1
|
Dong HW, Hintiryan H, Zhu M, Zhao P, Zhang M, Barry J, Nanda S, Rudd M, Wong A, Miller S, Gou L, Wei J, Zingg B, Sun J, Gutierrez A, Mun HS, Bowman I, Garcia L, Lo D, Boesen T, Cao C, Zhao Q, Foster N, Moradi K, Yamashita S, Estrada C, Dev A, Gonzalez J, Xu H, Yang G, Park C, Yang X, Levine M, Zhang L, Micevych P, Cepeda C, Golshani P, Hong W, Yang Y. Neural Networks of the Mouse Primary Visceromotor Cortex. RESEARCH SQUARE 2025:rs.3.rs-4125909. [PMID: 40343331 PMCID: PMC12060971 DOI: 10.21203/rs.3.rs-4125909/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
The medial prefrontal cortex (MPF) regulates emotions, stress responses, and goal-directed behaviors like attention and decision-making. However, the precise mechanisms underlying MPF function remain poorly understood, largely due to an incomplete characterization of its neural circuitry. Leveraging neuroanatomical, neurophysiological, and behavioral techniques, we present a detailed wiring diagram of the MPF, with a particular focus on the dorsal peduncular area (DP), an underexplored MPF area implicated in psychological stress, fear conditioning, anxiety, depression, and opioid addiction. Our analysis identifies the deep (DPd) and superficial (DPs) layers of the DP, together with the infralimbic area (ILA), as key components of the primary visceromotor cortex, that generate monosynaptic projections to regulate neuroendocrine, sympathetic, and parasympathetic functions in distinct, yet coordinated ways. Further, we demonstrate that the DP serves as a unique network hub for unidirectional cortical information flow, that integrates diverse cortical inputs and modulates social behavior. Based on the mesoscale connectome of entire MPF, we propose a unified MPF network model that regulates different aspects of motor actions associated with goal-directed behavior. This study provides novel insights into the complex role of the MPF in orchestrating physiological and behavioral responses to environmental stimuli in mammals.
Collapse
Affiliation(s)
| | | | - Muye Zhu
- David Geffen School of Medicine at UCLA
| | | | | | | | | | | | | | | | - Lin Gou
- USC Stevens Neuroimaging and Informatics Institute (INI), Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033
| | | | - Brian Zingg
- USC Stevens Neuroimaging and Informatics Institute (INI), Keck School of Medicine of USC, University of Southern California
| | | | | | | | - Ian Bowman
- USC Stevens Neuroimaging and Informatics Institute (INI), Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033
| | | | | | | | | | | | - Nicholas Foster
- Center for Integrative Connectomics, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California
| | | | | | | | | | | | | | | | - Chris Park
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior; Department of Psychiatry and Biobehavioral Sciences; Brain Research Institute, David Geffen S
| | | | | | - Li Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California
| | | | - Carlos Cepeda
- Jane and Terry Semel Institute for Neuroscience and Human Behavior; Department of Psychiatry and Biobehavioral Sciences; David Geffen School of Medicine at UCLA
| | | | | | - Yeji Yang
- David Geffen School of Medicine at UCLA
| |
Collapse
|
2
|
Arnal LH, Gonçalves N. Rough is salient: a conserved vocal niche to hijack the brain's salience system. Philos Trans R Soc Lond B Biol Sci 2025; 380:20240020. [PMID: 40176527 PMCID: PMC11966164 DOI: 10.1098/rstb.2024.0020] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 04/04/2025] Open
Abstract
The propensity to communicate extreme emotional states and arousal through salient, non-referential vocalizations is ubiquitous among mammals and beyond. Screams, whether intended to warn conspecifics or deter aggressors, require a rapid increase of air influx through vocal folds to induce nonlinear distortions of the signal. These distortions contain salient, temporally patterned acoustic features in a restricted range of the audible spectrum. These features may have a biological significance, triggering fast behavioural responses in the receivers. We present converging neurophysiological and behavioural evidence from humans and animals supporting that the properties emerging from nonlinear vocal phenomena are ideally adapted to induce efficient sensory, emotional and behavioural responses. We argue that these fast temporal-rough-modulations are unlikely to be an epiphenomenon of vocal production but rather the result of selective evolutionary pressure on vocal warning signals to promote efficient communication. In this view, rough features may have been selected and conserved as an acoustic trait to recruit ancestral sensory salience pathways and elicit optimal reactions in the receiver. By exploring the impact of rough vocalizations at the receiver's end, we review the perceptual, behavioural and neural factors that may have shaped these signals to evolve as powerful communication tools.This article is part of the theme issue 'Nonlinear phenomena in vertebrate vocalizations: mechanisms and communicative functions'.
Collapse
Affiliation(s)
- Luc H. Arnal
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l’Audition, IHU reConnect, Paris75012, France
| | - Noémi Gonçalves
- Université Paris Cité, Institut Pasteur, AP-HP, INSERM, CNRS, Fondation Pour l'Audition, Institut de l’Audition, IHU reConnect, Paris75012, France
| |
Collapse
|
3
|
Zhang NK, Zhang SK, Zhang LI, Tao HW, Zhang GW. The neural basis of neuropsychiatric symptoms in Alzheimer's disease. Front Aging Neurosci 2024; 16:1487875. [PMID: 39703925 PMCID: PMC11655510 DOI: 10.3389/fnagi.2024.1487875] [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: 08/28/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Neuropsychiatric symptoms (NPS) such as depression, anxiety, apathy and aggression affect up to 90% of Alzheimer's disease (AD) patients. These symptoms significantly increase caregiver stress and institutionalization rates, and more importantly they are correlated with faster cognitive decline. However, the neuronal basis of NPS in AD remains largely unknown. Here, we review current understanding of NPS and related pathology in studies of AD patients and AD mouse models. Clinical studies indicate that NPS prevalence and severity vary across different AD stages and types. Neuroimaging and postmortem studies have suggested that pathological changes in the anterior cingulate cortex, hippocampus, prefrontal cortex, and amygdala are linked to NPS, although the precise mechanisms remain unclear. Studies of AD mouse models have indicated that amyloid-beta and tau-related neurodegeneration in the hippocampus, prefrontal cortex, and anterior cingulate cortex are correlated with NPS-like behavioral deficits. A better understanding of the NPS phenotypes and related pathological changes will pave the way for developing a better management strategy for NPS in AD patients.
Collapse
Affiliation(s)
- Nicole K. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Selena K. Zhang
- Biomedical Engineering Program, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, United States
| | - Li I. Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Huizhong W. Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
4
|
Wu JL, Li ZM, Chen H, Chen WJ, Hu NY, Jin SY, Li XW, Chen YH, Yang JM, Gao TM. Distinct septo-hippocampal cholinergic projections separately mediate stress-induced emotional and cognitive deficits. SCIENCE ADVANCES 2024; 10:eado1508. [PMID: 39514666 PMCID: PMC11546849 DOI: 10.1126/sciadv.ado1508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
Patients suffering from chronic stress develop numerous symptoms, including emotional and cognitive deficits. The precise circuit mechanisms underlying different symptoms remain poorly understood. We identified two distinct basal forebrain cholinergic subpopulations in mice projecting to the dorsal hippocampus (dHPC) or ventral hippocampus (vHPC), which exhibited distinct input organizations, electrophysiological characteristics, transcriptomics, and responses to positive and negative valences of stimuli and were critical for cognitive and emotional modulation, respectively. Moreover, chronic stress induced elevated anxiety levels and cognitive deficits in mice, accompanied by enhanced vHPC but suppressed dHPC cholinergic projections. Chemogenetic activation of dHPC or inhibition of vHPC cholinergic projections alleviated stress-induced aberrant behaviors. Furthermore, we identified that the acetylcholinesterase inhibitor donepezil combined with blockade of muscarinic receptor 1-type muscarinic acetylcholine receptors in the vHPC rescued both stress-induced phenotypes. These data illuminated distinct septo-hippocampal cholinergic circuits mediated specific symptoms independently under stress, which may provide promising strategies for circuit-based treating of stress-related psychiatric disorders.
Collapse
Affiliation(s)
| | | | - Hao Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wen-Jun Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Neng-Yuan Hu
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Wen Li
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong Joint Laboratory for Psychiatric Disorders, Guangdong Provincial Key Laboratory of Psychiatric Disorders, Guangdong Basic Research Center of Excellence for Integrated Traditional and Western Medicine for Qingzhi Diseases, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Nguyen D, Wang G, Wafa T, Fitzgerald T, Gu Y. The medial entorhinal cortex encodes multisensory spatial information. Cell Rep 2024; 43:114813. [PMID: 39395171 PMCID: PMC11539853 DOI: 10.1016/j.celrep.2024.114813] [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: 12/27/2023] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024] Open
Abstract
Animals employ spatial information in multisensory modalities to navigate their natural environments. However, it is unclear whether the brain encodes such information in separate cognitive maps or integrates it all into a single, universal map. We address this question in the microcircuit of the medial entorhinal cortex (MEC), a cognitive map of space. Using cellular-resolution calcium imaging, we examine the MEC of mice navigating virtual reality tracks, where visual and auditory cues provide comparable spatial information. We uncover two cell types: "unimodality cells" and "multimodality cells." The unimodality cells specifically represent either auditory or visual spatial information. They are anatomically intermingled and maintain sensory preferences across multiple tracks and behavioral states. The multimodality cells respond to both sensory modalities, with their responses shaped differentially by auditory or visual information. Thus, the MEC enables accurate spatial encoding during multisensory navigation by computing spatial information in different sensory modalities and generating distinct maps.
Collapse
Affiliation(s)
- Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Talah Wafa
- Mouse Auditory Testing Core Facility, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tracy Fitzgerald
- Mouse Auditory Testing Core Facility, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Wei J, Xiao C, Zhang GW, Shen L, Tao HW, Zhang LI. A distributed auditory network mediated by pontine central gray underlies ultra-fast awakening in response to alerting sounds. Curr Biol 2024; 34:4597-4611.e5. [PMID: 39265569 PMCID: PMC11521200 DOI: 10.1016/j.cub.2024.08.020] [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: 03/11/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 09/14/2024]
Abstract
Sleeping animals can be woken up rapidly by external threat signals, which is an essential defense mechanism for survival. However, neuronal circuits underlying the fast transmission of sensory signals for this process remain unclear. Here, we report in mice that alerting sound can induce rapid awakening within hundreds of milliseconds and that glutamatergic neurons in the pontine central gray (PCG) play an important role in this process. These neurons exhibit higher sensitivity to auditory stimuli in sleep than wakefulness. Suppressing these neurons results in reduced sound-induced awakening and increased sleep in intrinsic sleep/wake cycles, whereas their activation induces ultra-fast awakening from sleep and accelerates awakening from anesthesia. Additionally, the sound-induced awakening can be attributed to the propagation of auditory signals from the PCG to multiple arousal-related regions, including the mediodorsal thalamus, lateral hypothalamus, and ventral tegmental area. Thus, the PCG serves as an essential distribution center to orchestrate a global auditory network to promote rapid awakening.
Collapse
Affiliation(s)
- Jinxing Wei
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Cuiyu Xiao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Li Shen
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
7
|
Polley DB, Chambers AR. Auditory circuits: Watchmen of the sleeping brain. Curr Biol 2024; 34:R924-R926. [PMID: 39437729 DOI: 10.1016/j.cub.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
A new study shows that glutamatergic neurons of the pontine central gray (PCG) play a key role in mediating rapid sound-induced awakenings from sleep by relaying short-latency auditory information to multiple arousal centers in the brain.
