. Here, we found that adolescent social isolation causes three abnormal phenotypes in female but not male mice: hypersociability, decreased PV+ neurons in the left orbitofrontal cortex (OFCL), and decreased socially evoked activity in the OFCL Moreover, parvalbumin (PV) deletion in the OFCL in vivo caused the same phenotypes in female mice by increasing excitation compared with inhibition within the OFCL Our data suggest that adolescent social experience is required for PV maturation in the OFCL, which is critical for evoking OFCL activity that shapes social behaviors in female mice.
Collapse
Affiliation(s)
- Yi-Seon Jeon
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Daun Jeong
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Hanseul Kweon
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| | - Jae-Hyun Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Choong Yeon Kim
- School of Electrical Engineering, KAIST, Daejeon 34141, Korea
| | - Youngbin Oh
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Young-Ho Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Chan Hyuk Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Sang-Gyu Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
| | - Jae-Woong Jeong
- School of Electrical Engineering, KAIST, Daejeon 34141, Korea
| | - Eunjoon Kim
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon 34141, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, KAIST, Daejeon 34141, Korea
| |
Collapse
|
53
|
Biojone C, C Casarotto P, Cannarozzo C, Fred SM, Herrera-Rodríguez R, Lesnikova A, Voipio M, Castrén E. nNOS-induced tyrosine nitration of TRKB impairs BDNF signaling and restrains neuronal plasticity. Prog Neurobiol 2023; 222:102413. [PMID: 36682419 DOI: 10.1016/j.pneurobio.2023.102413] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 12/01/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Nitric oxide (NO) has been long recognized as an important modulator of neural plasticity, but characterization of the molecular mechanisms involved - specially the guanylyl cyclase-independent ones - has been challenging. There is evidence that NO could modify BDNF-TRKB signaling, a key mediator of neuronal plasticity. However, the mechanism underlying the interplay of NO and TRKB remains unclear. Here we show that NO induces nitration of the tyrosine 816 in the TRKB receptor in vivo and in vitro, and that post-translational modification inhibits TRKB phosphorylation and binding of phospholipase Cγ1 (PLCγ1) to this same tyrosine residue. Additionally, nitration triggers clathrin-dependent endocytosis of TRKB through the adaptor protein AP-2 and ubiquitination, thereby increasing translocation of TRKB away from the neuronal surface and directing it towards lysosomal degradation. Accordingly, inhibition of nitric oxide increases TRKB phosphorylation and TRKB-dependent neurite branching in neuronal cultures. In vivo, chronic inhibition of neuronal nitric oxide synthase (nNOS) dramatically reduced TRKB nitration and facilitated TRKB signaling in the visual cortex, and promoted a shift in ocular dominance upon monocular deprivation - an indicator of increased plasticity. Altogether, our data describe and characterize a new molecular brake on plasticity, namely nitration of TRKB receptors.
Collapse
Affiliation(s)
- Caroline Biojone
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland; Aarhus University, Department of Biomedicine, Faculty of Health, and Translational Neuropsychiatry Unit, Department of Clinical Medicine.
| | - Plinio C Casarotto
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Cecilia Cannarozzo
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Senem Merve Fred
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | | | - Angelina Lesnikova
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Mikko Voipio
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Eero Castrén
- Neuroscience Center, HiLife, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland.
| |
Collapse
|
54
|
Woodburn SC, Asrat HS, Flurer JK, Schwierling HC, Bollinger JL, Vollmer LL, Wohleb ES. Depletion of microglial BDNF increases susceptibility to the behavioral and synaptic effects of chronic unpredictable stress. Brain Behav Immun 2023; 109:127-138. [PMID: 36681359 PMCID: PMC10023455 DOI: 10.1016/j.bbi.2023.01.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 12/22/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
In the medial prefrontal cortex (PFC), chronic stress reduces synaptic expression of glutamate receptors, leading to decreased excitatory signaling from layer V pyramidal neurons and working memory deficits. One key element driving these changes is a reduction in brain-derived neurotrophic factor (BDNF) signaling. BDNF is a potent mediator of synaptic growth and deficient BDNF signaling has been linked to stress susceptibility. Prior studies indicated that neurons are the primary source of BDNF, but more recent work suggests that microglia are also an important source of BDNF. Adding to this, our work showed that 14 days of chronic unpredictable stress (CUS) reduced Bdnf transcript in PFC microglia, evincing its relevance in the effects of stress. To explore this further, we utilized transgenic mice with microglia-specific depletion of BDNF (Cx3cr1Cre/+:Bdnffl/fl) and genotype controls (Cx3cr1Cre/+:Bdnf+/+). In the following experiments, mice were exposed to a shortened CUS paradigm (7 days) to determine if microglial Bdnf depletion promotes stress susceptibility. Analyses of PFC microglia revealed that Cx3cr1Cre/+:Bdnffl/fl mice had shifts in phenotypic markers and gene expression. In a separate cohort, synaptoneurosomes were collected from the PFC and western blotting was performed for synaptic markers. These experiments showed that Cx3cr1Cre/+:Bdnffl/fl mice had baseline deficits in GluN2B, and that 7 days of CUS additionally reduced GluN2A levels in Cx3cr1Cre/+:Bdnffl/fl mice, but not genotype controls. Behavioral and cognitive testing showed that this coincided with exacerbated stress effects on temporal object recognition in Cx3cr1Cre/+:Bdnffl/fl mice. These results indicate that microglial BDNF promotes glutamate receptor expression in the PFC. As such, mice with deficient microglial BDNF had increased susceptibility to the behavioral and cognitive consequences of stress.
Collapse
Affiliation(s)
- Samuel C Woodburn
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Helina S Asrat
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - James K Flurer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Hana C Schwierling
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Justin L Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Lauren L Vollmer
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
55
|
Moreno A, Rajagopalan S, Tucker MJ, Lunsford P, Liu RC. Hearing Vocalizations during First Social Experience with Pups Increase Bdnf Transcription in Mouse Auditory Cortex. Neural Plast 2023; 2023:5225952. [PMID: 36845359 PMCID: PMC9946766 DOI: 10.1155/2023/5225952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/30/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
While infant cues are often assumed to innately motivate maternal response, recent research highlights how the neural coding of infant cues is altered through maternal care. Infant vocalizations are important social signals for caregivers, and evidence from mice suggests that experience caring for mouse pups induces inhibitory plasticity in the auditory cortex (AC), though the molecular mediators for such AC plasticity during the initial pup experience are not well delineated. Here, we used the maternal mouse communication model to explore whether transcription in AC of a specific, inhibition-linked, memory-associated gene, brain-derived neurotrophic factor (Bdnf) changes due to the very first pup caring experience hearing vocalizations, while controlling for the systemic influence of the hormone estrogen. Ovariectomized and estradiol or blank-implanted virgin female mice hearing pup calls with pups present had significantly higher AC exon IV Bdnf mRNA compared to females without pups present, suggesting that the social context of vocalizations induces immediate molecular changes at the site of auditory cortical processing. E2 influenced the rate of maternal behavior but did not significantly affect Bdnf mRNA transcription in the AC. To our knowledge, this is the first time Bdnf has been associated with processing social vocalizations in the AC, and our results suggest that it is a potential molecular component responsible for enhancing future recognition of infant cues by contributing to AC plasticity.
Collapse
Affiliation(s)
- Amielle Moreno
- Neuroscience Graduate Program, Emory University, Atlanta, Georgia 30332, USA
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| | | | - Matthew J. Tucker
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
| | - Parker Lunsford
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
- College of Science Undergraduate Neuroscience Program, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Robert C. Liu
- Department of Biology, Emory University, Atlanta, Georgia 30322, USA
- Center for Translational Social Neuroscience, Emory University, Atlanta, Georgia 30322, USA
| |
Collapse
|
56
|
Rodriguez LA, Kim SH, Page SC, Nguyen CV, Pattie EA, Hallock HL, Valerino J, Maynard KR, Jaffe AE, Martinowich K. The basolateral amygdala to lateral septum circuit is critical for regulating social novelty in mice. Neuropsychopharmacology 2023; 48:529-539. [PMID: 36369482 PMCID: PMC9852457 DOI: 10.1038/s41386-022-01487-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 10/07/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022]
Abstract
The lateral septum (LS) is a basal forebrain GABAergic region that is implicated in social novelty. However, the neural circuits and cell signaling pathways that converge on the LS to mediate social behaviors aren't well understood. Multiple lines of evidence suggest that signaling of brain-derived neurotrophic factor (BDNF) through its receptor TrkB plays important roles in social behavior. BDNF is not locally produced in LS, but we demonstrate that nearly all LS GABAergic neurons express TrkB. Local TrkB knock-down in LS neurons decreased social novelty recognition and reduced recruitment of neural activity in LS neurons in response to social novelty. Since BDNF is not synthesized in LS, we investigated which inputs to LS could serve as potential BDNF sources for controlling social novelty recognition. We demonstrate that selectively ablating inputs to LS from the basolateral amygdala (BLA), but not from ventral CA1 (vCA1), impairs social novelty recognition. Moreover, depleting BDNF selectively in BLA-LS projection neurons phenocopied the decrease in social novelty recognition caused by either local LS TrkB knockdown or ablation of BLA-LS inputs. These data support the hypothesis that BLA-LS projection neurons serve as a critical source of BDNF for activating TrkB signaling in LS neurons to control social novelty recognition.
Collapse
Affiliation(s)
- Lionel A Rodriguez
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Sun-Hong Kim
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Stephanie C Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Claudia V Nguyen
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Elizabeth A Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Henry L Hallock
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Jessica Valerino
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Andrew E Jaffe
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Genetic Medicine, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Keri Martinowich
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- The Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, USA.
| |
Collapse
|
57
|
Liu S, Li Y. Deciphering functional roles of synaptic plasticity and intrinsic neural firing in developing mouse visual cortex layer IV microcircuit. J Comput Neurosci 2023; 51:23-42. [PMID: 35737171 DOI: 10.1007/s10827-022-00823-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/16/2022] [Accepted: 05/31/2022] [Indexed: 01/18/2023]
Abstract
Between the onset of the critical period of mouse primary visual cortex and eye opening at postnatal day 14 is a complex process and that is vital for the cognitive function of vision. The onset of the critical period of mouse primary visual cortex involves changes of the intrinsic firing property of each neuron and short term plasticity of synapses. In order to investigate the functional role of each factor in regulating the circuit firing activity during the critical period plasticity, we adopted the Markram's model for short term plasticity and Wilson's model for intrinsic neuron firing activity, and construct a microcircuit for mouse visual cortex layer IV based on the connection probabilities from experimental results. Our results indicate that, during CP development, the most critical factors that regulate the firing pattern of microcircuit is the short term plasticity of the synapse from PC to PV and SST interneurons, which upregulates the PV interneuron firing and produces new balance between excitation and inhibition; the intrinsic firing activity of PC and PV during development downregulates the firing frequency of the circuits. In addition, we have investigated the function of feedforward excitatory thalamic-cortical projection to PC and PV interneuron during CP, and found that neural firing activity largely depends on the TC input and the results are similar to the local circuit with minor differences. We conclude that the short term plasticity development during critical period plays a crucial role in regulating the circuit behavior.
