1
|
Mallick A, Tan HL, Epstein JM, Jing Ng CM, Cook OM, Gaudry Q, Dacks AM. Serotonin acts through multiple cellular targets during an olfactory critical period. iScience 2024; 27:111083. [PMID: 39524339 PMCID: PMC11550141 DOI: 10.1016/j.isci.2024.111083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/11/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Serotonin (5-HT) modulates early development during critical periods when experience drives heightened levels of plasticity in neurons. Here, we investigate the cellular mechanisms by which 5-HT modulates critical period plasticity (CPP) in the olfactory system of Drosophila. We first demonstrate that 5-HT is necessary for experience-dependent structural plasticity in response to chronic CO2 exposure and can re-open the critical period long after it normally closes. Knocking down 5-HT7 receptors in a subset of GABAergic local interneurons was sufficient to block CPP, as was knocking down GABA receptors expressed by CO2-sensing olfactory sensory neurons (OSNs). Furthermore, direct modulation of OSNs via 5-HT2B receptors in CO2-sensing OSNs and autoreceptor expression by serotonergic neurons was also required for CPP. Thus, 5-HT targets individual neuron types in the olfactory system via distinct receptors to enable sensory driven plasticity.
Collapse
Affiliation(s)
- Ahana Mallick
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Hua Leonhard Tan
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | | | | | - Oliver Mason Cook
- Departments of Biology and Neuroscience, West Virginia University, Morgantown, WV 26505, USA
| | - Quentin Gaudry
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Andrew M. Dacks
- Departments of Biology and Neuroscience, West Virginia University, Morgantown, WV 26505, USA
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
2
|
Baram TZ, Birnie MT. Enduring memory consequences of early-life stress / adversity: Structural, synaptic, molecular and epigenetic mechanisms. Neurobiol Stress 2024; 33:100669. [PMID: 39309367 PMCID: PMC11415888 DOI: 10.1016/j.ynstr.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/13/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Adverse early life experiences are strongly associated with reduced cognitive function throughout life. The link is strong in many human studies, but these do not enable assigning causality, and the limited access to the live human brain can impede establishing the mechanisms by which early-life adversity (ELA) may induce cognitive problems. In experimental models, artificially imposed chronic ELA/stress results in deficits in hippocampus dependent memory as well as increased vulnerability to the deleterious effects of adult stress on memory. This causal relation of ELA and life-long memory impairments provides a framework to probe the mechanisms by which ELA may lead to human cognitive problems. Here we focus on the consequences of a one-week exposure to adversity during early postnatal life in the rodent, the spectrum of the ensuing memory deficits, and the mechanisms responsible. We highlight molecular, cellular and circuit mechanisms using convergent trans-disciplinary approaches aiming to enable translation of the discoveries in experimental models to the clinic.
Collapse
Affiliation(s)
- Tallie Z. Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
- Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA
- Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Matthew T. Birnie
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
3
|
Gungor Aydin A, Lemenze A, Bieszczad KM. Functional diversities within neurons and astrocytes in the adult rat auditory cortex revealed by single-nucleus RNA sequencing. Sci Rep 2024; 14:25314. [PMID: 39455606 PMCID: PMC11511993 DOI: 10.1038/s41598-024-74732-7] [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: 04/24/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
The mammalian cerebral cortex is composed of a rich diversity of cell types. Sensory cortical cells are organized into networks that rely on their functional diversity to ultimately carry out a variety of sophisticated cognitive functions for perception, learning, and memory. The auditory cortex (AC) has been most extensively studied for its experience-dependent effects, including for perceptual learning and associative memory. Here, we used single-nucleus RNA sequencing (snRNA-seq) in the AC of the adult rat to investigate the breadth of transcriptionally diverse cell types that likely support the role of AC in experience-dependent functions. A variety of unique excitatory and inhibitory neuron subtypes were identified that harbor unique transcriptional profiles of genes with putative relevance for the adaptive neuroplasticity of cortical microcircuits. In addition, we report for the first time a diversity of astrocytes in AC that may represent functionally unique subtypes, including those that could integrate experience-dependent adult neuroplasticity at cortical synapses. Together, these results pave the way for building models of how cortical neurons work in concert with astrocytes to fulfill dynamic and experience-dependent cognitive functions.
Collapse
Affiliation(s)
- Aysegul Gungor Aydin
- Department of Psychology-Behavioral and Systems Neuroscience, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
| | - Alexander Lemenze
- Department of Pathology, Immunology, and Laboratory Medicine, Rutgers University, Newark, NJ, 07103, USA
| | - Kasia M Bieszczad
- Department of Psychology-Behavioral and Systems Neuroscience, Rutgers University, 152 Frelinghuysen Road, Piscataway, NJ, 08854, USA.
- Rutgers Center for Cognitive Science (RuCCS), Rutgers University, Piscataway, NJ, 08854, USA.
- Department of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
4
|
Glynn LM, Liu SR, Lucas CT, Davis EP. Leveraging the science of early life predictability to inform policies promoting child health. Dev Cogn Neurosci 2024; 69:101437. [PMID: 39260117 PMCID: PMC11415967 DOI: 10.1016/j.dcn.2024.101437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/30/2024] [Accepted: 08/12/2024] [Indexed: 09/13/2024] Open
Abstract
Addressing the tremendous burden of early-life adversity requires constructive dialogues between scientists and policy makers to improve population health. Whereas dialogues focused on several aspects of early-life adversity have been initiated, discussion of an underrecognized form of adversity that has been observed across multiple contexts and cultures is only now emerging. Here we provide evidence for "why unpredictability?", including: 1. Evidence that exposures to unpredictability affect child neurodevelopment, with influences that persist into adulthood. 2. The existence of a translational non-human animal model of exposure to early life unpredictability that can be capitalized upon to causally probe neurobiological mechanisms. 3. Evidence that patterns of signals in the early environment promote brain maturation across species. 4. The uneven distribution of unpredictability across demographic populations that illuminates a possible focal point for enhancing health equity. We then outline the potential of unpredictability in terms of the "what"; that is, how might the concept of unpredictability be leveraged to inform policy? We emphasize the importance of interdisciplinary and community partnerships to the success of this work and describe our community-engaged research project. Finally, we highlight opportunities for the science of unpredictability to inform policies in areas such as screening, immigration, criminal justice, education, childcare, child welfare, employment, healthcare and housing.
Collapse
Affiliation(s)
- Laura M Glynn
- Department of Psychology, Chapman University, United States.
| | - Sabrina R Liu
- Department of Human Development, California State University San Marcos, United States
| | | | - Elysia Poggi Davis
- Department of Pediatrics, University of California Irvine, United States; Department of Psychology, University of Denver, United States
| |
Collapse
|
5
|
Allen SJ, Morishita H. Local and long-range input balance: A framework for investigating frontal cognitive circuit maturation in health and disease. SCIENCE ADVANCES 2024; 10:eadh3920. [PMID: 39292771 PMCID: PMC11409946 DOI: 10.1126/sciadv.adh3920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 08/12/2024] [Indexed: 09/20/2024]
Abstract
Frontal cortical circuits undergo prolonged maturation across childhood and adolescence; however, it remains unknown what specific changes are occurring at the circuit level to establish adult cognitive function. With the recent advent of circuit dissection techniques, it is now feasible to examine circuit-specific changes in connectivity, activity, and function in animal models. Here, we propose that the balance of local and long-range inputs onto frontal cognitive circuits is an understudied metric of circuit maturation. This review highlights research on a frontal-sensory attention circuit that undergoes refinement of local/long-range connectivity, regulated by circuit activity and neuromodulatory signaling, and evaluates how this process may occur generally in the frontal cortex to support adult cognitive behavior. Notably, this balance can be bidirectionally disrupted through various mechanisms relevant to psychiatric disorders. Pharmacological or environmental interventions to normalize or reset the local and long-range balance could hold great therapeutic promise to prevent or rescue cognitive deficits.
Collapse
Affiliation(s)
- Samuel J. Allen
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Hirofumi Morishita
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
6
|
Furlanis E, Dai M, Leyva Garcia B, Vergara J, Pereira A, Pelkey K, Tran T, Gorissen BL, Vlachos A, Hairston A, Huang S, Dwivedi D, Du S, Wills S, McMahon J, Lee AT, Chang EF, Razzaq T, Qazi A, Vargish G, Yuan X, Caccavano A, Hunt S, Chittajallu R, McLean N, Hewit L, Paranzino E, Rice H, Cummins AC, Plotnikova A, Mohanty A, Tangen AC, Shin JH, Azadi R, Eldridge MA, Alvarez VA, Averbeck BB, Alyahyay M, Reyes Vallejo T, Soheib M, Vattino LG, MacGregor CP, Banks E, Olah VJ, Naskar S, Hill S, Liebergall S, Badiani R, Hyde L, Xu Q, Allaway KC, Goldberg EM, Nowakowski TJ, Lee S, Takesian AE, Ibrahim LA, Iqbal A, McBain CJ, Dimidschstein J, Fishell G, Wang Y. An enhancer-AAV toolbox to target and manipulate distinct interneuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.17.603924. [PMID: 39091835 PMCID: PMC11291062 DOI: 10.1101/2024.07.17.603924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
In recent years, we and others have identified a number of enhancers that, when incorporated into rAAV vectors, can restrict the transgene expression to particular neuronal populations. Yet, viral tools to access and manipulate fine neuronal subtypes are still limited. Here, we performed systematic analysis of single cell genomic data to identify enhancer candidates for each of the cortical interneuron subtypes. We established a set of enhancer-AAV tools that are highly specific for distinct cortical interneuron populations and striatal cholinergic neurons. These enhancers, when used in the context of different effectors, can target (fluorescent proteins), observe activity (GCaMP) and manipulate (opto- or chemo-genetics) specific neuronal subtypes. We also validated our enhancer-AAV tools across species. Thus, we provide the field with a powerful set of tools to study neural circuits and functions and to develop precise and targeted therapy.
Collapse
Affiliation(s)
- Elisabetta Furlanis
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- These authors contributed equally
| | - Min Dai
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- These authors contributed equally
| | - Brenda Leyva Garcia
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- These authors contributed equally
| | - Josselyn Vergara
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ana Pereira
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kenneth Pelkey
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Thien Tran
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Bram L. Gorissen
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anna Vlachos
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ariel Hairston
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Shuhan Huang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
| | - Deepanjali Dwivedi
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah Du
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sara Wills
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Justin McMahon
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anthony T. Lee
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, USA
| | - Edward F. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, USA
| | | | | | - Geoffrey Vargish
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Xiaoqing Yuan
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Adam Caccavano
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Steven Hunt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ramesh Chittajallu
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Nadiya McLean
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Lauren Hewit
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Emily Paranzino
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Haley Rice
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Alex C. Cummins
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Anya Plotnikova
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Arya Mohanty
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Anne Claire Tangen
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Jung Hoon Shin
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Reza Azadi
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Mark A.G. Eldridge
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Veronica A. Alvarez
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Bruno B. Averbeck
- National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland 20892-4415, USA
| | - Mansour Alyahyay
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | - Tania Reyes Vallejo
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | - Mohammed Soheib
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | - Lucas G. Vattino
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary Boston, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School Boston, USA
| | - Cathryn P. MacGregor
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary Boston, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School Boston, USA
| | - Emmie Banks
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Viktor Janos Olah
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Shovan Naskar
- Unit of Functional Neural Circuit, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sophie Hill
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Sophie Liebergall
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Rohan Badiani
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Lili Hyde
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Qing Xu
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
- Genomics & Systems Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Kathryn C. Allaway
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ethan M. Goldberg
- Department of Neuroscience, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Tomasz J. Nowakowski
- Department of Neurological Surgery, University of California San Francisco, San Francisco, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, USA
- Department of Anatomy, University of California, San Francisco (UCSF), San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Soohyun Lee
- Unit of Functional Neural Circuit, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E. Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear Infirmary Boston, USA
- Department of Otolaryngology - Head and Neck Surgery, Harvard Medical School Boston, USA
| | - Leena A. Ibrahim
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, 23955–6900, Kingdom of Saudi Arabia
| | | | - Chris J. McBain
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Jordane Dimidschstein
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yating Wang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
7
|
Iannone AF, Akgül G, Zhang R, Wacks S, Hussein N, Macias CG, Donatelle A, Bauriedel JMJ, Wright C, Abramov D, Johnson MA, Govek EE, Burré J, Milner TA, De Marco García NV. The chemokine Cxcl14 regulates interneuron differentiation in layer I of the somatosensory cortex. Cell Rep 2024; 43:114531. [PMID: 39058591 PMCID: PMC11373301 DOI: 10.1016/j.celrep.2024.114531] [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: 02/01/2024] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Spontaneous and sensory-evoked activity sculpts developing circuits. Yet, how these activity patterns intersect with cellular programs regulating the differentiation of neuronal subtypes is not well understood. Through electrophysiological and in vivo longitudinal analyses, we show that C-X-C motif chemokine ligand 14 (Cxcl14), a gene previously characterized for its association with tumor invasion, is expressed by single-bouquet cells (SBCs) in layer I (LI) of the somatosensory cortex during development. Sensory deprivation at neonatal stages markedly decreases Cxcl14 expression. Additionally, we report that loss of function of this gene leads to increased intrinsic excitability of SBCs-but not LI neurogliaform cells-and augments neuronal complexity. Furthermore, Cxcl14 loss impairs sensory map formation and compromises the in vivo recruitment of superficial interneurons by sensory inputs. These results indicate that Cxcl14 is required for LI differentiation and demonstrate the emergent role of chemokines as key players in cortical network development.