Collapse
Affiliation(s)
- Daniel B Polley
- Eaton-Peabody Laboratories, Mass Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA.
| | - Anna R Chambers
- Eaton-Peabody Laboratories, Mass Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
8
|
Hegedüs P, Király B, Schlingloff D, Lyakhova V, Velencei A, Szabó Í, Mayer MI, Zelenak Z, Nyiri G, Hangya B. Parvalbumin-expressing basal forebrain neurons mediate learning from negative experience. Nat Commun 2024; 15:4768. [PMID: 38849336 PMCID: PMC11161511 DOI: 10.1038/s41467-024-48755-7] [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: 04/17/2023] [Accepted: 05/11/2024] [Indexed: 06/09/2024] Open
Abstract
Parvalbumin (PV)-expressing GABAergic neurons of the basal forebrain (BFPVNs) were proposed to serve as a rapid and transient arousal system, yet their exact role in awake behaviors remains unclear. We performed bulk calcium measurements and electrophysiology with optogenetic tagging from the horizontal limb of the diagonal band of Broca (HDB) while male mice were performing an associative learning task. BFPVNs responded with a distinctive, phasic activation to punishment, but showed slower and delayed responses to reward and outcome-predicting stimuli. Optogenetic inhibition during punishment impaired the formation of cue-outcome associations, suggesting a causal role of BFPVNs in associative learning. BFPVNs received strong inputs from the hypothalamus, the septal complex and the median raphe region, while they synapsed on diverse cell types in key limbic structures, where they broadcasted information about aversive stimuli. We propose that the arousing effect of BFPVNs is recruited by aversive stimuli to serve crucial associative learning functions.
Collapse
Affiliation(s)
- Panna Hegedüs
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, H-1085, Budapest, Hungary
| | - Bálint Király
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Dániel Schlingloff
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Victoria Lyakhova
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Semmelweis University, H-1085, Budapest, Hungary
| | - Anna Velencei
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Írisz Szabó
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Márton I Mayer
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Zsofia Zelenak
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Gábor Nyiri
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary
| | - Balázs Hangya
- Lendület Laboratory of Systems Neuroscience, HUN-REN Institute of Experimental Medicine, H-1083, Budapest, Hungary.
| |
Collapse
|
9
|
Shipkov D, Nasretdinov A, Khazipov R, Valeeva G. Synchronous excitation in the superficial and deep layers of the medial entorhinal cortex precedes early sharp waves in the neonatal rat hippocampus. Front Cell Neurosci 2024; 18:1403073. [PMID: 38737704 PMCID: PMC11082381 DOI: 10.3389/fncel.2024.1403073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Early Sharp Waves (eSPWs) are the earliest pattern of network activity in the developing hippocampus of neonatal rodents. eSPWs were originally considered to be an immature prototype of adult SPWs, which are spontaneous top-down hippocampal events that are self-generated in the hippocampal circuitry. However, recent studies have shifted this paradigm to a bottom-up model of eSPW genesis, in which eSPWs are primarily driven by the inputs from the layers 2/3 of the medial entorhinal cortex (MEC). A hallmark of the adult SPWs is the relay of information from the CA1 hippocampus to target structures, including deep layers of the EC. Whether and how deep layers of the MEC are activated during eSPWs in the neonates remains elusive. In this study, we investigated activity in layer 5 of the MEC of neonatal rat pups during eSPWs using silicone probe recordings from the MEC and CA1 hippocampus. We found that neurons in deep and superficial layers of the MEC fire synchronously during MEC sharp potentials, and that neuronal firing in both superficial and deep layers of the MEC precedes the activation of CA1 neurons during eSPWs. Thus, the sequence of activation of CA1 hippocampal neurons and deep EC neurons during sharp waves reverses during development, from a lead of deep EC neurons during eSPWs in neonates to a lead of CA1 neurons during adult SPWs. These findings suggest another important difference in the generative mechanisms and possible functional roles of eSPWs compared to adult SPWs.
Collapse
Affiliation(s)
- Dmitrii Shipkov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Azat Nasretdinov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Roustem Khazipov
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
- INMED - INSERM, Aix-Marseille University, Marseille, France
| | - Guzel Valeeva
- Laboratory of Neurobiology, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
10
|
Clayton KK, Stecyk KS, Guo AA, Chambers AR, Chen K, Hancock KE, Polley DB. Sound elicits stereotyped facial movements that provide a sensitive index of hearing abilities in mice. Curr Biol 2024; 34:1605-1620.e5. [PMID: 38492568 PMCID: PMC11043000 DOI: 10.1016/j.cub.2024.02.057] [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: 09/07/2023] [Revised: 01/02/2024] [Accepted: 02/23/2024] [Indexed: 03/18/2024]
Abstract
Sound elicits rapid movements of muscles in the face, ears, and eyes that protect the body from injury and trigger brain-wide internal state changes. Here, we performed quantitative facial videography from mice resting atop a piezoelectric force plate and observed that broadband sounds elicited rapid and stereotyped facial twitches. Facial motion energy (FME) adjacent to the whisker array was 30 dB more sensitive than the acoustic startle reflex and offered greater inter-trial and inter-animal reliability than sound-evoked pupil dilations or movement of other facial and body regions. FME tracked the low-frequency envelope of broadband sounds, providing a means to study behavioral discrimination of complex auditory stimuli, such as speech phonemes in noise. Approximately 25% of layer 5-6 units in the auditory cortex (ACtx) exhibited firing rate changes during facial movements. However, FME facilitation during ACtx photoinhibition indicated that sound-evoked facial movements were mediated by a midbrain pathway and modulated by descending corticofugal input. FME and auditory brainstem response (ABR) thresholds were closely aligned after noise-induced sensorineural hearing loss, yet FME growth slopes were disproportionately steep at spared frequencies, reflecting a central plasticity that matched commensurate changes in ABR wave 4. Sound-evoked facial movements were also hypersensitive in Ptchd1 knockout mice, highlighting the use of FME for identifying sensory hyper-reactivity phenotypes after adult-onset hyperacusis and inherited deficiencies in autism risk genes. These findings present a sensitive and integrative measure of hearing while also highlighting that even low-intensity broadband sounds can elicit a complex mixture of auditory, motor, and reafferent somatosensory neural activity.
Collapse
Affiliation(s)
- Kameron K Clayton
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA.
| | - Kamryn S Stecyk
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Anna A Guo
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA
| | - Anna R Chambers
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Ke Chen
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Kenneth E Hancock
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA
| | - Daniel B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA 02114, USA; Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
11
|
Mocellin P, Barnstedt O, Luxem K, Kaneko H, Vieweg S, Henschke JU, Dalügge D, Fuhrmann F, Karpova A, Pakan JMP, Kreutz MR, Mikulovic S, Remy S. A septal-ventral tegmental area circuit drives exploratory behavior. Neuron 2024; 112:1020-1032.e7. [PMID: 38266645 DOI: 10.1016/j.neuron.2023.12.016] [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: 09/15/2022] [Revised: 11/10/2023] [Accepted: 12/20/2023] [Indexed: 01/26/2024]
Abstract
To survive, animals need to balance their exploratory drive with their need for safety. Subcortical circuits play an important role in initiating and modulating movement based on external demands and the internal state of the animal; however, how motivation and onset of locomotion are regulated remain largely unresolved. Here, we show that a glutamatergic pathway from the medial septum and diagonal band of Broca (MSDB) to the ventral tegmental area (VTA) controls exploratory locomotor behavior in mice. Using a self-supervised machine learning approach, we found an overrepresentation of exploratory actions, such as sniffing, whisking, and rearing, when this projection is optogenetically activated. Mechanistically, this role relies on glutamatergic MSDB projections that monosynaptically target a subset of both glutamatergic and dopaminergic VTA neurons. Taken together, we identified a glutamatergic basal forebrain to midbrain circuit that initiates locomotor activity and contributes to the expression of exploration-associated behavior.
Collapse
Affiliation(s)
- Petra Mocellin
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany.
| | - Oliver Barnstedt
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Kevin Luxem
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Hiroshi Kaneko
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Silvia Vieweg
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Julia U Henschke
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Dennis Dalügge
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; International Max Planck Research School for Brain and Behavior (IMPRS), Bonn 53175, Germany
| | - Falko Fuhrmann
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany
| | - Anna Karpova
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Janelle M P Pakan
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany
| | - Michael R Kreutz
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Sanja Mikulovic
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), Magdeburg 39118, Germany; German Center for Neurodegenerative Diseases (DZNE), Magdeburg 39120, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg 39106, Germany; German Center for Mental Health (DZPG), Magdeburg 39106, Germany.
| |
Collapse
|
12
|
Puhger K, Crestani AP, Diniz CRF, Wiltgen BJ. The hippocampus contributes to retroactive stimulus associations during trace fear conditioning. iScience 2024; 27:109035. [PMID: 38375237 PMCID: PMC10875141 DOI: 10.1016/j.isci.2024.109035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/04/2023] [Accepted: 01/23/2024] [Indexed: 02/21/2024] Open
Abstract
Binding events that occur at different times are essential for memory formation. In trace fear conditioning, animals associate a tone and footshock despite no temporal overlap. The hippocampus is thought to mediate this learning by maintaining a memory of the tone until shock occurrence, however, evidence for sustained hippocampal tone representations is lacking. Here, we demonstrate a retrospective role for the hippocampus in trace fear conditioning. Bulk calcium imaging revealed sustained increases in CA1 activity after footshock that were not observed after tone termination. Optogenetic silencing of CA1 immediately after footshock impaired subsequent memory. Additionally, footshock increased the number of sharp-wave ripples compared to baseline during conditioning. Therefore, post-shock hippocampal activity likely supports learning by reactivating and linking latent tone and shock representations. These findings highlight an underappreciated function of post-trial hippocampal activity in enabling retroactive temporal associations during new learning, as opposed to persistent maintenance of stimulus representations.
Collapse
Affiliation(s)
- Kyle Puhger
- Department of Psychology, University of California, Davis, 135 Young Hall, 1 Shields Avenue, Davis, CA 95616, USA
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA
| | - Ana P. Crestani
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA
| | - Cassiano R.A. F. Diniz
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA
| | - Brian J. Wiltgen
- Department of Psychology, University of California, Davis, 135 Young Hall, 1 Shields Avenue, Davis, CA 95616, USA
- Center for Neuroscience, University of California, Davis, 1544 Newton Court, Davis, CA 95618, USA
| |
Collapse
|
13
|
Nguyen D, Wang G, Gu Y. The medial entorhinal cortex encodes multisensory spatial information. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574924. [PMID: 38313299 PMCID: PMC10836072 DOI: 10.1101/2024.01.09.574924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Animals employ spatial information in multisensory modalities to navigate their natural environments. However, it is unclear whether the brain encodes such information in separate cognitive maps or integrates all into a single, universal map. We addressed this question in the microcircuit of the medial entorhinal cortex (MEC), a cognitive map of space. Using cellular-resolution calcium imaging, we examined the MEC of mice navigating virtual reality tracks, where visual and auditory cues provided comparable spatial information. We uncovered two cell types: "unimodality cells" and "multimodality cells". The unimodality cells specifically represent either auditory or visual spatial information. They are anatomically intermingled and maintain sensory preferences across multiple tracks and behavioral states. The multimodality cells respond to both sensory modalities with their responses shaped differentially by auditory and visual information. Thus, the MEC enables accurate spatial encoding during multisensory navigation by computing spatial information in different sensory modalities and generating distinct maps.
