1
|
Leman DP, Cary BA, Bissen D, Lane BJ, Shanley MR, Wong NF, Bhut KB, Turrigiano GG. Rapid prey capture learning drives a slow resetting of network activity in rodent binocular visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.11.648036. [PMID: 40291689 PMCID: PMC12027325 DOI: 10.1101/2025.04.11.648036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Neocortical neurons possess stable firing rate set points to which they faithfully return when perturbed. These set points are established early and are stable through adulthood, suggesting they are immutable. Here we challenge this idea using an ethological vision-dependent prey capture learning paradigm in juvenile rats. This learning required visual cortex (V1), and enhanced tuning of V1 neurons to specific behavioral epochs. Chronic recordings revealed a slow, state-dependent increase in V1 firing that began after learning was complete and persisted for days. This upward firing rate plasticity was gradual, gated by wake states, and in L2/3 was driven by a TNFα-dependent increase in excitatory synapses onto pyramidal neurons - all features of homeostatic plasticity within V1. Finally, TNFα inhibition after learning reduced retention of hunting skills. Thus, naturalistic learning in juvenile animals co-opts homeostatic forms of plasticity to reset firing rate setpoints within V1, in a process that facilitates skill consolidation.
Collapse
|
2
|
Wen W, Prada AM, Turrigiano GG. Modular Arrangement of Synaptic and Intrinsic Homeostatic Plasticity within Visual Cortical Circuits. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.01.596982. [PMID: 38853882 PMCID: PMC11160741 DOI: 10.1101/2024.06.01.596982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Neocortical circuits use synaptic and intrinsic forms of homeostatic plasticity to stabilize key features of network activity, but whether these different homeostatic mechanisms act redundantly, or can be independently recruited to stabilize different network features, is unknown. Here we used pharmacological and genetic perturbations both in vitro and in vivo to determine whether synaptic scaling and intrinsic homeostatic plasticity (IHP) are arranged and recruited in a hierarchical or modular manner within L2/3 pyramidal neurons in rodent V1. Surprisingly, although the expression of synaptic scaling and IHP was dependent on overlapping signaling pathways, they could be independently recruited by manipulating spiking activity or NMDAR signaling, respectively. Further, we found that changes in visual experience that affect NMDAR activation but not mean firing selectively trigger IHP, without recruiting synaptic scaling. These findings support a modular model in which synaptic and intrinsic homeostatic plasticity respond to and stabilize distinct aspects of network activity.
Collapse
Affiliation(s)
- Wei Wen
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Adriana M. Prada
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | | |
Collapse
|
3
|
Bissen D, Cary BA, Zhang A, Sailor KA, Van Hooser SD, Turrigiano GG. Prey capture learning drives critical period-specific plasticity in mouse binocular visual cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635373. [PMID: 39975102 PMCID: PMC11838381 DOI: 10.1101/2025.01.28.635373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Critical periods are developmental windows of high experience-dependent plasticity essential for the correct refinement of neuronal circuitry and function. While the consequences for the visual system of sensory deprivation during the critical period have been well-characterized, far less is known about the effects of enhanced sensory experience. Here, we use prey capture learning to assess structural and functional plasticity mediating visual learning in the primary visual cortex of critical period mice. We show that prey capture learning improves temporal frequency discrimination and drives a profound remodeling of visual circuitry through an increase in excitatory connectivity and spine turnover. This global and persistent rewiring is not observed in adult hunters and is mediated by TNFα-dependent mechanisms. Our findings demonstrate that enhanced visual experience in a naturalistic paradigm during the critical period can drive structural plasticity to improve visual function, and promotes a long-lasting increase in spine dynamics that could enhance subsequent plasticity.
Collapse
Affiliation(s)
- Diane Bissen
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Brian A Cary
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Amanda Zhang
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Kurt A Sailor
- Institut Pasteur, CNRS UMR 3571, Perception and Action Unit, F-75015, Paris, France
| | | | - Gina G Turrigiano
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
- Lead Contact
| |
Collapse
|
4
|
Álvarez-López AI, Cruz-Chamorro I, Lardone PJ, Bejarano I, Aspiazu-Hinostroza K, Ponce-España E, Santos-Sánchez G, Álvarez-Sánchez N, Carrillo-Vico A. Melatonin, an Antitumor Necrosis Factor Therapy. J Pineal Res 2025; 77:e70025. [PMID: 39740227 DOI: 10.1111/jpi.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Tumor necrosis factor (TNF) is a biomarker of inflammation whose levels are elevated in patients with several diseases associated with dysregulation of the immune response. The main limitations of currently used anti-TNF therapies are the induction of immunodepression, which in many cases leads to serious adverse effects such as infection and cancer, and the inability to cross the blood-brain barrier in neuroinflammatory conditions. Melatonin, in addition to being a chronobiotic compound, is widely known for its antioxidant and immunomodulatory capacity to control inflammatory processes in different pathological contexts. The aim of the present review is to address human-based studies that describe the effect of melatonin on TNF production. The review includes all the articles published in PubMed databases until April 15, 2024. After depuration, 45 studies were finally included in the review, 23 related to the in vitro action of melatonin in human cells and 22 in vivo studies in humans. Most of the data reviewed support the idea that melatonin has an immunosuppressive effect on TNF levels, which, together with its low toxicity profile, low cost, and ability to cross the blood-brain barrier, points to melatonin as a potential anti-TNF therapy. Therefore, improving our knowledge of the action of melatonin in regulating TNF through appropriate clinical trials would reveal the true potential of this molecule as a possible anti-TNF therapy.
Collapse
Grants
- This work was supported by the Andalusian Government Ministry of Health PC-0019-2017, PI-0015-2018 and PEMP-0085-2020 (co-financed with FEDER funds, call Resolution of 7 July 2021 of the General Secretary for Research, Development and Innovation in Health, which calls for grants to finance research, development and innovation in biomedicine and health sciences in Andalusia by 2021), the PAIDI Program from the Andalusian Government (CTS160) and Regional Ministry of Economy and Knowledge of Andalusia (US-1263804) into the European Regional Development Fund Operational Programme 2014 to 2020. A.I.A.L. was supported by grants US-1263804 and PEMP-0085-2020. I.C.C. was supported by a postdoctoral fellowship from the Andalusian Government Ministry of Economy, Knowledge, Business, and University (DOC_00587/2020). I.B. and E.P.E were supported by the VI Program of Inner Initiative for Research and Transfer of the University of Seville [VI PPIT-US]. G.S.S. was supported by a FPU grant from the Spanish Ministerio de Educación, Cultura y Deporte (FPU16/02339). N.A.-S. was supported by a fellowship from the Andalusian Regional Ministry of Health (PC-0111-2016-0111).
Collapse
Affiliation(s)
- Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Ignacio Bejarano
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Karla Aspiazu-Hinostroza
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Universidad Católica de Cuenca, Research Department, Cuenca-Azuay, Ecuador
| | - Eduardo Ponce-España
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Nuria Álvarez-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
5
|
Kwon HJ, Santhosh D, Huang Z. A novel monomeric amyloid β-activated signaling pathway regulates brain development via inhibition of microglia. eLife 2024; 13:RP100446. [PMID: 39635981 PMCID: PMC11620749 DOI: 10.7554/elife.100446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Amyloid β (Aβ) forms aggregates in the Alzheimer's disease brain and is well known for its pathological roles. Recent studies show that it also regulates neuronal physiology in the healthy brain. Whether Aβ also regulates glial physiology in the normal brain, however, has remained unclear. In this article, we describe the discovery of a novel signaling pathway activated by the monomeric form of Aβ in vitro that plays essential roles in the regulation of microglial activity and the assembly of neocortex during mouse development in vivo. We find that activation of this pathway depends on the function of amyloid precursor and the heterotrimeric G protein regulator Ric8a in microglia and inhibits microglial immune activation at transcriptional and post-transcriptional levels. Genetic disruption of this pathway during neocortical development results in microglial dysregulation and excessive matrix proteinase activation, leading to basement membrane degradation, neuronal ectopia, and laminar disruption. These results uncover a previously unknown function of Aβ as a negative regulator of brain microglia and substantially elucidate the underlying molecular mechanisms. Considering the prominence of Aβ and neuroinflammation in the pathology of Alzheimer's disease, they also highlight a potentially overlooked role of Aβ monomer depletion in the development of the disease.
Collapse
Affiliation(s)
- Hyo Jun Kwon
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Devi Santhosh
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| | - Zhen Huang
- Departments of Neurology and Neuroscience, University of Wisconsin-MadisonMadisonUnited States
| |
Collapse
|
6
|
Martinez-Navarrete G, Castaño-Castaño S, Morales-Navas M, Nieto-Escámez F, Sánchez-Santed F, Fernandez E. Impact of transcranial Direct Current Stimulation on stereoscopic vision and retinal structure in adult amblyopic rodents. Eye Brain 2024; 16:75-88. [PMID: 39498234 PMCID: PMC11533879 DOI: 10.2147/eb.s474573] [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: 04/19/2024] [Accepted: 09/23/2024] [Indexed: 11/07/2024] Open
Abstract
Purpose The impact of visual deprivation on retinal structure is widely debated. Experimental models, like monocular deprivation through lid suture, provide insights into the consequences of lacking visual experience during development. This deprivation delays primary visual cortex (CV1) maturation due to improper neural connection consolidation, which remains plastic beyond the critical period. However, few studies have used Optical Coherence Tomography (OCT) to investigate structural alterations in the retina of animal models following monocular deprivation. Instead, some studies have focused on the ganglion cell layer using post-mortem histological techniques in amblyopia models induced by monocular deprivation. Methods In this study, we used Cliff test to assess stereoscopic vision and spectral domain optical coherence tomography (SD-OCT) to evaluate retinal changes in an in vivo model of visual deprivation treated with Transcranial Direct Current Stimulation (tDCS). Results The depth perception test initially revealed differences between individuals with amblyopia and the control group. However, after 8 tDCS sessions, amblyopic subjects matched the control group's performance, which remained stable Additionally, significant changes were observed in retinal structures post-tDCS treatment. Specifically, the thickness of the Nerve Fiber Layer + Ganglion Cell Layer + Inner Plexiform Layer (NFL+GCL+IPL) increased significantly in amblyopic eyes (p<0.001). Moreover, significant retinal thickening, including the Nerve Fiber Layer + Ganglion Cell Layer + Inner Plexiform Layer (NFL+GCL+IPL) and the entire retina, was observed post-tDCS treatment (p<0.05), highlighting the critical role of tDCS in ameliorating amblyopia. Additionally, treated animals exhibited reduced thickness in the Inner Nuclear Layer (INL) and Outer Nuclear Layer (ONL). Conclusion tDCS treatment effectively restores amblyopic individuals' stereoscopic vision, aligning their performance with controls, while impacting retinal structure, highlighting its potential in ameliorating amblyopia's visual deficits.