Collapse
Affiliation(s)
- Andrew F Iannone
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA
| | - Gülcan Akgül
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Robin Zhang
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sam Wacks
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Nisma Hussein
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Carmen Ginelly Macias
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Alexander Donatelle
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Julia M J Bauriedel
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Cora Wright
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Debra Abramov
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY 10021, USA; Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Megan A Johnson
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Eve-Ellen Govek
- Laboratory of Developmental Neurobiology, The Rockefeller University, New York, NY 10065, USA
| | - Jacqueline Burré
- Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA
| | - Natalia V De Marco García
- Center for Neurogenetics, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10021, USA.
| |
Collapse
|
8
|
Vattino LG, MacGregor CP, Liu CJ, Sweeney CG, Takesian AE. Primary auditory thalamus relays directly to cortical layer 1 interneurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603741. [PMID: 39071266 PMCID: PMC11275971 DOI: 10.1101/2024.07.16.603741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Inhibitory interneurons within cortical layer 1 (L1-INs) integrate inputs from diverse brain regions to modulate sensory processing and plasticity, but the sensory inputs that recruit these interneurons have not been identified. Here we used monosynaptic retrograde tracing and whole-cell electrophysiology to characterize the thalamic inputs onto two major subpopulations of L1-INs in the mouse auditory cortex. We find that the vast majority of auditory thalamic inputs to these L1-INs unexpectedly arise from the ventral subdivision of the medial geniculate body (MGBv), the tonotopically-organized primary auditory thalamus. Moreover, these interneurons receive robust functional monosynaptic MGBv inputs that are comparable to those recorded in the L4 excitatory pyramidal neurons. Our findings identify a direct pathway from the primary auditory thalamus to the L1-INs, suggesting that these interneurons are uniquely positioned to integrate thalamic inputs conveying precise sensory information with top-down inputs carrying information about brain states and learned associations.
Collapse
Affiliation(s)
- Lucas G. Vattino
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Cathryn P. MacGregor
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- These authors contributed equally to this work
| | - Christine Junhui Liu
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
- Graduate Program in Speech and Hearing and Bioscience and Technologies, Harvard Medical School, Boston, MA, USA
- These authors contributed equally to this work
| | - Carolyn G. Sweeney
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| | - Anne E. Takesian
- Eaton-Peabody Laboratories, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Otolaryngology – Head and Neck Surgery, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Awad PN, Zerbi V, Johnson-Venkatesh EM, Damiani F, Pagani M, Markicevic M, Nickles S, Gozzi A, Umemori H, Fagiolini M. CDKL5 sculpts functional callosal connectivity to promote cognitive flexibility. Mol Psychiatry 2024; 29:1698-1709. [PMID: 36737483 PMCID: PMC11371650 DOI: 10.1038/s41380-023-01962-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 02/05/2023]
Abstract
Functional and structural connectivity alterations in short- and long-range projections have been reported across neurodevelopmental disorders (NDD). Interhemispheric callosal projection neurons (CPN) represent one of the major long-range projections in the brain, which are particularly important for higher-order cognitive function and flexibility. However, whether a causal relationship exists between interhemispheric connectivity alterations and cognitive deficits in NDD remains elusive. Here, we focused on CDKL5 Deficiency Disorder (CDD), a severe neurodevelopmental disorder caused by mutations in the X-linked Cyclin-dependent kinase-like 5 (CDKL5) gene. We found an increase in homotopic interhemispheric connectivity and functional hyperconnectivity across higher cognitive areas in adult male and female CDKL5-deficient mice by resting-state functional MRI (rs-fMRI) analysis. This was accompanied by an increase in the number of callosal synaptic inputs but decrease in local synaptic connectivity in the cingulate cortex of juvenile CDKL5-deficient mice, suggesting an impairment in excitatory synapse development and a differential role of CDKL5 across excitatory neuron subtypes. These deficits were associated with significant cognitive impairments in CDKL5 KO mice. Selective deletion of CDKL5 in the largest subtype of CPN likewise resulted in an increase of functional callosal inputs, without however significantly altering intracortical cingulate networks. Notably, such callosal-specific changes were sufficient to cause cognitive deficits. Finally, when CDKL5 was selectively re-expressed only in this CPN subtype, in otherwise CDKL5-deficient mice, it was sufficient to prevent the cognitive impairments of CDKL5 mutants. Together, these results reveal a novel role of CDKL5 by demonstrating that it is both necessary and sufficient for proper CPN connectivity and cognitive function and flexibility, and further validates a causal relationship between CPN dysfunction and cognitive impairment in a model of NDD.
Collapse
Affiliation(s)
- Patricia Nora Awad
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Valerio Zerbi
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Neuro-X Institute, School of Engineering (STI), École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- CIBM Center for Biomedical Imaging, Lausanne, Switzerland
| | - Erin M Johnson-Venkatesh
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francesca Damiani
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
- Autism Center, Child Mind Institute, New York, NY, USA
| | - Marija Markicevic
- Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sarah Nickles
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Rovereto, Italy
| | - Hisashi Umemori
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michela Fagiolini
- F. M. Kirby Neurobiology Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Hock E. Tan and K. Lisa Yang Center for Autism Research at Harvard University, Boston, MA, USA.
- International Research Center for Neurointelligence (IRCN), University of Tokyo Institutes for Advanced Study, Tokyo, Japan.
| |
Collapse
|
10
|
Aydin AG, Lemenze A, Bieszczad KM. Functional diversities within neurons and astrocytes in the adult rat auditory cortex revealed by single-nucleus RNA sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589831. [PMID: 38659766 PMCID: PMC11042262 DOI: 10.1101/2024.04.16.589831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The mammalian cerebral cortex is composed of a rich diversity of cell types. Cortical cells are organized into networks that rely on their functional diversity to ultimately carry out a variety of sophisticated cognitive functions. To investigate the breadth of transcriptional diverse cell types in the sensory cortex, we have used single-nucleus RNA sequencing (snRNA-seq) in the auditory cortex of the adult rat. A variety of unique excitatory and inhibitory neuron types were identified. In addition, we report for the first time a diversity of astrocytes in the auditory cortex that may represent functionally unique subtypes. Together, these results pave the way for building models of how neurons in the sensory cortex work in concert with astrocytes at synapses to fulfill high-cognitive functions like learning and memory.
Collapse
|
11
|
Burbridge TJ, Ratliff JM, Dwivedi D, Vrudhula U, Alvarado-Huerta F, Sjulson L, Ibrahim LA, Cheadle L, Fishell G, Batista-Brito R. Disruption of Cholinergic Retinal Waves Alters Visual Cortex Development and Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.05.588143. [PMID: 38644996 PMCID: PMC11030223 DOI: 10.1101/2024.04.05.588143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Retinal waves represent an early form of patterned spontaneous neural activity in the visual system. These waves originate in the retina before eye-opening and propagate throughout the visual system, influencing the assembly and maturation of subcortical visual brain regions. However, because it is technically challenging to ablate retina-derived cortical waves without inducing compensatory activity, the role these waves play in the development of the visual cortex remains unclear. To address this question, we used targeted conditional genetics to disrupt cholinergic retinal waves and their propagation to select regions of primary visual cortex, which largely prevented compensatory patterned activity. We find that loss of cholinergic retinal waves without compensation impaired the molecular and synaptic maturation of excitatory neurons located in the input layers of visual cortex, as well as layer 1 interneurons. These perinatal molecular and synaptic deficits also relate to functional changes observed at later ages. We find that the loss of perinatal cholinergic retinal waves causes abnormal visual cortex retinotopy, mirroring changes in the retinotopic organization of gene expression, and additionally impairs the processing of visual information. We further show that retinal waves are necessary for higher order processing of sensory information by impacting the state-dependent activity of layer 1 interneurons, a neuronal type that shapes neocortical state-modulation, as well as for state-dependent gain modulation of visual responses of excitatory neurons. Together, these results demonstrate that a brief targeted perinatal disruption of patterned spontaneous activity alters early cortical gene expression as well as synaptic and physiological development, and compromises both fundamental and, notably, higher-order functions of visual cortex after eye-opening.
Collapse
Affiliation(s)
- Timothy J Burbridge
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Jacob M Ratliff
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Deepanjali Dwivedi
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | | | - Lucas Sjulson
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| | - Leena Ali Ibrahim
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955–6900, KSA
| | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Howard Hughes Medical Institute, Cold Spring Harbor, NY 11724
| | - Gordon Fishell
- Department of Neurobiology, Harvard Medical School, 220 Longwood Ave., Boston, MA 02115
| | - Renata Batista-Brito
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Psychiatry and Behavioral Sciences, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
- Department of Genetics, Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461
| |
Collapse
|
12
|
Mallick A, Tan HL, Epstein JM, Gaudry Q, Dacks AM. Serotonin acts through multiple cellular targets during an olfactory critical period. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.14.589413. [PMID: 38645269 PMCID: PMC11030346 DOI: 10.1101/2024.04.14.589413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Serotonin (5-HT) is known to modulate early development during critical periods when experience drives heightened levels of plasticity in neurons. Here, we take advantage of the genetically tractable olfactory system of Drosophila to investigate how 5-HT modulates critical period plasticity in the CO2 sensing circuit of fruit flies. Our study reveals that 5HT modulation of multiple neuronal targets is necessary for experience-dependent structural changes in an odor processing circuit. The olfactory CPP is known to involve local inhibitory networks and consistent with this we found that knocking down 5-HT7 receptors in a subset of GABAergic local interneurons was sufficient to block CPP, as was knocking down GABA receptors expressed by olfactory sensory neurons (OSNs). Additionally, direct modulation of OSNs via 5-HT2B expression in the cognate OSNs sensing CO2 is also essential for CPP. Furthermore, 5-HT1B expression by serotonergic neurons in the olfactory system is also required during the critical period. Our study reveals that 5-HT modulation of multiple neuronal targets is necessary for experience-dependent structural changes in an odor processing circuit.
Collapse
Affiliation(s)
- Ahana Mallick
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Hua Leonhard Tan
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | | | - Quentin Gaudry
- Department of Biology, University of Maryland, College Park, MD 20742, USA
- Senior Author: These authors contributed equally
| | - Andrew M Dacks
- Departments of Biology and Neuroscience, West Virginia University, Morgantown, WV 26505, USA
- Senior Author: These authors contributed equally
| |
Collapse
|
13
|
Davis EP, Glynn LM. Annual Research Review: The power of predictability - patterns of signals in early life shape neurodevelopment and mental health trajectories. J Child Psychol Psychiatry 2024; 65:508-534. [PMID: 38374811 PMCID: PMC11283837 DOI: 10.1111/jcpp.13958] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 02/21/2024]
Abstract
The global burden of early life adversity (ELA) is profound. The World Health Organization has estimated that ELA accounts for almost 30% of all psychiatric cases. Yet, our ability to identify which individuals exposed to ELA will develop mental illness remains poor and there is a critical need to identify underlying pathways and mechanisms. This review proposes unpredictability as an understudied aspect of ELA that is tractable and presents a conceptual model that includes biologically plausible mechanistic pathways by which unpredictability impacts the developing brain. The model is supported by a synthesis of published and new data illustrating the significant impacts of patterns of signals on child development. We begin with an overview of the existing unpredictability literature, which has focused primarily on longer patterns of unpredictability (e.g. years, months, and days). We then describe our work testing the impact of patterns of parental signals on a moment-to-moment timescale, providing evidence that patterns of these signals during sensitive windows of development influence neurocircuit formation across species and thus may be an evolutionarily conserved process that shapes the developing brain. Next, attention is drawn to emerging themes which provide a framework for future directions of research including the evaluation of functions, such as effortful control, that may be particularly vulnerable to unpredictability, sensitive periods, sex differences, cross-cultural investigations, addressing causality, and unpredictability as a pathway by which other forms of ELA impact development. Finally, we provide suggestions for prevention and intervention, including the introduction of a screening instrument for the identification of children exposed to unpredictable experiences.