Collapse
Affiliation(s)
- Duc Nguyen
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
- Current address: Center of Neural Science, New York University, New York, NY, USA
| | - Garret Wang
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yi Gu
- Spatial Navigation and Memory Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
14
|
Blanco-Hernández E, Balsamo G, Preston-Ferrer P, Burgalossi A. Sensory and behavioral modulation of thalamic head-direction cells. Nat Neurosci 2024; 27:28-33. [PMID: 38177338 DOI: 10.1038/s41593-023-01506-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 10/24/2023] [Indexed: 01/06/2024]
Abstract
Head-direction (HD) neurons are thought to exclusively encode directional heading. In awake mice, we found that sensory stimuli evoked robust short-latency responses in thalamic HD cells, but not in non-HD neurons. The activity of HD cells, but not that of non-HD neurons, was tightly correlated to brain-state fluctuations and dynamically modulated during social interactions. These data point to a new role for the thalamic compass in relaying sensory and behavioral-state information.
Collapse
Affiliation(s)
- Eduardo Blanco-Hernández
- Institute of Neurobiology, University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
| | - Giuseppe Balsamo
- Institute of Neurobiology, University of Tübingen, Tübingen, Germany
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany
- Graduate Training Centre of Neuroscience, IMPRS, Tübingen, Germany
| | - Patricia Preston-Ferrer
- Institute of Neurobiology, University of Tübingen, Tübingen, Germany.
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany.
| | - Andrea Burgalossi
- Institute of Neurobiology, University of Tübingen, Tübingen, Germany.
- Werner-Reichardt Centre for Integrative Neuroscience, Tübingen, Germany.
| |
Collapse
|
15
|
Salgado-Puga K, Rothschild G. Exposure to sounds during sleep impairs hippocampal sharp wave ripples and memory consolidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568283. [PMID: 38045371 PMCID: PMC10690295 DOI: 10.1101/2023.11.22.568283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Sleep is critical for the consolidation of recent experiences into long-term memories. As a key underlying neuronal mechanism, hippocampal sharp-wave ripples (SWRs) occurring during sleep define periods of hippocampal reactivation of recent experiences and have been causally linked with memory consolidation. Hippocampal SWR-dependent memory consolidation during sleep is often referred to as occurring during an "offline" state, dedicated to processing internally generated neural activity patterns rather than external stimuli. However, the brain is not fully disconnected from the environment during sleep. In particular, sounds heard during sleep are processed by a highly active auditory system which projects to brain regions in the medial temporal lobe, reflecting an anatomical pathway for sound modulation of hippocampal activity. While neural processing of salient sounds during sleep, such as those of a predator or an offspring, is evolutionarily adaptive, whether ongoing processing of environmental sounds during sleep interferes with SWR-dependent memory consolidation remains unknown. To address this question, we used a closed-loop system to deliver non-waking sound stimuli during or following SWRs in sleeping rats. We found that exposure to sounds during sleep suppressed the ripple power and reduced the rate of SWRs. Furthermore, sounds delivered during SWRs (On-SWR) suppressed ripple power significantly more than sounds delivered 2 seconds after SWRs (Off-SWR). Next, we tested the influence of sound presentation during sleep on memory consolidation. To this end, SWR-triggered sounds were applied during sleep sessions following learning of a conditioned place preference paradigm, in which rats learned a place-reward association. We found that On-SWR sound pairing during post-learning sleep induced a complete abolishment of memory retention 24 h following learning, while leaving memory retention immediately following sleep intact. In contrast, Off-SWR pairing weakened memory 24 h following learning as well as immediately following learning. Notably, On-SWR pairing induced a significantly larger impairment in memory 24 h after learning as compared to Off-SWR pairing. Together, these findings suggest that sounds heard during sleep suppress SWRs and memory consolidation, and that the magnitude of these effects are dependent on sound-SWR timing. These results suggest that exposure to environmental sounds during sleep may pose a risk for memory consolidation processes.
Collapse
|
16
|
Ciralli B, Malfatti T, Lima TZ, Silva SRB, Cederroth CR, Leao KE. Alterations of auditory sensory gating in mice with noise-induced tinnitus treated with nicotine and cannabis extract. J Psychopharmacol 2023; 37:1116-1131. [PMID: 37837354 DOI: 10.1177/02698811231200879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Tinnitus is a phantom sound perception affecting both auditory and limbic structures. The mechanisms of tinnitus remain unclear and it is debatable whether tinnitus alters attention to sound and the ability to inhibit repetitive sounds, a phenomenon also known as auditory gating. Here we investigate if noise exposure interferes with auditory gating and whether natural extracts of cannabis or nicotine could improve auditory pre-attentional processing in noise-exposed mice. We used 22 male C57BL/6J mice divided into noise-exposed (exposed to a 9-11 kHz narrow band noise for 1 h) and sham (no sound during noise exposure) groups. Hearing thresholds were measured using auditory brainstem responses, and tinnitus-like behavior was assessed using Gap prepulse inhibition of acoustic startle. After noise exposure, mice were implanted with multi-electrodes in the dorsal hippocampus to assess auditory event-related potentials in response to paired clicks. The results showed that mice with tinnitus-like behavior displayed auditory gating of repetitive clicks, but with larger amplitudes and longer latencies of the N40 component of the aERP waveform. The combination of cannabis extract and nicotine improved the auditory gating ratio in noise-exposed mice without permanent hearing threshold shifts. Lastly, the longer latency of the N40 component appears due to an increased sensitivity to cannabis extract in noise-exposed mice compared to sham mice. The study suggests that the altered central plasticity in tinnitus is more sensitive to the combined actions on the cholinergic and the endocannabinoid systems. Overall, the findings contribute to a better understanding of pharmacological modulation of auditory sensory gating.
Collapse
Affiliation(s)
- Barbara Ciralli
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Thawann Malfatti
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, Brazil
- Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institutet, Sweden
| | - Thiago Z Lima
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, Brazil
- Department of Applied Mathematics and Statistics, Exact and Earth Sciences Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | | | - Christopher R Cederroth
- Experimental Audiology, Department of Physiology and Pharmacology, Karolinska Institutet, Sweden
- Translational Hearing Research, Tübingen Hearing Research Center, Department of Otolaryngology, Head and Neck Surgery, University of Tübingen, Tübingen, Germany
| | - Katarina E Leao
- Brain Institute, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
17
|
Tao C, Zhang GW, Huang JJ, Li Z, Tao HW, Zhang LI. The medial preoptic area mediates depressive-like behaviors induced by ovarian hormone withdrawal through distinct GABAergic projections. Nat Neurosci 2023; 26:1529-1540. [PMID: 37524978 PMCID: PMC11037266 DOI: 10.1038/s41593-023-01397-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 06/26/2023] [Indexed: 08/02/2023]
Abstract
Fluctuations in reproductive hormone levels are associated with mood disruptions in women, such as in postpartum and perimenopausal depression. However, the neural circuit mechanisms remain unclear. Here we report that medial preoptic area (MPOA) GABAergic neurons mediate multifaceted depressive-like behaviors in female mice after ovarian hormone withdrawal (HW), which can be attributed to downregulation of activity in Esr1 (estrogen receptor-1)-expressing GABAergic neurons. Enhancing activity of these neurons ameliorates depressive-like behaviors in HW-treated mice, whereas reducing their activity results in expression of these behaviors. Two separate subpopulations mediate different symptoms: a subpopulation projecting to the ventral tegmental area (VTA) mediates anhedonia and another projecting to the periaqueductal gray mediates immobility. These projections enhance activity of dopaminergic neurons in the VTA and serotonergic neurons in the dorsal raphe, respectively, with increased release of dopamine and serotonin, possibly through disinhibition mechanisms. Thus, the MPOA is a hub that mediates depressive-like behaviors resulting from transitions in reproductive hormone levels.
Collapse
Affiliation(s)
- Can Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Graduate Programs in Biological and Biomedical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhong Li
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Shi Y, Cui H, Li X, Chen L, Zhang C, Zhao X, Li X, Shao Q, Sun Q, Yan K, Wang G. Laminar and dorsoventral organization of layer 1 interneuronal microcircuitry in superficial layers of the medial entorhinal cortex. Cell Rep 2023; 42:112782. [PMID: 37436894 DOI: 10.1016/j.celrep.2023.112782] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/03/2023] [Accepted: 06/24/2023] [Indexed: 07/14/2023] Open
Abstract
Layer 1 (L1) interneurons (INs) participate in various brain functions by gating information flow in the neocortex, but their role in the medial entorhinal cortex (MEC) is still unknown, largely due to scant knowledge of MEC L1 microcircuitry. Using simultaneous triple-octuple whole-cell recordings and morphological reconstructions, we comprehensively depict L1IN networks in the MEC. We identify three morphologically distinct types of L1INs with characteristic electrophysiological properties. We dissect intra- and inter-laminar cell-type-specific microcircuits of L1INs, showing connectivity patterns different from those in the neocortex. Remarkably, motif analysis reveals transitive and clustered features of L1 networks, as well as over-represented trans-laminar motifs. Finally, we demonstrate the dorsoventral gradient of L1IN microcircuits, with dorsal L1 neurogliaform cells receiving fewer intra-laminar inputs but exerting more inhibition on L2 principal neurons. These results thus present a more comprehensive picture of L1IN microcircuitry, which is indispensable for deciphering the function of L1INs in the MEC.
Collapse
Affiliation(s)
- Yuying Shi
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Hui Cui
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaoyue Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Ligu Chen
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Chen Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xinran Zhao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Xiaowan Li
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qiming Shao
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Qiang Sun
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Kaiyue Yan
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Guangfu Wang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|
19
|
Xiao C, Wei J, Zhang GW, Tao C, Huang JJ, Shen L, Wickersham IR, Tao HW, Zhang LI. Glutamatergic and GABAergic neurons in pontine central gray mediate opposing valence-specific behaviors through a global network. Neuron 2023; 111:1486-1503.e7. [PMID: 36893756 PMCID: PMC10164086 DOI: 10.1016/j.neuron.2023.02.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/23/2023] [Accepted: 02/07/2023] [Indexed: 03/11/2023]
Abstract
Extracting the valence of environmental cues is critical for animals' survival. How valence in sensory signals is encoded and transformed to produce distinct behavioral responses remains not well understood. Here, we report that the mouse pontine central gray (PCG) contributes to encoding both negative and positive valences. PCG glutamatergic neurons were activated selectively by aversive, but not reward, stimuli, whereas its GABAergic neurons were preferentially activated by reward signals. The optogenetic activation of these two populations resulted in avoidance and preference behavior, respectively, and was sufficient to induce conditioned place aversion/preference. Suppression of them reduced sensory-induced aversive and appetitive behaviors, respectively. These two functionally opponent populations, receiving a broad range of inputs from overlapping yet distinct sources, broadcast valence-specific information to a distributed brain network with distinguishable downstream effectors. Thus, PCG serves as a critical hub to process positive and negative valences of incoming sensory signals and drive valence-specific behaviors with distinct circuits.