Collapse
Affiliation(s)
- Gema Martinez-Navarrete
- Neuroprosthesis and Neuroengineering Research Group, Institute of Bioengineering (IB), University Miguel Hernández (UMH), Elche, 03020, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Sergio Castaño-Castaño
- Department Psychology, University of Oviedo, Plaza Feijoo S/N, Oviedo, 33003, Spain
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), Oviedo, Spain
- Institute of Health Research of the Principality of Asturias (ISPA), Oviedo, Spain
| | - Miguel Morales-Navas
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Francisco Nieto-Escámez
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
- Neuropsychological Evaluation and Rehabilitation Center (CERNEP), Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Fernando Sánchez-Santed
- Department of Psychology, University of Almería, Ctra. Sacramento S/N, La Cañada de San Urbano, Almería, 04120, Spain
| | - Eduardo Fernandez
- Neuroprosthesis and Neuroengineering Research Group, Institute of Bioengineering (IB), University Miguel Hernández (UMH), Elche, 03020, Spain
- Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
7
|
Bridi MCD, Hong S, Severin D, Moreno C, Contreras A, Kirkwood A. Blockade of GluN2B-Containing NMDA Receptors Prevents Potentiation and Depression of Responses during Ocular Dominance Plasticity. J Neurosci 2024; 44:e0021232024. [PMID: 39117456 PMCID: PMC11376332 DOI: 10.1523/jneurosci.0021-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 07/03/2024] [Accepted: 07/27/2024] [Indexed: 08/10/2024] Open
Abstract
Monocular deprivation (MD) causes an initial decrease in synaptic responses to the deprived eye in juvenile mouse primary visual cortex (V1) through Hebbian long-term depression (LTD). This is followed by a homeostatic increase, which has been attributed either to synaptic scaling or to a slide threshold for Hebbian long-term potentiation (LTP) rather than scaling. We therefore asked in mice of all sexes whether the homeostatic increase during MD requires GluN2B-containing NMDA receptor activity, which is required to slide the plasticity threshold but not for synaptic scaling. Selective GluN2B blockade from 2-6 d after monocular lid suture prevented the homeostatic increase in miniature excitatory postsynaptic current (mEPSC) amplitude in monocular V1 of acute slices and prevented the increase in visually evoked responses in binocular V1 in vivo. The decrease in mEPSC amplitude and visually evoked responses during the first 2 d of MD also required GluN2B activity. Together, these results support the idea that GluN2B-containing NMDA receptors first play a role in LTD immediately following eye closure and then promote homeostasis during prolonged MD by sliding the plasticity threshold in favor of LTP.
Collapse
Affiliation(s)
- Michelle C D Bridi
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Su Hong
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Daniel Severin
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Cristian Moreno
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Altagracia Contreras
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| | - Alfredo Kirkwood
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, Maryland 21218
| |
Collapse
|
8
|
Wen W, Turrigiano GG. Keeping Your Brain in Balance: Homeostatic Regulation of Network Function. Annu Rev Neurosci 2024; 47:41-61. [PMID: 38382543 DOI: 10.1146/annurev-neuro-092523-110001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
To perform computations with the efficiency necessary for animal survival, neocortical microcircuits must be capable of reconfiguring in response to experience, while carefully regulating excitatory and inhibitory connectivity to maintain stable function. This dynamic fine-tuning is accomplished through a rich array of cellular homeostatic plasticity mechanisms that stabilize important cellular and network features such as firing rates, information flow, and sensory tuning properties. Further, these functional network properties can be stabilized by different forms of homeostatic plasticity, including mechanisms that target excitatory or inhibitory synapses, or that regulate intrinsic neuronal excitability. Here we discuss which aspects of neocortical circuit function are under homeostatic control, how this homeostasis is realized on the cellular and molecular levels, and the pathological consequences when circuit homeostasis is impaired. A remaining challenge is to elucidate how these diverse homeostatic mechanisms cooperate within complex circuits to enable them to be both flexible and stable.
Collapse
Affiliation(s)
- Wei Wen
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| | - Gina G Turrigiano
- Department of Biology, Brandeis University, Waltham, Massachusetts, USA;
| |
Collapse
|
9
|
Grieco M, Giorgi A, Giacovazzo G, Maggiore A, Ficchì S, d'Erme M, Mosca L, Mignogna G, Maras B, Coccurello R. β-Hexachlorocyclohexane triggers neuroinflammatory activity, epigenetic histone post-translational modifications and cognitive dysfunction. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 279:116487. [PMID: 38810285 DOI: 10.1016/j.ecoenv.2024.116487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/06/2024] [Accepted: 05/18/2024] [Indexed: 05/31/2024]
Abstract
Persistent organic pollutants (POPs), which encompass pesticides and industrial chemicals widely utilized across the globe, pose a covert threat to human health. β-hexachlorocyclohexane (β-HCH) is an organochlorine pesticide with striking stability, still illegally dumped in many countries, and recognized as responsible for several pathogenetic mechanisms. This study represents a pioneering exploration into the neurotoxic effects induced by the exposure to β-HCH specifically targeting neuronal cells (N2a), microglia (BV-2), and C57BL/6 mice. As shown by western blot and qPCR analyses, the administration of β-HCH triggered a modulation of NF-κB, a key factor influencing both inflammation and pro-inflammatory cytokines expression. We demonstrated by proteomic and western blot techniques epigenetic modifications in H3 histone induced by β-HCH. Histone acetylation of H3K9 and H3K27 increased in N2a, and in the prefrontal cortex of C57BL/6 mice administered with β-HCH, whereas it decreased in BV-2 cells and in the hippocampus. We also observed a severe detrimental effect on recognition memory and spatial navigation by the Novel Object Recognition Test (NORT) and the Object Place Recognition Task (OPRT) behavioural tests. Cognitive impairment was linked to decreased expression of the genes BDNF and SNAP-25, which are mediators involved in synaptic function and activity. The obtained results expand our understanding of the harmful impact produced by β-HCH exposure by highlighting its implication in the pathogenesis of neurological diseases. These findings will support intervention programs to limit the risk induced by exposure to POPs. Regulatory agencies should block further illicit use, causing environmental hazards and endangering human and animal health.
Collapse
Affiliation(s)
- Maddalena Grieco
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Alessandra Giorgi
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Giacomo Giacovazzo
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Anna Maggiore
- Department of Biochemical Sciences, Sapienza University, Rome, Italy; Department of Brain Sciences, Imperial College, London, UK
| | - Serena Ficchì
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Maria d'Erme
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | - Luciana Mosca
- Department of Biochemical Sciences, Sapienza University, Rome, Italy
| | | | - Bruno Maras
- Department of Biochemical Sciences, Sapienza University, Rome, Italy.
| | - Roberto Coccurello
- European Center for Brain Research, Santa Lucia Foundation IRCCS, Rome, Italy; Institute for Complex Systems, National Research Council (CNR), Roma, Italy
| |
Collapse
|
10
|
Kaneko M, Hoseini MS, Waschek JA, Stryker MP. Stimulus-specific enhancement in mouse visual cortex requires GABA but not VIP-peptide release from VIP interneurons. J Neurophysiol 2024; 132:34-44. [PMID: 38774975 PMCID: PMC11383382 DOI: 10.1152/jn.00463.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 06/26/2024] Open
Abstract
When adult mice are repeatedly exposed to a particular visual stimulus for as little as 1 h per day for several days while their visual cortex (V1) is in the high-gain state produced by locomotion, that specific stimulus elicits much stronger responses in V1 neurons for the following several weeks, even when measured in anesthetized animals. Such stimulus-specific enhancement (SSE) is not seen if locomotion is prevented. The effect of locomotion on cortical responses is mediated by vasoactive intestinal peptide (VIP) positive interneurons, which can release both the peptide and the inhibitory neurotransmitter GABA. Previous studies have examined the role of VIP-ergic interneurons, but none have distinguished the individual roles of peptide from GABA release. Here, we used genetic ablation to determine which of those molecules secreted by VIP-ergic neurons is responsible for SSE. SSE was not impaired by VIP deletion but was prevented by compromising release of GABA from VIP cells. This finding suggests that SSE may result from Hebbian mechanisms that remain present in adult V1.NEW & NOTEWORTHY Many neurons package and release a peptide along with a conventional neurotransmitter. The conventional view is that such peptides exert late, slow effects on plasticity. We studied a form of cortical plasticity that depends on the activity of neurons that express both vasoactive intestinal peptide (VIP) and the inhibitory neurotransmitter GABA. GABA release accounted for their action on plasticity, with no effect of deleting the peptide on this phenomenon.
Collapse
Affiliation(s)
- Megumi Kaneko
- Department of Physiology and Kavli Institute For Fundamental Neuroscience, University of California San Francisco, San Francisco, California, United States
| | - Mahmood S Hoseini
- Department of Physiology and Kavli Institute For Fundamental Neuroscience, University of California San Francisco, San Francisco, California, United States
| | - James A Waschek
- Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Michael P Stryker
- Department of Physiology and Kavli Institute For Fundamental Neuroscience, University of California San Francisco, San Francisco, California, United States
| |
Collapse
|
11
|
Zhao Y, Ji G, Zhou S, Cai S, Li K, Zhang W, Zhang C, Yan N, Zhang S, Li X, Song B, Qu L. IGF2BP2-Shox2 axis regulates hippocampal-neuronal senescence to alleviate microgravity-induced recognition disturbance. iScience 2024; 27:109917. [PMID: 38812544 PMCID: PMC11134919 DOI: 10.1016/j.isci.2024.109917] [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: 01/22/2024] [Revised: 03/06/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
During space travel, microgravity leads to disturbances in cognitive function, while the underlying mechanism is still unclear. Simulated microgravity mice showed neuronal age-like changes in the hippocampus of our study. In the context of microgravity, we discovered m6A modification reshapes in the hippocampal region. When paired with RNA-seq and MeRIP-seq, Shox2 was found to be a powerful regulator in hippocampal neuron that respondes to microgravity. Decreased expression of senescence-associated secretory phenotype factors and improved genes related to synapses led to the restoration of memory function in the hippocampus upon increased expression of Shox2. Moreover, we discovered that IGF2BP2 was required for the m6A modification of the Shox2, and overexpressed IGF2BP2 in the hippocampus protected against both neuronal senescence and learning and memory decline caused by loss of gravity. Accordingly, our research identified the hippocampal IGF2BP2-Shox2 axis as a possible therapeutic approach to maintaining cognitive function during space travel.