Collapse
Affiliation(s)
- Sanwu Liu
- School of Systems Science, Beijing Normal University, Beijing, China
| | - Yinyun Li
- School of Systems Science, Beijing Normal University, Beijing, China.
| |
Collapse
|
58
|
Mowery TM, Garraghty PE. Adult neuroplasticity employs developmental mechanisms. Front Syst Neurosci 2023; 16:1086680. [PMID: 36762289 PMCID: PMC9904365 DOI: 10.3389/fnsys.2022.1086680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 12/26/2022] [Indexed: 01/26/2023] Open
Abstract
Although neural plasticity is now widely studied, there was a time when the idea of adult plasticity was antithetical to the mainstream. The essential stumbling block arose from the seminal experiments of Hubel and Wiesel who presented convincing evidence that there existed a critical period for plasticity during development after which the brain lost its ability to change in accordance to shifts in sensory input. Despite the zeitgeist that mature brain is relatively immutable to change, there were a number of examples of adult neural plasticity emerging in the scientific literature. Interestingly, some of the earliest of these studies involved visual plasticity in the adult cat. Even earlier, there were reports of what appeared to be functional reorganization in adult rat somatosensory thalamus after dorsal column lesions, a finding that was confirmed and extended with additional experimentation. To demonstrate that these findings reflected more than a response to central injury, and to gain greater control of the extent of the sensory loss, peripheral nerve injuries were used that eliminated ascending sensory information while leaving central pathways intact. Merzenich, Kaas, and colleagues used peripheral nerve transections to reveal unambiguous reorganization in primate somatosensory cortex. Moreover, these same researchers showed that this plasticity proceeded in no less than two stages, one immediate, and one more protracted. These findings were confirmed and extended to more expansive cortical deprivations, and further extended to the thalamus and brainstem. There then began a series of experiments to reveal the physiological, morphological and neurochemical mechanisms that permitted this plasticity. Ultimately, Mowery and colleagues conducted a series of experiments that carefully tracked the levels of expression of several subunits of glutamate (AMPA and NMDA) and GABA (GABAA and GABAB) receptor complexes in primate somatosensory cortex at several time points after peripheral nerve injury. These receptor subunit mapping experiments revealed that membrane expression levels came to reflect those seen in early phases of critical period development. This suggested that under conditions of prolonged sensory deprivation the adult cells were returning to critical period like plastic states, i.e., developmental recapitulation. Here we outline the heuristics that drive this phenomenon.
Collapse
Affiliation(s)
- Todd M. Mowery
- Department of Otolaryngology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Preston E. Garraghty
- Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
59
|
Shaffery JP, Marks GA. Howard P. Roffwarg: sleep pioneer, legend, and ontogenetic hypothesis author. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2023; 4:zpad004. [PMID: 37193292 PMCID: PMC10108642 DOI: 10.1093/sleepadvances/zpad004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/15/2022] [Indexed: 05/18/2023]
Abstract
Narrated in this article are accounts of the many contributions Howard P. Roffwarg, MD, made to the field of sleep research and sleep medicine across his entire professional career as a student, a mentor, a leader in the Sleep Research Society, a sleep medicine clinician, and a scientist who performed experimental investigations in humans and animals. Dr Roffwarg was the originator of what is known as the "Ontogenetic Hypothesis" of sleep. His research over many years on physiology has contributed greatly to much of the experimental support substantiating a role for rapid eye-movement sleep (REMS) in the early development of the brain. Though much is still unknown, the Ontogenetic Hypothesis, still to this day, inspires many neuroscientists in their investigations. These studies have demonstrated roles for both REMS and NREMS in development as well as on brain function throughout his life span. Dr Howard P. Roffwarg, is one of the legends in the field of sleep research.
Collapse
Affiliation(s)
- James P Shaffery
- Department of Psychiatry and Human Behavior, University of Mississippi, Jackson, MS 39216-4505, USA
| | - Gerald A Marks
- Department of Psychiatry, University of Texas Southwestern, Dallas, TX 75390, USA
| |
Collapse
|
60
|
Comparison of the effect of glyphosate and glyphosate-based herbicide on hippocampal neurogenesis after developmental exposure in rats. Toxicology 2023; 483:153369. [PMID: 36332718 DOI: 10.1016/j.tox.2022.153369] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Increasing evidence indicates that glyphosate (GlyP)-based herbicides (GBHs) induce developmental neurotoxicity. The present study investigated the developmental exposure effect of GlyP and GBH on hippocampal neurogenesis in rats. Dams were treated from gestational day 6 to day 21 post-delivery on weaning with a diet containing 1.5% or 3.0% GlyP or drinking water with 1.0% GBH (containing 0.36% GlyP). Dams in the 1.5%-GlyP, 3.0%-GlyP, and GBH groups received 1.04, 2.16, and 0.25 g GlyP/kg body weight (BW)/day during gestation, and 2.27, 4.65, and 0.58 g GlyP/kg BW/day during lactation, respectively. On weaning, 3.0% GlyP- and GBH-exposed offspring decreased the BW, and the latter also decreased the brain weight. Both compounds suppressed neural progenitor cell proliferation in the neurogenic niche, and GlyP-exposed offspring showed a decreased number of TUBB3+ immature granule cells. In contrast, both compounds increased the number of ARC+ granule cells, suggesting increased synaptic plasticity. Both compounds downregulated antioxidant genes (Cat and Sod2) in the dentate gyrus, suggestive of increased sensitivity to oxidative stress, which might be related to the suppression of neurogenesis. At the adult age, GBH alone sustained decreases in body and brain weights. Both compounds increased hippocampal malondialdehyde levels and upregulated Cat in the dentate gyrus, suggesting induction of oxidative stress. Both compounds upregulated Casp9, and GBH increased neural progenitor cell apoptosis, suggesting disruption of neurogenesis related to oxidative stress. GBH increased the number of COX2+ granule cells, and both compounds upregulated Arc, suggesting increased synaptic plasticity. These results suggest that GlyP and GBH might cause similar effects on disruption of neurogenesis accompanying compensatory responses and induction of oxidative stress responses through the adult age in the hippocampus. However, effects on adult age were more evident with GBH, suggesting that the surfactants contained in GBH might have contributed to the enhanced neurotoxicity of GlyP, similar to the enhanced general toxicity.
Collapse
|
61
|
Takahashi H, Yamamoto T, Tsuboi A. Molecular mechanisms underlying activity-dependent ischemic tolerance in the brain. Neurosci Res 2023; 186:3-9. [PMID: 36244569 DOI: 10.1016/j.neures.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/05/2022] [Accepted: 10/10/2022] [Indexed: 11/06/2022]
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. The inhibition of cerebral blood flow triggers intertwined pathological events, resulting in cell death and loss of brain function. Interestingly, animals pre-exposed to short-term ischemia can tolerate subsequent severe ischemia. This phenomenon is called ischemic tolerance and is also triggered by other noxious stimuli. However, whether short-term exposure to non-noxious stimuli can induce ischemic tolerance remains unknown. Recently, we found that pre-exposing mice to an enriched environment for 40 min is sufficient to facilitate cell survival after a subsequent stroke. The neuroprotective process depends on the neuronal activity soon before stroke, of which the activity-dependent transcription factor Npas4 is essential. Excessive Ca2+ influx triggers Npas4 expression in ischemic neurons, leading to the activation of neuroprotective programs. Pre-induction of Npas4 in the normal brain effectively supports cell survival after stroke. Furthermore, our study revealed that Npas4 regulates L-type voltage-gated Ca2+ channels through expression of the small Ras-like GTPase Gem in ischemic neurons. Ischemic tolerance is a good model for understanding how to promote neuroprotective mechanisms in the normal and injured brain. Here, we highlight activity-dependent ischemic tolerance and discuss its role in promoting neuroprotection against stroke.
Collapse
Affiliation(s)
- Hiroo Takahashi
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan.
| | - Tohru Yamamoto
- Department of Molecular Neurobiology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan
| | - Akio Tsuboi
- Dynamic Brain Network Laboratory, Graduate School of Frontier Biosciences, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
62
|
An Early Enriched Experience Drives an Activated Microglial Profile at Site of Corrective Neuroplasticity in Ten-m3 Knock-Out Mice. eNeuro 2023; 10:ENEURO.0162-22.2022. [PMID: 36635245 PMCID: PMC9831145 DOI: 10.1523/eneuro.0162-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 12/15/2022] Open
Abstract
Environmental enrichment (EE) is beneficial for brain development and function, but our understanding of its capacity to drive circuit repair, the underlying mechanisms, and how this might vary with age remains limited. Ten-m3 knock-out (KO) mice exhibit a dramatic and stereotyped mistargeting of ipsilateral retinal inputs to the thalamus, resulting in visual deficits. We have recently shown a previously unexpected capacity for EE during early postnatal life (from birth for six weeks) to drive the partial elimination of miswired axonal projections, along with a recovery of visually mediated behavior, but the timeline of this repair was unclear. Here, we reveal that with just 3.5 weeks of EE from birth, Ten-m3 KOs exhibit a partial behavioral rescue, accompanied by pruning of the most profoundly miswired retinogeniculate terminals. Analysis suggests that the pruning is underway at this time point, providing an ideal opportunity to probe potential mechanisms. With the shorter EE-period, we found a localized increase in microglial density and activation profile within the identified geniculate region where corrective pruning was observed. No comparable response to EE was found in age-matched wild-type (WT) mice. These findings identify microglia as a potential mechanistic link through which EE drives the elimination of miswired neural circuits during early postnatal development. Activity driven, atypical recruitment of microglia to prune aberrant connectivity and restore function may have important therapeutic implications for neurodevelopmental disorders such as autistic spectrum disorder.
Collapse
|
63
|
Egbenya DL, Hussain S, Lai YC, Anderson AE, Davanger S. Synapse-specific changes in Arc and BDNF in rat hippocampus following chronic temporal lobe epilepsy. Neurosci Res 2022; 191:1-12. [PMID: 36535366 DOI: 10.1016/j.neures.2022.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 12/06/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Expression of immediate early genes (IEGs) in the brain is important for synaptic plasticity, and probably also in neurodegenerative conditions. To understand the cellular mechanisms of the underlying neuropathophysiological processes in epilepsy, we need to pinpoint changes in concentration of synaptic plasticity-related proteins at subsynaptic levels. In this study, we examined changes in synaptic expression of Activity-regulated cytoskeleton-associated (Arc) and Brai Derived Neurotrophic Factor (BDNF) in a rat model of kainate-induced temporal lobe epilepsy (TLE). Western blotting showed reduced concentrations of Arc and increased concentrations of BDNF in hippocampal synaptosomes in chronic TLE rats. Then, using quantitative electron microscopy, we found corresponding changes in subsynaptic regions in the hippocampus. Specifically, we detected significant reductions in the concentrations of Arc in the presynaptic terminal of Schaffer collateral glutamatergic synapses in the stratum radiatum of the CA1 area in TLE, as well as in their adjacent postsynaptic spines. In CA3, there was a significant reduction of Arc only in the presynaptic terminal cytoplasm. Conversely, in CA3, there was a significant increase in the expression of BDNF in the presynaptic terminal, but not in the postsynaptic spine. Significant increase in BDNF concentration in the CA1 postsynaptic density was also obtained. We hypothesize that the observed changes in Arc and BDNF may contribute to both cognitive impairment and increased excitotoxic vulnerability in chronic epilepsy.
Collapse
Affiliation(s)
- Daniel L Egbenya
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Physiology, School of Medical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Suleman Hussain
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway; Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.
| | - Yi-Chen Lai
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Anne E Anderson
- Jan and Dan Duncan Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Svend Davanger
- Laboratory for Synaptic Plasticity, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
64
|
5-HT-dependent synaptic plasticity of the prefrontal cortex in postnatal development. Sci Rep 2022; 12:21015. [PMID: 36470912 PMCID: PMC9723183 DOI: 10.1038/s41598-022-23767-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022] Open
Abstract
Important functions of the prefrontal cortex (PFC) are established during early life, when neurons exhibit enhanced synaptic plasticity and synaptogenesis. This developmental stage drives the organization of cortical connectivity, responsible for establishing behavioral patterns. Serotonin (5-HT) emerges among the most significant factors that modulate brain activity during postnatal development. In the PFC, activated 5-HT receptors modify neuronal excitability and interact with intracellular signaling involved in synaptic modifications, thus suggesting that 5-HT might participate in early postnatal plasticity. To test this hypothesis, we employed intracellular electrophysiological recordings of PFC layer 5 neurons to study the modulatory effects of 5-HT on plasticity induced by theta-burst stimulation (TBS) in two postnatal periods of rats. Our results indicate that 5-HT is essential for TBS to result in synaptic changes during the third postnatal week, but not later. TBS coupled with 5-HT2A or 5-HT1A and 5-HT7 receptors stimulation leads to long-term depression (LTD). On the other hand, TBS and synergic activation of 5-HT1A, 5-HT2A, and 5-HT7 receptors lead to long-term potentiation (LTP). Finally, we also show that 5-HT dependent synaptic plasticity of the PFC is impaired in animals that are exposed to early-life chronic stress.