Collapse
Affiliation(s)
- Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Laura M. Glynn
- Department of Psychology, Chapman University, Orange, CA, United States
| |
Collapse
|
14
|
Bayazitov IT, Teubner BJW, Feng F, Wu Z, Li Y, Blundon JA, Zakharenko SS. Sound-evoked adenosine release in cooperation with neuromodulatory circuits permits auditory cortical plasticity and perceptual learning. Cell Rep 2024; 43:113758. [PMID: 38358887 PMCID: PMC10939737 DOI: 10.1016/j.celrep.2024.113758] [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: 06/29/2023] [Revised: 11/21/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024] Open
Abstract
Meaningful auditory memories are formed in adults when acoustic information is delivered to the auditory cortex during heightened states of attention, vigilance, or alertness, as mediated by neuromodulatory circuits. Here, we identify that, in awake mice, acoustic stimulation triggers auditory thalamocortical projections to release adenosine, which prevents cortical plasticity (i.e., selective expansion of neural representation of behaviorally relevant acoustic stimuli) and perceptual learning (i.e., experience-dependent improvement in frequency discrimination ability). This sound-evoked adenosine release (SEAR) becomes reduced within seconds when acoustic stimuli are tightly paired with the activation of neuromodulatory (cholinergic or dopaminergic) circuits or periods of attentive wakefulness. If thalamic adenosine production is enhanced, then SEAR elevates further, the neuromodulatory circuits are unable to sufficiently reduce SEAR, and associative cortical plasticity and perceptual learning are blocked. This suggests that transient low-adenosine periods triggered by neuromodulatory circuits permit associative cortical plasticity and auditory perceptual learning in adults to occur.
Collapse
Affiliation(s)
- Ildar T Bayazitov
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brett J W Teubner
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Feng Feng
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhaofa Wu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Yulong Li
- School of Life Sciences, Peking University, Beijing 100871, China
| | - Jay A Blundon
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Division of Neural Circuits and Behavior, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
15
|
Aran Ö, Swales DA, Bailey NA, Korja R, Holmberg E, Eskola E, Nolvi S, Perasto L, Nordenswan E, Karlsson H, Karlsson L, Sandman CA, Stern HS, Baram TZ, Glynn LM, Davis EP. Across ages and places: Unpredictability of maternal sensory signals and child internalizing behaviors. J Affect Disord 2024; 347:557-567. [PMID: 38007106 PMCID: PMC10843791 DOI: 10.1016/j.jad.2023.11.068] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 09/21/2023] [Accepted: 11/18/2023] [Indexed: 11/27/2023]
Abstract
BACKGROUND Patterns of sensory inputs early in life play an integral role in shaping the maturation of neural circuits, including those implicated in emotion and cognition. In both experimental animal models and observational human research, unpredictable sensory signals have been linked to aberrant developmental outcomes, including poor memory and effortful control. These findings suggest that sensitivity to unpredictable sensory signals is conserved across species and sculpts the developing brain. The current study provides a novel investigation of unpredictable maternal sensory signals in early life and child internalizing behaviors. We tested these associations in three independent cohorts to probe the generalizability of associations across continents and cultures. METHOD The three prospective longitudinal cohorts were based in Orange, USA (n = 163, 47.2 % female, Mage = 1 year); Turku, Finland (n = 239, 44.8 % female, Mage = 5 years); and Irvine, USA (n = 129, 43.4 % female, Mage = 9.6 years). Unpredictability of maternal sensory signals was quantified during free-play interactions. Child internalizing behaviors were measured via parent report (Orange & Turku) and child self-report (Irvine). RESULTS Early life exposure to unpredictable maternal sensory signals was associated with greater child fearfulness/anxiety in all three cohorts, above and beyond maternal sensitivity and sociodemographic factors. The association between unpredictable maternal sensory signals and child sadness/depression was relatively weaker and did not reach traditional thresholds for statistical significance. LIMITATIONS The correlational design limits our ability to make causal inferences. CONCLUSIONS Findings across the three diverse cohorts suggest that unpredictable maternal signals early in life shape the development of internalizing behaviors, particularly fearfulness and anxiety.
Collapse
Affiliation(s)
- Özlü Aran
- Department of Psychology, University of Denver, Denver, CO, USA.
| | - Danielle A Swales
- Department of Psychiatry, University of North Carolina at Chapel Hill, NC, USA.
| | - Natasha A Bailey
- Department of Psychology, University of Virginia, Charlottesville, VA, USA
| | - Riikka Korja
- University of Turku, Department of Psychology and Speech-Language Pathology, Turku, Finland; University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland; Centre of Excellence in Learning Dynamics and Intervention Research (InterLearn), University of Turku, Turku, Finland
| | - Eeva Holmberg
- University of Turku, Department of Psychology and Speech-Language Pathology, Turku, Finland; University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland
| | - Eeva Eskola
- University of Turku, Department of Psychology and Speech-Language Pathology, Turku, Finland; University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland
| | - Saara Nolvi
- University of Turku, Department of Psychology and Speech-Language Pathology, Turku, Finland; University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland
| | - Laura Perasto
- University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Elisabeth Nordenswan
- University of Turku, Department of Psychology and Speech-Language Pathology, Turku, Finland; University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland
| | - Hasse Karlsson
- University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland; Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland; Department of Psychiatry, Turku University Hospital and University of Turku, Turku, Finland
| | - Linnea Karlsson
- University of Turku, Department of Clinical Medicine, Turku Brain and Mind Center, FinnBrain Birth Cohort Study, Turku, Finland; Centre of Excellence in Learning Dynamics and Intervention Research (InterLearn), University of Turku, Turku, Finland; Department of Clinical Medicine, Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku
| | - Curt A Sandman
- Department of Psychiatry and Human Behavior, University of California-Irvine, Irvine, CA, USA
| | - Hal S Stern
- Department of Statistics, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California-Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California-Irvine, Irvine, CA, USA; Department of Neurology, University of California-Irvine, Irvine, CA, USA
| | - Laura M Glynn
- Department of Psychology, Chapman University, Orange, CA, USA
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, USA; Department of Pediatrics, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
16
|
Ford AN, Czarny JE, Rogalla MM, Quass GL, Apostolides PF. Auditory Corticofugal Neurons Transmit Auditory and Non-auditory Information During Behavior. J Neurosci 2024; 44:e1190232023. [PMID: 38123993 PMCID: PMC10869159 DOI: 10.1523/jneurosci.1190-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 11/08/2023] [Accepted: 11/29/2023] [Indexed: 12/23/2023] Open
Abstract
Layer 5 pyramidal neurons of sensory cortices project "corticofugal" axons to myriad sub-cortical targets, thereby broadcasting high-level signals important for perception and learning. Recent studies suggest dendritic Ca2+ spikes as key biophysical mechanisms supporting corticofugal neuron function: these long-lasting events drive burst firing, thereby initiating uniquely powerful signals to modulate sub-cortical representations and trigger learning-related plasticity. However, the behavioral relevance of corticofugal dendritic spikes is poorly understood. We shed light on this issue using 2-photon Ca2+ imaging of auditory corticofugal dendrites as mice of either sex engage in a GO/NO-GO sound-discrimination task. Unexpectedly, only a minority of dendritic spikes were triggered by behaviorally relevant sounds under our conditions. Task related dendritic activity instead mostly followed sound cue termination and co-occurred with mice's instrumental licking during the answer period of behavioral trials, irrespective of reward consumption. Temporally selective, optogenetic silencing of corticofugal neurons during the trial answer period impaired auditory discrimination learning. Thus, auditory corticofugal systems' contribution to learning and plasticity may be partially nonsensory in nature.
Collapse
Affiliation(s)
- Alexander N Ford
- Department of Otolaryngology/Head and Neck Surgery, Kresge Hearing Research Institute, Ann Arbor, Michigan 48109
| | - Jordyn E Czarny
- Department of Otolaryngology/Head and Neck Surgery, Kresge Hearing Research Institute, Ann Arbor, Michigan 48109
| | - Meike M Rogalla
- Department of Otolaryngology/Head and Neck Surgery, Kresge Hearing Research Institute, Ann Arbor, Michigan 48109
| | - Gunnar L Quass
- Department of Otolaryngology/Head and Neck Surgery, Kresge Hearing Research Institute, Ann Arbor, Michigan 48109
| | - Pierre F Apostolides
- Department of Otolaryngology/Head and Neck Surgery, Kresge Hearing Research Institute, Ann Arbor, Michigan 48109
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
17
|
Huang S, Wu SJ, Sansone G, Ibrahim LA, Fishell G. Layer 1 neocortex: Gating and integrating multidimensional signals. Neuron 2024; 112:184-200. [PMID: 37913772 PMCID: PMC11180419 DOI: 10.1016/j.neuron.2023.09.041] [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: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/28/2023] [Indexed: 11/03/2023]
Abstract
Layer 1 (L1) of the neocortex acts as a nexus for the collection and processing of widespread information. By integrating ascending inputs with extensive top-down activity, this layer likely provides critical information regulating how the perception of sensory inputs is reconciled with expectation. This is accomplished by sorting, directing, and integrating the complex network of excitatory inputs that converge onto L1. These signals are combined with neuromodulatory afferents and gated by the wealth of inhibitory interneurons that either are embedded within L1 or send axons from other cortical layers. Together, these interactions dynamically calibrate information flow throughout the neocortex. This review will primarily focus on L1 within the primary sensory cortex and will use these insights to understand L1 in other cortical areas.
Collapse
Affiliation(s)
- Shuhan Huang
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Program in Neuroscience, Harvard University, Cambridge, MA 02138, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sherry Jingjing Wu
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Giulia Sansone
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Leena Ali Ibrahim
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia.
| | - Gord Fishell
- Harvard Medical School, Blavatnik Institute, Department of Neurobiology, Boston, MA 02115, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
18
|
Graham G, Chimenti MS, Knudtson KL, Grenard DN, Co L, Sumner M, Tchou T, Bieszczad KM. Learning induces unique transcriptional landscapes in the auditory cortex. Hear Res 2023; 438:108878. [PMID: 37659220 PMCID: PMC10529106 DOI: 10.1016/j.heares.2023.108878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 09/04/2023]
Abstract
Learning can induce neurophysiological plasticity in the auditory cortex at multiple timescales. Lasting changes to auditory cortical function that persist over days, weeks, or even a lifetime, require learning to induce de novo gene expression. Indeed, transcription is the molecular determinant for long-term memories to form with a lasting impact on sound-related behavior. However, auditory cortical genes that support auditory learning, memory, and acquired sound-specific behavior are largely unknown. Using an animal model of adult, male Sprague-Dawley rats, this report is the first to identify genome-wide changes in learning-induced gene expression within the auditory cortex that may underlie long-lasting discriminative memory formation of acoustic frequency cues. Auditory cortical samples were collected from animals in the initial learning phase of a two-tone discrimination sound-reward task known to induce sound-specific neurophysiological and behavioral effects. Bioinformatic analyses on gene enrichment profiles from bulk RNA sequencing identified cholinergic synapse (KEGG rno04725), extra-cellular matrix receptor interaction (KEGG rno04512), and neuroactive receptor interaction (KEGG rno04080) among the top biological pathways are likely to be important for auditory discrimination learning. The findings characterize candidate effectors underlying the early stages of changes in cortical and behavioral function to ultimately support the formation of long-term discriminative auditory memory in the adult brain. The molecules and mechanisms identified are potential therapeutic targets to facilitate experiences that induce long-lasting changes to sound-specific auditory function in adulthood and prime for future gene-targeted investigations.
Collapse
Affiliation(s)
- G Graham
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ, USA; Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - M S Chimenti
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - K L Knudtson
- Iowa Institute of Human Genetics, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - D N Grenard
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - L Co
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - M Sumner
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - T Tchou
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA
| | - K M Bieszczad
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ, USA; Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ, USA; Rutgers Center for Cognitive Science, Rutgers Univ., Piscataway, NJ, USA; Dept. of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA.
| |
Collapse
|
19
|
Benjamin AS, Kording KP. A role for cortical interneurons as adversarial discriminators. PLoS Comput Biol 2023; 19:e1011484. [PMID: 37768890 PMCID: PMC10538760 DOI: 10.1371/journal.pcbi.1011484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
The brain learns representations of sensory information from experience, but the algorithms by which it does so remain unknown. One popular theory formalizes representations as inferred factors in a generative model of sensory stimuli, meaning that learning must improve this generative model and inference procedure. This framework underlies many classic computational theories of sensory learning, such as Boltzmann machines, the Wake/Sleep algorithm, and a more recent proposal that the brain learns with an adversarial algorithm that compares waking and dreaming activity. However, in order for such theories to provide insights into the cellular mechanisms of sensory learning, they must be first linked to the cell types in the brain that mediate them. In this study, we examine whether a subtype of cortical interneurons might mediate sensory learning by serving as discriminators, a crucial component in an adversarial algorithm for representation learning. We describe how such interneurons would be characterized by a plasticity rule that switches from Hebbian plasticity during waking states to anti-Hebbian plasticity in dreaming states. Evaluating the computational advantages and disadvantages of this algorithm, we find that it excels at learning representations in networks with recurrent connections but scales poorly with network size. This limitation can be partially addressed if the network also oscillates between evoked activity and generative samples on faster timescales. Consequently, we propose that an adversarial algorithm with interneurons as discriminators is a plausible and testable strategy for sensory learning in biological systems.