Collapse
Affiliation(s)
- Cuiyu Xiao
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jinxing Wei
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Can Tao
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Graduate Program in Biological and Biomedical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Li Shen
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Center for Neural Circuits and Sensory Processing Disorders, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
20
|
Leprince E, Dard RF, Mortet S, Filippi C, Giorgi-Kurz M, Bourboulou R, Lenck-Santini PP, Picardo MA, Bocchio M, Baude A, Cossart R. Extrinsic control of the early postnatal CA1 hippocampal circuits. Neuron 2023; 111:888-902.e8. [PMID: 36608692 DOI: 10.1016/j.neuron.2022.12.013] [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: 06/03/2022] [Revised: 10/18/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023]
Abstract
The adult CA1 region of the hippocampus produces coordinated neuronal dynamics with minimal reliance on its extrinsic inputs. By contrast, neonatal CA1 is tightly linked to externally generated sensorimotor activity, but the circuit mechanisms underlying early synchronous activity in CA1 remain unclear. Here, using a combination of in vivo and ex vivo circuit mapping, calcium imaging, and electrophysiological recordings in mouse pups, we show that early dynamics in the ventro-intermediate CA1 are under the mixed influence of entorhinal (EC) and thalamic (VMT) inputs. Both VMT and EC can drive internally generated synchronous events ex vivo. However, movement-related population bursts detected in vivo are exclusively driven by the EC. These differential effects on synchrony reflect the different intrahippocampal targets of these inputs. Hence, cortical and subcortical pathways act differently on the neonatal CA1, implying distinct contributions to the development of the hippocampal microcircuit and related cognitive maps.
Collapse
Affiliation(s)
- Erwan Leprince
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Robin F Dard
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Salomé Mortet
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Caroline Filippi
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Marie Giorgi-Kurz
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Romain Bourboulou
- Department of Cell and Developmental Biology, University College London, London, UK
| | | | - Michel A Picardo
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Marco Bocchio
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France; Department of Psychology, Durham University, Durham, UK
| | - Agnès Baude
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France
| | - Rosa Cossart
- Aix Marseille University, INSERM, INMED (UMR1249), Turing Centre for Living systems, Marseille, France.
| |
Collapse
|
21
|
Paciello F, Pisani A, Rinaudo M, Cocco S, Paludetti G, Fetoni AR, Grassi C. Noise-induced auditory damage affects hippocampus causing memory deficits in a model of early age-related hearing loss. Neurobiol Dis 2023; 178:106024. [PMID: 36724860 DOI: 10.1016/j.nbd.2023.106024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/30/2023] Open
Abstract
Several studies identified noise-induced hearing loss (NIHL) as a risk factor for sensory aging and cognitive decline processes, including neurodegenerative diseases, such as dementia and age-related hearing loss (ARHL). Although the association between noise- and age-induced hearing impairment has been widely documented by epidemiological and experimental studies, the molecular mechanisms underlying this association are not fully understood as it is not known how these risk factors (aging and noise) can interact, affecting memory processes. We recently found that early noise exposure in an established animal model of ARHL (C57BL/6 mice) accelerates the onset of age-related cochlear dysfunctions. Here, we extended our previous data by investigating what happens in central brain structures (auditory cortex and hippocampus), to assess the relationship between hearing and memory impairment and the possible combined effect of noise and sensory aging on the cognitive domain. To this aim, we exposed juvenile C57BL/6 mice of 2 months of age to repeated noise sessions (60 min/day, pure tone of 100 dB SPL, 10 kHz, 10 consecutive days) and we monitored auditory threshold by measuring auditory brainstem responses (ABR), spatial working memory, by using the Y-maze test, and basal synaptic transmission by using ex vivo electrophysiological recordings, at different time points (1, 4 and 7 months after the onset of noise exposure, corresponding to 3, 6 and 9 months of age). We found that hearing loss, along with accelerated presbycusis onset, can induce persistent synaptic alterations in the auditory cortex. This was associated with decreased memory performance and oxidative-inflammatory injury in the hippocampus, the extra-auditory structure involved in memory processes. Collectively, our data confirm the critical relationship between auditory and memory circuits, suggesting that the combined detrimental effect of noise and sensory aging on hearing function can be considered a high-risk factor for both sensory and cognitive degenerative processes, given that early noise exposure accelerates presbycusis phenotype and induces hippocampal-dependent memory dysfunctions.
Collapse
Affiliation(s)
- Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Anna Pisani
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Marco Rinaudo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Gaetano Paludetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Anna Rita Fetoni
- Department of Head and Neck Surgery, Università Cattolica del Sacro Cuore, Roma, Italy; Department of Neuroscience, Unit of Audiology, Università degli Studi di Napoli Federico II, Naples, Italy.
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy; Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| |
Collapse
|
22
|
Billig AJ, Lad M, Sedley W, Griffiths TD. The hearing hippocampus. Prog Neurobiol 2022; 218:102326. [PMID: 35870677 PMCID: PMC10510040 DOI: 10.1016/j.pneurobio.2022.102326] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
Abstract
The hippocampus has a well-established role in spatial and episodic memory but a broader function has been proposed including aspects of perception and relational processing. Neural bases of sound analysis have been described in the pathway to auditory cortex, but wider networks supporting auditory cognition are still being established. We review what is known about the role of the hippocampus in processing auditory information, and how the hippocampus itself is shaped by sound. In examining imaging, recording, and lesion studies in species from rodents to humans, we uncover a hierarchy of hippocampal responses to sound including during passive exposure, active listening, and the learning of associations between sounds and other stimuli. We describe how the hippocampus' connectivity and computational architecture allow it to track and manipulate auditory information - whether in the form of speech, music, or environmental, emotional, or phantom sounds. Functional and structural correlates of auditory experience are also identified. The extent of auditory-hippocampal interactions is consistent with the view that the hippocampus makes broad contributions to perception and cognition, beyond spatial and episodic memory. More deeply understanding these interactions may unlock applications including entraining hippocampal rhythms to support cognition, and intervening in links between hearing loss and dementia.
Collapse
Affiliation(s)
| | - Meher Lad
- Translational and Clinical Research Institute, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - William Sedley
- Translational and Clinical Research Institute, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Timothy D Griffiths
- Biosciences Institute, Newcastle University Medical School, Newcastle upon Tyne, UK; Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, London, UK; Human Brain Research Laboratory, Department of Neurosurgery, University of Iowa Hospitals and Clinics, Iowa City, USA
| |
Collapse
|
23
|
Dong R, Lv P, Han Y, Jiang L, Wang Z, Peng L, Ma Z, Xia T, Zhang B, Gu X. Enhancement of astrocytic gap junctions Connexin43 coupling can improve long-term isoflurane anesthesia-mediated brain network abnormalities and cognitive impairment. CNS Neurosci Ther 2022; 28:2281-2297. [PMID: 36153812 PMCID: PMC9627365 DOI: 10.1111/cns.13974] [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] [Received: 07/22/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 02/06/2023] Open
Abstract
AIM Astrocytes are connected by gap junctions Connexin43 (GJs-Cx43) forming an extensive intercellular network and maintain brain homeostasis. Perioperative neurocognitive disorder (PND) occurs frequently after anesthesia/surgery and worsens patient outcome, but the neural circuit mechanisms remain unclear. This study aimed to determine the effects of the GJs-Cx43-mediated astrocytic network on PND and ascertain the underlying neural circuit mechanism. METHODS Male C57BL/6 mice were treated with long-term isoflurane exposure to construct a mouse model of PND. We also exposed primary mouse astrocytes to long-term isoflurane exposure to simulate the conditions of in vivo cognitive dysfunction. Behavioral tests were performed using the Y-maze and fear conditioning (FC) tests. Manganese-enhanced magnetic resonance imaging (MEMRI) and resting-state functional magnetic resonance imaging (rs-fMRI) were used to investigate brain activity and functional connectivity. Western blot and flow cytometry analysis were used to assess protein expression. RESULTS Reconfiguring the astrocytic network by increasing GJs-Cx43 expression can modulate 22 subregions affected by PND in three ways: reversed activation, reversed inhibition, and intensified activation. The brain functional connectivity analysis further suggests that PND is a brain network disorder that includes sleep-wake rhythm-related brain regions, contextual and fear memory-related subregions, the hippocampal-amygdala circuit, the septo-hippocampal circuit, and the entorhinal-hippocampal circuit. Notably, remodeling the astrocytic network by upregulation of GJs-Cx43 can partially reverse the abnormalities in the above circuits. Pathophysiological degeneration in hippocampus is one of the primary hallmarks of PND pathology, and long-term isoflurane anesthesia contributes to oxidative stress and neuroinflammation in the hippocampus. However, promoting the formation of GJs-Cx43 ameliorated cognitive dysfunction induced by long-term isoflurane anesthesia through the attenuation of oxidative stress in hippocampus. CONCLUSION Enhancing GJs-Cx43 coupling can improve brain network abnormalities and cognitive impairment induced by long-term isoflurane anesthesia, its mechanisms might be associated with the regulation of oxidative stress and neuroinflammation.
Collapse
Affiliation(s)
- Rui Dong
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Pin Lv
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yuqiang Han
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Linhao Jiang
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Zimo Wang
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Liangyu Peng
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Zhengliang Ma
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| | - Tianjiao Xia
- Medical SchoolNanjing UniversityNanjingChina,Jiangsu Key Laboratory of Molecular MedicineNanjingChina
| | - Bing Zhang
- Department of RadiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina,Jiangsu Key Laboratory of Molecular MedicineNanjingChina,Institute of Medical Imaging and Artificial IntelligenceNanjing UniversityNanjingChina,Institute of Brain ScienceNanjing UniversityNanjingChina
| | - Xiaoping Gu
- Department of AnesthesiologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjingChina
| |
Collapse
|
24
|
Kang SJ, Liu S, Ye M, Kim DI, Pao GM, Copits BA, Roberts BZ, Lee KF, Bruchas MR, Han S. A central alarm system that gates multi-sensory innate threat cues to the amygdala. Cell Rep 2022; 40:111222. [PMID: 35977501 PMCID: PMC9420642 DOI: 10.1016/j.celrep.2022.111222] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/16/2022] [Accepted: 07/22/2022] [Indexed: 12/31/2022] Open
Abstract
Perception of threats is essential for survival. Previous findings suggest that parallel pathways independently relay innate threat signals from different sensory modalities to multiple brain areas, such as the midbrain and hypothalamus, for immediate avoidance. Yet little is known about whether and how multi-sensory innate threat cues are integrated and conveyed from each sensory modality to the amygdala, a critical brain area for threat perception and learning. Here, we report that neurons expressing calcitonin gene-related peptide (CGRP) in the parvocellular subparafascicular nucleus in the thalamus and external lateral parabrachial nucleus in the brainstem respond to multi-sensory threat cues from various sensory modalities and relay negative valence to the lateral and central amygdala, respectively. Both CGRP populations and their amygdala projections are required for multi-sensory threat perception and aversive memory formation. The identification of unified innate threat pathways may provide insights into developing therapeutic candidates for innate fear-related disorders.