Collapse
Affiliation(s)
- Yujie Zhao
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Guohua Ji
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Sihai Zhou
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Shiou Cai
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Kai Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Wanyu Zhang
- Basic Medical Sciences, Capital Medical University School, Beijing, China
| | - Chuanjie Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Na Yan
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Shuhui Zhang
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Xiaopeng Li
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| | - Bo Song
- Department of Pathology and Forensics, Dalian Medical University, Dalian, China
| | - Lina Qu
- State Key Laboratory of Space Medicine, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
12
|
Sha C, Van Brunt T, Kudria J, Schmidt D, Yurovsky A, Bandovic J, Giarrizzo M, Lin J, Tsirka SA, Bialkowska AB, Wollmuth L, Speer E, Hsieh H. A graded neonatal mouse model of necrotizing enterocolitis demonstrates that mild enterocolitis is sufficient to activate microglia and increase cerebral cytokine expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.03.551849. [PMID: 38746118 PMCID: PMC11092491 DOI: 10.1101/2023.08.03.551849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal process that afflicts approximately 10% of preterm infants born in the United States each year, with a mortality rate of 30%. NEC severity is graded using Bell's classification system, from stage I mild NEC to stage III severe NEC. Over half of NEC survivors present with neurodevelopmental impairment during adolescence, a long-term complication that is poorly understood but can occur even after mild NEC. Although multiple animal models exist, none allow the experimenter to control nor represent the gradient of symptom severities seen in NEC patients. We bridge this knowledge gap by developing a graded murine model of NEC and studying its relationship with neuroinflammation across a range of NEC severities. Methods Postnatal day 3 (P3) C57BL/6 mice were fed a formula containing different concentrations (0% control, 0.25%, 1%, 2%, and 3%) of dextran sodium sulfate (DSS). P3 mice were fed every 3 hours for 72-hours. We collected data on weight gain and behavior (activity, response, body color) during feeding. At the end of the experiment, we collected tissues (intestine, liver, plasma, brain) for immunohistochemistry, immunofluorescence, and cytokine and chemokine analysis. Results Throughout NEC induction, mice fed higher concentrations of DSS died sooner, lost weight faster, and became sick or lethargic earlier. Intestinal characteristics (dilation, color, friability) were worse in mice fed with higher DSS concentrations. Histology revealed small intestinal disarray among mice fed all DSS concentrations, while higher DSS concentrations resulted in reduced small intestinal cellular proliferation and increased hepatic and systemic inflammation. In the brain, IL-2, G-CSF, and CXCL1 concentrations increased with higher DSS concentrations. Although the number of neurons and microglia in the CA1 hippocampal region did not differ, microglial branching was significantly reduced in DSS-fed mice. Conclusion We characterize a novel graded model of NEC that recapitulates the full range of NEC severities. We show that mild NEC is sufficient to initiate neuroinflammation and microglia activation. This model will facilitate studies on the neurodevelopmental effects of NEC.
Collapse
|
13
|
Heir R, Abbasi Z, Komal P, Altimimi HF, Franquin M, Moschou D, Chambon J, Stellwagen D. Astrocytes Are the Source of TNF Mediating Homeostatic Synaptic Plasticity. J Neurosci 2024; 44:e2278222024. [PMID: 38395613 PMCID: PMC10993029 DOI: 10.1523/jneurosci.2278-22.2024] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Tumor necrosis factor α (TNF) mediates homeostatic synaptic plasticity (HSP) in response to chronic activity blockade, and prior work has established that it is released from glia. Here we demonstrate that astrocytes are the necessary source of TNF during HSP. Hippocampal cultures from rats of both sexes depleted of microglia still will increase TNF levels following activity deprivation and still express TTX-driven HSP. Slice cultures from mice of either sex with a conditional deletion of TNF from microglia also express HSP, but critically, slice cultures with a conditional deletion of TNF from astrocytes do not. In astrocytes, glutamate signaling is sufficient to reduce NFκB signaling and TNF mRNA levels. Further, chronic TTX treatment increases TNF in an NFκB-dependent manner, although NFκB signaling is dispensable for the neuronal response to TTX-driven HSP. Thus, astrocytes can sense neuronal activity through glutamate spillover and increase TNF production when activity falls, to drive HSP through the production of TNF.
Collapse
Affiliation(s)
- Renu Heir
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Zahra Abbasi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Pragya Komal
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Haider F Altimimi
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Marie Franquin
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Dionysia Moschou
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - Julien Chambon
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| | - David Stellwagen
- Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Research Institute of the McGill University Health Center, Montréal, Quebec H3G 1A4, Canada
| |
Collapse
|
14
|
Shen J, Bian N, Zhao L, Wei J. The role of T-lymphocytes in central nervous system diseases. Brain Res Bull 2024; 209:110904. [PMID: 38387531 DOI: 10.1016/j.brainresbull.2024.110904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/04/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
The central nervous system (CNS) has been considered an immunologically privileged site. In the past few decades, research on inflammation in CNS diseases has mostly focused on microglia, innate immune cells that respond rapidly to injury and infection to maintain CNS homeostasis. Discoveries of lymphatic vessels within the dura mater and peripheral immune cells in the meningeal layer indicate that the peripheral immune system can monitor and intervene in the CNS. This review summarizes recent advances in the involvement of T lymphocytes in multiple CNS diseases, including brain injury, neurodegenerative diseases, and psychiatric disorders. It emphasizes that a deep understanding of the pathogenesis of CNS diseases requires intimate knowledge of T lymphocytes. Aiming to promote a better understanding of the relationship between the immune system and CNS and facilitate the development of therapeutic strategies targeting T lymphocytes in neurological diseases.
Collapse
Affiliation(s)
- Jianing Shen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Ning Bian
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China
| | - Lu Zhao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China.
| | - Jingkuan Wei
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan 650500, China; Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan 650500, China.
| |
Collapse
|
15
|
Gargas J, Janowska J, Gebala P, Maksymiuk W, Sypecka J. Reactive Gliosis in Neonatal Disorders: Friend or Foe for Neuroregeneration? Cells 2024; 13:131. [PMID: 38247822 PMCID: PMC10813898 DOI: 10.3390/cells13020131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
A developing nervous system is particularly vulnerable to the influence of pathophysiological clues and injuries in the perinatal period. Astrocytes are among the first cells that react to insults against the nervous tissue, the presence of pathogens, misbalance of local tissue homeostasis, and a lack of oxygen and trophic support. Under this background, it remains uncertain if induced astrocyte activation, recognized as astrogliosis, is a friend or foe for progressing neonatal neurodevelopment. Likewise, the state of astrocyte reactivity is considered one of the key factors discriminating between either the initiation of endogenous reparative mechanisms compensating for aberrations in the structures and functions of nervous tissue or the triggering of neurodegeneration. The responses of activated cells are modulated by neighboring neural cells, which exhibit broad immunomodulatory and pro-regenerative properties by secreting a plethora of active compounds (including interleukins and chemokines, neurotrophins, reactive oxygen species, nitric oxide synthase and complement components), which are engaged in cell crosstalk in a paracrine manner. As the developing nervous system is extremely sensitive to the influence of signaling molecules, even subtle changes in the composition or concentration of the cellular secretome can have significant effects on the developing neonatal brain. Thus, modulating the activity of other types of cells and their interactions with overreactive astrocytes might be a promising strategy for controlling neonatal astrogliosis.
Collapse
Affiliation(s)
| | | | | | | | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, A. Pawinskiego 5, 02-106 Warsaw, Poland; (J.G.); (J.J.)
| |
Collapse
|
16
|
Bottorff J, Padgett S, Turrigiano GG. Basal forebrain cholinergic activity is necessary for upward firing rate homeostasis in the rodent visual cortex. Proc Natl Acad Sci U S A 2024; 121:e2317987121. [PMID: 38147559 PMCID: PMC10769829 DOI: 10.1073/pnas.2317987121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/22/2023] [Indexed: 12/28/2023] Open
Abstract
Bidirectional homeostatic plasticity allows neurons and circuits to maintain stable firing in the face of developmental or learning-induced perturbations. In the primary visual cortex (V1), upward firing rate homeostasis (FRH) only occurs during active wake (AW) and downward during sleep, but how this behavioral state-dependent gating is accomplished is unknown. Here, we focus on how AW enables upward FRH in V1 of juvenile Long Evans rats. A major difference between quiet wake (QW), when upward FRH is absent, and AW, when it is present, is increased cholinergic (ACh) tone, and the main cholinergic projections to V1 arise from the horizontal diagonal band of the basal forebrain (HDB ACh). We therefore chemogenetically inhibited HDB ACh neurons while inducing upward homeostatic compensation using direct activity-suppression in V1. We found that synaptic scaling up and intrinsic homeostatic plasticity, two important cellular mediators of upward FRH, were both impaired when HDB ACh neurons were inhibited. Most strikingly, HDB ACh inhibition flipped the sign of intrinsic plasticity so that it became anti-homeostatic, and this effect was phenocopied by knockdown of the M1 ACh receptor in V1, indicating that this modulation of intrinsic plasticity is the result of direct actions of ACh within V1. Finally, we found that upward FRH induced by visual deprivation was completely prevented by HDB ACh inhibition. Together, our results show that HDB ACh modulation is a key enabler of upward homeostatic plasticity and FRH, and more broadly suggest that neuromodulatory inputs can segregate upward and downward homeostatic plasticity into distinct behavioral states.
Collapse
Affiliation(s)
- Juliet Bottorff
- Department of Biology and Neuroscience Program, Brandeis University, Waltham, MA02453
| | - Sydney Padgett
- Department of Biology and Neuroscience Program, Brandeis University, Waltham, MA02453
| | - Gina G. Turrigiano
- Department of Biology and Neuroscience Program, Brandeis University, Waltham, MA02453
| |
Collapse
|
17
|
Radulescu CI, Doostdar N, Zabouri N, Melgosa-Ecenarro L, Wang X, Sadeh S, Pavlidi P, Airey J, Kopanitsa M, Clopath C, Barnes SJ. Age-related dysregulation of homeostatic control in neuronal microcircuits. Nat Neurosci 2023; 26:2158-2170. [PMID: 37919424 PMCID: PMC10689243 DOI: 10.1038/s41593-023-01451-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/06/2023] [Indexed: 11/04/2023]
Abstract
Neuronal homeostasis prevents hyperactivity and hypoactivity. Age-related hyperactivity suggests homeostasis may be dysregulated in later life. However, plasticity mechanisms preventing age-related hyperactivity and their efficacy in later life are unclear. We identify the adult cortical plasticity response to elevated activity driven by sensory overstimulation, then test how plasticity changes with age. We use in vivo two-photon imaging of calcium-mediated cellular/synaptic activity, electrophysiology and c-Fos-activity tagging to show control of neuronal activity is dysregulated in the visual cortex in late adulthood. Specifically, in young adult cortex, mGluR5-dependent population-wide excitatory synaptic weakening and inhibitory synaptogenesis reduce cortical activity following overstimulation. In later life, these mechanisms are downregulated, so that overstimulation results in synaptic strengthening and elevated activity. We also find overstimulation disrupts cognition in older but not younger animals. We propose that specific plasticity mechanisms fail in later life dysregulating neuronal microcircuit homeostasis and that the age-related response to overstimulation can impact cognitive performance.