Collapse
Affiliation(s)
- Ari S. Benjamin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Konrad P. Kording
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
20
|
Kunnath AJ, Gifford RH, Wallace MT. Cholinergic modulation of sensory perception and plasticity. Neurosci Biobehav Rev 2023; 152:105323. [PMID: 37467908 PMCID: PMC10424559 DOI: 10.1016/j.neubiorev.2023.105323] [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: 04/03/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/21/2023]
Abstract
Sensory systems are highly plastic, but the mechanisms of sensory plasticity remain unclear. People with vision or hearing loss demonstrate significant neural network reorganization that promotes adaptive changes in other sensory modalities as well as in their ability to combine information across the different senses (i.e., multisensory integration. Furthermore, sensory network remodeling is necessary for sensory restoration after a period of sensory deprivation. Acetylcholine is a powerful regulator of sensory plasticity, and studies suggest that cholinergic medications may improve visual and auditory abilities by facilitating sensory network plasticity. There are currently no approved therapeutics for sensory loss that target neuroplasticity. This review explores the systems-level effects of cholinergic signaling on human visual and auditory perception, with a focus on functional performance, sensory disorders, and neural activity. Understanding the role of acetylcholine in sensory plasticity will be essential for developing targeted treatments for sensory restoration.
Collapse
Affiliation(s)
- Ansley J Kunnath
- Neuroscience Graduate Program, Vanderbilt University, Nashville, TN, USA; Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - René H Gifford
- Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark T Wallace
- Department of Hearing and Speech Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Psychology, Vanderbilt University, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN, USA; Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
21
|
Atsumi Y, Oisi Y, Odagawa M, Matsubara C, Saito Y, Uwamori H, Kobayashi K, Kato S, Kobayashi K, Murayama M. Anatomical identification of a corticocortical top-down recipient inhibitory circuitry by enhancer-restricted transsynaptic tracing. Front Neural Circuits 2023; 17:1245097. [PMID: 37720921 PMCID: PMC10502327 DOI: 10.3389/fncir.2023.1245097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/07/2023] [Indexed: 09/19/2023] Open
Abstract
Despite the importance of postsynaptic inhibitory circuitry targeted by mid/long-range projections (e.g., top-down projections) in cognitive functions, its anatomical properties, such as laminar profile and neuron type, are poorly understood owing to the lack of efficient tracing methods. To this end, we developed a method that combines conventional adeno-associated virus (AAV)-mediated transsynaptic tracing with a distal-less homeobox (Dlx) enhancer-restricted expression system to label postsynaptic inhibitory neurons. We called this method "Dlx enhancer-restricted Interneuron-SpECific transsynaptic Tracing" (DISECT). We applied DISECT to a top-down corticocortical circuit from the secondary motor cortex (M2) to the primary somatosensory cortex (S1) in wild-type mice. First, we injected AAV1-Cre into the M2, which enabled Cre recombinase expression in M2-input recipient S1 neurons. Second, we injected AAV1-hDlx-flex-green fluorescent protein (GFP) into the S1 to transduce GFP into the postsynaptic inhibitory neurons in a Cre-dependent manner. We succeeded in exclusively labeling the recipient inhibitory neurons in the S1. Laminar profile analysis of the neurons labeled via DISECT indicated that the M2-input recipient inhibitory neurons were distributed in the superficial and deep layers of the S1. This laminar distribution was aligned with the laminar density of axons projecting from the M2. We further classified the labeled neuron types using immunohistochemistry and in situ hybridization. This post hoc classification revealed that the dominant top-down M2-input recipient neuron types were somatostatin-expressing neurons in the superficial layers and parvalbumin-expressing neurons in the deep layers. These results demonstrate that DISECT enables the investigation of multiple anatomical properties of the postsynaptic inhibitory circuitry.
Collapse
Affiliation(s)
- Yusuke Atsumi
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
- Department of Life Science and Technology, School of Life Sciences and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Yasuhiro Oisi
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Maya Odagawa
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Chie Matsubara
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Yoshihito Saito
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
- Department of Biology, Graduate School of Science, Kobe University, Kobe-shi, Japan
| | - Hiroyuki Uwamori
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki-shi, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masanori Murayama
- Laboratory for Haptic Perception and Cognitive Physiology, RIKEN Center for Brain Science, Saitama, Japan
| |
Collapse
|
22
|
Graham G, Chimenti MS, Knudtson KL, Grenard DN, Co L, Sumner M, Tchou T, Bieszczad KM. Learning induces unique transcriptional landscapes in the auditory cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.15.536914. [PMID: 37090563 PMCID: PMC10120736 DOI: 10.1101/2023.04.15.536914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Learning can induce neurophysiological plasticity in the auditory cortex at multiple timescales. Lasting changes to auditory cortical function that persist over days, weeks, or even a lifetime, require learning to induce de novo gene expression. Indeed, transcription is the molecular determinant for long-term memories to form with a lasting impact on sound-related behavior. However, auditory cortical genes that support auditory learning, memory, and acquired sound-specific behavior are largely unknown. This report is the first to identify in young adult male rats (Sprague-Dawley) genome-wide changes in learning-induced gene expression within the auditory cortex that may underlie the formation of long-lasting discriminative memory for acoustic frequency cues. Auditory cortical samples were collected from animals in the initial learning phase of a two-tone discrimination sound-reward task known to induce sound-specific neurophysiological and behavioral effects (e.g., Shang et al., 2019). Bioinformatic analyses on gene enrichment profiles from bulk RNA sequencing identified cholinergic synapse (KEGG 04725), extra-cellular matrix receptor interaction (KEGG 04512) , and neuroactive ligand-receptor interaction (KEGG 04080) as top biological pathways for auditory discrimination learning. The findings characterize key candidate effectors underlying changes in cortical function that support the initial formation of long-term discriminative auditory memory in the adult brain. The molecules and mechanisms identified are potential therapeutic targets to facilitate lasting changes to sound-specific auditory function in adulthood and prime for future gene-targeted investigations.
Collapse
Affiliation(s)
- G Graham
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - M S Chimenti
- Iowa Institute of Human Genetics, Univ. of Iowa Carver College of Medicine, Iowa City, IA
| | - K L Knudtson
- Iowa Institute of Human Genetics, Univ. of Iowa Carver College of Medicine, Iowa City, IA
| | - D N Grenard
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - L Co
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - M Sumner
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - T Tchou
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
| | - K M Bieszczad
- Neuroscience Graduate Program, Rutgers Univ., Piscataway, NJ
- Behavioral and Systems Neuroscience, Dept. of Psychology, Rutgers Univ., Piscataway, NJ
- Rutgers Center for Cognitive Science, Rutgers Univ., Piscataway, NJ
- Dept. of Otolaryngology-Head and Neck Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ
| |
Collapse
|
23
|
Sahi RS, Eisenberger NI, Silvers JA. Peer facilitation of emotion regulation in adolescence. Dev Cogn Neurosci 2023; 62:101262. [PMID: 37302349 PMCID: PMC10276262 DOI: 10.1016/j.dcn.2023.101262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 06/02/2023] [Accepted: 06/04/2023] [Indexed: 06/13/2023] Open
Abstract
Emotion regulation is particularly important for adolescents as they undergo normative developmental changes in affective systems and experience heightened risk for psychopathology. Despite a high need for emotion regulation during adolescence, commonly studied emotion regulation strategies like cognitive reappraisal are less beneficial for adolescents than adults because they rely on neural regions that are still developing during this period (i.e., lateral prefrontal cortex). However, adolescence is also marked by increased valuation of peer relationships and sensitivity to social information and cues. In the present review, we synthesize research examining emotion regulation and peer influence across development to suggest that sensitivity to peers during adolescence could be leveraged to improve emotion regulation for this population. We first discuss developmental trends related to emotion regulation at the level of behavior and brain in adolescents, using cognitive reappraisal as an exemplar emotion regulation strategy. Next, we discuss social influences on adolescent brain development, describing caregiver influence and increasing susceptibility to peer influence, to describe how adolescent sensitivity to social inputs represents both a window of vulnerability and opportunity. Finally, we conclude by describing the promise of social (i.e., peer-based) interventions for enhancing emotion regulation in adolescence.
Collapse
Affiliation(s)
- Razia S Sahi
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| | - Naomi I Eisenberger
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jennifer A Silvers
- Department of Psychology, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Palumbo TB, Miwa JM. Lynx1 and the family of endogenous mammalian neurotoxin-like proteins and their roles in modulating nAChR function. Pharmacol Res 2023; 194:106845. [PMID: 37437646 DOI: 10.1016/j.phrs.2023.106845] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
The promise of nicotinic receptors as a therapeutic target has yet to be fully realized, despite solid data supporting their involvement in neurological and neuropsychiatric diseases. The reasons for this are likely complex and manifold, having to do with the widespread action of the cholinergic system and the biophysical mechanism of action of nicotinic receptors leading to fast desensitization and down-regulation. Conventional drug development strategies tend to focus on receptor subtype-specific action of candidate therapeutics, although the broad agonist, nicotine, is being explored in the clinic. The potential negative effects of nicotine make the search for alternate strategies warranted. Prototoxins are a promising yet little-explored avenue of nicotinic receptor drug development. Nicotinic receptors in the brain belong to a complex of proteins, including those that bind to the extracellular face of the receptor, as well as chaperones that bind the intracellular domain, etc. Lynx prototoxins have allosteric modularity effects on receptor function and number and have been implicated in complex in vivo processes such as neuroplasticity, learning, and memory. Their mechanism of action and binding specificity on sets of nAChR subtypes present intriguing possibilities for more efficacious and nuanced therapeutic targeting than nicotinic receptor subtypes alone. An allosteric drug may restrict its actions to physiologically relevant time points, which tend to be correlated with salient events which would be encoded into long-term memory storage. Rather than blanketing the brain with a steady and prolonged elevation of agonist, an allosteric nAChR compound could avoid side effects and loss of efficacy over time. This review details the potential strengths and challenges of prototoxin proteins as therapeutic targets, and some of the utility of such therapeutics based on the emerging understanding of cholinergic signaling in a growing number of complex neural processes.
Collapse
Affiliation(s)
- Talulla B Palumbo
- Department of Biological Sciences, Lehigh University, 111 Research Dr., Iacocca Hall, B-217, Bethlehem PA, USA.
| | - Julie M Miwa
- Department of Biological Sciences, Lehigh University, 111 Research Dr., Iacocca Hall, B-217, Bethlehem PA, USA.
| |
Collapse
|
25
|
Kumar M, Handy G, Kouvaros S, Zhao Y, Brinson LL, Wei E, Bizup B, Doiron B, Tzounopoulos T. Cell-type-specific plasticity of inhibitory interneurons in the rehabilitation of auditory cortex after peripheral damage. Nat Commun 2023; 14:4170. [PMID: 37443148 PMCID: PMC10345144 DOI: 10.1038/s41467-023-39732-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Peripheral sensory organ damage leads to compensatory cortical plasticity that is associated with a remarkable recovery of cortical responses to sound. The precise mechanisms that explain how this plasticity is implemented and distributed over a diverse collection of excitatory and inhibitory cortical neurons remain unknown. After noise trauma and persistent peripheral deficits, we found recovered sound-evoked activity in mouse A1 excitatory principal neurons (PNs), parvalbumin- and vasoactive intestinal peptide-expressing neurons (PVs and VIPs), but reduced activity in somatostatin-expressing neurons (SOMs). This cell-type-specific recovery was also associated with cell-type-specific intrinsic plasticity. These findings, along with our computational modelling results, are consistent with the notion that PV plasticity contributes to PN stability, SOM plasticity allows for increased PN and PV activity, and VIP plasticity enables PN and PV recovery by inhibiting SOMs.
Collapse
Affiliation(s)
- Manoj Kumar
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| | - Gregory Handy
- Departments of Neurobiology and Statistics, University of Chicago, Chicago, IL, 60637, USA
| | - Stylianos Kouvaros
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Yanjun Zhao
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Lovisa Ljungqvist Brinson
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Eric Wei
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Brandon Bizup
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Brent Doiron
- Departments of Neurobiology and Statistics, University of Chicago, Chicago, IL, 60637, USA
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center, Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
26
|
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: 4.0] [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
|
27
|
Batista-Brito R, Majumdar A, Nuño A, Ward C, Barnes C, Nikouei K, Vinck M, Cardin JA. Developmental loss of ErbB4 in PV interneurons disrupts state-dependent cortical circuit dynamics. Mol Psychiatry 2023; 28:3133-3143. [PMID: 37069344 PMCID: PMC10618960 DOI: 10.1038/s41380-023-02066-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/28/2023] [Accepted: 04/03/2023] [Indexed: 04/19/2023]
Abstract
GABAergic inhibition plays an important role in the establishment and maintenance of cortical circuits during development. Neuregulin 1 (Nrg1) and its interneuron-specific receptor ErbB4 are key elements of a signaling pathway critical for the maturation and proper synaptic connectivity of interneurons. Using conditional deletions of the ERBB4 gene in mice, we tested the role of this signaling pathway at two developmental timepoints in parvalbumin-expressing (PV) interneurons, the largest subpopulation of cortical GABAergic cells. Loss of ErbB4 in PV interneurons during embryonic, but not late postnatal development leads to alterations in the activity of excitatory and inhibitory cortical neurons, along with severe disruption of cortical temporal organization. These impairments emerge by the end of the second postnatal week, prior to the complete maturation of the PV interneurons themselves. Early loss of ErbB4 in PV interneurons also results in profound dysregulation of excitatory pyramidal neuron dendritic architecture and a redistribution of spine density at the apical dendritic tuft. In association with these deficits, excitatory cortical neurons exhibit normal tuning for sensory inputs, but a loss of state-dependent modulation of the gain of sensory responses. Together these data support a key role for early developmental Nrg1/ErbB4 signaling in PV interneurons as a powerful mechanism underlying the maturation of both the inhibitory and excitatory components of cortical circuits.