Collapse
|
65
|
Miranda JM, Cruz E, Bessières B, Alberini CM. Hippocampal parvalbumin interneurons play a critical role in memory development. Cell Rep 2022; 41:111643. [PMID: 36384113 PMCID: PMC9737056 DOI: 10.1016/j.celrep.2022.111643] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/16/2022] [Accepted: 10/19/2022] [Indexed: 11/18/2022] Open
Abstract
Episodic memories formed in early childhood rapidly decay, but their latent traces remain stored long term. These memories require the dorsal hippocampus (dHPC) and seem to undergo a developmental critical period. It remains to be determined whether the maturation of parvalbumin interneurons (PVIs), a major mechanism of critical periods, contributes to memory development. Here, we show that episodic infantile learning significantly increases the levels of parvalbumin in the dHPC 48 h after training. Chemogenetic inhibition of PVIs before learning indicated that these neurons are required for infantile memory formation. A bilateral dHPC injection of the γ-aminobutyric acid type A receptor agonist diazepam after training elicited long-term memory expression in infant rats, although direct PVI chemogenetic activation had no effect. Finally, PVI activity was required for brain-derived neurotrophic factor (BDNF)-dependent maturation of memory competence, i.e., adult-like long-term memory expression. Thus, dHPC PVIs are critical for the formation of infantile memories and for memory development.
Collapse
Affiliation(s)
- Janelle M Miranda
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Emmanuel Cruz
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Benjamin Bessières
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA
| | - Cristina M Alberini
- Center for Neural Science, New York University, 4 Washington Place, New York, NY 10003, USA.
| |
Collapse
|
66
|
Cutuli D, Sampedro-Piquero P. BDNF and its Role in the Alcohol Abuse Initiated During Early Adolescence: Evidence from Preclinical and Clinical Studies. Curr Neuropharmacol 2022; 20:2202-2220. [PMID: 35748555 PMCID: PMC9886842 DOI: 10.2174/1570159x20666220624111855] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/22/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a crucial brain signaling protein that is integral to many signaling pathways. This neurotrophin has shown to be highly involved in brain plastic processes such as neurogenesis, synaptic plasticity, axonal growth, and neurotransmission, among others. In the first part of this review, we revise the role of BDNF in different neuroplastic processes within the central nervous system. On the other hand, its deficiency in key neural circuits is associated with the development of psychiatric disorders, including alcohol abuse disorder. Many people begin to drink alcohol during adolescence, and it seems that changes in BDNF are evident after the adolescent regularly consumes alcohol. Therefore, the second part of this manuscript addresses the involvement of BDNF during adolescent brain maturation and how this process can be negatively affected by alcohol abuse. Finally, we propose different BNDF enhancers, both behavioral and pharmacological, which should be considered in the treatment of problematic alcohol consumption initiated during the adolescence.
Collapse
Affiliation(s)
- Debora Cutuli
- Department of Psychology, Medicine and Psychology Faculty, University Sapienza of Rome, Rome, Italy; ,I.R.C.C.S. Fondazione Santa Lucia, Laboratorio di Neurofisiologia Sperimentale e del Comportamento, Via del Fosso di Fiorano 64, 00143 Roma, Italy; ,Address correspondence to these authors at the Department of Biological and Health Psychology, Psychology Faculty, Autonomous University of Madrid, Madrid, Spain, Spain and Cutuli, D. at Fondazione Santa Lucia. Laboratorio di Neurofisiologia Sperimentale e del Comportamento. Via del Fosso di Fiorano 64, 00143 Roma, Italy; E-mails: ;
| | - Piquero Sampedro-Piquero
- Department of Biological and Health Psychology, Psychology Faculty, Autonomous University of Madrid, Madrid, Spain,Address correspondence to these authors at the Department of Biological and Health Psychology, Psychology Faculty, Autonomous University of Madrid, Madrid, Spain, Spain and Cutuli, D. at Fondazione Santa Lucia. Laboratorio di Neurofisiologia Sperimentale e del Comportamento. Via del Fosso di Fiorano 64, 00143 Roma, Italy; E-mails: ;
| |
Collapse
|
67
|
Ursini F, Bosello Travain V, Cozza G, Miotto G, Roveri A, Toppo S, Maiorino M. A white paper on Phospholipid Hydroperoxide Glutathione Peroxidase (GPx4) forty years later. Free Radic Biol Med 2022; 188:117-133. [PMID: 35718302 DOI: 10.1016/j.freeradbiomed.2022.06.227] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/25/2022]
Abstract
The purification of a protein inhibiting lipid peroxidation led to the discovery of the selenoperoxidase GPx4 forty years ago. Thus, the evidence of the enzymatic activity was reached after identifying the biological effect and unambiguously defined the relationship between the biological function and the enzymatic activity. In the syllogism where GPx4 inhibits lipid peroxidation and its inhibition is lethal, cell death is operated by lipid peroxidation. Based on this rationale, this form of cell death emerged as regulated iron-enforced oxygen toxicity and was named ferroptosis in 2012. In the last decades, we learned that reduction of lipid hydroperoxides is indispensable and, in cooperation with prooxidant systems, controls the critical steady state of lipid peroxidation. This concept defined the GPx4 reaction as both the target for possible anti-cancer therapy and if insufficient, as cause of degenerative diseases. We know the reaction mechanism, but the details of the interaction at the membrane cytosol interface are still poorly defined. We know the gene structure, but the knowledge about expression control is still limited. The same holds true for post-transcriptional modifications. Reverse genetics indicate that GPx4 has a role in inflammation, immunity, and differentiation, but the observations emerging from these studies need a more specifically addressed biochemical evidence. Finally, the role of GPx4 in spermatogenesis disclosed an area unconnected to lipid peroxidation. In its mitochondrial and nuclear form, the peroxidase catalyzes the oxidation of protein thiols in two specific aspects of sperm maturation: stabilization of the mid-piece and chromatin compaction. Thus, although available evidence converges to the notion that GPx4 activity is vital due to the inhibition of lipid peroxidation, it is reasonable to foresee other unknown aspects of the GPx4 reaction to be disclosed.
Collapse
Affiliation(s)
- Fulvio Ursini
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy
| | | | - Giorgio Cozza
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy
| | - Giovanni Miotto
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy
| | - Antonella Roveri
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy
| | - Stefano Toppo
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, Viale G. Colombo, 3, University of Padova, 35121, Padova, Italy.
| |
Collapse
|
68
|
Chang JT, Fitzpatrick D. Development of visual response selectivity in cortical GABAergic interneurons. Nat Commun 2022; 13:3791. [PMID: 35778379 PMCID: PMC9249896 DOI: 10.1038/s41467-022-31284-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 06/09/2022] [Indexed: 11/09/2022] Open
Abstract
The visual cortex of carnivores and primates displays a high degree of modular network organization characterized by local clustering and structured long-range correlations of activity and functional properties. Excitatory networks display modular organization before the onset of sensory experience, but the developmental timeline for modular networks of GABAergic interneurons remains under-explored. Using in vivo calcium imaging of the ferret visual cortex, we find evidence that before visual experience, interneurons display weak orientation tuning and widespread, correlated activity in response to visual stimuli. Robust modular organization and orientation tuning are evident with as little as one week of visual experience. Furthermore, we find that the maturation of orientation tuning requires visual experience, while the reduction in widespread, correlated network activity does not. Thus, the maturation of inhibitory cortical networks occurs in a delayed, parallel process relative to excitatory neurons. Excitatory neurons in the carnivore and primate visual cortex display orientation selectivity arranged in a modular fashion before the onset of visual experience, but the developmental timeline for visual response selectivity of inhibitory neurons is unknown. Using in vivo calcium imaging in ferret visual cortex, the authors show that experience-dependent emergence of orientation selectivity develops in a modular fashion for inhibitory interneurons over the first week of visual experience.
Collapse
Affiliation(s)
- Jeremy T Chang
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| | - David Fitzpatrick
- Department of Functional Architecture and Development of Cerebral Cortex, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| |
Collapse
|
69
|
Ueno H, Takahashi Y, Murakami S, Wani K, Matsumoto Y, Okamoto M, Ishihara T. Fingolimod increases parvalbumin-positive neurons in adult mice. IBRO Neurosci Rep 2022; 13:96-106. [PMID: 36590091 PMCID: PMC9795291 DOI: 10.1016/j.ibneur.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years, it has been shown that central nervous system agents, such as antidepressants and antiepileptic drugs, reopen a critical period in mature animals. Fingolimod, which is used for the treatment of multiple sclerosis, also restores neuroplasticity. In this study, we investigated the effects of parvalbumin (PV)-positive neurons and perineuronal nets (PNN) on fingolimod administration with respect to neuroplasticity. Fingolimod was chronically administered intraperitoneally to mature mice. PV-positive neurons and PNN in the hippocampus, prefrontal cortex, and somatosensory cortex were analyzed. An increase in PV-positive neurons was observed in the hippocampus, prefrontal cortex, and somatosensory cortex of the fingolimod-treated mice. An increase in Wisteria floribunda agglutinin-positive PNN was confirmed in mice treated with fingolimod in the somatosensory cortex only. Fingolimod increased the density of PV-positive neurons in the brains of mature mice. The results indicate that fingolimod may change the critical period in mature animals.
Collapse
Key Words
- CNS, central nervous system
- ECM, extracellular matrix
- Fingolimod
- GAD67, anti-glutamic acid decarboxylase
- GFAP, glial fibrillary acidic protein
- Hippocampus
- IL, infralimbic cortex
- NIH, National Institutes of Health, PBS, phosphate-buffered saline
- PL, prelimbic cortex
- PNN, perineuronal net
- PV neurons, parvalbumin-expressing interneurons
- Parvalbumin
- Perineuronal nets
- Prefrontal cortex
- Somatosensory cortex
- WFA, Wisteria floribunda agglutinin
- dAC, dorsal anterior cingulate cortex
Collapse
Affiliation(s)
- Hiroshi Ueno
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki 701–0193, Japan,Correspondence to: Department of Medical Technology, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama 701–0193, Japan, 193.
| | - Yu Takahashi
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Shinji Murakami
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Kenta Wani
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| | - Yosuke Matsumoto
- Department of Neuropsychiatry, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700–8558, Japan
| | - Motoi Okamoto
- Department of Medical Technology, Graduate School of Health Sciences, Okayama University, Okayama 700–8558, Japan
| | - Takeshi Ishihara
- Department of Psychiatry, Kawasaki Medical School, Kurashiki 701–0192, Japan
| |
Collapse
|
70
|
Llorca A, Deogracias R. Origin, Development, and Synaptogenesis of Cortical Interneurons. Front Neurosci 2022; 16:929469. [PMID: 35833090 PMCID: PMC9272671 DOI: 10.3389/fnins.2022.929469] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian cerebral cortex represents one of the most recent and astonishing inventions of nature, responsible of a large diversity of functions that range from sensory processing to high-order cognitive abilities, such as logical reasoning or language. Decades of dedicated study have contributed to our current understanding of this structure, both at structural and functional levels. A key feature of the neocortex is its outstanding richness in cell diversity, composed by multiple types of long-range projecting neurons and locally connecting interneurons. In this review, we will describe the great diversity of interneurons that constitute local neocortical circuits and summarize the mechanisms underlying their development and their assembly into functional networks.