Collapse
Affiliation(s)
- Sukjae J Kang
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Shijia Liu
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mao Ye
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Dong-Il Kim
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Gerald M Pao
- Molecular and Cellular Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Okinawa Institute of Science and Technology Graduate University, 1919-1 Tancha, Onna-son, Okinawa 904-0495, Japan
| | - Bryan A Copits
- Washington University Pain Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Benjamin Z Roberts
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Kuo-Fen Lee
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Michael R Bruchas
- Center of Excellence in the Neurobiology of Addiction, Pain, and Emotion, Departments of Anesthesiology and Pain Medicine, and Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Sung Han
- Peptide Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA; Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
25
|
Dard RF, Leprince E, Denis J, Rao Balappa S, Suchkov D, Boyce R, Lopez C, Giorgi-Kurz M, Szwagier T, Dumont T, Rouault H, Minlebaev M, Baude A, Cossart R, Picardo MA. The rapid developmental rise of somatic inhibition disengages hippocampal dynamics from self-motion. eLife 2022; 11:e78116. [PMID: 35856497 PMCID: PMC9363116 DOI: 10.7554/elife.78116] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Early electrophysiological brain oscillations recorded in preterm babies and newborn rodents are initially mostly driven by bottom-up sensorimotor activity and only later can detach from external inputs. This is a hallmark of most developing brain areas, including the hippocampus, which, in the adult brain, functions in integrating external inputs onto internal dynamics. Such developmental disengagement from external inputs is likely a fundamental step for the proper development of cognitive internal models. Despite its importance, the developmental timeline and circuit basis for this disengagement remain unknown. To address this issue, we have investigated the daily evolution of CA1 dynamics and underlying circuits during the first two postnatal weeks of mouse development using two-photon calcium imaging in non-anesthetized pups. We show that the first postnatal week ends with an abrupt shift in the representation of self-motion in CA1. Indeed, most CA1 pyramidal cells switch from activated to inhibited by self-generated movements at the end of the first postnatal week, whereas the majority of GABAergic neurons remain positively modulated throughout this period. This rapid switch occurs within 2 days and follows the rapid anatomical and functional surge of local somatic GABAergic innervation. The observed change in dynamics is consistent with a two-population model undergoing a strengthening of inhibition. We propose that this abrupt developmental transition inaugurates the emergence of internal hippocampal dynamics.
Collapse
Affiliation(s)
- Robin F Dard
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Erwan Leprince
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Julien Denis
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Shrisha Rao Balappa
- Turing Centre for Living systems, Aix-Marseille University, Université de Toulon, CNRS, CPT (UMR 7332)MarseilleFrance
| | - Dmitrii Suchkov
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Richard Boyce
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Catherine Lopez
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Marie Giorgi-Kurz
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Tom Szwagier
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
- Mines ParisTech, PSL Research UniversityParisFrance
| | - Théo Dumont
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
- Mines ParisTech, PSL Research UniversityParisFrance
| | - Hervé Rouault
- Turing Centre for Living systems, Aix-Marseille University, Université de Toulon, CNRS, CPT (UMR 7332)MarseilleFrance
| | - Marat Minlebaev
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Agnès Baude
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Rosa Cossart
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| | - Michel A Picardo
- Turing Centre for Living systems, Aix-Marseille University, INSERM, INMED U1249MarseilleFrance
| |
Collapse
|
26
|
Ojala KE, Staib M, Gerster S, Ruff CC, Bach DR. Inhibiting Human Aversive Memory by Transcranial Theta-Burst Stimulation to the Primary Sensory Cortex. Biol Psychiatry 2022; 92:149-157. [PMID: 35410762 DOI: 10.1016/j.biopsych.2022.01.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/14/2022] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
BACKGROUND Predicting adverse events from past experience is fundamental for many biological organisms. However, some individuals suffer from maladaptive memories that impair behavioral control and well-being, e.g., after psychological trauma. Inhibiting the formation and maintenance of such memories would have high clinical relevance. Previous preclinical research has focused on systemically administered pharmacological interventions, which cannot be targeted to specific neural circuits in humans. Here, we investigated the potential of noninvasive neural stimulation on the human sensory cortex in inhibiting aversive memory in a laboratory threat conditioning model. METHODS We build on an emerging nonhuman literature suggesting that primary sensory cortices may be crucially required for threat memory formation and consolidation. Immediately before conditioning innocuous somatosensory stimuli (conditioned stimuli [CS]) to aversive electric stimulation, healthy human participants received continuous theta-burst transcranial magnetic stimulation (cTBS) to individually localized primary somatosensory cortex in either the CS-contralateral (experimental) or CS-ipsilateral (control) hemisphere. We measured fear-potentiated startle to infer threat memory retention on the next day, as well as skin conductance and pupil size during learning. RESULTS After overnight consolidation, threat memory was attenuated in the experimental group compared with the control cTBS group. There was no evidence that this differed between simple and complex CS or that CS identification or initial learning were affected by cTBS. CONCLUSIONS Our results suggest that cTBS to the primary sensory cortex inhibits threat memory, likely by an impact on postlearning consolidation. We propose that noninvasive targeted stimulation of the sensory cortex may provide a new avenue for interfering with aversive memories in humans.
Collapse
Affiliation(s)
- Karita E Ojala
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland.
| | - Matthias Staib
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland
| | - Samuel Gerster
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland
| | - Christian C Ruff
- Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland; Zurich Center for Neuroeconomics, Department of Economics, University of Zürich, Zürich, Switzerland
| | - Dominik R Bach
- Computational Psychiatry Research, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zürich, Zürich, Switzerland; Neuroscience Centre Zurich, University of Zürich, Zürich, Switzerland; Wellcome Centre for Human Neuroimaging and Max-Planck UCL Centre for Computational Psychiatry and Ageing Research, University College London, London, United Kingdom.
| |
Collapse
|
27
|
Luo B, Li J, Liu J, Li F, Gu M, Xiao H, Lei S, Xiao Z. Frequency-Dependent Plasticity in the Temporal Association Cortex Originates from the Primary Auditory Cortex, and Is Modified by the Secondary Auditory Cortex and the Medial Geniculate Body. J Neurosci 2022; 42:5254-5267. [PMID: 35613891 PMCID: PMC9236291 DOI: 10.1523/jneurosci.1481-21.2022] [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: 07/21/2021] [Revised: 05/08/2022] [Accepted: 05/11/2022] [Indexed: 12/31/2022] Open
Abstract
The brain areas that mediate the formation of auditory threat memory and perceptual decisions remain uncertain to date. Candidates include the primary (A1) and secondary (A2) auditory cortex, the medial division of the medial geniculate body (MGm), amygdala, and the temporal association cortex. We used chemogenetic and optogenetic manipulations with in vivo and in vitro patch-clamp recordings to assess the roles of these brain regions in threat memory learning in female mice. We found that conditioned sound (CS) frequency-dependent plasticity resulted in the formation of auditory threat memory in the temporal association cortex. This neural correlated auditory threat memory depended on CS frequency information from A1 glutamatergic subthreshold monosynaptic inputs, CS lateral inhibition from A2 glutamatergic disynaptic inputs, and non-frequency-specific facilitation from MGm glutamatergic monosynaptic inputs. These results indicate that the A2 and MGm work together in an inhibitory-facilitative role.SIGNIFICANCE STATEMENT: The ability to recognize specific sounds to avoid predators or seek prey is a useful survival tool. Improving this ability through experiential learning is an added advantage requiring neural plasticity. As an example, humans must learn to distinguish the sound of a car horn, and thus avoid oncoming traffic. Our research discovered that the temporal association cortex can encode this kind of auditory information through tonal receptive field plasticity. In addition, the results revealed the underlying synaptic mechanisms of this process. These results extended our understanding of how meaningful auditory information is processed in an animal's brain.
Collapse
Affiliation(s)
- Bingmin Luo
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jing Li
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jingpeng Liu
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Funi Li
- General Practice Center, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong 528244, China
| | - Miaoqing Gu
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Haoran Xiao
- General Practice Center, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong 528244, China
| | - Shujun Lei
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhongju Xiao
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou, Guangdong 510515, China
- General Practice Center, The Seventh Affiliated Hospital, Southern Medical University, Foshan, Guangdong 528244, China
| |
Collapse
|
28
|
Sun W, Tang P, Liang Y, Li J, Feng J, Zhang N, Lu D, He J, Chen X. The anterior cingulate cortex directly enhances auditory cortical responses in air-puffing-facilitated flight behavior. Cell Rep 2022; 38:110506. [PMID: 35263590 DOI: 10.1016/j.celrep.2022.110506] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 11/11/2021] [Accepted: 02/16/2022] [Indexed: 12/17/2022] Open
Abstract
For survival, animals encode prominent events in complex environments, which modulates their defense behavior. Here, we design a paradigm that assesses how a mild aversive cue (i.e., mild air puff) interacts with sound-evoked flight behavior in mice. We find that air puffing facilitates sound-evoked flight behavior by enhancing the auditory responses of auditory cortical neurons. We then find that the anterior part of the anterior cingulate cortex (ACC) encodes the valence of air puffing and modulates the auditory cortex through anatomical examination, physiological recordings, and optogenetic/chemogenetic manipulations. Activating ACC projections to the auditory cortex simulates the facilitating effect of air puffing, whereas inhibiting the ACC or its projections to the auditory cortex neutralizes this facilitating effect. These findings show that the ACC regulates sound-evoked flight behavior by potentiating neuronal responses in the auditory cortex.
Collapse
Affiliation(s)
- Wenjian Sun
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 0000 Hong Kong SAR, P.R. China
| | - Peng Tang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 0000 Hong Kong SAR, P.R. China
| | - Ye Liang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China
| | - Jing Li
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 0000 Hong Kong SAR, P.R. China
| | - Jingyu Feng
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China
| | - Nan Zhang
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, 0000 Hong Kong SAR, P.R. China
| | - Danyi Lu
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China
| | - Jufang He
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen 518507, P.R. China.
| | - Xi Chen
- Department of Neuroscience, City University of Hong Kong, Kowloon Tong, 0000 Hong Kong SAR, P.R. China; City University of Hong Kong Shenzhen Research Institute, Shenzhen 518507, P.R. China.
| |
Collapse
|
29
|
Shen L, Zhang GW, Tao C, Seo MB, Zhang NK, Huang JJ, Zhang LI, Tao HW. A bottom-up reward pathway mediated by somatostatin neurons in the medial septum complex underlying appetitive learning. Nat Commun 2022; 13:1194. [PMID: 35256596 PMCID: PMC8901785 DOI: 10.1038/s41467-022-28854-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 02/16/2022] [Indexed: 12/11/2022] Open
Abstract
Valence detection and processing are essential for the survival of animals and their life quality in complex environments. Neural circuits underlying the transformation of external sensory signals into positive valence coding to generate appropriate behavioral responses remain not well-studied. Here, we report that somatostatin (SOM) subtype of GABAergic neurons in the mouse medial septum complex (MS), but not parvalbumin subtype or glutamatergic neurons, specifically encode reward signals and positive valence. Through an ascending pathway from the nucleus of solitary tract and then parabrachial nucleus, the MS SOM neurons receive rewarding taste signals and suppress the lateral habenula. They contribute essentially to appetitive associative learning via their projections to the lateral habenula: learning enhances their responses to reward-predictive sensory cues, and suppressing their responses to either conditioned or unconditioned stimulus impairs acquisition of reward learning. Thus, MS serves as a critical hub for transforming bottom-up sensory signals to mediate appetitive behaviors.
Collapse
Affiliation(s)
- Li Shen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Can Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Michelle B Seo
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA.,Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, 90089, USA
| | - Nicole K Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA.,Graduate Programs in Biomedical and Biological Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA. .,Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA.