Collapse
Affiliation(s)
- Renata Batista-Brito
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
- Department of Psychiatry and Behavioral Sciences, Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
- Department of Genetics, Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA.
| | - Antara Majumdar
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Department of Physiology, Anatomy and Genetics, University of Oxford, Sherrington Building, Sherrington Road, Oxford, OX1 3PT, England
| | - Alejandro Nuño
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
| | - Claire Ward
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, The Bronx, NY, 10461, USA
| | - Clayton Barnes
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
| | - Kasra Nikouei
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Martin Vinck
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA
- Ernst Strüngmann Institute (ESI) for Neuroscience in Cooperation with Max Planck Society, Deutschordenstraße 46, 60528, Frankfurt, Germany
| | - Jessica A Cardin
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06520, USA.
- Kavli Institute of Neuroscience, Yale University, 333 Cedar St., New Haven, CT, 06520, USA.
- Wu Tsai Institute, Yale University, 100 College St., New Haven, CT, 06520, USA.
| |
Collapse
|
28
|
Wu C, Gaier ED, Nihalani BR, Whitecross S, Hensch TK, Hunter DG. Durable recovery from amblyopia with donepezil. Sci Rep 2023; 13:10161. [PMID: 37349338 PMCID: PMC10287641 DOI: 10.1038/s41598-023-34891-5] [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: 05/24/2022] [Accepted: 05/09/2023] [Indexed: 06/24/2023] Open
Abstract
An elevated threshold for neuroplasticity limits visual gains with treatment of residual amblyopia in older children and adults. Acetylcholinesterase inhibitors (AChEI) can enable visual neuroplasticity and promote recovery from amblyopia in adult mice. Motivated by these promising findings, we sought to determine whether donepezil, a commercially available AChEI, can enable recovery in older children and adults with residual amblyopia. In this open-label pilot efficacy study, 16 participants (mean age 16 years; range 9-37 years) with residual anisometropic and/or strabismic amblyopia were treated with daily oral donepezil for 12 weeks. Donepezil dosage was started at 2.5 or 5.0 mg based on age and increased by 2.5 mg if the amblyopic eye visual acuity did not improve by 1 line from the visit 4 weeks prior for a maximum dosage of 7.5 or 10 mg. Participants < 18 years of age further patched the dominant eye. The primary outcome was visual acuity in the amblyopic eye at 22 weeks, 10 weeks after treatment was discontinued. Mean amblyopic eye visual acuity improved 1.2 lines (range 0.0-3.0), and 4/16 (25%) improved by ≥ 2 lines after 12 weeks of treatment. Gains were maintained 10 weeks after cessation of donepezil and were similar for children and adults. Adverse events were mild and self-limited. Residual amblyopia improves in older children and adults treated with donepezil, supporting the concept that the critical window of visual cortical plasticity can be pharmacologically manipulated to treat amblyopia. Placebo-controlled studies are needed.
Collapse
Affiliation(s)
- Carolyn Wu
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | - Eric D Gaier
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bharti R Nihalani
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Sarah Whitecross
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
| | - Takao K Hensch
- Center for Brain Science, Department of Molecular Cellular Biology, Harvard University, Cambridge, MA, USA
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo, Japan
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David G Hunter
- Department of Ophthalmology, Boston Children's Hospital, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo, Japan
| |
Collapse
|
29
|
Fossati G, Kiss-Bodolay D, Prados J, Chéreau R, Husi E, Cadilhac C, Gomez L, Silva BA, Dayer A, Holtmaat A. Bimodal modulation of L1 interneuron activity in anterior cingulate cortex during fear conditioning. Front Neural Circuits 2023; 17:1138358. [PMID: 37334059 PMCID: PMC10272719 DOI: 10.3389/fncir.2023.1138358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 05/16/2023] [Indexed: 06/20/2023] Open
Abstract
The anterior cingulate cortex (ACC) plays a crucial role in encoding, consolidating and retrieving memories related to emotionally salient experiences, such as aversive and rewarding events. Various studies have highlighted its importance for fear memory processing, but its circuit mechanisms are still poorly understood. Cortical layer 1 (L1) of the ACC might be a particularly important site of signal integration, since it is a major entry point for long-range inputs, which is tightly controlled by local inhibition. Many L1 interneurons express the ionotropic serotonin receptor 3a (5HT3aR), which has been implicated in post-traumatic stress disorder and in models of anxiety. Hence, unraveling the response dynamics of L1 interneurons and subtypes thereof during fear memory processing may provide important insights into the microcircuit organization regulating this process. Here, using 2-photon laser scanning microscopy of genetically encoded calcium indicators through microprisms in awake mice, we longitudinally monitored over days the activity of L1 interneurons in the ACC in a tone-cued fear conditioning paradigm. We observed that tones elicited responses in a substantial fraction of the imaged neurons, which were significantly modulated in a bidirectional manner after the tone was associated to an aversive stimulus. A subpopulation of these neurons, the neurogliaform cells (NGCs), displayed a net increase in tone-evoked responses following fear conditioning. Together, these results suggest that different subpopulations of L1 interneurons may exert distinct functions in the ACC circuitry regulating fear learning and memory.
Collapse
Affiliation(s)
- Giuliana Fossati
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Daniel Kiss-Bodolay
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Neurosurgery, Geneva University Hospitals, Geneva, Switzerland
- Lemanic Neuroscience Doctoral School, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Ronan Chéreau
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Elodie Husi
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Christelle Cadilhac
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Lucia Gomez
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Bianca A. Silva
- Neuro Center, IRCCS Humanitas Research Hospital, Milan, Italy
- National Research Council of Italy, Institute of Neuroscience, Milan, Italy
| | - Alexandre Dayer
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Anthony Holtmaat
- Department of Basic Neurosciences, and Neurocenter, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
30
|
Ghimire M, Cai R, Ling L, Brownell KA, Hackett TA, Llano DA, Caspary DM. Increased pyramidal and VIP neuronal excitability in rat primary auditory cortex directly correlates with tinnitus behaviour. J Physiol 2023; 601:2493-2511. [PMID: 37119035 PMCID: PMC10330441 DOI: 10.1113/jp284675] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023] Open
Abstract
Tinnitus affects roughly 15%-20% of the population while severely impacting 10% of those afflicted. Tinnitus pathology is multifactorial, generally initiated by damage to the auditory periphery, resulting in a cascade of maladaptive plastic changes at multiple levels of the central auditory neuraxis as well as limbic and non-auditory cortical centres. Using a well-established condition-suppression model of tinnitus, we measured tinnitus-related changes in the microcircuits of excitatory/inhibitory neurons onto layer 5 pyramidal neurons (PNs), as well as changes in the excitability of vasoactive intestinal peptide (VIP) neurons in primary auditory cortex (A1). Patch-clamp recordings from PNs in A1 slices showed tinnitus-related increases in spontaneous excitatory postsynaptic currents (sEPSCs) and decreases in spontaneous inhibitory postsynaptic currents (sIPSCs). Both measures could be correlated to the rat's behavioural evidence of tinnitus. Tinnitus-related changes in PN excitability were independent of changes in A1 excitatory or inhibitory cell numbers. VIP neurons, part of an A1 local circuit that can control the excitation of layer 5 PNs via disinhibitory mechanisms, showed significant tinnitus-related increases in excitability that directly correlated with the rat's behavioural tinnitus score. That PN and VIP changes directly correlated to tinnitus behaviour suggests an important role in A1 tinnitus pathology. Tinnitus-related A1 changes were similar to findings in studies of neuropathic pain in somatosensory cortex suggesting a common pathology of these troublesome perceptual impairments. Improved understanding between excitatory, inhibitory and disinhibitory sensory cortical circuits can serve as a model for testing therapeutic approaches to the treatment of tinnitus and chronic pain. KEY POINTS: We identified tinnitus-related changes in synaptic function of specific neuronal subtypes in a reliable animal model of tinnitus. The findings show direct and indirect tinnitus-related losses of normal inhibitory function at A1 layer 5 pyramidal cells, and increased VIP excitability. The findings are similar to what has been shown for neuropathic pain suggesting that restoring normal inhibitory function at synaptic inputs onto A1 pyramidal neurons (PNs) could conceptually reduce tinnitus discomfort.
Collapse
Affiliation(s)
- Madan Ghimire
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702
| | - Rui Cai
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702
| | - Lynne Ling
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702
| | - Kevin A. Brownell
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702
| | - Troy A. Hackett
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, Tennessee 37232
| | - Daniel A. Llano
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Donald M. Caspary
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, Illinois 62702
| |
Collapse
|
31
|
Zhu F, Elnozahy S, Lawlor J, Kuchibhotla KV. The cholinergic basal forebrain provides a parallel channel for state-dependent sensory signaling to auditory cortex. Nat Neurosci 2023; 26:810-819. [PMID: 36973512 PMCID: PMC10625791 DOI: 10.1038/s41593-023-01289-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023]
Abstract
Cholinergic basal forebrain (CBF) signaling exhibits multiple timescales of activity with classic slow signals related to brain and behavioral states and fast, phasic signals reflecting behavioral events, including movement, reinforcement and sensory-evoked responses. However, it remains unknown whether sensory cholinergic signals target the sensory cortex and how they relate to local functional topography. Here we used simultaneous two-channel, two-photon imaging of CBF axons and auditory cortical neurons to reveal that CBF axons send a robust, nonhabituating and stimulus-specific sensory signal to the auditory cortex. Individual axon segments exhibited heterogeneous but stable tuning to auditory stimuli allowing stimulus identity to be decoded from population activity. However, CBF axons displayed no tonotopy and their frequency tuning was uncoupled from that of nearby cortical neurons. Chemogenetic suppression revealed the auditory thalamus as a major source of auditory information to the CBF. Finally, slow fluctuations in cholinergic activity modulated the fast, sensory-evoked signals in the same axons, suggesting that a multiplexed combination of fast and slow signals is projected from the CBF to the auditory cortex. Taken together, our work demonstrates a noncanonical function of the CBF as a parallel channel for state-dependent sensory signaling to the sensory cortex that provides repeated representations of a broad range of sound stimuli at all points on the tonotopic map.
Collapse
Affiliation(s)
- Fangchen Zhu
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Sarah Elnozahy
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Jennifer Lawlor
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - Kishore V Kuchibhotla
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA.
- Department of Neuroscience, Johns Hopkins Medical Institute, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
32
|
Henton A, Zhao Y, Tzounopoulos T. A Role for KCNQ Channels on Cell Type-Specific Plasticity in Mouse Auditory Cortex after Peripheral Damage. J Neurosci 2023; 43:2277-2290. [PMID: 36813573 PMCID: PMC10072297 DOI: 10.1523/jneurosci.1070-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Damage to sensory organs triggers compensatory plasticity mechanisms in sensory cortices. These plasticity mechanisms result in restored cortical responses, despite reduced peripheral input, and contribute to the remarkable recovery of perceptual detection thresholds to sensory stimuli. Overall, peripheral damage is associated with a reduction of cortical GABAergic inhibition; however, less is known about changes in intrinsic properties and the underlying biophysical mechanisms. To study these mechanisms, we used a model of noise-induced peripheral damage in male and female mice. We uncovered a rapid, cell type-specific reduction in the intrinsic excitability of parvalbumin-expressing neurons (PVs) in layer (L) 2/3 of auditory cortex. No changes in the intrinsic excitability of either L2/3 somatostatin-expressing or L2/3 principal neurons (PNs) were observed. The decrease in L2/3 PV excitability was observed 1, but not 7, d after noise exposure, and was evidenced by a hyperpolarization of the resting membrane potential, depolarization of the action potential threshold, and reduction in firing frequency in response to depolarizing current. To uncover the underlying biophysical mechanisms, we recorded potassium currents. We found an increase in KCNQ potassium channel activity in L2/3 PVs of auditory cortex 1 d after noise exposure, associated with a hyperpolarizing shift in the minimal voltage activation of KCNQ channels. This increase contributes to the decreased intrinsic excitability of PVs. Our results highlight cell-type- and channel-specific mechanisms of plasticity after noise-induced hearing loss and will aid in understanding the pathologic processes involved in hearing loss and hearing loss-related disorders, such as tinnitus and hyperacusis.SIGNIFICANCE STATEMENT Noise-induced damage to the peripheral auditory system triggers central plasticity that compensates for the reduced peripheral input. The mechanisms of this plasticity are not fully understood. In the auditory cortex, this plasticity likely contributes to the recovery of sound-evoked responses and perceptual hearing thresholds. Importantly, other functional aspects of hearing do not recover, and peripheral damage may also lead to maladaptive plasticity-related disorders, such as tinnitus and hyperacusis. Here, after noise-induced peripheral damage, we highlight a rapid, transient, and cell type-specific reduction in the excitability of layer 2/3 parvalbumin-expressing neurons, which is due, at least in part, to increased KCNQ potassium channel activity. These studies may highlight novel strategies for enhancing perceptual recovery after hearing loss and mitigating hyperacusis and tinnitus.