Collapse
Affiliation(s)
- Alfredo Llorca
- Visual Neuroscience Laboratory, Centre for Discovery Brain Sciences, School of Biomedical Sciences, University of Edinburgh, Edinburg, United Kingdom
- *Correspondence: Alfredo Llorca
| | - Ruben Deogracias
- Neuronal Circuits Formation and Brain Disorders Laboratory, Institute of Neurosciences of Castilla y León (INCyL), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, Salamanca, Spain
- Department of Cell Biology and Pathology, School of Medicine, University of Salamanca, Salamanca, Spain
- Ruben Deogracias
| |
Collapse
|
71
|
Abdollahi M, Fahnestock M. Nurr1 Is Not an Essential Regulator of BDNF in Mouse Cortical Neurons. Int J Mol Sci 2022; 23:6853. [PMID: 35743300 PMCID: PMC9224520 DOI: 10.3390/ijms23126853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/30/2022] Open
Abstract
Nurr1 and brain-derived neurotrophic factor (BDNF) play major roles in cognition. Nurr1 regulates BDNF in midbrain dopaminergic neurons and cerebellar granule cells. Nurr1 and BDNF are also highly expressed in the cerebral cortex, a brain area important in cognition. Due to Nurr1 and BDNF tissue specificity, the regulatory effect of Nurr1 on BDNF in different brain areas cannot be generalized. The relationship between Nurr1 and BDNF in the cortex has not been investigated previously. Therefore, we examined Nurr1-mediated BDNF regulation in cortical neurons in activity-dependent and activity-independent states. Mouse primary cortical neurons were treated with the Nurr1 agonist, amodiaquine (AQ). Membrane depolarization was induced by KCl or veratridine and reversed by nimodipine. AQ and membrane depolarization significantly increased Nurr1 (p < 0.001) and BDNF (pAQ < 0.001, pKCl < 0.01) as assessed by real-time qRT-PCR. However, Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized neurons. Accordingly, the positive correlation between Nurr1 and BDNF expression in AQ and membrane depolarization experiments does not imply co-regulation because Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized cortical neurons. Therefore, in contrast to midbrain dopaminergic neurons and cerebellar granule cells, Nurr1 does not regulate BDNF in cortical neurons.
Collapse
Affiliation(s)
- Mona Abdollahi
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada;
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
72
|
Lipachev N, Melnikova A, Fedosimova S, Arnst N, Kochneva A, Shaikhutdinov N, Dvoeglazova A, Titova A, Mavlikeev M, Aganov A, Osin Y, Kiyasov A, Paveliev M. Postnatal development of the microstructure of cortical GABAergic synapses and perineuronal nets requires sensory input. Neurosci Res 2022; 182:32-40. [PMID: 35710035 DOI: 10.1016/j.neures.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/04/2022] [Accepted: 06/09/2022] [Indexed: 11/25/2022]
Abstract
The brain synaptic circuitry is formed as a result of pre-defined genetic programs and sensory experience during postnatal development. Perineuronal nets ensheath synaptic boutons and control several crucial features of the synapse physiology. Formation of the perineuronal net microstructure during the brain development remains largely unstudied. Here we provide a detailed quantitative description of the 3-dimensional geometry of the synapse and the surrounding perineuronal net in the mouse somatosensory cortex layer IV. We compare the morphology of the synapse+perineuronal net complex in the adult brain formed under normal conditions or in the whisker shaving model of somatosensory deprivation. We demonstrate that the sensory deprivation causes flattening of the 3D PNN mesh geometry and reduction of the VGAT-positive cluster volume in presynaptic boutons. These results reveal a mechanism of the sensory input-dependent synapse morphogenesis during the brain development.
Collapse
Affiliation(s)
- Nikita Lipachev
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O. Box 56, 00790 Helsinki, Finland; Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, 420111 Kazan, Tatarstan, Russia
| | - Anastasia Melnikova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia
| | - Svetlana Fedosimova
- Interdisciplinary Center for Analytic Microscopy, Kazan Federal University, Parizhskoy Kommuny 9, 420021 Kazan, Tatarstan, Russia
| | - Nikita Arnst
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia; Department of Biomedical Sciences, University of Padua, via U. Bassi 58/B, 35131 Padua, Italy
| | - Anastasia Kochneva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia
| | - Nurislam Shaikhutdinov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Anastasia Dvoeglazova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia
| | - Angelina Titova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia
| | - Mikhail Mavlikeev
- Department of Pathological Anatomy, North-Western State Medical University named after I.I.Mechnikov, Piskarevskiy prospect 47, Build. 23, 195067 Saint-Petersburg, Russia
| | - Albert Aganov
- Institute of Physics, Kazan Federal University, Kremlyovskaya 16a, 420111 Kazan, Tatarstan, Russia
| | - Yuri Osin
- Interdisciplinary Center for Analytic Microscopy, Kazan Federal University, Parizhskoy Kommuny 9, 420021 Kazan, Tatarstan, Russia
| | - Andrei Kiyasov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Karl Marx 74, 420012 Kazan, Tatarstan, Russia
| | - Mikhail Paveliev
- Neuroscience Center, University of Helsinki, Viikinkaari 4, P.O. Box 56, 00790 Helsinki, Finland.
| |
Collapse
|
73
|
Maffioli E, Angiulli E, Nonnis S, Grassi Scalvini F, Negri A, Tedeschi G, Arisi I, Frabetti F, D’Aniello S, Alleva E, Cioni C, Toni M. Brain Proteome and Behavioural Analysis in Wild Type, BDNF +/- and BDNF -/- Adult Zebrafish ( Danio rerio) Exposed to Two Different Temperatures. Int J Mol Sci 2022; 23:ijms23105606. [PMID: 35628418 PMCID: PMC9146406 DOI: 10.3390/ijms23105606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Experimental evidence suggests that environmental stress conditions can alter the expression of BDNF and that the expression of this neurotrophin influences behavioural responses in mammalian models. It has been recently demonstrated that exposure to 34 °C for 21 days alters the brain proteome and behaviour in zebrafish. The aim of this work was to investigate the role of BDNF in the nervous system of adult zebrafish under control and heat treatment conditions. For this purpose, zebrafish from three different genotypes (wild type, heterozygous BDNF+/- and knock out BDNF-/-) were kept for 21 days at 26 °C or 34 °C and then euthanized for brain molecular analyses or subjected to behavioural tests (Y-maze test, novel tank test, light and dark test, social preference test, mirror biting test) for assessing behavioural aspects such as boldness, anxiety, social preference, aggressive behaviour, interest for the novel environment and exploration. qRT-PCR analysis showed the reduction of gene expression of BDNF and its receptors after heat treatment in wild type zebrafish. Moreover, proteomic analysis and behavioural tests showed genotype- and temperature-dependent effects on brain proteome and behavioural responding. Overall, the absent expression of BDNF in KO alters (1) the brain proteome by reducing the expression of proteins involved in synapse functioning and neurotransmitter-mediated transduction; (2) the behaviour, which can be interpreted as bolder and less anxious and (3) the cellular and behavioural response to thermal treatment.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Elisa Angiulli
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
- CRC I-WE (Coordinating Research Centre: Innovation for Well-Being and Environment), University of Milan, 20134 Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Armando Negri
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
- CRC I-WE (Coordinating Research Centre: Innovation for Well-Being and Environment), University of Milan, 20134 Milan, Italy
| | - Ivan Arisi
- Bioinformatics Facility, European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, 00161 Rome, Italy;
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00131 Rome, Italy
| | - Flavia Frabetti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40136 Bologna, Italy;
| | - Salvatore D’Aniello
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy;
| | - Enrico Alleva
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Cioni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
| | - Mattia Toni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
- Correspondence:
| |
Collapse
|
74
|
Rupert DD, Shea SD. Parvalbumin-Positive Interneurons Regulate Cortical Sensory Plasticity in Adulthood and Development Through Shared Mechanisms. Front Neural Circuits 2022; 16:886629. [PMID: 35601529 PMCID: PMC9120417 DOI: 10.3389/fncir.2022.886629] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Parvalbumin-positive neurons are the largest class of GABAergic, inhibitory neurons in the central nervous system. In the cortex, these fast-spiking cells provide feedforward and feedback synaptic inhibition onto a diverse set of cell types, including pyramidal cells, other inhibitory interneurons, and themselves. Cortical inhibitory networks broadly, and cortical parvalbumin-expressing interneurons (cPVins) specifically, are crucial for regulating sensory plasticity during both development and adulthood. Here we review the functional properties of cPVins that enable plasticity in the cortex of adult mammals and the influence of cPVins on sensory activity at four spatiotemporal scales. First, cPVins regulate developmental critical periods and adult plasticity through molecular and structural interactions with the extracellular matrix. Second, they activate in precise sequence following feedforward excitation to enforce strict temporal limits in response to the presentation of sensory stimuli. Third, they implement gain control to normalize sensory inputs and compress the dynamic range of output. Fourth, they synchronize broad network activity patterns in response to behavioral events and state changes. Much of the evidence for the contribution of cPVins to plasticity comes from classic models that rely on sensory deprivation methods to probe experience-dependent changes in the brain. We support investigating naturally occurring, adaptive cortical plasticity to study cPVin circuits in an ethologically relevant framework, and discuss recent insights from our work on maternal experience-induced auditory cortical plasticity.
Collapse
Affiliation(s)
- Deborah D. Rupert
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
- Medical Scientist Training Program, Stony Brook University, Stony Brook, NY, United States
| | - Stephen D. Shea
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| |
Collapse
|
75
|
Gibel-Russo R, Benacom D, Di Nardo AA. Non-Cell-Autonomous Factors Implicated in Parvalbumin Interneuron Maturation and Critical Periods. Front Neural Circuits 2022; 16:875873. [PMID: 35601531 PMCID: PMC9115720 DOI: 10.3389/fncir.2022.875873] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/04/2022] [Indexed: 02/04/2023] Open
Abstract
From birth to adolescence, the brain adapts to its environmental stimuli through structural and functional remodeling of neural circuits during critical periods of heightened plasticity. They occur across modalities for proper sensory, motor, linguistic, and cognitive development. If they are disrupted by early-life adverse experiences or genetic deficiencies, lasting consequences include behavioral changes, physiological and cognitive deficits, or psychiatric illness. Critical period timing is orchestrated not only by appropriate neural activity but also by a multitude of signals that participate in the maturation of fast-spiking parvalbumin interneurons and the consolidation of neural circuits. In this review, we describe the various signaling factors that initiate critical period onset, such as BDNF, SPARCL1, or OTX2, which originate either from local neurons or glial cells or from extracortical sources such as the choroid plexus. Critical period closure is established by signals that modulate extracellular matrix and myelination, while timing and plasticity can also be influenced by circadian rhythms and by hormones and corticosteroids that affect brain oxidative stress levels or immune response. Molecular outcomes include lasting epigenetic changes which themselves can be considered signals that shape downstream cross-modal critical periods. Comprehensive knowledge of how these signals and signaling factors interplay to influence neural mechanisms will help provide an inclusive perspective on the effects of early adversity and developmental defects that permanently change perception and behavior.