Collapse
Affiliation(s)
- Carola I Radulescu
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Nazanin Doostdar
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Nawal Zabouri
- Department of Biomedical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Leire Melgosa-Ecenarro
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Xingjian Wang
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Sadra Sadeh
- Department of Biomedical Engineering, Imperial College London, South Kensington Campus, London, UK
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Pavlina Pavlidi
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
- Department of Pharmacology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Joe Airey
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK
| | | | - Claudia Clopath
- Department of Biomedical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Samuel J Barnes
- UK Dementia Research Institute, Department of Brain Sciences, Imperial College London, Hammersmith Hospital Campus, London, UK.
| |
Collapse
|
18
|
Li Y, Que M, Wang X, Zhan G, Zhou Z, Luo X, Li S. Exploring Astrocyte-Mediated Mechanisms in Sleep Disorders and Comorbidity. Biomedicines 2023; 11:2476. [PMID: 37760916 PMCID: PMC10525869 DOI: 10.3390/biomedicines11092476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Astrocytes, the most abundant cells in the brain, are integral to sleep regulation. In the context of a healthy neural environment, these glial cells exert a profound influence on the sleep-wake cycle, modulating both rapid eye movement (REM) and non-REM sleep phases. However, emerging literature underscores perturbations in astrocytic function as potential etiological factors in sleep disorders, either as protopathy or comorbidity. As known, sleep disorders significantly increase the risk of neurodegenerative, cardiovascular, metabolic, or psychiatric diseases. Meanwhile, sleep disorders are commonly screened as comorbidities in various neurodegenerative diseases, epilepsy, and others. Building on existing research that examines the role of astrocytes in sleep disorders, this review aims to elucidate the potential mechanisms by which astrocytes influence sleep regulation and contribute to sleep disorders in the varied settings of brain diseases. The review emphasizes the significance of astrocyte-mediated mechanisms in sleep disorders and their associated comorbidities, highlighting the need for further research.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Mengxin Que
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Xuan Wang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Zhiqiang Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyong Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China; (Y.L.); (M.Q.); (X.W.); (G.Z.); (Z.Z.)
| |
Collapse
|
19
|
Whitelaw BS, Stoessel MB, Majewska AK. Movers and shakers: Microglial dynamics and modulation of neural networks. Glia 2023; 71:1575-1591. [PMID: 36533844 PMCID: PMC10729610 DOI: 10.1002/glia.24323] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Microglia are multifaceted cells that act as immune sentinels, with important roles in pathological events, but also as integral contributors to the normal development and function of neural circuits. In the last decade, our understanding of the contributions these cells make to synaptic health and dysfunction has expanded at a dizzying pace. Here we review the known mechanisms that govern the dynamics of microglia allowing these motile cells to interact with synapses, and recruit microglia to specific sites on neurons. We then review the molecular signals that may underlie the function of microglia in synaptic remodeling. The emerging picture from the literature suggests that microglia are highly sensitive cells, reacting to neuronal signals with dynamic and specific actions tuned to the need of specific synapses and networks.
Collapse
Affiliation(s)
- Brendan Steven Whitelaw
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Mark Blohm Stoessel
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| | - Ania Katarzyna Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, New York, USA
| |
Collapse
|
20
|
Kaya ZB, Karakoc E, McLean PJ, Saka E, Atilla P. Post-inflammatory administration of N-acetylcysteine reduces inflammation and alters receptor levels in a cellular model of Parkinson's disease. FASEB Bioadv 2023; 5:263-276. [PMID: 37415931 PMCID: PMC10320847 DOI: 10.1096/fba.2022-00145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/10/2023] [Accepted: 04/28/2023] [Indexed: 07/08/2023] Open
Abstract
Parkinson's disease (PD) is a complex, multifactorial neurodegenerative disease with a prevalence of 1% over the age of 55. Neuropathological hallmarks of PD include the loss of dopaminergic neurons in the substantia nigra pars compacta and the accumulation of Lewy bodies that contain a variety of proteins and lipids including alpha-synuclein (α-syn). Although the formation of α-syn occurs intracellularly, it can also be found in the extracellular space where it can be taken up by neighboring cells. Toll-like receptor 2 (TLR2) is an immune system receptor that has been shown to recognize extracellular α-syn and modulate its uptake by other cells. Lymphocyte-activation gene 3 (LAG3), an immune checkpoint receptor, has also been proposed to play a role in extracellular α-syn internalization; however, a recent study has disputed this role. Internalized α-syn can trigger expression and secretion of inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, IL-2, and IL-6 and induce neuroinflammation, apoptosis, and mitophagy that results in cellular death. In this study, we tested if N-acetylcysteine (NAC), an anti-inflammatory and anti-carcinogenic drug, can circumvent the detrimental effects of neuroinflammation and induce an anti-inflammatory response by modulating transcription and expression of TLR2 and LAG3 receptors. Cells overexpressing wild-type α-syn were treated with TNF-α to induce inflammation followed by NAC to inhibit the deleterious effects of TNF-α-induced inflammation and apoptosis. SNCA gene transcription and α-syn protein expression were validated by q-PCR and Western blot (WB), respectively. Cell viability was measured, and apoptosis was evaluated by WB and terminal deoxynucleotidyl transferase nick end labeling methods. Alterations in LAG3 and TLR2 receptor levels were evaluated by immunofluorescent labeling, WB, and q-PCR. TNF-α not only increased inflammation but also increased endogenous and overexpressed α-syn levels. NAC treatment decreased expression of TLR2 and increased transcription of LAG3 receptor and diminished inflammation-mediated toxicity and cell death. Here, we demonstrate that NAC can reduce neuroinflammation that occurs as a result of alpha-synuclein overexpression, via a TLR2-associated pathway, making it a promising candidate for therapeutic intervention. Further studies are needed to elucidate molecular mechanisms and pathways related to neuroinflammation in PD and to develop possible new therapeutic approaches to slow the clinical progression of PD.
Collapse
Affiliation(s)
- Zeynep Bengisu Kaya
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Elif Karakoc
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| | | | - Esen Saka
- Department of NeurologyHacettepe University Faculty of MedicineAnkaraTurkey
| | - Pergin Atilla
- Department of Histology and EmbryologyHacettepe University Faculty of MedicineAnkaraTurkey
| |
Collapse
|
21
|
Wilmes KA, Clopath C. Dendrites help mitigate the plasticity-stability dilemma. Sci Rep 2023; 13:6543. [PMID: 37085642 PMCID: PMC10121616 DOI: 10.1038/s41598-023-32410-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/27/2023] [Indexed: 04/23/2023] Open
Abstract
With Hebbian learning 'who fires together wires together', well-known problems arise. Hebbian plasticity can cause unstable network dynamics and overwrite stored memories. Because the known homeostatic plasticity mechanisms tend to be too slow to combat unstable dynamics, it has been proposed that plasticity must be highly gated and synaptic strengths limited. While solving the issue of stability, gating and limiting plasticity does not solve the stability-plasticity dilemma. We propose that dendrites enable both stable network dynamics and considerable synaptic changes, as they allow the gating of plasticity in a compartment-specific manner. We investigate how gating plasticity influences network stability in plastic balanced spiking networks of neurons with dendrites. We compare how different ways to gate plasticity, namely via modulating excitability, learning rate, and inhibition increase stability. We investigate how dendritic versus perisomatic gating allows for different amounts of weight changes in stable networks. We suggest that the compartmentalisation of pyramidal cells enables dendritic synaptic changes while maintaining stability. We show that the coupling between dendrite and soma is critical for the plasticity-stability trade-off. Finally, we show that spatially restricted plasticity additionally improves stability.
Collapse
Affiliation(s)
- Katharina A Wilmes
- Imperial College London, London, United Kingdom.
- University of Bern, Bern, Switzerland.
| | | |
Collapse
|
22
|
Issa NP, Nunn KC, Wu S, Haider HA, Tao JX. Putative roles for homeostatic plasticity in epileptogenesis. Epilepsia 2023; 64:539-552. [PMID: 36617338 PMCID: PMC10015501 DOI: 10.1111/epi.17500] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/09/2023]
Abstract
Homeostatic plasticity allows neural circuits to maintain an average activity level while preserving the ability to learn new associations and efficiently transmit information. This dynamic process usually protects the brain from excessive activity, like seizures. However, in certain contexts, homeostatic plasticity might produce seizures, either in response to an acute provocation or more chronically as a driver of epileptogenesis. Here, we review three seizure conditions in which homeostatic plasticity likely plays an important role: acute drug withdrawal seizures, posttraumatic or disconnection epilepsy, and cyclic seizures. Identifying the homeostatic mechanisms active at different stages of development and in different circuits could allow better targeting of therapies, including determining when neuromodulation might be most effective, proposing ways to prevent epileptogenesis, and determining how to disrupt the cycle of recurring seizure clusters.
Collapse
Affiliation(s)
- Naoum P. Issa
- Comprehensive Epilepsy Center, Department of Neurology, 5841 S. Maryland Ave., MC 2030, University of Chicago, Chicago, IL 60637
| | | | - Shasha Wu
- Comprehensive Epilepsy Center, Department of Neurology, 5841 S. Maryland Ave., MC 2030, University of Chicago, Chicago, IL 60637
| | - Hiba A. Haider
- Comprehensive Epilepsy Center, Department of Neurology, 5841 S. Maryland Ave., MC 2030, University of Chicago, Chicago, IL 60637
| | - James X. Tao
- Comprehensive Epilepsy Center, Department of Neurology, 5841 S. Maryland Ave., MC 2030, University of Chicago, Chicago, IL 60637
| |
Collapse
|
23
|
Alomar HA, Nadeem A, Ansari MA, Attia SM, Bakheet SA, Al-Mazroua HA, Alhazzani K, Assiri MA, Alqinyah M, Almudimeegh S, Ahmad SF. Mitogen-activated protein kinase inhibitor PD98059 improves neuroimmune dysfunction in experimental autoimmune encephalomyelitis in SJL/J mice through the inhibition of nuclear factor-kappa B signaling in B cells. Brain Res Bull 2023; 194:45-53. [PMID: 36646144 DOI: 10.1016/j.brainresbull.2023.01.003] [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: 06/19/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Multiple sclerosis (MS) is a severe autoimmune disease leading to demyelination, followed by consequent axonal degeneration, causing sensory, motor, cognitive, and visual symptoms. Experimental autoimmune encephalomyelitis (EAE) is the most well-studied animal model of MS. Most current MS treatments are not completely effective, and severe side effects remain a great challenge. In this study, we report the therapeutic efficacy of PD98059, a potent mitogen-activated protein kinase inhibitor, on proteolipid protein (PLP)139-151-induced EAE in SJL/J mice. Following the induction of EAE, mice were intraperitoneally treated with PD98059 (5 mg/kg for 14 days) daily from day 14 to day 28. This study investigated the effects of PD98059 on C-C motif chemokine receptor 6 (CCR6), CD14, NF-κB p65, IκBα, GM-CSF, iNOS, IL-6, TNF-α in CD45R+ B lymphocytes using flow cytometry. Furthermore, we analyzed the effect of PD98059 on CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA and protein expression levels using qRT-PCR analysis in brain tissues. Mechanistic investigations revealed that PD98059-treated in mice with EAE had reduced CD45R+CCR6+, CD45R+CD14+, CD45R+NF-κB p65+, CD45R+GM-CSF+, CD45R+iNOS+, CD45R+IL-6+, and CD45R+TNF-α+ cells and increased CD45R+IκBα+ cells compared with vehicle-treated control mice in the spleen. Moreover, downregulation of CCR6, CD14, NF-κB p65, GM-CSF, iNOS, IL-6, and TNF-α mRNA expression level was observed in PD98059-treated mice with EAE compared with vehicle-treated control mice in the brain tissue. The results of this study demonstrate that PD98059 modulates inflammatory mediators through multiple cellular mechanisms. The results of this study suggest that PD98059 may be pursued as a therapeutic agent for the treatment of MS.