Collapse
Affiliation(s)
- Amanda Henton
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
- Center for Neuroscience, University of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Yanjun Zhao
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| | - Thanos Tzounopoulos
- Pittsburgh Hearing Research Center and Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
33
|
Haimson B, Mizrahi A. Plasticity in auditory cortex during parenthood. Hear Res 2023; 431:108738. [PMID: 36931020 DOI: 10.1016/j.heares.2023.108738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/09/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Most animals display robust parental behaviors that support the survival and well-being of their offspring. The manifestation of parental behaviors is accompanied by physiological and hormonal changes, which affect both the body and the brain for better care giving. Rodents exhibit a behavior called pup retrieval - a stereotyped sequence of perception and action - used to identify and retrieve their newborn pups back to the nest. Pup retrieval consists of a significant auditory component, which depends on plasticity in the auditory cortex (ACx). We review the evidence of neural changes taking place in the ACx of rodents during the transition to parenthood. We discuss how the plastic changes both in and out of the ACx support the encoding of pup vocalizations. Key players in the mechanism of this plasticity are hormones and experience, both of which have a clear dynamic signature during the transition to parenthood. Mothers, co caring females, and fathers have been used as models to understand parental plasticity at disparate levels of organization. Yet, common principles of cortical plasticity and the biological mechanisms underlying its involvement in parental behavior are just beginning to be unpacked.
Collapse
Affiliation(s)
- Baruch Haimson
- The Edmond and Lily Safra Center for Brain Sciences, and 2Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| | - Adi Mizrahi
- The Edmond and Lily Safra Center for Brain Sciences, and 2Department of Neurobiology, The Hebrew University of Jerusalem, Jerusalem 91904, Israel.
| |
Collapse
|
34
|
Tasaka GI, Maggi C, Taha E, Mizrahi A. The local and long-range input landscape of inhibitory neurons in mouse auditory cortex. J Comp Neurol 2023; 531:502-514. [PMID: 36453284 PMCID: PMC10107844 DOI: 10.1002/cne.25437] [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: 04/09/2022] [Revised: 09/27/2022] [Accepted: 11/04/2022] [Indexed: 12/05/2022]
Abstract
Roughly 20% of the neurons in the mouse cortex are inhibitory interneurons (INs). Of these, the three major subtypes are parvalbumin (PV), somatostatin (SST), and vasoactive intestinal polypeptide (VIP) expressing neurons. We used monosynaptic rabies tracing to compare the presynaptic input landscape onto these three IN subtypes in the mouse primary auditory cortex (A1). We compared both local patterns of monosynaptic inputs as well as long-range input patterns. The local monosynaptic input landscape to SST neurons was more widespread as compared to PV and VIP neurons. The brain-wide input landscape was rich and heterogeneous with >40 brain regions connecting to all the three INs subtypes from both hemispheres. The general pattern of the long-range input landscape was similar among the groups of INs. Nevertheless, a few differences could be identified. At low resolution, the proportion of local versus long-range inputs was smaller for PV neurons. At mesoscale resolution, we found fewer inputs from temporal association area to VIP INs, and more inputs to SST neurons from basal forebrain and lateral amygdala. Our work can be used as a resource for a quantitative comparison of the location and level of inputs impinging onto discrete populations of neurons in mouse A1.
Collapse
Affiliation(s)
- Gen-Ichi Tasaka
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Claudia Maggi
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elham Taha
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adi Mizrahi
- The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.,The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
35
|
Huerga-Gómez I, Martini FJ, López-Bendito G. Building thalamic neuronal networks during mouse development. Front Neural Circuits 2023; 17:1098913. [PMID: 36817644 PMCID: PMC9936079 DOI: 10.3389/fncir.2023.1098913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
The thalamic nuclear complex contains excitatory projection neurons and inhibitory local neurons, the two cell types driving the main circuits in sensory nuclei. While excitatory neurons are born from progenitors that reside in the proliferative zone of the developing thalamus, inhibitory local neurons are born outside the thalamus and they migrate there during development. In addition to these cell types, which occupy most of the thalamus, there are two small thalamic regions where inhibitory neurons target extra-thalamic regions rather than neighboring neurons, the intergeniculate leaflet and the parahabenular nucleus. Like excitatory thalamic neurons, these inhibitory neurons are derived from progenitors residing in the developing thalamus. The assembly of these circuits follows fine-tuned genetic programs and it is coordinated by extrinsic factors that help the cells find their location, associate with thalamic partners, and establish connections with their corresponding extra-thalamic inputs and outputs. In this review, we bring together what is currently known about the development of the excitatory and inhibitory components of the thalamocortical sensory system, in particular focusing on the visual pathway and thalamic interneurons in mice.
Collapse
Affiliation(s)
- Irene Huerga-Gómez
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Sant Joan d’Alacant, Spain
| | | | | |
Collapse
|
36
|
Quantitative Fluorescence Analysis Reveals Dendrite-Specific Thalamocortical Plasticity in L5 Pyramidal Neurons during Learning. J Neurosci 2023; 43:584-600. [PMID: 36639912 PMCID: PMC9888508 DOI: 10.1523/jneurosci.1372-22.2022] [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: 07/07/2022] [Revised: 10/28/2022] [Accepted: 11/23/2022] [Indexed: 12/14/2022] Open
Abstract
High-throughput anatomic data can stimulate and constrain new hypotheses about how neural circuits change in response to experience. Here, we use fluorescence-based reagents for presynaptic and postsynaptic labeling to monitor changes in thalamocortical synapses onto different compartments of layer 5 (L5) pyramidal (Pyr) neurons in somatosensory (barrel) cortex from mixed-sex mice during whisker-dependent learning (Audette et al., 2019). Using axonal fills and molecular-genetic tags for synapse identification in fixed tissue from Rbp4-Cre transgenic mice, we found that thalamocortical synapses from the higher-order posterior medial thalamic nucleus showed rapid morphologic changes in both presynaptic and postsynaptic structures at the earliest stages of sensory association training. Detected increases in thalamocortical synaptic size were compartment specific, occurring selectively in the proximal dendrites onto L5 Pyr and not at inputs onto their apical tufts in L1. Both axonal and dendritic changes were transient, normalizing back to baseline as animals became expert in the task. Anatomical measurements were corroborated by electrophysiological recordings at different stages of training. Thus, fluorescence-based analysis of input- and target-specific synapses can reveal compartment-specific changes in synapse properties during learning.SIGNIFICANCE STATEMENT Synaptic changes underlie the cellular basis of learning, experience, and neurologic diseases. Neuroanatomical methods to assess synaptic plasticity can provide critical spatial information necessary for building models of neuronal computations during learning and experience but are technically and fiscally intensive. Here, we describe a confocal fluorescence microscopy-based analytical method to assess input, cell type, and dendritic location-specific synaptic plasticity in a sensory learning assay. Our method not only confirms prior electrophysiological measurements but allows us to predict functional strength of synapses in a pathway-specific manner. Our findings also indicate that changes in primary sensory cortices are transient, occurring during early learning. Fluorescence-based synapse identification can be an efficient and easily adopted approach to study synaptic changes in a variety of experimental paradigms.
Collapse
|
37
|
Quast KB, Reh RK, Caiati MD, Kopell N, McCarthy MM, Hensch TK. Rapid synaptic and gamma rhythm signature of mouse critical period plasticity. Proc Natl Acad Sci U S A 2023; 120:e2123182120. [PMID: 36598942 PMCID: PMC9926253 DOI: 10.1073/pnas.2123182120] [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: 12/24/2021] [Accepted: 11/29/2022] [Indexed: 01/05/2023] Open
Abstract
Early-life experience enduringly sculpts thalamocortical (TC) axons and sensory processing. Here, we identify the very first synaptic targets that initiate critical period plasticity, heralded by altered cortical oscillations. Monocular deprivation (MD) acutely induced a transient (<3 h) peak in EEG γ-power (~40 Hz) specifically within the visual cortex, but only when the critical period was open (juvenile mice or adults after dark-rearing, Lynx1-deletion, or diazepam-rescued GAD65-deficiency). Rapid TC input loss onto parvalbumin-expressing (PV) inhibitory interneurons (but not onto nearby pyramidal cells) was observed within hours of MD in a TC slice preserving the visual pathway - again once critical periods opened. Computational TC modeling of the emergent γ-rhythm in response to MD delineated a cortical interneuronal gamma (ING) rhythm in networks of PV-cells bearing gap junctions at the start of the critical period. The ING rhythm effectively dissociated thalamic input from cortical spiking, leading to rapid loss of previously strong TC-to-PV connections through standard spike-timing-dependent plasticity rules. As a consequence, previously silent TC-to-PV connections could strengthen on a slower timescale, capturing the gradually increasing γ-frequency and eventual fade-out over time. Thus, ING enables cortical dynamics to transition from being dominated by the strongest TC input to one that senses the statistics of population TC input after MD. Taken together, our findings reveal the initial synaptic events underlying critical period plasticity and suggest that the fleeting ING accompanying a brief sensory perturbation may serve as a robust readout of TC network state with which to probe developmental trajectories.
Collapse
Affiliation(s)
- Kathleen B. Quast
- Department of Molecular Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA02138
- FM Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Rebecca K. Reh
- Department of Molecular Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA02138
- FM Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
| | - Maddalena D. Caiati
- Department of Molecular Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA02138
| | - Nancy Kopell
- Department of Mathematics, Boston University, Boston, MA02215
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Bunkyo-ku, Tokyo113, Japan
| | - Michelle M. McCarthy
- Department of Mathematics, Boston University, Boston, MA02215
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Bunkyo-ku, Tokyo113, Japan
| | - Takao K. Hensch
- Department of Molecular Cellular Biology, Center for Brain Science, Harvard University, Cambridge, MA02138
- FM Kirby Neurobiology Center, Boston Children’s Hospital, Harvard Medical School, Boston, MA02115
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Bunkyo-ku, Tokyo113, Japan
| |
Collapse
|
38
|
Holmberg E, Kataja EL, Davis EP, Pajulo M, Nolvi S, Lahtela H, Nordenswan E, Karlsson L, Karlsson H, Korja R. Unpredictable maternal sensory signals in caregiving behavior are associated with child effortful control. PLoS One 2022; 17:e0279384. [PMID: 36538558 PMCID: PMC9767348 DOI: 10.1371/journal.pone.0279384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence suggests that exposure to unpredictable patterns of maternal sensory signals during infancy is associated with child neurodevelopment, including poorer effortful control. However, longitudinal effects on child development and possible sex differences are understudied. The aims of the present study were to explore whether exposure to unpredictable maternal sensory signals during infancy is related to child effortful control at 5 years of age and whether child sex moderates these associations. In addition, we examined how exposure to very high vs. low/moderate unpredictability using categorical cut-offs is related to child effortful control. Participants (133 mother-child pairs, all Caucasian) were drawn from the FinnBrain Birth Cohort Study in Finland. Maternal sensory signals (auditory, visual, tactile) were coded from the 10-min free-play episode on a moment-on-moment basis using Observer XT 11 (Noldus), and the unpredictability of maternal sensory signals was characterized as the entropy rate when the infant was 8 months of age. Child effortful control was assessed via mother reports using the Child Behavior Questionnaire very short form (CBQ-VSF) when the child was 5 years old. Correlational analyses showed that higher unpredictability of maternal sensory signals had a modest association with children's poorer effortful control at 5 years of age. Notably, the linear regression model showed that child sex moderated these associations, as higher exposure to unpredictable maternal sensory signals was related to poorer effortful control among males, but not among females. Moreover, the general linear model showed that exposure to very high unpredictability was associated with poorer child effortful control at 5 years of age and remained significant when adjusted for possible confounding factors. These results are in line with previous findings and suggest that the unpredictability of maternal sensory signals is potentially an important aspect of early caregiving behavior associated with the development of child effortful control.
Collapse
Affiliation(s)
- Eeva Holmberg
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- * E-mail:
| | - Eeva-Leena Kataja
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States of America
- Department of Pediatrics, University of California, Irvine, CA, United States of America
| | - Marjukka Pajulo
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Child Psychiatry, University of Turku, Turku, Finland
| | - Saara Nolvi
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Hetti Lahtela
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Elisabeth Nordenswan
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
| | - Linnea Karlsson
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Hospital District of Southwest Finland, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Department of Pediatrics and Adolescent Medicine, University of Turku and Hospital District of Southwest Finland, Turku, Finland
| | - Hasse Karlsson
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Hospital District of Southwest Finland, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Riikka Korja
- Department of Clinical Medicine, FinnBrain Birth Cohort Study, Turku Brain and Mind Center, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| |
Collapse
|
39
|
Davis EP, McCormack K, Arora H, Sharpe D, Short AK, Bachevalier J, Glynn LM, Sandman CA, Stern HS, Sanchez M, Baram TZ. Early life exposure to unpredictable parental sensory signals shapes cognitive development across three species. Front Behav Neurosci 2022; 16:960262. [PMID: 36338881 PMCID: PMC9630745 DOI: 10.3389/fnbeh.2022.960262] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 09/21/2022] [Indexed: 08/23/2023] Open
Abstract
Exposure to early life adversity has long term consequences on cognitive function. Most research has focused on understanding components of early life adversities that contribute to later risk, including poverty, trauma, maltreatment, and neglect. Whereas these factors, in the aggregate, explain a significant proportion of emotional and cognitive problems, there are serious gaps in our ability to identify potential mechanisms by which early life adversities might promote vulnerability or resilience. Here we discuss early life exposure to unpredictable signals from the caretaker as an understudied type of adversity that is amenable to prevention and intervention. We employ a translational approach to discover underlying neurobiological mechanisms by which early life exposure to unpredictable signals sculpts the developing brain. First, we review evidence that exposure to unpredictable signals from the parent during sensitive periods impacts development of neural circuits. Second, we describe a method for characterizing early life patterns of sensory signals across species. Third, we present published and original data illustrating that patterns of maternal care predict memory function in humans, non-human primates, and rodents. Finally, implications are discussed for identifying individuals at risk so that early preventive-intervention can be provided.