Collapse
|
76
|
Cossart R, Garel S. Step by step: cells with multiple functions in cortical circuit assembly. Nat Rev Neurosci 2022; 23:395-410. [DOI: 10.1038/s41583-022-00585-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 12/23/2022]
|
77
|
Abstract
For four decades, investigations of the biological basis of critical periods in the developing mammalian visual cortex were dominated by study of the consequences of altered early visual experience in cats and nonhuman primates. The neural deficits thus revealed also provided insight into the origin and neural basis of human amblyopia that in turn motivated additional studies of humans with abnormal early visual input. Recent human studies point to deficits arising from alterations in all visual cortical areas and even in nonvisual cortical regions. As the new human data accumulated in parallel with a near-complete shift toward the use of rodent animal models for the study of neural mechanisms, it is now essential to review the human data and the earlier animal data obtained from cats and monkeys to infer general conclusions and to optimize future choice of the most appropriate animal model. Expected final online publication date for the Annual Review of Vision Science, Volume 8 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Donald E Mitchell
- Department of Psychology & Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada;
| | - Daphne Maurer
- Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, Ontario, Canada;
| |
Collapse
|
78
|
Ribot J, Breton R, Dallérac G, Rouach N. [Astrocytes, guardians of critical period plasticity]. Med Sci (Paris) 2022; 38:251-254. [PMID: 35333161 DOI: 10.1051/medsci/2022014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Jérôme Ribot
- Interactions neurogliales en physiologie et pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 11 place Marcelin Berthelot, 75005 Paris, France
| | - Rachel Breton
- Interactions neurogliales en physiologie et pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 11 place Marcelin Berthelot, 75005 Paris, France - École doctorale 568, Université Paris-Saclay, Université PSL, Le Kremlin Bicêtre, France - Adresse actuelle : Astrocytes et Cognition, Institut des neurosciences de Paris-Saclay, CNRS UMR 9197, Université Paris-Saclay, Orsay, France
| | - Glenn Dallérac
- Interactions neurogliales en physiologie et pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 11 place Marcelin Berthelot, 75005 Paris, France
| | - Nathalie Rouach
- Interactions neurogliales en physiologie et pathologies cérébrales, Centre interdisciplinaire de recherche en biologie, Collège de France, CNRS, Inserm, Labex Memolife, Université PSL, 11 place Marcelin Berthelot, 75005 Paris, France
| |
Collapse
|
79
|
Body Weight-Supported Treadmill Training Ameliorates Motoneuronal Hyperexcitability by Increasing GAD-65/67 and KCC2 Expression via TrkB Signaling in Rats with Incomplete Spinal Cord Injury. Neurochem Res 2022; 47:1679-1691. [PMID: 35320460 PMCID: PMC9124175 DOI: 10.1007/s11064-022-03561-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/29/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022]
Abstract
Spasticity is a typical consequence after spinal cord injury (SCI). The critical reasons are reducing the synthesis of Gamma-Aminobutyric Acid (GABA), glycine and potassium chloride co-transporter 2 (KCC2) inside the distal spinal cord. The current work aimed to test whether exercise training could increase the expression of glutamic acid decarboxylase 65/67 (GAD-65/67, the key enzymes in GABA synthesis) and KCC2 in the distal spinal cord via tropomyosin-related kinase B (TrkB) signaling. The experimental rats were randomly assigned to the following five groups: Sham, SCI/phosphate-buffered saline (PBS), SCI-treadmill training (TT)/PBS, SCI/TrkB-IgG, and SCI-TT/TrkB-IgG. After that, the model of T10 contusion SCI was used, then TrkB-IgG was used to prevent TrkB activity at 7 days post-SCI. Body weight-supported treadmill training started on the 8th day post-SCI for four weeks. The Hmax/Mmax ratio and the rate-dependent depression of H-reflex were used to assess the excitability of spinal motoneuronal networks. Western blotting and Immunohistochemistry techniques were utilized for measuring the expression of GAD-65, GAD-67, and KCC2. The findings revealed that exercise training could reduce motoneuronal excitability and boost GAD-65, GAD-67, and KCC2 production in the distal region of the spinal cord after SCI. The effects of exercise training were decreased after the TrkB signaling was inhibited. The present exploration demonstrated that exercise training increases GAD-65, GAD-67, and KCC2 expression in the spinal cord via TrkB signaling and that this method could also improve rats with motoneuronal hyperexcitability and spasticity induced by incomplete SCI.
Collapse
|
80
|
Jîtcă G, Ősz BE, Tero-Vescan A, Miklos AP, Rusz CM, Bătrînu MG, Vari CE. Positive Aspects of Oxidative Stress at Different Levels of the Human Body: A Review. Antioxidants (Basel) 2022; 11:antiox11030572. [PMID: 35326222 PMCID: PMC8944834 DOI: 10.3390/antiox11030572] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress is the subject of numerous studies, most of them focusing on the negative effects exerted at both molecular and cellular levels, ignoring the possible benefits of free radicals. More and more people admit to having heard of the term "oxidative stress", but few of them understand the meaning of it. We summarized and analyzed the published literature data in order to emphasize the importance and adaptation mechanisms of basal oxidative stress. This review aims to provide an overview of the mechanisms underlying the positive effects of oxidative stress, highlighting these effects, as well as the risks for the population consuming higher doses than the recommended daily intake of antioxidants. The biological dose-response curve in oxidative stress is unpredictable as reactive species are clearly responsible for cellular degradation, whereas antioxidant therapies can alleviate senescence by maintaining redox balance; nevertheless, excessive doses of the latter can modify the redox balance of the cell, leading to a negative outcome. It can be stated that the presence of oxidative status or oxidative stress is a physiological condition with well-defined roles, yet these have been insufficiently researched and explored. The involvement of reactive oxygen species in the pathophysiology of some associated diseases is well-known and the involvement of antioxidant therapies in the processes of senescence, apoptosis, autophagy, and the maintenance of cellular homeostasis cannot be denied. All data in this review support the idea that oxidative stress is an undesirable phenomenon in high and long-term concentrations, but regular exposure is consistent with the hormetic theory.
Collapse
Affiliation(s)
- George Jîtcă
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| | - Bianca E. Ősz
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (G.J.); (C.E.V.)
- Correspondence:
| | - Amelia Tero-Vescan
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (A.T.-V.); (A.P.M.)
| | - Amalia Pușcaș Miklos
- Department of Biochemistry, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (A.T.-V.); (A.P.M.)
| | - Carmen-Maria Rusz
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (C.-M.R.); (M.-G.B.)
| | - Mădălina-Georgiana Bătrînu
- Doctoral School of Medicine and Pharmacy, I.O.S.U.D, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (C.-M.R.); (M.-G.B.)
| | - Camil E. Vari
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540139 Târgu Mureș, Romania; (G.J.); (C.E.V.)
| |
Collapse
|
81
|
Tanti A, Belliveau C, Nagy C, Maitra M, Denux F, Perlman K, Chen F, Mpai R, Canonne C, Théberge S, McFarquhar A, Davoli MA, Belzung C, Turecki G, Mechawar N. Child abuse associates with increased recruitment of perineuronal nets in the ventromedial prefrontal cortex: a possible implication of oligodendrocyte progenitor cells. Mol Psychiatry 2022; 27:1552-1561. [PMID: 34799691 PMCID: PMC9095471 DOI: 10.1038/s41380-021-01372-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022]
Abstract
Child abuse (CA) is a strong predictor of psychopathologies and suicide, altering normal trajectories of brain development in areas closely linked to emotional responses such as the prefrontal cortex (PFC). Yet, the cellular underpinnings of these enduring effects are unclear. Childhood and adolescence are marked by the protracted formation of perineuronal nets (PNNs), which orchestrate the closure of developmental windows of cortical plasticity by regulating the functional integration of parvalbumin interneurons into neuronal circuits. Using well-characterized post-mortem brain samples, we show that a history of CA is specifically associated with increased densities and morphological complexity of WFL-labeled PNNs in the ventromedial PFC (BA11/12), possibly suggesting increased recruitment and maturation of PNNs. Through single-nucleus sequencing and fluorescent in situ hybridization, we found that the expression of canonical components of PNNs is enriched in oligodendrocyte progenitor cells (OPCs), and that they are upregulated in CA victims. These correlational findings suggest that early-life adversity may lead to persistent patterns of maladaptive behaviors by reducing the neuroplasticity of cortical circuits through the enhancement of developmental OPC-mediated PNN formation.
Collapse
Affiliation(s)
- Arnaud Tanti
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- UMR 1253, iBrain, Inserm, Université de Tours, Tours, France.
| | - Claudia Belliveau
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Corina Nagy
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Malosree Maitra
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Fanny Denux
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Kelly Perlman
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Frank Chen
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Refilwe Mpai
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Candice Canonne
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Stéphanie Théberge
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Ashley McFarquhar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Maria Antonietta Davoli
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | | | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montréal, QC, Canada
| | - Naguib Mechawar
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada.
- Department of Psychiatry, McGill University, Montréal, QC, Canada.
| |
Collapse
|
82
|
Enzymatic Degradation of Cortical Perineuronal Nets Reverses GABAergic Interneuron Maturation. Mol Neurobiol 2022; 59:2874-2893. [PMID: 35233718 PMCID: PMC9016038 DOI: 10.1007/s12035-022-02772-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 02/16/2022] [Indexed: 12/03/2022]
Abstract
Perineuronal nets (PNNs) are specialised extracellular matrix structures which preferentially enwrap fast-spiking (FS) parvalbumin interneurons and have diverse roles in the cortex. PNN maturation coincides with closure of the critical period of cortical plasticity. We have previously demonstrated that BDNF accelerates interneuron development in a c-Jun-NH2-terminal kinase (JNK)–dependent manner, which may involve upstream thousand-and-one amino acid kinase 2 (TAOK2). Chondroitinase-ABC (ChABC) enzymatic digestion of PNNs reportedly reactivates ‘juvenile-like’ plasticity in the adult CNS. However, the mechanisms involved are unclear. We show that ChABC produces an immature molecular phenotype in cultured cortical neurons, corresponding to the phenotype prior to critical period closure. ChABC produced different patterns of PNN-related, GABAergic and immediate early (IE) gene expression than well-characterised modulators of mature plasticity and network activity (GABAA-R antagonist, bicuculline, and sodium-channel blocker, tetrodotoxin (TTX)). ChABC downregulated JNK activity, while this was upregulated by bicuculline. Bicuculline, but not ChABC, upregulated Bdnf expression and ERK activity. Furthermore, we found that BDNF upregulation of semaphorin-3A and IE genes was TAOK mediated. Our data suggest that ChABC heightens structural flexibility and network disinhibition, potentially contributing to ‘juvenile-like’ plasticity. The molecular phenotype appears to be distinct from heightened mature synaptic plasticity and could relate to JNK signalling. Finally, we highlight that BDNF regulation of plasticity and PNNs involves TAOK signalling.
Collapse
|
83
|
Poh EZ, Green C, Agostinelli L, Penrose-Menz M, Karl AK, Harvey AR, Rodger J. Manipulating the Level of Sensorimotor Stimulation during LI-rTMS Can Improve Visual Circuit Reorganisation in Adult Ephrin-A2A5 -/- Mice. Int J Mol Sci 2022; 23:ijms23052418. [PMID: 35269561 PMCID: PMC8910719 DOI: 10.3390/ijms23052418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/15/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Repetitive transcranial magnetic stimulation (rTMS) is a non-invasive brain stimulation technique that has the potential to treat a variety of neurologic and psychiatric disorders. The extent of rTMS-induced neuroplasticity may be dependent on a subject's brain state at the time of stimulation. Chronic low intensity rTMS (LI-rTMS) has previously been shown to induce beneficial structural and functional reorganisation within the abnormal visual circuits of ephrin-A2A5-/- mice in ambient lighting. Here, we administered chronic LI-rTMS in adult ephrin-A2A5-/- mice either in a dark environment or concurrently with voluntary locomotion. One day after the last stimulation session, optokinetic responses were assessed and fluorescent tracers were injected to map corticotectal and geniculocortical projections. We found that LI-rTMS in either treatment condition refined the geniculocortical map. Corticotectal projections were improved in locomotion+LI-rTMS subjects, but not in dark + LI-rTMS and sham groups. Visuomotor behaviour was not improved in any condition. Our results suggest that the beneficial reorganisation of abnormal visual circuits by rTMS can be significantly influenced by simultaneous, ambient visual input and is enhanced by concomitant physical exercise. Furthermore, the observed pathway-specific effects suggest that regional molecular changes and/or the relative proximity of terminals to the induced electric fields influence the outcomes of LI-rTMS on abnormal circuitry.