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA. .,Department of Physiology & Neuroscience, Keck School of Medicine, University of Southern California, 1501 San Pablo Street, Los Angeles, CA, 90033, USA.
| |
Collapse
|
30
|
Neuronal activity in sensory cortex predicts the specificity of learning in mice. Nat Commun 2022; 13:1167. [PMID: 35246528 PMCID: PMC8897443 DOI: 10.1038/s41467-022-28784-w] [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: 02/05/2021] [Accepted: 01/27/2022] [Indexed: 11/08/2022] Open
Abstract
Learning to avoid dangerous signals while preserving normal responses to safe stimuli is essential for everyday behavior and survival. Following identical experiences, subjects exhibit fear specificity ranging from high (specializing fear to only the dangerous stimulus) to low (generalizing fear to safe stimuli), yet the neuronal basis of fear specificity remains unknown. Here, we identified the neuronal code that underlies inter-subject variability in fear specificity using longitudinal imaging of neuronal activity before and after differential fear conditioning in the auditory cortex of mice. Neuronal activity prior to, but not after learning predicted the level of specificity following fear conditioning across subjects. Stimulus representation in auditory cortex was reorganized following conditioning. However, the reorganized neuronal activity did not relate to the specificity of learning. These results present a novel neuronal code that determines individual patterns in learning. The neural mechanisms underpinning the specificity of fear memories remains poorly understood. Here, the authors highlight how neural activity prior to fear learning impacts fear memory specificity.
Collapse
|
31
|
Seeking motivation and reward: roles of dopamine, hippocampus and supramammillo-septal pathway. Prog Neurobiol 2022; 212:102252. [PMID: 35227866 PMCID: PMC8961455 DOI: 10.1016/j.pneurobio.2022.102252] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/09/2022] [Accepted: 02/23/2022] [Indexed: 01/07/2023]
Abstract
Reinforcement learning and goal-seeking behavior are thought to be mediated by midbrain dopamine neurons. However, little is known about neural substrates of curiosity and exploratory behavior, which occur in the absence of clear goal or reward. This is despite behavioral scientists having long suggested that curiosity and exploratory behaviors are regulated by an innate drive. We refer to such behavior as information-seeking behavior and propose 1) key neural substrates and 2) the concept of environment prediction error as a framework to understand information-seeking processes. The cognitive aspect of information-seeking behavior, including the perception of salience and uncertainty, involves, in part, the pathways from the posterior hypothalamic supramammillary region to the hippocampal formation. The vigor of such behavior is modulated by the following: supramammillary glutamatergic neurons; their projections to medial septal glutamatergic neurons; and the projections of medial septal glutamatergic neurons to ventral tegmental dopaminergic neurons. Phasic responses of dopaminergic neurons are characterized as signaling potentially important stimuli rather than rewards. This paper describes how novel stimuli and uncertainty trigger seeking motivation and how these neural substrates modulate information-seeking behavior.
Collapse
|
32
|
Yi GL, Zhu MZ, Cui HC, Yuan XR, Liu P, Tang J, Li YQ, Zhu XH. A hippocampus dependent neural circuit loop underlying the generation of auditory mismatch negativity. Neuropharmacology 2022; 206:108947. [PMID: 35026286 DOI: 10.1016/j.neuropharm.2022.108947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/11/2021] [Accepted: 01/06/2022] [Indexed: 11/16/2022]
Abstract
Extracting relevant information and transforming it into appropriate behavior, is a fundamental brain function, and requires the coordination between the sensory and cognitive systems, however, the underlying mechanisms of interplay between sensory and cognition systems remain largely unknown. Here, we developed a mouse model for mimicking human auditory mismatch negativity (MMN), a well-characterized translational biomarker for schizophrenia, and an index of early auditory information processing. We found that a subanesthetic dose of ketamine decreased the amplitude of MMN in adult mice. Using pharmacological and chemogenetic approaches, we identified an auditory cortex-entorhinal cortex-hippocampus neural circuit loop that is required for the generation of MMN. In addition, we found that inhibition of dCA1→MEC circuit impaired the auditory related fear discrimination. Moreover, we found that ketamine induced MMN deficiency by inhibition of long-range GABAergic projection from the CA1 region of the dorsal hippocampus to the medial entorhinal cortex. These results provided circuit insights for ketamine effects and early auditory information processing. As the entorhinal cortex is the interface between the neocortex and hippocampus, and the hippocampus is critical for the formation, consolidation, and retrieval of episodic memories and other cognition, our results provide a neural mechanism for the interplay between the sensory and cognition systems.
Collapse
Affiliation(s)
- Guo-Liang Yi
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Min-Zhen Zhu
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - He-Chen Cui
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin-Rui Yuan
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Peng Liu
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jie Tang
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Yuan-Qing Li
- Research Center for Brain-Computer Interface, Pazhou Lab, Guangzhou, 510330, China
| | - Xin-Hong Zhu
- Institute of Mental Health, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou, 510330, China; School of Psychology, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
33
|
Robert B, Kimchi EY, Watanabe Y, Chakoma T, Jing M, Li Y, Polley DB. A functional topography within the cholinergic basal forebrain for encoding sensory cues and behavioral reinforcement outcomes. eLife 2021; 10:e69514. [PMID: 34821218 PMCID: PMC8654357 DOI: 10.7554/elife.69514] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 11/16/2021] [Indexed: 01/01/2023] Open
Abstract
Basal forebrain cholinergic neurons (BFCNs) project throughout the cortex to regulate arousal, stimulus salience, plasticity, and learning. Although often treated as a monolithic structure, the basal forebrain features distinct connectivity along its rostrocaudal axis that could impart regional differences in BFCN processing. Here, we performed simultaneous bulk calcium imaging from rostral and caudal BFCNs over a 1-month period of variable reinforcement learning in mice. BFCNs in both regions showed equivalently weak responses to unconditioned visual stimuli and anticipated rewards. Rostral BFCNs in the horizontal limb of the diagonal band were more responsive to reward omission, more accurately classified behavioral outcomes, and more closely tracked fluctuations in pupil-indexed global brain state. Caudal tail BFCNs in globus pallidus and substantia innominata were more responsive to unconditioned auditory stimuli, orofacial movements, aversive reinforcement, and showed robust associative plasticity for punishment-predicting cues. These results identify a functional topography that diversifies cholinergic modulatory signals broadcast to downstream brain regions.
Collapse
Affiliation(s)
- Blaise Robert
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear InfirmaryBostonUnited States
| | - Eyal Y Kimchi
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear InfirmaryBostonUnited States
- Department of Neurology, Massachusetts General HospitalBostonUnited States
| | - Yurika Watanabe
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear InfirmaryBostonUnited States
| | - Tatenda Chakoma
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear InfirmaryBostonUnited States
| | - Miao Jing
- Chinese Institute for Brain ResearchBeijingChina
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research; Peking-Tsinghua Center for Life Sciences, BeijingBeijingChina
| | - Daniel B Polley
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear InfirmaryBostonUnited States
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
34
|
Feng H, Su J, Fang W, Chen X, He J. The entorhinal cortex modulates trace fear memory formation and neuroplasticity in the mouse lateral amygdala via cholecystokinin. eLife 2021; 10:69333. [PMID: 34779397 PMCID: PMC8629425 DOI: 10.7554/elife.69333] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/12/2021] [Indexed: 12/02/2022] Open
Abstract
Although fear memory formation is essential for survival and fear-related mental disorders, the neural circuitry and mechanism are incompletely understood. Here, we utilized trace fear conditioning to study the formation of trace fear memory in mice. We identified the entorhinal cortex (EC) as a critical component of sensory signaling to the amygdala. We adopted both loss-of-function and gain-of-function experiments to demonstrate that release of the cholecystokinin (CCK) from the EC is required for trace fear memory formation. We discovered that CCK-positive neurons project from the EC to the lateral nuclei of the amygdala (LA), and inhibition of CCK-dependent signaling in the EC prevented long-term potentiation of the auditory response in the LA and formation of trace fear memory. In summary, high-frequency activation of EC neurons triggers the release of CCK in their projection terminals in the LA, potentiating auditory response in LA neurons. The neural plasticity in the LA leads to trace fear memory formation.
Collapse
Affiliation(s)
- Hemin Feng
- Departments of Neuroscience and Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
| | - Junfeng Su
- Departments of Neuroscience and Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangzhou, China
| | - Wei Fang
- Departments of Neuroscience and Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Xi Chen
- Departments of Neuroscience and Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangzhou, China
| | - Jufang He
- Departments of Neuroscience and Biomedical Sciences, City University of Hong Kong, Hong Kong, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangzhou, China
| |
Collapse
|
35
|
Paciello F, Rinaudo M, Longo V, Cocco S, Conforto G, Pisani A, Podda MV, Fetoni AR, Paludetti G, Grassi C. Auditory sensory deprivation induced by noise exposure exacerbates cognitive decline in a mouse model of Alzheimer's disease. eLife 2021; 10:70908. [PMID: 34699347 PMCID: PMC8547960 DOI: 10.7554/elife.70908] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/15/2021] [Indexed: 12/18/2022] Open
Abstract
Although association between hearing impairment and dementia has been widely documented by epidemiological studies, the role of auditory sensory deprivation in cognitive decline remains to be fully understood. To address this issue we investigated the impact of hearing loss on the onset and time-course of cognitive decline in an animal model of Alzheimer's disease (AD), that is the 3×Tg-AD mice and the underlying mechanisms. We found that hearing loss induced by noise exposure in the 3×Tg-AD mice before the phenotype is manifested caused persistent synaptic and morphological alterations in the auditory cortex. This was associated with earlier hippocampal dysfunction, increased tau phosphorylation, neuroinflammation, and redox imbalance, along with anticipated memory deficits compared to the expected time-course of the neurodegenerative phenotype. Our data suggest that a mouse model of AD is more vulnerable to central damage induced by hearing loss and shows reduced ability to counteract noise-induced detrimental effects, which accelerates the neurodegenerative disease onset.
Collapse
Affiliation(s)
- Fabiola Paciello
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valentina Longo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sara Cocco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giulia Conforto
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Pisani
- Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Vittoria Podda
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Anna Rita Fetoni
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Gaetano Paludetti
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Otolaryngology Head and Neck Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
36
|
Wu X, Morishita W, Beier KT, Heifets BD, Malenka RC. 5-HT modulation of a medial septal circuit tunes social memory stability. Nature 2021; 599:96-101. [PMID: 34616037 DOI: 10.1038/s41586-021-03956-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022]
Abstract
Social memory-the ability to recognize and remember familiar conspecifics-is critical for the survival of an animal in its social group1,2. The dorsal CA2 (dCA2)3-5 and ventral CA1 (vCA1)6 subregions of the hippocampus, and their projection targets6,7, have important roles in social memory. However, the relevant extrahippocampal input regions remain poorly defined. Here we identify the medial septum (MS) as a dCA2 input region that is critical for social memory and reveal that modulation of the MS by serotonin (5-HT) bidirectionally controls social memory formation, thereby affecting memory stability. Novel social interactions increase activity in dCA2-projecting MS neurons and induce plasticity at glutamatergic synapses from MS neurons onto dCA2 pyramidal neurons. The activity of dCA2-projecting MS cells is enhanced by the neuromodulator 5-HT acting on 5-HT1B receptors. Moreover, optogenetic manipulation of median raphe 5-HT terminals in the MS bidirectionally regulates social memory stability. This work expands our understanding of the neural mechanisms by which social interactions lead to social memory and provides evidence that 5-HT has a critical role in promoting not only prosocial behaviours8,9, but also social memory, by influencing distinct target structures.