Collapse
Affiliation(s)
- Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Kai McCormack
- Department of Psychology, Spelman College, Atlanta, GA, United States
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Hina Arora
- Department of Statistics, University of California, Irvine, Irvine, CA, United States
| | - Desiree Sharpe
- Mary Frances Early College of Education (MFECOE) Torrance Center for Creativity and Talent Development, University of Georgia, Athens, GA, United States
| | - Annabel K. Short
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Jocelyne Bachevalier
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Laura M. Glynn
- Department of Psychology, Chapman University, Orange, CA, United States
| | - Curt A. Sandman
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Hal S. Stern
- Department of Statistics, University of California, Irvine, Irvine, CA, United States
| | - Mar Sanchez
- Emory National Primate Research Center, Emory University, Atlanta, GA, United States
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, Atlanta, GA, United States
| | - Tallie Z. Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
40
|
Baum GL, Flournoy JC, Glasser MF, Harms MP, Mair P, Sanders AFP, Barch DM, Buckner RL, Bookheimer S, Dapretto M, Smith S, Thomas KM, Yacoub E, Van Essen DC, Somerville LH. Graded Variation in T1w/T2w Ratio during Adolescence: Measurement, Caveats, and Implications for Development of Cortical Myelin. J Neurosci 2022; 42:5681-5694. [PMID: 35705486 PMCID: PMC9302463 DOI: 10.1523/jneurosci.2380-21.2022] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/07/2022] [Accepted: 06/04/2022] [Indexed: 01/22/2023] Open
Abstract
Adolescence is characterized by the maturation of cortical microstructure and connectivity supporting complex cognition and behavior. Axonal myelination influences brain connectivity during development by enhancing neural signaling speed and inhibiting plasticity. However, the maturational timing of cortical myelination during human adolescence remains poorly understood. Here, we take advantage of recent advances in high-resolution cortical T1w/T2w mapping methods, including principled correction of B1+ transmit field effects, using data from the Human Connectome Project in Development (HCP-D; N = 628, ages 8-21). We characterize microstructural changes relevant to myelination by estimating age-related differences in T1w/T2w throughout the cerebral neocortex from childhood to early adulthood. We apply Bayesian spline models and clustering analysis to demonstrate graded variation in age-dependent cortical T1w/T2w differences that are correlated with the sensorimotor-association (S-A) axis of cortical organization reported by others. In sensorimotor areas, T1w/T2w ratio measures start at high levels at early ages, increase at a fast pace, and decelerate at later ages (18-21). In intermediate multimodal areas along the S-A axis, T1w/T2w starts at intermediate levels and increases linearly at an intermediate pace. In transmodal/paralimbic association areas, T1w/T2w starts at low levels and increases linearly at the slowest pace. These data provide evidence for graded variation of the T1w/T2w ratio along the S-A axis that may reflect cortical myelination changes during adolescence underlying the development of complex information processing and psychological functioning. We discuss the implications of these results as well as caveats in interpreting magnetic resonance imaging (MRI)-based estimates of myelination.SIGNIFICANCE STATEMENT Myelin is a lipid membrane that is essential to healthy brain function. Myelin wraps axons to increase neural signaling speed, enabling complex neuronal functioning underlying learning and cognition. Here, we characterize the developmental timing of myelination across the cerebral cortex during adolescence using a noninvasive proxy measure, T1w/T2w mapping. Our results provide new evidence demonstrating graded variation across the cortex in the timing of T1w/T2w changes during adolescence, with rapid T1w/T2w increases in lower-order sensory areas and gradual T1w/T2w increases in higher-order association areas. This spatial pattern of microstructural brain development closely parallels the sensorimotor-to-association axis of cortical organization and plasticity during ontogeny.
Collapse
Affiliation(s)
- Graham L Baum
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA, 02138
| | - John C Flournoy
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA, 02138
| | - Matthew F Glasser
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA, 63110
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Michael P Harms
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Patrick Mair
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA, 02138
| | - Ashley F P Sanders
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Deanna M Barch
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA, 63110
- Department of Psychological and Brain Sciences, Washington University, St. Louis, MO, USA, MO 63130
| | - Randy L Buckner
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA, 02138
| | - Susan Bookheimer
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA, 90095
| | - Mirella Dapretto
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA, 90095
| | - Stephen Smith
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford, United Kingdom, OX3 9DU
| | - Kathleen M Thomas
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA, 55455
| | - Essa Yacoub
- Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA, 55455
| | - David C Van Essen
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA, 63110
| | - Leah H Somerville
- Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA, 02138
| |
Collapse
|
41
|
Abstract
Early-life environmental signals contribute to how the brain handles reward, stress, and fear.
Collapse
Affiliation(s)
- Matthew T Birnie
- Departments of Pediatrics, Anatomy and Neurobiology, and Neurology, University of California-Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Departments of Pediatrics, Anatomy and Neurobiology, and Neurology, University of California-Irvine, Irvine, CA, USA
| |
Collapse
|
42
|
Ferrer C, De Marco García NV. The Role of Inhibitory Interneurons in Circuit Assembly and Refinement Across Sensory Cortices. Front Neural Circuits 2022; 16:866999. [PMID: 35463203 PMCID: PMC9021723 DOI: 10.3389/fncir.2022.866999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/16/2022] [Indexed: 12/15/2022] Open
Abstract
Sensory information is transduced into electrical signals in the periphery by specialized sensory organs, which relay this information to the thalamus and subsequently to cortical primary sensory areas. In the cortex, microcircuits constituted by interconnected pyramidal cells and inhibitory interneurons, distributed throughout the cortical column, form the basic processing units of sensory information underlying sensation. In the mouse, these circuits mature shortly after birth. In the first postnatal week cortical activity is characterized by highly synchronized spontaneous activity. While by the second postnatal week, spontaneous activity desynchronizes and sensory influx increases drastically upon eye opening, as well as with the onset of hearing and active whisking. This influx of sensory stimuli is fundamental for the maturation of functional properties and connectivity in neurons allocated to sensory cortices. In the subsequent developmental period, spanning the first five postnatal weeks, sensory circuits are malleable in response to sensory stimulation in the so-called critical periods. During these critical periods, which vary in timing and duration across sensory areas, perturbations in sensory experience can alter cortical connectivity, leading to long-lasting modifications in sensory processing. The recent advent of intersectional genetics, in vivo calcium imaging and single cell transcriptomics has aided the identification of circuit components in emergent networks. Multiple studies in recent years have sought a better understanding of how genetically-defined neuronal subtypes regulate circuit plasticity and maturation during development. In this review, we discuss the current literature focused on postnatal development and critical periods in the primary auditory (A1), visual (V1), and somatosensory (S1) cortices. We compare the developmental trajectory among the three sensory areas with a particular emphasis on interneuron function and the role of inhibitory circuits in cortical development and function.
Collapse
|
43
|
Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, Beck J, Bai X, Law SM, Savage JC, Lambert JJ, Belelli D, Tremblay MÈ, Cahalan MD, Baram TZ. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep 2022; 38:110600. [PMID: 35354026 PMCID: PMC9014810 DOI: 10.1016/j.celrep.2022.110600] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Several mental illnesses, characterized by aberrant stress reactivity, often arise after early-life adversity (ELA). However, it is unclear how ELA affects stress-related brain circuit maturation, provoking these enduring vulnerabilities. We find that ELA increases functional excitatory synapses onto stress-sensitive hypothalamic corticotropin-releasing hormone (CRH)-expressing neurons, resulting from disrupted developmental synapse pruning by adjacent microglia. Microglial process dynamics and synaptic element engulfment were attenuated in ELA mice, associated with deficient signaling of the microglial phagocytic receptor MerTK. Accordingly, selective chronic chemogenetic activation of ELA microglia increased microglial process dynamics and reduced excitatory synapse density to control levels. Notably, selective early-life activation of ELA microglia normalized adult acute and chronic stress responses, including stress-induced hormone secretion and behavioral threat responses, as well as chronic adrenal hypertrophy of ELA mice. Thus, microglial actions during development are powerful contributors to mechanisms by which ELA sculpts the connectivity of stress-regulating neurons, promoting vulnerability to stress and stress-related mental illnesses.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA.
| | - Annabel K Short
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Cassandra L Kooiker
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Manlin Shao
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Benjamin G Gunn
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Jaclyn Beck
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Xinglong Bai
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Stephanie M Law
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Julie C Savage
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Marie-Ève Tremblay
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
44
|
Pedrosa LRR, Coimbra GDS, Corrêa MG, Dias IA, Bahia CP. Time Window of the Critical Period for Neuroplasticity in S1, V1, and A1 Sensory Areas of Small Rodents: A Systematic Review. Front Neuroanat 2022; 16:763245. [PMID: 35370567 PMCID: PMC8970055 DOI: 10.3389/fnana.2022.763245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/04/2022] [Indexed: 12/09/2022] Open
Abstract
The plasticity of the central nervous system (CNS) allows the change of neuronal organization and function after environmental stimuli or adaptation after sensory deprivation. The so-called critical period (CP) for neuroplasticity is the time window when each sensory brain region is more sensitive to changes and adaptations. This time window is usually different for each primary sensory area: somatosensory (S1), visual (V1), and auditory (A1). Several intrinsic mechanisms are also involved in the start and end of the CP for neuroplasticity; however, which is its duration in S1, VI, and A1? This systematic review evaluated studies on the determination of these time windows in small rodents. The careful study selection and methodological quality assessment indicated that the CP for neuroplasticity is different among the sensory areas, and the brain maps are influenced by environmental stimuli. Moreover, there is an overlap between the time windows of some sensory areas. Finally, the time window duration of the CP for neuroplasticity is predominant in S1.
Collapse
|
45
|
Doss SV, Barbat-Artigas S, Lopes M, Pradhan BS, Prószyński TJ, Robitaille R, Valdez G. Expression and Roles of Lynx1, a Modulator of Cholinergic Transmission, in Skeletal Muscles and Neuromuscular Junctions in Mice. Front Cell Dev Biol 2022; 10:838612. [PMID: 35372356 PMCID: PMC8967655 DOI: 10.3389/fcell.2022.838612] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 01/31/2022] [Indexed: 11/21/2022] Open
Abstract
Lynx1 is a glycosylphosphatidylinositol (GPI)-linked protein shown to affect synaptic plasticity through modulation of nicotinic acetylcholine receptor (nAChR) subtypes in the brain. Because of this function and structural similarity to α-bungarotoxin, which binds muscle-specific nAChRs with high affinity, Lynx1 is a promising candidate for modulating nAChRs in skeletal muscles. However, little is known about the expression and roles of Lynx1 in skeletal muscles and neuromuscular junctions (NMJs). Here, we show that Lynx1 is expressed in skeletal muscles, increases during development, and concentrates at NMJs. We also demonstrate that Lynx1 interacts with muscle-specific nAChR subunits. Additionally, we present data indicating that Lynx1 deletion alters the response of skeletal muscles to cholinergic transmission and their contractile properties. Based on these findings, we asked if Lynx1 deletion affects developing and adult NMJs. Loss of Lynx1 had no effect on NMJs at postnatal day 9 (P9) and moderately increased their size at P21. Thus, Lynx1 plays a minor role in the structural development of NMJs. In 7- and 12-month-old mice lacking Lynx1, there is a marked increase in the incidence of NMJs with age- and disease-associated morphological alterations. The loss of Lynx1 also reduced the size of adult muscle fibers. Despite these effects, Lynx1 deletion did not alter the rate of NMJ reinnervation and stability following motor axon injury. These findings suggest that Lynx1 is not required during fast remodeling of the NMJ, as is the case during reformation following crushing of motor axons and development. Instead, these data indicate that the primary role of Lynx1 may be to maintain the structure and function of adult and aging NMJs.