Collapse
Affiliation(s)
- Eugenia Z. Poh
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (E.Z.P.); (M.P.-M.); (A.-K.K.)
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (C.G.); (L.A.); (A.R.H.)
- Perron Institute for Neurological and Translational Research, 8 Verdun St, Nedlands, WA 6009, Australia
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, 1105 BA Amsterdam, The Netherlands
| | - Courtney Green
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (C.G.); (L.A.); (A.R.H.)
| | - Luca Agostinelli
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (C.G.); (L.A.); (A.R.H.)
| | - Marissa Penrose-Menz
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (E.Z.P.); (M.P.-M.); (A.-K.K.)
| | - Ann-Kathrin Karl
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (E.Z.P.); (M.P.-M.); (A.-K.K.)
- Department of Neurology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080 Würzburg, Germany
| | - Alan R. Harvey
- School of Human Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (C.G.); (L.A.); (A.R.H.)
- Perron Institute for Neurological and Translational Research, 8 Verdun St, Nedlands, WA 6009, Australia
| | - Jennifer Rodger
- School of Biological Sciences, The University of Western Australia, Crawley, WA 6009, Australia; (E.Z.P.); (M.P.-M.); (A.-K.K.)
- Perron Institute for Neurological and Translational Research, 8 Verdun St, Nedlands, WA 6009, Australia
- Correspondence: ; Tel.: +61-8-6488-2245
| |
Collapse
|
84
|
Strettoi E, Di Marco B, Orsini N, Napoli D. Retinal Plasticity. Int J Mol Sci 2022; 23:ijms23031138. [PMID: 35163059 PMCID: PMC8835074 DOI: 10.3390/ijms23031138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
Brain plasticity is a well-established concept designating the ability of central nervous system (CNS) neurons to rearrange as a result of learning, when adapting to changeable environmental conditions or else while reacting to injurious factors. As a part of the CNS, the retina has been repeatedly probed for its possible ability to respond plastically to a variably altered environment or to pathological insults. However, numerous studies support the conclusion that the retina, outside the developmental stage, is endowed with only limited plasticity, exhibiting, instead, a remarkable ability to maintain a stable architectural and functional organization. Reviewed here are representative examples of hippocampal and cortical paradigms of plasticity and of retinal structural rearrangements found in organization and circuitry following altered developmental conditions or occurrence of genetic diseases leading to neuronal degeneration. The variable rate of plastic changes found in mammalian retinal neurons in different circumstances is discussed, focusing on structural plasticity. The likely adaptive value of maintaining a low level of plasticity in an organ subserving a sensory modality that is dominant for the human species and that requires elevated fidelity is discussed.
Collapse
Affiliation(s)
- Enrica Strettoi
- CNR Neuroscience Institute, 56124 Pisa, Italy; (B.D.M.); (N.O.); (D.N.)
- Correspondence: ; Tel.: +39-0503153213
| | - Beatrice Di Marco
- CNR Neuroscience Institute, 56124 Pisa, Italy; (B.D.M.); (N.O.); (D.N.)
- Regional Doctorate School in Neuroscience, Universities of Florence, Pisa and Siena, 50134 Florence, Italy
| | - Noemi Orsini
- CNR Neuroscience Institute, 56124 Pisa, Italy; (B.D.M.); (N.O.); (D.N.)
- Regional Doctorate School in Neuroscience, Universities of Florence, Pisa and Siena, 50134 Florence, Italy
| | - Debora Napoli
- CNR Neuroscience Institute, 56124 Pisa, Italy; (B.D.M.); (N.O.); (D.N.)
- Regional Doctorate School in Neuroscience, Universities of Florence, Pisa and Siena, 50134 Florence, Italy
| |
Collapse
|
85
|
Abstract
Neuroplasticity, i.e., the modifiability of the brain, is different in development and adulthood. The first includes changes in: (i) neurogenesis and control of neuron number; (ii) neuronal migration; (iii) differentiation of the somato-dendritic and axonal phenotypes; (iv) formation of connections; (v) cytoarchitectonic differentiation. These changes are often interrelated and can lead to: (vi) system-wide modifications of brain structure as well as to (vii) acquisition of specific functions such as ocular dominance or language. Myelination appears to be plastic both in development and adulthood, at least, in rodents. Adult neuroplasticity is limited, and is mainly expressed as changes in the strength of excitatory and inhibitory synapses while the attempts to regenerate connections have met with limited success. The outcomes of neuroplasticity are not necessarily adaptive, but can also be the cause of neurological and psychiatric pathologies.
Collapse
|
86
|
Skirzewski M, Molotchnikoff S, Hernandez LF, Maya-Vetencourt JF. Multisensory Integration: Is Medial Prefrontal Cortex Signaling Relevant for the Treatment of Higher-Order Visual Dysfunctions? Front Mol Neurosci 2022; 14:806376. [PMID: 35110996 PMCID: PMC8801884 DOI: 10.3389/fnmol.2021.806376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/17/2021] [Indexed: 11/29/2022] Open
Abstract
In the mammalian brain, information processing in sensory modalities and global mechanisms of multisensory integration facilitate perception. Emerging experimental evidence suggests that the contribution of multisensory integration to sensory perception is far more complex than previously expected. Here we revise how associative areas such as the prefrontal cortex, which receive and integrate inputs from diverse sensory modalities, can affect information processing in unisensory systems via processes of down-stream signaling. We focus our attention on the influence of the medial prefrontal cortex on the processing of information in the visual system and whether this phenomenon can be clinically used to treat higher-order visual dysfunctions. We propose that non-invasive and multisensory stimulation strategies such as environmental enrichment and/or attention-related tasks could be of clinical relevance to fight cerebral visual impairment.
Collapse
Affiliation(s)
- Miguel Skirzewski
- Rodent Cognition Research and Innovation Core, University of Western Ontario, London, ON, Canada
| | - Stéphane Molotchnikoff
- Département de Sciences Biologiques, Université de Montréal, Montreal, QC, Canada
- Département de Génie Electrique et Génie Informatique, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Luis F. Hernandez
- Knoebel Institute for Healthy Aging, University of Denver, Denver, CO, United States
| | - José Fernando Maya-Vetencourt
- Department of Biology, University of Pisa, Pisa, Italy
- Centre for Synaptic Neuroscience, Istituto Italiano di Tecnologia (IIT), Genova, Italy
- *Correspondence: José Fernando Maya-Vetencourt
| |
Collapse
|
87
|
Cherubini E, Di Cristo G, Avoli M. Dysregulation of GABAergic Signaling in Neurodevelomental Disorders: Targeting Cation-Chloride Co-transporters to Re-establish a Proper E/I Balance. Front Cell Neurosci 2022; 15:813441. [PMID: 35069119 PMCID: PMC8766311 DOI: 10.3389/fncel.2021.813441] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 11/30/2021] [Indexed: 01/01/2023] Open
Abstract
The construction of the brain relies on a series of well-defined genetically and experience- or activity -dependent mechanisms which allow to adapt to the external environment. Disruption of these processes leads to neurological and psychiatric disorders, which in many cases are manifest already early in postnatal life. GABA, the main inhibitory neurotransmitter in the adult brain is one of the major players in the early assembly and formation of neuronal circuits. In the prenatal and immediate postnatal period GABA, acting on GABAA receptors, depolarizes and excites targeted cells via an outwardly directed flux of chloride. In this way it activates NMDA receptors and voltage-dependent calcium channels contributing, through intracellular calcium rise, to shape neuronal activity and to establish, through the formation of new synapses and elimination of others, adult neuronal circuits. The direction of GABAA-mediated neurotransmission (depolarizing or hyperpolarizing) depends on the intracellular levels of chloride [Cl−]i, which in turn are maintained by the activity of the cation-chloride importer and exporter KCC2 and NKCC1, respectively. Thus, the premature hyperpolarizing action of GABA or its persistent depolarizing effect beyond the postnatal period, leads to behavioral deficits associated with morphological alterations and an excitatory (E)/inhibitory (I) imbalance in selective brain areas. The aim of this review is to summarize recent data concerning the functional role of GABAergic transmission in building up and refining neuronal circuits early in development and its dysfunction in neurodevelopmental disorders such as Autism Spectrum Disorders (ASDs), schizophrenia and epilepsy. In particular, we focus on novel information concerning the mechanisms by which alterations in cation-chloride co-transporters (CCC) generate behavioral and cognitive impairment in these diseases. We discuss also the possibility to re-establish a proper GABAA-mediated neurotransmission and excitatory (E)/inhibitory (I) balance within selective brain areas acting on CCC.
Collapse
Affiliation(s)
- Enrico Cherubini
- European Brain Research Institute (EBRI)-Rita Levi-Montalcini, Roma, Italy
- *Correspondence: Enrico Cherubini
| | - Graziella Di Cristo
- Neurosciences Department, Université de Montréal and CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - Massimo Avoli
- Montreal Neurological Institute-Hospital and Departments of Neurology and Neurosurgery and of Physiology, McGill University, Montreal, QC, Canada
| |
Collapse
|
88
|
St. Pierre M, Rastogi N, Brown A, Parmar P, Lechner C, Fung C, Chavez-Valdez R. Intrauterine Growth Restriction Disrupts the Postnatal Critical Period of Synaptic Plasticity in the Mouse Dorsal Hippocampus in a Model of Hypertensive Disease of Pregnancy. Dev Neurosci 2022; 44:214-232. [PMID: 34933306 PMCID: PMC9209574 DOI: 10.1159/000521611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/16/2021] [Indexed: 01/03/2023] Open
Abstract
INTRODUCTION Intrauterine growth restriction (IUGR) from hypertensive disease of pregnancy complicates up to 10% of all pregnancies. Significant hippocampal-dependent cognitive and memory impairments as well as neuropsychiatric disorders have been linked to IUGR. Because disturbance of the hippocampal critical period (CPd) of synaptic plasticity leads to impairments similar to those described in IUGR human offspring, we hypothesized that IUGR would perturb the CPd of synaptic plasticity in the mouse hippocampus in our model. METHODS IUGR was produced by a micro-osmotic pump infusion of the potent vasoconstrictor U-46619, a thromboxane A2-agonist, at embryonic day 12.5 in C57BL/6J mouse dams to precipitate hypertensive disease of pregnancy and IUGR. Sham-operated mice acted as controls. At P10, P18, and P40, we assessed astrogliosis using GFAP-IHC. In dorsal CA1 and CA3 subfields, we assessed the immunoreactivities (IR) (IF-IHC) to (i) parvalbumin (PV) and glutamate decarboxylase (GAD) 65/67, involved in CPd onset; (ii) PSA-NCAM that antagonizes CPd onset; (iii) NPTX2, necessary for excitatory synapse formation and engagement of CPd; and (iv) MBP and WFA, staining perineural nets (PNNs), marking CPd closure. ImageJ/Fiji and IMARIS were used for image processing and SPSS v24 for statistical analysis. RESULTS Although PV+ interneuron numbers and IR intensity were unchanged, development of GAD65/67+ synaptic boutons was accelerated at P18 IUGR mice and inversely correlated with decreased expression of PSA-NCAM in the CA of P18 IUGR mice at P18. NPTX2+ puncta and total volume were persistently decreased in the CA3 pyramidal and radiatum layers of IUGR mice from P18 to P40. At P40, axonal myelination (MBP+) in CA3 of IUGR mice was decreased and correlated with NPTX2 deficits. Lastly, the volume and integrity of the PNNs in the dorsal CA was disrupted in IUGR mice at P40. DISCUSSION/CONCLUSION IUGR disrupts the molecular and structural initiation, consolidation, and closure of the CPd of synaptic plasticity in the mouse hippocampus in our model, which may explain the learning and memory deficits observed in juvenile IUGR mice and the cognitive disorders seen in human IUGR offspring. The mechanistic links warrant further investigation, to identify therapeutic targets to prevent neurodevelopmental deficits in patients affected by IUGR.