Collapse
Affiliation(s)
- Hatun A Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Haneen A Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed A Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed Alqinyah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sultan Almudimeegh
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
24
|
Zheng J, Zhang W, Liu L, Hung Yap MK. Low frequency repetitive transcranial magnetic stimulation promotes plasticity of the visual cortex in adult amblyopic rats. Front Neurosci 2023; 17:1109735. [PMID: 36743805 PMCID: PMC9892759 DOI: 10.3389/fnins.2023.1109735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
The decline of visual plasticity restricts the recovery of visual functions in adult amblyopia. Repetitive transcranial magnetic stimulation (rTMS) has been shown to be effective in treating adult amblyopia. However, the underlying mechanisms of rTMS on visual cortex plasticity remain unclear. In this study, we found that low-frequency rTMS reinstated the amplitude of visual evoked potentials, but did not influence the impaired depth perception of amblyopic rats. Furthermore, the expression of synaptic plasticity genes and the number of dendritic spines were significantly higher in amblyopic rats which received rTMS when compared with amblyopic rats which received sham stimulation, with reduced level of inhibition and perineuronal nets in visual cortex, as observed via molecular and histological investigations. The results provide further evidence that rTMS enhances functional recovery and visual plasticity in an adult amblyopic animal model.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Wenqiu Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | - Longqian Liu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
- Department of Optometry and Visual Science, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
25
|
Kaneko M, Stryker MP. Production of brain-derived neurotrophic factor gates plasticity in developing visual cortex. Proc Natl Acad Sci U S A 2023; 120:e2214833120. [PMID: 36634145 PMCID: PMC9934058 DOI: 10.1073/pnas.2214833120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/06/2022] [Indexed: 01/13/2023] Open
Abstract
We have previously shown that recovery of visual responses to a deprived eye during the critical period in mouse primary visual cortex requires phosphorylation of the TrkB receptor for BDNF [M. Kaneko, J. L. Hanover, P. M. England, M. P. Stryker, Nat. Neurosci. 11, 497-504 (2008)]. We have now studied the temporal relationship between the production of mature BDNF and the recovery of visual responses under several different conditions. Visual cortical responses to an eye whose vision has been occluded for several days during the critical period and is then re-opened recover rapidly during binocular vision or much more slowly following reverse occlusion, when the previously intact fellow eye is occluded in a model of "patch therapy" for amblyopia. The time to recovery of visual responses differed by more than 18 h between these two procedures, but in each, the production of mature BDNF preceded the physiological recovery. These findings suggest that a spurt of BDNF production is permissive for the growth of connections serving the deprived eye to restore visual responses. Attenuation of recovery of deprived-eye responses by interference with TrkB receptor activation or reduction of BDNF production by interference with homeostatic synaptic scaling had effects consistent with this suggestion.
Collapse
Affiliation(s)
- Megumi Kaneko
- Department of Physiology and Kavli Institute for Fundamental Neuroscience, University of California, San Francisco94143
| | - Michael P. Stryker
- Department of Physiology and Kavli Institute for Fundamental Neuroscience, University of California, San Francisco94143
| |
Collapse
|
26
|
Huang Z. A Function of Amyloid-β in Mediating Activity-Dependent Axon/Synapse Competition May Unify Its Roles in Brain Physiology and Pathology. J Alzheimers Dis 2023; 92:29-57. [PMID: 36710681 PMCID: PMC10023438 DOI: 10.3233/jad-221042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Amyloid-β protein precursor (AβPP) gives rise to amyloid-β (Aβ), a peptide at the center of Alzheimer's disease (AD). AβPP, however, is also an ancient molecule dating back in evolution to some of the earliest forms of metazoans. This suggests a possible ancestral function that may have been obscured by those that evolve later. Based on literature from the functions of Aβ/AβPP in nervous system development, plasticity, and disease, to those of anti-microbial peptides (AMPs) in bacterial competition as well as mechanisms of cell competition uncovered first by Drosophila genetics, I propose that Aβ/AβPP may be part of an ancient mechanism employed in cell competition, which is subsequently co-opted during evolution for the regulation of activity-dependent neural circuit development and plasticity. This hypothesis is supported by foremost the high similarities of Aβ to AMPs, both of which possess unique, opposite (i.e., trophic versus toxic) activities as monomers and oligomers. A large body of data further suggests that the different Aβ oligomeric isoforms may serve as the protective and punishment signals long predicted to mediate activity-dependent axonal/synaptic competition in the developing nervous system and that the imbalance in their opposite regulation of innate immune and glial cells in the brain may ultimately underpin AD pathogenesis. This hypothesis can not only explain the diverse roles observed of Aβ and AβPP family molecules, but also provide a conceptual framework that can unify current hypotheses on AD. Furthermore, it may explain major clinical observations not accounted for and identify approaches for overcoming shortfalls in AD animal modeling.
Collapse
Affiliation(s)
- Zhen Huang
- Departments of Neuroscience and Neurology, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
27
|
Groves Kuhnle C, Grimes M, Suárez Casanova VM, Turrigiano GG, Van Hooser SD. Juvenile Shank3 KO Mice Adopt Distinct Hunting Strategies during Prey Capture Learning. eNeuro 2022; 9:ENEURO.0230-22.2022. [PMID: 36446569 PMCID: PMC9768843 DOI: 10.1523/eneuro.0230-22.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/11/2022] [Accepted: 10/23/2022] [Indexed: 12/02/2022] Open
Abstract
Mice are opportunistic omnivores that readily learn to hunt and eat insects such as crickets. The details of how mice learn these behaviors and how these behaviors may differ in strains with altered neuroplasticity are unclear. We quantified the behavior of juvenile wild-type (WT) and Shank3 knock-out (KO) mice as they learned to hunt crickets during the critical period for ocular dominance plasticity. This stage involves heightened cortical plasticity including homeostatic synaptic scaling, which requires Shank3, a glutamatergic synaptic protein that, when mutated, produces Phelan-McDermid syndrome and is often comorbid with autism spectrum disorder (ASD). Both strains showed interest in examining live and dead crickets and learned to hunt. Shank3 knock-out mice took longer to become proficient, and, after 5 d, did not achieve the efficiency of wild-type mice in either time-to-capture or distance-to-capture. Shank3 knock-out mice also exhibited different characteristics when pursuing crickets that could not be explained by a simple motor deficit. Although both genotypes moved at the same average speed when approaching a cricket, Shank3 KO mice paused more often, did not begin final accelerations toward crickets as early, and did not close the distance gap to the cricket as quickly as wild-type mice. These differences in Shank3 KO mice are reminiscent of some behavioral characteristics of individuals with ASD as they perform complex tasks, such as slower action initiation and completion. This paradigm will be useful for exploring the neural circuit mechanisms that underlie these learning and performance differences in monogenic ASD rodent models.
Collapse
Affiliation(s)
| | - Micaela Grimes
- Department of Biology, Brandeis University, Waltham, MA 02453
| | | | | | | |
Collapse
|
28
|
Gonzalez A, Hammock EAD. Oxytocin and microglia in the development of social behaviour. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210059. [PMID: 35858111 PMCID: PMC9272152 DOI: 10.1098/rstb.2021.0059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 04/18/2022] [Indexed: 08/31/2023] Open
Abstract
Oxytocin is a well-established regulator of social behaviour. Microglia, the resident immune cells of the central nervous system, regulate brain development and maintenance in health and disease. Oxytocin and microglia interact: microglia appear to regulate the oxytocin system and are, in turn, regulated by oxytocin, which appears to have anti-inflammatory effects. Both microglia and oxytocin are regulated in sex-specific ways. Oxytocin and microglia may work together to promote experience-dependent circuit refinement through multiple developmental-sensitive periods contributing to individual differences in social behaviour. This article is part of the theme issue 'Interplays between oxytocin and other neuromodulators in shaping complex social behaviours'.
Collapse
Affiliation(s)
- Alicia Gonzalez
- Department of Psychology and Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 32306, USA
| | - Elizabeth A. D. Hammock
- Department of Psychology and Program in Neuroscience, Florida State University, 1107 West Call Street, Tallahassee, FL 32306, USA
| |
Collapse
|
29
|
A circuit mechanism for independent modulation of excitatory and inhibitory firing rates after sensory deprivation. Proc Natl Acad Sci U S A 2022; 119:e2116895119. [PMID: 35925891 PMCID: PMC9371725 DOI: 10.1073/pnas.2116895119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cortex is particularly vulnerable to perturbations during sensitive periods, such as the critical period when manipulating sensory experience can induce long-lasting changes in brain structure. Depriving rodents of vision in one eye (known as monocular deprivation [MD]) reduces network activity over two days, whereby inhibitory neurons decrease their firing rates one day after MD, while excitatory neurons are delayed by an additional day. We use spiking networks to mechanistically dissect the requirements for this independent firing-rate regulation after sensory deprivation. We find that in networks stabilized by recurrent inhibition, at least two interneuron subtypes (parvalbumin-expressing and somatostatin-expressing interneurons) are necessary to dynamically alter the circuit response after deprivation and generalize the result across sensory cortices. Diverse interneuron subtypes shape sensory processing in mature cortical circuits. During development, sensory deprivation evokes powerful synaptic plasticity that alters circuitry, but how different inhibitory subtypes modulate circuit dynamics in response to this plasticity remains unclear. We investigate how deprivation-induced synaptic changes affect excitatory and inhibitory firing rates in a microcircuit model of the sensory cortex with multiple interneuron subtypes. We find that with a single interneuron subtype (parvalbumin-expressing [PV]), excitatory and inhibitory firing rates can only be comodulated—increased or decreased together. To explain the experimentally observed independent modulation, whereby one firing rate increases and the other decreases, requires strong feedback from a second interneuron subtype (somatostatin-expressing [SST]). Our model applies to the visual and somatosensory cortex, suggesting a general mechanism across sensory cortices. Therefore, we provide a mechanistic explanation for the differential role of interneuron subtypes in regulating firing rates, contributing to the already diverse roles they serve in the cortex.