Collapse
Affiliation(s)
- Xiaoting Wu
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Wade Morishita
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Kevin T Beier
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, USA
| | - Robert C Malenka
- Nancy Pritzker Laboratory, Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Abstract
In mammals, the selective transformation of transient experience into stored memory occurs in the hippocampus, which develops representations of specific events in the context in which they occur. In this review, we focus on the development of hippocampal circuits and the self-organized dynamics embedded within them since the latter critically support the role of the hippocampus in learning and memory. We first discuss evidence that adult hippocampal cells and circuits are sculpted by development as early as during embryonic neurogenesis. We argue that these primary developmental programs provide a scaffold onto which later experience of the external world can be grafted. Next, we review the different sequences in the development of hippocampal cells and circuits at anatomical and functional levels. We cover a period extending from neurogenesis and migration to the appearance of phenotypic diversity within hippocampal cells, and their wiring into functional networks. We describe the progressive emergence of network dynamics in the hippocampus, from sensorimotor-driven early sharp waves to sequences of place cells tracking relational information. We outline the critical turn points and discontinuities in that developmental journey, and close by formulating open questions. We propose that rewinding the process of hippocampal development helps understand the main organization principles of memory circuits.
Collapse
Affiliation(s)
- Rosa Cossart
- Inserm, INMED, Turing Center for Living Systems, Aix Marseille University, Marseille, France
| | - Rustem Khazipov
- Inserm, INMED, Turing Center for Living Systems, Aix Marseille University, Marseille, France.,Laboratory of Neurobiology, Kazan Federal University, Kazan Russia
| |
Collapse
|
38
|
Brain–stomach coupling: Anatomy, functions, and future avenues of research. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100270] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
39
|
de Deus JL, Amorim MR, Ribeiro AB, Barcellos-Filho PCG, Ceballos CC, Branco LGS, Cunha AOS, Leão RM. Loss of Brain-Derived Neurotrophic Factor Mediates Inhibition of Hippocampal Long-Term Potentiation by High-Intensity Sound. Cell Mol Neurobiol 2021; 41:751-763. [PMID: 32445041 PMCID: PMC11448697 DOI: 10.1007/s10571-020-00881-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 05/16/2020] [Indexed: 01/15/2023]
Abstract
Exposure to noise produces cognitive and emotional disorders, and recent studies have shown that auditory stimulation or deprivation affects hippocampal function. Previously, we showed that exposure to high-intensity sound (110 dB, 1 min) strongly inhibits Schaffer-CA1 long-term potentiation (LTP). Here we investigated possible mechanisms involved in this effect. We found that exposure to 110 dB sound activates c-fos expression in hippocampal CA1 and CA3 neurons. Although sound stimulation did not affect glutamatergic or GABAergic neurotransmission in CA1, it did depress the level of brain-derived neurotrophic factor (BDNF), which is involved in promoting hippocampal synaptic plasticity. Moreover, perfusion of slices with BDNF rescued LTP in animals exposed to sound stimulation, whereas BDNF did not affect LTP in sham-stimulated rats. Furthermore, LM22A4, a TrkB receptor agonist, also rescued LTP from sound-stimulated animals. Our results indicate that depression of hippocampal BDNF mediates the inhibition of LTP produced by high-intensity sound stimulation.
Collapse
Affiliation(s)
- Júnia L de Deus
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Mateus R Amorim
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Aline B Ribeiro
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Procópio C G Barcellos-Filho
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - César C Ceballos
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Luiz Guilherme S Branco
- Department of Basic and Oral Biology, Dental School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14040-904, Brazil
| | - Alexandra O S Cunha
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Ricardo M Leão
- Department of Physiology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, 14049-900, Brazil.
| |
Collapse
|
40
|
Li Z, Wei JX, Zhang GW, Huang JJ, Zingg B, Wang X, Tao HW, Zhang LI. Corticostriatal control of defense behavior in mice induced by auditory looming cues. Nat Commun 2021; 12:1040. [PMID: 33589613 PMCID: PMC7884702 DOI: 10.1038/s41467-021-21248-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 01/14/2021] [Indexed: 01/31/2023] Open
Abstract
Animals exhibit innate defense behaviors in response to approaching threats cued by the dynamics of sensory inputs of various modalities. The underlying neural circuits have been mostly studied in the visual system, but remain unclear for other modalities. Here, by utilizing sounds with increasing (vs. decreasing) loudness to mimic looming (vs. receding) objects, we find that looming sounds elicit stereotypical sequential defensive reactions: freezing followed by flight. Both behaviors require the activity of auditory cortex, in particular the sustained type of responses, but are differentially mediated by corticostriatal projections primarily innervating D2 neurons in the tail of the striatum and corticocollicular projections to the superior colliculus, respectively. The behavioral transition from freezing to flight can be attributed to the differential temporal dynamics of the striatal and collicular neurons in their responses to looming sound stimuli. Our results reveal an essential role of the striatum in the innate defense control.
Collapse
Affiliation(s)
- Zhong Li
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jin-Xing Wei
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Guang-Wei Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Junxiang J Huang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Program in Biomedical and Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Brian Zingg
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Xiyue Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Noise Exposure Alters Glutamatergic and GABAergic Synaptic Connectivity in the Hippocampus and Its Relevance to Tinnitus. Neural Plast 2021; 2021:8833087. [PMID: 33510780 PMCID: PMC7822664 DOI: 10.1155/2021/8833087] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 12/16/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
Accumulating evidence implicates a role for brain structures outside the ascending auditory pathway in tinnitus, the phantom perception of sound. In addition to other factors such as age-dependent hearing loss, high-level sound exposure is a prominent cause of tinnitus. Here, we examined how noise exposure altered the distribution of excitatory and inhibitory synaptic inputs in the guinea pig hippocampus and determined whether these changes were associated with tinnitus. In experiment one, guinea pigs were overexposed to unilateral narrow-band noise (98 dB SPL, 2 h). Two weeks later, the density of excitatory (VGLUT-1/2) and inhibitory (VGAT) synaptic terminals in CA1, CA3, and dentate gyrus hippocampal subregions was assessed by immunohistochemistry. Overall, VGLUT-1 density primarily increased, while VGAT density decreased significantly in many regions. Then, to assess whether the noise-induced alterations were persistent and related to tinnitus, experiment two utilized a noise-exposure paradigm shown to induce tinnitus and assessed tinnitus development which was assessed using gap-prepulse inhibition of the acoustic startle (GPIAS). Twelve weeks after sound overexposure, changes in excitatory synaptic terminal density had largely recovered regardless of tinnitus status, but the recovery of GABAergic terminal density was dramatically different in animals expressing tinnitus relative to animals resistant to tinnitus. In resistant animals, inhibitory synapse density recovered to preexposure levels, but in animals expressing tinnitus, inhibitory synapse density remained chronically diminished. Taken together, our results suggest that noise exposure induces striking changes in the balance of excitatory and inhibitory synaptic inputs throughout the hippocampus and reveal a potential role for rebounding inhibition in the hippocampus as a protective factor leading to tinnitus resilience.
Collapse
|
42
|
Abstract
Tinnitus is a phantom auditory sensation in the absence of external sounds, while hyperacusis is an atypical sensitivity to external sounds that leads them to be perceived as abnormally loud or even painful. Both conditions may reflect the brain's over-compensation for reduced input from the ear. The present work differentiates between two compensation models: The additive central noise compensates for hearing loss and is likely to generate tinnitus, whereas the multiplicative central gain compensates for hidden hearing loss and is likely to generate hyperacusis. Importantly, both models predict increased variance in central representations of sounds, especially a nonlinear increase in variance by the central gain. The increased central variance limits the amount of central compensation and reduces temporal synchrony, which can explain the insufficient central gain reported in the literature. Future studies need to collect trial-by-trial firing variance data so that the present variance-based model can be falsified.
Collapse
Affiliation(s)
- Fan-Gang Zeng
- Departments of Anatomy and Neurobiology, Biomedical Engineering, Cognitive Sciences, and Otolaryngology - Head and Neck Surgery, Center for Hearing Research, University of California Irvine
| |
Collapse
|
43
|
Postal O, Dupont T, Bakay W, Dominique N, Petit C, Michalski N, Gourévitch B. Spontaneous Mouse Behavior in Presence of Dissonance and Acoustic Roughness. Front Behav Neurosci 2020; 14:588834. [PMID: 33132864 PMCID: PMC7578920 DOI: 10.3389/fnbeh.2020.588834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/08/2020] [Indexed: 11/13/2022] Open
Abstract
According to a novel hypothesis (Arnal et al., 2015, Current Biology 25:2051-2056), auditory roughness, or temporal envelope modulations between 30 and 150 Hz, are present in both natural and artificial human alarm signals, which boosts the detection of these alarms in various tasks. These results also shed new light on the unpleasantness of dissonant sounds to humans, which builds upon the high level of roughness present in such sounds. However, it is not clear whether this hypothesis also applies to other species, such as rodents. In particular, whether consonant/dissonant chords, and particularly whether auditory roughness, can trigger unpleasant sensations in mice remains unknown. Using an autonomous behavioral system, which allows the monitoring of mouse behavior over a period of weeks, we observed that C57Bl6J mice did not show any preference for consonant chords. In addition, we found that mice showed a preference for rough sounds over sounds having amplitude modulations in their temporal envelope outside the "rough" range. These results suggest that some emotional features carried by the acoustic temporal envelope are likely to be species-specific.
Collapse
Affiliation(s)
- Olivier Postal
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
- Sorbonne Université, Collège Doctoral, Paris, France
| | - Typhaine Dupont
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
| | - Warren Bakay
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
| | - Noémi Dominique
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
| | - Christine Petit
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
- Syndrome de Usher et Autres Atteintes Rétino-Cochléaires, Institut de la Vision, Paris, France
- Collège de France, Paris, France
| | | | - Boris Gourévitch
- Institut de l’Audition, Institut Pasteur, INSERM, Paris, France
- CNRS, Paris, France
| |
Collapse
|
44
|
Liang F, Li H, Chou XL, Zhou M, Zhang NK, Xiao Z, Zhang KK, Tao HW, Zhang LI. Sparse Representation in Awake Auditory Cortex: Cell-type Dependence, Synaptic Mechanisms, Developmental Emergence, and Modulation. Cereb Cortex 2020; 29:3796-3812. [PMID: 30307493 DOI: 10.1093/cercor/bhy260] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/29/2018] [Accepted: 09/19/2018] [Indexed: 01/25/2023] Open
Abstract
Sparse representation is considered an important coding strategy for cortical processing in various sensory modalities. It remains unclear how cortical sparseness arises and is being regulated. Here, unbiased recordings from primary auditory cortex of awake adult mice revealed salient sparseness in layer (L)2/3, with a majority of excitatory neurons exhibiting no increased spiking in response to each of sound types tested. Sparse representation was not observed in parvalbumin (PV) inhibitory neurons. The nonresponding neurons did receive auditory-evoked synaptic inputs, marked by weaker excitation and lower excitation/inhibition (E/I) ratios than responding cells. Sparse representation arises during development in an experience-dependent manner, accompanied by differential changes of excitatory input strength and a transition from unimodal to bimodal distribution of E/I ratios. Sparseness level could be reduced by suppressing PV or L1 inhibitory neurons. Thus, sparse representation may be dynamically regulated via modulating E/I balance, optimizing cortical representation of the external sensory world.