Collapse
Affiliation(s)
- Mark St. Pierre
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins School of Medicine. Baltimore, MD
| | - Neetika Rastogi
- Department of Neurosciences, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD
| | - Ashley Brown
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - Pritika Parmar
- Department of Neurosciences, Johns Hopkins University Krieger School of Arts and Sciences, Baltimore, MD
| | - Charles Lechner
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins School of Medicine. Baltimore, MD
| | - Camille Fung
- Division of Neonatology, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - Raul Chavez-Valdez
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, Johns Hopkins School of Medicine. Baltimore, MD,Corresponding author: Dr. Raul Chavez-Valdez. Associate Professor. Department of Pediatrics, Division of Neonatology, Johns Hopkins Hospital, 600 N. Wolfe Street, CMSC 6-104, Baltimore, MD 21287, USA. Telephone: (410) 955-7156,
| |
Collapse
|
89
|
Zhu Y, Wang MJ, Crawford KM, Ramírez-Tapia JC, Lussier AA, Davis KA, de Leeuw C, Takesian AE, Hensch TK, Smoller JW, Dunn EC. Sensitive period-regulating genetic pathways and exposure to adversity shape risk for depression. Neuropsychopharmacology 2022; 47:497-506. [PMID: 34689167 PMCID: PMC8674315 DOI: 10.1038/s41386-021-01172-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 07/23/2021] [Accepted: 08/30/2021] [Indexed: 01/03/2023]
Abstract
Animal and human studies have documented the existence of developmental windows (or sensitive periods) when experience can have lasting effects on brain structure or function, behavior, and disease. Although sensitive periods for depression likely arise through a complex interplay of genes and experience, this possibility has not yet been explored in humans. We examined the effect of genetic pathways regulating sensitive periods, alone and in interaction with common childhood adversities, on depression risk. Guided by a translational approach, we: (1) performed association analyses of three gene sets (60 genes) shown in animal studies to regulate sensitive periods using summary data from a genome-wide association study of depression (n = 807,553); (2) evaluated the developmental expression patterns of these genes using data from BrainSpan (n = 31), a transcriptional atlas of postmortem brain samples; and (3) tested gene-by-development interplay (dGxE) by analyzing the combined effect of common variants in sensitive period genes and time-varying exposure to two types of childhood adversity within a population-based birth cohort (n = 6254). The gene set regulating sensitive period opening associated with increased depression risk. Notably, 6 of the 15 genes in this set showed developmentally regulated gene-level expression. We also identified a statistical interaction between caregiver physical or emotional abuse during ages 1-5 years and genetic risk for depression conferred by the opening genes. Genes involved in regulating sensitive periods are differentially expressed across the life course and may be implicated in depression vulnerability. Our findings about gene-by-development interplay motivate further research in large, more diverse samples to further unravel the complexity of depression etiology through a sensitive period lens.
Collapse
Affiliation(s)
- Yiwen Zhu
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Min-Jung Wang
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | | | - Alexandre A Lussier
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Kathryn A Davis
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Christiaan de Leeuw
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Department of Complex Trait Genetics, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Anne E Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye & Ear and Department of Otorhinolaryngology and Head/Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Takao K Hensch
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
- F.M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jordan W Smoller
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Erin C Dunn
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
- Stanley Center for Psychiatric Research, The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Center on the Developing Child, Cambridge, MA, USA.
| |
Collapse
|
90
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
91
|
Jenks KR, Tsimring K, Ip JPK, Zepeda JC, Sur M. Heterosynaptic Plasticity and the Experience-Dependent Refinement of Developing Neuronal Circuits. Front Neural Circuits 2021; 15:803401. [PMID: 34949992 PMCID: PMC8689143 DOI: 10.3389/fncir.2021.803401] [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: 10/27/2021] [Accepted: 11/15/2021] [Indexed: 01/01/2023] Open
Abstract
Neurons remodel the structure and strength of their synapses during critical periods of development in order to optimize both perception and cognition. Many of these developmental synaptic changes are thought to occur through synapse-specific homosynaptic forms of experience-dependent plasticity. However, homosynaptic plasticity can also induce or contribute to the plasticity of neighboring synapses through heterosynaptic interactions. Decades of research in vitro have uncovered many of the molecular mechanisms of heterosynaptic plasticity that mediate local compensation for homosynaptic plasticity, facilitation of further bouts of plasticity in nearby synapses, and cooperative induction of plasticity by neighboring synapses acting in concert. These discoveries greatly benefited from new tools and technologies that permitted single synapse imaging and manipulation of structure, function, and protein dynamics in living neurons. With the recent advent and application of similar tools for in vivo research, it is now feasible to explore how heterosynaptic plasticity contribute to critical periods and the development of neuronal circuits. In this review, we will first define the forms heterosynaptic plasticity can take and describe our current understanding of their molecular mechanisms. Then, we will outline how heterosynaptic plasticity may lead to meaningful refinement of neuronal responses and observations that suggest such mechanisms are indeed at work in vivo. Finally, we will use a well-studied model of cortical plasticity—ocular dominance plasticity during a critical period of visual cortex development—to highlight the molecular overlap between heterosynaptic and developmental forms of plasticity, and suggest potential avenues of future research.
Collapse
Affiliation(s)
- Kyle R Jenks
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Katya Tsimring
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jacque Pak Kan Ip
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jose C Zepeda
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Mriganka Sur
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
92
|
Inhibition of Cdk5 in PV Neurons Reactivates Experience-Dependent Plasticity in Adult Visual Cortex. Int J Mol Sci 2021; 23:ijms23010186. [PMID: 35008611 PMCID: PMC8745415 DOI: 10.3390/ijms23010186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/18/2022] Open
Abstract
Cyclin-dependent kinase 5 (Cdk5) has been shown to play a critical role in brain development, learning, memory and neural processing in general. Cdk5 is widely distributed in many neuron types in the central nervous system, while its cell-specific role is largely unknown. Our previous study showed that Cdk5 inhibition restored ocular dominance (OD) plasticity in adulthood. In this study, we specifically knocked down Cdk5 in different types of neurons in the visual cortex and examined OD plasticity by optical imaging of intrinsic signals. Downregulation of Cdk5 in parvalbumin-expressing (PV) inhibitory neurons, but not other neurons, reactivated adult mouse visual cortical plasticity. Cdk5 knockdown in PV neurons reduced the evoked firing rate, which was accompanied by an increment in the threshold current for the generation of a single action potential (AP) and hyperpolarization of the resting membrane potential. Moreover, chemogenetic activation of PV neurons in the visual cortex can attenuate the restoration of OD plasticity by Cdk5 inhibition. Taken together, our results suggest that Cdk5 in PV interneurons may play a role in modulating the excitation and inhibition balance to control the plasticity of the visual cortex.
Collapse
|
93
|
Dubbioso R, Pellegrino G, Ranieri F, Di Pino G, Capone F, Dileone M, Iodice R, Ruggiero L, Tozza S, Uncini A, Manganelli F, Di Lazzaro V. BDNF polymorphism and inter hemispheric balance of motor cortex excitability: a preliminary study. J Neurophysiol 2021; 127:204-212. [PMID: 34936818 DOI: 10.1152/jn.00268.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preclinical studies have demonstrated that Brain-Derived Neurotrophic Factor (BDNF) plays a crucial role in the homeostatic regulation of cortical excitability and excitation/inhibition balance. Using transcranial magnetic stimulation (TMS) techniques we investigated whether BDNF polymorphism could influence cortical excitability of the left and right primary motor cortex in healthy humans. Twenty-nine participants were recruited and genotyped for the presence of the BDNF Val66Met polymorphism, namely homozygous for the valine allele (Val/Val), heterozygotes (Val/Met), and homozygous for the methionine allele (Met/Met). Blinded to the latter, we evaluated inhibitory and facilitatory circuits of the left (LH) and right motor cortex (RH) by measuring resting (RMT) and active motor threshold (AMT), short interval intracortical inhibition (SICI) and intracortical facilitation (ICF). For each neurophysiological metric we also considered the inter-hemispheric balance expressed by the Laterality Index (LI). Val/Val participants (n= 21) exhibited an overall higher excitability of the LH compared to the RH, as probed by lower motor thresholds, lower SICI and higher ICF. Val/Val participants displayed positive LI, especially for AMT and ICF (all p< 0.05), indicating higher LH excitability and more pronounced inter-hemispheric excitability imbalance as compared to Met carriers. Our preliminary results suggest that BDNF Val66Met polymorphism might influence interhemispheric balance of motor cortex excitability.
Collapse
Affiliation(s)
- Raffaele Dubbioso
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Giovanni Pellegrino
- Neurology and Neurosurgery Department, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Federico Ranieri
- Neurology Unit, Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Giovanni Di Pino
- Research Unit of Neurophysiology and Neuroengineering of Human-Technology Interaction (NeXTlab), Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Fioravante Capone
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Michele Dileone
- Faculty of Health Sciences, University of Castilla La Mancha, Talavera de la Reina, Spain
| | - Rosa Iodice
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Lucia Ruggiero
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Stefano Tozza
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Antonino Uncini
- Department of Neuroscience, Imaging and Clinical Sciences, University "G. d'Annunzio", Chieti-Pescara, Italy
| | - Fiore Manganelli
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples "Federico II", Naples, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
94
|
Mu L, Liu X, Yu H, Hu M, Friedman V, Kelly TJ, Zhao L, Liu QS. Ibudilast attenuates cocaine self-administration and prime- and cue-induced reinstatement of cocaine seeking in rats. Neuropharmacology 2021; 201:108830. [PMID: 34626665 PMCID: PMC8656241 DOI: 10.1016/j.neuropharm.2021.108830] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 11/21/2022]
Abstract
Ibudilast is a non-selective phosphodiesterase (PDE) inhibitor and glial cell modulator which has shown great promise for the treatment of drug and alcohol use disorders in recent clinical studies. However, it is unknown whether and how ibudilast affects cocaine seeking behavior. Here we show that systemic administration of ibudilast dose-dependently reduced cocaine self-administration under fixed- and progressive-ratio reinforcement schedules in rats and shifted cocaine dose-response curves downward. In addition, ibudilast decreased cocaine prime- and cue-induced reinstatement of cocaine seeking. These results indicate that ibudilast was effective in reducing the reinforcing effects of cocaine and relapse to cocaine seeking. Chronic cocaine exposure induces cAMP-related neuroadaptations in the reward circuitry of the brain. To investigate potential mechanisms for ibudilast-induced attenuation of cocaine self-administration, we recorded from ventral tegmental area (VTA) dopamine neurons in ex vivo midbrain slices prepared from rats that had undergone saline and cocaine self-administration. We found cocaine self-administration led to a decrease in inhibitory postsynaptic currents (IPSCs), an increase in the AMPAR/NMDAR ratio, and an increase in the excitation to inhibition (E/I) ratio. Ibudilast pretreatments enhanced GABAergic inhibition and did not further change cocaine-induced potentiation of excitation, leading to normalization of the E/I ratio. Restoration of the balance between excitation and inhibition in VTA dopamine neurons may contribute to the attenuation of cocaine self-administration by ibudilast.