Collapse
|
30
|
Castillo-Ruiz A, Cisternas CD, Sturgeon H, Forger NG. Birth triggers an inflammatory response in the neonatal periphery and brain. Brain Behav Immun 2022; 104:122-136. [PMID: 35661680 DOI: 10.1016/j.bbi.2022.05.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/11/2022] [Accepted: 05/30/2022] [Indexed: 10/18/2022] Open
Abstract
Birth is preceded by inflammation at the fetal/maternal interface. Additionally, the newborn experiences stimuli that under any other circumstance could elicit an immune response. It is unknown, however, whether birth elicits an inflammatory response in the newborn that extends to the brain. Moreover, it is unknown whether birth mode may alter such a response. To study these questions, we first measured corticosterone and pro- and anti-inflammatory cytokines in plasma of mouse offspring at several timepoints spaced closely before and after a vaginal or Cesarean birth. We found highest levels of IL-6 one day before birth and surges in corticosterone and IL-10 just after birth, regardless of birth mode. We next examined the neuroimmune response by measuring cytokine mRNA expression and microglial number and morphology in the paraventricular nucleus of the hypothalamus and hippocampus around the time of birth. We found a marked increase in TNF-α expression in both brain regions a day after birth, and rapid increases in microglial cell number in the first three days postnatal, with subtle differences by birth mode. To test whether the association between birth and cytokine production or expansion of microglia is causal, we manipulated birth timing. Remarkably, advancing birth by a day advanced the increases in all of the markers tested. Thus, birth triggers an immune response in the body and brain of offspring. Our results may provide a mechanism for effects of birth (e.g., acute changes in cell death and neural activation) previously reported in the newborn brain.
Collapse
Affiliation(s)
| | - Carla D Cisternas
- Instituto de Investigación Médica Mercedes y Martín Ferreyra INIMEC-CONICET-UNC, Córdoba, Argentina
| | - Hannah Sturgeon
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| | - Nancy G Forger
- Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
31
|
Lowery RL, Majewska AK. Synapse-specific plasticity relies on neuroimmune interactions. Proc Natl Acad Sci U S A 2022; 119:e2207817119. [PMID: 35737828 PMCID: PMC9271177 DOI: 10.1073/pnas.2207817119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Rebecca L. Lowery
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| | - Ania K. Majewska
- Department of Neuroscience, Center for Visual Science, University of Rochester, Rochester, NY 14642
| |
Collapse
|
32
|
Xiao SY, Liu YJ, Lu W, Sha ZW, Xu C, Yu ZH, Lee SD. Possible Neuropathology of Sleep Disturbance Linking to Alzheimer's Disease: Astrocytic and Microglial Roles. Front Cell Neurosci 2022; 16:875138. [PMID: 35755779 PMCID: PMC9218054 DOI: 10.3389/fncel.2022.875138] [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: 02/13/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep disturbances not only deteriorate Alzheimer’s disease (AD) progress by affecting cognitive states but also accelerate the neuropathological changes of AD. Astrocytes and microglia are the principal players in the regulation of both sleep and AD. We proposed that possible astrocyte-mediated and microglia-mediated neuropathological changes of sleep disturbances linked to AD, such as astrocytic adenosinergic A1, A2, and A3 regulation; astrocytic dopamine and serotonin; astrocyte-mediated proinflammatory status (TNFα); sleep disturbance-attenuated microglial CX3CR1 and P2Y12; microglial Iba-1 and astrocytic glial fibrillary acidic protein (GFAP); and microglia-mediated proinflammatory status (IL-1b, IL-6, IL-10, and TNFα). Furthermore, astrocytic and microglial amyloid beta (Aβ) and tau in AD were reviewed, such as astrocytic Aβ interaction in AD; astrocyte-mediated proinflammation in AD; astrocytic interaction with Aβ in the central nervous system (CNS); astrocytic apolipoprotein E (ApoE)-induced Aβ clearance in AD, as well as microglial Aβ clearance and aggregation in AD; proinflammation-induced microglial Aβ aggregation in AD; microglial-accumulated tau in AD; and microglial ApoE and TREM2 in AD. We reviewed astrocytic and microglial roles in AD and sleep, such as astrocyte/microglial-mediated proinflammation in AD and sleep; astrocytic ApoE in sleep and AD; and accumulated Aβ-triggered synaptic abnormalities in sleep disturbance. This review will provide a possible astrocytic and microglial mechanism of sleep disturbance linked to AD.
Collapse
Affiliation(s)
- Shu-Yun Xiao
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi-Jie Liu
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Institute of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wang Lu
- Department of Traditional Treatment, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhong-Wei Sha
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Che Xu
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhi-Hua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shin-Da Lee
- Department of Mental Diseases, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Physical Therapy, Graduate Institute of Rehabilitation Science, China Medical University, Taichung, Taiwan.,Department of Physical Therapy, Asia University, Taichung, Taiwan
| |
Collapse
|
33
|
Pandey A, Hardingham N, Fox K. Differentiation of Hebbian and homeostatic plasticity mechanisms within layer 5 visual cortex neurons. Cell Rep 2022; 39:110892. [PMID: 35649371 PMCID: PMC9637998 DOI: 10.1016/j.celrep.2022.110892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/05/2022] [Accepted: 05/09/2022] [Indexed: 11/26/2022] Open
Abstract
Cortical layer 5 contains two major types of projection neuron known as IB (intrinsic bursting) cells that project sub-cortically and RS (regular spiking) cells that project between cortical areas. This study describes the plasticity properties of RS and IB cells in the mouse visual cortex during the critical period for ocular dominance plasticity. We find that RS neurons exhibit synaptic depression in response to both dark exposure (DE) and monocular deprivation (MD), and their homeostatic recovery from depression is dependent on TNF-α. In contrast, IB cells demonstrate opposite responses to DE and MD, potentiating to DE and depressing to MD. IB cells' potentiation depends on CaMKII-autophosphorylation and not TNF-α. IB cells show mature synaptic properties at the start of the critical period while RS cells mature during the critical period. Together with observations in somatosensory cortex, these results suggest that differences in RS and IB plasticity mechanisms are a general cortical property.
Collapse
Affiliation(s)
- Anurag Pandey
- School of Biosciences, Cardiff University Museum Avenue, Cardiff CF10 3AX, UK
| | - Neil Hardingham
- School of Biosciences, Cardiff University Museum Avenue, Cardiff CF10 3AX, UK
| | - Kevin Fox
- School of Biosciences, Cardiff University Museum Avenue, Cardiff CF10 3AX, UK.
| |
Collapse
|
34
|
Guo A, Lau CG. TNF-α Orchestrates Experience-Dependent Plasticity of Excitatory and Inhibitory Synapses in the Anterior Piriform Cortex. Front Neurosci 2022; 16:824454. [PMID: 35557610 PMCID: PMC9086849 DOI: 10.3389/fnins.2022.824454] [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: 11/29/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Homeostatic synaptic plasticity, which induces compensatory modulation of synapses, plays a critical role in maintaining neuronal circuit function in response to changing activity patterns. Activity in the anterior piriform cortex (APC) is largely driven by ipsilateral neural activity from the olfactory bulb and is a suitable system for examining the effects of sensory experience on cortical circuits. Pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) can modulate excitatory and inhibitory synapses, but its role in APC is unexplored. Here we examined the role of TNF-α in adjusting synapses in the mouse APC after experience deprivation via unilateral naris occlusion. Immunofluorescent staining revealed that activity deprivation increased excitatory, and decreased inhibitory, synaptic density in wild-type mice, consistent with homeostatic regulation. Quantitative RT-PCR showed that naris occlusion increased the expression of Tnf mRNA in APC. Critically, occlusion-induced plasticity of excitatory and inhibitory synapses was completely blocked in the Tnf knockout mouse. Together, these results show that TNF-α is an important orchestrator of experience-dependent plasticity in the APC.
Collapse
Affiliation(s)
- Anni Guo
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| | - Chunyue Geoffrey Lau
- Department of Neuroscience, City University of Hong Kong, Kowloon, Hong Kong SAR, China.,Shenzhen Research Institute, City University of Hong Kong, Shenzhen, China
| |
Collapse
|
35
|
Renouard L, Hayworth C, Rempe M, Clegern W, Wisor J, Frank MG. REM sleep promotes bidirectional plasticity in developing visual cortex in vivo. Neurobiol Sleep Circadian Rhythms 2022; 12:100076. [PMID: 35592144 PMCID: PMC9112011 DOI: 10.1016/j.nbscr.2022.100076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 11/23/2022] Open
Abstract
Sleep is required for the full expression of plasticity during the visual critical period (CP). However, the precise role of rapid-eye-movement (REM) sleep in this process is undetermined. Previous studies in rodents indicate that REM sleep weakens cortical circuits following MD, but this has been explored in only one class of cortical neuron (layer 5 apical dendrites). We investigated the role of REM sleep in ocular dominance plasticity (ODP) in layer 2/3 neurons using 2-photon calcium imaging in awake CP mice. In contrast to findings in layer 5 neurons, we find that REM sleep promotes changes consistent with synaptic strengthening and weakening. This supports recent suggestions that the effects of sleep on plasticity are highly dependent upon the type of circuit and preceding waking experience.
Collapse
Affiliation(s)
- Leslie Renouard
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| | - Christopher Hayworth
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| | - Michael Rempe
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| | - Will Clegern
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| | - Jonathan Wisor
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| | - Marcos G. Frank
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, HSB 280M, Spokane, WA, 99223, USA
| |
Collapse
|
36
|
Anti-neuroinflammatory of Chloroform Extract of Panax ginseng Root Culture on Lipopolysaccharide-stimulated BV2 Microglia Cells. Rep Biochem Mol Biol 2022; 11:125-137. [PMID: 35765526 PMCID: PMC9208560 DOI: 10.52547/rbmb.11.1.125] [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: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 01/11/2023]
Abstract
Background It is believed that activation of microglia in the central nervous system upon detection of stimulus like lipopolysaccharides provokes neuroinflammation via the production of pro-inflammatory mediators and cytokines. The cytoprotective and anti-inflammatory properties of various folk medicine has been gaining attention as a strategy to combat various disease. This study aimed to assess the anti-neuroinflammatory properties of chloroform extract of in vitro Panax ginseng root culture based on nitric oxide and cytokines production. Methods The study was initiated with the determination of maximum non-toxic dose (MNTD) of P. ginseng root culture chloroform extract using the MTT assay. The lipopolysaccharides-stimulated BV2 microglia cells were treated with MNTD and ½MNTD of the extract and its anti-neuroinflammatory properties were assessed by measuring the production of nitric oxide (NO) via Griess assay, as well as TNF-α, IL-6 and IL-10 using Quantikine ELISA. Results It was found that the MNTD and ½MNTD of the extract did not play a significant role in the production of pro-inflammatory cytokines such as NO, TNF-α and IL-6. However, the MNTD and ½MNTD of chloroform extract significantly increased the anti-inflammatory IL-10 compared to the untreated cells. Conclusion With this, the chloroform extract of P. ginseng root culture potentially exerts anti-neuroinflammatory properties.