Collapse
Affiliation(s)
- Feixue Liang
- Department of Medical Engineering, School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Haifu Li
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiao-Lin Chou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Mu Zhou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089, USA
| | - Nicole K Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zhongju Xiao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Ke K Zhang
- Department of Pathology, the University of North Dakota, Grand Forks, ND, USA
| | - Huizhong W Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
45
|
Xie H, Wu D, Gao X, Wang N, Xiao Z. Auditory Fear Conditioning Alters Sensitivity of the Medial Prefrontal Cortex but this is not based on Frequency-dependent Integration. Neuroscience 2020; 442:237-252. [PMID: 32505746 DOI: 10.1016/j.neuroscience.2020.05.038] [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: 01/30/2020] [Revised: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 11/27/2022]
Abstract
Although many studies have shown that the prelimbic (PL) cortex of the mPFC is involved in the formation of conditioned freezing behavior, few have considered the acoustic response characteristics of PL cortex. Importantly, the change in auditory response characteristics of the PL cortex after conditional fear learning is largely unknown. Here we used in vivo cell-attached recordings targeting the mPFC during the waking state. We confirmed that the mPFC of adult C57 mice have neurons that respond to noise and tone in the waking state, especially in the PL cortex. Interestingly, the data also confirmed that these neurons responded well to the intensity of sound but did not have frequency topological distribution characteristics. Furthermore, we found that the number of c-fos positive neurons in the PL cortex increased significantly after auditory fear conditioning. The auditory-induced local field potential recordings and in vivo cell-attached recordings demonstrated that the PL cortex was more sensitive to the auditory conditioned stimulus after the acquisition of conditioned fear. The proportion of neurons responding to noise was significantly increased, and the signal to noise ratio of the spikes were also increased. These data reveal that PL neurons themselves responded to the main information (sound intensity), while the secondary information (frequency) response was almost negligible after auditory fear conditioning. This phenomenon may be the functional basis for handling this type of emotional memory, and this response characteristic is thought to be emotional sensitization but does not change the nature of this response.
Collapse
Affiliation(s)
- Haiting Xie
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China; Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Duobin Wu
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiaoya Gao
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Ningqian Wang
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| | - Zhongju Xiao
- Department of Physiology, School of Basic Medical Sciences, Key Laboratory of Psychiatric Disorders of Guangdong Province, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Key Laboratory of Mental Health of the Ministry of Education, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
46
|
Dunlap AG, Besosa C, Pascual LM, Chong KK, Walum H, Kacsoh DB, Tankeu BB, Lu K, Liu RC. Becoming a better parent: Mice learn sounds that improve a stereotyped maternal behavior. Horm Behav 2020; 124:104779. [PMID: 32502487 PMCID: PMC7487030 DOI: 10.1016/j.yhbeh.2020.104779] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/22/2020] [Accepted: 05/18/2020] [Indexed: 11/30/2022]
Abstract
While mothering is often instinctive and stereotyped in species-specific ways, evolution can favor genetically "open" behavior programs that allow experience to shape infant care. Among experience-dependent maternal behavioral mechanisms, sensory learning about infants has been hard to separate from motivational changes arising from sensitization with infants. We developed a paradigm in which sensory learning of an infant-associated cue improves a stereotypical maternal behavior in female mice. Mice instinctively employed a spatial memory-based strategy when engaged repetitively in a pup search and retrieval task. However, by playing a sound from a T-maze arm to signal where a pup will be delivered for retrieval, mice learned within 7 days and retained for at least 2 weeks the ability to use this specific cue to guide a more efficient search strategy. The motivation to retrieve pups also increased with learning on average, but their correlation did not explain performance at the trial level. Bilaterally silencing auditory cortical activity significantly impaired the utilization of new strategy without changing the motivation to retrieve pups. Finally, motherhood as compared to infant-care experience alone accelerated how quickly the new sensory-based strategy was acquired, suggesting a role for the maternal hormonal state. By rigorously establishing that newly formed sensory associations can improve the performance of a natural maternal behavior, this work facilitates future studies into the neurochemical and circuit mechanisms that mediate novel sensory learning in the maternal context, as well as more learning-based mechanisms of parental behavior in rodents.
Collapse
Affiliation(s)
- Alexander G Dunlap
- Bioengineering Interdisciplinary Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA; Department of Biology, Emory University, Atlanta, GA, USA
| | | | - Leila M Pascual
- Neuroscience Graduate Program, Emory University, Atlanta, GA, USA
| | - Kelly K Chong
- Department of Biology, Emory University, Atlanta, GA, USA; Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hasse Walum
- Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | | | - Brenda B Tankeu
- Department of Natural Sciences, Bowie State University, Bowie, MD, USA; Emory College Summer Undergraduate Research Experience Program, Atlanta, GA, USA
| | - Kai Lu
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Robert C Liu
- Department of Biology, Emory University, Atlanta, GA, USA; Silvio O. Conte Center for Oxytocin and Social Cognition and Center for Translational Social Neuroscience, Atlanta, GA, USA.
| |
Collapse
|
47
|
Nadhimi Y, Llano DA. Does hearing loss lead to dementia? A review of the literature. Hear Res 2020; 402:108038. [PMID: 32814645 DOI: 10.1016/j.heares.2020.108038] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/04/2020] [Accepted: 07/02/2020] [Indexed: 12/31/2022]
Abstract
Recent studies have revealed a correlation between aging-related hearing loss and the likelihood of developing Alzheimer Disease. However, it is not yet known if the correlation simply reflects the fact that these two disorders share common risk factors or whether there is a causal link between them. The answer to this question carries therapeutic implications. Unfortunately, it is not possible to study the question of causality between aging-related hearing loss and dementia in human subjects. Here, we evaluate the research surrounding induced-hearing loss in animal models on non-auditory cognition to help infer if there is any causal evidence linking hearing loss and a more general dementia. We find ample evidence that induction of hearing loss in animals produces cognitive decline, particularly hippocampal dysfunction. The data suggest that noise-exposure produces a toxic milieu in the hippocampus consisting of a spike in glucocorticoid levels, elevations of mediators of oxidative stress and excitotoxicity, which as a consequence induce cessation of neurogenesis, synaptic loss and tau hyperphosphorylation. These data suggest that hearing loss can lead to pathological hallmarks similar to those seen in Alzheimer's Disease and other dementias. However, the rodent data do not establish that hearing loss on its own can induce a progressive degenerative dementing illness. Therefore, we conclude that an additional "hit", such as aging, APOE genotype, microvascular disease or others, may be necessary to trigger an ongoing degenerative process such as Alzheimer Disease.
Collapse
Affiliation(s)
- Yosra Nadhimi
- Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, USA
| | - Daniel A Llano
- Molecular and Cellular Biology, University of Illinois at Urbana-Champaign, USA; Neuroscience Program, University of Illinois at Urbana-Champaign, USA; Carle Neuroscience Institute, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, Urbana, IL, USA.
| |
Collapse
|
48
|
Tinnitus Does Not Interfere with Auditory and Speech Perception. J Neurosci 2020; 40:6007-6017. [PMID: 32554549 DOI: 10.1523/jneurosci.0396-20.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/31/2020] [Accepted: 06/12/2020] [Indexed: 12/13/2022] Open
Abstract
Tinnitus is a sound heard by 15% of the general population in the absence of any external sound. Because external sounds can sometimes mask tinnitus, tinnitus is assumed to affect the perception of external sounds, leading to hypotheses such as "tinnitus filling in the temporal gap" in animal models and "tinnitus inducing hearing difficulty" in human subjects. Here we compared performance in temporal, spectral, intensive, masking and speech-in-noise perception tasks between 45 human listeners with chronic tinnitus (18 females and 27 males with a range of ages and degrees of hearing loss) and 27 young, normal-hearing listeners without tinnitus (11 females and 16 males). After controlling for age, hearing loss, and stimulus variables, we discovered that, contradictory to the widely held assumption, tinnitus does not interfere with the perception of external sounds in 32 of the 36 measures. We interpret the present result to reflect a bottom-up pathway for the external sound and a separate top-down pathway for tinnitus. We propose that these two perceptual pathways can be independently modulated by attention, which leads to the asymmetrical interaction between external and internal sounds, and several other puzzling tinnitus phenomena such as discrepancy in loudness between tinnitus rating and matching. The present results suggest not only a need for new theories involving attention and central noise in animal tinnitus models but also a shift in focus from treating tinnitus to managing its comorbid conditions when addressing complaints about hearing difficulty in individuals with tinnitus.SIGNIFICANCE STATEMENT Tinnitus, or ringing in the ears, is a neurologic disorder that affects 15% of the general population. Here we discovered an asymmetrical relationship between tinnitus and external sounds: although external sounds have been widely used to cover up tinnitus, tinnitus does not impair, and sometimes even improves, the perception of external sounds. This counterintuitive discovery contradicts the general belief held by scientists, clinicians, and even individuals with tinnitus themselves, who often report hearing difficulty, especially in noise. We attribute the counterintuitive discovery to two independent pathways: the bottom-up perception of external sounds and the top-down perception of tinnitus. Clinically, the present work suggests a shift in focus from treating tinnitus itself to treating its comorbid conditions and secondary effects.
Collapse
|
49
|
Behavioral and neuroanatomical effects on exposure to White noise in rats. Neurosci Lett 2020; 728:134898. [DOI: 10.1016/j.neulet.2020.134898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 12/13/2022]
|
50
|
Fang Q, Chou XL, Peng B, Zhong W, Zhang LI, Tao HW. A Differential Circuit via Retino-Colliculo-Pulvinar Pathway Enhances Feature Selectivity in Visual Cortex through Surround Suppression. Neuron 2019; 105:355-369.e6. [PMID: 31812514 DOI: 10.1016/j.neuron.2019.10.027] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/15/2019] [Accepted: 10/17/2019] [Indexed: 01/06/2023]
Abstract
In the mammalian visual system, information from the retina streams into parallel bottom-up pathways. It remains unclear how these pathways interact to contribute to contextual modulation of visual cortical processing. By optogenetic inactivation and activation of mouse lateral posterior nucleus (LP) of thalamus, a homolog of pulvinar, or its projection to primary visual cortex (V1), we found that LP contributes to surround suppression of layer (L) 2/3 responses in V1 by driving L1 inhibitory neurons. This results in subtractive suppression of visual responses and an overall enhancement of orientation, direction, spatial, and size selectivity. Neurons in V1-projecting LP regions receive bottom-up input from the superior colliculus (SC) and respond preferably to non-patterned visual noise. The noise-dependent LP activity allows V1 to "cancel" noise effects and maintain its orientation selectivity under varying noise background. Thus, the retina-SC-LP-V1 pathway forms a differential circuit with the canonical retino-geniculate pathway to achieve context-dependent sharpening of visual representations.
Collapse
Affiliation(s)
- Qi Fang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiao-Lin Chou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Bo Peng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Graduate Program in Neuroscience, University of Southern California, Los Angeles, CA 90089, USA
| | - Wen Zhong
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Li I Zhang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| | - Huizhong Whit Tao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA; Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|