Collapse
Affiliation(s)
- Lianwei Mu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Xiaojie Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Hao Yu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Mengming Hu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Vladislav Friedman
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Thomas J Kelly
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Li Zhao
- Department of Exercise Physiology, Beijing Sport University, Beijing, 100084, China
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
95
|
Prestigio C, Ferrante D, Marte A, Romei A, Lignani G, Onofri F, Valente P, Benfenati F, Baldelli P. REST/NRSF drives homeostatic plasticity of inhibitory synapses in a target-dependent fashion. eLife 2021; 10:e69058. [PMID: 34855580 PMCID: PMC8639147 DOI: 10.7554/elife.69058] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/22/2021] [Indexed: 12/31/2022] Open
Abstract
The repressor-element 1-silencing transcription/neuron-restrictive silencer factor (REST/NRSF) controls hundreds of neuron-specific genes. We showed that REST/NRSF downregulates glutamatergic transmission in response to hyperactivity, thus contributing to neuronal homeostasis. However, whether GABAergic transmission is also implicated in the homeostatic action of REST/NRSF is unknown. Here, we show that hyperactivity-induced REST/NRSF activation, triggers a homeostatic rearrangement of GABAergic inhibition, with increased frequency of miniature inhibitory postsynaptic currents (IPSCs) and amplitude of evoked IPSCs in mouse cultured hippocampal neurons. Notably, this effect is limited to inhibitory-onto-excitatory neuron synapses, whose density increases at somatic level and decreases in dendritic regions, demonstrating a complex target- and area-selectivity. The upscaling of perisomatic inhibition was occluded by TrkB receptor inhibition and resulted from a coordinated and sequential activation of the Npas4 and Bdnf gene programs. On the opposite, the downscaling of dendritic inhibition was REST-dependent, but BDNF-independent. The findings highlight the central role of REST/NRSF in the complex transcriptional responses aimed at rescuing physiological levels of network activity in front of the ever-changing environment.
Collapse
Affiliation(s)
- Cosimo Prestigio
- Department of Experimental Medicine, University of GenovaGenovaItaly
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Daniele Ferrante
- Department of Experimental Medicine, University of GenovaGenovaItaly
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Antonella Marte
- Department of Experimental Medicine, University of GenovaGenovaItaly
| | - Alessandra Romei
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, Queen Square HouseLondonUnited Kingdom
| | - Franco Onofri
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Pierluigi Valente
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di TecnologiaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| | - Pietro Baldelli
- Department of Experimental Medicine, University of GenovaGenovaItaly
- IRCCS, Ospedale Policlinico San MartinoGenovaItaly
| |
Collapse
|
96
|
Woodward EM, Coutellier L. Age- and sex-specific effects of stress on parvalbumin interneurons in preclinical models: Relevance to sex differences in clinical neuropsychiatric and neurodevelopmental disorders. Neurosci Biobehav Rev 2021; 131:1228-1242. [PMID: 34718048 PMCID: PMC8642301 DOI: 10.1016/j.neubiorev.2021.10.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/06/2021] [Accepted: 10/23/2021] [Indexed: 01/06/2023]
Abstract
Stress is a major risk factor for neurodevelopmental and neuropsychiatric disorders, with the capacity to impact susceptibility to disease as well as long-term neurobiological and behavioral outcomes. Parvalbumin (PV) interneurons, the most prominent subtype of GABAergic interneurons in the cortex, are uniquely responsive to stress due to their protracted development throughout the highly plastic neonatal period and into puberty and adolescence. Additionally, PV + interneurons appear to respond to stress in a sex-specific manner. This review aims to discuss existing preclinical studies that support our overall hypothesis that the sex-and age-specific impacts of stress on PV + interneurons contribute to differences in individual vulnerability to stress across the lifespan, particularly in regard to sex differences in the diagnostic rate of neurodevelopmental and neuropsychiatric diseases in clinical populations. We also emphasize the importance of studying sex as a biological variable to fully understand the mechanistic and behavioral differences between males and females in models of neuropsychiatric disease.
Collapse
Affiliation(s)
- Emma M Woodward
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States
| | - Laurence Coutellier
- Department of Neuroscience, Ohio State University, 255 Institute for Behavioral Medicine Research Building, 460 Medical Center Drive, Columbus, OH, 43210, United States; Department of Psychology, Ohio State University, 53 Psychology Building, 1835 Neil Avenue, Columbus, OH, 43210, United States.
| |
Collapse
|
97
|
Atefimanash P, Pourhamzeh M, Susanabadi A, Arabi M, Jamali-Raeufy N, Mehrabi S. Hippocampal chloride transporter KCC2 contributes to excitatory GABA dysregulation in the developmental rat model of schizophrenia. J Chem Neuroanat 2021; 118:102040. [PMID: 34695562 DOI: 10.1016/j.jchemneu.2021.102040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Recent studies have revealed an altered expression of NKCC1 and KCC2 in prefrontal cortex (PFC) and hippocampus of schizophrenic patients. Despite extensive considerations, the alteration of NKCC1 and KCC2 co-transporters at different stages of development has not been fully studied. Therefore, we evaluated the expression of these transporters in PFC and hippocampus at time points of four, eight, and twelve weeks in post-weaning social isolation rearing rat model. For this purpose, 23-25 days-old rats were classified into social- or isolation-reared groups. The levels of NKCC1 and KCC2 mRNA expression were evaluated at hippocampus or PFC regions at the time-points of four, eight, and twelve weeks following housing. Post-weaning isolation rearing decreased the hippocampal KCC2 mRNA expression level, but does not affect the NKCC1 mRNA expression. However, no significant difference was observed in the PFC mRNA levels of NKCC1 and KCC2 in the isolation-reared group compared to the socially-reared group during the course of modeling. Further, we assessed the therapeutic effect of selective NKCC1 inhibitor bumetanide (10 mg/kg), on improvement of prepulse inhibition (PPI) test on twelve weeks isolation-reared rats. Intraperitoneal administration of bumetanide (10 mg/kg) did not exert beneficial effects on PPI deficit. Our findings show that isolation rearing reduces hippocampal KCC2 expression level and may underlie hippocampal GABA excitatory. In addition, 10 mg/kg bumetanide is not effective in improving the reduced PPI of twelve weeks isolation-reared rats. Collectively, our findings show that hippocampal chloride transporter KCC2 contributes to excitatory GABA dysregulation in the developmental rat model of schizophrenia.
Collapse
Affiliation(s)
- Pezhman Atefimanash
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Susanabadi
- Department of Anesthesia and pain medicine, Arak University of Medical Sciences, Arak, Iran
| | - Mehrnoosh Arabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Radiology and Medical Physics, Faculty of Paramedicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Nida Jamali-Raeufy
- Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Soraya Mehrabi
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Physiology, Faculty of Medicine, Iran University of Medical Science, Tehran, Iran.
| |
Collapse
|
98
|
Chen BW, Zhang KW, Chen SJ, Yang C, Li PG. Vitamin A Deficiency Exacerbates Gut Microbiota Dysbiosis and Cognitive Deficits in Amyloid Precursor Protein/Presenilin 1 Transgenic Mice. Front Aging Neurosci 2021; 13:753351. [PMID: 34790112 PMCID: PMC8591312 DOI: 10.3389/fnagi.2021.753351] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/04/2021] [Indexed: 01/17/2023] Open
Abstract
Vitamin A deficiency (VAD) plays an essential role in the pathogenesis of Alzheimer’s disease (AD). However, the specific mechanism by which VAD aggravates cognitive impairment is still unknown. At the intersection of microbiology and neuroscience, the gut-brain axis is undoubtedly contributing to the formation and function of neurological systems, but most of the previous studies have ignored the influence of gut microbiota on the cognitive function in VAD. Therefore, we assessed the effect of VAD on AD pathology and the decline of cognitive function in AD model mice and determined the role played by the intestinal microbiota in the process. Twenty 8-week-old male C57BL/6J amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice were randomly assigned to either a vitamin A normal (VAN) or VAD diet for 45 weeks. Our results show that VAD aggravated the behavioral learning and memory deficits, reduced the retinol concentration in the liver and the serum, decreased the transcription of vitamin A (VA)-related receptors and VA-related enzymes in the cortex, increased amyloid-β peptides (Aβ40 and Aβ42) in the brain and gut, upregulate the translation of beta-site APP-cleaving enzyme 1 (BACE1) and phosphorylated Tau in the cortex, and downregulate the expression of brain-derived neurotrophic factor (BDNF) and γ-aminobutyric acid (GABA) receptors in the cortex. In addition, VAD altered the composition and functionality of the fecal microbiota as exemplified by a decreased abundance of Lactobacillus and significantly different α- and β-diversity. Of note, the functional metagenomic prediction (PICRUSt analysis) indicated that GABAergic synapse and retinol metabolism decreased remarkably after VAD intervention, which was in line with the decreased expression of GABA receptors and the decreased liver and serum retinol. In summary, the present study provided valuable facts that VAD exacerbated the morphological, histopathological, molecular biological, microbiological, and behavioral impairment in the APP/PS1 transgenic mice, and the intestinal microbiota may play a key mediator role in this mechanism.
Collapse
Affiliation(s)
- Bo-Wen Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Kai-Wen Zhang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Si-Jia Chen
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Chun Yang
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Peng-Gao Li
- School of Public Health, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Environmental Toxicology, Beijing, China.,Beijing Key Laboratory of Clinical Epidemiology, Beijing, China
| |
Collapse
|
99
|
Han D, Sun D, Xiu M, Su X, Wang J, Li J, Wang D. Association between the improvement in depressive symptoms and serum BDNF levels in drug-naive first episode patients with schizophrenia: A longitudinal follow-up. Psychoneuroendocrinology 2021; 133:105392. [PMID: 34428640 DOI: 10.1016/j.psyneuen.2021.105392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/13/2021] [Accepted: 08/15/2021] [Indexed: 11/28/2022]
Abstract
Depressive symptoms are frequent clinical manifestations in patients with schizophrenia. Decreased brain-derived neurotrophic factor (BDNF) has been shown to be involved in the development of depressive symptoms. However, the detailed molecular mechanism of BDNF in the depressive symptoms of schizophrenia patients remains to be fully elucidated. This study aimed to investigate the role of BDNF in the depressive symptoms in drug-naïve first-episode (DNFE) patients with schizophrenia and whether BDNF levels were associated with the improvement of depressive symptoms after olanzapine treatment. 50 DNFE schizophrenia patients and 55 healthy controls were recruited, and their serum BDNF levels were compared. All patients were treated with olanzapine monotherapy for 12 weeks, and 45 patients completed the trial. The serum BDNF levels and depressive symptoms were measured again at follow-up. We found that DNFE patients had lower BDNF levels, compared to controls. Last observation carried forward (LOCF) analysis was used for patients who dropped out after the second month, and 50 patients were included in the statistical analysis with LOCF. After 12 weeks of treatment with olanzapine, BDNF levels were significantly increased and depressive symptoms were significantly decreased. Correlation analysis showed that the change of BDNF levels after treatment was correlated with the change of HAMD total score from baseline. Further regression analysis showed that the change in BDNF levels was an independent predictor for the improvement in depressive symptoms, after controlling age, BMI change and the decrease of PANSS total score. However, the baseline BDNF levels were not associated with an improvement in depressive symptoms in patients. Our findings reveal that olanzapine treatment can increase BDNF levels and improve depressive symptoms in schizophrenia patients. Moreover, the changes in serum BDNF levels were related to the improvement in depressive symptoms.
Collapse
Affiliation(s)
- Dongmei Han
- Hebei Province Veterans Hospital, Baoding, China
| | - Daliang Sun
- Department of Psychiatry, Tianjin Anding Hospital, Tianjin Mental Health Center, Tianjin, China
| | - Meihong Xiu
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Xiu Su
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Jun Wang
- Peking University HuiLongGuan Clinical Medical School, Beijing HuiLongGuan Hospital, Beijing, China
| | - Jun Li
- Shanghai Xuhui District Mental Health Center, Shanghai, China.
| | - Dong Wang
- Department of Clinical Psychology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
100
|
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
|