Collapse
|
37
|
Paraschivescu C, Barbosa S, Van Steenwinckel J, Gressens P, Glaichenhaus N, Davidovic L. Early Life Exposure to Tumor Necrosis Factor Induces Precocious Sensorimotor Reflexes Acquisition and Increases Locomotor Activity During Mouse Postnatal Development. Front Behav Neurosci 2022; 16:845458. [PMID: 35368298 PMCID: PMC8964393 DOI: 10.3389/fnbeh.2022.845458] [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: 12/29/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation appears as a cardinal mediator of the deleterious effect of early life stress exposure on neurodevelopment. More generally, immune activation during the perinatal period, and most importantly elevations of pro-inflammatory cytokines levels could contribute to psychopathology and neurological deficits later in life. Cytokines are also required for normal brain function in homeostatic conditions and play a role in neurodevelopmental processes. Despite these latter studies, whether pro-inflammatory cytokines such as Tumor Necrosis Factor (TNF) impact neurodevelopmental trajectories and behavior during the immediate postnatal period remains to be elucidated. To address this issue, we have injected mouse pups daily with recombinant TNF from postnatal day (P)1 to P5. This yielded a robust increase in peripheral and central TNF at P5, and also an increase of additional pro-inflammatory cytokines. Compared to control pups injected with saline, mice injected with TNF acquired the righting and the acoustic startle reflexes more rapidly and exhibited increased locomotor activity 2 weeks after birth. Our results extend previous work restricted to adult behaviors and support the notion that cytokines, and notably TNF, modulate early neurodevelopmental trajectories.
Collapse
Affiliation(s)
- Cristina Paraschivescu
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Susana Barbosa
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | | | | | - Nicolas Glaichenhaus
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| | - Laetitia Davidovic
- Centre National de la Recherche Scientifique, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d'Azur, Valbonne, France
| |
Collapse
|
38
|
Chronic Monocular Deprivation Reveals MMP9-Dependent and -Independent Aspects of Murine Visual System Plasticity. Int J Mol Sci 2022; 23:ijms23052438. [PMID: 35269580 PMCID: PMC8909986 DOI: 10.3390/ijms23052438] [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: 12/31/2021] [Revised: 02/17/2022] [Accepted: 02/17/2022] [Indexed: 02/04/2023] Open
Abstract
The deletion of matrix metalloproteinase MMP9 is combined here with chronic monocular deprivation (cMD) to identify the contributions of this proteinase to plasticity in the visual system. Calcium imaging of supragranular neurons of the binocular region of primary visual cortex (V1b) of wild-type mice revealed that cMD initiated at eye opening significantly decreased the strength of deprived-eye visual responses to all stimulus contrasts and spatial frequencies. cMD did not change the selectivity of V1b neurons for the spatial frequency, but orientation selectivity was higher in low spatial frequency-tuned neurons, and orientation and direction selectivity were lower in high spatial frequency-tuned neurons. Constitutive deletion of MMP9 did not impact the stimulus selectivity of V1b neurons, including ocular preference and tuning for spatial frequency, orientation, and direction. However, MMP9-/- mice were completely insensitive to plasticity engaged by cMD, such that the strength of the visual responses evoked by deprived-eye stimulation was maintained across all stimulus contrasts, orientations, directions, and spatial frequencies. Other forms of experience-dependent plasticity, including stimulus selective response potentiation, were normal in MMP9-/- mice. Thus, MMP9 activity is dispensable for many forms of activity-dependent plasticity in the mouse visual system, but is obligatory for the plasticity engaged by cMD.
Collapse
|
39
|
Optimizing intact skull intrinsic signal imaging for subsequent targeted electrophysiology across mouse visual cortex. Sci Rep 2022; 12:2063. [PMID: 35136111 PMCID: PMC8826313 DOI: 10.1038/s41598-022-05932-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 01/18/2022] [Indexed: 12/16/2022] Open
Abstract
Understanding brain function requires repeatable measurements of neural activity across multiple scales and multiple brain areas. In mice, large scale cortical neural activity evokes hemodynamic changes readily observable with intrinsic signal imaging (ISI). Pairing ISI with visual stimulation allows identification of primary visual cortex (V1) and higher visual areas (HVAs), typically through cranial windows that thin or remove the skull. These procedures can diminish long-term mechanical and physiological stability required for delicate electrophysiological measurements made weeks to months after imaging (e.g., in subjects undergoing behavioral training). Here, we optimized and directly validated an intact skull ISI system in mice. We first assessed how imaging quality and duration affect reliability of retinotopic maps in V1 and HVAs. We then verified ISI map retinotopy in V1 and HVAs with targeted, multi-site electrophysiology several weeks after imaging. Reliable ISI maps of V1 and multiple HVAs emerged with ~ 60 trials of imaging (65 ± 6 min), and these showed strong correlation to local field potential (LFP) retinotopy in superficial cortical layers (r2 = 0.74–0.82). This system is thus well-suited for targeted, multi-area electrophysiology weeks to months after imaging. We provide detailed instructions and code for other researchers to implement this system.
Collapse
|
40
|
Pathway-specific TNF-mediated metaplasticity in hippocampal area CA1. Sci Rep 2022; 12:1746. [PMID: 35110639 PMCID: PMC8810872 DOI: 10.1038/s41598-022-05844-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/19/2022] [Indexed: 01/29/2023] Open
Abstract
Long-term potentiation (LTP) is regulated in part by metaplasticity, the activity-dependent alterations in neural state that coordinate the direction, amplitude, and persistence of future synaptic plasticity. Previously, we documented a heterodendritic metaplasticity effect whereby high-frequency priming stimulation in stratum oriens (SO) of hippocampal CA1 suppressed subsequent LTP in the stratum radiatum (SR). The cytokine tumor necrosis factor (TNF) mediated this heterodendritic metaplasticity in wild-type rodents and in a mouse model of Alzheimer’s disease. Here, we investigated whether LTP at other afferent synapses to CA1 pyramidal cells were similarly affected by priming stimulation. We found that priming stimulation in SO inhibited LTP only in SR and not in a second independent pathway in SO, nor in stratum lacunosum moleculare (SLM). Synapses in SR were also more sensitive than SO or SLM to the LTP-inhibiting effects of pharmacological TNF priming. Neither form of priming was sex-specific, while the metaplasticity effects were absent in TNFR1 knock-out mice. Our findings demonstrate an unexpected pathway specificity for the heterodendritic metaplasticity in CA1. That Schaffer collateral/commissural synapses in SR are particularly susceptible to such metaplasticity may reflect an important control of information processing in this pathway in addition to its sensitivity to neuroinflammation under disease conditions.
Collapse
|
41
|
Stress induced microglial activation contributes to depression. Pharmacol Res 2022; 179:106145. [DOI: 10.1016/j.phrs.2022.106145] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 02/08/2022] [Accepted: 02/22/2022] [Indexed: 02/06/2023]
|
42
|
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
|
43
|
Barnes SJ, Keller GB, Keck T. Homeostatic regulation through strengthening of neuronal network-correlated synaptic inputs. eLife 2022; 11:81958. [PMID: 36515269 PMCID: PMC9803349 DOI: 10.7554/elife.81958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Homeostatic regulation is essential for stable neuronal function. Several synaptic mechanisms of homeostatic plasticity have been described, but the functional properties of synapses involved in homeostasis are unknown. We used longitudinal two-photon functional imaging of dendritic spine calcium signals in visual and retrosplenial cortices of awake adult mice to quantify the sensory deprivation-induced changes in the responses of functionally identified spines. We found that spines whose activity selectively correlated with intrinsic network activity underwent tumor necrosis factor alpha (TNF-α)-dependent homeostatic increases in their response amplitudes, but spines identified as responsive to sensory stimulation did not. We observed an increase in the global sensory-evoked responses following sensory deprivation, despite the fact that the identified sensory inputs did not strengthen. Instead, global sensory-evoked responses correlated with the strength of network-correlated inputs. Our results suggest that homeostatic regulation of global responses is mediated through changes to intrinsic network-correlated inputs rather than changes to identified sensory inputs thought to drive sensory processing.
Collapse
Affiliation(s)
- Samuel J Barnes
- Department of Brain Sciences, Division of Neuroscience, Imperial College London, Hammersmith Hospital CampusLondonUnited Kingdom,UK Dementia Research Institute at Imperial CollegeLondonUnited Kingdom
| | - Georg B Keller
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Tara Keck
- Department of Neuroscience, Physiology and Pharmacology, University College LondonLondonUnited Kingdom
| |
Collapse
|
44
|
Stacy AK, Van Hooser SD. Development of Functional Properties in the Early Visual System: New Appreciations of the Roles of Lateral Geniculate Nucleus. Curr Top Behav Neurosci 2022; 53:3-35. [PMID: 35112333 DOI: 10.1007/7854_2021_297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In the years following Hubel and Wiesel's first reports on ocular dominance plasticity and amblyopia, much attention has been focused on understanding the role of cortical circuits in developmental and experience-dependent plasticity. Initial studies found few differences between retinal ganglion cells and neurons in the lateral geniculate nucleus and uncovered little evidence for an impact of altered visual experience on the functional properties of lateral geniculate nucleus neurons. In the last two decades, however, studies have revealed that the connectivity between the retina and lateral geniculate nucleus is much richer than was previously appreciated, even revealing visual plasticity - including ocular dominance plasticity - in lateral geniculate nucleus neurons. Here we review the development of the early visual system and the impact of experience with a distinct focus on recent discoveries about lateral geniculate nucleus, its connectivity, and evidence for its plasticity and rigidity during development.