Collapse
Affiliation(s)
- Sydney V. Doss
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, United States
| | | | - Mikayla Lopes
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Bhola Shankar Pradhan
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Laboratory of Synaptogenesis, Łukasiewicz Research Network—PORT Polish Center for Technology Development, Wrocław, Poland
| | - Tomasz J. Prószyński
- Laboratory of Synaptogenesis, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
- Laboratory of Synaptogenesis, Łukasiewicz Research Network—PORT Polish Center for Technology Development, Wrocław, Poland
| | - Richard Robitaille
- Département de Neurosciences, Université de Montréal, Montréal, QC, Canada
- Centre de Recherche Interdisciplinaire sur le Cerveau et L’Apprentissage (CIRCA), Montreal, QC, Canada
| | - Gregorio Valdez
- Department of Molecular Biology, Cellular Biology, and Biochemistry, Brown University, Providence, RI, United States
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI, United States
- Department of Neurology, Warren Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Gregorio Valdez,
| |
Collapse
|
46
|
Lesicko AMH, Angeloni CF, Blackwell JM, De Biasi M, Geffen MN. Cortico-fugal regulation of predictive coding. eLife 2022; 11:73289. [PMID: 35290181 PMCID: PMC8983050 DOI: 10.7554/elife.73289] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 03/12/2022] [Indexed: 11/13/2022] Open
Abstract
Sensory systems must account for both contextual factors and prior experience to adaptively engage with the dynamic external environment. In the central auditory system, neurons modulate their responses to sounds based on statistical context. These response modulations can be understood through a hierarchical predictive coding lens: responses to repeated stimuli are progressively decreased, in a process known as repetition suppression, whereas unexpected stimuli produce a prediction error signal. Prediction error incrementally increases along the auditory hierarchy from the inferior colliculus (IC) to the auditory cortex (AC), suggesting that these regions may engage in hierarchical predictive coding. A potential substrate for top-down predictive cues is the massive set of descending projections from the auditory cortex to subcortical structures, although the role of this system in predictive processing has never been directly assessed. We tested the effect of optogenetic inactivation of the auditory cortico-collicular feedback in awake mice on responses of IC neurons to stimuli designed to test prediction error and repetition suppression. Inactivation of the cortico-collicular pathway led to a decrease in prediction error in IC. Repetition suppression was unaffected by cortico-collicular inactivation, suggesting that this metric may reflect fatigue of bottom-up sensory inputs rather than predictive processing. We also discovered populations of IC units that exhibit repetition enhancement, a sequential increase in firing with stimulus repetition. Cortico-collicular inactivation led to a decrease in repetition enhancement in the central nucleus of IC, suggesting that it is a top-down phenomenon. Negative prediction error, a stronger response to a tone in a predictable rather than unpredictable sequence, was suppressed in shell IC units during cortico-collicular inactivation. These changes in predictive coding metrics arose from bidirectional modulations in the response to the standard and deviant contexts, such that units in IC responded more similarly to each context in the absence of cortical input. We also investigated how these metrics compare between the anesthetized and awake states by recording from the same units under both conditions. We found that metrics of predictive coding and deviance detection differ depending on the anesthetic state of the animal, with negative prediction error emerging in the central IC and repetition enhancement and prediction error being more prevalent in the absence of anesthesia. Overall, our results demonstrate that the auditory cortex provides cues about the statistical context of sound to subcortical brain regions via direct feedback, regulating processing of both prediction and repetition.
Collapse
Affiliation(s)
- Alexandria M H Lesicko
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| | | | - Jennifer M Blackwell
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, United States
| | - Mariella De Biasi
- Department of Psychiatry, University of Pennsylvania, Philadelphia, United States
| | - Maria N Geffen
- Department of Otorhinolaryngology, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
47
|
Holmberg E, Kataja EL, Davis EP, Pajulo M, Nolvi S, Hakanen H, Karlsson L, Karlsson H, Korja R. The Connection and Development of Unpredictability and Sensitivity in Maternal Care Across Early Childhood. Front Psychol 2022; 13:803047. [PMID: 35330718 PMCID: PMC8940198 DOI: 10.3389/fpsyg.2022.803047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/14/2022] [Indexed: 01/02/2023] Open
Abstract
Both patterns of maternal sensory signals and sensitive care have shown to be crucial elements shaping child development. However, research concerning these aspects of maternal care has focused mainly on maternal sensitivity with fewer studies evaluating the impact of patterns of maternal behaviors and changes in these indices across infancy and childhood. The aims of this study were to explore how maternal unpredictability of sensory signals and sensitivity develop and associate with each other from infancy to toddlerhood and whether elevated maternal depressive and anxiety symptoms relate to maternal unpredictable signals and sensitivity in toddlerhood. The study population consisted of 356 mother–child dyads assessed at 30 months; a subset of 103 mother–child dyads additionally participated in 8 months assessment. Maternal unpredictability and sensitivity were assessed from video-recorded free-play episodes at 8 and 30 months. Maternal depressive and anxiety symptoms were assessed with questionnaires at gestational weeks 14, 24, 34 and 3, 6, 12, and 24 months. Mean level of mothers’ unpredictability decreased on average whereas sensitivity did not change between infancy and toddlerhood. Both maternal unpredictability and sensitivity showed moderate level of individual stability from infancy to toddlerhood and these two measures were modestly correlated within each age. Elevated maternal depressive and anxiety symptoms were not related to unpredictability but related to lower maternal sensitivity in toddlerhood. These results identify unpredictable sensory signals as a characteristic of parental care that is independent of standard quality measures and suggest that it may be less influenced by maternal depressive and anxiety symptoms.
Collapse
Affiliation(s)
- Eeva Holmberg
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- *Correspondence: Eeva Holmberg,
| | - Eeva-Leena Kataja
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
| | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Pediatrics, University of California, Irvine, Irvine, CA, United States
| | - Marjukka Pajulo
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Child Psychiatry, University of Turku, Turku, Finland
| | - Saara Nolvi
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, Turku Institute for Advanced Studies, University of Turku, Turku, Finland
| | - Hetti Hakanen
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| | - Linnea Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Hospital District of Southwest Finland, Turku, Finland
| | - Hasse Karlsson
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Centre for Population Health Research, Turku University Hospital, University of Turku, Turku, Finland
- Department of Psychiatry, University of Turku and Hospital District of Southwest Finland, Turku, Finland
| | - Riikka Korja
- FinnBrain Birth Cohort Study, Turku Brain and Mind Center, Department of Clinical Medicine, University of Turku, Turku, Finland
- Department of Psychology and Speech-Language Pathology, University of Turku, Turku, Finland
| |
Collapse
|
48
|
Larsen B, Cui Z, Adebimpe A, Pines A, Alexander-Bloch A, Bertolero M, Calkins ME, Gur RE, Gur RC, Mahadevan AS, Moore TM, Roalf DR, Seidlitz J, Sydnor VJ, Wolf DH, Satterthwaite TD. A developmental reduction of the excitation:inhibition ratio in association cortex during adolescence. SCIENCE ADVANCES 2022; 8:eabj8750. [PMID: 35119918 PMCID: PMC8816330 DOI: 10.1126/sciadv.abj8750] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
Adolescence is hypothesized to be a critical period for the development of association cortex. A reduction of the excitation:inhibition (E:I) ratio is a hallmark of critical period development; however, it has been unclear how to assess the development of the E:I ratio using noninvasive neuroimaging techniques. Here, we used pharmacological fMRI with a GABAergic benzodiazepine challenge to empirically generate a model of E:I ratio based on multivariate patterns of functional connectivity. In an independent sample of 879 youth (ages 8 to 22 years), this model predicted reductions in the E:I ratio during adolescence, which were specific to association cortex and related to psychopathology. These findings support hypothesized shifts in E:I balance of association cortices during a neurodevelopmental critical period in adolescence.
Collapse
Affiliation(s)
- Bart Larsen
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zaixu Cui
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Azeez Adebimpe
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adam Pines
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aaron Alexander-Bloch
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Max Bertolero
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Monica E. Calkins
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Raquel E. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ruben C. Gur
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Arun S. Mahadevan
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tyler M. Moore
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David R. Roalf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jakob Seidlitz
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Valerie J. Sydnor
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel H. Wolf
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theodore D. Satterthwaite
- Penn Lifespan Neuroinformatics Center, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lifespan Brain Institute (LiBI) of Penn Medicine and CHOP, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
49
|
Mismatch negativity as an index of target engagement for excitation/inhibition-based treatment development: a double-blind, placebo-controlled, randomized, single-dose cross-over study of the serotonin type-3 receptor antagonist CVN058. Neuropsychopharmacology 2022; 47:711-718. [PMID: 34667294 PMCID: PMC8782925 DOI: 10.1038/s41386-021-01170-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 02/03/2023]
Abstract
Serotonin type-3 receptor (5-HT3R) antagonists show potential as a treatment for cognitive deficits in schizophrenia. CVN058, a brain-penetrant, potent and selective 5-HT3R antagonist, shows efficacy in rodent models of cognition and was well-tolerated in Phase-1 studies. We evaluated the target engagement of CVN058 using mismatch negativity (MMN) in a randomized, double-blind, placebo-controlled, cross-over study. Subjects were stable outpatients with schizophrenia or schizoaffective disorder treated with antipsychotics. Subjects were not permitted to use other 5-HT3R modulators or serotonin reuptake inhibitors. Each subject received a high (150 mg) and low (15 mg or 75 mg) oral dose of CVN058 and placebo in a randomized order across 3 single-day treatment visits separated by at least 1 week. The primary pre-registered outcome was amplitude of duration MMN. Amplitude of other MMN deviants (frequency, intensity, frequency modulation, and location), P50, P300 and auditory steady-state response (ASSR) were exploratory endpoints. 19 of 22 randomized subjects (86.4%) completed the study. Baseline PANSS scores indicated moderate impairment. CVN058 150 mg led to significant improvement vs. placebo on the primary outcome of duration MMN (p = 0.02, Cohen's d = 0.48). A significant treatment effect was also seen in a combined analysis across all MMN deviants (p < 0.001, d = 0.57). Effects on location MMN were independently significant (p < 0.007, d = 0.46). No other significant effects were seen for other deviants, doses or EEG measures. There were no clinically significant treatment related adverse effects. These results show MMN to be a sensitive target engagement biomarker for 5-HT3R, and support the potential utility of CVN058 in correcting the excitatory/inhibitory imbalance in schizophrenia.
Collapse
|
50
|
Knipper M, Singer W, Schwabe K, Hagberg GE, Li Hegner Y, Rüttiger L, Braun C, Land R. Disturbed Balance of Inhibitory Signaling Links Hearing Loss and Cognition. Front Neural Circuits 2022; 15:785603. [PMID: 35069123 PMCID: PMC8770933 DOI: 10.3389/fncir.2021.785603] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neuronal hyperexcitability in the central auditory pathway linked to reduced inhibitory activity is associated with numerous forms of hearing loss, including noise damage, age-dependent hearing loss, and deafness, as well as tinnitus or auditory processing deficits in autism spectrum disorder (ASD). In most cases, the reduced central inhibitory activity and the accompanying hyperexcitability are interpreted as an active compensatory response to the absence of synaptic activity, linked to increased central neural gain control (increased output activity relative to reduced input). We here suggest that hyperexcitability also could be related to an immaturity or impairment of tonic inhibitory strength that typically develops in an activity-dependent process in the ascending auditory pathway with auditory experience. In these cases, high-SR auditory nerve fibers, which are critical for the shortest latencies and lowest sound thresholds, may have either not matured (possibly in congenital deafness or autism) or are dysfunctional (possibly after sudden, stressful auditory trauma or age-dependent hearing loss linked with cognitive decline). Fast auditory processing deficits can occur despite maintained basal hearing. In that case, tonic inhibitory strength is reduced in ascending auditory nuclei, and fast inhibitory parvalbumin positive interneuron (PV-IN) dendrites are diminished in auditory and frontal brain regions. This leads to deficits in central neural gain control linked to hippocampal LTP/LTD deficiencies, cognitive deficits, and unbalanced extra-hypothalamic stress control. Under these conditions, a diminished inhibitory strength may weaken local neuronal coupling to homeostatic vascular responses required for the metabolic support of auditory adjustment processes. We emphasize the need to distinguish these two states of excitatory/inhibitory imbalance in hearing disorders: (i) Under conditions of preserved fast auditory processing and sustained tonic inhibitory strength, an excitatory/inhibitory imbalance following auditory deprivation can maintain precise hearing through a memory linked, transient disinhibition that leads to enhanced spiking fidelity (central neural gain⇑) (ii) Under conditions of critically diminished fast auditory processing and reduced tonic inhibitory strength, hyperexcitability can be part of an increased synchronization over a broader frequency range, linked to reduced spiking reliability (central neural gain⇓). This latter stage mutually reinforces diminished metabolic support for auditory adjustment processes, increasing the risks for canonical dementia syndromes.
Collapse
Affiliation(s)
- Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
- *Correspondence: Marlies Knipper,
| | - Wibke Singer
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Kerstin Schwabe
- Experimental Neurosurgery, Department of Neurosurgery, Hannover Medical School, Hanover, Germany
| | - Gisela E. Hagberg
- Department of Biomedical Magnetic Resonance, University Hospital Tübingen (UKT), Tübingen, Germany
- High-Field Magnetic Resonance, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Yiwen Li Hegner
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Tübingen Hearing Research Center (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Christoph Braun
- MEG Center, University of Tübingen, Tübingen, Germany
- Center of Neurology, Hertie-Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Rüdiger Land
- Department of Experimental Otology, Institute for Audioneurotechnology, Hannover Medical School, Hanover, Germany
| |
Collapse
|