Collapse
Affiliation(s)
- Andrea K Stacy
- Department of Biology, Brandeis University, Waltham, MA, USA
| | | |
Collapse
|
45
|
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
|
46
|
Abd-El-Basset EM, Rao MS, Alshawaf SM, Ashkanani HK, Kabli AH. Tumor necrosis factor (TNF) induces astrogliosis, microgliosis and promotes survival of cortical neurons. AIMS Neurosci 2021; 8:558-584. [PMID: 34877406 PMCID: PMC8611192 DOI: 10.3934/neuroscience.2021031] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/02/2021] [Indexed: 11/21/2022] Open
Abstract
Objectives Neuro-inflammation occurs as a sequence of brain injury and is associated with production of cytokines. Cytokines can modulate the function and survival of neurons, microglia and astrocytes. The objective of this study is to examine the effect of TNF on the neurons, microglia and astrocytes in normal brain and stab wound brain injury. Methods Normal BALB/c male mice (N) without any injury were subdivided into NA and NB groups. Another set mouse was subjected to stab wound brain injury (I) and were subdivided into IA and IB. NA and IA groups received intraperitoneal injections of TNF (1 µg/kg body weight/day) for nine days, whereas NB and IB groups received intraperitoneal injections of PBS. Animals were killed on 1st, 2nd, 3rd, 7th, and 9th day. Frozen brain sections through the injury site in IA and IB or corresponding region in NA and NB groups were stained for neurodegeneration, immunostained for astrocytes, microglia and neurons. Western blotting for GFAP and ELISA for BDNF were done from the tissues collected from all groups. Results The number of degenerating neurons significantly decreased in TNF treated groups. There was a significant increase in the number of astrocytes and microglia in TNF treated groups compared to PBS treated groups. In addition, it was found that TNF stimulated the expression of GFAP and BDNF in NA and IA groups. Conclusions TNF induces astrogliosis and microgliosis in normal and injured brain and promotes the survival of cortical neurons in stab wound brain injury, may be by upregulating the BDNF level.
Collapse
Affiliation(s)
- Ebtesam M Abd-El-Basset
- Department of Anatomy, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13100, Kuwait
| | - Muddanna Sakkattu Rao
- Department of Anatomy, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13100, Kuwait
| | | | | | | |
Collapse
|
47
|
Malada Edelstein YF, Solomonov Y, Hadad N, Alfahel L, Israelson A, Levy R. Early upregulation of cytosolic phospholipase A 2α in motor neurons is induced by misfolded SOD1 in a mouse model of amyotrophic lateral sclerosis. J Neuroinflammation 2021; 18:274. [PMID: 34823547 PMCID: PMC8620709 DOI: 10.1186/s12974-021-02326-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 11/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a fatal multifactorial neurodegenerative disease characterized by the selective death of motor neurons. Cytosolic phospholipase A2 alpha (cPLA2α) upregulation and activation in the spinal cord of ALS patients has been reported. We have previously shown that cPLA2α upregulation in the spinal cord of mutant SOD1 transgenic mice (SOD1G93A) was detected long before the development of the disease, and inhibition of cPLA2α upregulation delayed the disease's onset. The aim of the present study was to determine the mechanism for cPLA2α upregulation. METHODS Immunofluorescence analysis and western blot analysis of misfolded SOD1, cPLA2α and inflammatory markers were performed in the spinal cord sections of SOD1G93A transgenic mice and in primary motor neurons. Over expression of mutant SOD1 was performed by induction or transfection in primary motor neurons and in differentiated NSC34 motor neuron like cells. RESULTS Misfolded SOD1 was detected in the spinal cord of 3 weeks old mutant SOD1G93A mice before cPLA2α upregulation. Elevated expression of both misfolded SOD1 and cPLA2α was specifically detected in the motor neurons at 6 weeks with a high correlation between them. Elevated TNFα levels were detected in the spinal cord lysates of 6 weeks old mutant SOD1G93A mice. Elevated TNFα was specifically detected in the motor neurons and its expression was highly correlated with cPLA2α expression at 6 weeks. Induction of mutant SOD1 in primary motor neurons induced cPLA2α and TNFα upregulation. Over expression of mutant SOD1 in NSC34 cells caused cPLA2α upregulation which was prevented by antibodies against TNFα. The addition of TNFα to NSC34 cells caused cPLA2α upregulation in a dose dependent manner. CONCLUSIONS Motor neurons expressing elevated cPLA2α and TNFα are in an inflammatory state as early as at 6 weeks old mutant SOD1G93A mice long before the development of the disease. Accumulated misfolded SOD1 in the motor neurons induced cPLA2α upregulation via induction of TNFα.
Collapse
Affiliation(s)
- Yafa Fetfet Malada Edelstein
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, 84105, Beer Sheva, Israel
| | - Yulia Solomonov
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, 84105, Beer Sheva, Israel
| | - Nurit Hadad
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, 84105, Beer Sheva, Israel
| | - Leenor Alfahel
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Faculty of Health Sciences and The Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rachel Levy
- Immunology and Infectious Diseases Laboratory, Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, 84105, Beer Sheva, Israel.
| |
Collapse
|
48
|
The Health Hazards of Volcanoes: First Evidence of Neuroinflammation in the Hippocampus of Mice Exposed to Active Volcanic Surroundings. Mediators Inflamm 2021; 2021:5891095. [PMID: 34671225 PMCID: PMC8523235 DOI: 10.1155/2021/5891095] [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: 06/16/2021] [Accepted: 09/22/2021] [Indexed: 11/26/2022] Open
Abstract
Neuroinflammation is a process related to the onset of neurodegenerative diseases; one of the hallmarks of this process is microglial reactivation and the secretion by these cells of proinflammatory cytokines such as TNFα. Numerous studies report the relationship between neuroinflammatory processes and exposure to anthropogenic air pollutants, but few refer to natural pollutants. Volcanoes are highly inhabited natural sources of environmental pollution that induce changes in the nervous system, such as reactive astrogliosis or the blood-brain barrier breakdown in exposed individuals; however, no neuroinflammatory event has been yet defined. To this purpose, we studied resting microglia, reactive microglia, and TNFα production in the brains of mice chronically exposed to an active volcanic environment on the island of São Miguel (Azores, Portugal). For the first time, we demonstrate a proliferation of microglial cells and an increase in reactive microglia, as well an increase in TNFα secretion, in the central nervous system of individuals exposed to volcanogenic pollutants.
Collapse
|
49
|
All-or-none disconnection of pyramidal inputs onto parvalbumin-positive interneurons gates ocular dominance plasticity. Proc Natl Acad Sci U S A 2021; 118:2105388118. [PMID: 34508001 DOI: 10.1073/pnas.2105388118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2021] [Indexed: 12/16/2022] Open
Abstract
Disinhibition is an obligatory initial step in the remodeling of cortical circuits by sensory experience. Our investigation on disinhibitory mechanisms in the classical model of ocular dominance plasticity uncovered an unexpected form of experience-dependent circuit plasticity. In the layer 2/3 of mouse visual cortex, monocular deprivation triggers a complete, "all-or-none," elimination of connections from pyramidal cells onto nearby parvalbumin-positive interneurons (Pyr→PV). This binary form of circuit plasticity is unique, as it is transient, local, and discrete. It lasts only 1 d, and it does not manifest as widespread changes in synaptic strength; rather, only about half of local connections are lost, and the remaining ones are not affected in strength. Mechanistically, the deprivation-induced loss of Pyr→PV is contingent on a reduction of the protein neuropentraxin2. Functionally, the loss of Pyr→PV is absolutely necessary for ocular dominance plasticity, a canonical model of deprivation-induced model of cortical remodeling. We surmise, therefore, that this all-or-none loss of local Pyr→PV circuitry gates experience-dependent cortical plasticity.
Collapse
|
50
|
Mourão AA, Shimoura CG, Andrade MA, Truong TT, Pedrino GR, Toney GM. Local ionotropic glutamate receptors are required to trigger and sustain ramping of sympathetic nerve activity by hypothalamic paraventricular nucleus TNF α. Am J Physiol Heart Circ Physiol 2021; 321:H580-H591. [PMID: 34355986 DOI: 10.1152/ajpheart.00322.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tumor necrosis factor-α (TNFα) in the hypothalamic paraventricular nucleus (PVN) contributes to increased sympathetic nerve activity (SNA) in cardiovascular disease models, but mechanisms are incompletely understood. As previously reported, bilateral PVN TNFα (0.6 pmol, 50 nL) induced acute ramping of splanchnic SNA (SSNA) that averaged +64 ± 7% after 60 min and +109 ± 17% after 120 min (P < 0.0001, n = 10). Given that TNFα can rapidly strengthen glutamatergic transmission, we hypothesized that progressive activation of ionotropic glutamate receptors is critically involved. When compared with that of vehicle (n = 5), prior blockade of PVN AMPA or NMDA receptors in anesthetized (urethane/α-chloralose) adult male Sprague-Dawley rats dose-dependently (ED50: 2,3-dioxo-6-nitro-1,2,3,4-tetrahydrobenzo[f]quinoxaline-7-sulfonamide (NBQX), 2.48 nmol; D-(-)-2-amino-5-phosphonopentanoic acid (APV), 12.33 nmol), but incompletely (Emax: NBQX, 64%; APV, 41%), attenuated TNFα-induced SSNA ramping (n = 5/dose). By contrast, combined receptor blockade prevented ramping (1.3 ± 2.1%, P < 0.0001, n = 5). Whereas separate blockade of PVN AMPA or NMDA receptors (n = 5/group) had little effect on continued SSNA ramping when performed 60 min after TNFα injection, combined blockade (n = 5) or PVN inhibition with the GABA-A receptor agonist muscimol (n = 5) effectively stalled, without reversing, the SSNA ramp. Notably, PVN TNFα increased local TNFα immunofluorescence after 120, but not 60 min. Findings indicate that AMPA and NMDA receptors each contribute to SSNA ramping to PVN TNFα, and that their collective availability and ongoing activity are required to initiate and sustain the ramping response. We conclude that acute sympathetic activation by PVN TNFα involves progressive local glutamatergic excitation that recruits downstream neurons capable of maintaining heightened SSNA, but incapable of sustaining SSNA ramping.NEW & NOTEWORTHY The proinflammatory cytokine TNFα contributes to heightened SNA in cardiovascular disease models, but mechanisms remain obscure. Here, we demonstrate that TNFα injection into the hypothalamic PVN triggers SNA ramping by mechanisms dependent on local ionotropic glutamate receptor availability, but largely independent of TNFα autoinduction. Continued SNA ramping depends on ionotropic glutamate receptor and neuronal activity in PVN, indicating that strengthening and/or increased efficacy of glutamatergic transmission is necessary for acute sympathoexcitation by PVN TNFα.
Collapse
Affiliation(s)
- Aline A Mourão
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Department of Physiological Sciences, Center for Neuroscience and Cardiovascular Research, Federal University of Goias, Goiania, Goias, Brazil
| | - Caroline G Shimoura
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Mary Ann Andrade
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Tamara T Truong
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Gustavo R Pedrino
- Department of Physiological Sciences, Center for Neuroscience and Cardiovascular Research, Federal University of Goias, Goiania, Goias, Brazil
| | - Glenn M Toney
- Department of Cellular and Integrative Physiology, University of Texas Health San Antonio, San Antonio, Texas.,Center for Biomedical Neuroscience, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|